1
|
Alexander LM, Khalid S, Gallego-Lopez GM, Astmann TJ, Oh JH, Heggen M, Huss P, Fisher R, Mukherjee A, Raman S, Choi IY, Smith MN, Rogers CJ, Epperly MW, Knoll LJ, Greenberger JS, van Pijkeren JP. Development of a Limosilactobacillus reuteri therapeutic delivery platform with reduced colonization potential. Appl Environ Microbiol 2024; 90:e0031224. [PMID: 39480094 DOI: 10.1128/aem.00312-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/09/2024] [Indexed: 11/02/2024] Open
Abstract
Bacterial biotherapeutic delivery vehicles have the potential to treat a variety of diseases. This approach obviates the need to purify the recombinant effector molecule, allows delivery of therapeutics in situ via oral or intranasal administration, and protects the effector molecule during gastrointestinal transit. Lactic acid bacteria have been broadly developed as therapeutic delivery vehicles though risks associated with the colonization of a genetically modified microorganism have so-far not been addressed. Here, we present an engineered Limosilactobacillus reuteri strain with reduced colonization potential. We applied a dual-recombineering scheme for efficient barcoding and generated mutants in genes encoding five previously characterized and four uncharacterized putative adhesins. Compared with the wild type, none of the mutants were reduced in their ability to survive gastrointestinal transit in mice. CmbA was identified as a key protein in L. reuteri adhesion to HT-29 and enteroid cells. The nonuple mutant, a single strain with all nine genes encoding adhesins inactivated, had reduced capacity to adhere to enteroid monolayers. The nonuple mutant producing murine IFN-β was equally effective as its wild-type counterpart in mitigating radiation toxicity in mice. Thus, this work established a novel therapeutic delivery platform that lays a foundation for its application in other microbial therapeutic delivery candidates and furthers the progress of the L. reuteri delivery system towards human use.IMPORTANCEOne major advantage to leverage gut microbes that have co-evolved with the vertebrate host is that evolution already has taken care of the difficult task to optimize survival within a complex ecosystem. The availability of the ecological niche will support colonization. However, long-term colonization of a recombinant microbe may not be desirable. Therefore, strategies need to be developed to overcome this potential safety concern. In this work, we developed a single strain in which we inactivated the encoding sortase, and eight genes encoding characterized/putative adhesins. Each individual mutant was characterized for growth and adhesion to epithelial cells. On enteroid cells, the nonuple mutant has a reduced adhesion potential compared with the wild-type strain. In a model of total-body irradiation, the nonuple strain engineered to release murine interferon-β performed comparable to a derivative of the wild-type strain that releases interferon-β. This work is an important step toward the application of recombinant L. reuteri in humans.
Collapse
Affiliation(s)
- Laura M Alexander
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Saima Khalid
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Gina M Gallego-Lopez
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Theresa J Astmann
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mark Heggen
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Phil Huss
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Srivatsan Raman
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - In Young Choi
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Morgan N Smith
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Michael W Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Laura J Knoll
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joel S Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
2
|
Wu Q, Kan J, Cui Z, Ma Y, Liu X, Dong R, Huang D, Chen L, Du J, Fu C. Understanding the nutritional benefits through plant proteins-probiotics interactions: mechanisms, challenges, and perspectives. Crit Rev Food Sci Nutr 2024:1-19. [PMID: 38922612 DOI: 10.1080/10408398.2024.2369694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
The nutritional benefits of combining probiotics with plant proteins have sparked increasing research interest and drawn significant attention. The interactions between plant proteins and probiotics demonstrate substantial potential for enhancing the functionality of plant proteins. Fermented plant protein foods offer a unique blend of bioactive components and beneficial microorganisms that can enhance gut health and combat chronic diseases. Utilizing various probiotic strains and plant protein sources opens doors to develop innovative probiotic products with enhanced functionalities. Nonetheless, the mechanisms and synergistic effects of these interactions remain not fully understood. This review aims to delve into the roles of promoting health through the intricate interplay of plant proteins and probiotics. The regulatory mechanisms have been elucidated to showcase the synergistic effects, accompanied by a discussion on the challenges and future research prospects. It is essential to recognize that the interactions between plant proteins and probiotics encompass multiple mechanisms, highlighting the need for further research to address challenges in achieving a comprehensive understanding of these mechanisms and their associated health benefits.
Collapse
Affiliation(s)
- Qiming Wu
- Nutrilite Health Institute, Shanghai, China
| | - Juntao Kan
- Nutrilite Health Institute, Shanghai, China
| | - Zhengying Cui
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou, China
| | - Yuchen Ma
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou, China
| | - Xin Liu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou, China
| | - Ruifang Dong
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou, China
| | - Dejian Huang
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou, China
- Department of Food Science and Technology, National University of Singapore, Singapore
| | - Lin Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore
| | - Jun Du
- Nutrilite Health Institute, Shanghai, China
| | - Caili Fu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou, China
| |
Collapse
|
3
|
Mantel M, Durand T, Bessard A, Pernet S, Beaudeau J, Guimaraes-Laguna J, Maillard MB, Guédon E, Neunlist M, Le Loir Y, Jan G, Rolli-Derkinderen M. Propionibacterium freudenreichii CIRM-BIA 129 mitigates colitis through S layer protein B-dependent epithelial strengthening. Am J Physiol Gastrointest Liver Physiol 2024; 326:G163-G175. [PMID: 37988603 DOI: 10.1152/ajpgi.00198.2023] [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: 09/22/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
The growing incidence of human diseases involving inflammation and increased gut permeability makes the quest for protective functional foods more crucial than ever. Propionibacterium freudenreichii (P. freudenreichii) is a beneficial bacterium used in the dairy and probiotic industries. Selected strains exert anti-inflammatory effects, and the present work addresses whether the P. freudenreichii CIRM-BIA129, consumed daily in a preventive way, could protect mice from acute colitis induced by dextran sodium sulfate (DSS), and more precisely, whether it could protect from intestinal epithelial breakdown induced by inflammation. P. freudenreichii CIRM-BIA129 mitigated colitis severity and inhibited DSS-induced permeability. It limited crypt length reduction and promoted the expression of zonula occludens-1 (ZO-1), without reducing interleukin-1β mRNA (il-1β) expression. In vitro, P. freudenreichii CIRM-BIA129 prevented the disruption of a Caco-2 monolayer induced by proinflammatory cytokines. It increased transepithelial electrical resistance (TEER) and inhibited permeability induced by inflammation, along with an increased ZO-1 expression. Extracellular vesicles (EVs) from P. freudenreichii CIRM-BIA129, carrying the surface layer protein (SlpB), reproduced the protective effect of P. freudenreichii CIRM-BIA129. A mutant strain deleted for slpB (ΔslpB), or EVs from this mutant strain, had lost their protective effects and worsened both DSS-induced colitis and inflammation in vivo. These results shown that P. freudenreichii CIRM-BIA129 daily consumption has the potential to greatly alleviate colitis symptoms and, particularly, to counter intestinal epithelial permeability induced by inflammation by restoring ZO-1 expression through mechanisms involving S-layer protein B. They open new avenues for the use of probiotic dairy propionibacteria and/or postbiotic fractions thereof, in the context of gut permeability.NEW & NOTEWORTHY Propionibacterium freudenreichii reduces dextran sodium sulfate (DSS)-induced intestinal permeability in vivo. P. freudenreichii does not inhibit inflammation but damages linked to inflammation. P. freudenreichii inhibits intestinal epithelial breakdown through S-layer protein B. The protective effects of P. freudenreichii depend on S-layer protein B. Extracellular vesicles from P. freudenreichii CB 129 mimic the protective effect of the probiotic.
