1
|
Vashishth S, Ambasta RK, Kumar P. Deciphering the microbial map and its implications in the therapeutics of neurodegenerative disorder. Ageing Res Rev 2024; 100:102466. [PMID: 39197710 DOI: 10.1016/j.arr.2024.102466] [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: 07/31/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Every facet of biological anthropology, including development, ageing, diseases, and even health maintenance, is influenced by gut microbiota's significant genetic and metabolic capabilities. With current advancements in sequencing technology and with new culture-independent approaches, researchers can surpass older correlative studies and develop mechanism-based studies on microbiome-host interactions. The microbiota-gut-brain axis (MGBA) regulates glial functioning, making it a possible target for the improvement of development and advancement of treatments for neurodegenerative diseases (NDDs). The gut-brain axis (GBA) is accountable for the reciprocal communication between the gastrointestinal and central nervous system, which plays an essential role in the regulation of physiological processes like controlling hunger, metabolism, and various gastrointestinal functions. Lately, studies have discovered the function of the gut microbiome for brain health-different microbiota through different pathways such as immunological, neurological and metabolic pathways. Additionally, we review the involvement of the neurotransmitters and the gut hormones related to gut microbiota. We also explore the MGBA in neurodegenerative disorders by focusing on metabolites. Further, targeting the blood-brain barrier (BBB), intestinal barrier, meninges, and peripheral immune system is investigated. Lastly, we discuss the therapeutics approach and evaluate the pre-clinical and clinical trial data regarding using prebiotics, probiotics, paraprobiotics, fecal microbiota transplantation, personalised medicine, and natural food bioactive in NDDs. A comprehensive study of the GBA will felicitate the creation of efficient therapeutic approaches for treating different NDDs.
Collapse
Affiliation(s)
- Shrutikirti Vashishth
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Department of Medicine, School of Medicine, VUMC, Vanderbilt University, TN, USA
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India.
| |
Collapse
|
2
|
Mobasherpour P, Yavarmanesh M, Edalatian Dovom MR. Antitumor properties of traditional lactic acid bacteria: Short-chain fatty acid production and interleukin 12 induction. Heliyon 2024; 10:e36183. [PMID: 39253228 PMCID: PMC11382311 DOI: 10.1016/j.heliyon.2024.e36183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
This paper presents an in vitro evaluation of antitumor properties through producing short-chain fatty acids and inducing interleukin 12. In addition, it offers the most important and functional probiotic properties of 24 Lactobacillus gasseri, Lactiplantibacillus plantarum, Lactobacillus acidophilus, and Limosilactobacillus fermentum strains isolated from humans, foods, and fermented foods. To this end, survival in an acidic environment (pH = 2.5), tolerance in bile salt, viability in the presence of pepsin-pancreatin, adhesion percentage, antibiotic resistance, auto-aggregation, and potential percentage of co-aggregation are studied in contact with three human intestinal pathogens. These pathogens are Escherichia coli O157: H7 NCTC 12900, Salmonella enterica subsp. enterica ATCC 13076, and Listeria monocytogenes ATTC 7644. Also, in vitro induction amount of IL-12 in mouse splenocytes is investigated to evaluate antitumor properties by 19 strains of L. gasseri and L. plantarum along with the development of short-chain fatty acids (SCFA) by 5 strains of L. fermentum and L. acidophilus. Gas Chromatography Flame Ionization Detector (GC-FID) and enzyme-linked immunosorbent assay (ELISA) were used to measure short-chain fatty acids and IL-12, respectively. All strains had high viability under acidic conditions. The highest levels of pancreatin and pepsin resistance were found in strains LF56, LF57, LF55, OF, and F and strains LF56, LF57, and A7, respectively. All strains except LF56 had high resistance to bile salts. L. gasseri 54C had the highest average adhesion score (hydrophobicity) of 62.9 % among 19 strains. Despite the susceptibility of different strains of L. plantarum to the tested antibiotics, M8 and M11, S2G, A7, LF55, LF57, and 5G were resistant to kanamycin and chloramphenicol, respectively. Also, 21G was resistant to ampicillin, LF56 to tetracycline and M8, and M11, LF56, and 21G to Erythromycin. In addition, L. gasseri showed moderate resistance to ampicillin, erythromycin, and tetracycline, while L. fermentum ATCC 9338 showed good resistance to ampicillin, erythromycin, and chloramphenicol. In this respect, L. plantarum LF56 and gasseri 54C had the highest average auto-aggregation and co-aggregation against three pathogenic bacteria, respectively. The highest and lowest levels of acetic acid as short-chain fatty acids were produced by L. fermentum 19SH isolated from Horre 41.62 and L. fermentum 21SH from fermented seeds 27.047, respectively. Moreover, L. fermentum, with the OF code of traditional-fermented food origin, produced the most isobutyric acid, butyric acid, and valeric acid, with values of 0.6828, 0.74165, and 0.49915 mmol, respectively. L. fermentum isolated from the human origin with code F produced the most isovaleric acid of 1.1874 mmol. All the tested strains produced good propionic acid except L. fermentum 21SH from fermented seeds. Among strains, L. plantarum M11 isolated from milk and L. gasseri 52B from humans had the highest in vitro induction of IL-12, which is probably related to their cell wall compositions and structure.
Collapse
Affiliation(s)
- Parinaz Mobasherpour
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Masoud Yavarmanesh
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | | |
Collapse
|
3
|
Sarfi S, Azaryan E, Naseri M. Immune System of Dental Pulp in Inflamed and Normal Tissue. DNA Cell Biol 2024; 43:369-386. [PMID: 38959180 DOI: 10.1089/dna.2024.0044] [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] [Indexed: 07/05/2024] Open
Abstract
Teeth are vulnerable to structural compromise, primarily attributed to carious lesions, in which microorganisms originating from the oral cavity deteriorate the mineralized structures of enamel and dentin, subsequently infiltrating the underlying soft connective tissue, known as the dental pulp. Nonetheless, dental pulp possesses the necessary capabilities to detect and defend against bacteria and their by-products, using a variety of intricate defense mechanisms. The pulp houses specialized cells known as odontoblasts, which encounter harmful substances produced by oral bacteria. These cells identify pathogens at an early stage and commence the immune system response. As bacteria approach the pulp, various cell types within the pulp, such as different immune cells, stem cells, fibroblasts, as well as neuronal and vascular networks, contribute a range of defense mechanisms. Therefore, the immune system is present in the healthy pulp to restrain the initial spread of pathogens, and then in the inflamed pulp, it prepares the conditions for necrosis or regeneration, so inflammatory response mechanisms play a critical role in maintaining tissue homeostasis. This review aims to consolidate the existing literature on the immune system in dental pulp, encompassing current knowledge on this topic that explains the diverse mechanisms of recognition and defense against pathogens exhibited by dental pulp cells, elucidates the mechanisms of innate and adaptive immunity in inflamed pulp, and highlights the difference between inflamed and normal pulp tissue.
Collapse
Affiliation(s)
- Sepideh Sarfi
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Immunology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Ehsaneh Azaryan
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Naseri
- Cellular, and Molecular Research Center, Department of Molecular Medicine, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
4
|
Wang S, Wang P, Wang D, Shen S, Wang S, Li Y, Chen H. Postbiotics in inflammatory bowel disease: efficacy, mechanism, and therapeutic implications. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024. [PMID: 39007163 DOI: 10.1002/jsfa.13721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024]
Abstract
Inflammatory bowel disease (IBD) is one of the most challenging diseases in the 21st century, and more than 10 million people around the world suffer from IBD. Because of the limitations and adverse effects associated with conventional IBD therapies, there has been increased scientific interest in microbial-derived biomolecules, known as postbiotics. Postbiotics are defined as the preparation of inanimate microorganisms and/or their components that confer a health benefit on the host, comprising inactivated microbial cells, cell fractions, metabolites, etc. Postbiotics have shown potential in enhancing IBD treatment by reducing inflammation, modulating the immune system, stabilizing intestinal flora and maintaining the integrity of intestinal barriers. Consequently, they are considered promising adjunctive therapies for IBD. Recent studies indicate that postbiotics offer distinctive advantages, including spanning clinical (safe origin), technological (easy for storage and transportation) and economic (reduced production costs) dimensions, rendering them suitable for widespread applications in functional food/pharmaceutical. This review offers a comprehensive overview of the definition, classification and applications of postbiotics, with an emphasis on their biological activity in both the prevention and treatment of IBD. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Shuxin Wang
- Marine College, Shandong University, Weihai, China
| | - Pu Wang
- Marine College, Shandong University, Weihai, China
| | - Donghui Wang
- Marine College, Shandong University, Weihai, China
| | | | - Shiqi Wang
- Marine College, Shandong University, Weihai, China
| | - Yuanyuan Li
- Department of Food Science, Cornell University, Ithaca, NY, USA
| | - Hao Chen
- Marine College, Shandong University, Weihai, China
| |
Collapse
|
5
|
Nakagawa R, Gu W, Mizobuchi H, Kodera S, Takano T, Wang Y, Fujioka I, Uchida K, Nakajima-Adachi H, Hachimura S. Lactococcus lactis subsp. cremoris YRC3780 modifies function of mesenteric lymph node dendritic cells to modulate the balance of T cell differentiation inducing regulatory T cells. Front Immunol 2024; 15:1395380. [PMID: 39040096 PMCID: PMC11261344 DOI: 10.3389/fimmu.2024.1395380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction The intestinal immune system plays a pivotal role in the induction of immune responses against food. In the case of T cell response, dendritic cells (DCs) are especially important. However, the regulation of immune responses to food by intestinal DCs has been poorly described. In this study, we analyzed the effect of Lactococcus lactis subsp. cremoris YRC3780, a lactic acid bacterial strain isolated from kefir, a traditional fermented milk product, on the immune responses induced by antigen presentation by intestinal DCs to T cells as well as the mechanism of action of these immunomodulatory effects. It has been shown that L. cremoris YRC3780 ameliorates the symptoms of pollinosis in both animal and human studies. Methods CD11c+ cells from mesenteric lymph nodes (MLNs) of BALB/c mice were cultured as MLN DCs with L. cremoris YRC3780 and expression of genes inducing regulatory T cells (Tregs) was examined by qPCR. In addition, MLN DCs were cocultured with CD4+ T cells from DO11.10 transgenic mice expressing an ovalbumin (OVA)-specific TCR and the OVA antigen peptide and L. cremoris YRC3780. Induction of Tregs was examined by flow cytometry, gene expression was analyzed by DNA microarray and qPCR, and the production of cytokines was measured by ELISA. MLN DCs from TLR2-deficient mice and components of L. cremoris YRC3780 were used to examine the recognition of YRC3780 by MLN DCs. Results L. cremoris YRC3780 enhanced the expression of genes involved in Treg induction in MLN DCs and induced Foxp3+CD4+T cells in an MLN DC and CD4+ T-cell co-culture system. The effect on MLN DCs was likely mediated by receptors other than TLR2. Together with microarray analyses of CD4+ T cell gene expression and cytokine ELISA, it was demonstrated that L. cremoris YRC3780 promoted the induction of Th1 and Tregs, and regulated the balance of Th1/Th2 and Treg/Th17 cells involving multiple genes via the antigen-presentation of MLN DCs. Discussion Our findings provide insights into the modulation of intestinal immune responses mediated by DCs and the antiallergic effects of lactic acid bacteria.
