1
|
Amar Y, Grube J, Köberle M, Schaubeck M, Biedermann T, Volz T. Bifidobacterium breve DSM 32583 and Limosilactobacillus fermentum CECT5716 postbiotics attenuate S. aureus and IL-33-induced Th2 responses. Microbiol Res 2024; 289:127913. [PMID: 39316930 DOI: 10.1016/j.micres.2024.127913] [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: 07/12/2024] [Revised: 09/04/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
Over the past decades, the prevalence of allergic diseases noticeably increased in industrialized countries. The Th2 immune response plays a central role in these pathologies and its modulation using pro-/postbiotics constitutes a promising approach to prevent or alleviate disease symptoms. The aim of this in vitro study, was to investigate the ability of human milk-derived Bifidobacterium breve DSM 32583 (Bb) and Limosilactobacillus fermentum CECT5716 (Lf), to modulate the Th2 induced responses. To this end, Th2 cells were generated by co-culturing of human naïve Th cells with monocyte-derived dendritic cells (moDCs) either stimulated with Staphylococcus aureus or IL-33. The immunomodulatory effects of pro-/postbiotic preparations of Bb and Lf on moDCs and Th2 cells were evaluated in terms of maturation markers expression and cytokines production. Remarkably, the tested strains induced the anti-inflammatory cytokine IL-10 in moDCs, in a strain-, dose- and viability-dependent manner with no significant upregulation of IL-12p70 nor CD83, CD86 or HLA-DR. Interestingly, Bb and Lf postbiotics were able to dampen the Th2/Th1 response induced upon S. aureus- or IL-33 stimulation. They were also able to synergistically induce IL-10 in moDCs and T cells, upon co-stimulation with LPS. Finally, we observed that live probiotics triggered a mild Th1 response that was attenuated in the presence of galacto-oligosaccharides. Altogether, Bb and Lf pro-/postbiotics exhibited remarkable immune regulatory effects on both moDCs and Th2 cells. Therefore, further in vivo studies should be considered to validate these findings and assess their ability to prevent allergy or alleviate its symptoms in affected patients.
Collapse
Affiliation(s)
- Yacine Amar
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany.
| | - Jana Grube
- HiPP GmbH & Co. Vertrieb KG, Pfaffenhofen (Ilm) 85276, Germany
| | - Martin Köberle
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | | | - Tilo Biedermann
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | - Thomas Volz
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| |
Collapse
|
2
|
Qin M, Xing Y, Sun M, Ma L, Li X, Ma F, Li D, Duan C. An Exploration of the Antioxidative and Anti-Inflammatory Role of Lactiplantibacillus plantarum 106 via Improving Mitochondrial Function. Foods 2024; 13:1981. [PMID: 38998487 PMCID: PMC11241742 DOI: 10.3390/foods13131981] [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: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
In this present study, bioinformatics analysis and the experimental validation method were used to systematically explore the antioxidant activity and anti-inflammatory effect of Lactiplantibacillus plantarum A106, which was isolated from traditional Chinese pickles, on lipopolysaccharide (LPS)-induced RAW264.7 macrophages. L. plantarum A106 had a good scavenging ability for DPPH, ABTS, and hydroxyl radicals. Furthermore, L. plantarum A106 could increase the activity of RAW264.7 macrophages; raise the SOD and GSH levels, with or without LPS sensitization; or decrease the MDA, TNF-α, and IL-6 levels. In order to deeply seek the antioxidant and anti-inflammatory role and mechanism, bioinformatic analysis, including GO, KEGG, and GSEA analysis, was used to conduct an in-depth analysis, and the results showed that the LPS treatment of RAW264.7 macrophages significantly upregulated inflammatory-related genes and revealed an enrichment in the inflammatory signaling pathways. Additionally, a network analysis via the Cytoscape software (version 3.9.1) identified key central genes and found that LPS also disturbed apoptosis and mitochondrial function. Based on the above bioinformatics analysis, the effects of L. plantarum A106 on inflammation-related gene expression, mitochondrial function, apoptosis, etc., were detected. The results indicated that L. plantarum A106 restored the declined expression levels of crucial genes like TNF-α and IL-6; mitochondrial membrane potential; and apoptosis and the expression of apoptosis-related genes, Bcl-2, Caspase-3, and Bax. These results suggest that L. plantarum A106 exerts antioxidant activity and anti-inflammatory effects through regulating inflammatory and apoptosis-related gene expression, restoring the mitochondrial membrane potential.
