1
|
da Silva TF, Glória RDA, Americo MF, Freitas ADS, de Jesus LCL, Barroso FAL, Laguna JG, Coelho-Rocha ND, Tavares LM, le Loir Y, Jan G, Guédon É, Azevedo VADC. Unlocking the Potential of Probiotics: A Comprehensive Review on Research, Production, and Regulation of Probiotics. Probiotics Antimicrob Proteins 2024; 16:1687-1723. [PMID: 38539008 DOI: 10.1007/s12602-024-10247-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 10/02/2024]
Abstract
This review provides a comprehensive overview of the current state of probiotic research, covering a wide range of topics, including strain identification, functional characterization, preclinical and clinical evaluations, mechanisms of action, therapeutic applications, manufacturing considerations, and future directions. The screening process for potential probiotics involves phenotypic and genomic analysis to identify strains with health-promoting properties while excluding those with any factor that could be harmful to the host. In vitro assays for evaluating probiotic traits such as acid tolerance, bile metabolism, adhesion properties, and antimicrobial effects are described. The review highlights promising findings from in vivo studies on probiotic mitigation of inflammatory bowel diseases, chemotherapy-induced mucositis, dysbiosis, obesity, diabetes, and bone health, primarily through immunomodulation and modulation of the local microbiota in human and animal models. Clinical studies demonstrating beneficial modulation of metabolic diseases and human central nervous system function are also presented. Manufacturing processes significantly impact the growth, viability, and properties of probiotics, and the composition of the product matrix and supplementation with prebiotics or other strains can modify their effects. The lack of regulatory oversight raises concerns about the quality, safety, and labeling accuracy of commercial probiotics, particularly for vulnerable populations. Advancements in multi-omics approaches, especially probiogenomics, will provide a deeper understanding of the mechanisms behind probiotic functionality, allowing for personalized and targeted probiotic therapies. However, it is crucial to simultaneously focus on improving manufacturing practices, implementing quality control standards, and establishing regulatory oversight to ensure the safety and efficacy of probiotic products in the face of increasing therapeutic applications.
Collapse
Affiliation(s)
- Tales Fernando da Silva
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Rafael de Assis Glória
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Americo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria Dos Santos Freitas
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luis Claudio Lima de Jesus
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Guimarães Laguna
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Laisa Macedo Tavares
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Yves le Loir
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Gwénaël Jan
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Éric Guédon
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Vasco Ariston de Carvalho Azevedo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
2
|
Vahidi Z, Saghi E, Mahmoudi M, RezaieYazdi Z, Esmaeili SA, Zemorshidi F, Samadi M, Rastin M. Lactobacillus rhamnosus and Lactobacillus delbrueckii Ameliorate the Expression of miR-125a and miR-146a in Systemic Lupus Erythematosus Patients. Appl Biochem Biotechnol 2024; 196:6330-6341. [PMID: 38351428 DOI: 10.1007/s12010-023-04827-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 11/29/2024]
Abstract
The microRNAs are non-coding RNA molecules involved in physiological and pathological processes, causing autoimmune diseases such as systemic lupus erythematosus (SLE). Probiotics are living microorganisms that possess beneficial effects on the host immune system and modulate it. The effect of Lactobacillus rhamnosus and Lactobacillus delbrueckii on the expression of miR-125a and miR-146a was studied in peripheral blood mononuclear cells (PBMCs) from newly diagnosed lupus patients in this in vitro study. During this study, 20 recently diagnosed SLE patients and 20 healthy individuals participated. Ficoll method was used to isolate the PBMCs from whole blood, which were cultured for 48 h with Lactobacillus rhamnosus and Lactobacillus delbrueckii. In the next step, total RNA containing microRNA was extracted. cDNA was synthesized for miR-125a and miR-146a genes and analyzed by real-time PCR. Results were presented as fold changes. As compared to healthy controls, SLE patients expressed lower levels of miR-125a and miR-146a. PBMCs treated with Lactobacillus rhamnosus, Lactobacillus delbrueckii, or both probiotics had significantly higher levels of miR-125a and miR-146a compared to the untreated group. Treatment of PBMCs with both L. rhamnosus and L. delbrueckii upregulated the expression of miR-125a and miR-146a in treated cells compared with untreated cells in SLE patients (p = 0.02, p = 0.001). Lactobacillus rhamnosus and Lactobacillus delbrueckii modify lupus patients' immune responses and disease effects by regulating miR-125a and miR-146a.
Collapse
Affiliation(s)
- Zohreh Vahidi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Effat Saghi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Departments, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra RezaieYazdi
- Rheumatic Diseases Research Center, Ghaem Hospital, Internal Medicine Section, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Departments, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fariba Zemorshidi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Morteza Samadi
- Department of Immunology, Faculty of Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Rastin
- Immunology Departments, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
de Jesus LCL, Freitas ADS, Dutra JDCF, Campos GM, Américo MF, Laguna JG, Dornelas EG, Carvalho RDDO, Vital KD, Fernandes SOA, Cardoso VN, de Oliveira JS, de Oliveira MFA, Faria AMC, Ferreira E, Souza RDO, Martins FS, Barroso FAL, Azevedo V. Lactobacillus delbrueckii CIDCA 133 fermented milk modulates inflammation and gut microbiota to alleviate acute colitis. Food Res Int 2024; 186:114322. [PMID: 38729712 DOI: 10.1016/j.foodres.2024.114322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024]
Abstract
Lactobacillus delbrueckii subsp. lactis CIDCA 133 is a health-promoting bacterium that can alleviate gut inflammation and improve the epithelial barrier in a mouse model of mucositis. Despite these beneficial effects, the protective potential of this strain in other inflammation models, such as inflammatory bowel disease, remains unexplored. Herein, we examined for the first time the efficacy of Lactobacillus delbrueckii CIDCA 133 incorporated into a fermented milk formulation in the recovery of inflammation, epithelial damage, and restoration of gut microbiota in mice with dextran sulfate sodium-induced colitis. Oral administration of Lactobacillus delbrueckii CIDCA 133 fermented milk relieved colitis by decreasing levels of inflammatory factors (myeloperoxidase, N-acetyl-β-D-glucosaminidase, toll-like receptor 2, nuclear factor-κB, interleukins 10 and 6, and tumor necrosis factor), secretory immunoglobulin A levels, and intestinal paracellular permeability. This immunobiotic also modulated the expression of tight junction proteins (zonulin and occludin) and the activation of short-chain fatty acids-related receptors (G-protein coupled receptors 43 and 109A). Colonic protection was effectively associated with acetate production and restoration of gut microbiota composition. Treatment with Lactobacillus delbrueckii CIDCA 133 fermented milk increased the abundance of Firmicutes members (Lactobacillus genus) while decreasing the abundance of Proteobacteria (Helicobacter genus) and Bacteroidetes members (Bacteroides genus). These promising outcomes influenced the mice's mucosal healing, colon length, body weight, and disease activity index, demonstrating that this immunobiotic could be explored as an alternative approach for managing inflammatory bowel disease.
Collapse
Affiliation(s)
- Luís Cláudio Lima de Jesus
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Andria Dos Santos Freitas
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Joyce da Cruz Ferraz Dutra
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela Munis Campos
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Monique Ferrary Américo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana Guimarães Laguna
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Evandro Gonçalves Dornelas
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Kátia Duarte Vital
- Federal University of Minas Gerais, Department of Clinical and Toxicological Analysis, Belo Horizonte, Minas Gerais, Brazil
| | | | - Valbert Nascimento Cardoso
- Federal University of Minas Gerais, Department of Clinical and Toxicological Analysis, Belo Horizonte, Minas Gerais, Brazil
| | - Jamil Silvano de Oliveira
- Federal University of Minas Gerais, Department of Biochemistry and Immunology, Belo Horizonte, Minas Gerais, Brazil
| | | | - Ana Maria Caetano Faria
- Federal University of Minas Gerais, Department of Biochemistry and Immunology, Belo Horizonte, Minas Gerais, Brazil
| | - Enio Ferreira
- Federal University of Minas Gerais, Department of General Pathology, Belo Horizonte, Minas Gerais, Brazil
| | - Ramon de Oliveira Souza
- Federal University of Minas Gerais, Department of Microbiology, Belo Horizonte, Minas Gerais, Brazil; Ezequiel Dias Foundation, Research and Development Board, Belo Horizonte, Minas Gerais, Brazil
| | - Flaviano Santos Martins
- Federal University of Minas Gerais, Department of Microbiology, Belo Horizonte, Minas Gerais, Brazil
| | | | - Vasco Azevedo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
4
|
Laguna JG, Freitas ADS, Barroso FAL, De Jesus LCL, De Vasconcelos OAGG, Quaresma LS, Américo MF, Campos GM, Glória RDA, Dutra JDCF, Da Silva TF, Vital KD, Fernandes SO, Souza RO, Martins FDS, Ferreira E, Santos TM, Birbrair A, De Oliveira MFA, Faria AMC, Carvalho RDDO, Venanzi FM, Le Loir Y, Jan G, Guédon É, Azevedo VADC. Recombinant probiotic Lactococcus lactis delivering P62 mitigates moderate colitis in mice. Front Microbiol 2024; 15:1309160. [PMID: 38680913 PMCID: PMC11047439 DOI: 10.3389/fmicb.2024.1309160] [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: 10/07/2023] [Accepted: 02/21/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction and objective p62 is a human multifunctional adaptor protein involved in key cellular processes such as tissue homeostasis, inflammation, and cancer. It acts as a negative regulator of inflammasome complexes. It may thus be considered a good candidate for therapeutic use in inflammatory bowel diseases (IBD), such as colitis. Probiotics, including recombinant probiotic strains producing or delivering therapeutic biomolecules to the host mucosal surfaces, could help prevent and mitigate chronic intestinal inflammation. The objective of the present study was to combine the intrinsic immunomodulatory properties of the probiotic Lactococcus lactis NCDO2118 with its ability to deliver health-promoting molecules to enhance its protective and preventive effects in the context of ulcerative colitis (UC). Material and methods This study was realized in vivo in which mice were supplemented with the recombinant strain. The intestinal barrier function was analyzed by monitoring permeability, secretory IgA total levels, mucin expression, and tight junction genes. Its integrity was evaluated by histological analyses. Regarding inflammation, colonic cytokine levels, myeloperoxidase (MPO), and expression of key genes were monitored. The intestinal microbiota composition was investigated using 16S rRNA Gene Sequencing. Results and discussion No protective effect of L. lactis NCDO2118 pExu:p62 was observed regarding mice clinical parameters compared to the L. lactis NCDO2118 pExu: empty. However, the recombinant strain, expressing p62, increased the goblet cell counts, upregulated Muc2 gene expression in the colon, and downregulated pro-inflammatory cytokines Tnf and Ifng when compared to L. lactis NCDO2118 pExu: empty and inflamed groups. This recombinant strain also decreased colonic MPO activity. No difference in the intestinal microbiota was observed between all treatments. Altogether, our results show that recombinant L. lactis NCDO2118 delivering p62 protein protected the intestinal mucosa and mitigated inflammatory damages caused by dextran sodium sulfate (DSS). We thus suggest that p62 may constitute part of a therapeutic approach targeting inflammation.