Collapse
Affiliation(s)
- Marine Mantel
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Tony Durand
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Anne Bessard
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Ségolène Pernet
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Julie Beaudeau
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
- Centres de Recherche en Nutrition Humaine-Ouest, Nantes, France
| | - Juliana Guimaraes-Laguna
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Marie-Bernadette Maillard
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Eric Guédon
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Michel Neunlist
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Yves Le Loir
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Gwénaël Jan
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Malvyne Rolli-Derkinderen
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| |
Collapse
|
4
|
Budzinski L, von Goetze V, Chang HD. Single-cell phenotyping of bacteria combined with deep sequencing for improved contextualization of microbiome analyses. Eur J Immunol 2024; 54:e2250337. [PMID: 37863831 DOI: 10.1002/eji.202250337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/22/2023]
Abstract
Great effort was made to characterize the bacterial communities inhabiting the human body as a factor in disease, resulting in the realization that a wide spectrum of diseases is associated with an altered composition of the microbiome. However, the identification of disease-relevant bacteria has been hindered by the high cross-sectional diversity of individual microbiomes, and in most cases, it remains unclear whether the observed alterations are cause or consequence of disease. Hence, innovative analysis approaches are required that enable inquiries of the microbiome beyond mere taxonomic cataloging. This review highlights the utility of microbiota flow cytometry, a single-cell analysis platform to directly interrogate cellular interactions, cell conditions, and crosstalk with the host's immune system within the microbiome to take into consideration the role of microbes as critical interaction partners of the host and the spectrum of microbiome alterations, beyond compositional changes. In conjunction with advanced sequencing approaches it could reveal the genetic potential of target bacteria and advance our understanding of taxonomic diversity and gene usage in the context of the microenvironment. Single-cell bacterial phenotyping has the potential to change our perspective on the human microbiome and empower microbiome research for the development of microbiome-based therapy approaches and personalized medicine.
Collapse
Affiliation(s)
- Lisa Budzinski
- Schwiete Laboratory for Microbiota and Inflammation, German Rheumatism Research Centre Berlin - A Leibniz Institute, Berlin, Germany
| | - Victoria von Goetze
- Schwiete Laboratory for Microbiota and Inflammation, German Rheumatism Research Centre Berlin - A Leibniz Institute, Berlin, Germany
| | - Hyun-Dong Chang
- Schwiete Laboratory for Microbiota and Inflammation, German Rheumatism Research Centre Berlin - A Leibniz Institute, Berlin, Germany
| |
Collapse
|
5
|
Kosmerl E, González-Orozco BD, García-Cano I, Ortega-Anaya J, Jiménez-Flores R. Milk phospholipids protect Bifidobacterium longum subsp. infantis during in vitro digestion and enhance polysaccharide production. Front Nutr 2023; 10:1194945. [PMID: 38024346 PMCID: PMC10657999 DOI: 10.3389/fnut.2023.1194945] [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: 03/27/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Bifidobacterium longum subsp. infantis is associated with the gut microbiota of breast-fed infants. Bifidobacterium infantis promotes intestinal barrier and immune function through several proposed mechanisms, including interactions between their surface polysaccharides, the host, and other gut microorganisms. Dairy foods and ingredients are some of the most conspicuous food-based niches for this species and may provide benefits for their delivery and efficacy in the gut. Milk phospholipid (MPL)-rich ingredients have been increasingly recognized for their versatile benefits to health, including interactions with the gut microbiota and intestinal cells. Therefore, our objective was to investigate the capacity for MPL to promote survival of B. infantis during simulated digestion and to modulate bacterial polysaccharide production. To achieve these aims, B. infantis was incubated with or without 0.5% MPL in de Man, Rogosa, and Sharpe (MRS) media at 37°C under anaerobiosis. Survival across the oral, gastric, and intestinal phases using in vitro digestion was measured using plate count, along with adhesion to goblet-like intestinal cells. MPL increased B. infantis survival at the end of the intestinal phase by at least 7% and decreased adhesion to intestinal cells. The bacterial surface characteristics, which may contribute to these effects, were assessed by ζ-potential, changes in surface proteins using comparative proteomics, and production of bound polysaccharides. MPL decreased the surface charge of the bifidobacteria from -17 to -24 mV and increased a 50 kDa protein (3-fold) that appears to be involved in protection from stress. The production of bound polysaccharides was measured using FTIR, HPLC, and TEM imaging. These techniques all suggest an increase in bound polysaccharide production at least 1.7-fold in the presence of MPL. Our results show that MPL treatment increases B. infantis survival during simulated digestion, induces a stress resistance surface protein, and yields greater bound polysaccharide production, suggesting its use as a functional ingredient to enhance probiotic and postbiotic effects.
Collapse
Affiliation(s)
- Erica Kosmerl
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| | | | - Israel García-Cano
- Department of Food Science and Technology, National Institute of Medical Sciences and Nutrition Salvador Zubirán, Mexico City, Mexico
| | | | - Rafael Jiménez-Flores
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
6
|
Kim DY, Lee TS, Jung DH, Song EJ, Jang AR, Park JY, Ahn JH, Seo IS, Song SJ, Kim YJ, Lee YJ, Lee YJ, Park JH. Oral Administration of Lactobacillus sakei CVL-001 Improves Recovery from Dextran Sulfate Sodium-Induced Colitis in Mice by Microbiota Modulation. Microorganisms 2023; 11:1359. [PMID: 37317332 DOI: 10.3390/microorganisms11051359] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 06/16/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an intestinal chronic inflammatory disease, and its incidence is steadily increasing. IBD is closely related to the intestinal microbiota, and probiotics are known to be a potential therapeutic agent for IBD. In our study, we evaluated the protective effect of Lactobacillus sakei CVL-001, isolated from Baechu kimchi, on dextran sulfated sodium (DSS)-induced colitis in mice. The oral administration of L. sakei CVL-001 according to the experimental schedule alleviated weight loss and disease activity in the mice with colitis. Furthermore, the length and histopathology of the colon improved. The expression of tumor necrosis factor (TNF)-α and interleukin (IL)-1β genes decreased in the colons of mice that were administered L. sakei CVL-001, whereas that of IL-10 increased. The expressions of genes coding for E-cadherin, claudin3, occludin, and mucin were also restored. In co-housed conditions, L. sakei CVL-001 administration did not improve disease activity, colon length, and histopathology. Microbiota analysis revealed that L. sakei CVL-001 administration increased the abundance of microbiota and altered Firmicutes/Bacteroidetes ratio, and decreased Proteobacteria. In conclusion, L. sakei CVL-001 administration protects mice from DSS-induced colitis by regulating immune response and intestinal integrity via gut microbiota modulation.
Collapse
Affiliation(s)
- Dong-Yeon Kim
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Tae-Sung Lee
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Do-Hyeon Jung
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Eun-Jung Song
- Nodcure, Inc., 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Ah-Ra Jang
- Nodcure, Inc., 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Ji-Yeon Park
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Jae-Hun Ahn
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - In-Su Seo
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Seung-Ju Song
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Yeong-Jun Kim
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Yun-Ji Lee
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Yeon-Ji Lee
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, Animal Medical Institute, College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
- Nodcure, Inc., 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| |
Collapse
|
7
|
Assandri MH, Malamud M, Trejo FM, Serradell MDLA. S-layer proteins as immune players: tales from pathogenic and non-pathogenic bacteria. CURRENT RESEARCH IN MICROBIAL SCIENCES 2023; 4:100187. [PMID: 37064268 PMCID: PMC10102220 DOI: 10.1016/j.crmicr.2023.100187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
In bacteria, as in other microorganisms, surface compounds interact with different pattern recognition receptors expressed by host cells, which usually triggers a variety of cellular responses that result in immunomodulation. The S-layer is a two-dimensional macromolecular crystalline structure formed by (glyco)-protein subunits that covers the surface of many species of Bacteria and almost all Archaea. In Bacteria, the presence of S-layer has been described in both pathogenic and non-pathogenic strains. As surface components, special attention deserves the role that S-layer proteins (SLPs) play in the interaction of bacterial cells with humoral and cellular components of the immune system. In this sense, some differences can be predicted between pathogenic and non-pathogenic bacteria. In the first group, the S-layer constitutes an important virulence factor, which in turn makes it a potential therapeutic target. For the other group, the growing interest to understand the mechanisms of action of commensal microbiota and probiotic strains has prompted the studies of the role of the S-layer in the interaction between the host immune cells and bacteria bearing this surface structure. In this review, we aim to summarize the main latest reports and the perspectives of bacterial SLPs as immune players, focusing on those from pathogenic and commensal/probiotic most studied species.
