1
|
Perini HF, Pereira BDB, Sousa EG, Matos BS, Silva Prado LCD, Carvalho Azevedo VAD, Castro Soares SD, Silva MVD. Inhibitory effect of Bacillus velezensis 1273 strain cell-free supernatant against developing and preformed biofilms of Staphylococcus aureus and MRSA. Microb Pathog 2024; 197:107065. [PMID: 39447663 DOI: 10.1016/j.micpath.2024.107065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Microbial biofilms constitute a significant virulence factor and a substantial challenge in clinical environments due to their role in promoting antimicrobial resistance and their resilience to eradication efforts. Methicillin-resistant Staphylococcus aureus (MRSA) infections substantially increase healthcare costs, extend hospitalizations, and elevate morbidity and mortality rates. Therefore, developing innovative strategies to target and eliminate these bacteria and their biofilms effectively is imperative for robust epidemiological control. In this study, we evaluated the antibacterial and antibiofilm activities of cell-free supernatant (CFS) obtained from the Bacillus velezensis 1273 strain culture. Our data showed that CFS inhibited the growth of S. aureus ATCC 29213 and MRSA (clinical strain), with greater efficacy observed against S. aureus (1:16 dilution). Furthermore, CFS showed substantial potential in reducing biofilm formation in both strains (∼30 %) at subinhibitory concentrations. Additionally, the antibacterial activity against biofilm-formed cells showed that pure CFS treatment decreased the viability of S. aureus (60 %) and MRSA (45 %) sessile cells. We further demonstrated that CFS treatment induces the production of reactive oxygen species (ROS) and damages the membranes and cell walls of the pathogen cells. Genome analysis revealed the presence of genes encoding bacteriocins and secondary metabolites with antibacterial activity in the B. velezensis 1273 genome. These findings highlight the potential of probiotic bacterial metabolites as antibiofilm and anti-multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Hugo Felix Perini
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| | - Bianca de Barros Pereira
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Eduarda Guimarães Sousa
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Beatriz Sodré Matos
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | | | - Siomar de Castro Soares
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marcos Vinicius da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| |
Collapse
|
2
|
Wang P, Wang S, Wang D, Li Y, Yip RCS, Chen H. Postbiotics-peptidoglycan, lipoteichoic acid, exopolysaccharides, surface layer protein and pili proteins-Structure, activity in wounds and their delivery systems. Int J Biol Macromol 2024; 274:133195. [PMID: 38885869 DOI: 10.1016/j.ijbiomac.2024.133195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
Chronic wound healing is a pressing global public health concern. Abuse and drug resistance of antibiotics are the key problems in the treatment of chronic wounds at present. Postbiotics are a novel promising strategy. Previous studies have reported that postbiotics have a wide range of biological activities including antimicrobial, immunomodulatory, antioxidant and anti-inflammatory abilities. However, several aspects related to these postbiotic activities remain unexplored or poorly known. Therefore, this work aims to outline general aspects and emerging trends in the use of postbiotics for wound healing, such as the production, characterization, biological activities and delivery strategies of postbiotics. In this review, a comprehensive overview of the physiological activities and structures of postbiotic biomolecules that contribute to wound healing is provided, such as peptidoglycan, lipoteichoic acid, bacteriocins, exopolysaccharides, surface layer proteins, pili proteins, and secretory proteins (p40 and p75 proteins). Considering the presence of readily degradable components in postbiotics, potential natural polymer delivery materials and delivery systems are emphasized, followed by the potential applications and commercialization prospects of postbiotics. These findings suggest that the treatment of chronic wounds with postbiotic ingredients will help provide new insights into wound healing and better guidance for the development of postbiotic products.
Collapse
Affiliation(s)
- Pu Wang
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| | - Shuxin Wang
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| | - Donghui Wang
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| | - Yuanyuan Li
- Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Stocking Hall, 411 Tower Road, Ithaca, NY 14853, USA.
| | - Ryan Chak Sang Yip
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord St, Toronto, ON M5S 3G5, Canada.
| | - Hao Chen
- Marine College, Shandong University, No. 180 Wen Hua West Road, Gao Strict, Weihai 264209, China.
| |
Collapse
|
3
|
Nguyen MR, Ma E, Wyatt D, Knight KL, Osipo C. The effect of an exopolysaccharide probiotic molecule from Bacillus subtilis on breast cancer cells. Front Oncol 2023; 13:1292635. [PMID: 38074643 PMCID: PMC10702531 DOI: 10.3389/fonc.2023.1292635] [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: 09/11/2023] [Accepted: 10/27/2023] [Indexed: 02/12/2024] Open
Abstract
Introduction Many well-known risk factors for breast cancer are associated with dysbiosis (an aberrant microbiome). However, how bacterial products modulate cancer are poorly understood. In this study, we investigated the effect of an exopolysaccharide (EPS) produced by the commensal bacterium Bacillus subtilis on breast cancer phenotypes. Although B. subtilis is commonly included in probiotic preparations and its EPS protects against inflammatory diseases, it was virtually unknown whether B. subtilis-derived EPS affects cancer. Methods This work investigated effects of EPS on phenotypes of breast cancer cells as a cancer model. The phenotypes included proliferation, mammosphere formation, cell migration, and tumor growth in two immune compromised mouse models. RNA sequencing was performed on RNA from four breast cancer cells treated with PBS or EPS. IKKβ or STAT1 signaling was assessed using pharmacologic or RNAi-mediated knock down approaches. Results Short-term treatment with EPS inhibited proliferation of certain breast cancer cells (T47D, MDA-MB-468, HCC1428, MDA-MB-453) while having little effect on others (MCF-7, MDA-MB-231, BT549, ZR-75-30). EPS induced G1/G0 cell cycle arrest of T47D cells while increasing apoptosis of MDA-MB-468 cells. EPS also enhanced aggressive phenotypes in T47D cells including cell migration and cancer stem cell survival. Long-term treatment with EPS (months) led to resistance in vitro and promoted tumor growth in immunocompromised mice. RNA-sequence analysis showed that EPS increased expression of pro-inflammatory pathways including STAT1 and NF-κB. IKKβ and/or STAT1 signaling was necessary for EPS to modulate phenotypes of EPS sensitive breast cancer cells. Discussion These results demonstrate a multifaceted role for an EPS molecule secreted by the probiotic bacterium B. subtilis on breast cancer cell phenotypes. These results warrant future studies in immune competent mice and different cancer models to fully understand potential benefits and/or side effects of long-term use of probiotics.
