1
|
Alkhayyat R, Abbas A, Quinn CM, Rakha EA. Tumour 63 protein (p63) in breast pathology: biology, immunohistochemistry, diagnostic applications, and pitfalls. Histopathology 2024; 84:723-741. [PMID: 38012539 DOI: 10.1111/his.15101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023]
Abstract
Tumour protein 63 (p63) is a transcription factor of the p53 gene family, encoded by the TP63 gene located at chromosome 3q28, which regulates the activity of genes involved in growth and development of the ectoderm and derived tissues. p63 protein is normally expressed in the nuclei of the basal cell layer of glandular organs, including breast, in squamous epithelium and in urothelium. p63 immunohistochemical (IHC) staining has several applications in diagnostic breast pathology. It is commonly used to demonstrate myoepithelial cells at the epithelial stromal interface to differentiate benign and in situ lesions from invasive carcinoma and to characterize and classify papillary lesions including the distinction of breast intraduct papilloma from skin hidradenoma. p63 IHC is also used to identify and profile lesions showing myoepithelial cell and/or squamous differentiation, e.g. adenomyoepithelioma, salivary gland-like tumours including adenoid cystic carcinoma, and metaplastic breast carcinoma including low-grade adenosquamous carcinoma. This article reviews the applications of p63 IHC in diagnostic breast pathology and outlines a practical approach to the diagnosis and characterization of breast lesions through the identification of normal and abnormal p63 protein expression. The biology of p63, the range of available antibodies with emphasis on staining specificity and sensitivity, and pitfalls in interpretation are also discussed. The TP63 gene in humans, which shows a specific genomic structure, resulting in either TAp63 (p63) isoform or ΔNp63 (p40) isoform. As illustrated in the figure, both isoforms contain a DNA-binding domain (Orange box) and an oligomerization domain (Grey box). TAp63 contains an N-terminal transactivation (TA) domain (Green box), while ΔNp63 has an alternative terminus (Yellow box). Antibodies against conventional pan-p63 (TP63) bind to the DNA binding domain common to both isoforms (TAp63 and p40) and does not distinguish between them. Antibodies against TAp63 bind to the N-terminal TA domain, while antibodies specific to ΔNp63 (p40) bind to the alternative terminus. Each isoform has variant isotypes (α, β, γ, δ, and ε).
Collapse
Affiliation(s)
- Rabab Alkhayyat
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospitals, Nottingham, UK
- Department of Pathology, Salmaniya Medical Complex, Government Hospitals, Manama, Kingdom of Bahrain
| | - Areeg Abbas
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospitals, Nottingham, UK
| | - Cecily M Quinn
- Irish National Breast Screening Program, Department of Histopathology, St. Vincent's University Hospital, Dublin, School of Medicine, University College Dublin, Dublin, Ireland
| | - Emad A Rakha
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospitals, Nottingham, UK
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Pathology, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
2
|
Kaur H, Ali SA, Short SP, Williams CS, Goettel JA, Washington MK, Peek RM, Acra SA, Yan F. Identification of a functional peptide of a probiotic bacterium-derived protein for the sustained effect on preventing colitis. Gut Microbes 2023; 15:2264456. [PMID: 37815528 PMCID: PMC10566403 DOI: 10.1080/19490976.2023.2264456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 09/25/2023] [Indexed: 10/11/2023] Open
Abstract
Several probiotic-derived factors have been identified as effectors of probiotics for exerting beneficial effects on the host. However, there is a paucity of studies to elucidate mechanisms of their functions. p40, a secretory protein, is originally isolated from a probiotic bacterium, Lactobacillus rhamnosus GG. Thus, this study aimed to apply structure-functional analysis to define the functional peptide of p40 that modulates the epigenetic program in intestinal epithelial cells for sustained prevention of colitis. In silico analysis revealed that p40 is composed of a signal peptide (1-28 residues) followed by a coiled-coil domain with uncharacterized function on the N-terminus, a linker region, and a β-sheet domain with high homology to CHAP on the C-terminus. Based on the p40 three-dimensional structure model, two recombinant p40 peptides were generated, p40N120 (28-120 residues) and p40N180 (28-180 residues) that contain first two and first three coiled coils, respectively. Compared to full-length p40 (p40F) and p40N180, p40N120 showed similar or higher effects on up-regulating expression of Setd1b (encoding a methyltransferase), promoting mono- and trimethylation of histone 3 on lysine 4 (H3K4me1/3), and enhancing Tgfb gene expression and protein production that leads to SMAD2 phosphorylation in human colonoids and a mouse colonic epithelial cell line. Furthermore, supplementation with p40F and p40N120 in early life increased H3K4me1, Tgfb expression and differentiation of regulatory T cells (Tregs) in the colon, and mitigated disruption of epithelial barrier and inflammation induced by DSS in adult mice. This study reveals the structural feature of p40 and identifies a functional peptide of p40 that could maintain intestinal homeostasis.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Syed Azmal Ali
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Sarah P. Short
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Jeremy A. Goettel
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M. Kay Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Richard M. Peek
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sari A. Acra
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fang Yan
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
3
|
Lopez-Escalera S, Lund ML, Hermes GDA, Choi BSY, Sakamoto K, Wellejus A. In Vitro Screening for Probiotic Properties of Lactobacillus and Bifidobacterium Strains in Assays Relevant for Non-Alcoholic Fatty Liver Disease Prevention. Nutrients 2023; 15:nu15102361. [PMID: 37242245 DOI: 10.3390/nu15102361] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial metabolic disorder that poses health challenges worldwide and is expected to continue to rise dramatically. NAFLD is associated with metabolic syndrome, type 2 diabetes mellitus, and impaired gut health. Increased gut permeability, caused by disturbance of tight junction proteins, allows passage of damaging microbial components that, upon reaching the liver, have been proposed to trigger the release of inflammatory cytokines and generate cellular stress. A growing body of research has suggested the utilization of targeted probiotic supplements as a preventive therapy to improve gut barrier function and tight junctions. Furthermore, specific microbial interactions and metabolites induce the secretion of hormones such as GLP-1, resulting in beneficial effects on liver health. To increase the likelihood of finding beneficial probiotic strains, we set up a novel screening platform consisting of multiple in vitro and ex vivo assays for the screening of 42 bacterial strains. Analysis of transepithelial electrical resistance response via co-incubation of the 42 bacterial strains with human colonic cells (Caco-2) revealed improved barrier integrity. Then, strain-individual metabolome profiling was performed revealing species-specific clusters. GLP-1 secretion assay with intestinal secretin tumor cell line (STC-1) found at least seven of the strains tested capable of enhancing GLP-1 secretion in vitro. Gene expression profiling in human biopsy-derived intestinal organoids was performed using next generation sequencing transcriptomics post bacterial co-incubation. Here, different degrees of immunomodulation by the increase in certain cytokine and chemokine transcripts were found. Treatment of mouse primary hepatocytes with selected highly produced bacterial metabolites revealed that indole metabolites robustly inhibited de novo lipogenesis. Collectively, through our comprehensive bacterial screening pipeline, not previously ascribed strains from both Lactobacillus and Bifidobacterium genera were proposed as potential probiotics based on their ability to increase epithelial barrier integrity and immunity, promote GLP-1 secretion, and produce metabolites relevant to liver health.
Collapse
Affiliation(s)
- Silvia Lopez-Escalera
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
- Fakultät für Biowissenschaften, Friedrich-Schiller Universität Jena, Bachstraβe 18K, 07743 Jena, Germany
| | - Mari L Lund
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
| | - Gerben D A Hermes
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
| | - Béatrice S-Y Choi
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Anja Wellejus
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
| |
Collapse
|
4
|
da Silva Barreira D, Laurent J, Lourenço J, Novion Ducassou J, Couté Y, Guzzo J, Rieu A. Membrane vesicles released by Lacticaseibacillus casei BL23 inhibit the biofilm formation of Salmonella Enteritidis. Sci Rep 2023; 13:1163. [PMID: 36670157 PMCID: PMC9859808 DOI: 10.1038/s41598-023-27959-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/10/2023] [Indexed: 01/22/2023] Open
Abstract
Biofilms represent a major concern in the food industry and healthcare. The use of probiotic bacteria and their derivatives as an alternative to conventional treatments to fight biofilm development is a promising option that has provided convincing results in the last decades. Recently, membrane vesicles (MVs) produced by probiotics have generated considerable interest due to the diversity of roles they have been associated with. However, the antimicrobial activity of probiotic MVs remains to be studied. In this work, we showed that membrane vesicles produced by Lacticaseibacillus casei BL23 (LC-MVs) exhibited strong antibiofilm activity against Salmonella enterica serovar Enteritidis (S. Enteritidis) without affecting bacterial growth. Furthermore, we found that LC-MVs affected the early stages of S. Enteritidis biofilm development and prevented attachment of bacteria to polystyrene surfaces. Importantly, LC-MVs did not impact the biomass of already established biofilms. We also demonstrated that the antibiofilm activity depended on the proteins associated with the LC-MV fraction. Finally, two peptidoglycan hydrolases (PGHs) were found to be associated with the antibiofilm activity of LC-MVs. Overall, this work allowed to identify the antibiofilm properties of LC-MVs and paved the way for the use of probiotic MVs against the development of negative biofilms.
Collapse
Affiliation(s)
- David da Silva Barreira
- Université de Bourgogne Franche-Comté (UBFC), AgroSup Dijon, UMR PAM A 02.102, 21000, Dijon, France
| | - Julie Laurent
- Université de Bourgogne Franche-Comté (UBFC), AgroSup Dijon, UMR PAM A 02.102, 21000, Dijon, France
| | - Jessica Lourenço
- Université de Bourgogne Franche-Comté (UBFC), AgroSup Dijon, UMR PAM A 02.102, 21000, Dijon, France
| | - Julia Novion Ducassou
- Univ. Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, R2048, 38000, Grenoble, France
| | - Yohann Couté
- Univ. Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, R2048, 38000, Grenoble, France
| | - Jean Guzzo
- Université de Bourgogne Franche-Comté (UBFC), AgroSup Dijon, UMR PAM A 02.102, 21000, Dijon, France
| | - Aurélie Rieu
- Université de Bourgogne Franche-Comté (UBFC), AgroSup Dijon, UMR PAM A 02.102, 21000, Dijon, France.
| |
Collapse
|
5
|
Pérez Martínez G, Giner-Pérez L, Castillo-Romero KF. Bacterial extracellular vesicles and associated functional proteins in fermented dairy products with Lacticaseibacillus paracasei. Front Microbiol 2023; 14:1165202. [PMID: 37152726 PMCID: PMC10157241 DOI: 10.3389/fmicb.2023.1165202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Cells of all kingdoms produce extracellular vesicles (EVs); hence, they are present in most environments and body fluids. Lacticaseibacillus paracasei produces EVs that have attached biologically active proteins (P40 and P75). In this study, EV and functional proteins were found in five different commercial dairy-fermented products carrying L. paracasei. Strains present in those products were isolated, and with one exception, all produced small EVs (24-47 d.nm) carrying P40 and P75. In order to winnow bacterial EV from milk EV, products were subjected to centrifugal fractionation at 15,000 × g (15 K), 33,000 × g (33 K), and 100,000 × g (100 K). P75 was present in all supernatants and pellets, but P40 was only found in two products bound to the 15 and 33 K pellets, and 16S rDNA of L. paracasei could be amplified from all 100 K EVs, indicating the presence of L. paracasei EV. To investigate the interactions of bacterial EV and proteins with milk EV, L. paracasei BL23 EV was added to three commercial UHT milk products. Small-size vesicles (50-60 d.nm) similar to L. paracasei BL23 EV were found in samples from 100 K centrifugations, but intriguingly, P40 and P75 were bound to EV in 15 and 33 K pellets, containing bovine milk EV of larger size (200-300 d.nm). Sequencing 16S rDNA bands amplified from EV evidenced the presence of bacterial EVs of diverse origins in milk and fermented products. Furthermore, L. paracasei 16S rDNA could be amplified with species-specific primers from all samples, showing the presence of L. paracasei EV in all EV fractions (15, 33, and 100 K), suggesting that these bacterial EVs possibly aggregate and are co-isolated with EV from milk. P40 and P75 proteins would be interacting with specific populations of milk EV (15 and 33 K) because they were detected bound to them in fermented products and milk, and this possibly forced the sedimentation of part of L. paracasei EV at lower centrifugal forces. This study has solved technically complex problems and essential questions which will facilitate new research focusing on the molecular behavior of probiotics during fermentation and the mechanisms of action mediating the health benefits of fermented products.
