1
|
Li L, Meng Z, Huang Y, Xu L, Chen Q, Qiao D, Yue X. Chronic Sleep Deprivation Causes Anxiety, Depression and Impaired Gut Barrier in Female Mice-Correlation Analysis from Fecal Microbiome and Metabolome. Biomedicines 2024; 12:2654. [PMID: 39767560 PMCID: PMC11673394 DOI: 10.3390/biomedicines12122654] [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: 10/20/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chronic sleep deprivation (CSD) plays an important role in mood disorders. However, the changes in the gut microbiota and metabolites associated with CSD-induced anxiety/depression-like behavior in female mice have not been determined. Due to the influence of endogenous hormone levels, females are more susceptible than males to negative emotions caused by sleep deprivation. Here, we aim to investigate how CSD changes the gut microbiota and behavior and uncover the relationship between CSD and gut microbiota and its metabolites in female mice. METHODS We used a 48-day sleep deprivation (SD) model using the modified multiple platform method (MMPM) to induce anxiety/depression-like behavior in female C57BL/6J mice and verified our results using the open field test, elevated plus maze, novel object recognition test, forced swim test, and tail suspension test. We collected fecal samples of mice for 16S rDNA sequencing and untargeted metabolomic analysis and colons for histopathological observation. We used Spearmen analysis to find the correlations between differential bacterial taxa, fecal metabolites, and behaviors. RESULTS Our study demonstrates that CSD induced anxiety/depressive-like behaviors in female mice. The results of 16S rDNA sequencing suggested that the relative abundance of the harmful bacteria g_ Rothia, g_ Streptococcus, g_ Pantoea, and g_ Klebsiella were significantly increased, while the beneficial bacteria g_ Rikenella, g_ Eubacterium]-xylanophilum-group, and g_ Eisenbergiella were significantly decreased after SD. Glycerophospholipid metabolism and glutathione metabolism were identified as key pathways in the fecal metabolism related to oxidative stress and inflammatory states of the intestine. Histological observation showed hyperplasia of epithelial cells, a decrease in goblet cells, and glandular atrophy of the colon in SD mice. There were correlations between some of the differential bacterial taxa, fecal metabolites, and behaviors. CONCLUSION In summary, we found that CSD induced anxiety/depression-like behavior, caused gut microbiota dysbiosis, altered fecal metabolism, and damaged the colon barrier in female mice.
Collapse
Affiliation(s)
| | | | | | | | | | - Dongfang Qiao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; (L.L.); (Z.M.); (Y.H.); (L.X.); (Q.C.)
| | - Xia Yue
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; (L.L.); (Z.M.); (Y.H.); (L.X.); (Q.C.)
| |
Collapse
|
2
|
Pottash AE, Levy D, Powsner EH, Pirolli NH, Kuo L, Solomon TJ, Nowak R, Wang J, Kronstadt SM, Jay SM. Enhanced Extracellular Vesicle Cargo Loading via microRNA Biogenesis Pathway Modulation. ACS Biomater Sci Eng 2024; 10:6286-6298. [PMID: 39305230 DOI: 10.1021/acsbiomaterials.4c00821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Extracellular vesicles (EVs) are physiological vectors for the intercellular transport of a variety of molecules. Among these, small RNAs, and especially microRNAs (miRNAs), have been identified as prevalent components, and there has thus been a robust investigation of EVs for therapeutic miRNAs delivery. However, intrinsic levels of EV-associated miRNAs are generally too low to enable efficient and effective therapeutic outcomes. We hypothesized that miRNA localization to EVs could be improved by limiting competing interactions that occur throughout the miRNA biogenesis process. Using miR-146a-5p as a model, modulation of transcription, nuclear export, and enzymatic cleavage steps of miRNA biogenesis were tested for impact on EV miRNA loading. Working in HEK293T cells, various alterations in the EV biogenesis pathway were shown to impact miRNA localization to EVs. The system was then applied in induced pluripotent stem cells (iPSCs), a more promising substrate for therapeutic EV production, and EVs were separated and assessed for anti-inflammatory efficacy in vitro and in a murine colitis model, where the preservation of function was validated. Overall, the results highlight necessary considerations when designing a cell culture system for the devoted production of miRNA-loaded EVs.
Collapse
Affiliation(s)
- Alex Eli Pottash
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Daniel Levy
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Emily H Powsner
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Nicholas H Pirolli
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Leo Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Talia J Solomon
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Raith Nowak
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Jacob Wang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Stephanie M Kronstadt
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
- Program in Molecular and Cell Biology, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
3
|
Lu J, Shen X, Li H, Du J. Recent advances in bacteria-based platforms for inflammatory bowel diseases treatment. EXPLORATION (BEIJING, CHINA) 2024; 4:20230142. [PMID: 39439496 PMCID: PMC11491310 DOI: 10.1002/exp.20230142] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/18/2024] [Indexed: 10/25/2024]
Abstract
Inflammatory bowel disease (IBD) is a recurring chronic inflammatory disease. Current treatment strategies are aimed at alleviating clinical symptoms and are associated with gastrointestinal or systemic adverse effects. New delivery strategies are needed for the treatment of IBD. Bacteria are promising biocarriers, which can produce drugs in situ and sense the gut in real time. Herein, we focus on recent studies of engineered bacteria used for IBD treatment and introduce the application of engineered bacteria in the diagnosis. On this basis, the current dilemmas and future developments of bacterial delivery systems are discussed.