Collapse
Affiliation(s)
- Mai R. Nguyen
- M.D./Ph.D. Program, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Emily Ma
- M.D./Ph.D. Program, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Integrated Cell Biology Program, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Debra Wyatt
- Department of Cancer Biology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Katherine L. Knight
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Clodia Osipo
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Department of Cancer Biology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
4
|
Zamora-Pineda J, Kalinina O, Sperling AI, Knight KL. Mechanism of TLR4-Mediated Anti-Inflammatory Response Induced by Exopolysaccharide from the Probiotic Bacillus subtilis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1232-1239. [PMID: 37672039 DOI: 10.4049/jimmunol.2200855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 08/02/2023] [Indexed: 09/07/2023]
Abstract
Intestinal inflammatory diseases affect millions of people worldwide, and one class of drugs showing promise toward treatment of several inflammatory diseases is probiotics. Numerous studies have been performed using probiotics to prevent and treat intestinal inflammatory diseases. Most of these studies used intact bacteria, and neither the active molecule nor the molecular mechanisms by which they affect immune responses are known. We have shown that the probiotic Bacillus subtilis is anti-inflammatory and can protect mice from acute colitis induced by the enteric pathogen Citrobacter rodentium. We identified and purified the active molecule, exopolysaccharide (EPS), and showed that it protects mice from C. rodentium-induced colitis by inducing anti-inflammatory M2 macrophages or inhibitory dendritic cells (DCs), both of which inhibit excessive T cell responses. We showed previously that EPS affects macrophages and DCs in a TLR4-dependent manner, and in the current study we asked how EPS induces these anti-inflammatory cells and how they function to inhibit T cells. By investigating the signaling downstream of TLR4 that leads to acquisition of inhibitory properties of macrophages and DCs, we found that EPS induces expression of the inhibitory molecule IDO in bone marrow-derived DCs, and that inhibition of T cell proliferation by IDO-expressing bone marrow-derived DCs utilizes the kynurenine/aryl hydrocarbon receptor circuit. Furthermore, unlike LPS, EPS does not induce inflammatory cytokines upon injection in vivo, directly demonstrating different outcomes induced by two different TLR4 agonists.
Collapse
Affiliation(s)
- Jesus Zamora-Pineda
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Olga Kalinina
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Anne I Sperling
- Pulmonary and Critical Care Division, Department of Medicine, University of Virginia, Charlottesville, VA
| | - Katherine L Knight
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| |
Collapse
|
5
|
Niu Y, Liu W, Fan X, Wen D, Wu D, Wang H, Liu Z, Li B. Beyond cellulose: pharmaceutical potential for bioactive plant polysaccharides in treating disease and gut dysbiosis. Front Microbiol 2023; 14:1183130. [PMID: 37293228 PMCID: PMC10244522 DOI: 10.3389/fmicb.2023.1183130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/04/2023] [Indexed: 06/10/2023] Open
Abstract
Polysaccharides derived from plants, algae, or fungi serve as the major components of some human diets. Polysaccharides have been shown to exhibit diverse biological activities in improving human health, and have also been proposed to function as potent modulators of gut microbiota composition, thus playing a bi-directional regulatory role in host health. Here, we review a variety of polysaccharide structures potentially linked to biological functions, and cover current research progress in characterizing their pharmaceutical effects in various disease models, including antioxidant, anticoagulant, anti-inflammatory, immunomodulatory, hypoglycemic, and antimicrobial activities. We also highlight the effects of polysaccharides on modulating gut microbiota via enrichment for beneficial taxa and suppression of potential pathogens, leading to increased microbial expression of carbohydrate-active enzymes and enhanced short chain fatty acid production. This review also discusses polysaccharide-mediated improvements in gut function by influencing interleukin and hormone secretion in host intestinal epithelial cells.