Collapse
Affiliation(s)
- Juliana Guimarães Laguna
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria dos Santos Freitas
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Luís Cláudio Lima De Jesus
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Ludmila Silva Quaresma
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Américo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gabriela Munis Campos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rafael de Assis Glória
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Joyce da Cruz Ferraz Dutra
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Tales Fernando Da Silva
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Kátia Duarte Vital
- Department of Clinical Analysis and Toxicology, Federal University of Minas Gerais Belo Horizonte, Minas Gerais, Brazil
| | - Simone O. Fernandes
- Department of Clinical Analysis and Toxicology, Federal University of Minas Gerais Belo Horizonte, Minas Gerais, Brazil
| | - Ramon O. Souza
- Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Enio Ferreira
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Túlio Marcos Santos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alexander Birbrair
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Ana Maria Caetano Faria
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Franco Maria Venanzi
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | | | | | | | | |
Collapse
|
5
|
Mofrad LZ, Fateh A, Sotoodehnejadnematalahi F, Asbi DNS, Davar Siadat S. The Effect of Akkermansia muciniphila and Its Outer Membrane Vesicles on MicroRNAs Expression of Inflammatory and Anti-inflammatory Pathways in Human Dendritic Cells. Probiotics Antimicrob Proteins 2024; 16:367-382. [PMID: 36884184 DOI: 10.1007/s12602-023-10058-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Probiotics play a crucial role in immunomodulation by regulating dendritic cell (DC) maturation and inducing tolerogenic DCs. Akkermansia muciniphila affects inflammatory response by elevating inhibitory cytokines. We aimed to evaluate whether Akkermansia muciniphila and its outer membrane vesicles (OMVs) affect microRNA-155, microRNA-146a, microRNA-34a, and let-7i expression of inflammatory and anti-inflammatory pathways. Peripheral blood mononuclear cells (PBMCs) were isolated from the healthy volunteers. To produce DCs, monocytes were cultivated with granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4). DCs were allocated into six subgroups: DC + Lipopolysaccharide (LPS), DC + dexamethasone, DC + A. muciniphila (MOI 100, 50), DC + OMVs (50 µg/ml), and DC + PBS. The surface expression of human leukocyte antigen-antigen D related (HLA-DR), CD86, CD80, CD83, CD11c, and CD14 was examined using flow cytometry, and the expression of microRNAs was assessed using qRT-PCR, and the levels of IL-12 and IL-10 were measured using ELISA. A. muciniphila (MOIs 50, 100) could significantly decrease IL-12 levels relative to the LPS group. The IL-10 levels were decreased in the DC + LPS group than the DC + dexamethasone group. Treatment with A. muciniphila (MOI 100) and OMVs could elevate the concentrations of IL-10. DC treatment with LPS led to a significant increment in the expression of microRNA-155, microRNA-34a, and microRNA-146a. The expression of these microRNAs was reversed by A. muciniphilia and its OMVs treatment. Let-7i increased in treatment groups compared to the DC + LPS group. A. muciniphilia (MOI 50) had a substantial effect on the expression of HLA-DR, CD80, and CD83 on DCs. Therefore, DCs treatment with A. muciniphila led to induce tolerogenic DCs and the production of anti-inflammatory IL-10.
Collapse
Affiliation(s)
- Laya Zoghi Mofrad
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
6
|
Javanmardi Z, Mahmoudi M, Rafatpanah H, Rezaieyazdi Z, Shapouri-Moghaddam A, Ahmadi P, Mollazadeh S, Tabasi NS, Esmaeili SA. Tolerogenic probiotics Lactobacillus delbrueckii and Lactobacillus rhamnosus promote anti-inflammatory profile of macrophages-derived monocytes of newly diagnosed patients with systemic lupus erythematosus. Cell Biochem Funct 2024; 42:e3981. [PMID: 38509733 DOI: 10.1002/cbf.3981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
Systemic lupus erythematosus (SLE) is known as an autoimmune disorder that is characterized by the breakdown of self-tolerance, resulting in disease onset and progression. Macrophages have been implicated as a factor in the development of SLE through faulty phagocytosis of dead cells or an imbalanced M1/M2 ratio. The study aimed to investigate the immunomodulatory effects of Lactobacillus delbrueckii and Lactobacillus rhamnosus on M1 and M2 macrophages in new case lupus patients. For this purpose, blood monocytes were collected from lupus patients and healthy people and were cultured for 5 days to produce macrophages. For 48 h, the macrophages were then cocultured with either probiotics or lipopolysaccharides (LPS). Flow cytometry and real-time polymerase chain reaction were then used to analyze the expression of cluster of differentiation (CD) 14, CD80, and human leukocyte antigen - DR (HLADR) markers, as well as cytokine expression (interleukin [IL]1-β, IL-12, tumor necrosis factor α [TNF-α], IL-10, and transforming growth factor beta [TGF-β]). The results indicated three distinct macrophage populations, M0, M1, and M2. In both control and patient-derived macrophage-derived monocytes (MDMs), the probiotic groups showed a decrease in CD14, CD80, and HLADR expression compared to the LPS group. This decrease was particularly evident in M0 and M2 macrophages from lupus patients and M1 macrophages from healthy subjects. In addition, the probiotic groups showed increased levels of IL-10 and TGF-β and decreased levels of IL-12, IL1-β, and TNF-α in MDMs from both healthy and lupus subjects compared to the LPS groups. Although there was a higher expression of pro-inflammatory cytokines in lupus patients, there was a higher expression of anti-inflammatory cytokines in healthy subjects. In general, L. delbrueckii and L. rhamnosus could induce anti-inflammatory effects on MDMs from both healthy and lupus subjects.
Collapse
Affiliation(s)
- Zahra Javanmardi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Centre, Division of Inflammation and Inflammatory Diseases, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Rezaieyazdi
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Parisa Ahmadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Nafiseh Sadat Tabasi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Wu J, Huang H, Wang L, Gao M, Meng S, Zou S, Feng Y, Feng Z, Zhu Z, Cao X, Li B, Kang G. A tailored series of engineered yeasts for the cell-dependent treatment of inflammatory bowel disease by rational butyrate supplementation. Gut Microbes 2024; 16:2316575. [PMID: 38381494 PMCID: PMC10883098 DOI: 10.1080/19490976.2024.2316575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/17/2024] [Accepted: 02/06/2024] [Indexed: 02/22/2024] Open
Abstract
Intestinal microbiota dysbiosis and metabolic disruption are considered essential characteristics in inflammatory bowel disorders (IBD). Reasonable butyrate supplementation can help patients regulate intestinal flora structure and promote mucosal repair. Here, to restore microbiota homeostasis and butyrate levels in the patient's intestines, we modified the genome of Saccharomyces cerevisiae to produce butyrate. We precisely regulated the relevant metabolic pathways to enable the yeast to produce sufficient butyrate in the intestine with uneven oxygen distribution. A series of engineered strains with different butyrate synthesis abilities was constructed to meet the needs of different patients, and the strongest can reach 1.8 g/L title of butyrate. Next, this series of strains was used to co-cultivate with gut microbiota collected from patients with mild-to-moderate ulcerative colitis. After receiving treatment with engineered strains, the gut microbiota and the butyrate content have been regulated to varying degrees depending on the synthetic ability of the strain. The abundance of probiotics such as Bifidobacterium and Lactobacillus increased, while the abundance of harmful bacteria like Candidatus Bacilloplasma decreased. Meanwhile, the series of butyrate-producing yeast significantly improved trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice by restoring butyrate content. Among the series of engineered yeasts, the strain with the second-highest butyrate synthesis ability showed the most significant regulatory and the best therapeutic effect on the gut microbiota from IBD patients and the colitis mouse model. This study confirmed the existence of a therapeutic window for IBD treatment by supplementing butyrate, and it is necessary to restore butyrate levels according to the actual situation of patients to restore intestinal flora.
Collapse
Affiliation(s)
- Jiahao Wu
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - He Huang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Lina Wang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Mengxue Gao
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Shuxian Meng
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Shaolan Zou
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Yuanhang Feng
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Zeling Feng
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhixin Zhu
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bingzhi Li
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, China
| | - Guangbo Kang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, China
| |
Collapse
|
8
|
Hoseinzadeh A, Mahmoudi M, Rafatpanah H, Rezaieyazdi Z, Tavakol Afshari J, Hosseini S, Esmaeili SA. A new generation of mesenchymal stromal/stem cells differentially trained by immunoregulatory probiotics in a lupus microenvironment. Stem Cell Res Ther 2023; 14:358. [PMID: 38072921 PMCID: PMC10712058 DOI: 10.1186/s13287-023-03578-z] [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: 08/22/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Increasing evidence suggests that multipotent mesenchymal stem/stromal cells (MSCs) are a promising intervention strategy in treating autoimmune inflammatory diseases. It should be stated that systemic immunoregulation is increasingly recognized among the beneficial effects of MSCs and probiotics in treating morbid autoimmune disorders such as lupus. This study aimed to determine if immunoregulatory probiotics L. rhamnosus or L. delbrueckii can change the immunomodulatory effects of MSCs in lupus-like disease. METHODS Pristane-induced lupus (PIL) mice model was created via intraperitoneal injection of Pristane and then confirmed. Naïve MSCs (N-MSCs) were coincubated with two Lactobacillus strains, rhamnosus (R-MSCs) or delbrueckii (D-MSCs), and/or a combination of both (DR-MSCs) for 48 h, then administrated intravenously in separate groups. Negative (PBS-treated normal mice) and positive control groups (PBS-treated lupus mice) were also investigated. At the end of the study, flow cytometry and enzyme-linked immunosorbent assay (ELISA) analysis were used to determine the percentage of Th cell subpopulations in splenocytes and the level of their master cytokines in sera, respectively. Moreover, lupus nephritis was investigated and compared. Analysis of variance (ANOVA) was used for multiple comparisons. RESULTS Abnormalities in serum levels of anti-dsDNA antibodies, creatinine, and urine proteinuria were significantly suppressed by MSCs transplantation, whereas engrafted MSCs coincubation with both L. strains did a lesser effect on anti-dsDNA antibodies. L. rhamnosus significantly escalated the ability of MSCs to scale down the inflammatory cytokines (IFN-ɣ, IL-17), while L. delbrueckii significantly elevated the capacity of MSCs to scale down the percentage of Th cell subpopulations. However, incubation with both strains induced MSCs with augmented capacity in introducing inflammatory cytokines (IFN-ɣ, IL-17). Strikingly, R-MSCs directly restored the serum level of TGF-β more effectively and showed more significant improvement in disease parameters than N-MSCs. These results suggest that R-MSCs significantly attenuate lupus disease by further skew the immune phenotype of MSCs toward increased immunoregulation. CONCLUSIONS Results demonstrated that Lactobacillus strains showed different capabilities in training/inducing new abilities in MSCs, in such a way that pretreated MSCs with L. rhamnosus might benefit the treatment of lupus-like symptoms, given their desirable properties.
