1
|
Zhang Y, Liu K, Sun Q, Qi Y, Li F, Su X, Song M, Lv R, Sui H, Shi Y, Zhao L. Collagenase Degradable Biomimetic Nanocages Attenuate Porphyromonas gingivalis Mediated Neurocognitive Dysfunction via Targeted Intracerebral Antimicrobial Photothermal and Gas Therapy. ACS NANO 2025; 19:16448-16468. [PMID: 40285729 DOI: 10.1021/acsnano.4c17748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Porphyromonas gingivalis (P.g.), a pathogen linked to periodontitis, is reported to be associated with severe neurocognitive dysfunction. However, there are few reports focusing on improving neurological function in the brain by eliminating P.g.. Therefore, we developed a core-shell nanocomposite for targeted intracerebral P.g. clearance and ameliorating neurocognitive impairments, Pt-Au@C-P.g.-MM, consisting of platinum nanoparticles (Pt NPs) encapsulated within Au nanocages (Pt-Au) as the core and a shell made of collagen and macrophage membranes from macrophage pretreated with P.g. (C-P.g.-MM). This design enhanced the nanocomposite's ability to cross the blood-brain barrier (BBB) and specifically target intracerebral P.g. through coating of P.g.-MM. Pt-Au@C-P.g.-MM depended on collagen to neutralize excessive collagenase from P.g., promoting its directed migration toward P.g.. Au nanocages exhibited excellent photothermal effects under near-infrared (NIR) laser irradiation, while Pt NPs also provided an efficient antibacterial gas therapy by generating oxygen to expose anaerobic P.g.. As a result, Pt-Au@C-P.g.-MM contributed to a synergistic antibacterial therapy and significantly reduced P.g. mediated neurocognitive dysfunction in periodontitis mice induced by oral P.g. infection. Based on the insights provided by the transcriptome sequencing analysis, anti-P.g. activity of Pt-Au@C-P.g.-MM facilitated the transition of microglia from the M1 to M2 phenotype by stimulating the PI3K-Akt pathway and reducing neuronal damage through the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yifei Zhang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Kang Liu
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Qing Sun
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Yao Qi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Fang Li
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Xiangchen Su
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Mingzhu Song
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Ruizhen Lv
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Haijuan Sui
- Department of Pharmacology, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
- Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou 121000, P. R. China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, P. R. China
- Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou 121000, P. R. China
- Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou 121000, P. R. China
| |
Collapse
|
2
|
Ahmadi P, Mahmoudi M, Rafatpanah H, Rezaieyazdi Z, Ahmadi‐Khorram M, Javanmardi Z, Tabasi NS, Esmaeili S. The Impacts of Lactobacillus delbrueckii and Lactobacillus rhamnosus to Promote In Vitro Anti-Inflammatory Profile of RA-Macrophages. Food Sci Nutr 2025; 13:e70068. [PMID: 40099178 PMCID: PMC11911130 DOI: 10.1002/fsn3.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Rheumatoid arthritis (RA) is a prevalent and debilitating autoimmune disease. Numerous studies have demonstrated promising results regarding the use of probiotics as a therapeutic approach to alleviate RA symptoms. This study isolated monocytes from the PBMCs of RA patients and healthy donors. These monocytes were then differentiated into macrophages and divided into five groups: untreated, LPS-treated, L. delbrueckii (Del)-treated, L. rhamnosus (Ram)-treated, and a mixed treatment group. Three macrophage subpopulations-M0, M1, and M2-were identified in all treatment groups, with variations observed in the population percentages of each subpopulation and the expression levels of CD14, CD80, and HLA-DR. Flow cytometry results indicated that, compared to the untreated and LPS-treated groups, treatment with probiotic bacteria (Del, Ram, and Mix) stimulated the polarization of macrophages toward the M2 phenotype while suppressing the percentage of the M1 population. Additionally, the expression of CD14, a Pathogen-Associated Molecular Pattern (PAMP) and phagocytosis-inducing receptor, was significantly reduced in the probiotic-treated groups. Probiotic treatment also profoundly influenced antigen presentation by suppressing CD80, a ligand for the CD28 co-stimulatory marker on T cells, and HLA-DR, which presents antigens to the T cell receptors of Th4 cells. Interestingly, quantitative real-time PCR results indicated that probiotic treatment of macrophages significantly increased the expression of IL-10 and TGF-β, both anti-inflammatory cytokines, while significantly decreasing the expression of inflammatory cytokines, including IL-12, IL-1β, and TNF-α, in both healthy controls and RA patients. It seems that these probiotics may have a regulatory effect on macrophages, affecting their polarization, antigen presentation patterns, phagocytosis, and cytokine secretion profiles. This suggests that these probiotics may have therapeutic and prophylactic effects on RA.
Collapse
Affiliation(s)
- Parisa Ahmadi
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Immunology Department, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
| | - Mahmoud Mahmoudi
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Immunology Department, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Houshang Rafatpanah
- Division of Inflammation and Inflammatory Diseases, Immunology Research CentreMashhad University of Medical SciencesMashhadIran
| | - Zahra Rezaieyazdi
- Rheumatic Diseases Research CenterMashhad University of Medical SciencesMashhadIran
| | - Maryam Ahmadi‐Khorram
- Department of Nutrition, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Zahra Javanmardi
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Immunology Department, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
| | | | - Seyed‐Alireza Esmaeili
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Immunology Department, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
3
|
Ferreira RLPS, Nova BGV, Carmo MS, Abreu AG. Mechanisms of action of Lactobacillus spp. in the treatment of oral candidiasis. BRAZ J BIOL 2024; 84:e282609. [PMID: 39319927 DOI: 10.1590/1519-6984.282609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/16/2024] [Indexed: 09/26/2024] Open
Abstract
Candida albicans is often associated with oral candidiasis, and drug-resistance profiles have contributed to an increase in morbidity and mortality. It is known that Lactobacillus spp. acts by competing for adhesion to the epithelium, absorption of nutrients and modulation of the human microbiota. Therefore, they are important to assist in the host's microbiological balance and reduce the growth of Candida spp. Until now, there have been no reports in the literature of reviews correlating to the use of Lactobacillus spp. in the treatment of oral candidiasis. Thus, this review aims to highlight the mechanisms of action of Lactobacillus spp. and methods that can be used in the treatment of oral candidiasis. This is a study carried out through the databases PubMed Central and Scientific Electronic Library Online, using the following keywords: Oral Candidiasis and Lactobacillus. Original articles about oral candidiasis were included, with both in vitro and in vivo analyses, and published from 2012 to 2022. Lactobacillus rhamnosus was the most common microorganism used in the experiments against Candida, acting mainly in the reduction of biofilm, filamentation, and competing for adhesion sites of Candida spp. Among in vivo studies, most researchers used immunosuppressed mouse modelsof Candida infection. The studies showed that Lactobacillus has a great potential as a probiotic, acting mainly in the prevention and treatment of mucosal diseases. Thus, the use of Lactobacillus may be a good strategy for the treatment of oral candidiasis.