Collapse
Affiliation(s)
- Yuanlin Niu
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Wei Liu
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Xueni Fan
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Dongxu Wen
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Dan Wu
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Hongzhuang Wang
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Zhenjiang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Bin Li
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| |
Collapse
|
6
|
Pan N, Liu Y, Zhang H, Xu Y, Bao X, Sheng S, Liang Y, Liu B, Lyu Y, Li H, Ma F, Pan H, Wang X. Oral Vaccination with Engineered Probiotic Limosilactobacillus reuteri Has Protective Effects against Localized and Systemic Staphylococcus aureus Infection. Microbiol Spectr 2023; 11:e0367322. [PMID: 36723073 PMCID: PMC10100842 DOI: 10.1128/spectrum.03673-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/14/2023] [Indexed: 02/02/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive bacterium responsible for most hospital-acquired (nosocomial) and community-acquired infections worldwide. The only therapeutic strategy against S. aureus-induced infections, to date, is antibiotic treatment. A protective vaccine is urgently needed in view of the emergence of antibiotic-resistant strains associated with high-mortality cases; however, no such vaccine is currently available. In our previous work, the feasibility of implementing a Lactobacillus delivery system for development of S. aureus oral vaccine was first discussed. Here, we describe systematic screening and evaluation of protective effects of engineered Lactobacillus against S. aureus infection in terms of different delivery vehicle strains and S. aureus antigens and in localized and systemic infection models. Limosilactobacillus reuteri WXD171 was selected as the delivery vehicle strain based on its tolerance of the gastrointestinal environment, adhesion ability, and antimicrobial activities in vitro and in vivo. We designed, constructed, and evaluated engineered L. reuteri strains expressing various S. aureus antigens. Among these, engineered L. reuteri WXD171-IsdB displayed effective protection against S. aureus-induced localized infection (pneumonia and skin infection) and, furthermore, a substantial survival benefit in systemic infection (sepsis). WXD171-IsdB induced mucosal responses in gut-associated lymphoid tissues, as evidenced by increased production of secretory IgA and interleukin 17A (IL-17A) and proliferation of lymphocytes derived from Peyer's patches. The probiotic L. reuteri-based oral vaccine appears to have strong potential as a prophylactic agent against S. aureus infections. Our findings regarding utilization of Lactobacillus delivery system in S. aureus vaccine development support the usefulness of this live vaccination strategy and its potential application in next-generation vaccine development. IMPORTANCE We systematically screened and evaluated protective effects of engineered Lactobacillus against S. aureus infection in terms of differing delivery vehicle strains and S. aureus antigens and in localized and systemic infection models. Engineered L. reuteri was developed and showed strong protective effects against both types of S. aureus-induced infection. Our findings regarding the utilization of a Lactobacillus delivery system in S. aureus vaccine development support the usefulness of this live vaccination strategy and its potential application in next-generation vaccine development.
Collapse
Affiliation(s)
- Na Pan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yang Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Haochi Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Ying Xu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Xuemei Bao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Shouxin Sheng
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yanchen Liang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Bohui Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yueqing Lyu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Haotian Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Fangfei Ma
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Haiting Pan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
- Basic Medical College, Inner Mongolia Medical University, Hohhot, China
| | - Xiao Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
7
|
Bacillus subtilis Provides Long-Term Protection in a Murine Model of Allergic Lung Disease by Influencing Bacterial Composition. ALLERGIES 2022. [DOI: 10.3390/allergies3010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Probiotics are an attractive target for reducing the incidence of allergic disease. Bacillus subtilis is a gut-associated probiotic bacteria that can suppress allergic lung disease; however, it is not clear for how long this protection lasts. We exposed C57Bl/6 mice to B. subtilis via oral gavage and challenged them with intranasal house-dust mite for up to 8 weeks. We found that B. subtilis treatment was able to provide protection from eosinophil infiltration of the airways for 3 weeks. This loss of protection correlated with an increase in the eosinophil chemoattractant CCL24. Additionally, we demonstrate that B. subtilis treatment altered the bacterial composition by increasing the phylum Bacteroidetes and Verrucomicorbiota. The phylum Verrucomicorbiota was reduced in B. subtilis-treated mice at 8 weeks when protection was lost. These results support B. subtilis as a prophylactic for preventing the production of allergic lung disease and highlights that protection can last up to 3 weeks. This work also expands our understanding of how B. subtilis mediates protection and that in addition to modifying the immune system it is also altering the host microbiota.
Collapse
|
8
|
Probiotics Bring New Hope for Atherosclerosis Prevention and Treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3900835. [PMID: 36193065 PMCID: PMC9526629 DOI: 10.1155/2022/3900835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/12/2022] [Accepted: 09/17/2022] [Indexed: 11/26/2022]
Abstract
Cardiovascular disease is the leading cause of human mortality and morbidity worldwide. Atherosclerosis (AS) is the underlying pathological responsible in most acute and severe cardiovascular diseases including myocardial infarction and stroke. However, current drugs applied to the treatment of AS are not clinically effective, and there is a large residual risk of cardiovascular disease and multiple side effects. Increasing evidence supports a close relationship between microorganisms and the incidence of AS. Recent data have shown that probiotics can improve multiple key factors involved in the development and progression of AS, including cholesterol metabolism imbalance, endothelial dysfunction, proinflammatory factor production, macrophage polarization, intestinal flora disturbance, and infection with pathogenic microorganisms, and therefore probiotics have attracted great interest as a novel potential “medicine”. This review is aimed at summarizing the effects of probiotics on various influencing factors, and providing valuable insights in the search for early prevention and potential therapeutic strategies for AS.