Collapse
Affiliation(s)
- Mengchun Qin
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Yinfei Xing
- College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| | - Maocheng Sun
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Lin Ma
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Xiaolei Li
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Fumin Ma
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Dan Li
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Cuicui Duan
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| |
Collapse
|
3
|
Liu ZY, Yang HL, Cai GH, Li S, Ye JD, Zhang CX, Sun YZ. LTA and PGN from Bacillus siamensis can alleviate soybean meal-induced enteritis and microbiota dysbiosis in Lateolabrax maculatus. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109618. [PMID: 38729251 DOI: 10.1016/j.fsi.2024.109618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
An eight-week feeding trial was designed to assess which component of commensal Bacillus siamensis LF4 can mitigate SBM-induced enteritis and microbiota dysbiosis in spotted seabass (Lateolabrax maculatus) based on TLRs-MAPKs/NF-кB signaling pathways. Fish continuously fed low SBM (containing 16 % SBM) and high SBM (containing 40 % SBM) diets were used as positive (FM group) and negative (SBM group) control, respectively. After feeding high SBM diet for 28 days, fish were supplemented with B. siamensis LF4-derived whole cell wall (CW), cell wall protein (CWP), lipoteichoic acid (LTA) or peptidoglycan (PGN) until 56 days. The results showed that a high inclusion of SBM in the diet caused enteritis, characterized with significantly (P < 0.05) decreased muscular thickness, villus height, villus width, atrophied and loosely arranged microvillus. Moreover, high SBM inclusion induced an up-regulation of pro-inflammatory cytokines and a down-regulation of occludin, E-cadherin, anti-inflammatory cytokines, apoptosis related genes and antimicrobial peptides. However, dietary supplementation with CW, LTA, and PGN of B. siamensis LF4 could effectively alleviate enteritis caused by a high level of dietary SBM. Additionally, CWP and PGN administration increased beneficial Cetobacterium and decreased pathogenic Plesiomonas and Brevinema, while dietary LTA decreased Plesiomonas and Brevinema, suggesting that CWP, LTA and PGN positively modulated intestinal microbiota in spotted seabass. Furthermore, CW, LTA, and PGN application significantly stimulated TLR2, TLR5 and MyD88 expressions, and inhibited the downstream p38 and NF-κB signaling. Taken together, these results suggest that LTA and PGN from B. siamensis LF4 could alleviate soybean meal-induced enteritis and microbiota dysbiosis in L. maculatus, and p38 MAPK/NF-κB pathways might be involved in those processes.
Collapse
Affiliation(s)
- Zi-Yan Liu
- State Key Laboratory of Mariculture Breeding, Fisheries college of Jimei university, Xiamen, 361021, China; School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Hong-Ling Yang
- State Key Laboratory of Mariculture Breeding, Fisheries college of Jimei university, Xiamen, 361021, China; Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Guo-He Cai
- State Key Laboratory of Mariculture Breeding, Fisheries college of Jimei university, Xiamen, 361021, China; Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Sha Li
- State Key Laboratory of Mariculture Breeding, Fisheries college of Jimei university, Xiamen, 361021, China
| | - Ji-Dan Ye
- State Key Laboratory of Mariculture Breeding, Fisheries college of Jimei university, Xiamen, 361021, China; Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Chun-Xiao Zhang
- State Key Laboratory of Mariculture Breeding, Fisheries college of Jimei university, Xiamen, 361021, China; Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College of Jimei University, Xiamen, 361021, China
| | - Yun-Zhang Sun
- State Key Laboratory of Mariculture Breeding, Fisheries college of Jimei university, Xiamen, 361021, China; Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College of Jimei University, Xiamen, 361021, China; The Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College of Jimei University, Xiamen, 361021, China; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education, Xiamen, 361021, China.
| |
Collapse
|
4
|
OKANO A, TANAKA S, YAMADA K, HASHIMOTO N, WATANABE J. Mechanisms of interleukin-10 induction in murine spleen and RAW264 cells by Latilactobacillus curvatus K4G4 isolated from fermented Brassica rapa L. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:227-233. [PMID: 38966044 PMCID: PMC11220328 DOI: 10.12938/bmfh.2023-073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/18/2024] [Indexed: 07/06/2024]
Abstract
Lactic acid bacteria (LAB) are commonly used in fermented foods, and some LAB modulate the immune response. We aimed to investigate the mechanism by which LAB isolates from fermented Brassica rapa L. induce the production of anti-inflammatory interleukin (IL)-10 by the murine spleen and RAW264 cells. Spleen cells from BALB/c mice or the mouse macrophage cell line RAW264 were cultured with heat-killed LAB isolated from fermented B. rapa L., and the IL-10 level in the supernatant was measured. Latilactobacillus curvatus K4G4 provided the most potent IL-10 induction among 13 isolates. Cell wall components of K4G4 failed to induce IL-10, while treatment of the bacteria with RNase A under a high salt concentration altered K4G4 induction of IL-10 by spleen cells. In general, a low salt concentration diminished the IL-10 induction by all strains, including K4G4. In addition, chloroquine pretreatment and knock down of toll-like receptor 7 through small interfering RNA suppressed K4G4 induction of IL-10 production by RAW264 cells. Our results suggest that single-stranded RNA from K4G4 is involved, via endosomal toll-like receptor 7, in the induction of IL-10 production by macrophages. K4G4 is a promising candidate probiotic strain that modulates the immune response by inducing IL-10 from macrophages.