Collapse
Affiliation(s)
- Akram Hoseinzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Centre, Division of Inflammation and Inflammatory Diseases, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Rezaieyazdi
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Tavakol Afshari
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Hosseini
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Alaei A, Mahmoudi M, Sahebari M, Vahidi Z, Tabasi N, Rastin M. The effects of Lactobacillus delbrueckii and Lactobacillus rhamnosus on cytokines and their related molecules: An ex vivo study on patients with systemic lupus erythematosus. Arch Rheumatol 2023; 38:642-652. [PMID: 38125063 PMCID: PMC10728742 DOI: 10.46497/archrheumatol.2023.9941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/05/2023] [Indexed: 12/23/2023] Open
Abstract
Objectives This study aimed to assess the ex vivo impact of Lactobacillus delbrueckii (L. delbrueckii) and Lactobacillus rhamnosus (L. rhamnosus) on inflammatory and anti-inflammatory cytokines as well as their related molecules on the peripheral blood mononuclear cells (PBMCs) of systemic lupus erythematosus (SLE) patients. Patients and methods This study was conducted with 20 newly diagnosed SLE patients (18 females, 2 males; mean age: 33.3±12.4 years; range, 18 to 68 years) between September 2017 and September 2018. Extracted PBMCs from each patient were divided into 4 cell groups in our study. Three cell groups act as treatment groups receiving L. rhamnosus (107 CFU/mL), L. delbrueckii (105 CFU/mL) or a mixture of both, and one group act as our untreated control group in the absence of any probiotic agents. All cell groups were cultured in RPMI 1460 medium for 48 h. Then, total RNA was extracted, and cDNA was synthesized. Results The gene expression levels of forkhead box P3 (FOXP3), transforming growth factor beta (TGF-β), interleukin (IL)-6, IL-10, and IL-2 were evaluated by a quantitative real-time polymerase chain reaction. The results revealed that expression levels of FOXP3, TGF-β, IL-10, and IL-2 increased and the level of IL-6 decreased in probiotics-receiving groups compared to the control group. Lactobacillus delbrueckii and L. rhamnosus enhanced the expression of regulatory T cell-related molecules such as FOXP3 and IL-2 and also increased the expression of IL-10. These probiotics also reduced the expression of IL-6 as proinflammatory cytokines in the PBMCs of SLE patients. Conclusion The results of the present study show that these probiotics could be effective in regulating the balance of cytokine gene expression ex vivo , and due to their beneficial effects, they can be an intriguing option in the production of new complement drugs for SLE.
Collapse
Affiliation(s)
- Atefeh Alaei
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Sahebari
- Rheumatic Disease Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zohreh Vahidi
- Division of Inflammation and Inflammatory Diseases, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nafiseh Tabasi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rastin
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
das D Ribeiro JC, Drumond MM, Mancha-Agresti P, Guimarães JPF, da C Ferreira D, Martins MIA, de M Murata PM, de Carvalho AC, Pereira RT, Ribeiro Júnior V, de C Azevedo VA, de P Naves L. Diets Supplemented with Probiotics Improve the Performance of Broilers Exposed to Heat Stress from 15 Days of Age. Probiotics Antimicrob Proteins 2023; 15:1327-1341. [PMID: 36066817 DOI: 10.1007/s12602-022-09989-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2022] [Indexed: 01/10/2023]
Abstract
The poultry sector demands alternative additives to antibiotics that can be used as performance enhancers. Therefore, this experiment was conducted to evaluate the probiotics effects on performance, intestinal health, and redox status of 720 broilers exposed to heat stress from 15 days of age. Eight dietary treatments were evaluated: basal diet (BD) without antibiotic and probiotic (T1); BD supplemented with antibiotic zinc bacitracin (T2), BD supplemented with commercial probiotic of Bacillus subtilis DSM 17,299 (T3), BD supplemented with non-commercial probiotic of Lactococcus lactis NCDO 2118, Lactobacillus delbrueckii CNRZ 327, Escherichia coli CEC15, or Saccharomyces boulardii (T4 to T7), and BD simultaneously supplemented with the four non-commercial probiotics (T8). Feed intake, weight gain, and feed conversion were determined in the period from 1 to 42 days of age. Carcass and cuts yield, abdominal fat deposition, cloacal temperature, weight and length of intestine, activity of myeloperoxidase and eosinophilic peroxidase enzymes in the jejunum, jejunal histomorphometry, relative gene expression in the jejunum (occludin, zonulin, interleukin-8, cholecystokinin, ghrelin, and heat shock protein-70), and liver (heat shock protein-70), in addition to malondialdehyde level and superoxide dismutase activity in the intestine, liver, and blood, were measured in broilers at 42 days old. As main results, broilers fed T1 diet exhibited lower weight gain (3.222 kg) and worse feed conversion (1.70 kg/kg). However, diets containing non-commercial probiotics resulted in up to 3.584 kg of weight gain and improved feed conversion by up to 10%, similar to that observed for broilers of the T2 and T3 groups.
Collapse
Affiliation(s)
- Jéssica C das D Ribeiro
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, Minas Gerais, 37200-900, Brazil
| | - Mariana M Drumond
- Departamento de Ciências Biológicas, Centro Federal de Educação Tecnológica de Minas Gerais, Minas Gerais, Belo Horizonte, 30421-169, Brazil
| | - Pamela Mancha-Agresti
- Departamento de Ciências Biológicas, Centro Federal de Educação Tecnológica de Minas Gerais, Minas Gerais, Belo Horizonte, 30421-169, Brazil
| | - João P F Guimarães
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, Minas Gerais, 37200-900, Brazil
| | - Daiane da C Ferreira
- Departamento de Medicina Veterinária, Universidade Federal de Lavras, Lavras, Minas Gerais, 37200-900, Brazil
| | - Maria I A Martins
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, Minas Gerais, 37200-900, Brazil
| | - Pedro M de M Murata
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, Minas Gerais, 37200-900, Brazil
| | - Andressa C de Carvalho
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, Minas Gerais, 37200-900, Brazil
| | - Raquel T Pereira
- Departamento de Zootecnia, Universidade de São Paulo-ESALQ, Piracicaba, São Paulo, 13418-900, Brazil
| | - Valdir Ribeiro Júnior
- Departamento de Zootecnia, Universidade Federal de Sergipe, Nossa Senhora da Glória, Sergipe, 49680-000, Brazil
| | - Vasco A de C Azevedo
- Departamento de Genética, Universidade Federal de Minas Gerais, Minas Gerais, Ecologia e Evolução, Belo Horizonte, 31270-901, Brazil
| | - Luciana de P Naves
- Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, Minas Gerais, 37200-900, Brazil.
| |
Collapse
|
11
|
Mirfeizi Z, Mahmoudi M, Faridzadeh A. Probiotics as a complementary treatment in systemic lupus erythematosus: A systematic review. Health Sci Rep 2023; 6:e1640. [PMID: 37877130 PMCID: PMC10591540 DOI: 10.1002/hsr2.1640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/14/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that primarily affects young women. SLE has no recognized etiology but it is believed to be triggered by a number of factors, including genetic predisposition, hormonal influences, and environmental conditions. Dysbiosis in the gut microbiota has emerged as a potential mechanism connecting the intestinal microbiome to the breakdown of self-tolerance and chronic inflammation. This review aims to investigate the role of probiotics in modulating the gut microbiome and their potential therapeutic benefits in managing SLE, providing insights for future research and clinical practice. Methods We conducted a thorough search for papers published up to June 2023 in databases such as PubMed/MEDLINE, Web of Science, Scopus, and Cochrane Library. Results The systematic review identified 22 articles examining the effects of probiotics on SLE. These studies-which include in vivo tests, in vitro research, and clinical trials-indicate that probiotics may be effective against inflammation, and improve immunological responses and metabolic profiles in SLE patients. Most in vivo studies were assessed as medium to high quality, while the randomized controlled trial was deemed of high quality. Conclusion According to the findings of our systematic review, probiotics may be used in conjunction with other treatments to manage SLE. Nonetheless, current data is limited, and more randomized controlled trials would be required to fully examine their effectiveness.
Collapse
Affiliation(s)
- Zahra Mirfeizi
- Rheumatology Department, Rheumatic Diseases Research CenterMashhad University of Medical SciencesMashhadIran
| | - Mahmoud Mahmoudi
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Immunology and Allergy, School of MedicineMashhad University of Medical SciencesMashhadIran
| | - Arezoo Faridzadeh
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Immunology and Allergy, School of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
12
|
Illikoud N, Mantel M, Rolli-Derkinderen M, Gagnaire V, Jan G. Dairy starters and fermented dairy products modulate gut mucosal immunity. Immunol Lett 2022; 251-252:91-102. [DOI: 10.1016/j.imlet.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2022]
|
13
|
Paraprobiotics and Postbiotics of Lactobacillus delbrueckii CIDCA 133 Mitigate 5-FU-Induced Intestinal Inflammation. Microorganisms 2022; 10:microorganisms10071418. [PMID: 35889136 PMCID: PMC9324481 DOI: 10.3390/microorganisms10071418] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 12/02/2022] Open
Abstract
Intestinal mucositis is a commonly reported side effect in oncology practice. Probiotics are considered an excellent alternative therapeutic approach to this debilitating condition; however, there are safety questions regarding the viable consumption of probiotics in clinical practice due to the risks of systemic infections, especially in immune-compromised patients. The use of heat-killed or cell-free supernatants derived from probiotic strains has been evaluated to minimize these adverse effects. Thus, this work evaluated the anti-inflammatory properties of paraprobiotics (heat-killed) and postbiotics (cell-free supernatant) of the probiotic Lactobacillus delbrueckii CIDCA 133 strain in a mouse model of 5-Fluorouracil drug-induced mucositis. Administration of paraprobiotics and postbiotics reduced the neutrophil cells infiltrating into the small intestinal mucosa and ameliorated the intestinal epithelium architecture damaged by 5-FU. These ameliorative effects were associated with a downregulation of inflammatory markers (Tlr2, Nfkb1, Il12, Il17a, Il1b, Tnf), and upregulation of immunoregulatory Il10 cytokine and the epithelial barrier markers Ocln, Cldn1, 2, 5, Hp and Muc2. Thus, heat-killed L. delbrueckii CIDCA 133 and supernatants derived from this strain were shown to be effective in reducing 5-FU-induced inflammatory damage, demonstrating them to be an alternative approach to the problems arising from the use of live beneficial microorganisms in clinical practice.
Collapse
|
14
|
Peng X, Ed-Dra A, Yue M. Whole genome sequencing for the risk assessment of probiotic lactic acid bacteria. Crit Rev Food Sci Nutr 2022; 63:11244-11262. [PMID: 35694810 DOI: 10.1080/10408398.2022.2087174] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Probiotic bacteria exhibit beneficial effects on human and/or animal health, and have been widely used in foods and fermented products for decades. Most probiotics consist of lactic acid bacteria (LAB), which are used in the production of various food products but have also been shown to have the ability to prevent certain diseases. With the expansion of applications for probiotic LAB, there is an increasing concern with regard to safety, as cases with adverse effects, i.e., severe infections, transfer of antimicrobial resistance genes, etc., can occur. Currently, in vitro assays remain the primary way to assess the properties of LAB. However, such methodologies are not meeting the needs of strain risk assessment on a high-throughput scale, in the context of the evolving concept of food safety. Analyzing the complete genetic information, including potential virulence genes and other determinants with a negative impact on health, allows for assessing the safe use of the product, for which whole-genome sequencing (WGS) of individual LAB strains can be employed. Genomic data can also be used to understand subtle differences in the strain level important for beneficial effects, or protect patents. Here, we propose that WGS-based bioinformatics analyses are an ideal and cost-effective approach for the initial in silico microbial risk evaluation, while the technique may also increase our understanding of LAB strains for food safety and probiotic property evaluation.