Collapse
|
9
|
Exopolysaccharides of Bacillus amyloliquefaciens Amy-1 Mitigate Inflammation by Inhibiting ERK1/2 and NF-κB Pathways and Activating p38/Nrf2 Pathway. Int J Mol Sci 2022; 23:ijms231810237. [PMID: 36142159 PMCID: PMC9499622 DOI: 10.3390/ijms231810237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/28/2022] Open
Abstract
Bacillus amyloliquefaciens is a probiotic for animals. Evidence suggests that diets supplemented with B. amyloliquefaciens can reduce inflammation; however, the underlying mechanism is unclear and requires further exploration. The exopolysaccharides of B. amyloliquefaciens amy-1 displayed hypoglycemic activity previously, suggesting that they are bioactive molecules. In addition, they counteracted the effect of lipopolysaccharide (LPS) on inducing cellular insulin resistance in exploratory tests. Therefore, this study aimed to explore the anti-inflammatory effect and molecular mechanisms of the exopolysaccharide preparation of amy-1 (EPS). Consequently, EPS reduced the expression of proinflammatory factors, the phagocytic activity and oxidative stress of LPS-stimulated THP-1 cells. In animal tests, EPS effectively ameliorated ear inflammation of mice. These data suggested that EPS possess anti-inflammatory activity. A mechanism study revealed that EPS inhibited the nuclear factor-κB pathway, activated the mitogen-activated protein kinase (MAPK) p38, and prohibited the extracellular signal-regulated kinase 1/2, but had no effect on the c-Jun-N-terminal kinase 2 (JNK). EPS also activated the anti-oxidative nuclear factor erythroid 2–related factor 2 (Nrf2) pathway. Evidence suggested that p38, but not JNK, was involved in activating the Nrf2 pathway. Together, these mechanisms reduced the severity of inflammation. These findings support the proposal that exopolysaccharides may play important roles in the anti-inflammatory functions of probiotics.
Collapse
|
10
|
Cruz KCP, Enekegho LO, Stuart DT. Bioengineered Probiotics: Synthetic Biology Can Provide Live Cell Therapeutics for the Treatment of Foodborne Diseases. Front Bioeng Biotechnol 2022; 10:890479. [PMID: 35656199 PMCID: PMC9152101 DOI: 10.3389/fbioe.2022.890479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/29/2022] [Indexed: 11/15/2022] Open
Abstract
The rising prevalence of antibiotic resistant microbial pathogens presents an ominous health and economic challenge to modern society. The discovery and large-scale development of antibiotic drugs in previous decades was transformational, providing cheap, effective treatment for what would previously have been a lethal infection. As microbial strains resistant to many or even all antibiotic drug treatments have evolved, there is an urgent need for new drugs or antimicrobial treatments to control these pathogens. The ability to sequence and mine the genomes of an increasing number of microbial strains from previously unexplored environments has the potential to identify new natural product antibiotic biosynthesis pathways. This coupled with the power of synthetic biology to generate new production chassis, biosensors and “weaponized” live cell therapeutics may provide new means to combat the rapidly evolving threat of drug resistant microbial pathogens. This review focuses on the application of synthetic biology to construct probiotic strains that have been endowed with functionalities allowing them to identify, compete with and in some cases kill microbial pathogens as well as stimulate host immunity. Weaponized probiotics may have the greatest potential for use against pathogens that infect the gastrointestinal tract: Vibrio cholerae, Staphylococcus aureus, Clostridium perfringens and Clostridioides difficile. The potential benefits of engineered probiotics are highlighted along with the challenges that must still be met before these intriguing and exciting new therapeutic tools can be widely deployed.
Collapse
|
11
|
Probiotic Molecules That Inhibit Inflammatory Diseases. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Consumption of probiotics for health purposes has increased vastly in the past few decades, and yet the scientific evidence to support health benefits from probiotics is only beginning to emerge. As more probiotics are studied, we are beginning to understand the mechanisms of action by which they benefit human health, as well as to identify the bacterial molecules responsible for these benefits. A new era of therapeutics is on the horizon in which purified molecules from probiotics will be used to prevent and treat diseases. In this review, we summarize the active molecules from probiotic bacteria that have been shown to affect innate and adaptive immunity and have health benefits in experimental settings. We focus particularly on the cellular and molecular mechanisms of the probiotic Bacillus subtilis and its active molecule, exopolysaccharide (ESPBs).
Collapse
|
12
|
Arnaouteli S, Bamford NC, Stanley-Wall NR, Kovács ÁT. Bacillus subtilis biofilm formation and social interactions. Nat Rev Microbiol 2021; 19:600-614. [PMID: 33824496 DOI: 10.1038/s41579-021-00540-9] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2021] [Indexed: 02/03/2023]
Abstract
Biofilm formation is a process in which microbial cells aggregate to form collectives that are embedded in a self-produced extracellular matrix. Bacillus subtilis is a Gram-positive bacterium that is used to dissect the mechanisms controlling matrix production and the subsequent transition from a motile planktonic cell state to a sessile biofilm state. The collective nature of life in a biofilm allows emergent properties to manifest, and B. subtilis biofilms are linked with novel industrial uses as well as probiotic and biocontrol processes. In this Review, we outline the molecular details of the biofilm matrix and the regulatory pathways and external factors that control its production. We explore the beneficial outcomes associated with biofilms. Finally, we highlight major advances in our understanding of concepts of microbial evolution and community behaviour that have resulted from studies of the innate heterogeneity of biofilms.