Collapse
Affiliation(s)
- Gaspar Pérez Martínez
- Laboratory of Lactic Acid Bacteria and Probiotics, Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
- *Correspondence: Gaspar Pérez Martínez
| | - Lola Giner-Pérez
- Laboratory of Lactic Acid Bacteria and Probiotics, Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Keshia F. Castillo-Romero
- Laboratory of Lactic Acid Bacteria and Probiotics, Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
- School of Engineering and Sciences, Tecnológico de Monterrey, Monterrey, Mexico
| |
Collapse
|
6
|
Transcriptional responses of human intestinal epithelial HT-29 cells to spore-displayed p40 derived from Lacticaseibacillus rhamnosus GG. BMC Microbiol 2022; 22:316. [PMID: 36550414 PMCID: PMC9772600 DOI: 10.1186/s12866-022-02735-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUNDS The aims of this study were to construct spore-displayed p40, a Lacticaseibacillus rhamnosus GG-derived soluble protein, using spore surface display technology and to evaluate transcriptional responses in human intestinal epithelial cells. RESULTS p40 was displayed on the surface of Bacillus subtilis spores using spore coat protein CotG as an anchor protein. Effects of spore-displayed p40 (CotG-p40) on gene expression of intestinal epithelial cell line HT-29 were evaluated by transcriptome analysis using RNA-sequencing. As a result of differentially expressed gene (DEG) analysis, 81 genes were up-regulated and 82 genes were down-regulated in CotG-p40 stimulated cells than in unstimulated cells. Gene ontology enrichment analysis showed that CotG-p40 affected biological processes such as developmental process, metabolic process, cell surface receptor linked signaling pathway, and retinoic acid metabolic process. Gene-gene network analysis suggested that 10 DEGs (EREG, FOXF1, GLI2, PTGS2, SPP1, MMP19, TNFRSF1B, PTGER4, CLDN18, and ALDH1A3) activated by CotG-p40 were associated with probiotic action. CONCLUSIONS This study demonstrates the regulatory effects of CotG-p40 on proliferation and homeostasis of HT-29 cells. This study provided comprehensive insights into the transcriptional response of human intestinal epithelial cells stimulated by CotG-p40.
Collapse
|
7
|
Novel Horizons in Postbiotics: Lactobacillaceae Extracellular Vesicles and Their Applications in Health and Disease. Nutrients 2022; 14:nu14245296. [PMID: 36558455 PMCID: PMC9782203 DOI: 10.3390/nu14245296] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Lactobacillus probiotics contained in dietary supplements or functional foods are well-known for their beneficial properties exerted on host health and diverse pathological situations. Their capacity to improve inflammatory bowel disease (IBD) and regulate the immune system is especially remarkable. Although bacteria-host interactions have been thought to occur directly, the key role that extracellular vesicles (EVs) derived from probiotics play on this point is being unveiled. EVs are lipid bilayer-enclosed particles that carry a wide range of cargo compounds and act in different signalling pathways. Notably, these EVs have been recently proposed as a safe alternative to the utilisation of live bacteria since they can avoid the possible risks that probiotics may entail in vulnerable cases such as immunocompromised patients. Therefore, this review aims to give an updated overview of the existing knowledge about EVs from different Lactobacillus strains, their mechanisms and effects in host health and different pathological conditions. All of the information collected suggests that EVs could be considered as potential tools for the development of future novel therapeutic approaches.
Collapse
|
8
|
Kaur H, Ali SA, Yan F. Interactions between the gut microbiota-derived functional factors and intestinal epithelial cells - implication in the microbiota-host mutualism. Front Immunol 2022; 13:1006081. [PMID: 36159834 PMCID: PMC9492984 DOI: 10.3389/fimmu.2022.1006081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/23/2022] [Indexed: 12/13/2022] Open
Abstract
Mutual interactions between the gut microbiota and the host play essential roles in maintaining human health and providing a nutrient-rich environment for the gut microbial community. Intestinal epithelial cells (IECs) provide the frontline responses to the gut microbiota for maintaining intestinal homeostasis. Emerging evidence points to commensal bacterium-derived components as functional factors for the action of commensal bacteria, including protecting intestinal integrity and mitigating susceptibility of intestinal inflammation. Furthermore, IECs have been found to communicate with the gut commensal bacteria to shape the composition and function of the microbial community. This review will discuss the current understanding of the beneficial effects of functional factors secreted by commensal bacteria on IECs, with focus on soluble proteins, metabolites, and surface layer components, and highlight the impact of IECs on the commensal microbial profile. This knowledge provides a proof-of-concept model for understanding of mechanisms underlying the microbiota-host mutualism.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Syed Azmal Ali
- German Cancer Research Center, Division of Proteomics of Stem Cell and Cancer, Heidelberg, Germany
| | - Fang Yan
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States,*Correspondence: Fang Yan,
| |
Collapse
|
9
|
The Phagocytosis of Lacticaseibacillus casei and Its Immunomodulatory Properties on Human Monocyte-Derived Dendritic Cells Depend on the Expression of Lc-p75, a Bacterial Peptidoglycan Hydrolase. Int J Mol Sci 2022; 23:ijms23147620. [PMID: 35886967 PMCID: PMC9319067 DOI: 10.3390/ijms23147620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/02/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
The human gut symbiont Lacticaseibacillus (L.) casei (previously Lactobacillus casei) is under intense research due to its wide range of immunomodulatory effects on the human host. Dendritic cells (DCs) are crucial players in the direct and indirect communication with lactobacilli in the gastrointestinal tract. Here, we demonstrate that human monocyte-derived DCs (moDCs) are able to engulf L. casei BL23, in which the intact bacterial cell wall and morphology have a key role. The absence of the bacterial cell-wall-degrading enzyme, Lc-p75, in L. casei cells causes remarkable morphological changes, which have important consequences in the phagocytosis of L. casei by moDCs. Our results showed that the Lc-p75 mutation induced defective internalization and impaired proinflammatory and T-cell-polarizing cytokine secretion by bacteria-exposed moDCs. The T helper (Th) 1 and Th17 cell activating capacity of moDCs induced by the mutant L. casei was consequently reduced. Moreover, inhibition of the phagocytosis of wild-type bacteria showed similar results. Taken together, these data suggested that formation of short bacterial chains helps to exert the potent immunomodulatory properties of L. casei BL23.
Collapse
|
10
|
Effects of Spore-Displayed p75 Protein from Lacticaseibacillus rhamnosus GG on the Transcriptional Response of HT-29 Cells. Microorganisms 2022; 10:microorganisms10071276. [PMID: 35888995 PMCID: PMC9323162 DOI: 10.3390/microorganisms10071276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
A Lacticaseibacillus rhamnosus GG-derived protein, p75, is one of the key molecules exhibiting probiotic activity. However, the molecular mechanism and transcriptional response of p75 in human intestinal epithelial cells are not completely understood. To gain a deeper understanding of its potential probiotic action, this study investigated genome-wide responses of HT-29 cells to stimulation by spore-displayed p75 (CotG-p75) through a transcriptome analysis based on RNA sequencing. Analysis of RNA-seq data showed significant changes of gene expression in HT-29 cells stimulated by CotG-p75 compared to the control. A total of 189 up-regulated and 314 down-regulated genes was found as differentially expressed genes. Gene ontology enrichment analysis revealed that a large number of activated genes was involved in biological processes, such as epithelial cell differentiation, development, and regulation of cell proliferation. A gene–gene interaction network analysis showed that several DEGs, including AREG, EREG, HBEGF, EPGN, FASLG, GLI2, CDKN1A, FOSL1, MYC, SERPINE1, TNFSF10, BCL6, FLG, IVL, SPRR1A, SPRR1B, SPRR3, and MUC5AC, might play a critical role in these biological processes. RNA-seq results for selected genes were verified by reverse transcription-quantitative polymerase chain reaction. Overall, these results provide extensive knowledge about the transcriptional responses of HT-29 cells to stimulation by CotG-p75. This study showed that CotG-p75 can contribute to cell survival and epithelial development in human intestinal epithelial cells.
Collapse
|
11
|
Kiousi DE, Efstathiou C, Tegopoulos K, Mantzourani I, Alexopoulos A, Plessas S, Kolovos P, Koffa M, Galanis A. Genomic Insight Into Lacticaseibacillus paracasei SP5, Reveals Genes and Gene Clusters of Probiotic Interest and Biotechnological Potential. Front Microbiol 2022; 13:922689. [PMID: 35783439 PMCID: PMC9244547 DOI: 10.3389/fmicb.2022.922689] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/16/2022] [Indexed: 12/22/2022] Open
Abstract
The Lacticaseibacillus paracasei species is comprised by nomadic bacteria inhabiting a wide variety of ecological niches, from fermented foodstuffs to host-associated microenvironments. Lc. paracasei SP5 is a novel strain, originally isolated from kefir grains that presents desirable probiotic and biotechnological attributes. In this study, we applied genomic tools to further characterize the probiotic and biotechnological potential of the strain. Firstly, whole genome sequencing and assembly, were performed to construct the chromosome map of the strain and determine its genomic stability. Lc. paracasei SP5 carriers several insertion sequences, however, no plasmids or mobile elements were detected. Furthermore, phylogenomic and comparative genomic analyses were utilized to study the nomadic attributes of the strain, and more specifically, its metabolic capacity and ability to withstand environmental stresses imposed during food processing and passage through the gastrointestinal (GI) tract. More specifically, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Carbohydrate-active enzyme (CAZymes) analyses provided evidence for the ability of the stain to utilize an array of carbohydrates as growth substrates. Consequently, genes for heat, cold, osmotic shock, acidic pH, and bile salt tolerance were annotated. Importantly bioinformatic analysis showed that the novel strain does not harbor acquired antimicrobial resistance genes nor virulence factors, in agreement with previous experimental data. Putative bacteriocin biosynthesis clusters were identified using BAGEL4, suggesting its potential antimicrobial activity. Concerning microbe-host interactions, adhesins, moonlighting proteins, exopolysaccharide (EPS) biosynthesis genes and pilins mediating the adhesive phenotype were, also, pinpointed in the genome of Lc. paracasei SP5. Validation of this phenotype was performed by employing a microbiological method and confocal microscopy. Conclusively, Lc. paracasei SP5 harbors genes necessary for the manifestation of the probiotic character and application in the food industry. Upcoming studies will focus on the mechanisms of action of the novel strain at multiple levels.
Collapse
Affiliation(s)
- Despoina Eugenia Kiousi
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
| | - Christos Efstathiou
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
| | - Konstantinos Tegopoulos
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioanna Mantzourani
- Department of Agricultural Development, Democritus University of Thrace, Orestiada, Greece
| | - Athanasios Alexopoulos
- Department of Agricultural Development, Democritus University of Thrace, Orestiada, Greece
| | - Stavros Plessas
- Department of Agricultural Development, Democritus University of Thrace, Orestiada, Greece
- *Correspondence: Stavros Plessas,
| | - Petros Kolovos
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
| | - Maria Koffa
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
| | - Alex Galanis
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
- Alex Galanis,
| |
Collapse
|
12
|
Variability of Genetic Characters Associated with Probiotic Functions in Lacticaseibacillus Species. Microorganisms 2022; 10:microorganisms10051023. [PMID: 35630465 PMCID: PMC9145642 DOI: 10.3390/microorganisms10051023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/17/2022] Open
Abstract
This study aims to explore the intra-species distribution of genetic characteristics that favor the persistence in the gastrointestinal tract (GIT) and host interaction of bacteria belonging to species of the Lacticaseibacillus genus. These bacterial species comprise commercial probiotics with the widest use among consumers and strains naturally occurring in GIT and in fermented food. Since little is known about the distribution of genetic traits for adhesion capacity, polysaccharide production, biofilm formation, and utilization of substrates critically important for survival in GIT, which influence probiotic characteristics, a list of genetic determinants possibly involved in such functions was created by a search for specific genes involved in the above aspects in the genome of the extensively characterized probiotic L. rhamnosus GG. Eighty-two gene loci were retrieved and their presence and variability in other Lacticaseibacillus spp. genomes were assessed by alignment with the publicly available fully annotated genome sequences of L. casei, L. paracasei, L. rhamnosus, and L. zeae. Forty-nine of these genes were found to be absent in some strains or species. The remaining genes were conserved and covered almost all the functions considered, indicating that all strains of the genus may exert some probiotic effects. Among the variable loci, a taurine utilization operon and a α-L-fucosidase were examined for the presence/absence in 26 strains isolated from infant feces by PCR-based tests. Results were variable among the isolates, though their common origin indicated the capacity to survive in the intestinal niche. This study indicated that the capacity to exert probiotic actions of Lacticaseibacillus spp. depends on a conserved set of genes but variable genetic factors, whose role is only in part elucidated, are more numerous and can explain the enhanced probiotic characteristics for some strains. The selection of the most promising probiotic candidates to be used in food is feasible by analyzing the presence/absence of a set of variable traits.