Collapse
Affiliation(s)
- R L P S Ferreira
- Universidade Ceuma - UniCEUMA, Laboratório de Patogenicidade Microbiana, São Luís, MA, Brasil
- Universidade Federal do Maranhão - UFMA, Programa de Pós-graduação em Ciências da Saúde, São Luís, MA, Brasil
| | - B G V Nova
- Universidade Ceuma - UniCEUMA, Laboratório de Patogenicidade Microbiana, São Luís, MA, Brasil
| | - M S Carmo
- Universidade Ceuma - UniCEUMA, Laboratório de Patogenicidade Microbiana, São Luís, MA, Brasil
| | - A G Abreu
- Universidade Ceuma - UniCEUMA, Laboratório de Patogenicidade Microbiana, São Luís, MA, Brasil
- Universidade Federal do Maranhão - UFMA, Programa de Pós-graduação em Ciências da Saúde, São Luís, MA, Brasil
| |
Collapse
|
4
|
Xu T, Liu Y, Zhang W, Li M, Zhang L, Li X, Zhang Y, Yue L, Li S, Lin Y, Zou X, Chen F. Specific cell subclusters of dental pulp stem cells respond to distinct pathogens through the ROS pathway. Front Cell Infect Microbiol 2024; 14:1452124. [PMID: 39328360 PMCID: PMC11424553 DOI: 10.3389/fcimb.2024.1452124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/12/2024] [Indexed: 09/28/2024] Open
Abstract
Introduction Microbial pathogens invade various human organs, including the oral cavity. Candida albicans (C.a) and Streptococcus mutans (S.m) served respectively as representative oral pathogenic fungi and bacteria to stimulate dental pulp stem cells (DPSCs) and to screen the DPSC subcluster that specifically responded to fungal infection. Methods DPSCs were obtained from the impacted third molars of six healthy subjects. Then, cells were mixed and divided into three samples, two of which were stimulated with C.a and S.m, respectively; the third sample was exposed to cell medium only (Ctrl). Single-cell mRNA sequencing analysis of treated DPSCs was performed. Results DPSCs were composed of four major clusters of which one, DPSC.7, exhibited unique changes compared to those of other subclusters. The DPSC.7 cell percentage of the C.a sample was twice those of the Ctrl and S.m samples. DPSC.7 cells expressed genes associated with the response to reactive oxygen species (ROS) response. DPSC.7 subgroup cells established characteristic aggregation under the stimulation of different pathogens in UMAP. The MAPK/ERK1/2 and NF-κB pathways were up-regulated, DUSP1/5/6 expressions were suppressed, FOS synthesis was activated, the immune-related pathway was induced, and the levels of cytokines, including IL-6 and CCL2, were up-regulated in DPSC.7 cells when stimulated with C.a. Conclusions Our study analyzed the cellular and molecular properties of DPSCs infected by oral fungi and bacteria with single-cell RNA sequencing. A subcluster of DPSCs responded specifically to infections with different pathogens, activating the MAPK and NF-κB pathways to induce immune responses via the ROS pathway. This suggests novel treatment strategies for fungal infections.
Collapse
Affiliation(s)
- Tiansong Xu
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Fifth Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yangjia Liu
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Wen Zhang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Department of Stomatology, Peking University International Hospital, Beijing, China
| | - Murong Li
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Liqi Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Xueying Li
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yifei Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Lin Yue
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Sha Li
- Department of Implantology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Ye Lin
- Department of Implantology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Xiaoying Zou
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Center of Stomatology, Peking University Hospital, Beijing, China
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
5
|
Li B, Wang W, Zhao L, Wu Y, Li X, Yan D, Gao Q, Yan Y, Zhang J, Feng Y, Zheng J, Shu B, Wang J, Wang H, He L, Zhang Y, Pan M, Wang D, Tang BZ, Liao Y. Photothermal therapy of tuberculosis using targeting pre-activated macrophage membrane-coated nanoparticles. NATURE NANOTECHNOLOGY 2024; 19:834-845. [PMID: 38383890 DOI: 10.1038/s41565-024-01618-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024]
Abstract
Conventional antibiotics used for treating tuberculosis (TB) suffer from drug resistance and multiple complications. Here we propose a lesion-pathogen dual-targeting strategy for the management of TB by coating Mycobacterium-stimulated macrophage membranes onto polymeric cores encapsulated with an aggregation-induced emission photothermal agent that is excitable with a 1,064 nm laser. The coated nanoparticles carry specific receptors for Mycobacterium tuberculosis, which enables them to target tuberculous granulomas and internal M. tuberculosis simultaneously. In a mouse model of TB, intravenously injected nanoparticles image individual granulomas in situ in the lungs via signal emission in the near-infrared region IIb, with an imaging resolution much higher than that of clinical computed tomography. With 1,064 nm laser irradiation from outside the thoracic cavity, the photothermal effect generated by these nanoparticles eradicates the targeted M. tuberculosis and alleviates pathological damage and excessive inflammation in the lungs, resulting in a better therapeutic efficacy compared with a combination of first-line antibiotics. This precise photothermal modality that uses dual-targeted imaging in the near-infrared region IIb demonstrates a theranostic strategy for TB management.
Collapse
Affiliation(s)
- Bin Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China
- School of Inspection, Ningxia Medical University, Yinchuan, China
- Institute of Translational Medicine, Department of Clinical Laboratory & Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Wei Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Lu Zhao
- Institute of Translational Medicine, Department of Clinical Laboratory & Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Yunxia Wu
- Institute of Translational Medicine, Department of Clinical Laboratory & Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Xiaoxue Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Dingyuan Yan
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, China
| | - Qiuxia Gao
- Institute of Translational Medicine, Department of Clinical Laboratory & Department of Burn Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Yan Yan
- Department of Critical Care Medicine, Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Zhang
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, Xizang Minzu University, Xianyang, China
| | - Yi Feng
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Judun Zheng
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Bowen Shu
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Jiamei Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Huanhuan Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Lingjie He
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yunlong Zhang
- Department of Critical Care Medicine, Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingliang Pan
- Department of Critical Care Medicine, Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, China.
| | - Ben Zhong Tang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, China.
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen, China.
| | - Yuhui Liao
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, China.
- School of Inspection, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
6
|
Saito S, Cao DY, Maekawa T, Tsuji NM, Okuno A. Lactococcus lactis subsp. cremoris C60 Upregulates Macrophage Function by Modifying Metabolic Preference in Enhanced Anti-Tumor Immunity. Cancers (Basel) 2024; 16:1928. [PMID: 38792006 PMCID: PMC11120145 DOI: 10.3390/cancers16101928] [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: 04/19/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Lactococcus lactis subsp. cremoris C60 is a probiotic strain of lactic acid bacteria (LAB) which induces various immune modifications in myeloid lineage cells. These modifications subsequently regulate T cell function, resulting in enhanced immunity both locally and systemically. Here, we report that C60 suppresses tumor growth by enhancing macrophage function via metabolic alterations, thereby increasing adenosine triphosphate (ATP) production in a murine melanoma model. Intragastric (i.g.) administration of C60 significantly reduced tumor volume compared to saline administration in mice. The anti-tumor function of intratumor (IT) macrophage was upregulated in mice administered with C60, as evidenced by an increased inflammatory phenotype (M1) rather than an anti-inflammatory/reparative (M2) phenotype, along with enhanced antigen-presenting ability, resulting in increased tumor antigen-specific CD8+ T cells. Through this functional modification, we identified that C60 establishes a glycolysis-dominant metabolism, rather than fatty acid oxidation (FAO), in IT macrophages, leading to increased intracellular ATP levels. To address the question of why orally supplemented C60 exhibits functions in distal places, we found a possibility that bacterial cell wall components, which could be distributed throughout the body from the gut, may induce stimulatory signals in peripheral macrophages via Toll-like receptors (TLRs) signaling activation. Thus, C60 strengthens macrophage anti-tumor immunity by promoting a predominant metabolic shift towards glycolysis upon TLR-mediated stimulation, thereby increasing substantial energy production.
Collapse
Affiliation(s)
- Suguru Saito
- Department of Infection and Immunity, Division of Virology, Faculty of Medicine, Jichi Medical University, Shimotsuke, Tochigi 3290431, Japan
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Duo-Yao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Toshio Maekawa
- iFoodMed Inc., Tsuchiura, Ibaraki 3000873, Japan;
- Department of Pathology and Microbiology, Division of Immune Homeostasis, Nihon University School of Medicine, Itabashi, Tokyo 1738610, Japan;
| | - Noriko M. Tsuji
- Department of Pathology and Microbiology, Division of Immune Homeostasis, Nihon University School of Medicine, Itabashi, Tokyo 1738610, Japan;
- Department of Pathology and Microbiology, Division of Microbiology, Nihon University School of Medicine, Itabashi, Tokyo 1738610, Japan
- Department of Food Science, Jumonji University, Niiza, Saitama 3528510, Japan
| | - Alato Okuno
- Department of Health and Nutrition, Faculty of Human Design, Shibata Gakuen University, Hirosaki, Aomori 0368530, Japan
| |
Collapse
|
7
|
Bueno MR, Dudu-Silva G, Macedo TT, Gomes APDAP, Rodrigues Oliveira Braga A, Aguiar Silva LD, Bueno-Silva B. Lactobacillus acidophilus impairs the establishment of pathogens in a subgingival multispecies biofilm. FRONTIERS IN DENTAL MEDICINE 2023; 4:1212773. [PMID: 39916904 PMCID: PMC11797891 DOI: 10.3389/fdmed.2023.1212773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/13/2023] [Indexed: 02/09/2025] Open
Abstract
The present study evaluated the antibiofilm effects of Lactobacillus acidophilus within a subgingival multispecies biofilm. Lactobacillus acidophilus (La5) at 1 × 102, 1 × 104, and 1 × 106 were included at the beginning of biofilm formation, which lasted 7 days. The biofilms comprised 33 periodontitis-related bacterial species and the Calgary Biofilm device was used. At the end, DNA-DNA hybridization (checkerboard) was performed. A Kruskal-Wallis test followed by a Dunn post hoc test were performed (p ≤ 0.05). La5 at 1 × 104 and 1 × 106 reduced the total counts of biofilm and the proportions of red and green complexes when compared to the control biofilm without La5 (p ≤ 0.05). La5 at 1 × 104 increased the proportions of Actinomyces complex compared to the controls (p ≤ 0.05). Both La5 at 1 × 104 and 1 × 106 decreased levels of 20 and 14 distinct species, respectively, including Porphyromonas gingivalis, Prevotella intermedia, Fusobacterium nucleatum polymorphum, and Parvimonas micra compared to the control (p ≤ 0.05). Only La5 at 1 × 104 reduced the levels of Tannerella forsythia, Fusobacterium periodonticum, and Aggregatibacter actinomycetencomytans compared to the control (p ≤ 0.05). L. acidophilus inhibited establishing periodontic pathogens from red complex such as P. gingivalis and T. forsythia in a subgingival multispecies biofilm.