Collapse
Affiliation(s)
- Sofia Arnaouteli
- Division of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Natalie C Bamford
- Division of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Nicola R Stanley-Wall
- Division of Molecular Microbiology, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Ákos T Kovács
- Bacterial Interactions and Evolution Group, DTU Bioengineering, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
13
|
Antibiotic Resistance Crisis: An Update on Antagonistic Interactions between Probiotics and Methicillin-Resistant Staphylococcus aureus (MRSA). Curr Microbiol 2021; 78:2194-2211. [PMID: 33881575 DOI: 10.1007/s00284-021-02442-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
Antimicrobial resistance (AMR) havoc is a global multifaceted crisis endowing a significant challenge for the successful eradication of devastating pathogens. Methicillin-Resistant Staphylococcus aureus (MRSA) is an enduring superbug involved in causing devastating infections. Although MRSA is a frequent colonizer of human skin, wound, and anterior nares, the intestinal colonization of MRSA has greatly increased the risk of inducing MRSA-associated colitis besides creating a conducive environment for horizontal transfer of resistant genes to commensal microbes. On the other hand, staphylococcal resistance to last-resort antibiotics has urged the development of novel antimicrobial agents for the effective decolonization of MRSA. In response, probiotics and their metabolites (postbiotics) have been proposed as the adjunct therapeutic avenues. Probiotics exhibit a multitude of anti-MRSA actions (anti-bacterial, anti-biofilm, anti-virulence, anti-drug resistance, co-aggregation, and anti-quorum sensing) through the production of numerous antagonistic compounds such as organic acids, hydrogen peroxide, low molecular weight compounds, biosurfactants, bacteriocins, and bacteriocins like inhibitory substances. Besides, probiotics stabilize the epithelial barrier function and positively modulate the host immune system via regulating various signal transduction mechanisms. Preclinical and human intervention studies have suggested that probiotics outcompete with MRSA by exhibiting anti-colonization mechanisms via protective, competitive, and displacement mode. In this review, we aim to highlight the dynamics of MRSA associated virulence and drug resistance properties, and how probiotics antagonize MRSA through various mechanism of action.
Collapse
|
14
|
Kalinina O, Talley S, Zamora-Pineda J, Paik W, Campbell EM, Knight KL. Amelioration of Graft-versus-Host Disease by Exopolysaccharide from a Commensal Bacterium. THE JOURNAL OF IMMUNOLOGY 2021; 206:2101-2108. [PMID: 33846225 DOI: 10.4049/jimmunol.2000780] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 03/02/2021] [Indexed: 01/07/2023]
Abstract
Acute graft-versus-host disease (aGvHD) is a severe, often lethal, complication of hematopoietic stem cell transplantation, and although prophylactic regimens are given as standard pretransplantation therapy, up to 60% of these patients develop aGvHD, and require additional immunosuppressive intervention. We treated mice with a purified probiotic molecule, exopolysaccharide (EPS) from Bacillus subtilis, shortly before and after induction of aGvHD and found that, whereas only 10% of control mice survived to day 80, 70% of EPS-treated mice survived to 80 d. EPS treatment of donor-only mice resulted in ∼60% survival. Using a biosensor mouse model to assess inflammation in live mice during aGvHD, we found that EPS prevented the activation of alloreactive donor T cells. In vitro, EPS did not affect T cells directly but, instead, induced bone marrow-derived dendritic cells (BMDCs) that displayed characteristics of inhibitory dendritic cells (DCs). Development of these BMDCs required TLR4 signaling through both MyD88 and TRIF pathways. Using BMDCs derived from IDO knockout mice, we showed that T cell inhibition by EPS-treated BMDCs was mediated through the suppressive effects of IDO. These studies describe a bacterial molecule that modulates immune responses by inducing inhibitory DCs in a TLR4-dependent manner, and these cells have the capacity to inhibit T cell activation through IDO. We suggest that EPS or EPS-treated DCs can serve as novel agents for preventing aGvHD.
Collapse
Affiliation(s)
- Olga Kalinina
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Sarah Talley
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Jesus Zamora-Pineda
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Wonbeom Paik
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Edward M Campbell
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Katherine L Knight
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| |
Collapse
|
15
|
Deusenbery C, Wang Y, Shukla A. Recent Innovations in Bacterial Infection Detection and Treatment. ACS Infect Dis 2021; 7:695-720. [PMID: 33733747 DOI: 10.1021/acsinfecdis.0c00890] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bacterial infections are a major threat to human health, exacerbated by increasing antibiotic resistance. These infections can result in tremendous morbidity and mortality, emphasizing the need to identify and treat pathogenic bacteria quickly and effectively. Recent developments in detection methods have focused on electrochemical, optical, and mass-based biosensors. Advances in these systems include implementing multifunctional materials, microfluidic sampling, and portable data-processing to improve sensitivity, specificity, and ease of operation. Concurrently, advances in antibacterial treatment have largely focused on targeted and responsive delivery for both antibiotics and antibiotic alternatives. Antibiotic alternatives described here include repurposed drugs, antimicrobial peptides and polymers, nucleic acids, small molecules, living systems, and bacteriophages. Finally, closed-loop therapies are combining advances in the fields of both detection and treatment. This review provides a comprehensive summary of the current trends in detection and treatment systems for bacterial infections.