Collapse
Affiliation(s)
- Aki OKANO
- Department of Life and Food Sciences, Obihiro University of
Agriculture and Veterinary Medicine, Inadacho, Obihiro, Hokkaido 080-8555, Japan
| | - Sachi TANAKA
- Academic Assembly (Institute of Agriculture), Shinshu
University, Minami-Minowa, Nagano 399-4598, Japan
| | - Kazuha YAMADA
- Department of Life and Food Sciences, Obihiro University of
Agriculture and Veterinary Medicine, Inadacho, Obihiro, Hokkaido 080-8555, Japan
| | - Naoto HASHIMOTO
- Department of Life and Food Sciences, Obihiro University of
Agriculture and Veterinary Medicine, Inadacho, Obihiro, Hokkaido 080-8555, Japan
| | - Jun WATANABE
- Department of Life and Food Sciences, Obihiro University of
Agriculture and Veterinary Medicine, Inadacho, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
5
|
Lin L, Zhang K, Xiong Q, Zhang J, Cai B, Huang Z, Yang B, Wei B, Chen J, Niu Q. Gut microbiota in pre-clinical rheumatoid arthritis: From pathogenesis to preventing progression. J Autoimmun 2023; 141:103001. [PMID: 36931952 DOI: 10.1016/j.jaut.2023.103001] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/30/2022] [Accepted: 01/31/2023] [Indexed: 03/17/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by progressive polyarthritis that leads to cartilage and bone damage. Pre-clinical RA is a prolonged state before clinical arthritis and RA develop, in which autoantibodies (antibodies against citrullinated proteins, rheumatoid factors) can be present due to the breakdown of immunologic self-tolerance. As early treatment initiation before the onset of polyarthritis may achieve sustained remission, optimize clinical outcomes, and even prevent RA progression, the pre-clinical RA stage is showing the prospect to be the window of opportunity for RA treatment. Growing evidence has shown the role of the gut microbiota in inducing systemic inflammation and polyarthritis via multiple mechanisms, which may involve molecular mimicry, impaired intestinal barrier function, gut microbiota-derived metabolites mediated immune regulation, modulation of the gut microbiota's effect on immune cells, intestinal epithelial cells autophagy, and the interaction between the microbiome and human leukocyte antigen alleles as well as microRNAs. Since gut microbiota alterations in pre-clinical RA have been reported, potential therapies for modifying the gut microbiota in pre-clinical RA, including natural products, antibiotic therapy, fecal microbiota transplantation, probiotics, microRNAs therapy, vitamin D supplementation, autophagy inducer-based treatment, prebiotics, and diet, holds great promise for the successful treatment and even prevention of RA via altering ongoing inflammation. In this review, we summarized current studies that include pathogenesis of gut microbiota in RA progression and promising therapeutic strategies to provide novel ideas for the management of pre-clinical RA and possibly preventing arthritis progression.
Collapse
Affiliation(s)
- Liyan Lin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Keyi Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qiao Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Infection Control, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bei Cai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zhuochun Huang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Wei
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Qian Niu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Kim MY, Hyun IK, An S, Kim D, Kim KH, Kang SS. In vitro anti-inflammatory and antibiofilm activities of bacterial lysates from lactobacilli against oral pathogenic bacteria. Food Funct 2022; 13:12755-12765. [PMID: 36416047 DOI: 10.1039/d2fo00936f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Postbiotics are functional biological compounds, such as bacterial lysates (BLs) released from probiotic bacteria. Although postbiotics exert various bioactivities, the anti-inflammatory and antibiofilm activities of BLs against oral pathogenic bacteria have not been investigated. In the present study, pretreatment with BLs extracted from Lactobacillus plantarum and L. rhamnosus GG suppressed the mRNA and protein expression levels of inflammatory mediators induced by the lipopolysaccharide (LPS) of Porphyromonas gingivalis in RAW 264.7 cells. Both BLs attenuated P. gingivalis LPS-induced phosphorylation of mitogen-activated protein kinases (MAPKs) and activation of nuclear factor-κB (NF-κB), suggesting that BLs inhibit periodontal inflammatory responses by regulating the MAPK and NF-κB signaling pathways. Moreover, both BLs interfered with biofilm formation by Streptococcus mutans; however, they did not eradicate the established S. mutans biofilm. Furthermore, both BLs downregulated gtfB, gtfC, and gtfD responsible for biofilm formation by S. mutans, suggesting that BLs reduce the synthesis of extracellular polysaccharide and thereby reduce S. mutans biofilm. Taken together, these results suggest that BLs of L. plantarum and L. rhamnosus GG can attenuate periodontal inflammation and dental caries and thus contribute to the improvement of oral health.
