Dialogue of Probiotics with the Host 2.0

A special issue of Microorganisms (ISSN 2076-2607). This special issue belongs to the section "Gut Microbiota".

Deadline for manuscript submissions: closed (30 September 2023) | Viewed by 19714

Special Issue Editors


E-Mail Website
Guest Editor
Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos, Consejo Superior de Investigaciones Científicas (Spanish National Research Council), Valencia, Spain
Interests: functional food; molecular microbiology; biotechnology; gut microbiota; probiotics; host-bacteria communication
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos, Consejo Superior de Investigaciones Científicas (Spanish National Research Council), Valencia, Spain
Interests: gut microbiome; host-bacteria interactions; probiotics; postbiotics
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

This Special Issue is a continuation of our 2022 Special Issue "Dialogue of Probiotics with the Host".

The canonical concepts behind the definition of probiotics have remained unaltered for almost 20 years, establishing that probiotics must be well-characterized living microorganisms with proven beneficial effects that are safe to use. Different terms with the “biotic” root are used, referring to their pharmacological role (i.e. pharmabiotics), their target (i.e. psycobiotics) and their joint effect/administration with prebiotics (i.e. symbiotics). There are newer and broader concepts that involve non-living components or their metabolic products (i.e. postbiotics, parabiotics). Besides the terminology, this is one of the most dynamic scientific fields in the area of health and nutrition, which is heavily supported by robust scientific evidence, and where the forefront of omics, bioinformatics and molecular biology techniques has been put to work. Microbiome research works are on their way to overcome classical concepts and they have revealed a number of new bacterial species significantly related to health benefits that are considered the last generation of probiotics.

This Special Issue will be dealing with research works on diverse aspects of the interaction between well-established or new—to be appointed—probiotics with the host; this will include well founded contributions, from preclinical research (in vitro or in vivo assays) to human or animal trials.

As Guest Editors of this Special Issue, we would like to invite you to submit research articles, review articles and short communications dealing with different aspects of the communication between probiotics and the host.

Prof. Dr. Gaspar Pérez-Martínez
Dr. Christine Bäuerl
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Microorganisms is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • probiotics
  • postbiotics
  • symbiotics
  • disease
  • immune system
  • gut–brain axis
  • neurotransmitter
  • human trial
  • animal assay
  • health effect
  • bacteria–host interaction
  • proteomics
  • transcriptomics
  • metabolomics
  • metagenomics
  • gut microbiota

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

14 pages, 4971 KiB  
Article
The Effect of Probiotics in a Milk Replacer on Leukocyte Differential Counts, Phenotype, and Function in Neonatal Dairy Calves
by Susan D. Eicher, Janice E. Kritchevsky, Keith A. Bryan and Carol G. Chitko-McKown
Microorganisms 2023, 11(11), 2620; https://doi.org/10.3390/microorganisms11112620 - 24 Oct 2023
Viewed by 860
Abstract
Probiotics have been investigated for many health benefits; however, few studies have been performed to determine the effects of oral probiotics on peripheral blood and respiratory immune cells in cattle. Our objectives were to determine changes in health and growth status, differential blood [...] Read more.
Probiotics have been investigated for many health benefits; however, few studies have been performed to determine the effects of oral probiotics on peripheral blood and respiratory immune cells in cattle. Our objectives were to determine changes in health and growth status, differential blood cell counts and function, and blood and lung cell function using flow cytometry and PCR in dairy calves fed a milk replacer with (PRO, N = 10) or without (CON, N = 10) the addition of probiotics to the milk replacer and dry rations from birth to weaning. Performance and clinical scores were not different between the treatment groups. Treatment-by-day interactions for peripheral blood leukocyte populations differed in cell number and percentages. A greater percentage of leukocytes expressed the cell surface markers CD3, CD4, CD8, CD11b, and CD205 on d 21 in CON animals. Lung lavages were performed on five animals from each treatment group on d 52. There were no differences between treatment groups for the expression of cytokines and Toll-Like Receptors as measured using Polymerase Chain Reaction, possibly due to the small sample size. Oral probiotics appear to affect peripheral blood immune cells and function. Their effect on overall calf health remains to be determined. Full article
(This article belongs to the Special Issue Dialogue of Probiotics with the Host 2.0)
Show Figures