Collapse
Affiliation(s)
- Carly Deusenbery
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, Rhode Island 02912, United States
| | - Yingying Wang
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Anita Shukla
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
16
|
Raheem A, Liang L, Zhang G, Cui S. Modulatory Effects of Probiotics During Pathogenic Infections With Emphasis on Immune Regulation. Front Immunol 2021; 12:616713. [PMID: 33897683 PMCID: PMC8060567 DOI: 10.3389/fimmu.2021.616713] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
In order to inhibit pathogenic complications and to enhance animal and poultry growth, antibiotics have been extensively used for many years. Antibiotics applications not only affect target pathogens but also intestinal beneficially microbes, inducing long-lasting changes in intestinal microbiota associated with diseases. The application of antibiotics also has many other side effects like, intestinal barrier dysfunction, antibiotics residues in foodstuffs, nephropathy, allergy, bone marrow toxicity, mutagenicity, reproductive disorders, hepatotoxicity carcinogenicity, and antibiotic-resistant bacteria, which greatly compromise the efficacy of antibiotics. Thus, the development of new antibiotics is necessary, while the search for antibiotic alternatives continues. Probiotics are considered the ideal antibiotic substitute; in recent years, probiotic research concerning their application during pathogenic infections in humans, aquaculture, poultry, and livestock industry, with emphasis on modulating the immune system of the host, has been attracting considerable interest. Hence, the adverse effects of antibiotics and remedial effects of probiotics during infectious diseases have become central points of focus among researchers. Probiotics are live microorganisms, and when given in adequate quantities, confer good health effects to the host through different mechanisms. Among them, the regulation of host immune response during pathogenic infections is one of the most important mechanisms. A number of studies have investigated different aspects of probiotics. In this review, we mainly summarize recent discoveries and discuss two important aspects: (1) the application of probiotics during pathogenic infections; and (2) their modulatory effects on the immune response of the host during infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Abdul Raheem
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Lin Liang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Guangzhi Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Shangjin Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| |
Collapse
|
17
|
Liu Y, Sheng Y, Pan Q, Xue Y, Yu L, Tian F, Zhao J, Zhang H, Zhai Q, Chen W. Identification of the key physiological characteristics of Lactobacillus plantarum strains for ulcerative colitis alleviation. Food Funct 2020; 11:1279-1291. [PMID: 31984399 DOI: 10.1039/c9fo02935d] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lactobacillus plantarum is a probiotic that is widely used to prevent ulcerative colitis (UC). However, the effects of this species are strain-specific. We believe that the physiological characteristics of L. plantarum strains may affect their UC-alleviating function. Therefore, this study investigated the relationship between the alleviating effect of L. plantarum strains on UC and their physiological characteristics in vitro. The physiological characteristics of 14 L. plantarum strains were assayed in vitro, including gastrointestinal transit tolerance, oligosaccharide fermentation, HT-29 cell adhesion, generation time, exopolysaccharide production, acetic acid production, and conjugated linoleic acid (CLA) synthesis. To create animal models, colitis was established in C57BL/6 mice by adding 3.5% dextran sulfate sodium to drinking water for 7 days. L. plantarum strains with significantly different physiological characteristics were orally administered to the mice at a dose of 3 × 109 CFU. The results indicated that among the tested L. plantarum strains, L. plantarum N13 and L. plantarum CCFM8610 significantly alleviated colitis in the mice, as observed from the restoration of the body weight and disease activity index (DAI) score, recovery of the gut microbiota composition, reduced expression of pro-inflammatory cytokines, and significantly inhibited expression of p65. Correlation analysis indicated that four of the measured physiological characteristics (gastrointestinal transit tolerance, HT-29 cell adhesion, generation time, and CLA synthesis) were related to the UC-alleviating effects to different degrees. The strongest correlation was observed between the CLA synthesis ability and UC-alleviating effects (with Pearson correlation coefficients for IL-1β, IL-6, IL-17F, TNF-α, myeloperoxidase, and the DAI all below -0.95). The ability to synthesize CLA may be the key physiological characteristic of L. plantarum in UC alleviation. Our findings may contribute to the rapid screening of lactic acid bacterial strains with UC-alleviating effects.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yingyue Sheng
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province, China
| | - Qiqi Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China and International Joint Research Laboratory for Probiotics at Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China and Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, China and (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China and International Joint Research Laboratory for Probiotics at Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China and Beijing Innovation Center of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| |
Collapse
|
18
|
Regulating the Polarization of Macrophages: A Promising Approach to Vascular Dermatosis. J Immunol Res 2020; 2020:8148272. [PMID: 32775470 PMCID: PMC7407038 DOI: 10.1155/2020/8148272] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/04/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Macrophages, a kind of innate immune cells, derive from monocytes in circulation and play a crucial role in the innate and adaptive immunity. Under the stimulation of the signals from local microenvironment, macrophages generally tend to differentiate into two main functional phenotypes depending on their high plasticity and heterogeneity, namely, classically activated macrophage (M1) and alternatively activated macrophage (M2). This phenomenon is often called macrophage polarization. In pathological conditions, chronic persistent inflammation could induce an aberrant response of macrophage and cause a shift in their phenotypes. Moreover, this shift would result in the alteration of macrophage polarization in some vascular dermatoses; e.g., an increase in proinflammatory M1 emerges from Behcet's disease (BD), psoriasis, and systemic lupus erythematosus (SLE), whereas an enhancement in anti-inflammatory M2 appears in infantile hemangioma (IH). Individual polarized phenotypes and their complicated cytokine networks may crucially mediate in the pathological processes of some vascular diseases (vascular dermatosis in particular) by activation of T cell subsets (such as Th1, Th2, Th17, and Treg cells), deterioration of oxidative stress damage, and induction of angiogenesis, but the specific mechanism remains ambiguous. Therefore, in this review, we discuss the possible role of macrophage polarization in the pathological processes of vascular skin diseases. In addition, it is proposed that regulation of macrophage polarization may become a potential strategy for controlling these disorders.