Collapse
|
8
|
Illikoud N, Mantel M, Rolli-Derkinderen M, Gagnaire V, Jan G. Dairy starters and fermented dairy products modulate gut mucosal immunity. Immunol Lett 2022; 251-252:91-102. [DOI: 10.1016/j.imlet.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2022]
|
9
|
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: 0.5] [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
|
10
|
Surface Layer Protein Pattern of Levilactobacillus brevis Strains Investigated by Proteomics. Nutrients 2022; 14:nu14183679. [PMID: 36145058 PMCID: PMC9504196 DOI: 10.3390/nu14183679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
The outermost constituent of many bacterial cells is represented by an S-layer, i.e., a semiporous lattice-like layer composed of self-assembling protein subunits called S-layer proteins (Slps). These proteins are involved in several processes, such as protecting against environmental stresses, mediating bacterial adhesion to host cells, and modulating gut immune response. Slps may also act as a scaffold for the external display of additional cell surface proteins also named S-layer associated proteins (SLAPs). Levilactobacillus brevis is an S-layer forming lactic acid bacterium present in many different environments, such as sourdough, milk, cheese, and the intestinal tract of humans and animals. This microorganism exhibits probiotic features including the inhibition of bacterial infection and the improvement of human immune function. The potential role of Slps in its probiotic and biotechnological features was documented. A shotgun proteomic approach was applied to identify in a single experiment both the Slps and the SLAPs pattern of five different L. brevis strains isolated from traditional sourdoughs of the Southern Italian region. This study reveals that these closely related strains expressed a specific pattern of surface proteins, possibly affecting their peculiar properties.
Collapse
|
11
|
Factors Affecting Spontaneous Endocytosis and Survival of Probiotic Lactobacilli in Human Intestinal Epithelial Cells. Microorganisms 2022; 10:microorganisms10061142. [PMID: 35744660 PMCID: PMC9230732 DOI: 10.3390/microorganisms10061142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/29/2022] Open
Abstract
Mutualistic bacteria have different forms of interaction with the host. In contrast to the invasion of pathogenic bacteria, naturally occurring internalization of commensal bacteria has not been studied in depth. Three in vitro methods, gentamicin protection, flow cytometry and confocal laser scanning microscopy, have been implemented to accurately assess the internalization of two lactobacillus strains—Lacticaseibacillus paracasei BL23 and Lacticaseibacillus rhamnosus GG—in Caco-2 and T84 intestinal epithelial cells (IECs) under a variety of physiological conditions and with specific inhibitors. First and most interesting, internalization occurred at a variable rate that depends on the bacterial strain and IEC line, and the most efficient was BL23 internalization by T84 and, second, efficient internalization required active IEC proliferation, as it improved naturally at the early confluence stages and by stimulation with epidermal growth factor (EGF). IFN-γ is bound to innate immune responses and autolysis; this cytokine had a significant effect on internalization, as shown by flow cytometry, but increased internalization was not perceived in all conditions, possibly because it was also stimulating autolysis and, as a consequence, the viability of bacteria after uptake could be affected. Bacterial uptake required actin polymerization, as shown by cytochalasin D inhibition, and it was partially bound to clathrin and caveolin dependent endocytosis. It also showed partial inhibition by ML7 indicating the involvement of cholesterol lipid rafts and myosin light chain kinase (MLCK) activation, at least in the LGG uptake by Caco-2. Most interestingly, bacteria remained viable inside the IEC for as long as 72 h without damaging the epithelial cells, and paracellular transcytosis was observed. These results stressed the fact that internalization of commensal and mutualistic bacteria is a natural, nonpathogenic process that may be relevant in crosstalk processes between the intestinal populations and the host, and future studies could determine its connection to processes such as commensal tolerance, resilience of microbial populations or transorganic bacterial migration.
Collapse
|
12
|
Khalifa A, Sheikh A, Ibrahim HIM. Bacillus amyloliquefaciens Enriched Camel Milk Attenuated Colitis Symptoms in Mice Model. Nutrients 2022; 14:1967. [PMID: 35565934 PMCID: PMC9101272 DOI: 10.3390/nu14091967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/30/2022] [Accepted: 05/04/2022] [Indexed: 12/22/2022] Open
Abstract
Fermented camel's milk has various health beneficial prebiotics and probiotics. This study aimed to evaluate the preventive efficacy of Bacillus amyloliquefaciens enriched camel milk (BEY) in 2-, 4- and 6-Trinitrobenzenesulfonic acid (TNBS)-induced colitis mice models. To this end, the immune modulatory effects of Bacillus amyloliquefaciens (BA) on TNF-α challenged HT29 colon cells were estimated using the cell proliferation and cytokines ELISA method. BEY was prepared using the incubation method and nutritional value was quantified by comparing it to commercial yogurt. Furthermore, TNBS-induced colitis was established and the level of disease index, pathological scores, and inflammatory markers of BEY-treated mice using macroscopic and microscopic examinations, qPCR and immunoblot were investigated. The results demonstrate that BA is non-toxic to HT29 colon cells and balanced the inflammatory cytokines. BEY reduced the colitis disease index, and improved the body weight and colon length of the TNBS-induced mice. Additionally, Myeloperoxidase (MPO) and pro-inflammatory cytokines (IL1β, IL6, IL8 and TNF-α) were attenuated by BEY treatment. Moreover, the inflammatory progress mRNA and protein markers nuclear factor kappa B (NFκB), phosphatase and tensin homolog (PTEN), proliferating cell nuclear antigen (PCNA), cyclooxygenase-2 (COX-2) and occludin were significantly down-regulated by BEY treatment. Interestingly, significant suppression of PCNA was observed in colonic tissues using the immunohistochemical examination. Treatment with BEY increased the epigenetic (microRNA217) interactions with PCNA. In conclusion, the BEY clearly alleviated the colitis symptoms and in the future could be used to formulate a probiotic-based diet for the host gut health and control the inflammatory bowel syndrome in mammals.
Collapse
Affiliation(s)
- Ashraf Khalifa
- Biological Science Department, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Abdullah Sheikh
- Camel Research Center, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
| | - Hairul Islam Mohamed Ibrahim
- Biological Science Department, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- Molecular Biology Division, Pondicherry Centre for Biological Sciences and Educational Trust, Kottakuppam 605104, India
| |
Collapse
|
13
|
Allouche R, Hafeez Z, Papier F, Dary-Mourot A, Genay M, Miclo L. In Vitro Anti-Inflammatory Activity of Peptides Obtained by Tryptic Shaving of Surface Proteins of Streptococcus thermophilus LMD-9. Foods 2022; 11:foods11081157. [PMID: 35454744 PMCID: PMC9030335 DOI: 10.3390/foods11081157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 12/16/2022] Open
Abstract
Streptococcus thermophilus, a lactic acid bacterium widely used in the dairy industry, is consumed regularly by a significant proportion of the population. Some strains show in vitro anti-inflammatory activity which is not fully understood. We hypothesized that peptides released from the surface proteins of this bacterium during digestion could be implied in this activity. Consequently, we prepared a peptide hydrolysate by shaving and hydrolysis of surface proteins using trypsin, and the origin of peptides was checked by liquid chromatography–tandem mass spectrometry (LC-MS/MS) analysis. Most of the identified peptides originated from bacterial cell surface proteins. The anti-inflammatory activity of peptide hydrolysate was investigated under inflammatory conditions in two cell models. Peptide hydrolysate significantly decreased secretion of pro-inflammatory cytokine IL-8 in lipopolysaccharide (LPS)-stimulated human colon epithelial HT-29 cells. It also reduced the production of pro-inflammatory cytokines IL-8, IL-1β and the protein expression levels of Pro-IL-1β and COX-2 in LPS-stimulated THP-1 macrophages. The results showed that peptides released from bacterial surface proteins by a pancreatic protease could therefore participate in an anti-inflammatory activity of S. thermophilus LMD-9 and could prevent low-grade inflammation.