Collapse
|
13
|
Requena T, Pérez Martínez G. Probiotics, Prebiotics, Synbiotics, Postbiotics and Other Biotics. What's Next? COMPREHENSIVE GUT MICROBIOTA 2022:197-210. [DOI: 10.1016/b978-0-12-819265-8.00094-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Dou X, Qiao L, Chang J, Yan S, Song X, Chen Y, Xu Q, Xu C. Lactobacillus casei ATCC 393 and it's metabolites alleviate dextran sulphate sodium-induced ulcerative colitis in mice through the NLRP3-(Caspase-1)/IL-1β pathway. Food Funct 2021; 12:12022-12035. [PMID: 34755743 DOI: 10.1039/d1fo02405a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease (IBD) represents a broad group of intestinal disorders, including ulcerative colitis (UC) and Crohn's disease (CD). Probiotics are increasingly being recognized as a means of treatment for people suffering from IBD. Our previous studies demonstrated that Lactobacillus casei ATCC 393 (L. casei ATCC 393) effectively alleviated enterotoxigenic Escherichia coli K88-induced intestinal barrier dysfunction. This study was conducted to investigate the protective effects of L. casei ATCC 393 and its metabolites on dextran sulfate sodium (DSS)-induced UC in C57BL/6 mice and the potential mechanism of these effects. The results showed that oral administration of L. casei ATCC 393 and its metabolites both effectively reversed the DSS-induced weight loss, and the reduction in the disease activity index (DAI), colon length, and villus height of colon tissue in mice. Compared to the DSS-induced model group, L. casei ATCC 393 and its metabolites significantly inhibited the infiltration of immune cells into the intestinal mucosa, decreased the production of pro-inflammatory factors, and increased the expression of anti-inflammatory factors in the serum and colon tissue, increased the expression levels of occludin, ZO-1, and claudin-1, and reduced the expression of nucleotide binding oligomeric domain-like receptor protein 3 (NLRP3), cysteine proteinase-1 (Caspase-1), IL-1β, and IL-18. In addition, L. casei ATCC 393 and its metabolites effectively improved DSS-induced gut microbiota dysbiosis. These results suggested that L. casei ATCC 393 and its metabolites alleviated the DSS-induced ulcerative inflammatory response in C57BL/6 mice through the NLRP3-(Caspase-1)/IL-1β signaling pathway.
Collapse
Affiliation(s)
- Xina Dou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Lei Qiao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Jiajing Chang
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Shuqi Yan
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Xiaofan Song
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Yue Chen
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Qinhong Xu
- Department of Geriatric Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
15
|
Ye S, Si C, Deng J, Chen X, Kong L, Zhou X, Wang W. Understanding the Effects of Metabolites on the Gut Microbiome and Severe Acute Pancreatitis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1516855. [PMID: 34712726 PMCID: PMC8548099 DOI: 10.1155/2021/1516855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 10/09/2021] [Indexed: 12/11/2022]
Abstract
Acute pancreatitis (AP) is an inflammatory disease of the pancreas. The severity is classified as mild (MAP), moderately severe (MSAP), or severe (SAP). In patients with SAP, organ dysfunction can occur in the early stage of the disease course, accompanied by secondary infection, with a mortality rate of 36%-50%. In the late stage SAP, infection-related complications caused by pancreatic necrotic tissue and peripancreatic effusion are the main causes of death in patients. Dysbacteriosis of intestinal microflora, barrier dysfunction of intestinal mucosa, and translocation of enteric bacteria are considered to be the main causes of infection of pancreatic necrotic tissue and peripancreatic effusion. During the past few years, increasing attention has been paid to the metabolic activities of intestinal microflora in SAP, which plays an important role in the metabolic activities of the human body. This review is aimed at bringing together the most recent findings and advances regarding the gut microbial community and associated gut microbial community metabolites and illustrating the role of these metabolites in disease progression in severe acute pancreatitis. We hope that this review will provide new ideas and schemes for the treatment of SAP in the clinical settings.
Collapse
Affiliation(s)
- Shijie Ye
- Wenzhou Medical University, Wenzhou, China
| | - Chenli Si
- Wenzhou Medical University, Wenzhou, China
| | - Jie Deng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaohu Chen
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | | | - Xiang Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiming Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
16
|
Lei C, Teng Y, He L, Sayed M, Mu J, Xu F, Zhang X, Kumar A, Sundaram K, Sriwastva MK, Zhang L, Chen SY, Feng W, Zhang S, Yan J, Park JW, Merchant ML, Zhang X, Zhang HG. Lemon exosome-like nanoparticles enhance stress survival of gut bacteria by RNase P-mediated specific tRNA decay. iScience 2021; 24:102511. [PMID: 34142028 PMCID: PMC8188359 DOI: 10.1016/j.isci.2021.102511] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/29/2021] [Accepted: 04/30/2021] [Indexed: 11/30/2022] Open
Abstract
Diet and bile play critical roles in shaping gut microbiota, but the molecular mechanism underlying interplay with intestinal microbiota is unclear. Here, we showed that lemon-derived exosome-like nanoparticles (LELNs) enhance lactobacilli toleration to bile. To decipher the mechanism, we used Lactobacillus rhamnosus GG (LGG) as proof of concept to show that LELNs enhance LGG bile resistance via limiting production of Msp1 and Msp3, resulting in decrease of bile accessibility to cell membrane. Furthermore, we found that decline of Msps protein levels was regulated through specific tRNAser UCC and tRNAser UCG decay. We identified RNase P, an essential housekeeping endonuclease, being responsible for LELNs-induced tRNAser UCC and tRNAser UCG decay. We further identified galacturonic acid-enriched pectin-type polysaccharide as the active factor in LELNs to increase bile resistance and downregulate tRNAser UCC and tRNAser UCG level in the LGG. Our study demonstrates a tRNA-based gene expression regulation mechanism among lactobacilli to increase bile resistance.
Collapse
Affiliation(s)
- Chao Lei
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Yun Teng
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Liqing He
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Mohammed Sayed
- Department of Computer Engineering and Computer Science, University of Louisville, Louisville, KY 40202, USA
| | - Jingyao Mu
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Fangyi Xu
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Xiangcheng Zhang
- Department of ICU, the Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu 223300, China
| | - Anil Kumar
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Kumaran Sundaram
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Mukesh K. Sriwastva
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Lifeng Zhang
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Shao-yu Chen
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Wenke Feng
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Shuangqin Zhang
- Peeples Cancer Institute, 215 Memorial Drive, Dalton, GA 30720, USA
| | - Jun Yan
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Juw Won Park
- Department of Computer Engineering and Computer Science, University of Louisville, Louisville, KY 40202, USA
- KBRIN Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA
| | - Michael L. Merchant
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Xiang Zhang
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Huang-Ge Zhang
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA
- James Graham Brown Cancer Center, Department of Microbiology & Immunology, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| |
Collapse
|
17
|
Kim J, Balasubramanian I, Bandyopadhyay S, Nadler I, Singh R, Harlan D, Bumber A, He Y, Kerkhof LJ, Gao N, Su X, Ferraris RP. Lactobacillus rhamnosus GG modifies the metabolome of pathobionts in gnotobiotic mice. BMC Microbiol 2021; 21:165. [PMID: 34082713 PMCID: PMC8176599 DOI: 10.1186/s12866-021-02178-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 02/25/2021] [Indexed: 12/31/2022] Open
Abstract
Background Lactobacillus rhamnosus GG (LGG) is the most widely used probiotic, but the mechanisms underlying its beneficial effects remain unresolved. Previous studies typically inoculated LGG in hosts with established gut microbiota, limiting the understanding of specific impacts of LGG on host due to numerous interactions among LGG, commensal microbes, and the host. There has been a scarcity of studies that used gnotobiotic animals to elucidate LGG-host interaction, in particular for gaining specific insights about how it modifies the metabolome. To evaluate whether LGG affects the metabolite output of pathobionts, we inoculated with LGG gnotobiotic mice containing Propionibacterium acnes, Turicibacter sanguinis, and Staphylococcus aureus (PTS). Results 16S rRNA sequencing of fecal samples by Ion Torrent and MinION platforms showed colonization of germ-free mice by PTS or by PTS plus LGG (LTS). Although the body weights and feeding rates of mice remained similar between PTS and LTS groups, co-associating LGG with PTS led to a pronounced reduction in abundance of P. acnes in the gut. Addition of LGG or its secretome inhibited P. acnes growth in culture. After optimizing procedures for fecal metabolite extraction and metabolomic liquid chromatography-mass spectrometry analysis, unsupervised and supervised multivariate analyses revealed a distinct separation among fecal metabolites of PTS, LTS, and germ-free groups. Variables-important-in-projection scores showed that LGG colonization robustly diminished guanine, ornitihine, and sorbitol while significantly elevating acetylated amino acids, ribitol, indolelactic acid, and histamine. In addition, carnitine, betaine, and glutamate increased while thymidine, quinic acid and biotin were reduced in both PTS and LTS groups. Furthermore, LGG association reduced intestinal mucosal expression levels of inflammatory cytokines, such as IL-1α, IL-1β and TNF-α. Conclusions LGG co-association had a negative impact on colonization of P. acnes, and markedly altered the metabolic output and inflammatory response elicited by pathobionts. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02178-2.
Collapse
Affiliation(s)
- Jinhee Kim
- Department of Pharmacology, Physiology and Neurosciences, Medical Science Building, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | | | - Sheila Bandyopadhyay
- Department of Biological Sciences, Life Science Center, Rutgers University, Newark, NJ, 07102, USA
| | - Ian Nadler
- Department of Pharmacology, Physiology and Neurosciences, Medical Science Building, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Rajbir Singh
- Department of Biological Sciences, Life Science Center, Rutgers University, Newark, NJ, 07102, USA
| | - Danielle Harlan
- Department of Pharmacology, Physiology and Neurosciences, Medical Science Building, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Amanda Bumber
- Comparative Medicine Resources, Rutgers University, Newark, NJ, 07103, USA
| | - Yuling He
- Department of Medicine, Clinical Academic Building, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA.,Present address: Geriatric Endocrinology Division, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Lee J Kerkhof
- Department of Marine and Coastal Sciences, Rutgers University, 71 Dudley Rd, New Brunswick, NJ, 08901, USA
| | - Nan Gao
- Department of Biological Sciences, Life Science Center, Rutgers University, Newark, NJ, 07102, USA
| | - Xiaoyang Su
- Department of Medicine, Clinical Academic Building, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Ronaldo P Ferraris
- Department of Pharmacology, Physiology and Neurosciences, Medical Science Building, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA.
| |
Collapse
|
18
|
Coll-Marqués JM, Bäuerl C, Zúñiga M, Pérez-Martínez G. Differences in the expression of cell envelope proteinases (CEP) in two Lactobacillus paracasei probiotic strains. FEMS Microbiol Lett 2021; 367:5861318. [PMID: 32573688 DOI: 10.1093/femsle/fnaa102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022] Open
Abstract
Proteinase PrtP (EC:3.4.21.96) is a cell envelope proteinase (CEP) highly expressed in the probiotic strain Lactobacillus paracasei BL312(VSL#3) that accounts for its anti-inflammatory properties. The main aim of this work is to understand differences in CEP expression between this strain and L. paracasei BL23. Hence, differences in the regulation by amino acid sources of four proteinase related genes (prtP, prsA, prtR1 and prtR2) were determined by RT-qPCR in BL312(VSL#3) and BL23 using as a reference BL368, a BL23 derepressed mutant lacking the response regulator (RR) PrcR. BL312(VSL#3) showed greater expression of prtP (2- to 3-fold) than BL23, and prtP was highly repressed by peptone in both strains. Two other putative CEP genes, prtR1 and prtR2, showed a low expression profile. Interestingly, when the prsA-prtP promoter region from both strains, and deleted mutants, were cloned in vector pT1GR, expression of the gfp and mrfp fluorescent reporters was always repressed in BL23 (high or low peptone) and derepressed in BL368, revealing an interesting mechanism of regulation affecting specifically to this promoter. In conclusion, BL312(VSL#3) has higher expression of prtP and other CEP related genes than BL23, that could respond to a natural deregulation in this strain, possibly independent from the RR PrcR.
Collapse
Affiliation(s)
- José María Coll-Marqués
- Department of Food Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos, Spanish National Research Council (CSIC), Agustín Escardino Benlloch, 7, 46980 Paterna, Valencia, Spain
| | - Christine Bäuerl
- Department of Food Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos, Spanish National Research Council (CSIC), Agustín Escardino Benlloch, 7, 46980 Paterna, Valencia, Spain
| | - Manuel Zúñiga
- Department of Food Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos, Spanish National Research Council (CSIC), Agustín Escardino Benlloch, 7, 46980 Paterna, Valencia, Spain
| | - Gaspar Pérez-Martínez
- Department of Food Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos, Spanish National Research Council (CSIC), Agustín Escardino Benlloch, 7, 46980 Paterna, Valencia, Spain
| |
Collapse
|
19
|
Petrova MI, Reid G, Ter Haar JA. Lacticaseibacillus rhamnosus GR-1, a.k.a. Lactobacillus rhamnosus GR-1: Past and Future Perspectives. Trends Microbiol 2021; 29:747-761. [PMID: 33865678 DOI: 10.1016/j.tim.2021.03.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/14/2022]
Abstract
Lacticaseibacillus rhamnosus GR-1 (LGR-1) (previously classified as Lactobacillus rhamnosus GR-1) is the most researched probiotic strain for women's health. Its various urogenital health effects, including a reduction in the recurrence of bacterial vaginosis and urinary-tract infection, are well documented. The strain has also been safely used by HIV-positive subjects, a portion of whom have reported reduced diarrhea and increased CD4 counts. Unlike most probiotic strains used for urogenital health, LGR-1 has been extensively studied for its properties, including its genomic and metabolic traits and its surface properties. This review aims to highlight the totality of research performed with LGR-1, to act as a rigorous scientific benchmark for probiotic microbes, especially for application to women's health.