Collapse
|
19
|
Song Y, Sun M, Feng L, Liang X, Song X, Mu G, Tuo Y, Jiang S, Qian F. Antibiofilm Activity of Lactobacillus plantarum 12 Exopolysaccharides against Shigella flexneri. Appl Environ Microbiol 2020; 86:e00694-20. [PMID: 32444475 PMCID: PMC7376565 DOI: 10.1128/aem.00694-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
In developing countries, Shigella flexneri is the most common enteric pathogen causing bacillary dysentery. Biofilm formation by S. flexneri can cause the emergence of antibiotic-resistant strains, which poses serious threats to food safety and human health. In this study, the effects of Lactobacillus plantarum 12 exopolysaccharides (L-EPSs) and S. flexneri exopolysaccharides (S-EPSs) on S. flexneri CMCC51574 biofilm formation were investigated. The results showed that L-EPS could decrease polysaccharide production in the extracellular polymeric matrix of S. flexneri and inhibit biofilm formation by S. flexneri L-EPS could decrease the minimum biofilm elimination concentration (MBEC) of antibiotics against S. flexneri biofilm and inhibit S. flexneri adhesion to and invasion into HT-29 cell monolayers, which might be ascribed to S. flexneri biofilm disturbance by L-EPS. In contrast, S-EPS exhibited the opposite effects compared to L-EPS. The monosaccharide composition analysis showed that L-EPS was composed of mannose, glucuronic acid, galactosamine, glucose, galactose, and xylose, with the molar ratio of 32.26:0.99:1.79:5.63:0.05:4.07, while S-EPS was composed of mannose, glucuronic acid, galactosamine, glucose, and galactose, with the molar ratio of 25.43:2.28:7.13:5.35. L-EPS was separated into the neutral polysaccharide L-EPS 1-1 and the acidic polysaccharide L-EPS 2-1 by ion-exchange chromatography and gel chromatography. L-EPS 2-1 exerted higher antibiofilm activity than L-EPS 1-1. The antibiofilm activity of L-EPS might be associated with its structure.IMPORTANCES. flexneri is a widespread foodborne pathogen causing food contamination and responsible for food poisoning outbreaks related to various foods in developing countries. Not only has biofilm formation by S. flexneri been difficult to eliminate, but it has also increased the drug resistance of the strain. In the present study, it was demonstrated that L-EPSs secreted by Lactobacillus plantrum 12 could inhibit S. flexneri biofilm formation on, adhesion to, and invasion into HT-29 cells. Also, L-EPSs could decrease the minimum biofilm elimination concentration (MBEC) of the antibiotics used against S. flexneri biofilm. Therefore, L-EPSs were shown to be bioactive macromolecules with the potential ability to act against S. flexneri infections.
Collapse
Affiliation(s)
- Yinglong Song
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, People's Republic of China
| | - Mengying Sun
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, People's Republic of China
| | - Lu Feng
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, People's Republic of China
| | - Xue Liang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, People's Republic of China
| | - Xing Song
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, People's Republic of China
| | - Guangqing Mu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, People's Republic of China
- Dalian Probiotics Function Research Key Laboratory, Dalian Polytechnic University, Dalian, People's Republic of China
| | - Yanfeng Tuo
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, People's Republic of China
| | - Shujuan Jiang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, People's Republic of China
| | - Fang Qian
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, People's Republic of China
| |
Collapse
|
20
|
Nowak B, Śróttek M, Ciszek-Lenda M, Skałkowska A, Gamian A, Górska S, Marcinkiewicz J. Exopolysaccharide from Lactobacillus rhamnosus KL37 Inhibits T Cell-dependent Immune Response in Mice. Arch Immunol Ther Exp (Warsz) 2020; 68:17. [PMID: 32448979 PMCID: PMC7246254 DOI: 10.1007/s00005-020-00581-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 05/10/2020] [Indexed: 01/06/2023]
Abstract
Exopolysaccharides (EPSs), major components of the bacterial biofilm, display strong strain-specific immunomodulatory properties. Previously, we have shown that crude EPS derived from Lactobacillus rhamnosus KL37 depresses the production of arthritogenic anti-collagen IgG and ameliorates collagen-induced arthritis (CIA) in DBA/1 mice, when lipopolysaccharide (LPS) was used as adjuvant. In this study, we used highly purified EPS from L. rhamnosus KL37 (EPS-37) to verify its anti-inflammatory properties and the ability to suppress T cell-dependent humoral response. We have employed the model of active CIA, in which mice immunized with type II collagen (CII) along with LPS were treated with pure EPS-37. Intravenous administration of purified EPS-37 markedly ameliorated arthritis and reduced CII-specific antibody production. EPS-37 injected subcutaneously reduced the clinical symptoms of CIA but without the reduction of arthritogenic antibodies. In addition, the effect of EPS-37 on T-cell functions was tested ex vivo and in vitro. EPS-37 inhibited the in vitro proliferation of T cells activated both in vivo (CII immunization) and in vitro (antigen/mitogen), and markedly reduced the production of interferon (IFN)-γ. These results together with other reports suggest that anti-inflammatory potential of EPS-37 depends on its ability to inhibit either one or the other or both possible inflammatory signaling pathways. Namely, Th1 → IFN-γ → M1 inflammatory macrophages → arthritis and/or Th1 → IFN-γ → B cells → arthritogenic antibodies → arthritis. We suggest that L. rhamnosus KL37 EPS might be utilized to control T cell-dependent immune responses in various inflammatory diseases. However, the most effective route of EPS-37 administration needs to be tailored for a given disorder.