Collapse
|
14
|
Gut health benefit and application of postbiotics in animal production. J Anim Sci Biotechnol 2022; 13:38. [PMID: 35392985 PMCID: PMC8991504 DOI: 10.1186/s40104-022-00688-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/04/2022] [Indexed: 01/05/2023] Open
Abstract
Gut homeostasis is of importance to host health and imbalance of the gut usually leads to disorders or diseases for both human and animal. Postbiotics have been applied in manipulating of gut health, and utilization of postbiotics threads new lights into the host health. Compared with the application of probiotics, the characteristics such as stability and safety of postbiotics make it a potential alternative to probiotics. Studies have reported the beneficial effects of components derived from postbiotics, mainly through the mechanisms including inhibition of pathogens, strengthen gut barrier, and/or regulation of immunity of the host. In this review, we summarized the characteristics of postbiotics, main compounds of postbiotics, potential mechanisms in gut health, and their application in animal production.
Collapse
|
15
|
Heavey MK, Durmusoglu D, Crook N, Anselmo AC. Discovery and delivery strategies for engineered live biotherapeutic products. Trends Biotechnol 2022; 40:354-369. [PMID: 34481657 PMCID: PMC8831446 DOI: 10.1016/j.tibtech.2021.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
Genetically engineered microbes that secrete therapeutics, sense and respond to external environments, and/or target specific sites in the gut fall under an emergent class of therapeutics, called live biotherapeutic products (LBPs). As live organisms that require symbiotic host interactions, LBPs offer unique therapeutic opportunities, but also face distinct challenges in the gut microenvironment. In this review, we describe recent approaches (often demonstrated using traditional probiotic microorganisms) to discover LBP chassis and genetic parts utilizing omics-based methods and highlight LBP delivery strategies, with a focus on addressing physiological challenges that LBPs encounter after oral administration. Finally, we share our perspective on the opportunity to apply an integrated approach, wherein discovery and delivery strategies are utilized synergistically, towards tailoring and optimizing LBP efficacy.
Collapse
Affiliation(s)
- Mairead K. Heavey
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Deniz Durmusoglu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Nathan Crook
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, USA.
| | - Aaron C. Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA,Correspondence: (A.C. Anselmo), (N. Crook)
| |
Collapse
|
16
|
Bozzetti V, Senger S. Organoid technologies for the study of intestinal microbiota–host interactions. Trends Mol Med 2022; 28:290-303. [PMID: 35232671 PMCID: PMC8957533 DOI: 10.1016/j.molmed.2022.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022]
Abstract
Postbiotics have recently emerged as critical effectors of the activity of probiotics and, because of their safety profile, they are considered potential therapeutics for the treatment of fragile patients. Here, we present recent studies on probiotics and postbiotics in the context of novel discovery tools, such as organoids and organoid-based platforms, and nontransformed preclinical models, that can be generated from intestinal stem cells. The implementation of organoid-related techniques is the next gold standard for unraveling the effect of microbial communities on homeostasis, inflammation, idiopathic diseases, and cancer in the gut. We also summarize recent studies on biotics in organoid-based models and offer our perspective on future directions.
Collapse
|
17
|
Belo GA, Cordeiro BF, Oliveira ER, Braga MP, da Silva SH, Costa BG, Martins FDS, Jan G, Le Loir Y, Gala-García A, Ferreira E, Azevedo V, do Carmo FLR. SlpB Protein Enhances the Probiotic Potential of L. lactis NCDO 2118 in Colitis Mice Model. Front Pharmacol 2021; 12:755825. [PMID: 34987390 PMCID: PMC8721164 DOI: 10.3389/fphar.2021.755825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/29/2021] [Indexed: 01/30/2023] Open
Abstract
Bacteria used in the production of fermented food products have been investigated for their potential role as modulators of inflammation in gastrointestinal tract disorders such as inflammatory bowel diseases (IBD) that cause irreversible changes in the structure and function of gut tissues. Ulcerative colitis (UC) is the most prevalent IBD in the population of Western countries, and it is marked by symptoms such as weight loss, rectal bleeding, diarrhea, shortening of the colon, and destruction of the epithelial layer. The strain Propionibacterium freudenreichii CIRM-BIA 129 recently revealed promising immunomodulatory properties that greatly rely on surface-layer proteins (Slp), notably SlpB. We, thus, cloned the sequence encoding the SlpB protein into the pXIES-SEC expression and secretion vector, and expressed the propionibacterial protein in the lactic acid bacterium Lactococcus lactis NCDO 2118. The probiotic potential of L. lactis NCDO 2118 harboring pXIES-SEC:slpB (L. lactis-SlpB) was evaluated in a UC-mice model induced by Dextran Sulfate Sodium (DSS). During colitis induction, mice receiving L. lactis-SlpB exhibited reduced severity of colitis, with lower weight loss, lower disease activity index, limited shortening of the colon length, and reduced histopathological score, with significant differences, compared with the DSS group and the group treated with L. lactis NCDO 2118 wild-type strain. Moreover, L. lactis-SlpB administration increased the expression of genes encoding tight junction proteins zo-1, cln-1, cln-5, ocln, and muc-2 in the colon, increased IL-10 and TGF-β, and decreased IL-17, TNF-α, and IL-12 cytokines in the colon. Therefore, this work demonstrates that SlpB recombinant protein is able to increase the probiotic potential of the L. lactis strain to alleviate DSS-induced colitis in mice. This opens perspectives for the development of new approaches to enhance the probiotic potential of strains by the addition of SlpB protein.
Collapse
Affiliation(s)
- Giovanna A. Belo
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Bárbara F. Cordeiro
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Emiliano R. Oliveira
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Marina P. Braga
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Sara H. da Silva
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Bruno G. Costa
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Flaviano dos S. Martins
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gwénaël Jan
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| | - Yves Le Loir
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| | - Alfonso Gala-García
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
- School of Dentistry, Federal University of Bahia (UFBA), Salvador, Brazil
| | - Enio Ferreira
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vasco Azevedo
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Fillipe L. R. do Carmo
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
- *Correspondence: Fillipe L. R. do Carmo,
| |
Collapse
|
18
|
Zheng D, Wang Z, Sui L, Xu Y, Wang L, Qiao X, Cui W, Jiang Y, Zhou H, Tang L, Li Y. Lactobacillus johnsonii activates porcine monocyte derived dendritic cells maturation to modulate Th cellular immune response. Cytokine 2021; 144:155581. [PMID: 34029942 DOI: 10.1016/j.cyto.2021.155581] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 02/02/2023]
Abstract
Lactobacilli are abundant in the intestinal tract where they constantly regulate immune system via interacting with a great diversity of immune cells, such as dendritic cells (DCs). Notably, DCs are powerful antigen-presenting cells and they are capable of initiating primary immune responses. In this study, we studied the effects of Lactobacillus johnsonii (L. johnsonii) and Lactobacillus johnsonii cell-free supernatant (L. johnsonii-CFS) on the activation of porcine monocyte-derived dendritic cells (MoDCs) and their regulation of Th cellular immune responses in vitro. The MoDCs generated from porcine peripheral blood monocytes were stimulated by L. johnsonii and L. johnsonii-CFS, respectively. Pre-incubation with L. johnsonii increased expression of CD172a, CD80, major histocompatibility complex class II (MHCII) in MoDCs, and enhanced the ability of MoDCs to induce the proliferation of CD4+ T cell, while pre-incubation with L. johnsonii-CFS merely upregulated the expression of MHCII. Analysis of the cytokines showed that L. johnsonii stimulated up-regulation of Th1-type cytokines (IL-12p40, IFN-γ, TNF-α), pro-inflammatory cytokine IL-1β, chemokine CCL20, and Treg-type / anti-inflammatory cytokines IL-10 in MoDCs. Notably, a high production of IL-10 was observed in the MoDCs treated with L. johnsonii-CFS, indicating L. johnsonii-CFS exerted anti-inflammatory effects. Furthermore, L. johnsonii induced up-regulation of TLR2 and TLR6, but L. johnsonii-CFS not. Moreover, MoDCs stimulated by L. johnsonii mainly promoted T cell differentiate into Th1/Th2/Treg cells and plays an important role in improving the balance between Th1/Th2/Treg-type cells, whereas MoDCs stimulated by L. johnsonii-CFS mainly directed T cell to Th2/Treg subset polarization. In conclusion, L. johnsonii and L. johnsonii-CFS exhibited the ability of modulating innate immunity by regulating immunological functions of MoDCs in vitro, suggesting their potential ability to use as microecological preparations and medicines.