Collapse
Affiliation(s)
- Mariya I Petrova
- Microbiome Insights and Probiotics Consultancy, Karlovo, Bulgaria.
| | - Gregor Reid
- Canadian Research and Development Centre for Human Microbiome and Probiotics, Lawson Health Research Institute, London, Canada; Department of Microbiology and Immunology, The University of Western Ontario, London, Canada; Department of Surgery, The University of Western Ontario, London, Canada
| | | |
Collapse
|
20
|
Effect of culturing lactic acid bacteria with varying skim milk concentration on bacteria survival during heat treatment. J FOOD ENG 2021. [DOI: 10.1016/j.jfoodeng.2020.110396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
21
|
Dadfarma N, Nowroozi J, Kazemi B, Bandehpour M. Identification of the effects of acid-resistant Lactobacillus casei metallopeptidase gene under colon-specific promoter on the colorectal and breast cancer cell lines. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:506-513. [PMID: 34094033 PMCID: PMC8143706 DOI: 10.22038/ijbms.2021.53015.11950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/02/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Anti-tumor effects of Lactobacilli as normal flora have been described. In a previous study, we identified a protein isolated from the bacterium Lactobacillus casei ATCC 39392 in acidic pH conditions named metallopeptidase. Therefore, we decided to evaluate the effect of the recombinant plasmid coding metallopeptidase protein on the inhibition, proliferation, or apoptosis of the colorectal and breast cancer cell lines. MATERIALS AND METHODS Identified metallopeptidase gene of L. casei under the specific colon cancer promoter was transferred to the Human SW480 and MDA-MB231 cells. Cell viability was evaluated in these two cancer cell lines via MTT assay, apoptotic changes, and expression level of p53 and MAP2K1 genes in comparison with healthy blood cells as a control group. RESULTS Viability of SW480 and MDA-MB231 cells was identified at 25% and 7%, respectively. An increase in apoptotic cell death in the SW480 cell line was observed as revealed by Tunnel staining. The expression assay of TP53 and MAP2K1 genes showed that MPL protein altered gene expression in a cell type-specific manner. Tunnel analyses showed that the pronounced cytotoxic effect of pEGFP-C2/MPL plasmid on SW480 cells was mediated through apoptosis. CONCLUSION These results suggest that endogenous recombinant MPL under colon specific promoter inhibits the proliferation of SW480 colorectal cancer cells by increase in MAP2K1 and P53 activation. L. casei metallopeptidase under the same circumstances could not affect the growth rate and viability of MDA-MB231 breast cancer cells in vitro.
Collapse
Affiliation(s)
- Narges Dadfarma
- Department of Microbiology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Jamileh Nowroozi
- Department of Microbiology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Bahram Kazemi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojgan Bandehpour
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Daliri EBM, Ofosu FK, Xiuqin C, Chelliah R, Oh DH. Probiotic Effector Compounds: Current Knowledge and Future Perspectives. Front Microbiol 2021; 12:655705. [PMID: 33746935 PMCID: PMC7965967 DOI: 10.3389/fmicb.2021.655705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/12/2021] [Indexed: 12/22/2022] Open
Abstract
Understanding the mechanism behind probiotic action will enable a rational selection of probiotics, increase the chances of success in clinical studies and make it easy to substantiate health claims. However, most probiotic studies over the years have rather focused on the effects of probiotics in health and disease, whereas little is known about the specific molecules that trigger effects in hosts. This makes it difficult to describe the detailed mechanism by which a given probiotic functions. Probiotics communicate with their hosts through molecular signaling. Meanwhile, since the molecules produced by probiotics under in vitro conditions may differ from those produced in vivo, in vitro mechanistic studies would have to be conducted under conditions that mimic gastrointestinal conditions as much as possible. The ideal situation would, however, be to carry out well-designed clinical trials in humans (or the target animal) using adequate quantities of the suspected probiotic molecule(s) or adequate quantities of isogenic knock-out or knock-in probiotic mutants. In this review, we discuss our current knowledge about probiotic bacteria and yeast molecules that are involved in molecular signaling with the host. We also discuss the challenges and future perspectives in the search for probiotic effector molecules.
Collapse
Affiliation(s)
- Eric Banan-Mwine Daliri
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Fred Kwame Ofosu
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Chen Xiuqin
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Ramachandran Chelliah
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
23
|
Moreira CF, Cassini-Vieira P, Canesso MCC, Felipetto M, Ranfley H, Teixeira MM, Nicoli JR, Martins FS, Barcelos LS. Lactobacillus rhamnosus CGMCC 1.3724 (LPR) Improves Skin Wound Healing and Reduces Scar Formation in Mice. Probiotics Antimicrob Proteins 2021; 13:709-719. [PMID: 33433898 DOI: 10.1007/s12602-020-09713-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2020] [Indexed: 01/16/2023]
Abstract
Skin wounds are an important clinical problem which affects millions of people worldwide. The search for new therapeutic approaches to improve wound healing is needed. The present study aimed to evaluate the effects of the oral treatment with the skin-related probiotics Lactobacillus johnsonii LA1 (LJ), L. paracasei ST11 (LP), and L. rhamnosus LPR (LR) in a model of excisional skin wounds in Swiss mice. The animals received daily oral gavage of PBS or 1 × 107 colony-forming units of LJ, LP, or LR, singly, beginning just after the creation of wounds until euthanasia. Blood flow was evaluated by laser Doppler perfusion imaging. Myeloperoxidase and N-acetyl-β-D-glucosaminidase activities were used to assess the accumulation of neutrophils and macrophages, respectively. The wound tissue was also collected for histological analyses (H&E, Toluidine blue, and Picrosirius red staining). The macroscopic wound closure rate was faster only in mice treated with LR, but not with LJ and LP, when compared to mice treated with PBS. Histological evaluations showed that treatment with LR stimulated wound epithelization when compared to PBS. Further analyses showed that wounds from LR-treated mice presented a significant decrease in macrophage (p < 0.001) and mast cell (p < 0.001) infiltration, along with improved angiogenesis (p < 0.001) and blood flow (p < 0.01). Of note, collagen deposition and scarring were reduced in LR-treated mice when compared to PBS-treated mice. In conclusion, our results show that the oral treatment with Lactobacillus rhamnosus accelerates skin wound closure and reduces scar, besides to reducing inflammation and fibrogenesis and improving angiogenesis in the wounded skin.
Collapse
Affiliation(s)
- Camila Francisco Moreira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Puebla Cassini-Vieira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Maria Cecília Campos Canesso
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Mariane Felipetto
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Hedden Ranfley
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Minas Gerais, Belo Horizonte, Brazil
| | - Jacques Robert Nicoli
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Minas Gerais, Belo Horizonte, Brazil
| | - Flaviano Santos Martins
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Minas Gerais, Belo Horizonte, Brazil
| | - Lucíola Silva Barcelos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av. Antônio Carlos, 6627 - Pampulha, Minas Gerais, 31270-901, Belo Horizonte, Brazil.
| |
Collapse
|
24
|
Lactobacillus casei extracellular vesicles stimulate EGFR pathway likely due to the presence of proteins P40 and P75 bound to their surface. Sci Rep 2020; 10:19237. [PMID: 33159116 PMCID: PMC7648624 DOI: 10.1038/s41598-020-75930-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 10/16/2020] [Indexed: 12/20/2022] Open
Abstract
In the complex interplay of beneficial bacteria with the host, there are few examples of bacterial metabolites and effector molecules that have been consistently identified. Protective effects on the intestinal epithelium have been ascribed to P40 and P75, two well characterized cell wall muramidases, present in the culture supernatant of strains belonging to the taxon Lactobacillus casei/paracasei/rhamnosus. This work reports that Lactobacillus casei BL23 extracellular vesicles (BL23 EVs) have a small size (17–20 nm or 24–32 nm, depending on the method used) and contain lipoteichoic acid (LTA). Interestingly, all detected P40 and most of P75 were associated to EVs and possibly located at their external surface, as shown by proteinase K digestion. Biosensor assays showed that both proteins bind LTA and vesicles, suggesting that they could bind to ligands like LTA present on BL23 EVs. Native BL23 EVs have a moderate proinflammatory effect and they were able to induce phosphorylation of the epidermal growth factor receptor (EGFR), showing an effect similar to purified P40 and P75 and leading to the conclusion that the activity described in the supernatant (postbiotic) of these bacteria would be mainly due to P40 and P75 bound to EVs.
Collapse
|
25
|
Teame T, Wang A, Xie M, Zhang Z, Yang Y, Ding Q, Gao C, Olsen RE, Ran C, Zhou Z. Paraprobiotics and Postbiotics of Probiotic Lactobacilli, Their Positive Effects on the Host and Action Mechanisms: A Review. Front Nutr 2020; 7:570344. [PMID: 33195367 PMCID: PMC7642493 DOI: 10.3389/fnut.2020.570344] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
Lactobacilli comprise an important group of probiotics for both human and animals. The emerging concern regarding safety problems associated with live microbial cells is enhancing the interest in using cell components and metabolites derived from probiotic strains. Here, we define cell structural components and metabolites of probiotic bacteria as paraprobiotics and postbiotics, respectively. Paraprobiotics and postbiotics produced from Lactobacilli consist of a wide range of molecules including peptidoglycans, surface proteins, cell wall polysaccharides, secreted proteins, bacteriocins, and organic acids, which mediate positive effect on the host, such as immunomodulatory, anti-tumor, antimicrobial, and barrier-preservation effects. In this review, we systematically summarize the paraprobiotics and postbiotics derived from Lactobacilli and their beneficial functions. We also discuss the mechanisms underlying their beneficial effects on the host, and their interaction with the host cells. This review may boost our understanding on the benefits and molecular mechanisms associated with paraprobiotics and probiotics from Lactobacilli, which may promote their applications in humans and animals.
Collapse
Affiliation(s)
- Tsegay Teame
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Tigray Agricultural Research Institute, Mekelle, Ethiopia
| | - Anran Wang
- AgricultureIsLife/EnvironmentIsLife and Precision Livestock and Nutrition Unit, AgroBioChem/TERRA, Gembloux Agro-Bio Tech, University of Liege, Passage des Deportes, Gembloux, Belgium
| | - Mingxu Xie
- Norway-China Fish Gastrointestinal Microbiota Joint Lab, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qianwen Ding
- Norway-China Fish Gastrointestinal Microbiota Joint Lab, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Chenchen Gao
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Rolf Erik Olsen
- Norway-China Fish Gastrointestinal Microbiota Joint Lab, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhigang Zhou
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
26
|
Paveljšek D, Ivičak-Kocjan K, Treven P, Benčina M, Jerala R, Rogelj I. Distinctive probiotic features share common TLR2-dependent signalling in intestinal epithelial cells. Cell Microbiol 2020; 23:e13264. [PMID: 32945079 PMCID: PMC7757178 DOI: 10.1111/cmi.13264] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 12/14/2022]
Abstract
The underlying mechanisms of probiotics and postbiotics are not well understood, but it is known that both affect the adaptive and innate immune responses. In addition, there is a growing concept that some probiotic strains have common core mechanisms that provide certain health benefits. Here, we aimed to elucidate the signalization of the probiotic bacterial strains Lactobacillus paragasseri K7, Limosilactobacillus fermentum L930BB, Bifidobacterium animalis subsp. animalis IM386 and Lactiplantibacillus plantarum WCFS1. We showed in in vitro experiments that the tested probiotics exhibit common TLR2- and TLR10-dependent downstream signalling cascades involving inhibition of NF-κB signal transduction. Under inflammatory conditions, the probiotics activated phosphatidylinositol 3-kinase (PI3K)/Akt anti-apoptotic pathways and protein kinase C (PKC)-dependent pathways, which led to regulation of the actin cytoskeleton and tight junctions. These pathways contribute to the regeneration of the intestinal epithelium and modulation of the mucosal immune system, which, together with the inhibition of canonical TLR signalling, promote general immune tolerance. With this study we identified shared probiotic mechanisms and were the first to pinpoint the role of anti-inflammatory probiotic signalling through TLR10.