Collapse
Affiliation(s)
- Manuela Rocha Bueno
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | - Bruno Bueno-Silva
- Dental Research Division, Guarulhos University, Guarulhos, Brazil
- Department of Bioscience, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| |
Collapse
|
8
|
Hong SH. Influence of Microbiota on Vaccine Effectiveness: "Is the Microbiota the Key to Vaccine-induced Responses?". J Microbiol 2023:10.1007/s12275-023-00044-6. [PMID: 37052795 PMCID: PMC10098251 DOI: 10.1007/s12275-023-00044-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023]
Abstract
Vaccines are one of the most powerful tools for preventing infectious diseases. To effectively fight pathogens, vaccines should induce potent and long-lasting immune responses that are specific to the pathogens. However, not all vaccines can induce effective immune responses, and the responses vary greatly among individuals and populations. Although several factors, such as age, host genetics, nutritional status, and region, affect the effectiveness of vaccines, increasing data have suggested that the gut microbiota is critically associated with vaccine-induced immune responses. In this review, I discuss how gut microbiota affects vaccine effectiveness based on the clinical and preclinical data, and summarize possible underlying mechanisms related to the adjuvant effects of microbiota. A better understanding of the link between vaccine-induced immune responses and the gut microbiota using high-throughput technology and sophisticated system vaccinology approaches could provide crucial insights for designing effective personalized preventive and therapeutic vaccination strategies.
Collapse
Affiliation(s)
- So-Hee Hong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, 07084, Republic of Korea.
| |
Collapse
|
9
|
Qing F, Xie T, Xie L, Guo T, Liu Z. How Gut Microbiota Are Shaped by Pattern Recognition Receptors in Colitis and Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14153821. [PMID: 35954484 PMCID: PMC9367250 DOI: 10.3390/cancers14153821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The pathogenesis of intestinal inflammatory disorders such as colitis and colorectal cancer is complicated and dysregulation of gut microbiota is considered an important contributing factor. Inflammation is often initiated by the activation of pattern recognition receptors. However, the relationship between these innate immune receptors and gut microbiota is not fully understood. Here, we show that pattern recognition receptors not only recognize pathogens and initiate inflammatory signal transduction to induce immune responses, but also influence the composition of intestinal microorganisms, thus affecting the development of intestinal inflammation and cancer through various mechanisms. This suggests that the modification of innate immune receptors and relevant molecules could be therapeutic targets for the treatment of colitis and colorectal cancer by regulating gut microbiota. Abstract Disorders of gut microbiota have been closely linked to the occurrence of various intestinal diseases including colitis and colorectal cancer (CRC). Specifically, the production of beneficial bacteria and intestinal metabolites may slow the development of some intestinal diseases. Recently, it has been proposed that pattern recognition receptors (PRRs) not only recognize pathogens and initiate inflammatory signal transduction to induce immune responses but also influence the composition of intestinal microorganisms. However, the mechanisms through which PRRs regulate gut microbiota in the setting of colitis and CRC have rarely been systematically reviewed. Therefore, in this paper, we summarize recent advances in our understanding of how PRRs shape gut microbiota and how this influences the development of colitis and CRC.
Collapse
Affiliation(s)
- Furong Qing
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- School of Graduate, Gannan Medical University, Ganzhou 341000, China
| | - Tao Xie
- Center for Scientific Research, Gannan Medical University, Ganzhou 341000, China
| | - Lu Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Tianfu Guo
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
- Correspondence: (T.G.); (Z.L.)
| | - Zhiping Liu
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Center for Scientific Research, Gannan Medical University, Ganzhou 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
- Correspondence: (T.G.); (Z.L.)
| |
Collapse
|
10
|
Bueno MR, Ishikawa KH, Almeida-Santos G, Ando-Suguimoto ES, Shimabukuro N, Kawamoto D, Mayer MPA. Lactobacilli Attenuate the Effect of Aggregatibacter actinomycetemcomitans Infection in Gingival Epithelial Cells. Front Microbiol 2022; 13:846192. [PMID: 35602018 PMCID: PMC9116499 DOI: 10.3389/fmicb.2022.846192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/29/2022] [Indexed: 01/10/2023] Open
Abstract
Probiotics may be considered as an additional strategy to achieve a balanced microbiome in periodontitis. However, the mechanisms underlying the use of probiotics in the prevention or control of periodontitis are still not fully elucidated. This in vitro study aimed to evaluate the effect of two commercially available strains of lactobacilli on gingival epithelial cells (GECs) challenged by Aggregatibacter actinomycetemcomitans. OBA-9 GECs were infected with A. actinomycetemcomitans strain JP2 at an MOI of 1:100 and/or co-infected with Lactobacillus acidophilus La5 (La5) or Lacticaseibacillus rhamnosus Lr32 (Lr32) at an MOI of 1:10 for 2 and 24 h. The number of adherent/internalized bacteria to GECs was determined by qPCR. Production of inflammatory mediators (CXCL-8, IL-1β, GM-CSF, and IL-10) by GECs was determined by ELISA, and the expression of genes encoding cell receptors and involved in apoptosis was determined by RT-qPCR. Apoptosis was also analyzed by Annexin V staining. There was a slight loss in OBA-9 cell viability after infection with A. actinomycetemcomitans or the tested probiotics after 2 h, which was magnified after 24-h co-infection. Adherence of A. actinomycetemcomitans to GECs was 1.8 × 107 (± 1.2 × 106) cells/well in the mono-infection but reduced to 1.2 × 107 (± 1.5 × 106) in the co-infection with Lr32 and to 6 × 106 (± 1 × 106) in the co-infection with La5 (p < 0.05). GECs mono-infected with A. actinomycetemcomitans produced CXCL-8, GM-CSF, and IL-1β, and the co-infection with both probiotic strains altered this profile. While the co-infection of A. actinomycetemcomitans with La5 resulted in reduced levels of all mediators, the co-infection with Lr32 promoted reduced levels of CXCL-8 and GM-CSF but increased the production of IL-1β. The probiotics upregulated the expression of TLR2 and downregulated TLR4 in cells co-infected with A. actinomycetemcomitans. A. actinomycetemcomitans-induced the upregulation of NRLP3 was attenuated by La5 but increased by Lr32. Furthermore, the transcription of the anti-apoptotic gene BCL-2 was upregulated, whereas the pro-apoptotic BAX was downregulated in cells co-infected with A. actinomycetemcomitans and the probiotics. Infection with A. actinomycetemcomitans induced apoptosis in GECs, whereas the co-infection with lactobacilli attenuated the apoptotic phenotype. Both tested lactobacilli may interfere in A. actinomycetemcomitans colonization of the oral cavity by reducing its ability to interact with gingival epithelial cells and modulating cells response. However, L. acidophilus La5 properties suggest that this strain has a higher potential to control A. actinomycetemcomitans-associated periodontitis than L. rhamnosus Lr32.