Collapse
Affiliation(s)
- Ryogo Nakagawa
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Wenting Gu
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hibine Mizobuchi
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shuhei Kodera
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomohiro Takano
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yimei Wang
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ikumi Fujioka
- R&D Center, Yotsuba Milk Products, Co., Ltd., Kitahiroshima, Japan
| | - Kenji Uchida
- R&D Center, Yotsuba Milk Products, Co., Ltd., Kitahiroshima, Japan
| | - Haruyo Nakajima-Adachi
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Satoshi Hachimura
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Watanabe M, Nakai H, Ohara T, Kawasaki K, Murosaki S, Hirose Y. Beneficial effect of heat-killed Lactiplantibacillus plantarum L-137 on intestinal barrier function of rat small intestinal epithelial cells. Sci Rep 2024; 14:12319. [PMID: 38811623 PMCID: PMC11136994 DOI: 10.1038/s41598-024-62657-0] [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: 02/06/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
Heat-killed Lactiplantibacillus plantarum L-137 (HK L-137) has been suggested to enhance the intestinal barrier in obese mice, leading to improvement of metabolic abnormalities and adipose tissue inflammation, and in healthy humans with overweight, leading to improvement of systemic inflammation. However, its detailed mechanism of action has not been clarified. Therefore, this study investigated the effects of HK L-137 on the permeability of rat small intestinal epithelial IEC-6 cells, tight junction-related gene and protein expression and localization, and intracellular signaling pathways involved in barrier function. Treatment of IEC-6 cells with HK L-137 for 26 h significantly reduced the permeability to fluorescein isothiocyanate-dextran (FD-4). HK L-137 also increased gene and protein expression of zonula occludens-1 (ZO-1), an important tight junction protein, without affecting the localization. Furthermore, inhibition of the extracellular signal-regulated kinase (ERK)1/2 pathway in IEC-6 cells canceled the HK L-137-related reduction in permeability to FD-4. Phosphorylation of ERK in IEC-6 cells was induced 15 min after the addition of HK L-137. These results suggest that HK L-137 reduces intestinal permeability partly through activating the ERK pathway and increasing expression of the ZO-1 gene and protein. Enhancement of intestinal barrier function with HK L-137 might be effective in preventing and treating leaky gut, for which no specific therapeutic tool has been established.
Collapse
Affiliation(s)
- Mototsugu Watanabe
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan.
| | - Hiroko Nakai
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| | - Tatsuya Ohara
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| | - Kengo Kawasaki
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| | - Shinji Murosaki
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| | - Yoshitaka Hirose
- Research & Development Institute, House Wellness Foods Corporation, 3-20 Imoji, Itami, Hyogo, 664-0011, Japan
| |
Collapse
|
7
|
Sangiorgio G, Nicitra E, Bivona D, Bonomo C, Bonacci P, Santagati M, Musso N, Bongiorno D, Stefani S. Interactions of Gram-Positive Bacterial Membrane Vesicles and Hosts: Updates and Future Directions. Int J Mol Sci 2024; 25:2904. [PMID: 38474151 DOI: 10.3390/ijms25052904] [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: 01/24/2024] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayers derived from cell membranes, released by both eukaryotic cells and bacteria into the extracellular environment. During production, EVs carry proteins, nucleic acids, and various compounds, which are then released. While Gram-positive bacteria were traditionally thought incapable of producing EVs due to their thick peptidoglycan cell walls, recent studies on membrane vesicles (MVs) in Gram-positive bacteria have revealed their significant role in bacterial physiology and disease progression. This review explores the current understanding of MVs in Gram-positive bacteria, including the characterization of their content and functions, as well as their interactions with host and bacterial cells. It offers a fresh perspective to enhance our comprehension of Gram-positive bacterial EVs.
Collapse
Affiliation(s)
- Giuseppe Sangiorgio
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Emanuele Nicitra
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Dalida Bivona
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Carmelo Bonomo
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Paolo Bonacci
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Maria Santagati
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Dafne Bongiorno
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| |
Collapse
|
8
|
Xie W, Zhong YS, Li XJ, Kang YK, Peng QY, Ying HZ. Postbiotics in colorectal cancer: intervention mechanisms and perspectives. Front Microbiol 2024; 15:1360225. [PMID: 38450163 PMCID: PMC10914944 DOI: 10.3389/fmicb.2024.1360225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024] Open
Abstract
Colorectal cancer (CRC) is a common malignancy affecting the gastrointestinal tract worldwide. The etiology and progression of CRC are related to factors such as environmental influences, dietary structure, and genetic susceptibility. Intestinal microbiota can influence the integrity of the intestinal mucosal barrier and modulate intestinal immunity by secreting various metabolites. Dysbiosis of the intestinal microbiota can affect the metabolites of the microbial, leading to the accumulation of toxic metabolites, which can trigger chronic inflammation or DNA damage and ultimately lead to cellular carcinogenesis and the development of CRC. Postbiotics are preparations of inanimate microorganisms or their components that are beneficial to the health of the host, with the main components including bacterial components (e.g., exopolysaccharides, teichoic acids, surface layer protein) and metabolites (e.g., short-chain fatty acids, tryptophan metabolite, bile acids, vitamins and enzymes). Compared with traditional probiotics, it has a more stable chemical structure and higher safety. In recent years, it has been demonstrated that postbiotics are involved in regulating intestinal microecology and improving the progression of CRC, which provides new ideas for the prevention and diagnosis of CRC. In this article, we review the changes in intestinal microbiota in different states of the gut and the mechanisms of anti-tumor activity of postbiotic-related components, and discuss the potential significance of postbiotics in the diagnosis and treatment of CRC. This reviews the changes and pathogenesis of intestinal microbiota in the development of CRC, and summarizes the relevant mechanisms of postbiotics in resisting the development of CRC in recent years, as well as the advantages and limitations of postbiotics in the treatment process of CRC.
Collapse
Affiliation(s)
| | | | | | | | | | - Hua-Zhong Ying
- Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
9
|
Han J, Zhao X, Zhao X, Li P, Gu Q. Insight into the structure, biosynthesis, isolation method and biological function of teichoic acid in different gram-positive microorganisms: A review. Int J Biol Macromol 2023; 253:126825. [PMID: 37696369 DOI: 10.1016/j.ijbiomac.2023.126825] [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: 07/11/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023]
Abstract
Teichoic acid (TA) is a weakly anionic polymer present in the cell walls of Gram-positive bacteria. It can be classified into wall teichoic acid (WTA) and lipoteichoic acid (LTA) based on its localization in the cell wall. The structure and biosynthetic pathway of TAs are strain-specific and have a significant role in maintaining cell wall stability. TAs have various beneficial functions, such as immunomodulatory, anticancer and antioxidant activities. However, the purity and yield of TAs are generally not high, and different isolation methods may even affect their structural integrity, which limits the research progress on the probiotic functions of TA. This paper reviews an overview of the structure and biosynthetic pathway of TAs in different strains, as well as the research progress of the isolation and purification methods of TAs. Furthermore, this review also highlights the current research status on the biological functions of TAs. Through a comprehensive understanding of this review, it is expected to pave the way for advancements in isolating and purifying high-quality TAs and, in turn, lay a foundation for contributing to the development of targeted probiotic therapies.
Collapse
Affiliation(s)
- Jiarun Han
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, People's Republic of China
| | - Xin Zhao
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, People's Republic of China
| | - Xilian Zhao
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, People's Republic of China
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, People's Republic of China
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, People's Republic of China.
| |
Collapse
|
10
|
Gurunathan S, Thangaraj P, Kim JH. Postbiotics: Functional Food Materials and Therapeutic Agents for Cancer, Diabetes, and Inflammatory Diseases. Foods 2023; 13:89. [PMID: 38201117 PMCID: PMC10778838 DOI: 10.3390/foods13010089] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Postbiotics are (i) "soluble factors secreted by live bacteria, or released after bacterial lysis, such as enzymes, peptides, teichoic acids, peptidoglycan-derived muropeptides, polysaccharides, cell-surface proteins and organic acids"; (ii) "non-viable metabolites produced by microorganisms that exert biological effects on the hosts"; and (iii) "compounds produced by microorganisms, released from food components or microbial constituents, including non-viable cells that, when administered in adequate amounts, promote health and wellbeing". A probiotic- and prebiotic-rich diet ensures an adequate supply of these vital nutrients. During the anaerobic fermentation of organic nutrients, such as prebiotics, postbiotics act as a benevolent bioactive molecule matrix. Postbiotics can be used as functional components in the food industry by offering a number of advantages, such as being added to foods that are harmful to probiotic survival. Postbiotic supplements have grown in popularity in the food, cosmetic, and healthcare industries because of their numerous health advantages. Their classification depends on various factors, including the type of microorganism, structural composition, and physiological functions. This review offers a succinct introduction to postbiotics while discussing their salient features and classification, production, purification, characterization, biological functions, and applications in the food industry. Furthermore, their therapeutic mechanisms as antibacterial, antiviral, antioxidant, anticancer, anti-diabetic, and anti-inflammatory agents are elucidated.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641021, Tamil Nadu, India;
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641021, Tamil Nadu, India;
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
11
|
Liang B, Xing D. The Current and Future Perspectives of Postbiotics. Probiotics Antimicrob Proteins 2023; 15:1626-1643. [PMID: 36763279 PMCID: PMC9913028 DOI: 10.1007/s12602-023-10045-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 02/11/2023]
Abstract
With the emphasis on intestinal health, probiotics have exploded into a vast market potential. However, new scientific evidence points out that the beneficial health benefits of probiotics are not necessarily directly related to viable bacteria. However, the metabolites or bacterial components of the live bacteria are the driving force behind health promotion. Therefore, scientists gradually noticed that the beneficial effects of probiotics are based on bacteria itself, metabolites, or cell lysates, and these factors are officially named "postbiotics" by the ISAPP. Postbiotic components are diverse and outperform live probiotics in terms of technology, safety, and cost due to their good absorption, metabolism, and organismal distribution. Postbiotics have been shown to have bioactivities such as antimicrobial, antioxidant, anti-inflammatory, anti-proliferative, and immunomodulation. Moreover, numerous studies have revealed the significant potential of postbiotics for disease treatment. This paper first presents the production and classification of postbiotics with examples from lactic acid bacteria (LAB), followed by the mechanisms of action with the most recent pre-clinical and clinical studies and the wide range of non-clinical and clinical applications of postbiotics. Furthermore, the current and future prospects of the postbiotic market with commercial available products are discussed. Finally, we comment on the knowledge gaps and future clinical applications with several examples.
Collapse
Affiliation(s)
- Bing Liang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.
- Cancer Institute, Qingdao University, Qingdao, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Cancer Institute, Qingdao University, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
12
|
Chen S, Nai Z, Qin Z, Li G, He X, Wang W, Tian Y, Liu D, Jiang X. The extracellular polysaccharide inhibit porcine epidemic diarrhea virus with extract and gene editing Lacticaseibacillus. Microb Cell Fact 2023; 22:225. [PMID: 37924089 PMCID: PMC10625274 DOI: 10.1186/s12934-023-02226-8] [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: 12/17/2022] [Accepted: 10/07/2023] [Indexed: 11/06/2023] Open
Abstract
Lacticaseibacillus is one of the predominant microorganisms in gut from human and animal, and the lacticaseibacillus have effective applications against the viral diarrhea of piglets in the farm. However, the function and the concrete cell single pathways of the active ingredient from lacticaseibacillus was not clear within anti-infection in the postbiotics research. Here, we compared the biological function of extracellular polysaccharides (EPS) purified from lacticaseibacillus casei (L. casei) and gene editing lacticaseibacillus casei with the CRISPER-Cas9 technology, which were with the ability of antioxidation and anti-inflammation, and the EPS could also inhibit the ROS production within the Porcine Small Intestinal Epithelial Cells-J2 (IPEC-J2). Interestingly, we found that both of EPS and genome editing lacticaseibacillus casei could specifically target the IFN-λ expression in the IPEC-J2, which was beneficial against the PEDV infection in the virus replication and production with the qRT-PCR and indirect immunofluorescence methods. Finally, the STAT3 cell single pathway was stimulated to transcribe IFN-λ with the EPS to elucidate the detailed mechanism of activating type III IFN signals receptor of IL-10R2, which play the function between anti-inflammation and anti-virus in the PEDV infection. Taken together, our research linked a postbiotics of EPS with the antiviral infection of PEDV, which suggest that the lacticaseibacillus itself still have displayed the potential immunomodulatory activities, and highlight the immunomodulatory potential of EPS-producing microbes.