Collapse
Affiliation(s)
- Diana Paveljšek
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| | - Karolina Ivičak-Kocjan
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Primož Treven
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Irena Rogelj
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| |
Collapse
|
27
|
Kang SJ, Jun JS, Moon JA, Hong KW. Surface display of p75, a Lactobacillus rhamnosus GG derived protein, on Bacillus subtilis spores and its antibacterial activity against Listeria monocytogenes. AMB Express 2020; 10:139. [PMID: 32770428 PMCID: PMC7415045 DOI: 10.1186/s13568-020-01073-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/25/2020] [Indexed: 01/05/2023] Open
Abstract
Lactobacillus rhamnosus p75 protein with peptidoglycan hydrolase (PGH) activity is one of the key molecules exhibiting anti-apoptotic and cell-protective activity for human intestinal epithelial cells. In this study, with the goal of developing new probiotics, the p75 protein was displayed on the surface of Bacillus subtilis spores using spore coat protein CotG as an anchoring motif. The PGH activity, stability, and the antibacterial activity of the spore-displayed p75 (CotG-p75) protein were also investigated. The PGH activity of the CotG-p75 against peptidoglycan extracted from B. subtilis was confirmed by the ninhydrin test. Under various harsh conditions, compared to the control groups, the PGH activities of CotG-p75 were very stable in the range of pH 3–7 and maintained at 70% at 50 °C. In addition, the antibacterial activity of CotG-p75 against Listeria monocytogenes was evaluated by a time-kill assay. After 6 h incubation in phosphate-buffered saline, CotG-p75 reduced the number of viable cells of L. monocytogenes by up to 2.0 log. Scanning electron microscopy analysis showed that the cell wall of L. monocytogenes was partially damaged by the treatment with CotG-p75. Our preliminary results show that CotG-p75 could be a good candidate for further research to develop new genetically engineered probiotics.
Collapse
|
28
|
Adu KT, Wilson R, Baker AL, Bowman J, Britz ML. Prolonged Heat Stress of Lactobacillus paracasei GCRL163 Improves Binding to Human Colorectal Adenocarcinoma HT-29 Cells and Modulates the Relative Abundance of Secreted and Cell Surface-Located Proteins. J Proteome Res 2020; 19:1824-1846. [PMID: 32108472 DOI: 10.1021/acs.jproteome.0c00107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lactobacillus casei group bacteria improve cheese ripening and may interact with host intestinal cells as probiotics, where surface proteins play a key role. Three complementary methods [trypsin shaving (TS), LiCl-sucrose (LS) extraction, and extracellular culture fluid precipitation] were used to analyze cell surface proteins of Lactobacillus paracasei GCRL163 by label-free quantitative proteomics after culture to the mid-exponential phase in bioreactors at pH 6.5 and temperatures of 30-45 °C. A total of 416 proteins, including 300 with transmembrane, cell wall anchoring, and secretory motifs and 116 cytoplasmic proteins, were quantified as surface proteins. Although LS caused significantly greater cell lysis as growth temperature increased, higher numbers of extracytoplasmic proteins were exclusively obtained by LS treatment. Together with the increased positive surface charge of cells cultured at supra-optimal temperatures, proteins including cell wall hydrolases Msp1/p75 and Msp2/p40, α-fucosidase AlfB, SecA, and a PspC-domain putative adhesin were upregulated in surface or secreted protein fractions, suggesting that cell adhesion may be altered. Prolonged heat stress (PHS) increased binding of L. paracasei GCRL163 to human colorectal adenocarcinoma HT-29 cells, relative to acid-stressed cells. This study demonstrates that PHS influences cell adhesion and relative abundance of proteins located on the surface, which may impact probiotic functionality, and the detected novel surface proteins likely linked to the cell cycle and envelope stress.
Collapse
Affiliation(s)
- Kayode T Adu
- Tasmanian Institute of Agriculture, University of Tasmania, Hobart, Tasmania 7001, Australia
| | - Richard Wilson
- Central Science Laboratory, University of Tasmania, Hobart, Tasmania 7001, Australia
| | - Anthony L Baker
- Tasmanian Institute of Agriculture, University of Tasmania, Hobart, Tasmania 7001, Australia
| | - John Bowman
- Tasmanian Institute of Agriculture, University of Tasmania, Hobart, Tasmania 7001, Australia
| | - Margaret L Britz
- Tasmanian Institute of Agriculture, University of Tasmania, Hobart, Tasmania 7001, Australia
| |
Collapse
|
29
|
Caruana JC, Walper SA. Bacterial Membrane Vesicles as Mediators of Microbe - Microbe and Microbe - Host Community Interactions. Front Microbiol 2020; 11:432. [PMID: 32265873 PMCID: PMC7105600 DOI: 10.3389/fmicb.2020.00432] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/02/2020] [Indexed: 01/18/2023] Open
Abstract
Bacterial membrane vesicles are proteoliposomal nanoparticles produced by both Gram-negative and Gram-positive bacteria. As they originate from the outer surface of the bacteria, their composition and content is generally similar to the parent bacterium’s membrane and cytoplasm. However, there is ample evidence that preferential packaging of proteins, metabolites, and toxins into vesicles does occur. Incorporation into vesicles imparts a number of benefits to the cargo, including protection from degradation by other bacteria, the host organism, or environmental factors, maintenance of a favorable microenvironment for enzymatic activity, and increased potential for long-distance movement. This enables vesicles to serve specialized functions tailored to changing or challenging environments, particularly in regard to microbial community interactions including quorum sensing, biofilm formation, antibiotic resistance, antimicrobial peptide expression and deployment, and nutrient acquisition. Additionally, based on their contents, vesicles play crucial roles in host-microbe interactions as carriers of virulence factors and other modulators of host cell function. Here, we discuss recent advances in our understanding of how vesicles function as signals both within microbial communities and between pathogenic or commensal microbes and their mammalian hosts. We also highlight a few areas that are currently ripe for additional research, including the mechanisms of selective cargo packaging into membrane vesicles and of cargo processing once it enters mammalian host cells, the function of vesicles in transfer of nucleic acids among bacteria, and the possibility of engineering commensal bacteria to deliver cargo of interest to mammalian hosts in a controlled manner.
Collapse
Affiliation(s)
- Julie C Caruana
- American Society for Engineering Education, Washington, DC, United States
| | - Scott A Walper
- US Naval Research Laboratory, Center for Biomolecular Science and Engineering, Washington, DC, United States
| |
Collapse
|
30
|
Qureshi N, Gu Q, Li P. Whole genome sequence analysis and in vitro probiotic characteristics of a Lactobacillus strain Lactobacillus paracasei ZFM54. J Appl Microbiol 2020; 129:422-433. [PMID: 32119175 DOI: 10.1111/jam.14627] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 12/11/2022]
Abstract
AIM The aim was to identify a Lactobacillus strain with potential probiotic characteristics by whole-genome sequence analysis and in vitro experimental studies. METHODS AND RESULTS The whole-genome sequencing was carried out using PacBio RSII sequencing method and Illumina's paired-end sequencing technology. Gene prediction and annotation were achieved using GlimmerVersion 3.02 and NCBI prokaryotic Genome Annotation Pipeline. Identification was done by biochemical tests and 16S rRNA sequence analysis. mega 6 software was used to build phylogenetic tree. Antagonism against pathogen was determined by agar well diffusion method. Resistance and stability to bile, simulated gastric acid, different salt concentration and thermostability were investigated. Hydrophobicity assay, aggregation assay and anti-oxidation assay were performed to check further probiotic traits. Finally antibiotic susceptibility and acute oral toxicity of the strain in mice were investigated to check its safety status. The strain showed >99% similarity to Lactobacillus paracasei which was further confirmed by biochemical tests. It significantly inhibited pathogens in agar well diffusion assay. It showed tolerance to simulated gastric acid (pH 3), 0·3% bile salt and 10% NaCl. Significant hydrophobic, aggregation and anti-oxidizing activities were observed. No resistance to antibiotics tested was observed and no adverse effects during acute oral toxicity in mice were detected. CONCLUSIONS Lactobacillus paracasei ZFM 54, a new and safe Lactobacillus strain was identified with numerous probiotic-associated genes and characteristics confirmed by experimental studies. SIGNIFICANCE AND IMPACT OF THE STUDY A new probiotic strain has been identified which is highly stable, safe and suitable to be used in health and food industries.
Collapse
Affiliation(s)
- N Qureshi
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Department of Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Q Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Department of Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - P Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Department of Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
31
|
Scornec H, Palud A, Pédron T, Wheeler R, Petitgonnet C, Boneca IG, Cavin JF, Sansonetti PJ, Licandro H. Study of the cwaRS-ldcA Operon Coding a Two-Component System and a Putative L,D-Carboxypeptidase in Lactobacillus paracasei. Front Microbiol 2020; 11:156. [PMID: 32194510 PMCID: PMC7062640 DOI: 10.3389/fmicb.2020.00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/22/2020] [Indexed: 11/22/2022] Open
Abstract
The cell surface is the primary recognition site between the bacterium and the host. An operon of three genes, LSEI_0219 (cwaR), LSEI_0220 (cwaS), and LSEI_0221 (ldcA), has been previously identified as required for the establishment of Lactobacillus paracasei in the gut. The genes cwaR and cwaS encode a predicted two-component system (TCS) and ldcA a predicted D-alanyl-D-alanine carboxypeptidase which is a peptidoglycan (PG) biosynthesis enzyme. We explored the functionality and the physiological role of these three genes, particularly their impact on the bacterial cell wall architecture and on the bacterial adaptation to environmental perturbations in the gut. The functionality of CwaS/R proteins as a TCS has been demonstrated by biochemical analysis. It is involved in the transcriptional regulation of several genes of the PG biosynthesis. Analysis of the muropeptides of PG in mutants allowed us to re-annotate LSEI_0221 as a putative L,D-carboxypeptidase (LdcA). The absence of this protein coincided with a decrease of two surface antigens: LSEI_0020, corresponding to p40 or msp2 whose implication in the host epithelial homeostasis has been recently studied, and LSEI_2029 which has never been functionally characterized. The inactivation of each of these three genes induces susceptibility to antimicrobial peptides (hBD1, hBD2, and CCL20), which could be the main cause of the gut establishment deficiency. Thus, this operon is necessary for the presence of two surface antigens and for a suitable cell wall architecture.
Collapse
Affiliation(s)
- Hélène Scornec
- PAM UMR, AgroSup Dijon, Université de Bourgogne Franche-Comté, Dijon, France
| | - Aurore Palud
- PAM UMR, AgroSup Dijon, Université de Bourgogne Franche-Comté, Dijon, France
| | - Thierry Pédron
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
- Unité INSERM, Institut Pasteur, Paris, France
| | - Richard Wheeler
- Unité de Biologie et Génétique de la Paroi Bactérienne, Institut Pasteur, Paris, France
- Avenir Group, INSERM, Paris, France
| | - Clément Petitgonnet
- PAM UMR, AgroSup Dijon, Université de Bourgogne Franche-Comté, Dijon, France
| | - Ivo Gomperts Boneca
- Unité de Biologie et Génétique de la Paroi Bactérienne, Institut Pasteur, Paris, France
- Avenir Group, INSERM, Paris, France
| | - Jean-François Cavin
- PAM UMR, AgroSup Dijon, Université de Bourgogne Franche-Comté, Dijon, France
| | - Philippe J. Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
- Unité INSERM, Institut Pasteur, Paris, France
- Chaire de Microbiologie et Maladies Infectieuses, Collège de France, Paris, France
| | - Hélène Licandro
- PAM UMR, AgroSup Dijon, Université de Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
32
|
West CL, Stanisz AM, Mao YK, Champagne-Jorgensen K, Bienenstock J, Kunze WA. Microvesicles from Lactobacillus reuteri (DSM-17938) completely reproduce modulation of gut motility by bacteria in mice. PLoS One 2020; 15:e0225481. [PMID: 31910436 PMCID: PMC6946587 DOI: 10.1371/journal.pone.0225481] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023] Open
Abstract
Microvesicles are small lipid, bilayer structures (20–400 nm in diameter) secreted by bacteria, fungi, archaea and parasites involved in inter-bacterial communication and host-pathogen interactions. Lactobacillus reuteri DSM-17938 (DSM) has been shown to have clinical efficacy in the treatment of infantile colic, diarrhea and constipation. We have shown previously that luminal administration to the mouse gut promotes reduction of jejunal motility but increases that in the colon. The production of microvesicles by DSM has been characterized, but the effect of these microvesicles on gastrointestinal motility has yet to be evaluated. To investigate a potential mechanism for the effects of DSM on the intestine, the bacteria and its products have here been tested for changes in velocity, frequency, and amplitude of contractions in intact segments of jejunum and colon excised from mice. The effect of the parent bacteria (DSM) was compared to the conditioned media in which it was grown, and the microvesicles it produced. The media used to culture the bacteria (broth) was tested as a negative control and the conditioned medium was tested after the microvesicles had been removed. DSM, conditioned medium, and the microvesicles all produced comparable effects in both the jejunum and the colon. The treatments individually decreased the velocity and frequency of propagating contractile cluster contractions in the jejunum and increased them in the colon to a similar degree. The broth control had little effect in both tissues. Removal of the microvesicles from the conditioned medium almost completely eradicated their effect on motility in both tissues. These results show that the microvesicles from DSM alone can completely reproduce the effects of the whole bacteria on gut motility. Furthermore, they suggest a new approach to the formulation of orally active bacterial therapeutics and offer a novel way to begin to identify the active bacterial components.