Collapse
Affiliation(s)
- Manuela R. Bueno
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Karin H. Ishikawa
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gislane Almeida-Santos
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ellen S. Ando-Suguimoto
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Natali Shimabukuro
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Dione Kawamoto
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcia P. A. Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Bueno-Silva B, Bueno MR, Kawamoto D, Casarin RC, Pingueiro JMS, Alencar SM, Rosalen PL, Mayer MPA. Anti-Inflammatory Effects of (3S)-Vestitol on Peritoneal Macrophages. Pharmaceuticals (Basel) 2022; 15:ph15050553. [PMID: 35631379 PMCID: PMC9145271 DOI: 10.3390/ph15050553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
The isoflavone (3S)-vestitol, obtained from red propolis, has exhibited anti-inflammatory, antimicrobial, and anti-caries activity; however, few manuscripts deal with its anti-inflammatory mechanisms in macrophages. The objective is to elucidate the anti-inflammatory mechanisms of (3S)-vestitol on those cells. Peritoneal macrophages of C57BL6 mice, stimulated with lipopolysaccharide, were treated with 0.37 to 0.59 µM of (3S)-vestitol for 48 h. Then, nitric oxide (NO) quantities, macrophages viability, the release of 20 cytokines and the transcription of several genes related to cytokine production and inflammatory response were evaluated. The Tukey–Kramer variance analysis test statistically analyzed the data. (3S)-vestitol 0.55 µM (V55) lowered NO release by 60% without altering cell viability and diminished IL-1β, IL-1α, G-CSF, IL-10 and GM-CSF levels. V55 reduced expression of Icam-1, Wnt5a and Mmp7 (associated to inflammation and tissue destruction in periodontitis) and Scd1, Scd2, Egf1 (correlated to atherosclerosis). V55 increased expression of Socs3 and Dab2 genes (inhibitors of cytokine signaling and NF-κB pathway), Apoe (associated to atherosclerosis control), Igf1 (encoder a protein with analogous effects to insulin) and Fgf10 (fibroblasts growth factor). (3S)-vestitol anti-inflammatory mechanisms involve cytokines and NF-κB pathway inhibition. Moreover, (3S)-vestitol may be a candidate for future in vivo investigations about the treatment/prevention of persistent inflammatory diseases such as atherosclerosis and periodontitis.
Collapse
Affiliation(s)
- Bruno Bueno-Silva
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, SP, Brazil; (M.R.B.); (D.K.); (M.P.A.M.)
- Dental Research Division, Guarulhos University, Guarulhos 07023-070, SP, Brazil;
- Correspondence:
| | - Manuela Rocha Bueno
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, SP, Brazil; (M.R.B.); (D.K.); (M.P.A.M.)
| | - Dione Kawamoto
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, SP, Brazil; (M.R.B.); (D.K.); (M.P.A.M.)
| | - Renato C. Casarin
- Piracicaba Dental School, University of Campinas-UNICAMP, Piracicaba 13414-903, SP, Brazil; (R.C.C.); (P.L.R.)
| | | | - Severino Matias Alencar
- College of Agriculture “Luiz de Queiroz” (ESALQ/USP), University of São Paulo, Piracicaba 13418-900, SP, Brazil;
| | - Pedro Luiz Rosalen
- Piracicaba Dental School, University of Campinas-UNICAMP, Piracicaba 13414-903, SP, Brazil; (R.C.C.); (P.L.R.)
| | - Marcia Pinto Alves Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, SP, Brazil; (M.R.B.); (D.K.); (M.P.A.M.)
| |
Collapse
|
12
|
Archambault LS, Dongari-Bagtzoglou A. Probiotics for Oral Candidiasis: Critical Appraisal of the Evidence and a Path Forward. FRONTIERS IN ORAL HEALTH 2022; 3:880746. [PMID: 35495563 PMCID: PMC9046664 DOI: 10.3389/froh.2022.880746] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/29/2022] Open
Abstract
Oropharyngeal Candidiasis (OPC) is a mucosal fungal infection that is prevalent among patients with compromised immunity. The success of probiotics in treating chronic diseases with a microbial etiology component at other mucosal sites (i.e., gastro-intestinal, genitourinary and alveolar mucosae) has inspired research into the use of probiotics in the treatment of OPC. A growing body of research in vitro and in animal models indicates that some probiotic species and strains have inhibitory activities against Candida albicans growth, morphological switching, and biofilm formation. However, recent review and meta-analysis studies reveal a dearth of human randomized, controlled clinical trials on the efficacy of probiotics to treat or prevent OPC, while the majority of these have not based their selection of probiotic strains or the type of administration on sound pre-clinical evidence. In this mini-review, we assess the state of the field, outline some of the difficulties in translating lab results to clinical efficacy, and make recommendations for future research needed in order to move the field forward.
Collapse
Affiliation(s)
- Linda S. Archambault
- Department of Craniofacial Sciences, University of Connecticut Health Center, Farmington, CT, United States
- Center for Quantitative Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Anna Dongari-Bagtzoglou
- Department of Craniofacial Sciences, University of Connecticut Health Center, Farmington, CT, United States
- *Correspondence: Anna Dongari-Bagtzoglou
| |
Collapse
|
13
|
Duarte ME, Kim SW. Intestinal microbiota and its interaction to intestinal health in nursery pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 8:169-184. [PMID: 34977387 PMCID: PMC8683651 DOI: 10.1016/j.aninu.2021.05.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/20/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023]
Abstract
The intestinal microbiota has gained increased attention from researchers within the swine industry due to its role in promoting intestinal maturation, immune system modulation, and consequently the enhancement of the health and growth performance of the host. This review aimed to provide updated scientific information on the interaction among intestinal microbiota, dietary components, and intestinal health of pigs. The small intestine is a key site to evaluate the interaction of the microbiota, diet, and host because it is the main site for digestion and absorption of nutrients and plays an important role within the immune system. The diet and its associated components such as feed additives are the main factors affecting the microbial composition and is central in stimulating a beneficial population of microbiota. The microbiota–host interaction modulates the immune system, and, concurrently, the immune system helps to modulate the microbiota composition. The direct interaction between the microbiota and the host is an indication that the mucosa-associated microbiota can be more effective in evaluating its effect on health parameters. It was demonstrated that the mucosa-associated microbiota should be evaluated when analyzing the interaction among diets, microbiota, and health. In addition, supplementation of feed additives aimed to promote the intestinal health of pigs should consider their roles in the modulation of mucosa-associated microbiota as biomarkers to predict the response of growth performance to dietary interventions.
Collapse
Affiliation(s)
- Marcos Elias Duarte
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, United States
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, United States
| |
Collapse
|
14
|
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: 2.0] [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
|
15
|
Zhao W, Wang X, Zhao C, Yan Z. Immunomodulatory mechanism of Bacillus subtilis R0179 in RAW 264.7 cells against Candida albicans challenge. Microb Pathog 2021; 157:104988. [PMID: 34044051 DOI: 10.1016/j.micpath.2021.104988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/26/2022]
Abstract
This study was aimed to explore the immunomodulatory and anti-Candida mechanisms of Bacillus subtilis (B. subtilis) R0179 in macrophages. RAW 264.7 cells were first challenged with B. subtilis R0179. B. subtilis R0179 was found to down-regulate the signals of Dectin-1, Card9, P-Iκ-Bα, Iκ-Bα, and NF-κB. Meanwhile, it reduced the levels of cytokines interleukin (IL)-1β, IL-6, IL-12, and tumor necrosis factor (TNF)-α, but increased the level of cytokine IL-10. Then RAW 264.7 cells were pretreated with B. subtilis R0179 before challenged with Candida albicans (C. albicans) or RAW 264.7 cells were co-treated with B. subtilis R0179 and C. albicans. In the presence of C. albicans, B. subtilis R0179 also showed the similar immunomodulatory effects on RAW 264.7 cells. Hence, this study provides the first insight into the immunomodulatory mechanisms of B. subtilis R0179 on the Dectin-1-related downstream signaling pathways in macrophages, which may prevent tissue damage caused by excessive pro-inflammatory response during the infection of C. albicans.