Collapse
Affiliation(s)
- Shaojun Chen
- Northeast Agricultural University, Harbin, 150030, Heilongjiang, People's Republic of China
| | - Zida Nai
- Yanbian University, Yanji, 133002, Jilin, People's Republic of China
| | - Ziliang Qin
- Northeast Agricultural University, Harbin, 150030, Heilongjiang, People's Republic of China
| | - Gang Li
- Northeast Agricultural University, Harbin, 150030, Heilongjiang, People's Republic of China
| | - Xinmiao He
- Key Laboratory of Combining Farming and Animal Husbandry, Ministry of Agriculture, Animal Husbandry Research Institute, Heilongjiang Academy of Agricultural Sciences No, 368 Xuefu Road, Harbin, 150086, People's Republic of China
| | - Wentao Wang
- Key Laboratory of Combining Farming and Animal Husbandry, Ministry of Agriculture, Animal Husbandry Research Institute, Heilongjiang Academy of Agricultural Sciences No, 368 Xuefu Road, Harbin, 150086, People's Republic of China
| | - Yaguang Tian
- Northeast Agricultural University, Harbin, 150030, Heilongjiang, People's Republic of China
- Undergraduate Experimental and Teaching Center, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, Heilongjiang, People's Republic of China
| | - Di Liu
- Northeast Agricultural University, Harbin, 150030, Heilongjiang, People's Republic of China.
- Key Laboratory of Combining Farming and Animal Husbandry, Ministry of Agriculture, Animal Husbandry Research Institute, Heilongjiang Academy of Agricultural Sciences No, 368 Xuefu Road, Harbin, 150086, People's Republic of China.
| | - Xinpeng Jiang
- Northeast Agricultural University, Harbin, 150030, Heilongjiang, People's Republic of China.
| |
Collapse
|
13
|
Choi YH, Kim BS, Kang SS. Genomic DNA Extracted from Lactiplantibacillus plantarum Attenuates Porphyromonas gingivalis Lipopolysaccharide (LPS)-Induced Inflammatory Responses via Suppression of Toll-Like Receptor (TLR)-Mediated Mitogen-Activated Protein Kinase (MAPK) and Nuclear Factor-κB (NF-κB) Signaling Pathways. Food Sci Anim Resour 2023; 43:938-947. [PMID: 37701749 PMCID: PMC10493568 DOI: 10.5851/kosfa.2023.e43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/14/2023] Open
Abstract
In the present study, we aimed to examine the inhibition of genomic DNA from Lactiplantibacillus plantarum (LpDNA) on Porphyromonas gingivalis lipopolysaccharide (PgLPS)-induced inflammatory responses in RAW264.7 cells. Pretreatment with LpDNA for 15 h significantly inhibited PgLPS-induced mRNA expression and protein secretion of interleukin (IL)-1β, IL-6, and monocyte chemoattractant protein-1. LpDNA pretreatment also reduced the mRNA expression of Toll-like receptor (TLR)2 and TLR4. Furthermore, LpDNA inhibited the phosphorylation of mitogen-activated protein kinases (MAPKs) and the activation of nuclear factor-κB (NF-κB) induced by PgLPS. Taken together, these findings demonstrate that LpDNA attenuates PgLPS-induced inflammatory responses by regulating MAPKs and NF-κB signaling pathways through the suppression of TLR2 and TLR4 expression.
Collapse
Affiliation(s)
- Young Hyeon Choi
- Department of Food Science and
Biotechnology, College of Life Science and Biotechnology, Dongguk
University, Goyang 10326, Korea
| | - Bong Sun Kim
- Division of Applied Food System, Major in
Food Science & Technology, Seoul Women’s
University, Seoul 01797, Korea
| | - Seok-Seong Kang
- Department of Food Science and
Biotechnology, College of Life Science and Biotechnology, Dongguk
University, Goyang 10326, Korea
| |
Collapse
|
14
|
Pradhan D, Gulati G, Avadhani R, H M R, Soumya K, Kumari A, Gupta A, Dwivedi D, Kaushik JK, Grover S. Postbiotic Lipoteichoic acid of probiotic Lactobacillus origin ameliorates inflammation in HT-29 cells and colitis mice. Int J Biol Macromol 2023; 236:123962. [PMID: 36907160 DOI: 10.1016/j.ijbiomac.2023.123962] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/21/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023]
Abstract
Lipoteichoic acid (LTA) is a key surface component of probiotic lactobacilli that is involved in important cellular functions including cross talk with the host immune cells. In this study, the anti-inflammatory and ameliorative properties of LTA from probiotic lactobacilli strains were assessed in in vitro HT-29 cells and in vivo colitis mice. The LTA was extracted with n-butanol and its safety was confirmed based on its endotoxin content and cytotoxicity in HT-29 cells. In the Lipopolysaccharide stimulated HT-29 cells, the LTA from the test probiotics evoked a visible but non-significant increase in IL-10 and decrease in TNF-α levels. During the colitis mice study, probiotic LTA treated mice showed substantial improvement in external colitis symptoms, disease activity score and weight gain. The treated mice also showed improvements in key inflammatory markers such as the gut permeability, myeloperoxidase activity and histopathological damages in colon, although non-significant improvements were recorded for the inflammatory cytokines. Furthermore, structural studies by NMR and FTIR revealed increased level of D-alanine substitution in the LTA of LGG strain over MTCC5690. The present study demonstrates the ameliorative effect of LTA as a postbiotic component from probiotics which can be helpful in building effective strategies for combating gut inflammatory disorders.
Collapse
Affiliation(s)
- Diwas Pradhan
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India.
| | - Ganga Gulati
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Rashmi Avadhani
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Rashmi H M
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Kandukuri Soumya
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Anisha Kumari
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Archita Gupta
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | | | - Jai K Kaushik
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Sunita Grover
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India.
| |
Collapse
|
15
|
Fan X, Mai C, Zuo L, Huang J, Xie C, Jiang Z, Li R, Yao X, Fan X, Wu Q, Yan P, Liu L, Chen J, Xie Y, Leung ELH. Herbal formula BaWeiBaiDuSan alleviates polymicrobial sepsis-induced liver injury via increasing the gut microbiota Lactobacillus johnsonii and regulating macrophage anti-inflammatory activity in mice. Acta Pharm Sin B 2023; 13:1164-1179. [PMID: 36970196 PMCID: PMC10031256 DOI: 10.1016/j.apsb.2022.10.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/19/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Sepsis-induced liver injury (SILI) is an important cause of septicemia deaths. BaWeiBaiDuSan (BWBDS) was extracted from a formula of Panax ginseng C. A. Meyer, Lilium brownie F. E. Brown ex Miellez var. viridulum Baker, Polygonatum sibiricum Delar. ex Redoute, Lonicera japonica Thunb., Hippophae rhamnoides Linn., Amygdalus Communis Vas, Platycodon grandiflorus (Jacq.) A. DC., and Cortex Phelloderdri. Herein, we investigated whether the BWBDS treatment could reverse SILI by the mechanism of modulating gut microbiota. BWBDS protected mice against SILI, which was associated with promoting macrophage anti-inflammatory activity and enhancing intestinal integrity. BWBDS selectively promoted the growth of Lactobacillus johnsonii (L. johnsonii) in cecal ligation and puncture treated mice. Fecal microbiota transplantation treatment indicated that gut bacteria correlated with sepsis and was required for BWBDS anti-sepsis effects. Notably, L. johnsonii significantly reduced SILI by promoting macrophage anti-inflammatory activity, increasing interleukin-10+ M2 macrophage production and enhancing intestinal integrity. Furthermore, heat inactivation L. johnsonii (HI-L. johnsonii) treatment promoted macrophage anti-inflammatory activity and alleviated SILI. Our findings revealed BWBDS and gut microbiota L. johnsonii as novel prebiotic and probiotic that may be used to treat SILI. The potential underlying mechanism was at least in part, via L. johnsonii-dependent immune regulation and interleukin-10+ M2 macrophage production.
Collapse
Affiliation(s)
- Xiaoqing Fan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Chutian Mai
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Ling Zuo
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jumin Huang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Chun Xie
- Cancer Center, Faculty of Health Science; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, China
| | - Zebo Jiang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Runze Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Xiaojun Yao
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xingxing Fan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Qibiao Wu
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Peiyu Yan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Liang Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Jianxin Chen
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ying Xie
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, China
| |
Collapse
|
16
|
Rahman Z, Dandekar MP. Implication of Paraprobiotics in Age-Associated Gut Dysbiosis and Neurodegenerative Diseases. Neuromolecular Med 2023; 25:14-26. [PMID: 35879588 DOI: 10.1007/s12017-022-08722-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/13/2022] [Indexed: 10/16/2022]
Abstract
Neurodegenerative diseases, including Alzheimer's and Parkinson's disease, are major age-related concerns in elderly people. Since no drug fully addresses the progression of neurodegenerative diseases, advance treatment strategies are urgently needed. Several studies have noted the senescence of immune system and the perturbation of gut microbiota in the aged population. In recent years, the role of gut microbiota has been increasingly studied in the manifestation of age-related CNS disorders. In this context, prebiotics, probiotics, and paraprobiotics are reported to improve the behavioural and neurobiological abnormalities in elderly patients. As live microbiota, prescribed in the form of probiotics, shows some adverse effects like sepsis, translocation, and horizontal gene transfer, paraprobiotics could be a possible alternative strategy in designing microbiome-based therapeutics. This review describes the health-beneficial effects of paraprobiotics in age-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
17
|
Huang R, Wu F, Zhou Q, Wei W, Yue J, Xiao B, Luo Z. Lactobacillus and intestinal diseases: mechanisms of action and clinical applications. Microbiol Res 2022; 260:127019. [DOI: 10.1016/j.micres.2022.127019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
|
18
|
Zhao C, Chen H, Liang H, Zhao X, Tang W, Wei M, Li Y, Zhang J, Yu X, Chen G, Zhu H, Jiang L, Zhang X. Lactobacillus plantarum RS-09 Induces M1-Type Macrophage Immunity Against Salmonella Typhimurium Challenge via the TLR2/NF-κB Signalling Pathway. Front Pharmacol 2022; 13:832245. [PMID: 35355723 PMCID: PMC8959098 DOI: 10.3389/fphar.2022.832245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/26/2022] [Indexed: 12/30/2022] Open
Abstract
Lactobacillus plantarum can interact with macrophages against bacterial enteropathy due to its potential ability to modulate macrophage polarization. However, this mechanism is not completely understood. TLR2 can recognize microbial components and trigger macrophage cytokine responses to different gram-positive strains. The aim of this study was to investigate whether probiotic Lactobacillus plantarum RS-09 can induce macrophage polarization against Salmonella Typhimurium infection via TLR2 signalling. BALB/c mice were preadministered RS-09 continuously for 7 days and then infected with Salmonella Typhimurium ATCC14028. Mouse RAW264.7 mononuclear macrophages were stimulated with RS-09 and coincubated with ATCC14028 or PBS controls. The results of the in vivo study indicated that RS-09 could relieve S. Typhimurium-induced splenomegaly, body weight loss and death rate. RS-09 also limited the colonization and translocation of S. Typhimurium in the gastrointestinal tract and thereby protected against infection. We also observed that RS-09 upregulated the production of M1 macrophage characteristics (e.g., CD11c and IL-6) against S. Typhimurium. Furthermore, RS-09 induced the expression of TLR2 in macrophages. In an in vitro study, treatment of RAW264.7 cells with RS-09 either concurrently with or before S. Typhimurium challenge enhanced the secretion of Reactive oxygen species and Nitric oxide. This effect was related to TLR2 and NF-κB activation. Based on these findings, Lactobacillus plantarum RS-09 was shown to modulate M1 macrophage polarization and induce TLR2-linked NF-κB signalling activity in the innate immune response to S. Typhimurium infection.