Collapse
Affiliation(s)
- Dianzhong Zheng
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhaorui Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ling Sui
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yigang Xu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China
| | - Li Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China
| | - Xinyuan Qiao
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Wen Cui
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yanping Jiang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Han Zhou
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Lijie Tang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China.
| | - Yijing Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin 150030, China.
| |
Collapse
|
19
|
Nataraj BH, Ramesh C, Mallappa RH. Extractable surface proteins of indigenous probiotic strains confer anti-adhesion knack and protect against methicillin-resistant Staphylococcus aureus induced epithelial hyperpermeability in HT-29 cell line. Microb Pathog 2021; 158:104974. [PMID: 34015494 DOI: 10.1016/j.micpath.2021.104974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 11/28/2022]
Abstract
Probiotic intervention has been long believed to have beneficial effects on human health by curbing the intestinal colonization of pathogens. However, the application of live probiotics therapy may not be an ideal approach to circumvent the infections of superbug origin due to the risk of horizontal antibiotic resistance genes transfer. In this study, the anti-adhesion ability of extractable cell surface proteins from two indigenous potential probiotic strains (Lactiplantibacillus plantarum A5 and Limosilactobacillus fermentum Lf1) and two standard reference strains (Lactobacillus acidophilus NCFM and Lacticaseibacillus rhamnosus LGG) was evaluated against clinical isolates of Methicillin-Resistant Staphylococcus aureus (MRSA) on porcine gastric mucin and HT-29 cells. The surface proteins from the probiotic strains were extracted by treatment with 5 M lithium chloride. The surface protein quantification and SDS-PAGE profiling indicated that the yield and protein patterns were strain-specific. Surface proteins significantly hampered the mucoadhesion of MRSA isolates via protective, competitive, and displacement. Similarly, the treatment with surface proteins probiotic strains displayed anti-adhesion against MRSA isolates on HT-29 cells without affecting the viability of the cell line. Surface proteins treatment to the confluent monolayer of HT-29 cells maintained the epithelial integrity; however, MRSA isolates (109 cells/mL) showed considerable alteration in the epithelial integrity by exacerbating the FITC-dextran transflux. Contrarily, the co-treatment with surface proteins with MRSA isolates significantly lowered the FITC-dextran transflux across the differentiated HT-29 monolayer. Overall, the findings of this study suggest that probiotic-derived surface proteins could be the novel biotherapeutics to combat the MRSA colonization and their concomitant intestinal infections.
Collapse
Affiliation(s)
| | - Chette Ramesh
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Rashmi Hogarehalli Mallappa
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India.
| |
Collapse
|
20
|
Drolia R, Amalaradjou MAR, Ryan V, Tenguria S, Liu D, Bai X, Xu L, Singh AK, Cox AD, Bernal-Crespo V, Schaber JA, Applegate BM, Vemulapalli R, Bhunia AK. Receptor-targeted engineered probiotics mitigate lethal Listeria infection. Nat Commun 2020; 11:6344. [PMID: 33311493 PMCID: PMC7732855 DOI: 10.1038/s41467-020-20200-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022] Open
Abstract
Probiotic bacteria reduce the intestinal colonization of pathogens. Yet, their use in preventing fatal infection caused by foodborne Listeria monocytogenes (Lm), is inconsistent. Here, we bioengineered Lactobacillus probiotics (BLP) to express the Listeria adhesion protein (LAP) from a non-pathogenic Listeria (L. innocua) and a pathogenic Listeria (Lm) on the surface of Lactobacillus casei. The BLP strains colonize the intestine, reduce Lm mucosal colonization and systemic dissemination, and protect mice from lethal infection. The BLP competitively excludes Lm by occupying the surface presented LAP receptor, heat shock protein 60 and ameliorates the Lm-induced intestinal barrier dysfunction by blocking the nuclear factor-κB and myosin light chain kinase-mediated redistribution of the major epithelial junctional proteins. Additionally, the BLP increases intestinal immunomodulatory functions by recruiting FOXP3+T cells, CD11c+ dendritic cells and natural killer cells. Engineering a probiotic strain with an adhesion protein from a non-pathogenic bacterium provides a new paradigm to exclude pathogens and amplify their inherent health benefits.
Collapse
Affiliation(s)
- Rishi Drolia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Mary Anne Roshni Amalaradjou
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
- Department of Animal Science, University of Connecticut, Storrs, CT, USA
| | - Valerie Ryan
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Shivendra Tenguria
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Dongqi Liu
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Xingjian Bai
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Luping Xu
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Atul K Singh
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Abigail D Cox
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Victor Bernal-Crespo
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - James A Schaber
- Bindley Bioscience Research Center, Purdue University, West Lafayette, IN, USA
| | - Bruce M Applegate
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
- Purdue University Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, USA
| | - Ramesh Vemulapalli
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, USA
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA.
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA.
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA.
- Purdue University Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
21
|
Tarnaud F, Gaucher F, do Carmo FLR, Illikoud N, Jardin J, Briard-Bion V, Guyomarc'h F, Gagnaire V, Jan G. Differential Adaptation of Propionibacterium freudenreichii CIRM-BIA129 to Cow's Milk Versus Soymilk Environments Modulates Its Stress Tolerance and Proteome. Front Microbiol 2020; 11:549027. [PMID: 33335514 PMCID: PMC7736159 DOI: 10.3389/fmicb.2020.549027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Propionibacterium freudenreichii is a beneficial bacterium that modulates the gut microbiota, motility and inflammation. It is traditionally consumed within various fermented dairy products. Changes to consumer habits in the context of food transition are, however, driving the demand for non-dairy fermented foods, resulting in a considerable development of plant-based fermented products that require greater scientific knowledge. Fermented soymilks, in particular, offer an alternative source of live probiotics. While the adaptation of lactic acid bacteria (LAB) to such vegetable substrates is well documented, little is known about that of propionibacteria. We therefore investigated the adaptation of Propionibacterium freudenreichii to soymilk by comparison to cow's milk. P. freudenreichii grew in cow's milk but not in soymilk, but it did grow in soymilk when co-cultured with the lactic acid bacterium Lactobacillus plantarum. When grown in soymilk ultrafiltrate (SUF, the aqueous phase of soymilk), P. freudenreichii cells appeared thinner and rectangular-shaped, while they were thicker and more rounded in cow's milk utltrafiltrate (MUF, the aqueous phase of cow milk). The amount of extractable surface proteins (SlpA, SlpB, SlpD, SlpE) was furthermore reduced in SUF, when compared to MUF. This included the SlpB protein, previously shown to modulate adhesion and immunomodulation in P. freudenreichii. Tolerance toward an acid and toward a bile salts challenge were enhanced in SUF. By contrast, tolerance toward an oxidative and a thermal challenge were enhanced in MUF. A whole-cell proteomic approach further identified differential expression of 35 proteins involved in amino acid transport and metabolism (including amino acid dehydrogenase, amino acid transporter), 32 proteins involved in carbohydrate transport and metabolism (including glycosyltransferase, PTS), indicating metabolic adaptation to the substrate. The culture medium also modulated the amount of stress proteins involved in stress remediation: GroEL, OpuCA, CysK, DnaJ, GrpE, in line with the modulation of stress tolerance. Changing the fermented substrate may thus significantly affect the fermentative and probiotic properties of dairy propionibacteria. This needs to be considered when developing new fermented functional foods.