Collapse
Affiliation(s)
- Min Young Kim
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea.
| | - In Kyung Hyun
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea.
| | - Sunghyun An
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea.
| | - Donghan Kim
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea.
| | - Ki Hwan Kim
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea.
| | - Seok-Seong Kang
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Republic of Korea.
| |
Collapse
|
7
|
Gao K, Chen CL, Ke XQ, Yu YX, Chen S, Liu GC, Wang HF, Li YJ. Ingestion of Lactobacillus helveticus WHH1889 improves depressive and anxiety symptoms induced by chronic unpredictable mild stress in mice. Benef Microbes 2022; 13:473-488. [PMID: 36377577 DOI: 10.3920/bm2022.0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Emerging evidence indicates that the alterations in the gut microbiota-brain axis (GBA), which is the bilateral connection between the gut microbial communities and brain function, are involved in several mental illnesses, including depression. Certain probiotic strains have been revealed to improve depressive behaviours and the dysregulation of 5-hydroxytryptamine (5-HT) metabolism in depression. Here we evaluated the potential antidepressant effects of Lactobacillus helveticus strains using an in vitro enterochromaffin cell model (RIN14B). The L. helveticus strain WHH1889 was shown to significantly promote the level of 5-hydroxytryptamine (5-HTP, 5-HT precursor) and the gene expression of tryptophan hydroxylase 1 (Tph1), which is the key synthetase in the 5-HT biosynthesis in RIN14B cells. Ingestion of 0.2 ml WHH1889 (1´109 cfu/ml) in a chronic unpredictable mild stress (CUMS) mouse model of depression for five weeks normalised depressive and anxiety-like behaviours in the forced swim test, tail suspension test, sucrose preference test, and open field test. Meanwhile, the CUMS-induced elevated level of serum corticosterone and declined levels of hippocampal 5-HT and 5-HTP were reversed by WHH1889. Furthermore, the disturbances of the gut microbiome composition with reduced microbial diversity were also improved by WHH1889, accompanied by the increased colonic 5-HTP level and Tph1 gene expression. In summary, these findings indicate that WHH1889 exerts antidepressant-like effects on CUMS mice, which is associated with the modulations of the 5-HT/5-HTP metabolism and gut microbiome composition. Therefore, ingestion of the L. helveticus strain WHH1889 with antidepressant potentials may become an encouraging therapeutic option in the treatment of depression.
Collapse
Affiliation(s)
- K Gao
- Key Laboratory of Food And Biological Engineering of Zhejiang Province, Hangzhou 310018, China P.R.,Research and Development Department, Hangzhou Wahaha Group Co., Ltd, Hangzhou 310018, China P.R.,College of Animal Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310058, China P.R
| | - C-L Chen
- Key Laboratory of Food And Biological Engineering of Zhejiang Province, Hangzhou 310018, China P.R.,Research and Development Department, Hangzhou Wahaha Group Co., Ltd, Hangzhou 310018, China P.R
| | - X-Q Ke
- Key Laboratory of Food And Biological Engineering of Zhejiang Province, Hangzhou 310018, China P.R.,Research and Development Department, Hangzhou Wahaha Group Co., Ltd, Hangzhou 310018, China P.R
| | - Y-X Yu
- Key Laboratory of Food And Biological Engineering of Zhejiang Province, Hangzhou 310018, China P.R.,Research and Development Department, Hangzhou Wahaha Group Co., Ltd, Hangzhou 310018, China P.R
| | - S Chen
- Key Laboratory of Food And Biological Engineering of Zhejiang Province, Hangzhou 310018, China P.R.,Research and Development Department, Hangzhou Wahaha Group Co., Ltd, Hangzhou 310018, China P.R
| | - G-C Liu
- Key Laboratory of Food And Biological Engineering of Zhejiang Province, Hangzhou 310018, China P.R.,Research and Development Department, Hangzhou Wahaha Group Co., Ltd, Hangzhou 310018, China P.R
| | - H-F Wang
- College of Animal Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310058, China P.R
| | - Y-J Li
- Key Laboratory of Food And Biological Engineering of Zhejiang Province, Hangzhou 310018, China P.R.,Research and Development Department, Hangzhou Wahaha Group Co., Ltd, Hangzhou 310018, China P.R.,College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China P.R
| |
Collapse
|
8
|
Illikoud N, Mantel M, Rolli-Derkinderen M, Gagnaire V, Jan G. Dairy starters and fermented dairy products modulate gut mucosal immunity. Immunol Lett 2022; 251-252:91-102. [DOI: 10.1016/j.imlet.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2022]
|
9
|
Modulation of Alveolar Macrophages by Postimmunobiotics: Impact on TLR3-Mediated Antiviral Respiratory Immunity. Cells 2022; 11:cells11192986. [PMID: 36230948 PMCID: PMC9562200 DOI: 10.3390/cells11192986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Beneficial microbes with immunomodulatory capacities (immunobiotics) and their non-viable forms (postimmunobiotics) could be effectively utilized in formulations towards the prevention of respiratory viral infections. In this study, novel immunobiotic strains with the ability to increase antiviral immunity in porcine alveolar macrophages were selected from a library of Lactobacillus gasseri. Postimmunobiotics derived from the most remarkable strains were also evaluated in their capacity to modulate the immune response triggered by Toll-like receptor 3 (TLR3) in alveolar macrophages and to differentially regulate TLR3-mediated antiviral respiratory immunity in infant mice. We provide evidence that porcine alveolar macrophages (3D4/31 cells) are a useful in vitro tool for the screening of new antiviral immunobiotics and postimmunobiotics by assessing their ability to modulate the expression IFN-β, IFN-λ1, RNAseL, Mx2, and IL-6, which can be used as prospective biomarkers. We also demonstrate that the postimmunobiotics derived from the Lactobacillus gasseri TMT36, TMT39 and TMT40 (HK36, HK39 or HK40) strains modulate the innate antiviral immune response of alveolar macrophages and reduce lung inflammatory damage triggered by TLR3 activation in vivo. Although our findings should be deepened and expanded, the results of the present work provide a scientific rationale for the use of nasally administered HK36, HK39 or HK40 to beneficially modulate TLR3-triggerd respiratory innate immune response.