Collapse
Affiliation(s)
- Weiwei Zhao
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China; Central Laboratory, Peking University School and Hospital of Stomatology, China; National Center of Stomatology, Peking University School and Hospital of Stomatology, China
| | - Xu Wang
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China; Central Laboratory, Peking University School and Hospital of Stomatology, China; National Center of Stomatology, Peking University School and Hospital of Stomatology, China
| | - Chen Zhao
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China; Department of Oral Medicine, The Affiliated Stomatology Hospital of Tongji University, Shanghai, 200070, PR China
| | - Zhimin Yan
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China; Central Laboratory, Peking University School and Hospital of Stomatology, China; National Center of Stomatology, Peking University School and Hospital of Stomatology, China.
| |
Collapse
|
16
|
Demin KA, Refeld AG, Bogdanova AA, Prazdnova EV, Popov IV, Kutsevalova OY, Ermakov AM, Bren AB, Rudoy DV, Chistyakov VA, Weeks R, Chikindas ML. Mechanisms of Candida Resistance to Antimycotics and Promising Ways to Overcome It: The Role of Probiotics. Probiotics Antimicrob Proteins 2021; 13:926-948. [PMID: 33738706 DOI: 10.1007/s12602-021-09776-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 12/12/2022]
Abstract
Pathogenic Candida and infections caused by those species are now considered as a serious threat to public health. The treatment of candidiasis is significantly complicated by the increasing resistance of pathogenic strains to current treatments and the stagnant development of new antimycotic drugs. Many species, such as Candida auris, have a wide range of resistance mechanisms. Among the currently used synthetic and semi-synthetic antifungal drugs, the most effective are azoles, echinocandins, polyenes, nucleotide analogs, and their combinations. However, the use of probiotic microorganisms and/or the compounds they produce is quite promising, although underestimated by modern pharmacology, to control the spread of pathogenic Candida species.
Collapse
Affiliation(s)
- Konstantin A Demin
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Aleksandr G Refeld
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Anna A Bogdanova
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Evgenya V Prazdnova
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Igor V Popov
- Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia
| | | | - Alexey M Ermakov
- Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia
| | - Anzhelica B Bren
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia.,Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia
| | - Dmitry V Rudoy
- Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia
| | - Vladimir A Chistyakov
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Richard Weeks
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA
| | - Michael L Chikindas
- Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia. .,Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA. .,I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
17
|
Prado Acosta M, Goyette-Desjardins G, Scheffel J, Dudeck A, Ruland J, Lepenies B. S-Layer From Lactobacillus brevis Modulates Antigen-Presenting Cell Functions via the Mincle-Syk-Card9 Axis. Front Immunol 2021; 12:602067. [PMID: 33732234 PMCID: PMC7957004 DOI: 10.3389/fimmu.2021.602067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
C-type lectin receptors (CLRs) are pattern recognition receptors that are crucial in the innate immune response. The gastrointestinal tract contributes significantly to the maintenance of immune homeostasis; it is the shelter for billions of microorganisms including many genera of Lactobacillus sp. Previously, it was shown that host-CLR interactions with gut microbiota play a crucial role in this context. The Macrophage-inducible C-type lectin (Mincle) is a Syk-coupled CLR that contributes to sensing of mucosa-associated commensals. In this study, we identified Mincle as a receptor for the Surface (S)-layer of the probiotic bacteria Lactobacillus brevis modulating GM-CSF bone marrow-derived cells (BMDCs) functions. We found that the S-layer/Mincle interaction led to a balanced cytokine response in BMDCs by triggering the release of both pro- and anti-inflammatory cytokines. In contrast, BMDCs derived from Mincle−/−, CARD9−/− or conditional Syk−/− mice failed to maintain this balance, thus leading to an increased production of the pro-inflammatory cytokines TNF and IL-6, whereas the levels of the anti-inflammatory cytokines IL-10 and TGF-β were markedly decreased. Importantly, this was accompanied by an altered CD4+ T cell priming capacity of Mincle−/− BMDCs resulting in an increased CD4+ T cell IFN-γ production upon stimulation with L. brevis S-layer. Our results contribute to the understanding of how commensal bacteria regulate antigen-presenting cell (APC) functions and highlight the importance of the Mincle/Syk/Card9 axis in APCs as a key factor in host-microbiota interactions.
Collapse
Affiliation(s)
- Mariano Prado Acosta
- Research Center for Emerging Infections and Zoonoses, Institute for Immunology, University of Veterinary Medicine, Hannover, Germany
| | - Guillaume Goyette-Desjardins
- Research Center for Emerging Infections and Zoonoses, Institute for Immunology, University of Veterinary Medicine, Hannover, Germany
| | - Jörg Scheffel
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany
| | - Jürgen Ruland
- School of Medicine, Institute of Clinical Chemistry and Pathobiochemistry, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Bernd Lepenies
- Research Center for Emerging Infections and Zoonoses, Institute for Immunology, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
18
|
Garcia-Gonzalez N, Nuñez-Sanchez MA, Villoria Recio M, Battista N, Gahan CGM, Corsetti A. Immunomodulation of J774A.1 Murine Macrophages by Lactiplantibacillus plantarum Strains Isolated From the Human Gastrointestinal Tract and Fermented Foods. Front Microbiol 2021; 11:557143. [PMID: 33510712 PMCID: PMC7835322 DOI: 10.3389/fmicb.2020.557143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022] Open
Abstract
Lactobacillus plantarum species (recently re-named Lactiplantibacillus (Lpb.) plantarum subsp. plantarum) can be isolated from both either the mammalian gut or specific fermented foods where they may be present at high concentrations. Whilst Lpb. plantarum strains have been proposed as potential probiotic candidates, the ability of resident strains consumed in fermented foods to interact with the host is unclear. The main objective of this study was to investigate the cellular location and ability of three different food-borne Lpb. plantarum strains isolated from different sources (table olives and cheese) to modulate the immune response of a murine macrophage-like cell line (J774A.1). For that purpose, macrophages were exposed to the three different Lpb. plantarum strains for 24 h and the expression of a panel of genes involved in the immune response, including genes encoding pattern-recognition receptors (TLRs and NLRs) and cytokines was evaluated by qRT-PCR. We also utilized chemical inhibitors of intracellular pathways to gain some insight into potential signaling mechanisms. Results showed that the native food strains of Lpb. plantarum were able to modulate the response of J774A.1 murine macrophages through a predominately NOD signaling pathway that reflects the transient intracellular location of these strains within the macrophage. The data indicate the capacity of food-dwelling Lpb. plantarum strains to influence macrophage-mediated host responses if consumed in sufficient quantities.
Collapse
Affiliation(s)
- Natalia Garcia-Gonzalez
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy.,School of Microbiology, University College Cork, Cork, Ireland
| | | | - Miguel Villoria Recio
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Natalia Battista
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Cormac G M Gahan
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Aldo Corsetti
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
19
|
De Rudder C, Garcia-Tímermans C, De Boeck I, Lebeer S, Van de Wiele T, Calatayud Arroyo M. Lacticaseibacillus casei AMBR2 modulates the epithelial barrier function and immune response in a donor-derived nasal microbiota manner. Sci Rep 2020; 10:16939. [PMID: 33037304 PMCID: PMC7547715 DOI: 10.1038/s41598-020-73857-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/17/2020] [Indexed: 01/06/2023] Open
Abstract
Live biotherapeutic products (LBP) are emerging as alternative treatment strategies for chronic rhinosinusitis. The selection of interesting candidate LBPs often involves model systems that do not include the polymicrobial background (i.e. the host microbiota) in which they will be introduced. Here, we performed a screening in a simplified model system of upper respiratory epithelium to assess the effect of nasal microbiota composition on the ability to attach and grow of a potential LBP, Lacticaseibacillus casei AMBR2, in this polymicrobial background. After selecting the most permissive and least permissive donor, L. casei AMBR2 colonisation in their respective polymicrobial backgrounds was assessed in more physiologically relevant model systems. We examined cytotoxicity, epithelial barrier function, and cytokine secretion, as well as bacterial cell density and phenotypic diversity in differentiated airway epithelium based models, with or without macrophage-like cells. L. casei AMBR2 could colonize in the presence of both selected donor microbiota and increased epithelial barrier resistance in presence of donor-derived nasal bacteria, as well as anti-inflammatory cytokine secretion in the presence of macrophage-like cells. This study highlights the potential of L. casei AMBR2 as LBP and the necessity to employ physiologically relevant model systems to investigate host–microbe interaction in LBP research.