Collapse
Affiliation(s)
- Bernadeta Nowak
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121, Kraków, Poland
| | - Małgorzata Śróttek
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121, Kraków, Poland
| | - Marta Ciszek-Lenda
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121, Kraków, Poland
| | - Anna Skałkowska
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121, Kraków, Poland
| | - Andrzej Gamian
- Laboratory of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Sabina Górska
- Laboratory of Microbiome Immunology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Janusz Marcinkiewicz
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121, Kraków, Poland.
| |
Collapse
|
21
|
Hsieh SA, Allen PM. Immunomodulatory Roles of Polysaccharide Capsules in the Intestine. Front Immunol 2020; 11:690. [PMID: 32351514 PMCID: PMC7174666 DOI: 10.3389/fimmu.2020.00690] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
The interplay between the immune system and the microbiota in the human intestine dictates states of health vs. disease. Polysaccharide capsules are critical elements of bacteria that protect bacteria against environmental and host factors, including the host immune system. This review summarizes the mechanisms by which polysaccharide capsules from commensal and pathogenic bacteria in the gut microbiota modulate the innate and adaptive immune systems in the intestine. A deeper understanding of the roles of polysaccharide capsules in microbiota-immune interactions will provide a basis to harness their therapeutic potential to advance human health.
Collapse
Affiliation(s)
- Samantha A Hsieh
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Paul M Allen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
22
|
Suppression of Staphylococcus aureus Superantigen-Independent Interferon Gamma Response by a Probiotic Polysaccharide. Infect Immun 2020; 88:IAI.00661-19. [PMID: 31932326 DOI: 10.1128/iai.00661-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/03/2020] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive opportunistic pathogen that causes a variety of diseases. Bloodstream infection is the most severe, with mortality rates reaching 20 to 50%. Exopolysaccharide (EPS) from the probiotic Bacillus subtilis reduces bacterial burden and inflammation during S. aureus bloodstream infection in mice. Protection is due, in part, to hybrid macrophages that restrict S. aureus growth through reactive oxygen species and to limiting superantigen-induced T cell activation and interferon gamma (IFN-γ) production during infection. A decrease in IFN-γ production was observed within 24 h after infection, and here, we investigated how EPS abrogates its production. We discovered that S. aureus uses a rapid, superantigen-independent mechanism to induce host IFN-γ and that this is mediated by interleukin-12 (IL-12) activation of NK cells. Furthermore, we found that EPS limits IFN-γ production by modulating host immunity in a Toll-like receptor 4 (TLR4)-dependent manner, a signaling pathway that is required for EPS-mediated protection from S. aureus infection in vivo We conclude that EPS protects hosts from acute bloodstream S. aureus infection not only by inducing macrophages that restrict S. aureus growth and inhibit superantigen-activated T cells but also by limiting NK cell production of IFN-γ after S. aureus infection in a TLR4-dependent manner.
Collapse
|
23
|
Macrophage Polarization Induced by Probiotic Bacteria: a Concise Review. Probiotics Antimicrob Proteins 2019; 12:798-808. [DOI: 10.1007/s12602-019-09612-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
24
|
Grayczyk JP, Alonzo F. Staphylococcus aureus Lipoic Acid Synthesis Limits Macrophage Reactive Oxygen and Nitrogen Species Production To Promote Survival during Infection. Infect Immun 2019; 87:e00344-19. [PMID: 31308080 PMCID: PMC6759302 DOI: 10.1128/iai.00344-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/05/2019] [Indexed: 01/29/2023] Open
Abstract
Macrophages are critical mediators of innate immunity and must be overcome for bacterial pathogens to cause disease. The Gram-positive bacterium Staphylococcus aureus produces virulence factors that impede macrophages and other immune cells. We previously determined that production of the metabolic cofactor lipoic acid by the lipoic acid synthetase, LipA, blunts macrophage activation. A ΔlipA mutant was attenuated during infection and was more readily cleared from the host. We hypothesized that bacterial lipoic acid synthesis perturbs macrophage antimicrobial functions and therefore hinders the clearance of S. aureus Here, we found that enhanced innate immune cell activation after infection with a ΔlipA mutant was central to attenuation in vivo, whereas a growth defect imparted by the lipA mutation made a negligible contribution to overall clearance. Macrophages recruited to the site of infection with the ΔlipA mutant produced larger amounts of bactericidal reactive oxygen species (ROS) and reactive nitrogen species (RNS) than those recruited to the site of infection with the wild-type strain or the mutant strain complemented with lipA ROS derived from the NADPH phagocyte oxidase complex and RNS derived from the inducible nitric oxide synthetase, but not mitochondrial ROS, were critical for the restriction of bacterial growth under these conditions. Despite enhanced antimicrobial immunity upon primary infection with the ΔlipA mutant, we found that the host failed to mount an improved recall response to secondary infection. Our data suggest that lipoic acid synthesis in S. aureus promotes bacterial persistence during infection through limitation of ROS and RNS generation by macrophages. Broadly, this work furthers our understanding of the intersections between bacterial metabolism and immune responses to infection.
Collapse
Affiliation(s)
- James P Grayczyk
- Department of Microbiology and Immunology, Loyola University Chicago-Stritch School of Medicine, Maywood, Illinois, USA
| | - Francis Alonzo
- Department of Microbiology and Immunology, Loyola University Chicago-Stritch School of Medicine, Maywood, Illinois, USA
| |
Collapse
|