Collapse
Affiliation(s)
| | - Floriane Gaucher
- INRAE, Institut Agro, STLO, Rennes, France
- Bioprox, Levallois-Perret, France
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Lactobacillus Cell Surface Proteins Involved in Interaction with Mucus and Extracellular Matrix Components. Curr Microbiol 2020; 77:3831-3841. [PMID: 33079206 PMCID: PMC7677277 DOI: 10.1007/s00284-020-02243-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/03/2020] [Indexed: 12/18/2022]
Abstract
The gut microbiota is a complex microbial ecosystem where bacteria, through mutual interactions, cooperate in maintaining of wellbeing and health. Lactobacilli are among the most important constituents of human and animal intestinal microbiota and include many probiotic strains. Their presence ensures protection from invasion of pathogens, as well as stimulation of the immune system and protection of the intestinal flora, often exerted through the ability to interact with mucus and extracellular matrix components. The main factors responsible for mediating adhesion of pathogens and commensals to the gut are cell surface proteins that recognize host targets, as mucus layer and extracellular matrix proteins. In the last years, several adhesins have been reported to be involved in lactobacilli–host interaction often miming the same mechanism used by pathogens.
Collapse
|
23
|
Strategies to display heterologous proteins on the cell surface of lactic acid bacteria using as anchor the C-terminal domain of Lactobacillus acidophilus SlpA. World J Microbiol Biotechnol 2020; 36:169. [DOI: 10.1007/s11274-020-02945-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/30/2020] [Indexed: 01/19/2023]
|
24
|
Mantziari A, Salminen S, Szajewska H, Malagón-Rojas JN. Postbiotics against Pathogens Commonly Involved in Pediatric Infectious Diseases. Microorganisms 2020; 8:E1510. [PMID: 33008065 PMCID: PMC7601467 DOI: 10.3390/microorganisms8101510] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
The Sustainable Development goals for 2020 included reducing all causes associated with infant and perinatal mortality in their priorities. The use of compounds with bioactive properties has been proposed as a therapeutic strategy due to their stimulating effect on the host's immune system. Additionally, biotherapeutic products such as postbiotics, tentatively defined as compounds produced during a fermentation process that support health and well-being, promote intestinal barrier integrity without posing considerable risks to children's health. Although this is a concept in development, there are increasing studies in the field of nutrition, chemistry, and health that aim to understand how postbiotics can help prevent different types of infections in priority populations such as minors under the age of five. The present review aims to describe the main mechanisms of action of postbiotics. In addition, it presents the available current evidence regarding the effects of postbiotics against pathogens commonly involved in pediatric infections. Postbiotics may constitute a safe alternative capable of modulating the cellular response and stimulating the host's humoral response.
Collapse
Affiliation(s)
- Anastasia Mantziari
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland;
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland;
| | - Hania Szajewska
- Department of Paediatrics at the Medical University of Warsaw, 02091 Warsaw, Poland;
| | - Jeadran Nevardo Malagón-Rojas
- Facultad de Medicina, Universidad El Bosque, 110121 Bogotá, Colombia;
- Instituto Nacional de Salud de Colombia, 111321 Bogotá, Colombia
| |
Collapse
|
25
|
Dubey V, Mishra AK, Ghosh AR. Cell adherence efficacy of probiotic Pediococcus pentosaceus GS4 (MTCC 12683) and demonstrable role of its surface layer protein (Slp). J Proteomics 2020; 226:103894. [PMID: 32652219 DOI: 10.1016/j.jprot.2020.103894] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 06/08/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022]
Abstract
The current study examined the cell adherence property of probiotic Pediococcus pentosaceus GS4 (MTCC12683) with the characterization and functionality in adherence of its surface layer protein (GS4-Slp). The Slp of P. pentosaceus GS4 was extracted purified and detected using SDS-PAGE (98 kDa) and size exclusion chromatography. The cell adherence property of probiotic GS4 (Slp+/Slp-) was evaluated on buccal cells and HCT-116. Purified Slp was found neutralized with raised anti-Slp showing reduced adherence to HCT-116 as evident from SEM analysis. The structure of GS4-Slp was determined by MALDI-TOF analysis, CD analysis, atomic force microscopy (AFM), and FT-IR spectrometry. In Silico approach revealed its indirect similarity with cell membrane protein of Helicobacter pylori. Results thus reveal that GS4 has the potential of the production of 98 kDa Slp which facilitates the cell adherence property. This added probiotic attribute will enhance the probiotic potentials of P. pentosaceus GS4 to use it biotechnologically. SIGNIFICANCE: Probiotic Pediococcus pentosaceus GS4 facilitates demonstrable colonization by the elaboration of Slp. This property imparts a value to the strain and claims to be more useful biotechnologically.
Collapse
Affiliation(s)
- Vinay Dubey
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Alok Kumar Mishra
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Asit Ranjan Ghosh
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
26
|
Regulating colonic dendritic cells by commensal glycosylated large surface layer protein A to sustain gut homeostasis against pathogenic inflammation. Mucosal Immunol 2020; 13:34-46. [PMID: 31619761 PMCID: PMC6917853 DOI: 10.1038/s41385-019-0210-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/30/2019] [Accepted: 09/23/2019] [Indexed: 02/04/2023]
Abstract
Microbial interaction with the host through sensing receptors, including SIGNR1, sustains intestinal homeostasis against pathogenic inflammation. The newly discovered commensal Propionibacterium strain, P. UF1, regulates the intestinal immunity against pathogen challenge. However, the molecular events driving intestinal phagocytic cell response, including colonic dendritic cells (DCs), by this bacterium are still elusive. Here, we demonstrate that the glycosylation of bacterial large surface layer protein A (LspA) by protein O-mannosyltransferase 1 (Pmt1) regulates the interaction with SIGNR1, resulting in the control of DC transcriptomic and metabolomic machineries. Programmed DCs promote protective T cell response to intestinal Listeria infection and resist chemically induced colitis in mice. Thus, our findings may highlight a novel molecular mechanism by which commensal surface glycosylation interacting with SIGNR1 directs the intestinal homeostasis to potentially protect the host against proinflammatory signals inducing colonic tissue damage.
Collapse
|
27
|
Probiotic Propionibacterium freudenreichii requires SlpB protein to mitigate mucositis induced by chemotherapy. Oncotarget 2019; 10:7198-7219. [PMID: 31921383 PMCID: PMC6944450 DOI: 10.18632/oncotarget.27319] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Propionibacterium freudenreichii CIRM-BIA 129 (P. freudenreichii wild type, WT) is a probiotic bacterium, which exerts immunomodulatory effects. This strain possesses extractable surface proteins, including SlpB, which are involved in anti-inflammatory effect and in adhesion to epithelial cells. We decided to investigate the impact of slpB gene mutation on immunomodulation in vitro and in vivo. In an in vitro assay, P. freudenreichii WT reduced expression of IL-8 (p<0.0001) and TNF-α (p<0.0001) cytokines in LPS-stimulated HT-29 cells. P. freudenreichii ΔslpB, lacking the SlpB protein, failed to do so. Subsequently, both strains were investigated in vivo in a 5-FU-induced mucositis mice model. Mucositis is a common side effect of cytotoxic chemotherapy with 5-FU, characterized by mucosal injury, inflammation, diarrhea, and weight loss. The WT strain prevented weight loss, reduced inflammation and consequently histopathological scores. Furthermore, it regulated key markers, including Claudin-1 (cld1, p<0.0005) and IL-17a (Il17a, p<0.0001) genes, as well as IL-12 (p<0.0001) and IL-1β (p<0.0429) cytokines levels. Mutant strain displayed opposite regulatory effect on cld1 expression and on IL-12 levels. This work emphasizes the importance of SlpB in P. freudenreichii ability to reduce mucositis inflammation. It opens perspectives for the development of probiotic products to decrease side effects of chemotherapy using GRAS bacteria with immunomodulatory surface protein properties.