Collapse
Affiliation(s)
- Charlotte De Rudder
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Coupure Links 653, Ghent University, 9000, Ghent, Belgium
| | - Cristina Garcia-Tímermans
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Coupure Links 653, Ghent University, 9000, Ghent, Belgium
| | - Ilke De Boeck
- Research Group of Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Sarah Lebeer
- Research Group of Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Coupure Links 653, Ghent University, 9000, Ghent, Belgium.
| | - Marta Calatayud Arroyo
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Coupure Links 653, Ghent University, 9000, Ghent, Belgium.,Group of Lactic Bacteria and Probiotics, Department of Biotechnology, Institute of Agrochemistry and Food Technology (IATA), Spanish Research Council (CSIC), Valencia, Spain
| |
Collapse
|
20
|
Ghavami SB, Yadegar A, Aghdaei HA, Sorrentino D, Farmani M, Mir AS, Azimirad M, Balaii H, Shahrokh S, Zali MR. Immunomodulation and Generation of Tolerogenic Dendritic Cells by Probiotic Bacteria in Patients with Inflammatory Bowel Disease. Int J Mol Sci 2020; 21:6266. [PMID: 32872480 PMCID: PMC7503552 DOI: 10.3390/ijms21176266] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
In inflammatory bowel diseases (IBD), the therapeutic benefit and mucosal healing from specific probiotics may relate to the modulation of dendritic cells (DCs). Herein, we assessed the immunomodulatory effects of four probiotic strains including Lactobacillus salivarius, Bifidobacterium bifidum, Bacillus coagulans and Bacillus subtilis natto on the expression of co-stimulatory molecules, cytokine production and gene expression of signal-transducing receptors in DCs from IBD patients. Human monocyte-derived DCs from IBD patients and healthy controls were exposed to four probiotic strains. The expression of co-stimulatory molecules was assessed and supernatants were analyzed for anti-inflammatory cytokines. The gene expression of toll-like receptors (TLRs), IL-12p40 and integrin αvβ8 were also analyzed. CD80 and CD86 were induced by most probiotic strains in ulcerative colitis (UC) patients whereas only B. bifidum induced CD80 and CD86 expression in Crohn's disease (CD) patients. IL-10 and TGF-β production was increased in a dose-independent manner while TLR expression was decreased by all probiotic bacteria except B. bifidum in DCs from UC patients. TLR-4 and TLR-9 expression was significantly downregulated while integrin ß8 was significantly increased in the DCs from CD patients. IL-12p40 expression was only significantly downregulated in DCs from CD patients. Our findings point to the general beneficial effects of probiotics in DC immunomodulation and indicate that probiotic bacteria favorably modulate the expression of co-stimulatory molecules, proinflammatory cytokines and TLRs in DCs from IBD patients.
Collapse
Affiliation(s)
- Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran; (S.B.G.); (H.A.A.); (M.F.)
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran; (A.Y.); (M.A.)
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran; (S.B.G.); (H.A.A.); (M.F.)
| | - Dario Sorrentino
- IBD Center, Division of Gastroenterology, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Clinical and Experimental Medical Sciences, University of Udine School of Medicine, 33100 Udine, Italy
| | - Maryam Farmani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran; (S.B.G.); (H.A.A.); (M.F.)
| | - Adil Shamim Mir
- Department of Internal Medicine, Roanoke Memorial Hospital, Carilion Clinic, VA 24014, USA;
| | - Masoumeh Azimirad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran; (A.Y.); (M.A.)
| | - Hedieh Balaii
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran; (H.B.); (S.S.); (M.R.Z.)
| | - Shabnam Shahrokh
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran; (H.B.); (S.S.); (M.R.Z.)
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran; (H.B.); (S.S.); (M.R.Z.)
| |
Collapse
|
21
|
Ishikawa KH, Mita D, Kawamoto D, Nicoli JR, Albuquerque-Souza E, Lorenzetti Simionato MR, Mayer MPA. Probiotics alter biofilm formation and the transcription of Porphyromonas gingivalis virulence-associated genes. J Oral Microbiol 2020; 12:1805553. [PMID: 32944156 PMCID: PMC7482675 DOI: 10.1080/20002297.2020.1805553] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background and Objective The potential of probiotics on the prevention and control of periodontitis and other chronic inflammatory conditions has been suggested. Lactobacillus and Bifidobacterium species influence P. gingivalis interaction with gingival epithelial cells (GECs) but may not act in a unique way. In order to select the most appropriate probiotic against P. gingivalis, we aimed to evaluate the effect of several strains on Porphyromonas gingivalis biofilm formation and transcription virulence-associated factors (PgVAFs). Methods Cell-free pH neutralized supernatants (CFS) and living Lactobacillus spp. and Bifidobacterium spp. were tested against P. gingivalis ATCC 33277 and W83, in mono- and multi-species (with Streptococcus oralis and S. gordonii) biofilms. Relative transcription of P. gingivalis genes (fimA, mfa1, kgp, rgp, ftsH and luxS) was determined in biofilms and under GECs co-infection. Results Probiotics CFS reduced P. gingivalis ATCC 33277 levels in mono-species biofilms and living probiotics reduced P. gingivalis abundance in multi-species biofilms. L. acidophilus LA5 down-regulated transcription of most PgVAFs in biofilms and GECs. Conclusions Probiotics affect P. gingivalis biofilm formation by down-regulating overall PgVAFs with the most pronounced effect observed for L. acidophilus LA5.
Collapse
Affiliation(s)
- Karin Hitomi Ishikawa
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela Mita
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Dione Kawamoto
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jacques Robert Nicoli
- Department of Microbiology, Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Emmanuel Albuquerque-Souza
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | | | - Marcia Pinto Alves Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
22
|
Veisseire P, Bonnet M, Saraoui T, Poupet C, Camarès O, Gachinat M, Callon C, Febvre G, Chassard C, Bornes S. Investigation into In Vitro and In Vivo Caenorhabditis elegans Models to Select Cheese Yeasts as Probiotic Candidates for their Preventive Effects against Salmonella Typhimurium. Microorganisms 2020; 8:microorganisms8060922. [PMID: 32570901 PMCID: PMC7356738 DOI: 10.3390/microorganisms8060922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 12/17/2022] Open
Abstract
The design of multiscale strategies integrating in vitro and in vivo models is necessary for the selection of new probiotics. In this regard, we developed a screening assay based on the investigation of the potential of yeasts from cheese as probiotics against the pathogen Salmonella Typhimurium UPsm1 (ST). Two yeasts isolated from raw-milk cheese (Saccharomyces cerevisiae 16, Sc16; Debaryomyces hansenii 25, Dh25), as well as S. cerevisiae subspecies boulardii (CNCM I-1079, Sb1079), were tested against ST by applying in vitro and in vivo tests. Adherence measurements to Caco-2 and HT29-MTX intestinal cells indicated that the two tested cheese yeasts presented a better adhesion than the probiotic Sb1079 as the control strain. Further, the Dh25 was the cheese yeast most likely to survive in the gastrointestinal tract. What is more, the modulation of the TransEpithelial Electrical Resistance (TEER) of differentiated Caco-2 cell monolayers showed the ability of Dh25 to delay the deleterious effects of ST. The influence of microorganisms on the in vivo model Caenorhabditis elegans was evaluated by measuring the longevity of the worm. This in vivo approach revealed that this yeast increased the worm’s lifespan and protected it against ST infection, confirming that this in vivo model can be useful for screening probiotic cheese yeasts.