Collapse
|
10
|
Zhang X, Li X, Yu Y, Zhang X, Wang X, Zhang N, Chen M, Gong P, Li J. Giardia lamblia
regulates the production of proinflammatory cytokines through activating the NOD2–Rip2–ROS signaling pathway in mouse macrophages. Immunol Cell Biol 2022; 100:440-452. [DOI: 10.1111/imcb.12550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/17/2022] [Accepted: 03/31/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Xu Zhang
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University 5333 Xian Road Changchun Jilin 130062 China
| | - Xin Li
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University 5333 Xian Road Changchun Jilin 130062 China
| | - Yanhui Yu
- The Second Hospital of Jilin University Changchun Jilin 130021 China
| | - Xichen Zhang
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University 5333 Xian Road Changchun Jilin 130062 China
| | - Xiaocen Wang
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University 5333 Xian Road Changchun Jilin 130062 China
| | - Nan Zhang
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University 5333 Xian Road Changchun Jilin 130062 China
| | - Mengge Chen
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University 5333 Xian Road Changchun Jilin 130062 China
| | - Pengtao Gong
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University 5333 Xian Road Changchun Jilin 130062 China
| | - Jianhua Li
- Key Laboratory of Zoonosis Research Ministry of Education College of Veterinary Medicine Jilin University 5333 Xian Road Changchun Jilin 130062 China
| |
Collapse
|
11
|
Niu X, Yin L, Yang X, Yang Y, Gu Y, Sun Y, Yang M, Wang Y, Zhang Q, Ji H. SAA induces suppressive neutrophils via the TLR2-mediated signaling pathway to promote progression of breast cancer. Cancer Sci 2022; 113:1140-1153. [PMID: 35102665 PMCID: PMC8990718 DOI: 10.1111/cas.15287] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/11/2022] [Accepted: 01/27/2022] [Indexed: 11/29/2022] Open
Abstract
Immune inflammation plays a key role in breast cancer development, progression, and therapeutic efficacy. Neutrophils are crucial for the regulation of the suppressive tumor microenvironment and are associated with poor clinical survival. However, the mechanisms underlying the activation of suppressive neutrophils in breast cancer are poorly understood. Here, we report that breast cancer cells secrete abundant serum amyloid A 1 (SAA1), which is associated with the accumulation of suppressive neutrophils. High expression of SAA1 in breast cancer induces neutrophil immunosuppressive cytokine production through the activation of toll like receptor 2 (TLR2)-mediated signaling pathways. These include the TLR2/myeloid differentiation primary response 88 (MYD88)-mediated phosphatidylinositol 3-kinase (PI3K)/nuclear factor κB (NF-κB) signaling pathway and p38 mitogen-activated protein kinase (MAPK)-associated apoptosis resistance pathway, which eventually promote the progression of breast cancer. Our study demonstrates a mechanistic link between breast cancer cell secretion of SAA1 and suppressive neutrophils that potentiate tumor progression. These findings provide potential therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Xingjian Niu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin Heilongjiang, 150081, PR China
| | - Lei Yin
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin Heilongjiang, 150081, PR China.,Heilongjiang Academy of Medical Sciences, Harbin Heilongjiang, 150081, PR China
| | - Xudong Yang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin Heilongjiang, 150081, PR China.,Heilongjiang Academy of Medical Sciences, Harbin Heilongjiang, 150081, PR China
| | - Yue Yang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin Heilongjiang, 150081, PR China.,Heilongjiang Academy of Medical Sciences, Harbin Heilongjiang, 150081, PR China
| | - Yucui Gu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin Heilongjiang, 150081, PR China
| | - Yutian Sun
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin Heilongjiang, 150081, PR China
| | - Ming Yang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin Heilongjiang, 150081, PR China
| | - Yiran Wang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin Heilongjiang, 150081, PR China.,Heilongjiang Academy of Medical Sciences, Harbin Heilongjiang, 150081, PR China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin Heilongjiang, 150081, PR China.,Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin Heilongjiang, 150081, PR China.,Heilongjiang Academy of Medical Sciences, Harbin Heilongjiang, 150081, PR China
| | - Hongfei Ji
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin Heilongjiang, 150081, PR China.,Heilongjiang Academy of Medical Sciences, Harbin Heilongjiang, 150081, PR China
| |
Collapse
|
12
|
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
|
13
|
Host defense against Neospora caninum infection via IL-12p40 production through TLR2/TLR3-AKT-ERK signaling pathway in C57BL/6 mice. Mol Immunol 2021; 139:140-152. [PMID: 34509754 DOI: 10.1016/j.molimm.2021.08.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022]
Abstract
Neospora caninum is an intracellular parasite which can cause neosporosis and significant economic losses in both dairy and beef industries worldwide. A better understanding of the immune response by host cells against N. caninum could help to design better strategies for the prevention and treatment of neosporosis. Although previous studies have shown TLR2/TLR3 were involved in controlling N. caninum infection in mice, the precise mechanisms of the AKT and MAPK pathways controlled by TLR2/TLR3 to regulate N. caninum-induced IL-12p40 production and the role of TLR2/TLR3 in anti-N. caninum infection in bovine macrophages remain unclear. In the present study, TLR2-/- mice displayed more parasite burden and lower level of IL-12p40 production compared to TLR3-/- mice. N. caninum could activate AKT and ERK signaling pathways in WT mouse macrophages, which were inhibited in TLR2-/- and TLR3-/- mouse macrophages. In N. caninum-infected WT mouse macrophages, AKT inhibitor or AKT siRNA could decrease the phosphorylation of ERK. AKT or ERK inhibitors reduced the production of IL-12p40 and increased the number of parasites. The productions of ROS, NO, and GBP2 were significantly reduced in TLR2-/- and TLR3-/- mouse macrophages. Supplementation of rIL-12p40 inhibited N. caninum proliferation and rescued the productions of IFN-γ, NO, and GBP2 in WT, TLR2-/-, and TLR3-/- mouse macrophages. In bovine macrophages, the expressions of TLR2, TLR3, and IL-12p40 mRNA were significantly enhanced by N. caninum, and N. caninum proliferation was inhibited by TLR2/TLR3 agonists. Taken together, the proliferation of N. caninum in mouse macrophages was controlled by the TLR2/TLR3-AKT-ERK signal pathway via increased IL-12p40 production, which in turn lead to the productions of NO, GBP2, and IFN-γ during N. caninum infection. And in bovine macrophages, TLR2 and TLR3 contributed to inhibiting N. caninum proliferation via increased IL-12p40 production.