Collapse
|
28
|
Aggregation, adhesion and efficacy studies of probiotic candidate Pediococcus acidilactici NCDC 252: a strain of dairy origin. World J Microbiol Biotechnol 2019; 36:10. [DOI: 10.1007/s11274-019-2785-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 12/13/2019] [Indexed: 11/26/2022]
|
29
|
Fina Martin J, Palomino MM, Cutine AM, Modenutti CP, Fernández Do Porto DA, Allievi MC, Zanini SH, Mariño KV, Barquero AA, Ruzal SM. Exploring lectin-like activity of the S-layer protein of Lactobacillus acidophilus ATCC 4356. Appl Microbiol Biotechnol 2019; 103:4839-4857. [PMID: 31053916 DOI: 10.1007/s00253-019-09795-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/22/2019] [Accepted: 03/23/2019] [Indexed: 02/06/2023]
Abstract
The surface layer (S-layer) protein of Lactobacillus acidophilus is a crystalline array of self-assembling, proteinaceous subunits non-covalently bound to the outmost bacterial cell wall envelope and is involved in the adherence of bacteria to host cells. We have previously described that the S-layer protein of L. acidophilus possesses anti-viral and anti-bacterial properties. In this work, we extracted and purified S-layer proteins from L. acidophilus ATCC 4356 cells to study their interaction with cell wall components from prokaryotic (i.e., peptidoglycan and lipoteichoic acids) and eukaryotic origin (i.e., mucin and chitin), as well as with viruses, bacteria, yeast, and blood cells. Using chimeric S-layer fused to green fluorescent protein (GFP) from different parts of the protein, we analyzed their binding capacity. Our results show that the C-terminal part of the S-layer protein presents lectin-like activity, interacting with different glycoepitopes. We further demonstrate that lipoteichoic acid (LTA) serves as an anchor for the S-layer protein. Finally, a structure for the C-terminal part of S-layer and possible binding sites were predicted by a homology-based model.
Collapse
Affiliation(s)
- Joaquina Fina Martin
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Cdad. Universitaria, Pabellón II, 4 piso, Lab QB40, C1428EGA, CABA, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria Mercedes Palomino
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Cdad. Universitaria, Pabellón II, 4 piso, Lab QB40, C1428EGA, CABA, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Anabella M Cutine
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Carlos P Modenutti
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Cdad. Universitaria, Pabellón II, 4 piso, Lab QB40, C1428EGA, CABA, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dario A Fernández Do Porto
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Cdad. Universitaria, Pabellón II, 4 piso, Lab QB40, C1428EGA, CABA, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Instituto de Cálculo, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariana C Allievi
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Cdad. Universitaria, Pabellón II, 4 piso, Lab QB40, C1428EGA, CABA, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sofia H Zanini
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Cdad. Universitaria, Pabellón II, 4 piso, Lab QB40, C1428EGA, CABA, Buenos Aires, Argentina
| | - Karina V Mariño
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Andrea A Barquero
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Cdad. Universitaria, Pabellón II, 4 piso, Lab QB40, C1428EGA, CABA, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sandra M Ruzal
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Cdad. Universitaria, Pabellón II, 4 piso, Lab QB40, C1428EGA, CABA, Buenos Aires, Argentina.
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
30
|
Siciliano RA, Lippolis R, Mazzeo MF. Proteomics for the Investigation of Surface-Exposed Proteins in Probiotics. Front Nutr 2019; 6:52. [PMID: 31069232 PMCID: PMC6491629 DOI: 10.3389/fnut.2019.00052] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/05/2019] [Indexed: 01/08/2023] Open
Abstract
Probiotics are commensal microorganisms that are present in the intestinal tract and in many fermented foods and positively affect human health, promoting digestion and uptake of dietary nutrients, strengthening intestinal barrier function, modulating immune response, and enhancing antagonism toward pathogens. The proteosurfaceome, i.e., the complex set of proteins present on the bacterial surface, is directly involved as leading actor in the dynamic communication between bacteria and host. In the last decade, the biological relevance of surface-exposed proteins prompted research activities exploiting the potentiality of proteomics to define the complex network of proteins that are involved in the molecular mechanisms at the basis of the adaptation to gastrointestinal environment and the probiotic effects. These studies also took advantages of the recent technological improvements in proteomics, mass spectrometry and bioinformatics that triggered the development of ad hoc designed innovative strategies to characterize the bacterial proteosurfaceome. This mini-review is aimed at describing the key role of proteomics in depicting the cell wall protein architecture and the involvement of surface-exposed proteins in the intimate and dynamic molecular dialogue between probiotics and intestinal epithelial and immune cells.
Collapse
Affiliation(s)
- Rosa Anna Siciliano
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Rosa Lippolis
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council (CNR-IBIOM), Bari, Italy
| | | |
Collapse
|
31
|
Gaucher F, Bonnassie S, Rabah H, Marchand P, Blanc P, Jeantet R, Jan G. Review: Adaptation of Beneficial Propionibacteria, Lactobacilli, and Bifidobacteria Improves Tolerance Toward Technological and Digestive Stresses. Front Microbiol 2019; 10:841. [PMID: 31068918 PMCID: PMC6491719 DOI: 10.3389/fmicb.2019.00841] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 04/02/2019] [Indexed: 01/15/2023] Open
Abstract
This review deals with beneficial bacteria, with a focus on lactobacilli, propionibacteria, and bifidobacteria. As being recognized as beneficial bacteria, they are consumed as probiotics in various food products. Some may also be used as starters in food fermentation. In either case, these bacteria may be exposed to various environmental stresses during industrial production steps, including drying and storage, and during the digestion process. In accordance with their adaptation to harsh environmental conditions, they possess adaptation mechanisms, which can be induced by pretreatments. Adaptive mechanisms include accumulation of compatible solutes and of energy storage compounds, which can be largely modulated by the culture conditions. They also include the regulation of energy production pathways, as well as the modulation of the cell envelop, i.e., membrane, cell wall, surface layers, and exopolysaccharides. They finally lead to the overexpression of molecular chaperones and of stress-responsive proteases. Triggering these adaptive mechanisms can improve the resistance of beneficial bacteria toward technological and digestive stresses. This opens new perspectives for the improvement of industrial processes efficiency with regard to the survival of beneficial bacteria. However, this bibliographical survey evidenced that adaptive responses are strain-dependent, so that growth and adaptation should be optimized case-by-case.
Collapse
Affiliation(s)
- Floriane Gaucher
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
- Bioprox, Levallois-Perret, France
| | - Sylvie Bonnassie
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
- Science de la Vie et de la Terre, Université de Rennes 1, Rennes, France
| | - Houem Rabah
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
- Pôle Agronomique Ouest, Bba, Rennes, France
| | | | | | - Romain Jeantet
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
| | - Gwénaël Jan
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
| |
Collapse
|
32
|
Mathipa MG, Thantsha MS, Bhunia AK. Lactobacillus casei expressing Internalins A and B reduces Listeria monocytogenes interaction with Caco-2 cells in vitro. Microb Biotechnol 2019; 12:715-729. [PMID: 30989823 PMCID: PMC6559204 DOI: 10.1111/1751-7915.13407] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/28/2022] Open
Abstract
Listeria monocytogenes has been implicated in a number of outbreaks including the recent largest outbreak in South Africa. Current methods for prevention of foodborne L. monocytogenes infection are inadequate, thus raising a need for an alternative strategy. Probiotic bioengineering is considered a prevailing approach to enhance the efficacy of probiotics for targeted control of pathogens. Here, the ability of Lactobacillus casei expressing the L. monocytogenes invasion proteins Internalins A and B (inlAB) to prevent infection was investigated. The inlAB operon was cloned and surface‐expressed on L. casei resulting in a recombinant strain, LbcInlAB, and subsequently, its ability to inhibit adhesion, invasion and translocation of L. monocytogenes through enterocyte‐like Caco‐2 cells was examined. Cell surface expression of InlAB on the LbcInlAB was confirmed by Western blotting and immunofluorescence staining. The LbcInlAB strain showed significantly higher (P < 0.0001) adherence, invasion and translocation of Caco‐2 cells than the wild‐type L. casei strain (LbcWT), as well as reduced L. monocytogenes adhesion, invasion and transcellular passage through the cell monolayer than LbcWT. Furthermore, pre‐exposure of Caco‐2 cells to LbcInlAB significantly reduced L. monocytogenes‐induced cell cytotoxicity and epithelial barrier dysfunction. These results suggest that InlAB‐expressing L. casei could be a potential practical approach for prevention of listeriosis.