Collapse
Affiliation(s)
- Christine L. West
- McMaster Brain-Body Institute, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Department of Biology, McMaster University, Hamilton, ON, Canada
- * E-mail:
| | - Andrew M. Stanisz
- McMaster Brain-Body Institute, St. Joseph’s Healthcare, Hamilton, ON, Canada
| | - Yu-Kang Mao
- McMaster Brain-Body Institute, St. Joseph’s Healthcare, Hamilton, ON, Canada
| | | | - John Bienenstock
- McMaster Brain-Body Institute, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Wolfgang A. Kunze
- McMaster Brain-Body Institute, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Department of Biology, McMaster University, Hamilton, ON, Canada
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
33
|
Cell wall hydrolase as a surface-associated protein target for the specific detection of Lactobacillus rhamnosus using flow cytometry. INNOV FOOD SCI EMERG 2020. [DOI: 10.1016/j.ifset.2019.102240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
34
|
Diez-Gutiérrez L, San Vicente L, R. Barrón LJ, Villarán MDC, Chávarri M. Gamma-aminobutyric acid and probiotics: Multiple health benefits and their future in the global functional food and nutraceuticals market. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103669] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
35
|
Comparative genomics and functional analysis of a highly adhesive dairy Lactobacillus paracasei subsp. paracasei IBB3423 strain. Appl Microbiol Biotechnol 2019; 103:7617-7634. [PMID: 31359102 PMCID: PMC6717177 DOI: 10.1007/s00253-019-10010-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 01/04/2023]
Abstract
Various Lactobacillus paracasei strains are found in diverse environments, including dairy and plant materials and the intestinal tract of humans and animals, and are also used in the food industry or as probiotics. In this study, we have isolated a new strain L. paracasei subsp. paracasei IBB3423 from samples of raw cow milk collected in a citizen science project. IBB3423 showed some desired probiotic features such as high adhesion capacity and ability to metabolize inulin. Its complete genome sequence comprising the chromosome of 3,183,386 bp and two plasmids of 5986 bp and 51,211 bp was determined. In silico analysis revealed numerous genes encoding proteins involved in carbohydrate metabolism and of extracellular localization likely supporting interaction with host tissues. In vitro tests confirmed the high adhesion capacity of IBB3423 and showed that it even exceeds that of the highly adhesive L. rhamnosus GG. Curing of the larger plasmid indicated that the adhesive properties depend on the plasmid and thus could be determined by its pilus-encoding spaCBA genes.
Collapse
|
36
|
Bäuerl C, Abitayeva G, Sosa-Carrillo S, Mencher-Beltrán A, Navarro-Lleó N, Coll-Marqués JM, Zúñiga-Cabrera M, Shaikhin S, Pérez-Martinez G. P40 and P75 Are Singular Functional Muramidases Present in the Lactobacillus casei /paracasei/rhamnosus Taxon. Front Microbiol 2019; 10:1420. [PMID: 31297099 PMCID: PMC6607858 DOI: 10.3389/fmicb.2019.01420] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/05/2019] [Indexed: 01/29/2023] Open
Abstract
Lactobacillus casei and Lactobacillus rhamnosus proteins P40 and P75 belong to a large family of secreted cell wall proteins that contain a carboxy(C)-terminal CHAP or NlpC/P60 superfamily domains. In addition to their peptidoglycan hydrolases activity, proteins in this family are specific antigens of pathogens, frequently responsible of interactions with the host. L. rhamnosus GG and L. casei BL23 purified P40 and P75 proteins have antiapoptotic activity by inducing the EGF/Akt pathway. The aim of this work was to study the genetics, phylogeny and dissemination of this family of proteins in the genus Lactobacillus as well as their characteristics and likely function. The scrutiny of their DNA encoding sequences revealed the presence of minisatellite DNA in the P75 encoding gene of L. casei/paracasei strains (cmuB) with intraspecific indels that gave raise to four different alleles (cmuB1-4), which are exclusive of this species. Phylogenic analyses suggest that both proteins are present mainly in the L. casei and Lactobacillus sakei phylogenomic groups. A P40 ancestral gene was possibly present in the common ancestor of Enterococcaceae, Lactobacillaceae and Streptococcaceae. P75 is also present in L. casei and L. sakei groups, but its evolution is difficult to explain only by vertical transmission. Antibodies raised against the N-terminal regions of P40 and P75 improved their immunological detection in culture supernatants as they recognized almost exclusively proteins of L. casei/paracasei/rhamnosus strains, highlighting their structural similarity, that allowed to detect them in different fermented dairy products that contained probiotic L. casei strains. Purified P40 and P75 proteins showed no evident lytic activity but they complemented L. casei BL23 cmuA and cmuB defective mutants, respectively, thus proving that they actively participate in cell division.
Collapse
Affiliation(s)
- Christine Bäuerl
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| | - Gulyaim Abitayeva
- Department of Microbiology, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan.,Laboratory of Genetics and Biochemistry of Microorganisms, Republican Collection of Microorganisms at Science Committee of Ministry of Education and Science RK, Astana, Kazakhstan
| | - Sebastián Sosa-Carrillo
- Computational Biology Department, Inria, Institut Pasteur and Université Paris Diderot, Paris, France
| | | | - Noemí Navarro-Lleó
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| | - José M Coll-Marqués
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| | - Manuel Zúñiga-Cabrera
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| | - Serik Shaikhin
- Department of Microbiology, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan.,Laboratory of Genetics and Biochemistry of Microorganisms, Republican Collection of Microorganisms at Science Committee of Ministry of Education and Science RK, Astana, Kazakhstan
| | - Gaspar Pérez-Martinez
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
| |
Collapse
|
37
|
Gimeno-Alcañiz JV, Collado MC. Impact of human milk on the transcriptomic response of fetal intestinal epithelial cells reveals expression changes of immune-related genes. Food Funct 2019; 10:140-150. [PMID: 30499575 PMCID: PMC6350622 DOI: 10.1039/c8fo01107a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Human milk, the best food for infants, is a dynamic and complex fluid that directly influences the immune system and microbiota establishment.
Human milk, the best food for infants, is a dynamic and complex fluid that directly influences the immune system and microbiota establishment. The protective role of human milk is well known although the mechanisms behind it still need to be uncovered. This study aimed to characterize the impact of human milk in the immature intestine of newborns by analyzing the global transcriptomic response of the FHs 74 int cell line (ATCC CCL-241). The expression of intestinal keratins and other genes with a well-annotated intestinal or epithelial function validated FHs 74 int derived from the fetal small intestine as a model of the intestinal epithelium of newborns. Cells exposed to skimmed human milk showed seventeen differentially expressed genes, most of them up-regulated, including four chemokine genes (CXCL1, CXCL2, CXCL3 and CXCL10) and other immune-related genes. qRT-PCR and ELISA analysis confirmed the microarray data and indicated a different pattern of expression upon milk exposure in FHs 74 int as compared to the adult tumorigenic Caco-2 cell line. The evaluation of the functional significance of these transcriptomic changes reveals that human milk exposure may contribute to the regulation of the inflammatory response in the intestine during the perinatal period, which is characterized by the immaturity of the immune system and a pro-inflammatory phenotype.
Collapse
Affiliation(s)
- José V Gimeno-Alcañiz
- Instituto de agroquímica y tecnología de alimentos (IATA-CSIC), Department of Biotechnology, Avenida Agustín Escardino 7, 46980 Paterna, Spain.
| | | |
Collapse
|
38
|
An Acid Up-Regulated Surface Protein of Lactobacillus paracasei Strain GCRL 46 is Phylogenetically Related to the Secreted Glucan- (GpbB) and Immunoglobulin-Binding (SibA) Protein of Pathogenic Streptococci. Int J Mol Sci 2019; 20:ijms20071610. [PMID: 30935131 PMCID: PMC6479570 DOI: 10.3390/ijms20071610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/22/2019] [Accepted: 03/28/2019] [Indexed: 02/07/2023] Open
Abstract
Bacterial cell wall hydrolases, including amidases and peptidases, play a critical role in peptidoglycan turnover during growth, impacting daughter cell separation, and cell death, through autolysis. When exploring the regulation of protein expression across the growth cycle of an acid-resistant strain of Lactobacillus paracasei, GCRL 46, we observed temporal up-regulation of proteins in the 40⁻45 kDa molecular weight range for whole-cell extracts when culturing in fermenters at a controlled pH of 4.0 versus optimum growth pH of 6.3. Up-regulation of proteins in this size range was not detected in SDS-PAGE gels of the cytosolic fraction, but was routinely detected following growth at low pH in whole cells and cell debris obtained after bead beating and centrifugation, indicating a cell surface location. N-terminal sequencing and in silico analyses showed sequence similarity with proteins in the L. casei group (L. casei, L. paracasei and L. rhamnosus) which were variously annotated as uncharacterized proteins, surface antigens, possible TrsG proteins, CHAP (cysteine, histidine-dependent amidohydrolases/peptidases)-domain proteins or putative peptidoglycan d,l-endopeptidase due to the presence of a CwlO domain. This protein is a homologue of the p40 (Msp2) secreted protein of L. rhamnosus LGG, which is linked to probiotic functionality in this species, and is phylogenetically related to structurally-similar proteins found in Enterococcus, Streptococcus and Bifidobacterium species, including the glucan-binding (GbpB), surface antigen (SagA) proteins detected in pathogenic group A streptococci species as secreted, immunoglobulin-binding (SibA) proteins (also named PcsB). Three-dimensional (3D) modelling predicted structural similarities in the CHAP proteins from the L. casei group and streptococcal species, indicating retention of overall architecture despite sequence divergence, and an implied retention of function during evolution. A phylogenetically-related hydrolase also contained the CwlO domain with a NLPC_P60 domain, and showed similar overall but distinct architecture to the CHAP proteins. We concluded that the surface-located, CHAP protein in L. casei is up-regulated during long-term exposure to acidic conditions during growth but not during acid shock.
Collapse
|
39
|
Huang S, Gaucher F, Cauty C, Jardin J, Le Loir Y, Jeantet R, Chen XD, Jan G. Growth in Hyper-Concentrated Sweet Whey Triggers Multi Stress Tolerance and Spray Drying Survival in Lactobacillus casei BL23: From the Molecular Basis to New Perspectives for Sustainable Probiotic Production. Front Microbiol 2018; 9:2548. [PMID: 30405593 PMCID: PMC6204390 DOI: 10.3389/fmicb.2018.02548] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/05/2018] [Indexed: 12/14/2022] Open
Abstract
Lactobacillus casei BL23 has a recognized probiotic potential, which includes immune modulation, protection toward induced colitis, toward induced colon cancer and toward dissemination of pathogens. In L. casei, as well as in other probiotics, both probiotic and technological abilities are highly dependent (1) on the substrate used to grow bacteria and (2) on the process used to dry and store this biomass. Production and storage of probiotics, at a reasonable financial and environmental cost, becomes a crucial challenge. Food-grade media must be used, and minimal process is preferred. In this context, we have developed a “2-in-1” medium used both to grow and to dry L. casei BL23, considered a fragile probiotic strain. This medium consists in hyper-concentrated sweet whey (HCSW). L. casei BL23 grows in HCSW up to 30% dry matter, which is 6 times-concentrated sweet whey. Compared to isotonic sweet whey (5% dry matter), these growth conditions enhanced tolerance of L. casei BL23 toward heat, acid and bile salts stress. HCSW also triggered intracellular accumulation of polyphosphate, of glycogen and of trehalose. A gel-free global proteomic differential analysis further evidenced overexpression of proteins involved in pathways known to participate in stress adaptation, including environmental signal transduction, oxidative and metal defense, DNA repair, protein turnover and repair, carbohydrate, phosphate and amino acid metabolism, and in osmoadaptation. Accordingly, HCSW cultures of L. casei BL23 exhibited enhanced survival upon spray drying, a process known to drastically affect bacterial viability. This work opens new perspectives for sustainable production of dried probiotic lactobacilli, using food industry by-products and lowering energy costs.