Collapse
|
14
|
de Jesus LCL, Drumond MM, Aburjaile FF, Sousa TDJ, Coelho-Rocha ND, Profeta R, Brenig B, Mancha-Agresti P, Azevedo V. Probiogenomics of Lactobacillus delbrueckii subsp. lactis CIDCA 133: In Silico, In Vitro, and In Vivo Approaches. Microorganisms 2021; 9:microorganisms9040829. [PMID: 33919849 PMCID: PMC8070793 DOI: 10.3390/microorganisms9040829] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 11/16/2022] Open
Abstract
Lactobacillus delbrueckii subsp. lactis CIDCA 133 (CIDCA 133) has been reported as a potential probiotic strain, presenting immunomodulatory properties. This study investigated the possible genes and molecular mechanism involved with a probiotic profile of CIDCA 133 through a genomic approach associated with in vitro and in vivo analysis. Genomic analysis corroborates the species identification carried out by the classical microbiological method. Phenotypic assays demonstrated that the CIDCA 133 strain could survive acidic, osmotic, and thermic stresses. In addition, this strain shows antibacterial activity against Salmonella Typhimurium and presents immunostimulatory properties capable of upregulating anti-inflammatory cytokines Il10 and Tgfb1 gene expression through inhibition of Nfkb1 gene expression. These reported effects can be associated with secreted, membrane/exposed to the surface and cytoplasmic proteins, and bacteriocins-encoding genes predicted in silico. Furthermore, our results showed the genes and the possible mechanisms used by CIDCA 133 to produce their beneficial host effects and highlight its use as a probiotic microorganism.
Collapse
Affiliation(s)
- Luís Cláudio Lima de Jesus
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
| | - Mariana Martins Drumond
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Departamento de Ciências Biológicas, Belo Horizonte 31421-169, Brazil;
| | - Flávia Figueira Aburjaile
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
- Laboratório de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| | - Thiago de Jesus Sousa
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
| | - Nina Dias Coelho-Rocha
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
| | - Rodrigo Profeta
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
| | - Bertram Brenig
- Institute of Veterinary Medicine, University of Göttingen, D-37077 Göttingen, Germany;
| | | | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
- Correspondence:
| |
Collapse
|
15
|
Cristofori F, Dargenio VN, Dargenio C, Miniello VL, Barone M, Francavilla R. Anti-Inflammatory and Immunomodulatory Effects of Probiotics in Gut Inflammation: A Door to the Body. Front Immunol 2021; 12:578386. [PMID: 33717063 PMCID: PMC7953067 DOI: 10.3389/fimmu.2021.578386] [Citation(s) in RCA: 304] [Impact Index Per Article: 101.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/14/2021] [Indexed: 02/05/2023] Open
Abstract
Hosting millions of microorganisms, the digestive tract is the primary and most important part of bacterial colonization. On one side, in cases of opportunistic invasion, the abundant bacterial population inside intestinal tissues may face potential health problems such as inflammation and infections. Therefore, the immune system has evolved to sustain the host-microbiota symbiotic relationship. On the other hand, to maintain host immune homeostasis, the intestinal microflora often exerts an immunoregulatory function that cannot be ignored. A field of great interest is the association of either microbiota or probiotics with the immune system concerning clinical uses. This microbial community regulates some of the host's metabolic and physiological functions and drives early-life immune system maturation, contributing to their homeostasis throughout life. Changes in gut microbiota can occur through modification in function, composition (dysbiosis), or microbiota-host interplays. Studies on animals and humans show that probiotics can have a pivotal effect on the modulation of immune and inflammatory mechanisms; however, the precise mechanisms have not yet been well defined. Diet, age, BMI (body mass index), medications, and stress may confound the benefits of probiotic intake. In addition to host gut functions (permeability and physiology), all these agents have profound implications for the gut microbiome composition. The use of probiotics could improve the gut microbial population, increase mucus-secretion, and prevent the destruction of tight junction proteins by decreasing the number of lipopolysaccharides (LPSs). When LPS binds endothelial cells to toll-like receptors (TLR 2, 4), dendritic cells and macrophage cells are activated, and inflammatory markers are increased. Furthermore, a decrease in gut dysbiosis and intestinal leakage after probiotic therapy may minimize the development of inflammatory biomarkers and blunt unnecessary activation of the immune system. In turn, probiotics improve the differentiation of T-cells against Th2 and development of Th2 cytokines such as IL-4 and IL-10. The present narrative review explores the interactions between gut microflora/probiotics and the immune system starting from the general perspective of a biological plausibility to get to the in vitro and in vivo demonstrations of a probiotic-based approach up to the possible uses for novel therapeutic strategies.