Figure 1

17 pages, 3509 KiB  
Article
Lactobacillus helveticus Isolated from Raw Milk Improves Liver Function, Hepatic Steatosis, and Lipid Metabolism in Non-Alcoholic Fatty Liver Disease Mouse Model
by Hyeonji Kim, Kippeum Lee, Ju-Yeon Kim, Jae-Jung Shim, Junghyun Lim, Joo-Yun Kim and Jung-Lyoul Lee
Microorganisms 2023, 11(10), 2466; https://doi.org/10.3390/microorganisms11102466 - 30 Sep 2023
Viewed by 1489
Abstract
Here, we show that Lactiplantibacillus plantarum LP158 (LP158), Lactobacillus helveticus HY7804 (HY7804), and Lacticaseibacillus paracasei LPC226 (LPC226) isolated from raw milk alleviate non-alcoholic fatty acid disease (NAFLD) in a C57BL/6 mouse model. Lactic acid bacteria (LAB) were screened for their ability to inhibit [...] Read more.
Here, we show that Lactiplantibacillus plantarum LP158 (LP158), Lactobacillus helveticus HY7804 (HY7804), and Lacticaseibacillus paracasei LPC226 (LPC226) isolated from raw milk alleviate non-alcoholic fatty acid disease (NAFLD) in a C57BL/6 mouse model. Lactic acid bacteria (LAB) were screened for their ability to inhibit fatty acid accumulation in palmitic acid (PA)-treated HepG2 cells, and three strains were selected based on the results. We also investigated hemolytic activity and antibiotic resistance of the three strains. LP158, HY7804, and LPC226 suppressed expression of mRNA encoding genes related to lipogenesis, and increased expression of genes related to β-oxidation, in a PA-induced HepG2 cell model. Moreover, when LP158, HY7804, and LPC226 were administered at 109 CFU/kg/day for 8 weeks to mice with dietary-induced NAFLD, they all modulated blood biochemistry markers and reduced steatosis in liver tissue. Also, all three strains significantly reduced expression of mRNA encoding lipogenesis genes (Fasn, Acaca, and Srebp-1c) and inflammatory factors (Tnfα and Ccl-2) and fibrosis factors, and increased expression of a β-oxidation gene (Acox1) in the liver. In particular, HY7804 showed the strongest effects both in vitro and in vivo. Therefore, HY7804, LP158, and LPC226 can be proposed as potential supplements that can improve NAFLD through anti-steatosis, anti-inflammatory, and anti-fibrotic effects. Full article
(This article belongs to the Special Issue Dialogue of Probiotics with the Host 2.0)
Show Figures

Figure 1

16 pages, 3178 KiB  
Article
Lactobacillus Probiotic Strains Differ in Their Ability to Adhere to Human Lung Epithelial Cells and to Prevent Adhesion of Clinical Isolates of Pseudomonas aeruginosa from Cystic Fibrosis Lung
by Giovanna Batoni, Esingül Kaya, Elisa Catelli, Sabrina Quinti, Matteo Botti, Alessandro De Carli, Marta Bianchi, Giuseppantonio Maisetta and Semih Esin
Microorganisms 2023, 11(7), 1707; https://doi.org/10.3390/microorganisms11071707 - 29 Jun 2023
Cited by 3 | Viewed by 1381
Abstract
The field of probiotic applications is rapidly expanding, including their use for the control of respiratory tract infections. Nevertheless, probiotics ability to colonize the lung environment and to compete with pulmonary pathogens is still a poorly investigated research area. In this study, we [...] Read more.
The field of probiotic applications is rapidly expanding, including their use for the control of respiratory tract infections. Nevertheless, probiotics ability to colonize the lung environment and to compete with pulmonary pathogens is still a poorly investigated research area. In this study, we aimed to evaluate the adhesion ability of a number of commercial probiotic strains to the human lung epithelial cell line A549. Furthermore, we assessed probiotic ability to prevent host cell adhesion of one of the major lung pathogens in cystic fibrosis, Pseudomonas aeruginosa, and to reduce the pathogen-induced inflammatory response of human peripheral blood mononuclear cells (PBMCs) in terms of cytokine release. Lactobacillus acidophilus displayed the highest adhesion ability to A549 cells evaluated as percent of adhered bacteria compared to the inoculum. In agreement with such an observation, L. acidophilus was the most efficient in preventing adhesion to A549 cells of a P. aeruginosa isolate from CF sputum. Three-color fluorescence labeling of A549 cells, P. aeruginosa, and L. acidophilus, and confocal microcopy image analyses revealed a likely exclusion effect played by both live and UV-killed L. acidophilus towards P. aeruginosa. Such results were confirmed by CFU count. When co-cultured with PBMCs, both live and UV-killed L. acidophilus reduced the amount of IL-1β and IL-6 in culture supernatants in a statistically significant manner. Overall, the results obtained point to L. acidophilus as an interesting candidate for further studies for a potential aerogenous administration to control P. aeruginosa infections. Full article
(This article belongs to the Special Issue Dialogue of Probiotics with the Host 2.0)
Show Figures