Collapse
Affiliation(s)
- Xianqi Peng
- Department of Veterinary Medicine & Institute of Preventive Veterinary Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | | | - Min Yue
- Department of Veterinary Medicine & Institute of Preventive Veterinary Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, China
- Hainan Institute of Zhejiang University, Sanya, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou, China
| |
Collapse
|
15
|
De Jesus LCL, Aburjaile FF, Sousa TDJ, Felice AG, Soares SDC, Alcantara LCJ, Azevedo VADC. Genomic Characterization of Lactobacillus delbrueckii Strains with Probiotics Properties. FRONTIERS IN BIOINFORMATICS 2022; 2:912795. [PMID: 36304288 PMCID: PMC9580953 DOI: 10.3389/fbinf.2022.912795] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/16/2022] [Indexed: 01/22/2023] Open
Abstract
Probiotics are health-beneficial microorganisms with mainly immunomodulatory and anti-inflammatory properties. Lactobacillus delbrueckii species is a common bacteria used in the dairy industry, and their benefits to hosting health have been reported. This study analyzed the core genome of nine strains of L. delbrueckii species with documented probiotic properties, focusing on genes related to their host health benefits. For this, a combined methodology including several software and databases (BPGA, SPAAN, BAGEL4, BioCyc, KEEG, and InterSPPI) was used to predict the most important characteristics related to L. delbrueckii strains probiose. Comparative genomics analyses revealed that L. delbrueckii probiotic strains shared essential genes related to acid and bile stress response and antimicrobial activity. Other standard features shared by these strains are surface layer proteins and extracellular proteins-encoding genes, with high adhesion profiles that interacted with human proteins of the inflammatory signaling pathways (TLR2/4-MAPK, TLR2/4-NF-κB, and NOD-like receptors). Among these, the PrtB serine protease appears to be a strong candidate responsible for the anti-inflammatory properties reported for these strains. Furthermore, genes with high proteolytic and metabolic activity able to produce beneficial metabolites, such as acetate, bioactive peptides, and B-complex vitamins were also identified. These findings suggest that these proteins can be essential in biological mechanisms related to probiotics’ beneficial effects of these strains in the host.
Collapse
Affiliation(s)
- Luís Cláudio Lima De Jesus
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Flávia Figueira Aburjaile
- Department of Preventive Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Thiago De Jesus Sousa
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andrei Giacchetto Felice
- Department of Immunology, Microbiology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Siomar De Castro Soares
- Department of Immunology, Microbiology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Luiz Carlos Junior Alcantara
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Flavivirus Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- *Correspondence: Luiz Carlos Junior Alcantara, ; Vasco Ariston De Carvalho Azevedo,
| | - Vasco Ariston De Carvalho Azevedo
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Luiz Carlos Junior Alcantara, ; Vasco Ariston De Carvalho Azevedo,
| |
Collapse
|
16
|
Alatawi H, Mosli M, Saadah OI, Annese V, Al-Hindi R, Alatawy M, Al-Amrah H, Alshehri D, Bahieldin A, Edris S. Attributes of intestinal microbiota composition and their correlation with clinical primary non-response to anti-TNF-α agents in inflammatory bowel disease patients. Bosn J Basic Med Sci 2022; 22:412-426. [PMID: 34761733 PMCID: PMC9162754 DOI: 10.17305/bjbms.2021.6436] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
The largest microbial aggregation in the human body exists in the gastrointestinal tract. The microbiota in the host gastrointestinal tract comprises a diverse ecosystem, and the intestinal microbiota plays a vital role in maintaining gut homeostasis. This study aims to examine whether the gut microbiota influences unresponsiveness to anti-TNF-α treatments in primary nonresponder patients, and consequently identify the responsible microbes as biomarkers of unresponsiveness. Stool samples were collected from a cohort of patients with an established diagnosis of IBD, either ulcerative colitis (UC) or Crohn's disease (CD), following completion of the induction phase of anti TNF therapy. 16S rRNA sequencing analysis was used to examine the pattern of microbiota communities in fecal samples. The quality and quantity of fecal microbiota were compared in responder and primary nonresponder IBD patients following anti-TNF-α therapy. As per our hypothesis, a difference in gut microbiome composition between the two patient subgroups was observed. A decreased abundance of short-chain fatty acid (SCFA)-producing bacteria, including Anaerostipes, Coprococcus, Lachnospira, Roseburia, and Ruminococcus, was detected in non-responsive patients, which was the hallmark of dysbiosis. Biomarkers of dysbiosis that were identified as predictors of clinical nonresponse, included Klebsiella, Eubacteriaceae, RF32, Bifidobacterium_animalis, and Muribaculaceae-previously known as S24-7. Signature biomarkers showed dramatic alteration in the composition of gut microbiota in patients who demonstrated primary nonresponse to anti-TNF-α agents. Dysbiosis, with features including a dropped biodiversity, augmentation in opportunistic pathogenic microbiota, and a lack of SCFA-producing bacteria, is a prominent feature of the microbiome of primary nonresponders to anti-TNF-α therapy.
Collapse
Affiliation(s)
- Hanan Alatawi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, University College of Haqel, University of Tabuk, Tabuk, Saudi Arabia
| | - Mahmoud Mosli
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Inflammatory Bowel Disease Research Group, King Abduklaziz University, Jeddah, Saudi Arabia
| | - Omar I. Saadah
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Inflammatory Bowel Disease Research Group, King Abduklaziz University, Jeddah, Saudi Arabia
| | - Vito Annese
- Department of Internal Medicine, Fakeeh University Hospital, Dubai, United Arab Emirates
| | - Rashad Al-Hindi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Marfat Alatawy
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, University College of Haqel, University of Tabuk, Tabuk, Saudi Arabia
| | - Hadba Al-Amrah
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Dikhnah Alshehri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, University College of Haqel, University of Tabuk, Tabuk, Saudi Arabia
| | - Ahmad Bahieldin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Genetics, Ain Shams University, Cairo, Egypt
| | - Sherif Edris
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al Jawhara Albrahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
17
|
Zheng S, Song J, Qin X, Yang K, Liu M, Yang C, Nyachoti CM. Dietary supplementation of red-osier dogwood polyphenol extract changes the ileal microbiota structure and increases Lactobacillus in a pig model. AMB Express 2021; 11:145. [PMID: 34714436 PMCID: PMC8556438 DOI: 10.1186/s13568-021-01303-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/16/2021] [Indexed: 01/22/2023] Open
Abstract
Red-osier dogwood (ROD) extract contains a lot of polyphenols that have the potential for modulation of gut microbiota. However, little information is available about its prebiotic properties. This study investigated the impact of ROD polyphenol extract on the ileal microbiota with dietary supplementation of ROD polyphenol extract in a pig model. The data indicated that supplementation of ROD polyphenol extract significantly increased class Bacilli, order Lactobacillales and family lactobacillaceae. Within family lactobacillaceae, Lactobacillus was the main responder by increasing from 5.92% to 35.09%. Further analysis showed that ROD polyphenol extract improved two species Lactobacillus delbrueckii and Lactobacillus mucus. The results of this study suggested that ROD polyphenol extract has the potential to play prebiotic role and confer health benefit through modifying gut microbiota.
Collapse
|
18
|
Safety Evaluation of Lactobacillus delbrueckii subsp. lactis CIDCA 133: a Health-Promoting Bacteria. Probiotics Antimicrob Proteins 2021; 14:816-829. [PMID: 34403080 DOI: 10.1007/s12602-021-09826-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2021] [Indexed: 12/12/2022]
Abstract
Lactobacillus delbrueckii subsp. lactis CIDCA is a new potential probiotic strain whose molecular basis attributed to the host's benefit has been reported. This study investigated the safety aspects of Lactobacillus delbrueckii subsp. lactis CIDCA 133 based on whole-genome sequence and phenotypic analysis to avoid future questions about the harmful effects of this strain consumption. Genomic analysis showed that L. delbrueckii subsp. lactis CIDCA 133 harbors virulence, harmful metabolites, and antimicrobial resistance-associated genes. However, none of these genetic elements is flanked or located within prophage regions and plasmid sequence. At a phenotypic level, it was observed L. delbrueckii subsp. lactis CIDCA 133 antimicrobial resistance to aminoglycosides streptomycin and gentamicin antibiotics, but no hemolytic and mucin degradation activity was exhibited by strain. Furthermore, no adverse effects were observed regarding mice clinical and histopathological analysis after the strain consumption (5 × 107 CFU/mL). Overall, these findings reveal the safety of Lactobacillus delbrueckii subsp. lactis CIDCA 133 for consumption and future probiotic applications.
Collapse
|
19
|
de Jesus LCL, Drumond MM, Aburjaile FF, Sousa TDJ, Coelho-Rocha ND, Profeta R, Brenig B, Mancha-Agresti P, Azevedo V. Probiogenomics of Lactobacillus delbrueckii subsp. lactis CIDCA 133: In Silico, In Vitro, and In Vivo Approaches. Microorganisms 2021; 9:microorganisms9040829. [PMID: 33919849 PMCID: PMC8070793 DOI: 10.3390/microorganisms9040829] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 11/16/2022] Open
Abstract
Lactobacillus delbrueckii subsp. lactis CIDCA 133 (CIDCA 133) has been reported as a potential probiotic strain, presenting immunomodulatory properties. This study investigated the possible genes and molecular mechanism involved with a probiotic profile of CIDCA 133 through a genomic approach associated with in vitro and in vivo analysis. Genomic analysis corroborates the species identification carried out by the classical microbiological method. Phenotypic assays demonstrated that the CIDCA 133 strain could survive acidic, osmotic, and thermic stresses. In addition, this strain shows antibacterial activity against Salmonella Typhimurium and presents immunostimulatory properties capable of upregulating anti-inflammatory cytokines Il10 and Tgfb1 gene expression through inhibition of Nfkb1 gene expression. These reported effects can be associated with secreted, membrane/exposed to the surface and cytoplasmic proteins, and bacteriocins-encoding genes predicted in silico. Furthermore, our results showed the genes and the possible mechanisms used by CIDCA 133 to produce their beneficial host effects and highlight its use as a probiotic microorganism.
Collapse
Affiliation(s)
- Luís Cláudio Lima de Jesus
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
| | - Mariana Martins Drumond
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Departamento de Ciências Biológicas, Belo Horizonte 31421-169, Brazil;
| | - Flávia Figueira Aburjaile
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
- Laboratório de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| | - Thiago de Jesus Sousa
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
| | - Nina Dias Coelho-Rocha
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
| | - Rodrigo Profeta
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
| | - Bertram Brenig
- Institute of Veterinary Medicine, University of Göttingen, D-37077 Göttingen, Germany;
| | | | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (L.C.L.d.J.); (F.F.A.); (T.d.J.S.); (N.D.C.-R.); (R.P.)
- Correspondence:
| |
Collapse
|
20
|
Cordeiro BF, Alves JL, Belo GA, Oliveira ER, Braga MP, da Silva SH, Lemos L, Guimarães JT, Silva R, Rocha RS, Jan G, Le Loir Y, Silva MC, Freitas MQ, Esmerino EA, Gala-García A, Ferreira E, Faria AMC, Cruz AG, Azevedo V, do Carmo FLR. Therapeutic Effects of Probiotic Minas Frescal Cheese on the Attenuation of Ulcerative Colitis in a Murine Model. Front Microbiol 2021; 12:623920. [PMID: 33737918 PMCID: PMC7960676 DOI: 10.3389/fmicb.2021.623920] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/18/2021] [Indexed: 01/14/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) constitute disturbances of gastrointestinal tract that cause irreversible changes in the structure and function of tissues. Ulcerative colitis (UC), the most frequent IBD in the population, is characterized by prominent inflammation of the human colon. Functional foods containing probiotic bacteria have been studied as adjuvants to the treatment or prevention of IBDs. The selected probiotic strain Lactococcus lactis NCDO 2118 (L. lactis NCDO 2118) exhibits immunomodulatory effects, with promising results in UC mouse model induced by dextran sodium sulfate (DSS). Additionally, cheese is a dairy food that presents high nutritional value, besides being a good delivery system that can be used to improve survival and enhance the therapeutic effects of probiotic bacteria in the host. Therefore, this work investigated the probiotic therapeutic effects of an experimental Minas Frescal cheese containing L. lactis NCDO 2118 in DSS-induced colitis in mice. During colitis induction, mice that consumed the probiotic cheese exhibited reduced in the severity of colitis, with attenuated weight loss, lower disease activity index, limited shortening of the colon length, and reduced histopathological score. Moreover, probiotic cheese administration increased gene expression of tight junctions’ proteins zo-1, zo-2, ocln, and cln-1 in the colon and increase IL-10 release in the spleen and lymph nodes. In this way, this work demonstrates that consumption of probiotic Minas Frescal cheese, containing L. lactis NCDO 2118, prevents the inflammatory process during DSS-induced colitis in mice, opening perspectives for the development of new probiotic functional foods for personalized nutrition in the context of IBD.