Collapse
Affiliation(s)
- Fernanda Cristofori
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Vanessa Nadia Dargenio
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Costantino Dargenio
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Vito Leonardo Miniello
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Michele Barone
- Gastroenterology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Ruggiero Francavilla
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
16
|
An evaluation of the effects of probiotics on tumoral necrosis factor (TNF-α) signaling and gene expression. Cytokine Growth Factor Rev 2020; 57:27-38. [PMID: 33162326 DOI: 10.1016/j.cytogfr.2020.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/16/2022]
Abstract
The search for functional foods containing probiotics has been growing due to numerous benefits they provide to health, such as modulation of the immune system and of the anti-inflammatory activity by inhibiting the release of pro-inflammatory cytokines, such as TNF-α. However, the mechanisms of actions of the probiotics responsible for this inhibition have not been completely explained so far. A better understanding of the interaction between probiotics and cell signaling pathways related to inflammatory processes shall help to prevent inflammatory bowel diseases. Therefore, the aim of this revision is to help understand the mechanisms of action of probiotics in cell signaling pathways that regulate TNF-α expression. Probiotics might act at different points of the MAPK pathway, on NF-kB, on proteasome activity, on Toll-like receptors, and on their regulators and stimuli. The present revision reaches the conclusion that probiotics act through multiple mechanisms, especially by inhibiting IkB phosphorylation and degradation, thus preventing the translocation of NF-kB. Effects are also shown to be strain-specific, and probiotics of the genus Lactobacillus are proved to play and essential role in anti-inflammatory activity.
Collapse
|
17
|
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
|
18
|
Interactions between Gut Microbiota and Immunomodulatory Cells in Rheumatoid Arthritis. Mediators Inflamm 2020; 2020:1430605. [PMID: 32963490 PMCID: PMC7499318 DOI: 10.1155/2020/1430605] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is one of the most common autoimmune diseases caused by abnormal immune activation and immune tolerance. Immunomodulatory cells (ICs) play a critical role in the maintenance and homeostasis of normal immune function and in the pathogenesis of RA. The human gastrointestinal tract is inhabited by trillions of commensal microbiota on the mucosal surface that play a fundamental role in the induction, maintenance, and function of the host immune system. Gut microbiota dysbiosis can impact both the local and systemic immune systems and further contribute to various diseases, such as RA. The neighbouring intestinal ICs located in distinct intestinal mucosa may be the most likely intermediary by which the gut microbiota can affect the occurrence and development of RA. However, the reciprocal interaction between the components of the gut microbiota and their microbial metabolites with distinct ICs and how this interaction may impact the development of RA are not well studied. Therefore, a better understanding of the gut microbiota, ICs, and their interactions might improve our knowledge of the mechanisms by which the gut microbiota contribute to RA and facilitate the further development of novel therapeutic approaches. In this review, we have summarized the roles of the gut microbiota in the immunopathogenesis of RA, especially the interactions between the gut microbiota and ICs, and further discussed the strategies for treating RA by targeting/regulating the gut microbiota.