Collapse
Affiliation(s)
- Chenpei Zhao
- School of Life Sciences, Ludong University, Yantai, China
| | - Huan Chen
- School of Life Sciences, Ludong University, Yantai, China
| | - Hao Liang
- Department of Microbiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyu Zhao
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Wenli Tang
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Maolian Wei
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Youzhi Li
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Jianlong Zhang
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Xin Yu
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
- Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China
| | - Guozhong Chen
- School of Life Sciences, Ludong University, Yantai, China
- Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
| | - Hongwei Zhu
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
| | - Linlin Jiang
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
- *Correspondence: Linlin Jiang, ; Xingxiao Zhang,
| | - Xingxiao Zhang
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
- *Correspondence: Linlin Jiang, ; Xingxiao Zhang,
| |
Collapse
|
19
|
Hu J, Deng F, Zhao B, Lin Z, Sun Q, Yang X, Wu M, Qiu S, Chen Y, Yan Z, Luo S, Zhao J, Liu W, Li C, Liu KX. Lactobacillus murinus alleviate intestinal ischemia/reperfusion injury through promoting the release of interleukin-10 from M2 macrophages via Toll-like receptor 2 signaling. MICROBIOME 2022; 10:38. [PMID: 35241180 PMCID: PMC8896269 DOI: 10.1186/s40168-022-01227-w] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/07/2022] [Indexed: 05/05/2023]
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) injury has high morbidity and mortality rates. Gut microbiota is a potential key factor affecting intestinal I/R injury. Populations exhibit different sensitivities to intestinal I/R injury; however, whether this interpopulation difference is related to variation in gut microbiota is unclear. Here, to elucidate the interaction between the gut microbiome and intestinal I/R injury, we performed 16S DNA sequencing on the preoperative feces of C57BL/6 mice and fecal microbiota transplantation (FMT) experiments in germ-free mice. The transwell co-culture system of small intestinal organoids extracted from control mice and macrophages extracted from control mice or Toll-like receptor 2 (TLR2)-deficient mice or interleukin-10 (IL-10)-deficient mice were established separately to explore the potential mechanism of reducing intestinal I/R injury. RESULTS Intestinal I/R-sensitive (Sen) and intestinal I/R-resistant (Res) mice were first defined according to different survival outcomes of mice suffering from intestinal I/R. Fecal microbiota composition and diversity prior to intestinal ischemia differed between Sen and Res mice. The relative abundance of Lactobacillus murinus (L. murinus) at the species level was drastically higher in Res than that in Sen mice. Clinically, the abundance of L. murinus in preoperative feces of patients undergoing cardiopulmonary bypass surgery was closely related to the degree of intestinal I/R injury after surgery. Treatment with L. murinus significantly prevented intestinal I/R-induced intestinal injury and improved mouse survival, which depended on macrophages involvement. Further, in vitro experiments indicated that promoting the release of IL-10 from macrophages through TLR2 may be a potential mechanism for L. murinus to reduce intestinal I/R injury. CONCLUSION The gut microbiome is involved in the postoperative outcome of intestinal I/R. Lactobacillus murinus alleviates mice intestinal I/R injury through macrophages, and promoting the release of IL-10 from macrophages through TLR2 may be a potential mechanism for L. murinus to reduce intestinal I/R injury. This study revealed a novel mechanism of intestinal I/R injury and a new therapeutic strategy for clinical practice. Video Abstract.
Collapse
Affiliation(s)
- Jingjuan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Fan Deng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Bingcheng Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Zebin Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Qishun Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xiao Yang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Mei Wu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Shida Qiu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Yu Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Zhengzheng Yan
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Sidan Luo
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Jin Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Weifeng Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| | - Ke Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
20
|
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
|
21
|
Song X, Li F, Zhang M, Xia Y, Ai L, Wang G. Effect of D-Ala-Ended Peptidoglycan Precursors on the Immune Regulation of Lactobacillus plantarum Strains. Front Immunol 2022; 12:825825. [PMID: 35126378 PMCID: PMC8807649 DOI: 10.3389/fimmu.2021.825825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/20/2021] [Indexed: 11/21/2022] Open
Abstract
The resistance of Lactobacillus plantarum to vancomycin depends on its peptidoglycan composition. Vancomycin has poor binding affinity with peptidoglycan precursors ending in D-alanyl-D-lactate (D-Ala-D-Lac) but binds strongly to peptidoglycan precursors ending in D-alanyl-D-alanine (D-Ala-D-Ala), resulting in resistance and sensitivity, respectively. The ligase Ddl, which generates D-Ala-D-Lac or D-Ala-D-Ala incorporated into the peptidoglycan precursor chain, is responsible for this specificity. To study the effect of peptidoglycan precursors on immunity, we constructed several strains of L. plantarum expressing the ddl gene of Lactococcus lactis to change their peptidoglycan precursors. The change in the termini of the peptidoglycan precursors was determined by the sensitivity of the strains to vancomycin. The overexpression of ddl increased the susceptibility of the strains to vancomycin. We further explored the regulation of the macrophage inflammatory response pathway by the wild-type and constructed strains, and found that these strains induced the MyD88-dependent TRAF6/MAPK pathway, and the increase in D-Ala L. plantarum peptidoglycan precursors increased the secretion of the inflammatory factors IL-6, IL-1β and TNF-α. These results indicate that D-Ala-ended peptidoglycan precursors play a central role in the variable immunomodulatory ability of L. plantarum.
Collapse
|
22
|
Mohseni AH, Casolaro V, Bermúdez-Humarán LG, Keyvani H, Taghinezhad-S S. Modulation of the PI3K/Akt/mTOR signaling pathway by probiotics as a fruitful target for orchestrating the immune response. Gut Microbes 2022; 13:1-17. [PMID: 33615993 PMCID: PMC7899637 DOI: 10.1080/19490976.2021.1886844] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) and the phosphatidylinositol-3-kinase (PI3K)/protein kinase B or Akt (PKB/Akt) signaling pathways are considered as two but somewhat interconnected significant immune pathways which play complex roles in a variety of physiological processes as well as pathological conditions. Aberrant activation of PI3K/Akt/mTOR signaling pathways has been reported to be associated in a wide variety of human diseases. Over the past few years, growing evidence in in vitro and in vivo models suggest that this sophisticated and subtle cascade mediates the orchestration of the immune response in health and disease through exposure to probiotics. An expanding body of literature has highlighted the contribution of probiotics and PI3K/Akt/mTOR signaling pathways in gastrointestinal disorders, metabolic syndrome, skin diseases, allergy, salmonella infection, and aging. However, longitudinal human studies are possibly required to verify more conclusively whether the investigational tools used to understand the regulation of these pathways might provide effective approaches in the prevention and treatment of various disorders. In this Review, we summarize the experimental evidence from recent peer-reviewed studies and provide a brief overview of the causal relationship between the effects of probiotics and their metabolites on the components of PI3K/Akt/mTOR signaling pathways and human disease.
Collapse
Affiliation(s)
- Amir Hossein Mohseni
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Salerno, Italy
| | | | - Hossein Keyvani
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran,Hossein Keyvani Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran, Tel +98 21 88715350
| | - Sedigheh Taghinezhad-S
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran,CONTACT Sedigheh Taghinezhad-S Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran
| |
Collapse
|
23
|
Afifirad R, Darb Emamie A, Golmoradi Zadeh R, Asadollahi P, Ghanavati R, Darbandi A. Effects of Pro/Prebiotics Alone over Pro/Prebiotics Combined with Conventional Antibiotic Therapy to Treat Bacterial Vaginosis: A Systematic Review. Int J Clin Pract 2022; 2022:4774783. [PMID: 35685517 PMCID: PMC9159122 DOI: 10.1155/2022/4774783] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/26/2022] [Accepted: 03/08/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Bacterial vaginosis (BV), caused by an imbalance in the vaginal microbiota, can be treated and prevented by probiotics. Pregnant women with BV can experience premature labor and spontaneous abortions. Probiotics and prebiotics promote the proliferation of beneficial microorganisms, alter the composition of the vaginal microbiota, and prevent intravaginal infections in postmenopausal women. In addition to reducing infection symptoms, pre/probiotics can also help prevent vaginal infections. MATERIALS AND METHODS A systematic review was conducted on studies from 2010 to 2020 to determine the efficacy of pre/probiotics on the treatment of BV in pregnant and nonpregnant women. The databases Medline, Scopus, Embase, and Google Scholar were systematically searched using the following keywords: "bacterial vaginosis," "probiotics," "prebiotics," and "synbiotics." RESULTS A total of 1,871 articles were found in the initial search, and 24 clinical trials were considered eligible. In studies comparing the effects of pre/probiotics and placebos with or without antibiotic therapy in patients with BV, significant differences in clinical outcomes were observed. Probiotics reduced the levels of IL-1β and IL-6, as well as the overall Nugent score and Amsel's criteria for restitution of a balanced vaginal microbiota. In addition, probiotics can reduce the vaginal colonization of Group B streptococci among pregnant women. In subjects treated with probiotics, BV cure rates were higher than those in subjects treated with antibiotics. There were no additional adverse events. CONCLUSION Pre/probiotic regimens, when used for BV treatment, are usually safe and can exhibit long-term and short-term benefits. In order to prove the benefits of pre/probiotics in BV treatment, additional high-quality research is required.
Collapse
Affiliation(s)
- Roghayeh Afifirad
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Darb Emamie
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Rezvan Golmoradi Zadeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Parisa Asadollahi
- Department of Microbiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Atieh Darbandi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Kaur H, Ali SA. Probiotics and gut microbiota: mechanistic insights into gut immune homeostasis through TLR pathway regulation. Food Funct 2022; 13:7423-7447. [DOI: 10.1039/d2fo00911k] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Consumption of probiotics as a useful functional food improves the host's wellbeing, and, when paired with prebiotics (indigestible dietary fibre/carbohydrate), often benefits the host through anaerobic fermentation.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-NDRI, 132001, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
25
|
Scarpellini E, Rinninella E, Basilico M, Colomier E, Rasetti C, Larussa T, Santori P, Abenavoli L. From Pre- and Probiotics to Post-Biotics: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:37. [PMID: 35010297 PMCID: PMC8750841 DOI: 10.3390/ijerph19010037] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/14/2021] [Accepted: 12/18/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND AND AIMS gut microbiota (GM) is a complex ecosystem containing bacteria, viruses, fungi, and yeasts. It has several functions in the human body ranging from immunomodulation to metabolic. GM derangement is called dysbiosis and is involved in several host diseases. Pre-, probiotics, and symbiotics (PRE-PRO-SYMB) have been extensively developed and studied for GM re-modulation. Herein, we review the literature data regarding the new concept of postbiotics, starting from PRE-PRO-SYMB. METHODS we conducted a search on the main medical databases for original articles, reviews, meta-analyses, randomized clinical trials, and case series using the following keywords and acronyms and their associations: gut microbiota, prebiotics, probiotics, symbiotic, and postbiotics. RESULTS postbiotics account for PRO components and metabolic products able to beneficially affect host health and GM. The deeper the knowledge about them, the greater their possible uses: the prevention and treatment of atopic, respiratory tract, and inflammatory bowel diseases. CONCLUSIONS better knowledge about postbiotics can be useful for the prevention and treatment of several human body diseases, alone or as an add-on to PRE-PRO-SYMB.