Collapse
Affiliation(s)
- Bárbara F Cordeiro
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Juliana L Alves
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Giovanna A Belo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Emiliano R Oliveira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Marina P Braga
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Sara H da Silva
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Luisa Lemos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.,Department of Infectious Diseases, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Jonas T Guimarães
- Faculdade de Medicina Veterinária, Universidade Federal Fluminense (UFF), Niterói, Brazil
| | - Ramon Silva
- Faculdade de Medicina Veterinária, Universidade Federal Fluminense (UFF), Niterói, Brazil.,Departamento de Alimentos, Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Rio de Janeiro, Brazil
| | - Ramon S Rocha
- Faculdade de Medicina Veterinária, Universidade Federal Fluminense (UFF), Niterói, Brazil.,Departamento de Alimentos, Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Rio de Janeiro, Brazil
| | - Gwénaël Jan
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| | - Yves Le Loir
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| | - Marcia Cristina Silva
- Departamento de Alimentos, Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Rio de Janeiro, Brazil
| | - Mônica Q Freitas
- Faculdade de Medicina Veterinária, Universidade Federal Fluminense (UFF), Niterói, Brazil
| | - Erick A Esmerino
- Faculdade de Medicina Veterinária, Universidade Federal Fluminense (UFF), Niterói, Brazil
| | - Alfonso Gala-García
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Enio Ferreira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Maria C Faria
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Adriano G Cruz
- Departamento de Alimentos, Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Rio de Janeiro, Brazil
| | - Vasco Azevedo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Fillipe L R do Carmo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.,INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| |
Collapse
|
21
|
Minj J, Chandra P, Paul C, Sharma RK. Bio-functional properties of probiotic Lactobacillus: current applications and research perspectives. Crit Rev Food Sci Nutr 2020; 61:2207-2224. [PMID: 32519883 DOI: 10.1080/10408398.2020.1774496] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lactic acid bacteria as a starter culture are very important component in the fermentation process of dairy and food industry. Application of lactic acid bacteria as probiotic bacteria adds more functionality to the developed product. Gut colonizing bacteria have attractive benefits related to human health. Bio-functional properties such as antimicrobial activity, anti-inflammatory, ACE-inhibitory, antioxidant, antidiarrheal, antiviral, immunomodulatory, hypocholesterolemic, anti-diabetic and anti-cancer activities are the most applicable research areas of lactic acid bacteria. Different strains of Lactobacillus are generally consumed as probiotics and colonize the gastrointestinal tract. Sometimes these bacteria may possess antimicrobial activity and may positively influence the effect of antibiotics. Use of Lactobacillus spp. for the development of functional foods is one of the promising areas of current research and applications. Individual bacterial species have unique biological activity, which may vary from strains to strains and identification of this uniqueness could be helpful in the development of functional and therapeutic food products.
Collapse
Affiliation(s)
- Jagrani Minj
- Department of Food Science and Technology, Nebraska Innovation Campus (NIC), University of Nebraska, Lincoln, Nebraska, USA
| | | | - Catherine Paul
- Department of Food Science and Technology, Nebraska Innovation Campus (NIC), University of Nebraska, Lincoln, Nebraska, USA
| | - Rakesh Kumar Sharma
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan, India
| |
Collapse
|
22
|
Rabah H, do Carmo FLR, Carvalho RDDO, Cordeiro BF, da Silva SH, Oliveira ER, Lemos L, Cara DC, Faria AMC, Garric G, Harel-Oger M, Le Loir Y, Azevedo V, Bouguen G, Jan G. Beneficial Propionibacteria within a Probiotic Emmental Cheese: Impact on Dextran Sodium Sulphate-Induced Colitis in Mice. Microorganisms 2020; 8:E380. [PMID: 32156075 PMCID: PMC7142753 DOI: 10.3390/microorganisms8030380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUNDS AND AIMS Inflammatory Bowel Diseases (IBD), including Ulcerative Colitis (UC), coincide with alterations in the gut microbiota. Consumption of immunomodulatory strains of probiotic bacteria may induce or prolong remission in UC patients. Fermented foods, including cheeses, constitute major vectors for bacteria consumption. New evidences revealed anti-inflammatory effects in selected strains of Propionibacterium freudenreichii. We thus hypothesized that consumption of a functional cheese, fermented by such a strain, may exert a positive effect on IBD. METHODS We investigated the impact of cheese fermented by P. freudenreichii on gut inflammation. We developed an experimental single-strain cheese solely fermented by a selected immunomodulatory strain of P. freudenreichii, CIRM-BIA 129. We moreover produced, in industrial conditions, an Emmental cheese using the same strain, in combination with Lactobacillus delbrueckii CNRZ327 and Streptococcus thermophilus LMD-9, as starters. Consumption of both cheeses was investigated with respect to prevention of Dextran Sodium Sulphate (DSS)-induced colitis in mice. RESULTS Consumption of the single-strain experimental cheese, or of the industrial Emmental, both fermented by P. freudenreichii CIRM-BIA 129, reduced severity of subsequent DSS-induced colitis, weight loss, disease activity index and histological score. Both treatments, in a preventive way, reduced small bowel Immunoglobulin A (IgA) secretion, restored occludin gene expression and prevented induction of Tumor Necrosis Factor α (TNFα), Interferon γ (IFNγ) and Interleukin-17 (IL-17). CONCLUSIONS A combination of immunomodulatory strains of starter bacteria can be used to manufacture an anti-inflammatory cheese, as revealed in an animal model of colitis. This opens new perspectives for personalised nutrition in the context of IBD.
Collapse
Affiliation(s)
- Houem Rabah
- STLO, INRA, Agrocampus Ouest, 35 000 Rennes, France
- Pôle Agronomique Ouest, Régions Bretagne et Pays de la Loire, F-35 042 Rennes, France
| | - Fillipe Luiz Rosa do Carmo
- Departamento de Genética, Ecologia e evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais CP 486 CEP 31270-901, Brazil
| | | | - Barbara Fernandes Cordeiro
- Departamento de Genética, Ecologia e evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais CP 486 CEP 31270-901, Brazil
| | - Sara Heloisa da Silva
- Departamento de Genética, Ecologia e evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais CP 486 CEP 31270-901, Brazil
| | - Emiliano Rosa Oliveira
- Departamento de Genética, Ecologia e evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais CP 486 CEP 31270-901, Brazil
| | - Luisa Lemos
- Departamento de Genética, Ecologia e evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais CP 486 CEP 31270-901, Brazil
| | - Denise Carmona Cara
- Departamento de Genética, Ecologia e evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais CP 486 CEP 31270-901, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Genética, Ecologia e evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais CP 486 CEP 31270-901, Brazil
| | | | | | - Yves Le Loir
- STLO, INRA, Agrocampus Ouest, 35 000 Rennes, France
| | - Vasco Azevedo
- Departamento de Genética, Ecologia e evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais CP 486 CEP 31270-901, Brazil
| | - Guillaume Bouguen
- CHU Rennes, Univ Rennes, INSERM, CIC1414, Institut NUMECAN (Nutrition Metabolism and Cancer), F-35000 Rennes, France
| | - Gwénaël Jan
- STLO, INRA, Agrocampus Ouest, 35 000 Rennes, France
| |
Collapse
|
23
|
Probiotic Propionibacterium freudenreichii requires SlpB protein to mitigate mucositis induced by chemotherapy. Oncotarget 2019; 10:7198-7219. [PMID: 31921383 PMCID: PMC6944450 DOI: 10.18632/oncotarget.27319] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Propionibacterium freudenreichii CIRM-BIA 129 (P. freudenreichii wild type, WT) is a probiotic bacterium, which exerts immunomodulatory effects. This strain possesses extractable surface proteins, including SlpB, which are involved in anti-inflammatory effect and in adhesion to epithelial cells. We decided to investigate the impact of slpB gene mutation on immunomodulation in vitro and in vivo. In an in vitro assay, P. freudenreichii WT reduced expression of IL-8 (p<0.0001) and TNF-α (p<0.0001) cytokines in LPS-stimulated HT-29 cells. P. freudenreichii ΔslpB, lacking the SlpB protein, failed to do so. Subsequently, both strains were investigated in vivo in a 5-FU-induced mucositis mice model. Mucositis is a common side effect of cytotoxic chemotherapy with 5-FU, characterized by mucosal injury, inflammation, diarrhea, and weight loss. The WT strain prevented weight loss, reduced inflammation and consequently histopathological scores. Furthermore, it regulated key markers, including Claudin-1 (cld1, p<0.0005) and IL-17a (Il17a, p<0.0001) genes, as well as IL-12 (p<0.0001) and IL-1β (p<0.0429) cytokines levels. Mutant strain displayed opposite regulatory effect on cld1 expression and on IL-12 levels. This work emphasizes the importance of SlpB in P. freudenreichii ability to reduce mucositis inflammation. It opens perspectives for the development of probiotic products to decrease side effects of chemotherapy using GRAS bacteria with immunomodulatory surface protein properties.
Collapse
|
24
|
Cordeiro BF, Lemos L, Oliveira ER, Silva SH, Savassi B, Figueiroa A, Faria AMC, Ferreira E, Esmerino EA, Rocha RS, Freitas MQ, Silva MC, Cruz AG, do Carmo FLR, Azevedo V. Prato cheese containing Lactobacillus casei 01 fails to prevent dextran sodium sulphate-induced colitis. Int Dairy J 2019. [DOI: 10.1016/j.idairyj.2019.104551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
25
|
Ahn SI, Cho S, Choi NJ. Effect of dietary probiotics on colon length in an inflammatory bowel disease-induced murine model: A meta-analysis. J Dairy Sci 2019; 103:1807-1819. [PMID: 31785874 DOI: 10.3168/jds.2019-17356] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/11/2019] [Indexed: 12/15/2022]
Abstract
We investigated the effect of probiotic supplementation on inflammatory bowel disease (IBD) by meta-analysis. We included 30 studies to assess the effect of probiotic administration. We estimated the effect size using standardized mean difference, and we evaluated the statistical heterogeneity of the effect size using Cochran's Q test, followed by meta-ANOVA and meta-regression analysis to explain the heterogeneity of the effect size using a mixed-effects model. We conducted Egger's linear regression test to evaluate publication bias. Among the factors evaluated, colon length and myeloperoxidase showed the greatest Q statistic and I2 index, respectively. Colon length, transforming growth factor-β, IL-10, superoxide dismutase, and glutathione showed positive effect sizes in the fixed- and random-effects models. The others (spleen weight, tumor necrosis factor α, IL-1β, IL-6, IL-12, IL-17, IFN-γ, disease activity index, thiobarbituric acid reactive substances, nitric oxide, myeloperoxidase, malondialdehyde, histological score, and macroscopic inflammatory score) showed negative effect sizes in the fixed- and random-effects models. Probiotics showed a significant effect on all investigated factors, except IL-10. In meta-ANOVA and meta-regression analysis, Lactobacillus paracasei was the most effective probiotic for colon length. Lactobacillus paracasei, Lactobacillus reuteri, Lactobacillus fermentum, and a mixture of Lactobacillus bulgaricus and Saccharomyces boulardii (LC + SB) were effective for colon length, tumor necrosis factor α, IL-6, IL-10, IFN-γ, and disease activity index. Lactobacillus rhamnosus was most effective for IL-10 and IFN-γ. Dietary probiotics are effective in improving the symptoms of IBD. Although the results of this meta-analysis had some limitations due to a lack of animal experiments, they will be meaningful to people with IBD.