Collapse
|
19
|
Rodovalho VDR, da Luz BSR, Rabah H, do Carmo FLR, Folador EL, Nicolas A, Jardin J, Briard-Bion V, Blottière H, Lapaque N, Jan G, Le Loir Y, de Carvalho Azevedo VA, Guédon E. Extracellular Vesicles Produced by the Probiotic Propionibacterium freudenreichii CIRM-BIA 129 Mitigate Inflammation by Modulating the NF-κB Pathway. Front Microbiol 2020; 11:1544. [PMID: 32733422 PMCID: PMC7359729 DOI: 10.3389/fmicb.2020.01544] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) are nanometric spherical structures involved in intercellular communication, whose production is considered to be a widespread phenomenon in living organisms. Bacterial EVs are associated with several processes that include survival, competition, pathogenesis, and immunomodulation. Among probiotic Gram-positive bacteria, some Propionibacterium freudenreichii strains exhibit anti-inflammatory activity, notably via surface proteins such as the surface-layer protein B (SlpB). We have hypothesized that, in addition to surface exposure and secretion of proteins, P. freudenreichii may produce EVs and thus export immunomodulatory proteins to interact with the host. In order to demonstrate their production in this species, EVs were purified from cell-free culture supernatants of the probiotic strain P. freudenreichii CIRM-BIA 129, and their physicochemical characterization, using transmission electron microscopy and nanoparticle tracking analysis (NTA), revealed shapes and sizes typical of EVs. Proteomic characterization showed that EVs contain a broad range of proteins, including immunomodulatory proteins such as SlpB. In silico protein-protein interaction predictions indicated that EV proteins could interact with host proteins, including the immunomodulatory transcription factor NF-κB. This potential interaction has a functional significance because EVs modulate inflammatory responses, as shown by IL-8 release and NF-κB activity, in HT-29 human intestinal epithelial cells. Indeed, EVs displayed an anti-inflammatory effect by modulating the NF-κB pathway; this was dependent on their concentration and on the proinflammatory inducer (LPS-specific). Moreover, while this anti-inflammatory effect partly depended on SlpB, it was not abolished by EV surface proteolysis, suggesting possible intracellular sites of action for EVs. This is the first report on identification of P. freudenreichii-derived EVs, alongside their physicochemical, biochemical and functional characterization. This study has enhanced our understanding of the mechanisms associated with the probiotic activity of P. freudenreichii and identified opportunities to employ bacterial-derived EVs for the development of bioactive products with therapeutic effects.
Collapse
Affiliation(s)
- Vinícius de Rezende Rodovalho
- INRAE, Institut Agro, STLO, Rennes, France.,Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Brenda Silva Rosa da Luz
- INRAE, Institut Agro, STLO, Rennes, France.,Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Fillipe Luiz Rosa do Carmo
- Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Edson Luiz Folador
- Biotechnology Center, Federal University of Paraíba, João Pessoa, Brazil
| | | | | | | | - Hervé Blottière
- INRAE, AgroParisTech, Paris-Saclay University, Micalis Institute, Jouy-en-Josas, France
| | - Nicolas Lapaque
- INRAE, AgroParisTech, Paris-Saclay University, Micalis Institute, Jouy-en-Josas, France
| | | | | | - Vasco Ariston de Carvalho Azevedo
- Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
20
|
Li Y, Mooney EC, Xia XJ, Gupta N, Sahingur SE. A20 Restricts Inflammatory Response and Desensitizes Gingival Keratinocytes to Apoptosis. Front Immunol 2020; 11:365. [PMID: 32218782 PMCID: PMC7078700 DOI: 10.3389/fimmu.2020.00365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 02/14/2020] [Indexed: 12/16/2022] Open
Abstract
The pathophysiology of periodontal disease involves a perturbed immune system to a dysbiotic microflora leading to unrestrained inflammation, collateral tissue damage, and various systemic complications. Gingival epithelial cells function as an important part of immunity to restrict microbial invasion and orchestrate the subsequent innate responses. A20 (TNFAIP3), an ubiquitin-editing enzyme, is one of the key regulators of inflammation and cell death in numerous tissues including gastrointestinal tract, skin, and lungs. Emerging evidence indicates A20 as an essential molecule in the oral mucosa as well. In this study, we characterized the role of A20 in human telomerase immortalized gingival keratinocytes (TIGKs) through loss and gain of function assays in preclinical models of periodontitis. Depletion of A20 through gene editing in TIGKs significantly increased IL-6 and IL-8 secretion in response to Porphyromonas gingivalis infection while A20 over-expression dampened the cytokine production compared to A20 competent cells through modulating NF-κB signaling pathway. In the subsequent experiments which assessed apoptosis, A20 depleted TIGKs displayed increased levels of cleaved caspase 3 and DNA fragmentation following P. gingivalis infection and TNF/CHX challenge compared to A20 competent cells. Consistently, there was reduced apoptosis in the cells overexpressing A20 compared to the control cells expressing GFP further substantiating the role of A20 in regulating gingival epithelial cell fate in response to exogenous insult. Collectively, our findings reveal first systematic evidence and demonstrate that A20 acts as a regulator of inflammatory response in gingival keratinocytes through its effect on NF-κB signaling and desensitizes cells to bacteria and cytokine induced apoptosis in the oral mucosa. As altered A20 levels can have profound effect on different cellular responses, future studies will determine whether A20-targeted therapies can be exploited to restrain periodontal inflammation and maintain oral mucosa tissue homeostasis.
Collapse
Affiliation(s)
- Yajie Li
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Erin C Mooney
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, VA, United States
| | - Xia-Juan Xia
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nitika Gupta
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sinem Esra Sahingur
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|