Collapse
Affiliation(s)
- Emidio Scarpellini
- Internal Medicine Unit, "Madonna del Soccorso" General Hospital, 63074 San Benedetto del Tronto, Italy
- TARGID, KU Leuven, 3000 Leuven, Belgium
| | - Emanuele Rinninella
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Martina Basilico
- Internal Medicine Unit, "Madonna del Soccorso" General Hospital, 63074 San Benedetto del Tronto, Italy
| | | | - Carlo Rasetti
- Internal Medicine Unit, "Madonna del Soccorso" General Hospital, 63074 San Benedetto del Tronto, Italy
| | - Tiziana Larussa
- Department of Health Sciences, Magna Græcia University, 88100 Catanzaro, Italy
| | - Pierangelo Santori
- Internal Medicine Unit, "Madonna del Soccorso" General Hospital, 63074 San Benedetto del Tronto, Italy
| | - Ludovico Abenavoli
- Department of Health Sciences, Magna Græcia University, 88100 Catanzaro, Italy
| |
Collapse
|
26
|
Campos-Sánchez JC, Mayor-Lafuente J, González-Silvera D, Guardiola FA, Esteban MÁ. Acute inflammatory response in the skin of gilthead seabream (Sparus aurata) caused by carrageenin. FISH & SHELLFISH IMMUNOLOGY 2021; 119:623-634. [PMID: 34656758 DOI: 10.1016/j.fsi.2021.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 06/13/2023]
Abstract
Although inflammation is a well-characterized process in mammals, few studies have dealt with the mechanisms involved in this process in fish. The present study evaluated the expression of inflammation-related genes in the skin of fish injected with carrageenin, which has previously been used in inflammatory models in mammals. In our case, fish were injected subcutaneously with PBS (as control) or carrageenin (1%), and skin samples from the injection site were collected 1.5, 3 and 6 h post-injection. The gene expression of inflammatory markers (csfr1, mhc-ii and phox40), several pro-inflammatory cytokines (il1b, tnfa, il6, il8 and il18) and other molecules related (such as myd88 and c-rel) were up-regulated at 1.5 and 3 h in fish injected with carrageenin compared with control levels. By contrast, the gene expression of anti-inflammatory molecules (nlrx1, nlrc5 isoform 1, ctsd and ctss) was down-regulated in fish injected with carrageenin and sampled 3 h post injection, again compared to the gene expression in control fish. According to our results, carrageenin can be considered not only a good stimulator to study skin inflammation in gilthead seabream but also this method might be use to study the modulation of fish inflammatory process caused by internal or external factors.
Collapse
Affiliation(s)
- Jose Carlos Campos-Sánchez
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Javier Mayor-Lafuente
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Daniel González-Silvera
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Francisco A Guardiola
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - María Ángeles Esteban
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
27
|
Kaur S, Thukral SK, Kaur P, Samota MK. Perturbations associated with hungry gut microbiome and postbiotic perspectives to strengthen the microbiome health. FUTURE FOODS 2021. [DOI: 10.1016/j.fufo.2021.100043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
28
|
Importance of particle size of oligomannose-coated liposomes for induction of Th1 immunity. Int Immunopharmacol 2021; 99:108068. [PMID: 34426114 DOI: 10.1016/j.intimp.2021.108068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/09/2021] [Accepted: 08/09/2021] [Indexed: 12/21/2022]
Abstract
Oligomannose-coated liposomes (OMLs) comprised of dipalmitoylphosphatidylcholine, cholesterol and Man3-DPPE at a molar ratio of 1:1:0.1 and particle diameters of about 1000 nm can induce liposome-encased antigen-specific strong Th1 immunity. In this study, we evaluated the effect of particle sizes of OMLs on induction of Th1 immune responses in mice. Spleen cells obtained from mice immunized with antigen-encapsulating OMLs with 1000- and 800-nm diameters secreted remarkably high levels of IFN-γ upon in vitro stimulation. In addition, sera of mice that received these OMLs had significantly higher titers of antigen-specific IgG2a than those of IgG1, which are commonly associated with Th1 responses. In contrast, treatment with antigen-encapsulating OMLs with 400- and 200-nm diameters failed to induce IFN-γ secretion from spleen cells, although these OMLs did elicit elevation of antigen-specific IgGs. In addition, the titers of serum antigen-specific IgG2a were the same as those of IgG1 in mice that received 400-nm OMLs. Resident peritoneal mononuclear phagocytes (MNPs) treated with OMLs of diameter ≥ 600 nm secreted IL-12, which is essential for induction of Th1 immune responses, while those treated with OMLs of ≤ 400 nm failed to produce this cytokine. However, 400-nm OMLs did induce enhanced expression of MHC class II and costimulatory molecules on MNPs, similarly to OMLs of ≥ 600 nm. Taken together, these results strongly indicate that OMLs of diameter ≥ 600 nm are required to induce Th1 immune responses against OML-encased antigens, although OMLs of diameter ≤ 400 nm can activate MNPs.
Collapse
|
29
|
Kaur H, Gupta T, Kapila S, Kapila R. Protective effects of potential probiotic Lactobacillus rhamnosus (MTCC-5897) fermented whey on reinforcement of intestinal epithelial barrier function in a colitis-induced murine model. Food Funct 2021; 12:6102-6116. [PMID: 34047732 DOI: 10.1039/d0fo02641g] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fermented foods provide essential nutritional components and bioactive molecules that have beneficial effects on several gastrointestinal disorders. In the present investigation, the potential protective effects of whey fermented with probiotic Lactobacillus rhamnosus MTCC-5897 on gastrointestinal health in a murine ulcerative colitis model induced by dextran sulfate sodium (DSS) were evaluated. Pre-consumption of whey fermented with probiotic L. rhamnosus (PFW) before colitis induction significantly reduced (p < 0.01) the disease activity index and improved (p < 0.05) the hematological parameters and histological scores. The considerably diminished levels (p < 0.01) of pro-inflammatory markers (IL-4, TNF-α, CRP and MPO activity) and the enhanced (p < 0.05) levels of the anti-inflammatory cytokine TGF-β with IgA in the intestine upon feeding PFW appeared to prevent inflammation on colitis induction. Transcriptional modulations in pathogen recognition receptors (TLR-2/4) and tight junctional genes (ZO-1, occludin, claudin-1) along with localized distribution of junctional (claudin-1, occludin and ZO-1) and cytoskeleton (actin) proteins improved immune homeostasis and intestinal barrier integrity. Besides, significantly reduced (p < 0.05) levels of the FITC-dextran marker in serum upon consumption of PFW directly confirmed the healthy status of the host gut.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India.
| | - Taruna Gupta
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India.
| | - Suman Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India.
| | - Rajeev Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India.
| |
Collapse
|
30
|
Kang X, Liang H, Luo Y, Li Z, He F, Han X, Zhang L. Streptococcus thermophilus MN-ZLW-002 Can Inhibit Pre-adipocyte Differentiation through Macrophage Activation. Biol Pharm Bull 2021; 44:316-324. [PMID: 33390424 DOI: 10.1248/bpb.b20-00335] [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] [Indexed: 11/22/2022]
Abstract
It is well documented that obesity and metabolic syndrome have a deep association with the intestinal immune system of the host animal. Recent studies indicate that some selected probiotics can modulate the immune responses of the host animal, thereby altering its lipid metabolism. However, the underlying mechanisms are still not fully understood. This study was conducted to investigate the possibility of probiotics to activate macrophages in the hosts, thus alter the differentiation of pre-adipocytes. In this study, Streptococcus thermophilus MN-ZLW-002 (MN-ZLW-002) was co-cultured with RAW264.7 macrophages, with Lactobacillus rhamnosus GG (LGG) as a control. The conditioned medium (CM) of the co-culture was collected and then added to 3T3-L1 pre-adipocytes. Viable and heat-killed (80 °C, 30 min) MN-ZLW-002 stimulated RAW264.7 cells to produce significant amounts of interleukin (IL)-6 and tumor necrosis factor (TNF)-α and induced intense phosphorylation of P38, p44/42 mitogen-activated protein kinase (MAPK) (extracellular signal-regulated kinase (ERK)) and nuclear factor κB (NF-κB). Cytokine production reduced dramatically when heat-killed MN-ZLW-002 was treated with Ribonuclease. Viable and heat-killed LGG induced less cytokine production and little signaling protein activation. Viable and heat-killed MN-ZLW-002-stimulated RAW264.7-CM notably suppressed pre-adipocytes differentiation. However, viable LGG-stimulated RAW264.7-CM had a weaker effect and heat-killed LGG-stimulated RAW264.7-CM had no effect. These findings suggest that viable and heat-killed (80 °C, 30 min) MN-ZLW-002 may alter its lipid metabolism by regulating its immune response, possibly via the release of cytokine, particularly TNF-α. The RNA of heat-killed MN-ZLW-002 may be a key component in its immune activation effect.
Collapse
Affiliation(s)
- Xiaohong Kang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology
| | - Huijing Liang
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health and West China Forth Hospital, Sichuan University
| | - Yating Luo
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health and West China Forth Hospital, Sichuan University
| | - Zhouyong Li
- R&D Center, Inner Mongolia Meng Niu Dairy Industry (Group) Co., Ltd
| | - Fang He
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health and West China Forth Hospital, Sichuan University
| | - Xue Han
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology
| | - Lanwei Zhang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology.,College of Food Science and Engineering, Ocean University of China
| |
Collapse
|
31
|
Characterization and function of membrane vesicles in Gram-positive bacteria. Appl Microbiol Biotechnol 2021; 105:1795-1801. [PMID: 33547922 DOI: 10.1007/s00253-021-11140-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/15/2022]
Abstract
In recent years, extracellular vesicles have gained more attention. However, studies on membrane vesicles in Gram-positive bacteria were carried out relatively late because of the thick bacterial wall and the low production of membrane vesicles. Thanks to the research in recent years, the cognition of the composition and function of the membrane vesicles of Gram-positive bacteria has made significant progress. Membrane vesicles are spherical in shape comprising bilayer membranous structures with a diameter of 20-400 nm. Components of membrane vesicles are diverse, including proteins, nucleic acids, lipids, and metabolites. It also has been reported that membrane vesicles are involved in various pathophysiological processes and serve as communication tools in pathophysiological activities between the bacteria and the host. This review provided the current understanding of components and functions of membrane vesicles in Gram-positive bacteria. The findings might facilitate further research in the emerging field of membrane vesicles in Gram-positive bacteria. KEY POINTS: • Membrane vesicles in Gram-positive bacteria contain proteins, nucleic acids, lipids, and metabolites, suggesting their biological significance. • Membrane vesicles in Gram-positive bacteria are thought to be involved in stress response, biofilm formation, immune regulation, and so on.
Collapse
|
32
|
Galler KM, Weber M, Korkmaz Y, Widbiller M, Feuerer M. Inflammatory Response Mechanisms of the Dentine-Pulp Complex and the Periapical Tissues. Int J Mol Sci 2021; 22:ijms22031480. [PMID: 33540711 PMCID: PMC7867227 DOI: 10.3390/ijms22031480] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 02/08/2023] Open
Abstract
The macroscopic and microscopic anatomy of the oral cavity is complex and unique in the human body. Soft-tissue structures are in close interaction with mineralized bone, but also dentine, cementum and enamel of our teeth. These are exposed to intense mechanical and chemical stress as well as to dense microbiologic colonization. Teeth are susceptible to damage, most commonly to caries, where microorganisms from the oral cavity degrade the mineralized tissues of enamel and dentine and invade the soft connective tissue at the core, the dental pulp. However, the pulp is well-equipped to sense and fend off bacteria and their products and mounts various and intricate defense mechanisms. The front rank is formed by a layer of odontoblasts, which line the pulp chamber towards the dentine. These highly specialized cells not only form mineralized tissue but exert important functions as barrier cells. They recognize pathogens early in the process, secrete antibacterial compounds and neutralize bacterial toxins, initiate the immune response and alert other key players of the host defense. As bacteria get closer to the pulp, additional cell types of the pulp, including fibroblasts, stem and immune cells, but also vascular and neuronal networks, contribute with a variety of distinct defense mechanisms, and inflammatory response mechanisms are critical for tissue homeostasis. Still, without therapeutic intervention, a deep carious lesion may lead to tissue necrosis, which allows bacteria to populate the root canal system and invade the periradicular bone via the apical foramen at the root tip. The periodontal tissues and alveolar bone react to the insult with an inflammatory response, most commonly by the formation of an apical granuloma. Healing can occur after pathogen removal, which is achieved by disinfection and obturation of the pulp space by root canal treatment. This review highlights the various mechanisms of pathogen recognition and defense of dental pulp cells and periradicular tissues, explains the different cell types involved in the immune response and discusses the mechanisms of healing and repair, pointing out the close links between inflammation and regeneration as well as between inflammation and potential malignant transformation.