Figure 1

21 pages, 2343 KiB  
Article
Bifidobacterium breve PRL2020: Antibiotic-Resistant Profile and Genomic Detection of Antibiotic Resistance Determinants
by Francesco Di Pierro, Ilenia Campedelli, Patrick De Marta, Fabio Fracchetti, Antonio Del Casale, Ilaria Cavecchia, Mariarosaria Matera, Massimiliano Cazzaniga, Alexander Bertuccioli, Luigina Guasti and Nicola Zerbinati
Microorganisms 2023, 11(7), 1649; https://doi.org/10.3390/microorganisms11071649 - 24 Jun 2023
Cited by 2 | Viewed by 1689
Abstract
Antibiotics are one of the greatest scientific achievements of modern medicine, but excessive use is creating challenges for the future of medicine. Antibiotic resistance (AR) is thought to cause changes in bowel habits and an increased risk of gastroenteritis, but it may also [...] Read more.
Antibiotics are one of the greatest scientific achievements of modern medicine, but excessive use is creating challenges for the future of medicine. Antibiotic resistance (AR) is thought to cause changes in bowel habits and an increased risk of gastroenteritis, but it may also increase the risk of overweight, obesity, autoimmune and atopic diseases, and a low response to vaccines and cancer, likely mediated by antibiotic-induced gut dysbiosis. Probiotic add-on therapy could partially prevent antibiotic-induced gut dysbiosis, but their antibiotic sensitivity features likely limits this potential. The EFSA (European Food Safety Authority) guidelines consider the use of probiotics whose antibiotic-resistant profile could be transferable an important hazard. Recently, a strain of B. breve (PRL2020) has shown to be resistant to amoxicillin and amoxicillin-clavulanate (AC) by applying the microdilution protocol according EFSA guidelines. After verifying that horizontal gene transfer is unlikely to take place, this feature suggests its concomitant use with these specific antibiotics. The results of our tests demonstrated that the strain PRL2020 is indeed endowed with amoxicillin- and AC-resistant properties and that it is also insensitive to ampicillin. In-depth analysis of the annotated genome sequence of B. breve PRL2020 was employed to query the Comprehensive Antibiotic Resistance Database (CARD) using Resistance Gene Identifier (RGI) software (version 5.2.1). The similarity among the AR determinants found was studied through nucleotide sequence alignment, and it was possible to verify not only the absence of genes explaining these features in the flanking regions but also the presence of genetic sequences (rpoB and erm(X)) putatively responsible for rifampicin and erythromycin resistance. Both features are not phenotypically expressed, and for these antibiotics, the strain is within the EFSA limits. Analysis of the flanking regions of these genes revealed possible mobile elements upstream and downstream only in the case of the erm(X) gene, but the features of the Insertion Sequences (IS) are described as not to cause horizontal transfer. Our findings on strain PRL2020 demonstrate that its AR profile is compatible with antibiotics when taken with the aim of reducing the risk of dysbiosis. Full article
(This article belongs to the Special Issue Dialogue of Probiotics with the Host 2.0)
Show Figures