Collapse
Affiliation(s)
- Philippe Veisseire
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
- Correspondence: ; Tel.: +33-(0)4-43-79-11-28
| | - Muriel Bonnet
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Taous Saraoui
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Cyril Poupet
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Olivier Camarès
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Marylise Gachinat
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Cécile Callon
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Guy Febvre
- Université Clermont Auvergne, Laboratoire Météorologie Physique, CNRS, F-15000 Aurillac, France;
| | - Christophe Chassard
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Stéphanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| |
Collapse
|
23
|
Fidel PL, Yano J, Esher SK, Noverr MC. Applying the Host-Microbe Damage Response Framework to Candida Pathogenesis: Current and Prospective Strategies to Reduce Damage. J Fungi (Basel) 2020; 6:jof6010035. [PMID: 32168864 PMCID: PMC7151217 DOI: 10.3390/jof6010035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/16/2022] Open
Abstract
Disease is a complex outcome that can occur as a result of pathogen-mediated damage, host-mediated damage or both. This has led to the revolutionary concept of the damage response framework (DRF) that defines microbial virulence as a function of host immunity. The DRF outlines six scenarios (classes) of host damage or beneficial outcomes, depending on the microbe and the strength of the immune response. Candida albicans is uniquely adapted to its human host and can exist as either a commensal, colonizing various anatomical sites without causing notable damage, or as a pathogen, with the ability to cause a diverse array of diseases, ranging from mucosal to invasive systemic infections that result in varying levels of microbe-mediated and/or host-mediated damage. We recently categorized six different forms of candidiasis (oropharyngeal, hematogenous, intra-abdominal, gastrointestinal, denture stomatitis, and vulvovaginitis) into independent DRF classes, supporting a contemporary view of unique mechanisms of pathogenesis for these Candida infections. In this review, we summarize the evidence for the pathogenesis of these various forms of candidiasis in the context of the DRF with the further intent to provide insights into strategies to achieve a level of host response or outcome otherwise, that limits host damage.
Collapse
Affiliation(s)
- Paul L. Fidel
- Center of Excellence in Oral and Craniofacial Biology, Louisiana State University Health Sciences Center School of Dentistry, New Orleans, LA 70119, USA;
- Correspondence: ; Tel.: +1-504-941-8425
| | - Junko Yano
- Center of Excellence in Oral and Craniofacial Biology, Louisiana State University Health Sciences Center School of Dentistry, New Orleans, LA 70119, USA;
| | - Shannon K. Esher
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.K.E.); (M.C.N.)
| | - Mairi C. Noverr
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (S.K.E.); (M.C.N.)
| |
Collapse
|
24
|
Dual and Triple Epithelial Coculture Model Systems with Donor-Derived Microbiota and THP-1 Macrophages To Mimic Host-Microbe Interactions in the Human Sinonasal Cavities. mSphere 2020; 5:5/1/e00916-19. [PMID: 31941815 PMCID: PMC6968656 DOI: 10.1128/msphere.00916-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Despite the relevance of the resident microbiota in sinonasal health and disease and the need for cross talk between immune and epithelial cells in the upper respiratory tract, these parameters have not been combined in a single in vitro model system. We have developed a coculture system of differentiated respiratory epithelium and natural nasal microbiota and incorporated an immune component. As indicated by absence of cytotoxicity and stable cytokine profiles and epithelial integrity, nasal microbiota from human origin appeared to be well tolerated by host cells, while microbial community composition remained representative for that of the human (sino)nasal cavity. Importantly, the introduction of macrophage-like cells enabled us to obtain a differential readout from the epithelial cells dependent on the donor microbial background to which the cells were exposed. We conclude that both model systems offer the means to investigate host-microbe interactions in the upper respiratory tract in a more representative way. The epithelium of the human sinonasal cavities is colonized by a diverse microbial community, modulating epithelial development and immune priming and playing a role in respiratory disease. Here, we present a novel in vitro approach enabling a 3-day coculture of differentiated Calu-3 respiratory epithelial cells with a donor-derived bacterial community, a commensal species (Lactobacillus sakei), or a pathobiont (Staphylococcus aureus). We also assessed how the incorporation of macrophage-like cells could have a steering effect on both epithelial cells and the microbial community. Inoculation of donor-derived microbiota in our experimental setup did not pose cytotoxic stress on the epithelial cell layers, as demonstrated by unaltered cytokine and lactate dehydrogenase release compared to a sterile control. Epithelial integrity of the differentiated Calu-3 cells was maintained as well, with no differences in transepithelial electrical resistance observed between coculture with donor-derived microbiota and a sterile control. Transition of nasal microbiota from in vivo to in vitro conditions maintained phylogenetic richness, and yet a decrease in phylogenetic and phenotypic diversity was noted. Additional inclusion and coculture of THP-1-derived macrophages did not alter phylogenetic diversity, and yet donor-independent shifts toward higher Moraxella and Mycoplasma abundance were observed, while phenotypic diversity was also increased. Our results demonstrate that coculture of differentiated airway epithelial cells with a healthy donor-derived nasal community is a viable strategy to mimic host-microbe interactions in the human upper respiratory tract. Importantly, including an immune component allowed us to study host-microbe interactions in the upper respiratory tract more in depth. IMPORTANCE Despite the relevance of the resident microbiota in sinonasal health and disease and the need for cross talk between immune and epithelial cells in the upper respiratory tract, these parameters have not been combined in a single in vitro model system. We have developed a coculture system of differentiated respiratory epithelium and natural nasal microbiota and incorporated an immune component. As indicated by absence of cytotoxicity and stable cytokine profiles and epithelial integrity, nasal microbiota from human origin appeared to be well tolerated by host cells, while microbial community composition remained representative for that of the human (sino)nasal cavity. Importantly, the introduction of macrophage-like cells enabled us to obtain a differential readout from the epithelial cells dependent on the donor microbial background to which the cells were exposed. We conclude that both model systems offer the means to investigate host-microbe interactions in the upper respiratory tract in a more representative way.
Collapse
|
25
|
Extracellular Membrane Vesicles from Lactobacilli Dampen IFN-γ Responses in a Monocyte-Dependent Manner. Sci Rep 2019; 9:17109. [PMID: 31745234 PMCID: PMC6864076 DOI: 10.1038/s41598-019-53576-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/01/2019] [Indexed: 12/01/2022] Open
Abstract
Secreted factors derived from Lactobacillus are able to dampen pro-inflammatory cytokine responses. Still, the nature of these components and the underlying mechanisms remain elusive. Here, we aimed to identify the components and the mechanism involved in the Lactobacillus-mediated modulation of immune cell activation. PBMC were stimulated in the presence of the cell free supernatants (CFS) of cultured Lactobacillus rhamnosus GG and Lactobacillus reuteri DSM 17938, followed by evaluation of cytokine responses. We show that lactobacilli-CFS effectively dampen induced IFN-γ and IL-17A responses from T- and NK cells in a monocyte dependent manner by a soluble factor. A proteomic array analysis highlighted Lactobacillus-induced IL-1 receptor antagonist (ra) as a potential candidate responsible for the IFN-γ dampening activity. Indeed, addition of recombinant IL-1ra to stimulated PBMC resulted in reduced IFN-γ production. Further characterization of the lactobacilli-CFS revealed the presence of extracellular membrane vesicles with a similar immune regulatory activity to that observed with the lactobacilli-CFS. In conclusion, we have shown that lactobacilli produce extracellular MVs, which are able to dampen pro-inflammatory cytokine responses in a monocyte-dependent manner.
Collapse
|
26
|
The Dysbiosis and Inter-Kingdom Synergy Model in Oropharyngeal Candidiasis, a New Perspective in Pathogenesis. J Fungi (Basel) 2019; 5:jof5040087. [PMID: 31546600 PMCID: PMC6958497 DOI: 10.3390/jof5040087] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 12/28/2022] Open
Abstract
As more information emerges on oral microbiota using advanced sequencing methodologies, it is imperative to examine how organisms modulate the capacity of each other to colonize or trigger infection. Most mouse models of oral C. albicans infection have focused on interactions with single bacterial species. Thus, little is known about the microbiome-mediated interactions that control the switch of C. albicans from commensalism to infection. Evidence is accumulating that in immunosuppression where mucosal candidiasis is more prevalent, there is an altered oral bacterial microbiome with reduced diversity, but not an altered mycobiome. Oropharyngeal candidiasis in immunosuppressed humans and mice is associated with a further reduction in oral bacterial diversity and a dysbiotic shift with significant enrichment of streptococcal and enterococcal species. Our recent studies in a cancer chemotherapy mouse model supported the combined profound effect of immunosuppression and C. albicans in reducing oral bacterial diversity and provided the first direct evidence that these changes contribute to pathogenesis, representing dysbiosis. There is still a gap in understanding the relationship between Candida and the oral bacterial microbiome. We propose that certain oral commensal bacteria contribute to fungal pathogenesis and we identify gaps in our understanding of the mechanisms involved in this cooperative virulence.