Collapse
Affiliation(s)
- Kerstin M. Galler
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, 93093 Regensburg, Germany;
- Correspondence:
| | - Manuel Weber
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Yüksel Korkmaz
- Department of Periodontology and Operative Dentistry, University of Mainz, 55131 Mainz, Germany;
| | - Matthias Widbiller
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, 93093 Regensburg, Germany;
| | - Markus Feuerer
- Department for Immunology, University Hospital Regensburg, 93053 Regensburg, Germany;
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
33
|
Keogh CE, Rude KM, Gareau MG. Role of pattern recognition receptors and the microbiota in neurological disorders. J Physiol 2021; 599:1379-1389. [PMID: 33404072 DOI: 10.1113/jp279771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, the gut microbiota has been increasingly implicated in the development of many extraintestinal disorders, including neurodevelopmental and neurodegenerative disorders. Despite this growing connection, our understanding of the precise mechanisms behind these effects is currently lacking. Pattern recognition receptors (PRRs) are important innate immune proteins expressed on the surface and within the cytoplasm of a multitude of cells, both immune and otherwise, including epithelial, endothelial and neuronal. PRRs comprise four major subfamilies: the Toll-like receptors (TLRs), the nucleotide-binding oligomerization domain leucine rich repeats-containing receptors (NLRs), the retinoic acid inducible gene 1-like receptors and the C-type lectin receptors. Recognition of commensal bacteria by PRRs is critical for maintaining host-microbe interactions and homeostasis, including behaviour. The expression of PRRs on multiple cell types makes them a highly interesting and novel target for regulation of host-microbe signalling, which may lead to gut-brain signalling. Emerging evidence indicates that two of the four known families of PRRs (the NLRs and the TLRs) are involved in the pathogenesis of neurodevelopmental and neurodegenerative disorders via the gut-brain axis. Taken together, increasing evidence supports a role for these PRRs in the development of neurological disorders, including Alzheimer's disease, Parkinson's disease and multiple sclerosis, via the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Ciara E Keogh
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Kavi M Rude
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Mélanie G Gareau
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| |
Collapse
|
34
|
Inatomi T, Otomaru K. Effects of heat-killed Enterococcus faecalis T-110 supplementation on gut immunity, gut flora, and intestinal infection in naturally aged hamsters. PLoS One 2020; 15:e0240773. [PMID: 33378402 PMCID: PMC7773277 DOI: 10.1371/journal.pone.0240773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/14/2020] [Indexed: 12/25/2022] Open
Abstract
Infectious diseases are a threat to elderly individuals, whose immune systems weaken with age. Among the various infectious diseases, Clostridium difficile infection is associated with a high rate of mortality in elderly individuals and is a serious health problem worldwide, owing to the increasing infection rates. Probiotic use has been proposed as an effective countermeasure for C. difficile infection. The aim of this study was to evaluate the effects of heat-killed Enterococcus faecalis T-110 on intestinal immunity, intestinal flora, and intestinal infections, especially C. difficile infections, in naturally ageing animals, for extrapolating the results to elderly human subjects. Twenty female hamsters were randomly distributed into two groups. Group 1 was fed a basal diet and group 2 was fed a basal diet supplemented with heat-killed E. faecalis for 7 days. Heat-killed E. faecalis T-110 improved the gut immunity and microflora, especially Clostridium perfringens and C. difficile, in naturally aged hamsters. Therefore, heat-killed E. faecalis T-110 use may be a countermeasure against age-related immune dysfunction and intestinal infections, especially C. difficile infection, in elderly humans. However, further investigation in this regard is needed in humans.
Collapse
Affiliation(s)
| | - Konosuke Otomaru
- Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima, Japan
| |
Collapse
|
35
|
Kang X, Liang H, Luo Y, Li Z, He F, Han X, Zhang L. Anti-adipogenesis and metabolism-regulating effects of heat-inactivated Streptococcus thermophilus MN-ZLW-002. Lett Appl Microbiol 2020; 72:677-687. [PMID: 32981107 DOI: 10.1111/lam.13398] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/27/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome and obesity have become serious threats to public health worldwide. This study was conducted to evaluate the anti-adipogenesis and metabolism-regulating effects of heat-inactivated Streptococcus thermophilus MN-ZLW-002 (MN-ZLW-002), which can be used as a yogurt starter. In vitro study suggested that MN-ZLW-002 stimulated the RAW264.7 macrophages to produce significant amounts of interleukin (IL)-6, IL-10 and tumour necrosis factor (TNF)-α and induced intense phosphorylation of P38, p44/42 MAPK and nuclear factor κB. MN-ZLW-002-stimulated RAW264.7-conditioned medium (CM) notably suppressed the differentiation and adipogenesis of 3T3-L1 pre-adipocytes. The 12-week in vivo study suggested that orally administered MN-ZLW-002 significantly reduced the weight gain of mice caused by the high-fat diet (HFD) at weeks 3-8; decreased fasting blood glucose levels at week 4 and week 8; decreased serum total triglyceride level at week 12. MN-ZLW-002 also reduced serum IL-1β and chemokine ligand 3 levels in the HFD-fed mice. These findings suggest that heat-inactivated MN-ZLW-002 can suppress adipocytes differentiation and lipid accumulation by regulating the immune response, possibly via the release of cytokines, particularly TNF-α; MN-ZLW-002 can improve metabolism-related indicators in the early stage of HFD intervention and regulate the related pro-inflammatory immune response.
Collapse
Affiliation(s)
- X Kang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - H Liang
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health and West China Forth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Y Luo
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health and West China Forth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Z Li
- R&D Center, Inner Mongolia Meng Niu Dairy Industry (Group) Co., Ltd, Hohhot, Inner Mongolia, China
| | - F He
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health and West China Forth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - X Han
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - L Zhang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, China.,College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| |
Collapse
|
36
|
Lactococcus lactis subsp. Cremoris C60 restores T Cell Population in Small Intestinal Lamina Propria in Aged Interleukin-18 Deficient Mice. Nutrients 2020; 12:nu12113287. [PMID: 33121026 PMCID: PMC7693701 DOI: 10.3390/nu12113287] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022] Open
Abstract
Lactic acid bacteria (LAB), a major commensal bacterium in the small intestine, are well known beneficial bacteria which promote establishment of gut-centric immunity, such as anti-inflammation and anti-infection. In this report, we show that a LAB strain Lactococcus lactis subsp. Cremoris C60 possess an ability to activate antigen presenting cells, such as dendritic cells (DCs), and intestinal T cells which possibly support to maintain healthy intestinal immunological environment in aging process. We found that CD4+ T cells in the small intestine are dramatically decreased in aged Interleukin-18 knock out (IL-18KO) mice, associated with the impairment of IFN-γ production in the CD4+ T cells, especially in small intestinal lamina propria (LP). Surprisingly, heat killed-C60 (HK-C60) diet completely recovered the CD4+ T cells population and activity in SI-LP and over activated the population in Peyer's patches (PPs) of IL-18KO mice. The HK-C60 diet was effective approach not only to restore the number of cells, but also to recover IFN-γ production in the CD4+ T cell population in the small intestine of IL-18-deficient mice. As a possible cause in the age-associated impairment of CD4+ T cells activity in IL-18KO mice, we found that the immunological activity was downregulated in the IL-18-deficient DCs. The cytokines production and cellular activation markers expression were downregulated in the IL-18-deficient bone marrow derived dendritic cells (BMDCs) at the basal level, however, both activities were highly upregulated in HK-C60 stimulation as compared to those of WT cells. Antigen uptake was also attenuated in the IL-18-deficient BMDCs, and it was significantly enhanced in the cells as compared to WT cells in HK-60 stimulation. An in vitro antigen presentation assay showed that IFN-γ production in the CD4+ T cells was significantly enhanced in the culture of IL-18-deficient BMDCs compared with WT cells in the presence of HK-C60. Thus, we conclude that HK-C60 diet possesses an ability to restore T cells impairment in the small intestine of IL-18-deficient environment. In addition, the positive effect is based on the immunological modification of DCs function which directory influences into the promotion of effector CD4+ T cells generation in the small intestine.
Collapse
|
37
|
Lockyer S, Aguirre M, Durrant L, Pot B, Suzuki K. The role of probiotics on the roadmap to a healthy microbiota: a symposium report. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2020; 1:e2. [PMID: 39296722 PMCID: PMC11406418 DOI: 10.1017/gmb.2020.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/23/2020] [Accepted: 05/01/2020] [Indexed: 09/21/2024]
Abstract
The ninth International Yakult Symposium was held in Ghent, Belgium in April 2018. Keynote lectures were from Professor Wijmenga on using biobanks to understand the relationship between the gut microbiota and health; and Professor Hill on phage-probiotic interactions. Session one included talks from Professor Plӧsch on epigenetic programming by nutritional and environmental factors; Professor Wilmes on the use of "omics" methodologies in microbiome research and Professor Rescigno on the gut vascular barrier. Session two explored the evidence behind Lactobacillus casei Shirota with Dr Nanno explaining the plasticity in immunomodulation that enables the strain to balance immune functions; Dr Macnaughtan outlining its potential therapeutic use in cirrhosis and Professor Nishida detailing effects in subjects under stress. The third session saw Professor Marchesi describing that both the host genes and the gut microbiota can play a role in cancer; Professor Bergheim highlighting crosstalk between the gut and the liver and Professor Cani describing the relationship between the gut microbiota and the endocrine system. The final session explored probiotic mechanisms, with Professor Lebeer dissecting the challenges in conducting mechanistic studies; Professor Wehkamp describing the mucosal defence system and Professor Van de Wiele detailing methods for modelling the gut microbiota in vitro.
Collapse
Affiliation(s)
| | | | | | - Bruno Pot
- Yakult Europe B.V., Almere, The Netherlands
| | | |
Collapse
|
38
|
Wang B, Wu Y, Liu R, Xu H, Mei X, Shang Q, Liu S, Yu D, Li W. Lactobacillus rhamnosus GG promotes M1 polarization in murine bone marrow-derived macrophages by activating TLR2/MyD88/MAPK signaling pathway. Anim Sci J 2020; 91:e13439. [PMID: 32779289 DOI: 10.1111/asj.13439] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/23/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022]
Abstract
Lactobacillus rhamnosus GG (LGG) is increasingly applied in functional food products and acts as a probiotic model in nutritious and clinical studies. Increasing evidences have revealed the immune modulation of LGG on macrophages. The aim of this study is to investigate the effect of LGG on macrophage polarization of murine bone marrow-derived macrophages (BMDMs). BMDMs were treated with 108 colony-forming units (CFU)/ml LGG for 1.5, 3, and 6 hr. Results showed that LGG obviously upregulated the mRNA expression of M1-associated cytokines (p < .05), including interleukin-1 beta (IL-1β), IL-6, tumor necrosis factor-alpha (TNF-α), and inducible nitric oxide synthase (iNOS), whereas had no effect on the expression of M2-associated markers (p > .05), including arginase 1 (Arg1), mannose receptor, and chitinase-like protein 3 (YM1). Furthermore, LGG markedly increased the expression of pro-inflammatory cytokines (IL-12p40, cyclooxygenase-2 [COX-2], and interferon-γ [IFN-γ]) (p < .05) and anti-inflammatory cytokines (IL-10, IL-4, and transforming growth factor-β [TGF-β]) (p < .05). In addition, we also found that TLR2/MyD88/MAPK signaling pathway was required for LGG-induced M1 macrophage polarization and M1-related cytokines expression. Together, these findings demonstrate that probiotic LGG facilitates M1 polarization of BMDMs, suggesting that LGG may have an immunotherapeutic potential in regulating the host defense against pathogen invasion.