Collapse
Affiliation(s)
- Moloko G Mathipa
- Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Pretoria, South Africa.,Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Mapitsi S Thantsha
- Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN, USA.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA.,Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
33
|
Gharaibeh RZ, Jobin C. Microbiota and cancer immunotherapy: in search of microbial signals. Gut 2019; 68:385-388. [PMID: 30530851 PMCID: PMC6580757 DOI: 10.1136/gutjnl-2018-317220] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 01/22/2023]
Affiliation(s)
- Raad Z Gharaibeh
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, Florida, USA,Department of Infectious Diseases and Immunology, University of Florida, Gainesville, Florida, USA,Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
34
|
Rabah H, Ferret-Bernard S, Huang S, Le Normand L, Cousin FJ, Gaucher F, Jeantet R, Boudry G, Jan G. The Cheese Matrix Modulates the Immunomodulatory Properties of Propionibacterium freudenreichii CIRM-BIA 129 in Healthy Piglets. Front Microbiol 2018; 9:2584. [PMID: 30420848 PMCID: PMC6215859 DOI: 10.3389/fmicb.2018.02584] [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: 08/03/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022] Open
Abstract
Propionibacterium freudenreichii is a beneficial bacterium, used as a cheese starter, which presents versatile probiotic properties. These properties are strain-dependent. We hypothesized they may also be delivery vehicle-dependent. In this study, we thus explored in healthy piglets how the cheese matrix affects the immunomodulatory properties of P. freudenreichii. During 2 weeks, three groups of weaned piglets consumed, respectively, P. freudenreichii as a liquid culture (PF-culture), P. freudenreichii under the form of a cheese (PF-cheese), or a control sterile cheese matrix (Cheese-matrix). The in vivo metabolic activity of P. freudenreichii was assessed by determining short chain fatty acids (SCFA) concentration and bifidobacteria population in feces. Whatever the delivery vehicle, P. freudenreichii was metabolically active in piglets' colon and enhanced both bifidobacteria and SCFA in feces. P. freudenreichii consumption decreased the secretion of TNFα and of IL-10 by peripheral blood mononuclear cells (PBMC). It did not alter IL-10, IFNγ, IL-17, and TNFα secretion in mesenteric lymph node immune cells (MLNC). PF-cheese enhanced significantly Treg phenotype, while PF-culture decreased significantly Th17 phenotype in PBMC and MLNC. Remarkably, only PF-cheese induced an increase of Th2 phenotype in PBMC and MLNC. Ex vivo stimulation of PBMC and MLNC by Lipopolysaccharides and Concanavalin A emphasized the difference in the immunomodulatory responses between PF-culture and PF-cheese group, as well as between PBMC and MLNC. This study shows the importance to consider the delivery vehicle for probiotic administration. It confirms the anti-inflammatory potential of P. freudenreichii. It opens new perspectives for the use propionibacteria-fermented products as preventive agents for inflammatory bowel diseases and intestinal infectious diseases.
Collapse
Affiliation(s)
- Houem Rabah
- STLO, INRA, Agrocampus Ouest, Rennes, France
- Pôle Agronomique Ouest, Rennes, France
| | | | - Song Huang
- STLO, INRA, Agrocampus Ouest, Rennes, France
| | - Laurence Le Normand
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, France
| | | | - Floriane Gaucher
- STLO, INRA, Agrocampus Ouest, Rennes, France
- Bioprox, Levallois-Perret, France
| | | | - Gaëlle Boudry
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, France
| | - Gwénaël Jan
- STLO, INRA, Agrocampus Ouest, Rennes, France
| |
Collapse
|
35
|
do Carmo FLR, Silva WM, Tavares GC, Ibraim IC, Cordeiro BF, Oliveira ER, Rabah H, Cauty C, da Silva SH, Canário Viana MV, Caetano ACB, Dos Santos RG, de Oliveira Carvalho RD, Jardin J, Pereira FL, Folador EL, Le Loir Y, Figueiredo HCP, Jan G, Azevedo V. Mutation of the Surface Layer Protein SlpB Has Pleiotropic Effects in the Probiotic Propionibacterium freudenreichii CIRM-BIA 129. Front Microbiol 2018; 9:1807. [PMID: 30174657 PMCID: PMC6107788 DOI: 10.3389/fmicb.2018.01807] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/18/2018] [Indexed: 01/19/2023] Open
Abstract
Propionibacterium freudenreichii is a beneficial Gram-positive bacterium, traditionally used as a cheese-ripening starter, and currently considered as an emerging probiotic. As an example, the P. freudenreichii CIRM-BIA 129 strain recently revealed promising immunomodulatory properties. Its consumption accordingly exerts healing effects in different animal models of colitis, suggesting a potent role in the context of inflammatory bowel diseases. This anti-inflammatory effect depends on surface layer proteins (SLPs). SLPs may be involved in key functions in probiotics, such as persistence within the gut, adhesion to host cells and mucus, or immunomodulation. Several SLPs coexist in P. freudenreichii CIRM-BIA 129 and mediate immunomodulation and adhesion. A mutant P. freudenreichii CIRM-BIA 129ΔslpB (CB129ΔslpB) strain was shown to exhibit decreased adhesion to intestinal epithelial cells. In the present study, we thoroughly analyzed the impact of this mutation on cellular properties. Firstly, we investigated alterations of surface properties in CB129ΔslpB. Surface extractable proteins, surface charges (ζ-potential) and surface hydrophobicity were affected by the mutation. Whole-cell proteomics, using high definition mass spectrometry, identified 1,288 quantifiable proteins in the wild-type strain, i.e., 53% of the theoretical proteome predicted according to P. freudenreichii CIRM-BIA 129 genome sequence. In the mutant strain, we detected 1,252 proteins, including 1,227 proteins in common with the wild-type strain. Comparative quantitative analysis revealed 97 proteins with significant differences between wild-type and mutant strains. These proteins are involved in various cellular process like signaling, metabolism, and DNA repair and replication. Finally, in silico analysis predicted that slpB gene is not part of an operon, thus not affecting the downstream genes after gene knockout. This study, in accordance with the various roles attributed in the literature to SLPs, revealed a pleiotropic effect of a single slpB mutation, in the probiotic P. freudenreichii. This suggests that SlpB may be at a central node of cellular processes and confirms that both nature and amount of SLPs, which are highly variable within the P. freudenreichii species, determine the probiotic abilities of strains.
Collapse
Affiliation(s)
- Fillipe L R do Carmo
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Institut National de la Recherche Agronomique, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France.,Agrocampus Ouest, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France
| | - Wanderson M Silva
- Instituto de Biotecnología, CICVyA - Instituto Nacional de Tecnología Agropecuaria, Hurlingham, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Guilherme C Tavares
- AQUACEN, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela C Ibraim
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Barbara F Cordeiro
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Emiliano R Oliveira
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Houem Rabah
- Institut National de la Recherche Agronomique, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France.,Agrocampus Ouest, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France
| | - Chantal Cauty
- Institut National de la Recherche Agronomique, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France
| | - Sara H da Silva
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcus V Canário Viana
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana C B Caetano
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Roselane G Dos Santos
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Julien Jardin
- Institut National de la Recherche Agronomique, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France
| | - Felipe L Pereira
- AQUACEN, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Edson L Folador
- Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Yves Le Loir
- Institut National de la Recherche Agronomique, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France.,Agrocampus Ouest, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France
| | - Henrique C P Figueiredo
- AQUACEN, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gwénaël Jan
- Institut National de la Recherche Agronomique, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France.,Agrocampus Ouest, UMR1253 Science & Technologie du Lait & de l'Oeuf, Rennes, France
| | - Vasco Azevedo
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
36
|
Whole fractions from probiotic bacteria induce in vitro Th17 responses in human peripheral blood mononuclear cells. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|