Collapse
Affiliation(s)
- Song Huang
- Suzhou Key Laboratory of Green Chemical Engineering, School of Chemical and Environmental Engineering, College of Chemistry, Chemical Engineering and Material Science, Soochow University, Jiangsu, China.,UMR1253 STLO, Agrocampus Ouest, INRA, Rennes, France
| | - Floriane Gaucher
- UMR1253 STLO, Agrocampus Ouest, INRA, Rennes, France.,Bioprox, Levallois-Perret, France
| | - Chantal Cauty
- UMR1253 STLO, Agrocampus Ouest, INRA, Rennes, France
| | - Julien Jardin
- UMR1253 STLO, Agrocampus Ouest, INRA, Rennes, France
| | - Yves Le Loir
- UMR1253 STLO, Agrocampus Ouest, INRA, Rennes, France
| | - Romain Jeantet
- Suzhou Key Laboratory of Green Chemical Engineering, School of Chemical and Environmental Engineering, College of Chemistry, Chemical Engineering and Material Science, Soochow University, Jiangsu, China.,UMR1253 STLO, Agrocampus Ouest, INRA, Rennes, France
| | - Xiao Dong Chen
- Suzhou Key Laboratory of Green Chemical Engineering, School of Chemical and Environmental Engineering, College of Chemistry, Chemical Engineering and Material Science, Soochow University, Jiangsu, China
| | - Gwénaël Jan
- UMR1253 STLO, Agrocampus Ouest, INRA, Rennes, France
| |
Collapse
|
40
|
Bron PA, Kleerebezem M. Lactic Acid Bacteria for Delivery of Endogenous or Engineered Therapeutic Molecules. Front Microbiol 2018; 9:1821. [PMID: 30123213 PMCID: PMC6085456 DOI: 10.3389/fmicb.2018.01821] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
Food-grade lactic acid bacteria (LAB) are considered suitable vehicles for the production and/or delivery of health promoting or therapeutic, bioactive molecules. The molecules considered for health-beneficial use include the endogenous effector molecules produced by probiotics (mostly lactobacilli), as well as heterologous bioactives that can be produced in LAB by genetic engineering (mostly using lactococci). Both strategies aim to deliver appropriate dosages of specific bioactive molecules to the site of action. This review uses specific examples of both strategies to illustrate the different avenues of research involved in these applications as well as their translation to human health-promoting applications. These examples pinpoint that despite the promising perspectives of these approaches, the evidence for their effective applications in human populations is lagging behind.
Collapse
Affiliation(s)
- Peter A Bron
- NIZO Food Research BV, Ede, Netherlands.,BE-Basic Foundation, Delft, Netherlands
| | - Michiel Kleerebezem
- BE-Basic Foundation, Delft, Netherlands.,Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
41
|
Petrova MI, van den Broek MFL, Spacova I, Verhoeven TLA, Balzarini J, Vanderleyden J, Schols D, Lebeer S. Engineering Lactobacillus rhamnosus GG and GR-1 to express HIV-inhibiting griffithsin. Int J Antimicrob Agents 2018; 52:599-607. [PMID: 30040991 DOI: 10.1016/j.ijantimicag.2018.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 06/14/2018] [Accepted: 07/08/2018] [Indexed: 11/30/2022]
Abstract
Probiotic bacteria are being explored for the in situ delivery of various therapeutic agents. In this study, we aimed to express two HIV-inhibiting lectins, actinohivin (AH) and griffithsin (GRFT), in the probiotic strains Lactobacillus rhamnosus GG and L. rhamnosus GR-1 for gastrointestinal and vaginal mucosal delivery, respectively. Constructs were generated for the intracellular and extracellular production of AH and GRFT under the control of the promoter of their Major Secreted Protein Msp1. Also, intracellular expression of GRFT was investigated under the control of the nisA promoter from the inducible nisin-controlled expression (NICE) system. For the extracellular localization, the signal leader peptide of Msp1/p75 from L. rhamnosus GG was translationally fused with the genes encoding AH and GRFT. Construction of recombinant strains expressing the AH monomer and dimer was unsuccessful, probably due to the intracellular toxicity of AH for the lactobacilli. On the other hand, recombinant strains for intra- and extracellular production of GRFT by L. rhamnosus GG and GR-1 were successfully constructed. The highest expression levels of recombinant GRFT were observed for the constructs under the control of the inducible nisA promoter and we demonstrated anti-HIV activity against an M-tropic and a T-tropic HIV-1 strain. We can conclude that recombinant Lactobacillus expressing anti-HIV lectins could contribute to the development of enhanced probiotic strains that are able to inhibit HIV transmission and subsequent replication, although further research and development are required.
Collapse
Affiliation(s)
- Mariya I Petrova
- KU Leuven, Centre of Microbial and Plant Genetics, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium; University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium.
| | - Marianne F L van den Broek
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Irina Spacova
- KU Leuven, Centre of Microbial and Plant Genetics, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium; University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Tine L A Verhoeven
- KU Leuven, Centre of Microbial and Plant Genetics, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium
| | - Jan Balzarini
- KU Leuven, Rega Institute for Medical Research, Herestraat 49, B-3000 Leuven, Belgium
| | - Jos Vanderleyden
- KU Leuven, Centre of Microbial and Plant Genetics, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium
| | - Dominique Schols
- KU Leuven, Rega Institute for Medical Research, Herestraat 49, B-3000 Leuven, Belgium
| | - Sarah Lebeer
- KU Leuven, Centre of Microbial and Plant Genetics, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium; University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium.
| |
Collapse
|
42
|
Vinusha KS, Deepika K, Johnson TS, Agrawal GK, Rakwal R. Proteomic studies on lactic acid bacteria: A review. Biochem Biophys Rep 2018; 14:140-148. [PMID: 29872746 PMCID: PMC5986552 DOI: 10.1016/j.bbrep.2018.04.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/02/2018] [Accepted: 04/17/2018] [Indexed: 02/07/2023] Open
Abstract
Probiotics are amongst the most common microbes in the gastro-intestinal tract of humans and other animals. Prominent among probiotics are Lactobacillus and Bifidobacterium. They offer wide-ranging health promoting benefits to the host which include reduction in pathological alterations, stimulation of mucosal immunity and interaction with mediators of inflammation among others. Proteomics plays a vital role in understanding biological functions of a cell. Proteomics is also slowly and steadily adding to the existing knowledge on role of probiotics. In this paper, the proteomics of probiotics, with special reference to lactic acid bacteria is reviewed with a view to understand i) proteome map, ii) mechanism of adaptation to harsh gut environment such as low pH and bile acid, iii) role of cell surface proteins in adhering to intestinal epithelial cells, and iv) as a tool to answer basic cell functions. We have also reviewed various analytical methods used to carry out proteome analysis, in which 2D-MS and LC-MS/MS approaches were found to be versatile methods to perform high-throughput sample analyses even for a complex gut samples. Further, we present future road map of understanding gut microbes combining meta-proteomics, meta-genomics, meta-transcriptomics and -metabolomics.
Collapse
Affiliation(s)
- K Sri Vinusha
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - K Deepika
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - T Sudhakar Johnson
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - Ganesh K Agrawal
- Research Laboratory for Biotechnology and Biochemistry (RLABB), GPO Box 13265, Kathmandu, Nepal.,GRADE Academy Private Limited, Adarsh Nagar-13, Birgunj, Nepal
| | - Randeep Rakwal
- Research Laboratory for Biotechnology and Biochemistry (RLABB), GPO Box 13265, Kathmandu, Nepal.,GRADE Academy Private Limited, Adarsh Nagar-13, Birgunj, Nepal.,Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8574, Japan.,Global Research Center for Innovative Life Science, Peptide Drug Innovation, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 4-41 Ebara 2-chome, Shinagawa, Tokyo 142-8501, Japan
| |
Collapse
|
43
|
Engevik MA, Versalovic J. Biochemical Features of Beneficial Microbes: Foundations for Therapeutic Microbiology. Microbiol Spectr 2017; 5:10.1128/microbiolspec.BAD-0012-2016. [PMID: 28984235 PMCID: PMC5873327 DOI: 10.1128/microbiolspec.bad-0012-2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Indexed: 12/15/2022] Open
Abstract
Commensal and beneficial microbes secrete myriad products which target the mammalian host and other microbes. These secreted substances aid in bacterial niche development, and select compounds beneficially modulate the host and promote health. Microbes produce unique compounds which can serve as signaling factors to the host, such as biogenic amine neuromodulators, or quorum-sensing molecules to facilitate inter-bacterial communication. Bacterial metabolites can also participate in functional enhancement of host metabolic capabilities, immunoregulation, and improvement of intestinal barrier function. Secreted products such as lactic acid, hydrogen peroxide, bacteriocins, and bacteriocin-like substances can also target the microbiome. Microbes differ greatly in their metabolic potential and subsequent host effects. As a result, knowledge about microbial metabolites will facilitate selection of next-generation probiotics and therapeutic compounds derived from the mammalian microbiome. In this article we describe prominent examples of microbial metabolites and their effects on microbial communities and the mammalian host.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 and Department of Pathology, Texas Children's Hospital, Houston, TX 77030
| | - James Versalovic
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 and Department of Pathology, Texas Children's Hospital, Houston, TX 77030
| |
Collapse
|
44
|
Domínguez Rubio AP, Martínez JH, Martínez Casillas DC, Coluccio Leskow F, Piuri M, Pérez OE. Lactobacillus casei BL23 Produces Microvesicles Carrying Proteins That Have Been Associated with Its Probiotic Effect. Front Microbiol 2017; 8:1783. [PMID: 28979244 PMCID: PMC5611436 DOI: 10.3389/fmicb.2017.01783] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/04/2017] [Indexed: 12/20/2022] Open
Abstract
Archaea, bacteria, and eukarya secrete membrane microvesicles (MVs) as a mechanism for intercellular communication. We report the isolation and characterization of MVs from the probiotic strain Lactobacillus casei BL23. MVs were characterized using analytical high performance techniques, DLS, AFM and TEM. Similar to what has been described for other Gram-positive bacteria, MVs were on the nanometric size range (30–50 nm). MVs carried cytoplasmic components such as DNA, RNA and proteins. Using a proteomic approach (LC-MS), we identified a total of 103 proteins; 13 exclusively present in the MVs. The MVs content included cell envelope associated and secretory proteins, heat and cold shock proteins, several metabolic enzymes, proteases, structural components of the ribosome, membrane transporters, cell wall-associated hydrolases and phage related proteins. In particular, we identified proteins described as mediators of Lactobacillus’ probiotic effects such as p40, p75 and the product of LCABL_31160, annotated as an adhesion protein. The presence of these proteins suggests a role for the MVs in the bacteria-gastrointestinal cells interface. The expression and further encapsulation of proteins into MVs of GRAS (Generally Recognized as Safe) bacteria could represent a scientific novelty, with applications in food, nutraceuticals and clinical therapies.
Collapse
Affiliation(s)
- A Paula Domínguez Rubio
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina.,Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Jimena H Martínez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina.,Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Diana C Martínez Casillas
- Departamento de Física de la Materia Condensada, Centro Nacional de Energía AtómicaBuenos Aires, Argentina
| | - Federico Coluccio Leskow
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina.,Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina.,Departamento de Ciencias Básicas, Universidad Nacional de LujánBuenos Aires, Argentina
| | - Mariana Piuri
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina.,Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Oscar E Pérez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos AiresBuenos Aires, Argentina.,Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina.,Departamento de Desarrollo Productivo y Tecnológico, Universidad Nacional de LanúsBuenos Aires, Argentina
| |
Collapse
|
45
|
Ilinskaya ON, Ulyanova VV, Yarullina DR, Gataullin IG. Secretome of Intestinal Bacilli: A Natural Guard against Pathologies. Front Microbiol 2017; 8:1666. [PMID: 28919884 PMCID: PMC5586196 DOI: 10.3389/fmicb.2017.01666] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/17/2017] [Indexed: 12/12/2022] Open
Abstract
Current studies of human gut microbiome usually do not consider the special functional role of transient microbiota, although some of its members remain in the host for a long time and produce broad spectrum of biologically active substances. Getting into the gastrointestinal tract (GIT) with food, water and probiotic preparations, two representatives of Bacilli class, genera Bacillus and Lactobacillus, colonize epithelium blurring the boundaries between resident and transient microbiota. Despite their minor proportion in the microbiome composition, these bacteria can significantly affect both the intestinal microbiota and the entire body thanks to a wide range of secreted compounds. Recently, insufficiency and limitations of pure genome-based analysis of gut microbiota became known. Thus, the need for intense functional studies is evident. This review aims to characterize the Bacillus and Lactobacillus in GIT, as well as the functional roles of the components released by these members of microbial intestinal community. Complex of their secreted compounds is referred by us as the "bacillary secretome." The composition of the bacillary secretome, its biological effects in GIT and role in counteraction to infectious diseases and oncological pathologies in human organism is the subject of the review.