Collapse
Affiliation(s)
- Baikui Wang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China) of the Ministry of Agriculture, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yanping Wu
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China) of the Ministry of Agriculture, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Rongrong Liu
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China) of the Ministry of Agriculture, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Han Xu
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China) of the Ministry of Agriculture, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoqiang Mei
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China) of the Ministry of Agriculture, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qinqin Shang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China) of the Ministry of Agriculture, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Shijie Liu
- National Animal Husbandry Service, Beijing, China
| | - Dongyou Yu
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China) of the Ministry of Agriculture, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China) of the Ministry of Agriculture, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Żółkiewicz J, Marzec A, Ruszczyński M, Feleszko W. Postbiotics-A Step Beyond Pre- and Probiotics. Nutrients 2020; 12:E2189. [PMID: 32717965 PMCID: PMC7468815 DOI: 10.3390/nu12082189] [Citation(s) in RCA: 290] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
As an imbalance in the intestinal microbiota can lead to the development of several diseases (e.g., type 1 diabetes, cancer, among others), the use of prebiotics, probiotics, and postbiotics to alter the gut microbiome has attracted recent interest. Postbiotics include any substance released by or produced through the metabolic activity of the microorganism, which exerts a beneficial effect on the host, directly or indirectly. As postbiotics do not contain live microorganisms, the risks associated with their intake are minimized. Here, we provided a critical review of postbiotics described in the literature, including their mechanisms of action, clinical characteristics, and potential therapeutic applications. We detailed the pleiotropic effects of postbiotics, including their immunomodulatory, anti-inflammatory, antioxidant, and anti-cancer properties. Although the use of postbiotics is an attractive strategy for altering the microbiome, further study into its efficacy and safety is warranted.
Collapse
Affiliation(s)
- Jakub Żółkiewicz
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Żwirki i Wigury 63A, 02-091 Warsaw, Poland; (J.Ż.); (A.M.)
| | - Aleksandra Marzec
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Żwirki i Wigury 63A, 02-091 Warsaw, Poland; (J.Ż.); (A.M.)
| | - Marek Ruszczyński
- Department of Paediatrics, Medical University of Warsaw, Żwirki i Wigury 63A, 02-091 Warsaw, Poland;
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Żwirki i Wigury 63A, 02-091 Warsaw, Poland; (J.Ż.); (A.M.)
| |
Collapse
|
40
|
Total RNA and genomic DNA of Lactobacillus gasseri OLL2809 induce interleukin-12 production in the mouse macrophage cell line J774.1 via toll-like receptors 7 and 9. BMC Microbiol 2020; 20:217. [PMID: 32689952 PMCID: PMC7372888 DOI: 10.1186/s12866-020-01900-w] [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: 02/09/2020] [Accepted: 07/12/2020] [Indexed: 12/04/2022] Open
Abstract
Background Lactobacillus gasseri OLL2809 can highly induce interleukin (IL)-12 production in immune cells. Even though beneficial properties of this strain for both humans and animals have been reported, the mechanism by which the bacteria induces the production of IL-12 in immune cells remains elusive. In this study, we investigated the mechanism of induction of IL-12 using a mouse macrophage cell line J774.1. Results Inhibition of phagocytosis of L. gasseri OLL2809, and myeloid differentiation factor 88 and Toll-like receptors (TLRs) 7 and 9 signalling attenuated IL-12 production in J774.1 cells. Total RNA and genomic DNA of L. gasseri OLL2809, when transferred to the J774.1 cells, also induced IL-12 production. The difference in the IL-12-inducing activity of Lactobacilli is attributed to the susceptibility to phagocytosis, but not to a difference in the total RNA and genomic DNA of each strain. Conclusion We concluded that total RNA and genomic DNA of phagocytosed L. gasseri OLL2809 induce IL-12 production in J774.1 cell via TLRs 7 and 9, and the high IL-12-inducing activity of L. gasseri OLL2809 is due to its greater susceptibility to phagocytosis.
Collapse
|
41
|
Ren C, Faas MM, de Vos P. Disease managing capacities and mechanisms of host effects of lactic acid bacteria. Crit Rev Food Sci Nutr 2020; 61:1365-1393. [PMID: 32366110 DOI: 10.1080/10408398.2020.1758625] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Consumption of lactic acid bacteria (LAB) has been suggested to confer health-promoting effects on the host. However, effects of LABs have been reported to be species- and strain-specific and the mechanisms involved are subjects of discussion. Here, the possible mechanisms by which LABs induce antipathogenic, gut barrier enhancing and immune modulating effects in consumers are reviewed. Specific strains for which it has been proven that health is improved by these mechanisms are discussed. However, most strains probably act via several or combinations of mechanisms depending on which effector molecules they express. Current insight is that these effector molecules are either present on the cell wall of LAB or are excreted. These molecules are reviewed as well as the ligand binding receptors in the host. Also postbiotics are discussed. Finally, we provide an overview of the efficacy of LABs in combating infections caused by Helicobacter pylori, Salmonella, Escherichia coli, Streptococcus pneumoniae, and influenza virus, in controlling gut inflammatory diseases, in managing allergic disorders, and in alleviating cancer.
Collapse
Affiliation(s)
- Chengcheng Ren
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Marijke M Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
42
|
Takano T, Endo R, Wang Y, Nakajima-Adachi H, Hachimura S. Lactobacillus plantarum OLL2712 induces IL-10 production by intestinal dendritic cells. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2020; 39:39-44. [PMID: 32328399 PMCID: PMC7162692 DOI: 10.12938/bmfh.19-019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/10/2019] [Indexed: 12/27/2022]
Abstract
Recently many researchers have revealed that certain lactic acid bacteria (LAB) have beneficial effects on the immune system. Understanding the mechanisms of how certain LAB induce
immunomodulatory functions is important for the development of food ingredients that improve our health. Lactobacillus plantarum OLL2712 has been shown to induce production
of interleukin (IL)-10, an anti-inflammatory cytokine, by murine in vitro-induced dendritic cells (DCs) and peritoneal macrophages. However, it is probable that in
vitro-induced DCs have different properties compared with intestinal DCs, and the effects of the LAB on intestinal DCs are not fully understood. In this report, we investigated
whether L. plantarum OLL2712 had efficacy for inducing intestinal DCs to produce IL-10 in vitro and whether oral administration of the bacteria induced the
same effect. Co-culture of L. plantarum OLL2712 with purified DCs from the mesenteric lymph node (MLN) or Peyer’s patch (PP) elevated IL-10 mRNA expression and protein
production by both kinds of DCs. Addition of the LAB enhanced IL-10 production by T cells during antigen-specific responses in co-culture of MLN or PP DCs and T cells. Oral administration of
L. plantarum OLL2712 for 6 days increased IL-10 gene expression in MLN DCs, and upregulated IL-10 gene expression in PP DCs was observed 12 hr after oral administration of
the LAB. Our results suggested that L. plantarum OLL2712 could modulate immune responses by enhancing IL-10 production from intestinal DCs.
Collapse
Affiliation(s)
- Tomohiro Takano
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.,Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ryunosuke Endo
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.,Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yimei Wang
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.,Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Haruyo Nakajima-Adachi
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.,Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Satoshi Hachimura
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.,Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
43
|
Lactic acid bacteria secrete toll like receptor 2 stimulating and macrophage immunomodulating bioactive factors. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103783] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
44
|
Hernandez GV, Smith VA, Melnyk M, Burd MA, Sprayberry KA, Edwards MS, Peterson DG, Bennet DC, Fanter RK, Columbus DA, Steibel JP, Glanz H, Immoos C, Rice MS, Santiago-Rodriguez TM, Blank J, VanderKelen JJ, Kitts CL, Piccolo BD, La Frano MR, Burrin DG, Maj M, Manjarin R. Dysregulated FXR-FGF19 signaling and choline metabolism are associated with gut dysbiosis and hyperplasia in a novel pig model of pediatric NASH. Am J Physiol Gastrointest Liver Physiol 2020; 318:G582-G609. [PMID: 32003601 PMCID: PMC7099491 DOI: 10.1152/ajpgi.00344.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 02/08/2023]
Abstract
To investigate the role of bile acids (BAs) in the pathogenesis of diet-induced nonalcoholic steatohepatitis (NASH), we fed a "Western-style diet" [high fructose, high fat (HFF)] enriched with fructose, cholesterol, and saturated fat for 10 wk to juvenile Iberian pigs. We also supplemented probiotics with in vitro BA deconjugating activity to evaluate their potential therapeutic effect in NASH. Liver lipid and function, cytokines, and hormones were analyzed using commercially available kits. Metabolites, BAs, and fatty acids were measured by liquid chromatography-mass spectrometry. Histology and gene and protein expression analyses were performed using standard protocols. HFF-fed pigs developed NASH, cholestasis, and impaired enterohepatic Farnesoid-X receptor (FXR)-fibroblast growth factor 19 (FGF19) signaling in the absence of obesity and insulin resistance. Choline depletion in HFF livers was associated with decreased lipoprotein and cholesterol in serum and an increase of choline-containing phospholipids in colon contents and trimethylamine-N-oxide in the liver. Additionally, gut dysbiosis and hyperplasia increased with the severity of NASH, and were correlated with increased colonic levels of choline metabolites and secondary BAs. Supplementation of probiotics in the HFF diet enhanced NASH, inhibited hepatic autophagy, increased excretion of taurine and choline, and decreased gut microbial diversity. In conclusion, dysregulation of BA homeostasis was associated with injury and choline depletion in the liver, as well as increased biliary secretion, gut metabolism and excretion of choline-based phospholipids. Choline depletion limited lipoprotein synthesis, resulting in hepatic steatosis, whereas secondary BAs and choline-containing phospholipids in colon may have promoted dysbiosis, hyperplasia, and trimethylamine synthesis, causing further damage to the liver.NEW & NOTEWORTHY Impaired Farnesoid-X receptor (FXR)-fibroblast growth factor 19 (FGF19) signaling and cholestasis has been described in nonalcoholic fatty liver disease (NAFLD) patients. However, therapeutic interventions with FXR agonists have produced contradictory results. In a swine model of pediatric nonalcoholic steatohepatitis (NASH), we show that the uncoupling of intestinal FXR-FGF19 signaling and a decrease in FGF19 levels are associated with a choline-deficient phenotype of NASH and increased choline excretion in the gut, with the subsequent dysbiosis, colonic hyperplasia, and accumulation of trimethylamine-N-oxide in the liver.
Collapse
Affiliation(s)
- Gabriella V Hernandez
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Victoria A Smith
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Megan Melnyk
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California
| | - Matthew A Burd
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Kimberly A Sprayberry
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Mark S Edwards
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Daniel G Peterson
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Darin C Bennet
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Rob K Fanter
- Center for Health Research, California Polytechnic State University, San Luis Obispo, California
| | | | - Juan P Steibel
- Department of Animal Science and Department of Fisheries and Wildlife, Michigan State University, East Lansing, Michigan
| | - Hunter Glanz
- Department of Statistics, California Polytechnic State University, San Luis Obispo, California
| | - Chad Immoos
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California
| | - Margaret S Rice
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California
| | | | - Jason Blank
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California
| | - Jennifer J VanderKelen
- Center for Applications in Biotechnology, California Polytechnic State University, San Luis Obispo, California
| | - Christopher L Kitts
- Center for Applications in Biotechnology, California Polytechnic State University, San Luis Obispo, California
| | - Brian D Piccolo
- United States Department of Agriculture-Agricultural Research Services, Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Michael R La Frano
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, California
- Center for Health Research, California Polytechnic State University, San Luis Obispo, California
| | - Douglas G Burrin
- United States Department of Agriculture-Agricultural Research Services, Children's Nutrition Research Center, Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Magdalena Maj
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California
- Center for Applications in Biotechnology, California Polytechnic State University, San Luis Obispo, California
| | - Rodrigo Manjarin
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| |
Collapse
|
45
|
Rocha-Ramírez LM, Hernández-Ochoa B, Gómez-Manzo S, Marcial-Quino J, Cárdenas-Rodríguez N, Centeno-Leija S, García-Garibay M. Evaluation of Immunomodulatory Activities of the Heat-Killed Probiotic Strain Lactobacillus casei IMAU60214 on Macrophages In Vitro. Microorganisms 2020; 8:microorganisms8010079. [PMID: 31936101 PMCID: PMC7022880 DOI: 10.3390/microorganisms8010079] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/02/2020] [Accepted: 01/05/2020] [Indexed: 12/16/2022] Open
Abstract
Most Lactobacillus species have beneficial immunological (“immunoprobiotic”) effects in the host. However, it is unclear how probiotic bacteria regulate immune responses. The present study investigated the effects of heat-killed Lactobacillus casei IMAU60214 on the activity of human monocyte-derived macrophages (MDMs). Human MDMs were treated with heat-killed L. casei at a ratio (bacteria/MDM) of 50:1, 100:1, 250:1, and 500:1, and then evaluated for the following: NO production, by Griess reaction; phagocytosis of FITC-labeled Staphylococcus aureus particles; cytokine secretion profile (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-12p70, IL-10, and transforming growth factor (TGF)-β) by ELISA; and costimulatory molecule (CD80 and CD86) surface expression, by flow cytometry. Heat-killed L. casei IMAU60214 enhanced phagocytosis, NO production, cytokine release, and surface expression of CD80 and CD86 in a dose-dependent manner. All products were previously suppressed by pretreatment with a Toll-like receptor 2 (TLR2)-neutralizing antibody. Overall, our findings suggest that this probiotic strain promotes an M1-like pro-inflammatory phenotype through the TLR2 signaling pathway. These effects on macrophage phenotype help explain the probiotic efficacy of Lactobacillus and provide important information for the selection of therapeutic targets and treatments compatible with the immunological characteristics of this probiotic strain.