Collapse
Affiliation(s)
- Sung-Il Ahn
- Department of Animal Science, Jeonbuk National University, Jeonju 54896, Korea
| | - Sangbuem Cho
- Department of Animal Science, Jeonbuk National University, Jeonju 54896, Korea
| | - Nag-Jin Choi
- Department of Animal Science, Jeonbuk National University, Jeonju 54896, Korea.
| |
Collapse
|
26
|
Breyner NM, Vilas Boas PB, Fernandes G, de Carvalho RD, Rochat T, Michel ML, Chain F, Sokol H, de Azevedo M, Myioshi A, Azevedo VA, Langella P, Bermúdez-Humarán LG, Chatel JM. Oral delivery of pancreatitis-associated protein by Lactococcus lactis displays protective effects in dinitro-benzenesulfonic-acid-induced colitis model and is able to modulate the composition of the microbiota. Environ Microbiol 2019; 21:4020-4031. [PMID: 31325218 PMCID: PMC6899824 DOI: 10.1111/1462-2920.14748] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 07/14/2019] [Indexed: 12/26/2022]
Abstract
Antimicrobial peptides secreted by intestinal immune and epithelial cells are important effectors of innate immunity. They play an essential role in the maintenance of intestinal homeostasis by limiting microbial epithelium interactions and preventing unnecessary microbe‐driven inflammation. Pancreatitis‐associated protein (PAP) belongs to Regenerating islet‐derived III proteins family and is a C‐type (Ca+2 dependent) lectin. PAP protein plays a protective effect presenting anti‐inflammatory properties able to reduce the severity of colitis, preserving gut barrier and epithelial inflammation. Here, we sought to determine whether PAP delivered at intestinal lumen by recombinant Lactococcus lactis strain (LL‐PAP) before and after chemically induced colitis is able to reduce the severity in two models of colitis. After construction and characterization of our recombinant strains, we tested their effects in dinitro‐benzenesulfonic‐acid (DNBS) and Dextran sulfate sodium (DSS) colitis model. After the DNBS challenge, mice treated with LL‐PAP presented less severe colitis compared with PBS and LL‐empty‐treated mice groups. After the DSS challenge, no protective effects of LL‐PAP could be detected. We determined that after 5 days administration, LL‐PAP increase butyrate producer's bacteria, especially Eubacterium plexicaudatum. Based on our findings, we hypothesize that a treatment with LL‐PAP shifts the microbiota preventing the severity of colon inflammation in DNBS colitis model. These protective roles of LL‐PAP in DNBS colitis model might be through intestinal microbiota modulation.
Collapse
Affiliation(s)
- Natalia M Breyner
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, MG, Brazil
| | - Priscilla Bagano Vilas Boas
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, MG, Brazil
| | | | | | | | - Marie-Laure Michel
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Florian Chain
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Harry Sokol
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Marcela de Azevedo
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Anderson Myioshi
- Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, MG, Brazil
| | - Vasco A Azevedo
- Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, MG, Brazil
| | - Philippe Langella
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Luis G Bermúdez-Humarán
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Jean-Marc Chatel
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| |
Collapse
|
27
|
Khorasani S, Mahmoudi M, Kalantari MR, Lavi Arab F, Esmaeili S, Mardani F, Tabasi N, Rastin M. Amelioration of regulatory T cells by
Lactobacillus delbrueckii
and
Lactobacillus rhamnosus
in pristane‐induced lupus mice model. J Cell Physiol 2019. [DOI: 10.1002/jcp.27663 10.1002/jcp.27663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Sahar Khorasani
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Mahmoud Reza Kalantari
- Pathology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Fahimeh Lavi Arab
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed‐Alireza Esmaeili
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Fatemeh Mardani
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Nafiseh Tabasi
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Rastin
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
28
|
De Jesus LCL, Drumond MM, de Carvalho A, Santos SS, Martins FS, Ferreira Ê, Fernandes RS, de Barros ALB, do Carmo FL, Perez PF, Azevedo V, Mancha-Agresti P. Protective effect of Lactobacillus delbrueckii subsp. Lactis CIDCA 133 in a model of 5 Fluorouracil-Induced intestinal mucositis. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
29
|
Moreira TG, Horta LS, Gomes-Santos AC, Oliveira RP, Queiroz NMGP, Mangani D, Daniel B, Vieira AT, Liu S, Rodrigues AM, Gomes DA, Gabriely G, Ferreira E, Weiner HL, Rezende RM, Nagy L, Faria AMC. CLA-supplemented diet accelerates experimental colorectal cancer by inducing TGF-β-producing macrophages and T cells. Mucosal Immunol 2019; 12:188-199. [PMID: 30279515 DOI: 10.1038/s41385-018-0090-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/24/2018] [Accepted: 09/07/2018] [Indexed: 02/07/2023]
Abstract
Conjugated linoleic acid (CLA) has been shown to activate the nuclear receptor PPAR-γ and modulate metabolic and immune functions. Despite the worldwide use of CLA dietary supplementation, strong scientific evidence for its proposed beneficial actions are missing. We found that CLA-supplemented diet reduced mucosal damage and inflammatory infiltrate in the dextran sodium sulfate (DSS)-induced colitis model. Conditional deletion of PPAR-γ in macrophages from mice supplemented with CLA diet resulted in loss of this protective effect of CLA, suggesting a PPAR-γ-dependent mechanism mediated by macrophages. However, CLA supplementation significantly worsened colorectal tumor formation induced by azoxymethane and DSS by inducing macrophage and T-cell-producing TGF-β via PPAR-γ activation. Accordingly, either macrophage-specific deletion of PPAR-γ or in vivo neutralization of latency-associated peptide (LAP, a membrane-bound TGF-β)-expressing cells abrogated the protumorigenic effect of CLA. Thus, the anti-inflammatory properties of CLA are associated with prevention of colitis but also with development of colorectal cancer.
Collapse
Affiliation(s)
- T G Moreira
- Departamento de Alimentos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Campus Pampulha, 31270-901, Belo Horizonte, MG, Brazil. .,Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil. .,Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Biochemistry and Molecular Biology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary.
| | - L S Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - A C Gomes-Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - R P Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - N M G P Queiroz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - D Mangani
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - B Daniel
- Department of Biochemistry and Molecular Biology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary.,Department of Medicine, School of Medicine, Johns Hopkins All Children's Hospital, Johns Hopkins University, St. Petersburg, FL, 33701, USA
| | - A T Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - S Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - A M Rodrigues
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - D A Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - G Gabriely
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - E Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - H L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - R M Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - L Nagy
- Diabetes and Obesity Research Center, Sanford Burnham Medical Research Institute, Lake Nona, Orlando, FL, USA.,Department of Medicine, School of Medicine, Johns Hopkins All Children's Hospital, Johns Hopkins University, St. Petersburg, FL, 33701, USA
| | - A M C Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
30
|
Khorasani S, Mahmoudi M, Kalantari MR, Lavi Arab F, Esmaeili S, Mardani F, Tabasi N, Rastin M. Amelioration of regulatory T cells by
Lactobacillus delbrueckii
and
Lactobacillus rhamnosus
in pristane‐induced lupus mice model. J Cell Physiol 2018; 234:9778-9786. [DOI: 10.1002/jcp.27663] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Sahar Khorasani
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Mahmoud Reza Kalantari
- Pathology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Fahimeh Lavi Arab
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed‐Alireza Esmaeili
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Fatemeh Mardani
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Nafiseh Tabasi
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Rastin
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
31
|
Mardani F, Mahmoudi M, Esmaeili SA, Khorasani S, Tabasi N, Rastin M. In vivo study: Th1-Th17 reduction in pristane-induced systemic lupus erythematosus mice after treatment with tolerogenic Lactobacillus probiotics. J Cell Physiol 2018; 234:642-649. [PMID: 30078223 DOI: 10.1002/jcp.26819] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 04/30/2018] [Indexed: 12/17/2022]
Abstract
Uncontrolled inflammation in systemic lupus erythematosus (SLE) could cause dysfunction in multiple organs. T helper 17 (Th17) cells are a main branch of inflammatory responses in the pathogenesis of SLE, and by producing interleukin 17 (IL-17), represent a major functional tool in the progression of inflammation. Animal models provide a special field for better studies of the pathogenesis of diseases. Tolergenic probiotics could decrease inflammation in autoimmune diseases by modulating the immune system and maintaining homeostasis. The aim of this project was to evaluate the effects of Lactobacillus rhamnosus and Lactobacillus delbrueckii on Th17 cells and their related mediators in a pristane-induced BALB/c mice model of SLE. The mice were divided into pretreatment groups, which received probiotics or prednisolone at Day 0, and treatment groups, which received probiotics and prednisolone 2 months after injection. The presence of antinuclear antibody (ANA), anti-double-stranded DNA (anti-dsDNA), and anti-ribonucleoprotein (anti-RNP) and lipogranuloma was evaluated; also, the population of Th1-Th17 cells as well as interferon γ (IFN-γ), IL-17, and IL-10 levels, and the expression of RAR-related orphan related receptor gamma (RORγt) and IL-17 were determined. We observed that probiotics and prednisolone could delay SLE in pretreatment and treatment mice groups, with a reduction in ANA, anti-dsDNA, anti-RNP, and mass of lipogranuloma. Probiotics and prednisolone decreased the population of Th1-Th17 cells and reduced IFN-γ and IL-17 as inflammatory cytokines in the pretreatment and treatment groups in comparison with SLE-induced mice. Our results indicated that, due to their anti-inflammatory properties and reduction of Th17, Th1, and cytotoxic T lymphocyte (CTL) cells, the use of these probiotics could probably represent a new tool for the better management of SLE.
Collapse
Affiliation(s)
- Fatemeh Mardani
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committeen, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committeen, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Khorasani
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committeen, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nafiseh Tabasi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rastin
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Esmaeili S, Mahmoudi M, Rezaieyazdi Z, Sahebari M, Tabasi N, Sahebkar A, Rastin M. Generation of tolerogenic dendritic cells using
Lactobacillus rhamnosus
and
Lactobacillus delbrueckii
as tolerogenic probiotics. J Cell Biochem 2018; 119:7865-7872. [DOI: 10.1002/jcb.27203] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Seyed‐Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee, Mashhad University of Medical Sciences Mashhad Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Zahra Rezaieyazdi
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Sahebari
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences Mashhad Iran
| | - Nafiseh Tabasi
- Immunology Research Center, Mashhad University of Medical Sciences Mashhad Iran
| | - Amirhossein Sahebkar
- Neurogenic inflammation Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Rastin
- Immunology Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
33
|
Probiotic Mixture Protects Dextran Sulfate Sodium-Induced Colitis by Altering Tight Junction Protein Expressions and Increasing Tregs. Mediators Inflamm 2018; 2018:9416391. [PMID: 29849501 PMCID: PMC5925202 DOI: 10.1155/2018/9416391] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/27/2017] [Accepted: 03/15/2018] [Indexed: 02/07/2023] Open
Abstract
Bifico is a probiotic mixture containing Bifidobacterium, Lactobacillus acidophilus, and Enterococcus. Studies support that Bifico has a protective effect in experimental colitis (IL-10-deficient and TNBS) models and in patients with inflammatory bowel disease (IBD). However, the mechanism underlying the protective effects of this mixture of probiotic bacteria remains incompletely clear. Here, we investigated the effect of Bifico on intestinal inflammation. In an in vivo experiment, dextran sulfate sodium was used to induce colitis. Bifico treatment significantly attenuated the severity of colitis in this model. Bifico increased the expression of tight junction proteins (TJs). In addition, Bifico increased the number of Tregs, but reduced the number of total CD4+ T cells in the peripheral blood. Furthermore, the expression of colonic CD4 protein was decreased while the level of forkhead box P3 (Foxp3) was upregulated. These results suggested that Bifico exerts beneficial effects on experimental colitis by increasing the expressions of TJs, upregulating the number of Tregs, and reducing the total CD4+ T cell number in both colon and peripheral blood. The intestinal damage in the pretreated + treated-Bifico-colitis group was more severe than that in only the pretreated-Bifico-colitis group. This suggested that Bifico might aggravate intestinal damage when the mucosal barrier is impaired.