Figure 1

26 pages, 8581 KiB  
Article
Synergistic Immunostimulatory Activities of Probiotic Strains, Leuconostoc lactis and Weissella cibaria, and the Prebiotic Oligosaccharides They Produce
by Seoyoung Jeong, Ayeon Kwon, Huijin Jeong and Young-Seo Park
Microorganisms 2023, 11(5), 1354; https://doi.org/10.3390/microorganisms11051354 - 22 May 2023
Cited by 1 | Viewed by 1410
Abstract
Synbiotics contain health-beneficial bacteria, i.e., probiotics and prebiotics selectively utilized by the probiotics. Herein, three probiotic strains, Leuconostoc lactis CCK940, L. lactis SBC001, and Weissella cibaria YRK005, and the oligosaccharides produced by these strains (CCK, SBC, and YRK, respectively) were used to prepare [...] Read more.
Synbiotics contain health-beneficial bacteria, i.e., probiotics and prebiotics selectively utilized by the probiotics. Herein, three probiotic strains, Leuconostoc lactis CCK940, L. lactis SBC001, and Weissella cibaria YRK005, and the oligosaccharides produced by these strains (CCK, SBC, and YRK, respectively) were used to prepare nine synbiotic combinations. Macrophages (RAW 264.7) were treated with these synbiotic combinations and the corresponding lactic acid bacteria and oligosaccharides alone to evaluate the treatments’ immunostimulatory activities. The level of nitric oxide (NO) production was significantly higher in the macrophages treated with the synbiotics than in those treated with the corresponding probiotic strains and the oligosaccharide alone. The immunostimulatory activities of the synbiotics increased regardless of the probiotic strain and the type of oligosaccharide used. The expressions of tissue necrosis factor-α, interleukin-1β, cyclooxygenase-2, inducible NO synthase genes, and extracellular-signal-regulated and c-Jun N-terminal kinases were significantly higher in the macrophages treated with the three synbiotics than in those treated with the corresponding strains or with the oligosaccharides alone. These results indicate that the synergistic immunostimulatory activities of probiotics and the prebiotics they produced in the studied synbiotic preparations resulted from the activation of the mitogen-activated protein-kinase-signaling pathway. This study suggests the combined use of these probiotics and prebiotics in the development of synbiotic preparations as health supplements. Full article
(This article belongs to the Special Issue Dialogue of Probiotics with the Host 2.0)
Show Figures

Figure 1

Review

Jump to: Research

24 pages, 1274 KiB  
Review
Probiotics and Their Bioproducts: A Promising Approach for Targeting Methicillin-Resistant Staphylococcus aureus and Vancomycin-Resistant Enterococcus
by Manav Jain, Gideon Stitt, Luke Son and Elena Y. Enioutina
Microorganisms 2023, 11(10), 2393; https://doi.org/10.3390/microorganisms11102393 - 25 Sep 2023
Cited by 4 | Viewed by 3470
Abstract
Antibiotic resistance is a serious global health problem that poses a threat to the successful treatment of various bacterial infections, especially those caused by methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE). Conventional treatment of MRSA and VRE infections is challenging and often [...] Read more.
Antibiotic resistance is a serious global health problem that poses a threat to the successful treatment of various bacterial infections, especially those caused by methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE). Conventional treatment of MRSA and VRE infections is challenging and often requires alternative or combination therapies that may have limited efficacy, higher costs, and/or more adverse effects. Therefore, there is an urgent need to find new strategies to combat antibiotic-resistant bacteria. Probiotics and antimicrobial peptides (AMPs) are two promising approaches that have shown potential benefits in various diseases. Probiotics are live microorganisms that confer health benefits to the host when administered in adequate amounts. AMPs, usually produced with probiotic bacteria, are short amino acid sequences that have broad-spectrum activity against bacteria, fungi, viruses, and parasites. Both probiotics and AMPs can modulate the host immune system, inhibit the growth and adhesion of pathogens, disrupt biofilms, and enhance intestinal barrier function. In this paper, we review the current knowledge on the role of probiotics and AMPs in targeting multi-drug-resistant bacteria, with a focus on MRSA and VRE. In addition, we discuss future directions for the clinical use of probiotics. Full article
(This article belongs to the Special Issue Dialogue of Probiotics with the Host 2.0)
Show Figures