Collapse
Affiliation(s)
| | - Vera V. Ulyanova
- Department of Microbiology, Kazan Federal UniversityKazan, Russia
| | | | - Ilgiz G. Gataullin
- Department of Surgery and Oncology, Regional Clinical Cancer CenterKazan, Russia
| |
Collapse
|
46
|
Zhang L, Yu Y, Li X, Li X, Zhang H, Zhang Z, Xu Y. Starch Flocculation by the Sweet Potato Sour Liquid Is Mediated by the Adhesion of Lactic Acid Bacteria to Starch. Front Microbiol 2017; 8:1412. [PMID: 28791000 PMCID: PMC5524740 DOI: 10.3389/fmicb.2017.01412] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 07/11/2017] [Indexed: 01/09/2023] Open
Abstract
In the current study, we focused on the mechanism underlying starch flocculation by the sweet potato sour liquid. The traditional microbial techniques and 16S rDNA sequencing revealed that Lactobacillus was dominant flocculating microorganism in sour liquid. In total, 86 bacteria, 20 yeasts, and 10 molds were isolated from the sour liquid and only eight Lactobacillus species exhibited flocculating activity. Lactobacillus paracasei subsp. paracasei L1 strain with a high flocculating activity was isolated and identified, and the mechanism of starch flocculation was examined. L. paracasei subsp. paracasei L1 cells formed chain-like structures on starch granules. Consequently, these cells connected the starch granules to one another, leading to formation of large flocs. The results of various treatments of L1 cells indicated that bacterial surface proteins play a role in flocculation and L1 cells adhered to the surface of starch granules via specific surface proteins. These surface starch-binding proteins were extracted using the guanidine hydrochloride method; 10 proteins were identified by mass spectrometry: three of these proteins were glycolytic enzymes; two were identified as the translation elongation factor Tu; one was a cell wall hydrolase; one was a surface antigen; one was lyzozyme M1; one was a glycoside hydrolase; and one was an uncharacterized proteins. This study will paves the way for future industrial application of the L1 isolate in starch processing and food manufacturing.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Food Science and Engineering, Jinzhou Medical UniversityJinzhou, China.,Liaoning Provincial Research Center of Meat Processing and Quality ControlJinzhou, China
| | - Yang Yu
- Department of Food Science and Engineering, Jinzhou Medical UniversityJinzhou, China
| | - Xinhua Li
- Department of Food Science, Shenyang Agricultural UniversityShenyang, China
| | - Xiaona Li
- Department of Food Science, Shenyang Agricultural UniversityShenyang, China
| | - Huajiang Zhang
- Department of Food Science, Northeast Agricultural UniversityHarbin, China
| | - Zhen Zhang
- Department of Food Science and Engineering, Jinzhou Medical UniversityJinzhou, China
| | - Yunhe Xu
- Department of Food Science and Engineering, Jinzhou Medical UniversityJinzhou, China
| |
Collapse
|
47
|
Salvetti E, O'Toole PW. The Genomic Basis of Lactobacilli as Health-Promoting Organisms. Microbiol Spectr 2017; 5:10.1128/microbiolspec.bad-0011-2016. [PMID: 28643623 PMCID: PMC11687495 DOI: 10.1128/microbiolspec.bad-0011-2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Indexed: 12/11/2022] Open
Abstract
Lactobacilli occupy a unique position in human culture and scientific history. Like brewer's and baker's yeast, lactobacilli have been associated with food production and preservation for thousands of years. Lactobacillus species are used in mixed microbial cultures, such as the classical Lactobacillus bulgaricus/Streptococcus thermophilus inoculum for yogurt fermentation, or combinations of diverse lactobacilli/yeasts in kefir grains. The association of lactobacilli consumption with greater longevity and improved health formed the basis for developing lactobacilli as probiotics, whose market has exploded worldwide in the past 10 years. The decade that followed the determination of the first genome sequence of a food-associated species, Lactobacillus plantarum, saw the application to lactobacilli of a full range of functional genomics methods to identify the genes and gene products that govern their distinctive phenotypes and health associations. In this review, we will briefly remind the reader of the range of beneficial effects attributed to lactobacilli, and then explain the phylogenomic basis for the distribution of these traits across the genus. Recognizing the strain specificity of probiotic effects, we review studies of intraspecific genomic variation and their contributions to identifying probiotic traits. Finally we offer a perspective on classification of lactobacilli into new genera in a scheme that will make attributing probiotic properties to clades, taxa, and species more logical and more robust.
Collapse
Affiliation(s)
- Elisa Salvetti
- School of Microbiology and APC Microbiome Institute, University College Cork, Ireland
| | - Paul W O'Toole
- School of Microbiology and APC Microbiome Institute, University College Cork, Ireland
| |
Collapse
|
48
|
van Beek AA, Sovran B, Hugenholtz F, Meijer B, Hoogerland JA, Mihailova V, van der Ploeg C, Belzer C, Boekschoten MV, Hoeijmakers JHJ, Vermeij WP, de Vos P, Wells JM, Leenen PJM, Nicoletti C, Hendriks RW, Savelkoul HFJ. Supplementation with Lactobacillus plantarum WCFS1 Prevents Decline of Mucus Barrier in Colon of Accelerated Aging Ercc1-/Δ7 Mice. Front Immunol 2016; 7:408. [PMID: 27774093 PMCID: PMC5054004 DOI: 10.3389/fimmu.2016.00408] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/22/2016] [Indexed: 11/23/2022] Open
Abstract
Although it is clear that probiotics improve intestinal barrier function, little is known about the effects of probiotics on the aging intestine. We investigated effects of 10-week bacterial supplementation of Lactobacillus plantarum WCFS1, Lactobacillus casei BL23, or Bifidobacterium breve DSM20213 on gut barrier and immunity in 16-week-old accelerated aging Ercc1−/Δ7 mice, which have a median lifespan of ~20 weeks, and their wild-type littermates. The colonic barrier in Ercc1−/Δ7 mice was characterized by a thin (< 10 μm) mucus layer. L. plantarum prevented this decline in mucus integrity in Ercc1−/Δ7 mice, whereas B. breve exacerbated it. Bacterial supplementations affected the expression of immune-related genes, including Toll-like receptor 4. Regulatory T cell frequencies were increased in the mesenteric lymph nodes of L. plantarum- and L. casei-treated Ercc1−/Δ7 mice. L. plantarum- and L. casei-treated Ercc1−/Δ7 mice showed increased specific antibody production in a T cell-dependent immune response in vivo. By contrast, the effects of bacterial supplementation on wild-type control mice were negligible. Thus, supplementation with L. plantarum – but not with L. casei and B. breve – prevented the decline in the mucus barrier in Ercc1−/Δ7 mice. Our data indicate that age is an important factor influencing beneficial or detrimental effects of candidate probiotics. These findings also highlight the need for caution in translating beneficial effects of probiotics observed in young animals or humans to the elderly.
Collapse
Affiliation(s)
- Adriaan A van Beek
- Cell Biology and Immunology Group, Wageningen University, Wageningen, Netherlands; Top Institute Food and Nutrition, Wageningen, Netherlands; Gut Health and Food Safety, Institute of Food Research, Norwich, UK
| | - Bruno Sovran
- Top Institute Food and Nutrition, Wageningen, Netherlands; Host Microbe Interactomics, Wageningen University, Wageningen, Netherlands
| | - Floor Hugenholtz
- Top Institute Food and Nutrition, Wageningen, Netherlands; Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Ben Meijer
- Cell Biology and Immunology Group, Wageningen University , Wageningen , Netherlands
| | - Joanne A Hoogerland
- Cell Biology and Immunology Group, Wageningen University , Wageningen , Netherlands
| | - Violeta Mihailova
- Cell Biology and Immunology Group, Wageningen University , Wageningen , Netherlands
| | - Corine van der Ploeg
- Cell Biology and Immunology Group, Wageningen University , Wageningen , Netherlands
| | - Clara Belzer
- Top Institute Food and Nutrition, Wageningen, Netherlands; Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Mark V Boekschoten
- Top Institute Food and Nutrition, Wageningen, Netherlands; Human Nutrition, Wageningen University, Wageningen, Netherlands
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands; CECAD Forschungszentrum, Universität zu Köln, Köln, Germany
| | - Wilbert P Vermeij
- Department of Molecular Genetics, Erasmus University Medical Center , Rotterdam , Netherlands
| | - Paul de Vos
- Top Institute Food and Nutrition, Wageningen, Netherlands; University of Groningen, Groningen, Netherlands
| | - Jerry M Wells
- Top Institute Food and Nutrition, Wageningen, Netherlands; Host Microbe Interactomics, Wageningen University, Wageningen, Netherlands
| | - Pieter J M Leenen
- Department of Immunology, Erasmus University Medical Center , Rotterdam , Netherlands
| | - Claudio Nicoletti
- Gut Health and Food Safety, Institute of Food Research, Norwich, UK; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center , Rotterdam , Netherlands
| | - Huub F J Savelkoul
- Cell Biology and Immunology Group, Wageningen University, Wageningen, Netherlands; Top Institute Food and Nutrition, Wageningen, Netherlands
| |
Collapse
|
49
|
Tytgat HLP, Schoofs G, Vanderleyden J, Van Damme EJM, Wattiez R, Lebeer S, Leroy B. Systematic Exploration of the Glycoproteome of the Beneficial Gut Isolate Lactobacillus rhamnosus GG. J Mol Microbiol Biotechnol 2016; 26:345-58. [PMID: 27463506 DOI: 10.1159/000447091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/25/2016] [Indexed: 11/19/2022] Open
Abstract
Glycoproteins form an interesting class of macromolecules involved in bacterial-host interactions, but they are not yet widely explored in Gram-positive and beneficial species. Here, an integrated and widely applicable approach was followed to identify putative bacterial glycoproteins, combining proteome fractionation with 2D protein and glycostained gels and lectin blots. This approach was validated for the microbiota isolate Lactobacillus rhamnosus GG. The approach resulted in a list of putative glycosylated proteins receiving a 'glycosylation score'. Ultimately, we could identify 41 unique glycosylated proteins in L. rhamnosus GG (6 top-confidence, 10 high-confidence and 25 putative hits; classification based on glycosylation score). Most glycoproteins are associated with the cell wall and membrane. Identified glycoproteins include proteins involved in transport, translation, and sugar metabolism processes. A robust screening resulted in a comprehensive mapping of glycoproteins in L. rhamnosus GG. Our results reflect the glycosylation of sugar metabolism enzymes, transporters, and other proteins crucial for cell physiology. We hypothesize that protein glycosylation can confer an extra level of regulation, for example by affecting enzyme functions. This is the first systematic study of the glycoproteome of a probiotic and beneficial gut isolate.
Collapse
Affiliation(s)
- Hanne L P Tytgat
- Centre of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
50
|
Ceapa C, Davids M, Ritari J, Lambert J, Wels M, Douillard FP, Smokvina T, de Vos WM, Knol J, Kleerebezem M. The Variable Regions of Lactobacillus rhamnosus Genomes Reveal the Dynamic Evolution of Metabolic and Host-Adaptation Repertoires. Genome Biol Evol 2016; 8:1889-905. [PMID: 27358423 PMCID: PMC4943194 DOI: 10.1093/gbe/evw123] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lactobacillus rhamnosus is a diverse Gram-positive species with strains isolated from different ecological niches. Here, we report the genome sequence analysis of 40 diverse strains of L. rhamnosus and their genomic comparison, with a focus on the variable genome. Genomic comparison of 40 L. rhamnosus strains discriminated the conserved genes (core genome) and regions of plasticity involving frequent rearrangements and horizontal transfer (variome). The L. rhamnosus core genome encompasses 2,164 genes, out of 4,711 genes in total (the pan-genome). The accessory genome is dominated by genes encoding carbohydrate transport and metabolism, extracellular polysaccharides (EPS) biosynthesis, bacteriocin production, pili production, the cas system, and the associated clustered regularly interspaced short palindromic repeat (CRISPR) loci, and more than 100 transporter functions and mobile genetic elements like phages, plasmid genes, and transposons. A clade distribution based on amino acid differences between core (shared) proteins matched with the clade distribution obtained from the presence–absence of variable genes. The phylogenetic and variome tree overlap indicated that frequent events of gene acquisition and loss dominated the evolutionary segregation of the strains within this species, which is paralleled by evolutionary diversification of core gene functions. The CRISPR-Cas system could have contributed to this evolutionary segregation. Lactobacillus rhamnosus strains contain the genetic and metabolic machinery with strain-specific gene functions required to adapt to a large range of environments. A remarkable congruency of the evolutionary relatedness of the strains’ core and variome functions, possibly favoring interspecies genetic exchanges, underlines the importance of gene-acquisition and loss within the L. rhamnosus strain diversification.
Collapse
Affiliation(s)
- Corina Ceapa
- Gut Biology & Microbiology Platform, Nutricia Research Centre, Utrecht, the Netherlands Laboratory of Microbiology, Wageningen University, the Netherlands
| | - Mark Davids
- Laboratory of Systems and Synthetic Biology, Wageningen University, the Netherlands
| | - Jarmo Ritari
- RPU Immunobiology, Department of Bacteriology and Immunology, University of Helsinki, Finland
| | - Jolanda Lambert
- Gut Biology & Microbiology Platform, Nutricia Research Centre, Utrecht, the Netherlands
| | | | | | | | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, the Netherlands RPU Immunobiology, Department of Bacteriology and Immunology, University of Helsinki, Finland Department of Veterinary Biosciences, University of Helsinki, Finland
| | - Jan Knol
- Gut Biology & Microbiology Platform, Nutricia Research Centre, Utrecht, the Netherlands Laboratory of Microbiology, Wageningen University, the Netherlands
| | - Michiel Kleerebezem
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research Centre, Wageningen, the Netherlands
| |
Collapse
|