Collapse
|
34
|
Bazin T, Hooks KB, Barnetche T, Truchetet ME, Enaud R, Richez C, Dougados M, Hubert C, Barré A, Nikolski M, Schaeverbeke T. Microbiota Composition May Predict Anti-Tnf Alpha Response in Spondyloarthritis Patients: an Exploratory Study. Sci Rep 2018; 8:5446. [PMID: 29615661 PMCID: PMC5882885 DOI: 10.1038/s41598-018-23571-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/13/2018] [Indexed: 02/07/2023] Open
Abstract
Spondyloarthritis (SpA) pathophysiology remains largely unknown. While the association with genetic factors has been established for decades, the influence of gut microbiota is only an emerging direction of research. Despite the remarkable efficacy of anti-TNF-α treatments, non-responders are frequent and no predictive factors of patient outcome have been identified. Our objective was to investigate the modifications of intestinal microbiota composition in patients suffering from SpA three months after an anti-TNF-α treatment. We performed 16S rDNA sequencing of 38 stool samples from 19 spondyloarthritis patients before and three months after anti-TNF-α treatment onset. SpA activity was assessed at each time using ASDAS and BASDAI scores. Some modifications of the microbiota composition were observed after three months of anti-TNF-α treatment, but no specific taxon was modified, whatever the clinical response. We identified a particular taxonomic node before anti-TNF-α treatment that can predict the clinical response as a biomarker, with a higher proportion of Burkholderiales order in future responder patients. This study suggests a cross-influence between anti-TNF-α treatment and intestinal microbiota. If its results are confirmed on larger groups of patients, it may pave the way to the development of predictive tests suitable for clinical practices.
Collapse
Affiliation(s)
- Thomas Bazin
- Univ. Bordeaux, INRA, Mycoplasmal and chlamydial infections in humans, EA 3671, 33000, Bordeaux, France
- Bordeaux Hospital University Center, Department of Hepato-gastroenterology, 33600, Pessac, France
| | - Katarzyna B Hooks
- Univ. Bordeaux, Bordeaux Bioinformatics Center, 33000, Bordeaux, France
- Univ. Bordeaux, CNRS, Immunoconcept, UMR 5164, 33000, Bordeaux, France
| | - Thomas Barnetche
- Bordeaux Hospital University Center, Department of Rheumatology, 33000, Bordeaux, France
| | - Marie-Elise Truchetet
- Bordeaux Hospital University Center, Department of Rheumatology, 33000, Bordeaux, France
| | - Raphaël Enaud
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, FHU ACRONIM, Laboratoire 8 de Parasitologie-Mycologie, F-33000, Bordeaux, France
- CHU Bordeaux, Unité d'Hépatologie, Gastroentérologie et Nutrition Pédiatriques, CRCM Pédiatrique, Service 10 de Rhumatologie, Service d'Exploration Fonctionnelle Respiratoire, 33000, Bordeaux, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33000, Bordeaux, France
| | - Christophe Richez
- Bordeaux Hospital University Center, Department of Rheumatology, 33000, Bordeaux, France
| | - Maxime Dougados
- AP-HP, Cochin Hospital University Center, Department of Rheumatology, 75014, Paris, France
| | - Christophe Hubert
- Univ. Bordeaux, INSERM, Rare Diseases, genetic and metabolism, U1211, 33000, Bordeaux, France
- Univ. Bordeaux, Genome Transcriptome Facility of Bordeaux, 33000, Bordeaux, France
| | - Aurélien Barré
- Univ. Bordeaux, Bordeaux Bioinformatics Center, 33000, Bordeaux, France
| | - Macha Nikolski
- Univ. Bordeaux, Bordeaux Bioinformatics Center, 33000, Bordeaux, France
- Univ. Bordeaux, CNRS, LaBRI, UMR 5800, 33400, Talence, France
| | - Thierry Schaeverbeke
- Univ. Bordeaux, INRA, Mycoplasmal and chlamydial infections in humans, EA 3671, 33000, Bordeaux, France.
- Bordeaux Hospital University Center, Department of Rheumatology, 33000, Bordeaux, France.
| |
Collapse
|
35
|
Cortes-Perez N, Lozano-Ojalvo D, Maiga M, Hazebrouck S, Adel-Patient K. Intragastric administration of Lactobacillus casei BL23 induces regulatory FoxP3+RORγt+ T cells subset in mice. Benef Microbes 2017; 8:433-438. [DOI: 10.3920/bm2016.0174] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Many studies have highlighted the immunomodulatory properties of the probiotic strain Lactobacillus casei BL23. Recently, we demonstrated the ability of this strain to modulate the Th2-oriented immune response in a mouse model of cow’s milk allergy based on the induction of a Th17-biased immune response. The probiotic function of L. casei has been also linked to gut-microbiota modifications which could been potentially involved in the immune regulation; however, its precise mechanism of action remains poorly understood. In this regard, recent studies suggest that gut microbiota induces a specific subset of CD4+FoxP3+ Treg cells that also express RORγt+, the specific transcription factor of Th17 cells. This new type of regulatory T cells, called type 3 Treg, displays suppressive function during intestinal inflammation, participating in inflammation control. We thus explored the ability of L. casei BL23 to specifically induce type 3 Treg cells, both in vitro and in vivo. Our results showed that intragastric administration of L. casei BL23 to mice induces local and systemic FoxP3+ RORγt+ type 3 Treg cells that could then participate in the beneficial effects of L. casei BL23 in different intestinal-related disorders.
Collapse
Affiliation(s)
- N.G. Cortes-Perez
- UMR Service de Pharmacologie et d’Immuno-Analyse, CEA, INRA, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - D. Lozano-Ojalvo
- UMR Service de Pharmacologie et d’Immuno-Analyse, CEA, INRA, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - M.A. Maiga
- UMR Service de Pharmacologie et d’Immuno-Analyse, CEA, INRA, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - S. Hazebrouck
- UMR Service de Pharmacologie et d’Immuno-Analyse, CEA, INRA, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - K. Adel-Patient
- UMR Service de Pharmacologie et d’Immuno-Analyse, CEA, INRA, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| |
Collapse
|
36
|
Carvalho RDDO, do Carmo FLR, de Oliveira Junior A, Langella P, Chatel JM, Bermúdez-Humarán LG, Azevedo V, de Azevedo MS. Use of Wild Type or Recombinant Lactic Acid Bacteria as an Alternative Treatment for Gastrointestinal Inflammatory Diseases: A Focus on Inflammatory Bowel Diseases and Mucositis. Front Microbiol 2017; 8:800. [PMID: 28536562 PMCID: PMC5422521 DOI: 10.3389/fmicb.2017.00800] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/19/2017] [Indexed: 12/26/2022] Open
Abstract
The human gastrointestinal tract (GIT) is highly colonized by bacterial communities, which live in a symbiotic relationship with the host in normal conditions. It has been shown that a dysfunctional interaction between the intestinal microbiota and the host immune system, known as dysbiosis, is a very important factor responsible for the development of different inflammatory conditions of the GIT, such as the idiopathic inflammatory bowel diseases (IBD), a complex and multifactorial disorder of the GIT. Dysbiosis has also been implicated in the pathogenesis of other GIT inflammatory diseases such as mucositis usually caused as an adverse effect of chemotherapy. As both diseases have become a great clinical problem, many research groups have been focusing on developing new strategies for the treatment of IBD and mucositis. In this review, we show that lactic acid bacteria (LAB) have been capable in preventing and treating both disorders in animal models, suggesting they may be ready for clinical trials. In addition, we present the most current studies on the use of wild type or genetically engineered LAB strains designed to express anti-inflammatory proteins as a promising strategy in the treatment of IBD and mucositis.
Collapse
Affiliation(s)
| | - Fillipe L R do Carmo
- Federal University of Minas Gerais - Instituto de Ciências BiológicasBelo Horizonte, Brazil
| | | | - Philippe Langella
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Jean-Marc Chatel
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Luis G Bermúdez-Humarán
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Vasco Azevedo
- Federal University of Minas Gerais - Instituto de Ciências BiológicasBelo Horizonte, Brazil
| | - Marcela S de Azevedo
- Federal University of Minas Gerais - Instituto de Ciências BiológicasBelo Horizonte, Brazil
| |
Collapse
|
37
|
El Kafsi H, Loux V, Mariadassou M, Blin C, Chiapello H, Abraham AL, Maguin E, van de Guchte M. Unprecedented large inverted repeats at the replication terminus of circular bacterial chromosomes suggest a novel mode of chromosome rescue. Sci Rep 2017; 7:44331. [PMID: 28281695 PMCID: PMC5345009 DOI: 10.1038/srep44331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/07/2017] [Indexed: 12/29/2022] Open
Abstract
The first Lactobacillus delbrueckii ssp. bulgaricus genome sequence revealed the presence of a very large inverted repeat (IR), a DNA sequence arrangement which thus far seemed inconceivable in a non-manipulated circular bacterial chromosome, at the replication terminus. This intriguing observation prompted us to investigate if similar IRs could be found in other bacteria. IRs with sizes varying from 38 to 76 kbp were found at the replication terminus of all 5 L. delbrueckii ssp. bulgaricus chromosomes analysed, but in none of 1373 other chromosomes. They represent the first naturally occurring very large IRs detected in circular bacterial genomes. A comparison of the L. bulgaricus replication terminus regions and the corresponding regions without IR in 5 L. delbrueckii ssp. lactis genomes leads us to propose a model for the formation and evolution of the IRs. The DNA sequence data are consistent with a novel model of chromosome rescue after premature replication termination or irreversible chromosome damage near the replication terminus, involving mechanisms analogous to those proposed in the formation of very large IRs in human cancer cells. We postulate that the L. delbrueckii ssp. bulgaricus-specific IRs in different strains derive from a single ancestral IR of at least 93 kbp.
Collapse
Affiliation(s)
- Hela El Kafsi
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Valentin Loux
- MaIAGE, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | | | - Camille Blin
- MaIAGE, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Hélène Chiapello
- MaIAGE, INRA, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Anne-Laure Abraham
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Emmanuelle Maguin
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Maarten van de Guchte
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| |
Collapse
|
38
|
Mancha-Agresti P, Drumond MM, Carmo FLRD, Santos MM, Santos JSCD, Venanzi F, Chatel JM, Leclercq SY, Azevedo V. A New Broad Range Plasmid for DNA Delivery in Eukaryotic Cells Using Lactic Acid Bacteria: In Vitro and In Vivo Assays. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 4:83-91. [PMID: 28344994 PMCID: PMC5363290 DOI: 10.1016/j.omtm.2016.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/15/2016] [Indexed: 12/30/2022]
Abstract
Lactococcus lactis is well documented as a promising candidate for development of novel oral live vaccines. It has been broadly engineered for heterologous expression, as well as for plasmid expression vector delivery, directly inside eukaryotic cells, for DNA vaccine, or as therapeutic vehicle. This work describes the characteristics of a new plasmid, pExu (extra chromosomal unit), for DNA delivery using L. lactis and evaluates its functionality both by in vitro and in vivo assays. This plasmid exhibits the following features: (1) a theta origin of replication and (2) an expression cassette containing a multiple cloning site and a eukaryotic promoter, the cytomegalovirus (pCMV). The functionality of pExu:egfp was evaluated by fluorescence microscopy. The L. lactis MG1363 (pExu:egfp) strains were administered by gavage to Balb/C mice and the eGFP expression was monitored by fluorescence microscopy. The pExu vector has demonstrated an excellent stability either in L. lactis or in Escherichia coli. The eGFP expression at different times in in vitro assay showed that 15.8% of CHO cells were able to express the protein after transfection. The enterocytes of mice showed the expression of eGFP protein. Thus, L. lactis carrying the pExu is a good candidate to deliver genes into eukaryotic cells.