Collapse
Affiliation(s)
- Luz María Rocha-Ramírez
- Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Secretaría de Salud Dr. Márquez No. 162, Col Doctores, Delegación Cuauhtémoc, Ciudad de México 06720, Mexico
- Correspondence: ; Tel.: +52-55-5228-9917 (ext. 2084)
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica y Biología Celular, Hospital Infantil de México Federico Gómez, Secretaría de Salud. Dr. Márquez No. 162, Col Doctores, Delegación Cuauhtémoc, Ciudad de México 06720, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaria de Salud, Ciudad de México 04530, Mexico;
| | - Jaime Marcial-Quino
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | - Sara Centeno-Leija
- Consejo Nacional Ciencia y Tecnologia (CONACYT) Laboratorio de Agrobiotecnología, Tecnoparque CLQ, Universidad de Colima, Carretera Los Limones-Loma de Juárez, Colima 28629, Mexico;
| | - Mariano García-Garibay
- Departamento de Ciencias de la Alimentación, Unidad Lerma, Universidad Autónoma Metropolitana, Av. San Rafael Atlixco No. 186. Col Vicentina, Ciudad de México 09340, Mexico;
| |
Collapse
|
46
|
Lin C, Zhao S, Zhu Y, Fan Z, Wang J, Zhang B, Chen Y. Microbiota-gut-brain axis and toll-like receptors in Alzheimer's disease. Comput Struct Biotechnol J 2019; 17:1309-1317. [PMID: 31921396 PMCID: PMC6944716 DOI: 10.1016/j.csbj.2019.09.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 09/19/2019] [Accepted: 09/22/2019] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial disease which involves both the periphery and central nervous system (CNS). It has been recently recognized that gut microbiota interacts with the gut and brain (microbiota-gut-brain axis), contributing to the pathogenesis of neurodegenerative diseases, such as AD. Dysbiosis of gut microbiota can induce increased intestinal permeability and systemic inflammation, which may lead to the development of AD pathologies and cognitive impairment via the neural, immune, endocrine, and metabolic pathways. Toll-like receptors (TLRs) play an important role in the innate immune system via recognizing microbes-derived pathogens and initiating the inflammatory process. TLRs have also been found in the brain, especially in the microglia, and have been indicated in the development of AD. In this review, we summarized the relationship between microbiota-gut-brain axis and AD, as well as the complex role of TLRs in AD. Intervention of the gut microbiota or modulation of TLRs properly might emerge as promising preventive and therapeutic strategies for AD.
Collapse
Affiliation(s)
- Caixiu Lin
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuai Zhao
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yueli Zhu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziqi Fan
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Wang
- Department of Geriatric, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanxing Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
47
|
Fawzy El-Sayed KM, Elsalawy R, Ibrahim N, Gadalla M, Albargasy H, Zahra N, Mokhtar S, El Nahhas N, El Kaliouby Y, Dörfer CE. The Dental Pulp Stem/Progenitor Cells-Mediated Inflammatory-Regenerative Axis. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:445-460. [DOI: 10.1089/ten.teb.2019.0106] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Karim M. Fawzy El-Sayed
- Oral Medicine and Periodontology Department, Faculty of Oral and Dental Medicine, Cairo University, Cairo, Egypt
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| | | | | | | | | | - Nehal Zahra
- Faculty of Dentistry, New Giza University, Giza, Egypt
| | | | | | | | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| |
Collapse
|
48
|
Sandagdorj B, Hamajima C, Kawahara T, Watanabe J, Tanaka S. Characterization of Microbiota that Influence Immunomodulatory Effects of Fermented Brassica rapa L. Microbes Environ 2019; 34:206-214. [PMID: 31167991 PMCID: PMC6594740 DOI: 10.1264/jsme2.me19003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/09/2019] [Indexed: 02/06/2023] Open
Abstract
Lactic acid bacteria (LAB) exert beneficial health effects by regulating immune responses. Brassica rapa L., known as Nozawana, is commonly consumed as a lactic acid-fermented food called nozawana-zuke. Few studies have investigated changes in the bacterial community and cytokine production activities during the fermentation of B. rapa L. In order to obtain more detail information, we herein conducted a study on fresh B. rapa L. fermented for 28 d. An amplicon analysis of the 16S rRNA gene revealed that Lactobacillales predominated during fermentation, and the microbiota became less diverse on day 7 or later. Fermented B. rapa L. promoted the production of interferon (IFN)-γ and interleukin (IL)-10 by mouse spleen cells more than non-fermented vegetables. Lactobacillus curvatus was the predominant species during fermentation, followed by L. plantarum and L. brevis. L. sakei was occasionally detected. A correlation analysis showed that IFN-γ concentrations positively correlated with the numbers of L. curvatus and L. plantarum, while those of IL-10 correlated with the numbers of L. sakei in addition to these 2 species. Significantly higher levels of IFN-γ and IL-10 were induced by fermented B. rapa L. when isolated Lactobacillus strains were added as starter cultures. These results suggest that the Lactobacillus species present in fermented B. rapa L. are beneficial for manufacturing vegetables with immunomodulatory effects.
Collapse
Affiliation(s)
- Bayanjargal Sandagdorj
- Food Research Institute, National Agriculture and Food Research OrganizationTsukuba, Ibaraki 305–8642Japan
- School of Integrative and Global Majors, University of TsukubaTsukuba, 305–8577Japan
| | - Chisato Hamajima
- Academic Assembly (Institute of Agriculture), Shinshu UniversityMinamiminowa, Nagano 399–4598Japan
| | - Takeshi Kawahara
- Academic Assembly (Institute of Agriculture), Shinshu UniversityMinamiminowa, Nagano 399–4598Japan
- Supramolecular Complexes Unit, Research Center for Fungal and Microbial Dynamism, Shinshu UniversityMinamiminowa, Nagano 399–4598Japan
| | - Jun Watanabe
- Food Research Institute, National Agriculture and Food Research OrganizationTsukuba, Ibaraki 305–8642Japan
- School of Integrative and Global Majors, University of TsukubaTsukuba, 305–8577Japan
| | - Sachi Tanaka
- Academic Assembly (Institute of Agriculture), Shinshu UniversityMinamiminowa, Nagano 399–4598Japan
- Supramolecular Complexes Unit, Research Center for Fungal and Microbial Dynamism, Shinshu UniversityMinamiminowa, Nagano 399–4598Japan
| |
Collapse
|
49
|
Tang C, Lu Z. Health promoting activities of probiotics. J Food Biochem 2019; 43:e12944. [PMID: 31368544 DOI: 10.1111/jfbc.12944] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022]
Abstract
In recent years, probiotics have received increasing attention and become one type of popular functional food because of their many biological functions. Among these desirable biological functions, the immune regulation, antioxidative activities, and antimicrobial effects are essential properties to maintain host health. Probiotics can regulate the immune system and improve the antioxidative system by producing microbial components and metabolites. Meanwhile, probiotics also possess antimicrobial abilities owing to their competition for nutrient requirements and mucus adherence, reducing pathogenic toxins, producing antimicrobial metabolites (short-chain fatty acids, bacteriocins, reuterin, linoleic acid, and secondary bile acids) and enhancing intestinal, or systemic immunity. Therefore, probiotics could be used to alleviate heavy metal toxicity and metabolic disorders by improving immunity, the antioxidative system, and intestinal micro-environment. This comprehensive review mainly highlights the potential health promoting activities of probiotics based on their antioxidative, antimicrobial, and immune regulatory effects. PRACTICAL APPLICATIONS: The antioxidative defense and the immune system are essential to maintain human health. However, many factors may result in microbial dysbiosis in the gut, which subsequently leads to pathogenic expansion, oxidative stress, and inflammatory responses. Therefore, it is important to explore beneficial foods to prevent or suppress these abnormal responses. Successful application of probiotics in the functional foods has attracted increasing attention due to their immune regulatory, antioxidative, and antimicrobial properties. The aim of this review is to introduce immune regulatory antioxidative and antimicrobial effects of probiotics, which provides some basic theories for scientific research and development of potential functional foods.
Collapse
Affiliation(s)
- Chao Tang
- Laboratory of Enzyme Engineering, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhaoxin Lu
- Laboratory of Enzyme Engineering, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
50
|
Surface Layer of Lactobacillus helveticus MIMLh5 Promotes Endocytosis by Dendritic Cells. Appl Environ Microbiol 2019; 85:AEM.00138-19. [PMID: 30824443 DOI: 10.1128/aem.00138-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/13/2019] [Indexed: 01/09/2023] Open
Abstract
Surface layers (S-layers) are proteinaceous arrays covering the cell walls of numerous bacteria. Their suggested properties, such as interactions with the host immune system, have been only poorly described. Here, we aimed to elucidate the role of the S-layer from the probiotic bacterial strain Lactobacillus helveticus MIMLh5 in the stimulation of murine bone-marrow-derived dendritic cells (DCs). MIMLh5 induced greater production of interferon beta (IFN-β), interleukin 10 (IL-10), and IL-12p70, compared to S-layer-depleted MIMLh5 (naked MIMLh5 [n-MIMLh5]), whereas the isolated S-layer was a poor immunostimulator. No differences in the production of tumor necrosis factor alpha (TNF-α) or IL-1β were found. Inhibition of the mitogen-activated protein kinases JNK1/2, p38, and ERK1/2 modified IL-12p70 production similarly in MIMLh5 and n-MIMLh5, suggesting the induction of the same signaling pathways by the two bacterial preparations. Treatment of DCs with cytochalasin D to inhibit endocytosis before the addition of fluorescently labeled MIMLh5 cells led to a dramatic reduction in the proportion of fluorescence-positive DCs and decreased IL-12 production. Endocytosis and IL-12 production were only marginally affected by cytochalasin D pretreatment when fluorescently labeled n-MIMLh5 was used. Treatment of DCs with fluorescently labeled S-layer-coated polystyrene beads (Sl-beads) resulted in much greater uptake of beads, compared to noncoated beads. Prestimulation of DCs with cytochalasin D reduced the uptake of Sl-beads more than plain beads. These findings indicate that the S-layer plays a role in the endocytosis of MIMLh5 by DCs. In conclusion, this study provides evidence that the S-layer of L. helveticus MIMLh5 is involved in endocytosis of the bacterium, which is important for strong Th1-inducing cytokine production.IMPORTANCE Beneficial microbes may positively affect host physiology at various levels, e.g., by participating in immune system maturation and modulation, boosting defenses and dampening reactions, thus affecting the whole homeostasis. As a consequence, the use of probiotics is increasingly regarded as suitable for more extended applications for health maintenance, not only microbiota balancing. This implies a deep knowledge of the mechanisms and molecules involved in host-microbe interactions, for the final purpose of fine tuning the choice of a probiotic strain for a specific outcome. With this aim, studies targeted to the description of strain-related immunomodulatory effects and the identification of bacterial molecules responsible for specific responses are indispensable. This study provides new insights in the characterization of the food-origin probiotic bacterium L. helveticus MIMLh5 and its S-layer protein as a driver for the cross-talk with DCs.
Collapse
|