Collapse
|
27
|
Santecchia I, Vernel-Pauillac F, Rasid O, Quintin J, Gomes-Solecki M, Boneca IG, Werts C. Innate immune memory through TLR2 and NOD2 contributes to the control of Leptospira interrogans infection. PLoS Pathog 2019; 15:e1007811. [PMID: 31107928 PMCID: PMC6544334 DOI: 10.1371/journal.ppat.1007811] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/31/2019] [Accepted: 05/02/2019] [Indexed: 12/25/2022] Open
Abstract
Leptospira interrogans are pathogenic spirochetes responsible for leptospirosis, a worldwide reemerging zoonosis. Many Leptospira serovars have been described, and prophylaxis using inactivated bacteria provides only short-term serovar-specific protection. Therefore, alternative approaches to limit severe leptospirosis in humans and morbidity in cattle would be welcome. Innate immune cells, including macrophages, play a key role in fighting infection and pathogen clearance. Recently, it has been shown that functional reprograming of innate immune cells through the activation of pattern recognition receptors leads to enhanced nonspecific antimicrobial responses upon a subsequent microbial encounter. This mechanism is known as trained immunity or innate immune memory. We have previously shown that oral treatment with Lactobacillus plantarum confers a beneficial effect against acute leptospirosis. Here, using a macrophage depletion protocol and live imaging in mice, we established the role of peritoneal macrophages in limiting the initial dissemination of leptospires. We further showed that intraperitoneal priming of mice with CL429, a TLR2 and NOD2 agonist known to mimic the modulatory effect of Lactobacillus, alleviated acute leptospiral infection. The CL429 treatment was characterized as a training effect since i.) it was linked to peritoneal macrophages that produced ex vivo more pro-inflammatory cytokines and chemokines against 3 different pathogenic serovars of Leptospira, independently of the presence of B and T cells, ii.) it had systemic effects on splenic cells and bone marrow derived macrophages, and iii.) it was sustained for 3 months. Importantly, trained macrophages produced more nitric oxide, a potent antimicrobial compound, which has not been previously linked to trained immunity. Accordingly, trained macrophages better restrict leptospiral survival. Finally, we could use CL429 to train ex vivo human monocytes that produced more cytokines upon leptospiral stimulation. In conclusion, host-directed treatment using a TLR2/NOD2 agonist could be envisioned as a novel prophylactic strategy against acute leptospirosis.
Collapse
Affiliation(s)
- Ignacio Santecchia
- Unité Biologie et Génétique de la Paroi Bactérienne, Institut Pasteur, Groupe Avenir, INSERM, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Frédérique Vernel-Pauillac
- Unité Biologie et Génétique de la Paroi Bactérienne, Institut Pasteur, Groupe Avenir, INSERM, Paris, France
| | - Orhan Rasid
- Chromatine et Infection G5, Institut Pasteur, Paris, France
| | - Jessica Quintin
- Immunologie des infections fongiques G5, Institut Pasteur, Paris, France
| | - Maria Gomes-Solecki
- University of Tennessee Health Science Center, Department of Microbiology, Immunology and Biochemistry, Memphis, Tennessee, United States of America
| | - Ivo G. Boneca
- Unité Biologie et Génétique de la Paroi Bactérienne, Institut Pasteur, Groupe Avenir, INSERM, Paris, France
| | - Catherine Werts
- Unité Biologie et Génétique de la Paroi Bactérienne, Institut Pasteur, Groupe Avenir, INSERM, Paris, France
- * E-mail:
| |
Collapse
|
28
|
Piatek J, Sommermeyer H, Bernatek M, Ciechelska-Rybarczyk A, Oleskow B, Mikkelsen LS, Barken KB. Persistent infection by Salmonella enterica servovar Typhimurium: are synbiotics a therapeutic option? – a case report. Benef Microbes 2019; 10:211-217. [DOI: 10.3920/bm2018.0080] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Little is known about the prevalence of persistent human infections by nontyphoidal Salmonella (NTS). Recently published study results indicate that a small fraction (at least 2.2%) of NTS-infected patients continue to shed Salmonella for an extended period of time (up to years). Despite the recommendation not to use antibiotics for the treatment of uncomplicated NTS-Salmonella infection, little treatment guidance is available. Clinical findings from a NTS-patient indicate that administration of synbiotics (probiotic bacteria plus prebiotic) might be considered as a treatment option. We report data of a patient who was treated with a synbiotic preparation containing nine different probiotic bacteria and the prebiotic fructooligosaccharides (FOS). Starting from day one of the treatment, the patient experienced an improvement of symptoms and was symptom-free at the end of a 10 days treatment course. After finishing the treatment, the stool proved to be Salmonella enterica serovar Typhimurium negative. In vitro pathogenic inhibition studies showed the inhibitory effects of the multistrain synbiotic mixture against S. Typhimurium. Growth of S. Typhimurium was also inhibited by individual bacterial strains making part of the composition of the mixture. However, he inhibitory effects of individual strains varied significantly, with those of Streptococcus thermophilus St-21 and Lactobacillus helveticus SP-27 exhibiting the strongest inhibitory effect. Persistent infections by S. Typhimurium are a severe concern for the affected patients. Besides the symptomatic burden, infected persons are banned from work in certain areas (e.g. food related service). In addition, patients with persistent S. Typhimurium infections are a threat for the public health in general, as they serve as a reservoir for NTS transmission. The findings indicate that treatment with a synbiotic preparation might provide a treatment option for persistent S. Typhimurium infections. More clinical data have to be gathered to confirm the relevance of this potential treatment approach.
Collapse
Affiliation(s)
- J. Piatek
- The President Stanisław Wojciechowski State University of Applied Sciences in Kalisz, Nowy Świat 4 st., Kalisz 62-800, Poland
| | - H. Sommermeyer
- Vivatrex GmbH, Martinstrasse 10-12, 52062 Aachen, Germany
| | - M. Bernatek
- State Hospital Jarocin, Szpitalna 1, Jarocin 63-200, Poland
| | | | - B. Oleskow
- District Sanitary-Epidemiological Station in Jarocin, Waska 2, Jarocin 63-200, Poland
| | | | | |
Collapse
|
29
|
Xu C, Guo Y, Qiao L, Ma L, Cheng Y, Roman A. Biogenic Synthesis of Novel Functionalized Selenium Nanoparticles by Lactobacillus casei ATCC 393 and Its Protective Effects on Intestinal Barrier Dysfunction Caused by Enterotoxigenic Escherichia coli K88. Front Microbiol 2018; 9:1129. [PMID: 29967593 PMCID: PMC6015882 DOI: 10.3389/fmicb.2018.01129] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/14/2018] [Indexed: 11/13/2022] Open
Abstract
Selenium (Se) is an essential element for human and animal health. Biogenic selenium nanoparticles (SeNPs) by microorganism possess unique physical and chemical properties and biological activities compared with inorganic Se and organic Se. The study was conducted to investigate the mainly biological activities of SeNPs by Lactobacillus casei ATCC 393 (L. casei 393). The results showed that L. casei 393 transformed sodium selenite to red SeNPs with the size of 50–80 nm, and accumulated them intracellularly. L. casei 393-SeNPs promoted the growth and proliferation of porcine intestinal epithelial cells (IPEC-J2), human colonic epithelial cells (NCM460), and human acute monocytic leukemia cell (THP-1)-derived macrophagocyte. L. casei 393-SeNPs significantly inhibited the growth of human liver tumor cell line-HepG2, and alleviated diquat-induced IPEC-J2 oxidative damage. Moreover, in vivo and in vitro experimental results showed that administration with L. casei 393-SeNPs protected against Enterotoxigenic Escherichia coli K88 (ETEC K88)-caused intestinal barrier dysfunction. ETEC K88 infection-associated oxidative stress (glutathione peroxidase activity, total superoxide dismutase activity, total antioxidant capacity, and malondialdehyde) was ameliorated in L. casei 393-SeNPs-treated mice. These findings suggest that L. casei 393-SeNPs with no cytotoxicity play a key role in maintaining intestinal epithelial integrity and intestinal microflora balance in response to oxidative stress and infection.
Collapse
Affiliation(s)
- Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yu Guo
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Lei Qiao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Li Ma
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yiyi Cheng
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Alexandra Roman
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|