Figure 1

18 pages, 2468 KiB  
Review
The Possible Role of Probiotic Supplementation in Inflammation: A Narrative Review
by Alessandro Colletti, Marzia Pellizzato and Arrigo Francesco Cicero
Microorganisms 2023, 11(9), 2160; https://doi.org/10.3390/microorganisms11092160 - 26 Aug 2023
Cited by 2 | Viewed by 3156
Abstract
The fine balance between symbiotic and potentially opportunistic and/or pathogenic microorganisms can undergo quantitative alterations, which, when associated with low intestinal biodiversity, could be responsible for the development of gut inflammation and the so-called “intestinal dysbiosis”. This condition is characterized by the disbalance [...] Read more.
The fine balance between symbiotic and potentially opportunistic and/or pathogenic microorganisms can undergo quantitative alterations, which, when associated with low intestinal biodiversity, could be responsible for the development of gut inflammation and the so-called “intestinal dysbiosis”. This condition is characterized by the disbalance of a fine synergistic mechanism involving the mucosal barrier, the intestinal neuroendocrine system, and the immune system that results in an acute inflammatory response induced by different causes, including viral or bacterial infections of the digestive tract. More frequently, however, dysbiosis is induced slowly and subtly by subliminal causal factors, resulting in a chronic condition related to different diseases affecting the digestive tract and other organs and apparatuses. Studies on animal models, together with studies on humans, highlight the significant role of the gut microbiota and microbiome in the occurrence of inflammatory conditions such as metabolic syndrome and cardiovascular diseases (CVDs); neurodegenerative, urologic, skin, liver, and kidney pathologies; and premature aging. The blood translocation of bacterial fragments has been found to be one of the processes linked to gut dysbiosis and responsible for the possible occurrence of “metabolic endotoxemia” and systemic inflammation, associated with an increased risk of oxidative stress and related diseases. In this context, supplementation with different probiotic strains has been shown to restore gut eubiosis, especially if administered in long-term treatments. The aim of this review is to describe the anti-inflammatory effects of specific probiotic strains observed in clinical trials and the respective indications, highlighting the differences in efficacy depending on strain, formulation, time and duration of treatment, and dosage used. Full article
(This article belongs to the Special Issue Dialogue of Probiotics with the Host 2.0)
Show Figures

Figure 1

14 pages, 304 KiB  
Review
Lactobacillus Bacteremia and Probiotics: A Review
by Ravina Kullar, Ellie J. C. Goldstein, Stuart Johnson and Lynne V. McFarland
Microorganisms 2023, 11(4), 896; https://doi.org/10.3390/microorganisms11040896 - 30 Mar 2023
Cited by 19 | Viewed by 4381
Abstract
Lactobacilli are widely found in nature, are commensal microbes in humans, and are commonly used as probiotics. Concerns about probiotic safety have arisen due to reports of bacteremia and other Lactobacillus-associated infections. We reviewed the literature for articles on the pathogenicity of [...] Read more.
Lactobacilli are widely found in nature, are commensal microbes in humans, and are commonly used as probiotics. Concerns about probiotic safety have arisen due to reports of bacteremia and other Lactobacillus-associated infections. We reviewed the literature for articles on the pathogenicity of Lactobacillus spp. bacteremia and reports of probiotics in these patients. Our aim is to review these articles and update the present knowledge on the epidemiology of Lactobacillus spp. bacteremia and determine the role of probiotics in Lactobacillus bacteremia. Lactobacillus bacteremia is infrequent but has a higher risk of mortality and risk factors, including severe underlying diseases, immune system suppression, admission to intensive care units, and use of central venous catheters. A variety of Lactobacillus species may cause bacteremia and may or may not be associated with probiotic exposure. To determine if oral probiotics are the source of these infections, the blood isolates and the oral probiotic strain(s) must be compared by sensitive identification methods. The prevalence of Lactobacillus bacteremia is infrequent but is more common in patients taking probiotics compared to those not taking probiotics. Three probiotics (Lacticaseibacillus rhamnosus GG, Lactiplantibacillus plantarum, and Lacticaseibacillus paracasei) were directly linked with blood isolates from bacteremia patients using molecular identification assays. Full article
(This article belongs to the Special Issue Dialogue of Probiotics with the Host 2.0)
Back to TopTop