Collapse
Affiliation(s)
- Pamela Mancha-Agresti
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (ICB/UFMG), 31270-901 Belo Horizonte, Brazil
| | - Mariana Martins Drumond
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (ICB/UFMG), 31270-901 Belo Horizonte, Brazil; CEFET - Centro Federal de Educação Tecnológica de Minas Gerais, Coordenação de Ciências, Campus I, 30421-169 Belo Horizonte, Brazil
| | - Fillipe Luiz Rosa do Carmo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (ICB/UFMG), 31270-901 Belo Horizonte, Brazil
| | - Monica Morais Santos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (ICB/UFMG), 31270-901 Belo Horizonte, Brazil
| | | | - Franco Venanzi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Jean-Marc Chatel
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Sophie Yvette Leclercq
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel Dias (FUNED), Belo Horizonte, 30510-010 Minas Gerais, Brazil
| | - Vasco Azevedo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (ICB/UFMG), 31270-901 Belo Horizonte, Brazil
| |
Collapse
|
39
|
Uhde AK, Herder V, Akram Khan M, Ciurkiewicz M, Schaudien D, Teich R, Floess S, Baumgärtner W, Huehn J, Beineke A. Viral Infection of the Central Nervous System Exacerbates Interleukin-10 Receptor Deficiency-Mediated Colitis in SJL Mice. PLoS One 2016; 11:e0161883. [PMID: 27611574 PMCID: PMC5017624 DOI: 10.1371/journal.pone.0161883] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/12/2016] [Indexed: 12/16/2022] Open
Abstract
Theiler´s murine encephalomyelitis virus (TMEV)-infection is a widely used animal model for studying demyelinating disorders, including multiple sclerosis (MS). The immunosuppressive cytokine Interleukin (IL)-10 counteracts hyperactive immune responses and critically controls immune homeostasis in infectious and autoimmune disorders. In order to investigate the effect of signaling via Interleukin-10 receptor (IL-10R) in infectious neurological diseases, TMEV-infected SJL mice were treated with IL-10R blocking antibody (Ab) in the acute and chronic phase of the disease. The findings demonstrate that (i) Ab-mediated IL-10 neutralization leads to progressive colitis with a reduction in Foxp3+ regulatory T cells and increased numbers of CD8+CD44+ memory T cells as well as activated CD4+CD69+ and CD8+CD69+ T cells in uninfected mice. (ii) Concurrent acute TMEV-infection worsened enteric disease-mediated by IL-10R neutralization. Virus-triggered effects were associated with an enhanced activation of CD4+ T helper cells and CD8+ cytotoxic T lymphocytes and augmented cytokine expression. By contrast, (iii) IL-10R neutralization during chronic TMEV-infection was not associated with enhanced peripheral immunopathology but an increased CD3+ T cell influx in the spinal cord. IL-10R neutralization causes a breakdown in peripheral immune tolerance in genetically predisposed mice, which leads to immune-mediated colitis, resembling inflammatory bowel disease. Hyperactive immune state following IL-10R blockade is enhanced by central nervous system-restricted viral infection in a disease phase-dependent manner.
Collapse
Affiliation(s)
- Ann-Kathrin Uhde
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Vanessa Herder
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Muhammad Akram Khan
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
- Department of Pathobiology, Faculty of Veterinary & Animal Sciences, PMAS—Arid Agriculture University, Rawalpindi, Pakistan
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer—Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - René Teich
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Floess
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
- * E-mail:
| |
Collapse
|
40
|
Plé C, Breton J, Richoux R, Nurdin M, Deutsch SM, Falentin H, Hervé C, Chuat V, Lemée R, Maguin E, Jan G, Van de Guchte M, Foligné B. Combining selected immunomodulatory Propionibacterium freudenreichii
and Lactobacillus delbrueckii
strains: Reverse engineering development of an anti-inflammatory cheese. Mol Nutr Food Res 2015; 60:935-48. [DOI: 10.1002/mnfr.201500580] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/25/2015] [Accepted: 11/29/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Coline Plé
- Institut Pasteur de Lille, Lactic Acid Bacteria & Mucosal Immunity; Center for Infection and Immunity of Lille; Lille France
| | - Jérôme Breton
- Institut Pasteur de Lille, Lactic Acid Bacteria & Mucosal Immunity; Center for Infection and Immunity of Lille; Lille France
| | - Romain Richoux
- Actalia Produits Laitiers; Agrocampus Ouest; Rennes cedex France
| | - Marine Nurdin
- Actalia Produits Laitiers; Agrocampus Ouest; Rennes cedex France
| | - Stéphanie-Marie Deutsch
- Science et Technologie du Lait et de l’Œuf; INRA; Rennes France
- Agrocampus Ouest; Rennes France
| | - Hélène Falentin
- Science et Technologie du Lait et de l’Œuf; INRA; Rennes France
- Agrocampus Ouest; Rennes France
| | | | - Victoria Chuat
- Science et Technologie du Lait et de l’Œuf; INRA; Rennes France
- Agrocampus Ouest; Rennes France
- CIRM-BIA; Centre Internationale de Ressources Microbiennes-Bactéries d'Intérêt Alimentaire; Rennes France
| | | | - Emmanuelle Maguin
- INRA; Jouy-en-Josas France
- AgroParisTech; UMR Micalis; Jouy-en-Josas France
| | - Gwénaël Jan
- Science et Technologie du Lait et de l’Œuf; INRA; Rennes France
- Agrocampus Ouest; Rennes France
| | | | - Benoit Foligné
- Institut Pasteur de Lille, Lactic Acid Bacteria & Mucosal Immunity; Center for Infection and Immunity of Lille; Lille France
| |
Collapse
|
41
|
Lee HA, Bong YJ, Kim H, Jeong JK, Kim HY, Lee KW, Park KY. Effect of NanometricLactobacillus plantarumin Kimchi on Dextran Sulfate Sodium-Induced Colitis in Mice. J Med Food 2015; 18:1073-80. [DOI: 10.1089/jmf.2015.3509] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Hyun Ah Lee
- Kimchi Research Institute, Pusan National University, Busan, Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, Korea
| | - Yeon-Ju Bong
- Department of Food Science and Nutrition, Pusan National University, Busan, Korea
| | | | - Ji-Kang Jeong
- Department of Food Science and Nutrition, Pusan National University, Busan, Korea
| | - Hee-Young Kim
- Department of Food Science and Nutrition, Pusan National University, Busan, Korea
| | - Kwang-Won Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, Korea
| | - Kun-Young Park
- Kimchi Research Institute, Pusan National University, Busan, Korea
- Department of Food Science and Nutrition, Pusan National University, Busan, Korea
| |
Collapse
|
42
|
Boer MC, Joosten SA, Ottenhoff THM. Regulatory T-Cells at the Interface between Human Host and Pathogens in Infectious Diseases and Vaccination. Front Immunol 2015; 6:217. [PMID: 26029205 PMCID: PMC4426762 DOI: 10.3389/fimmu.2015.00217] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/20/2015] [Indexed: 12/20/2022] Open
Abstract
Regulatory T-cells (Tregs) act at the interface of host and pathogen interactions in human infectious diseases. Tregs are induced by a wide range of pathogens, but distinct effects of Tregs have been demonstrated for different pathogens and in different stages of infection. Moreover, Tregs that are induced by a specific pathogen may non-specifically suppress immunity against other microbes and parasites. Thus, Treg effects need to be assessed not only in homologous but also in heterologous infections and vaccinations. Though Tregs protect the human host against excessive inflammation, they probably also increase the risk of pathogen persistence and chronic disease, and the possibility of disease reactivation later in life. Mycobacterium leprae and Mycobacterium tuberculosis, causing leprosy and tuberculosis, respectively, are among the most ancient microbes known to mankind, and are master manipulators of the immune system toward tolerance and pathogen persistence. The majority of mycobacterial infections occur in settings co-endemic for viral, parasitic, and (other) bacterial coinfections. In this paper, we discuss recent insights in the activation and activity of Tregs in human infectious diseases, with emphasis on early, late, and non-specific effects in disease, coinfections, and vaccination. We highlight mycobacterial infections as important models of modulation of host responses and vaccine-induced immunity by Tregs.
Collapse
Affiliation(s)
- Mardi C Boer
- Department of Infectious Diseases, Leiden University Medical Center , Leiden , Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center , Leiden , Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
43
|
Genome Sequence of Lactobacillus delbrueckii subsp. lactis CNRZ327, a Dairy Bacterium with Anti-Inflammatory Properties. GENOME ANNOUNCEMENTS 2014; 2:2/4/e00328-14. [PMID: 25035318 PMCID: PMC4102855 DOI: 10.1128/genomea.00328-14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lactobacillus delbrueckii subsp. lactis CNRZ327 is a dairy bacterium with anti-inflammatory properties both in vitro and in vivo. Here, we report the genome sequence of this bacterium, which appears to contain no less than 215 insertion sequence (IS) elements, an exceptionally high number regarding the small genome size of the strain.
Collapse
|
44
|
Di Luccia B, Manzo N, Baccigalupi L, Calabrò V, Crescenzi E, Ricca E, Pollice A. Lactobacillus gasseri SF1183 affects intestinal epithelial cell survival and growth. PLoS One 2013. [PMID: 23894414 DOI: 10.1016/j.jff.2017.12.049] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
It is now commonly accepted that the intestinal microbiota plays a crucial role in the gut physiology and homeostasis, and that both qualitative and quantitative alterations in the compositions of the gut flora exert profound effects on the host's intestinal cells. In spite of this, the details of the interaction between commensal bacteria and intestinal cells are still largely unknown and only in few cases the molecular mechanisms have been elucidated. Here we analyze the effects of molecules produced and secreted by Lactobacillus gasseri SF1183 on human intestinal HCT116 cells. L. gasseri is a well known species of lactic acid bacteria, commonly associated to the human intestine and SF1183 is a human strain previously isolated from an ileal biopsy of an healthy volunteer. SF1183 produces and secretes, in a growth phase-dependent way, molecule(s) able to drastically interfere with HCT116 cell proliferation. Although several attempts to purify and identify the bioactive molecule(s) have been so far unsuccessful, a partial characterization has indicated that it is smaller than 3 kDa, thermostable and of proteinaceous nature. L. gasseri molecule(s) stimulate a G1-phase arrest of the cell cycle by up-regulation of p21WAF1 rendering cells protected from intrinsic and extrinsic apoptosis. A L. gasseri-mediated reduction of apoptosis and of cell proliferation could be relevant in protecting epithelial barrier integrity and helping in reconstituting tissutal homeostasis.
Collapse
Affiliation(s)
- Blanda Di Luccia
- Department of Biology, University of Naples Federico II-MSA-Via Cinthia, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|