Collapse
Affiliation(s)
- Jiaoying Lu
- Department of GastroenterologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Xinyuan Shen
- National Key Laboratory of Advanced Drug Delivery and Release SystemsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Department of BioengineeringUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Hongjun Li
- National Key Laboratory of Advanced Drug Delivery and Release SystemsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
| | - Juan Du
- Department of GastroenterologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
4
|
Perruzza L, Rezzonico Jost T, Raneri M, Gargari G, Palatella M, De Ponte Conti B, Seehusen F, Heckmann J, Viemann D, Guglielmetti S, Grassi F. Protection from environmental enteric dysfunction and growth improvement in malnourished newborns by amplification of secretory IgA. Cell Rep Med 2024; 5:101639. [PMID: 38959887 PMCID: PMC11293325 DOI: 10.1016/j.xcrm.2024.101639] [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: 12/10/2023] [Revised: 04/04/2024] [Accepted: 06/11/2024] [Indexed: 07/05/2024]
Abstract
Environmental enteric dysfunction (EED) is a condition associated with malnutrition that can progress to malabsorption and villous atrophy. Severe EED results in linear growth stunting, slowed neurocognitive development, and unresponsiveness to oral vaccines. Prenatal exposure to malnutrition and breast feeding by malnourished mothers replicates EED. Pups are characterized by deprivation of secretory IgA (SIgA) and altered development of the gut immune system and microbiota. Extracellular ATP (eATP) released by microbiota limits T follicular helper (Tfh) cell activity and SIgA generation in Peyer's patches (PPs). Administration of a live biotherapeutic releasing the ATP-degrading enzyme apyrase to malnourished pups restores SIgA levels and ameliorates stunted growth. SIgA is instrumental in improving the growth and intestinal immune competence of mice while they are continuously fed a malnourished diet. The analysis of microbiota composition suggests that amplification of endogenous SIgA may exert a dominant function in correcting malnourishment dysbiosis and its consequences on host organisms, irrespective of the actual microbial ecology.
Collapse
Affiliation(s)
- Lisa Perruzza
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland.
| | - Tanja Rezzonico Jost
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Matteo Raneri
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Giorgio Gargari
- Division of Food Microbiology and Bioprocesses, Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, 20133 Milan, Italy
| | - Martina Palatella
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Benedetta De Ponte Conti
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland; Graduate School of Cellular and Molecular Sciences, University of Bern, 3012 Bern, Switzerland
| | - Frauke Seehusen
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Julia Heckmann
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Dorothee Viemann
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; Cluster of Excellence RESIST (EXC 2355), Hannover Medical School, 30625 Hannover, Germany; Center for Infection Research, University Würzburg, 97080 Würzburg, Germany
| | - Simone Guglielmetti
- Department of Biotechnology and Biosciences (BtBs), University of Milano-Bicocca, 20126 Milan, Italy
| | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland.
| |
Collapse
|
5
|
Fu Y, Zhang C, Xie H, Wu Z, Tao Y, Wang Z, Gu M, Wei P, Lin S, Li R, He Y, Sheng J, Xu J, Wang J, Pan Y. Human umbilical cord mesenchymal stem cells alleviated TNBS-induced colitis in mice by restoring the balance of intestinal microbes and immunoregulation. Life Sci 2023; 334:122189. [PMID: 37865178 DOI: 10.1016/j.lfs.2023.122189] [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: 05/01/2023] [Revised: 09/07/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
AIMS Human umbilical cord mesenchymal stem cells (HUMSCs) have been documented to be effective for several immune disorders including inflammatory bowel diseases (IBD). However, it remains unclear how HUMSCs function in regulating immune responses and intestinal flora in the trinitrobenzene sulfonic acid (TNBS)-induced IBD model. MATERIALS AND METHODS We assessed the regulatory effects of HUMSCs on the gut microbiota, T lymphocyte subpopulations and related immune cytokines in the TNBS-induced IBD model. The mice were divided into the normal, TNBS, and HUMSC-treated groups. The effect of HUMSCs was evaluated by Hematoxylin and Eosin (H&E) staining, fluorescence-activated cell sorting (FACS), and enzyme-linked immunosorbent assay (ELISA) analyses. Metagenomics Illumina sequencing was conducted for fecal samples. KEY FINDINGS We demonstrated that the disease symptoms and pathological changes in the colon tissues of TNBS-induced colitis mice were dramatically ameliorated by HUMSCs, which improved the gut microbiota and rebalanced the immune system, increasing the abundance of healthy bacteria (such as Lactobacillus murinus and Lactobacillus johnsonii), the Firmicutes/Bacteroidetes ratio, and the proportion of Tregs; the Th1/Th17 ratio was decreased. Consistently, the expression levels of IFN-γ and IL-17 were significantly decreased, and transforming growth factor-β1 (TGF-β1) levels were significantly increased in the plasma of colitis mice HUMSC injection. SIGNIFICANCE Our experiment revealed that HUMSCs mitigate acute colitis by regulating the rebalance of Th1/Th17/Treg cells and related cytokines and remodeling the gut microbiota, providing potential future therapeutic targets in IBD.
Collapse
Affiliation(s)
- Yanxia Fu
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China
| | - Chen Zhang
- Chinese PLA General Hospital and Medical School, Beijing 100853, China; Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Hui Xie
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Zisheng Wu
- Chinese PLA General Hospital and Medical School, Beijing 100853, China
| | - Yurong Tao
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Ziyu Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Meng Gu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Panjian Wei
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Shuye Lin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Ruoran Li
- Chinese PLA General Hospital and Medical School, Beijing 100853, China
| | - Yuqi He
- Department of Gastroenterology, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Jianqiu Sheng
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Junfeng Xu
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Jinghui Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China.
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China.
| |
Collapse
|
6
|
Plavec TV, Klemenčič K, Kuchař M, Malý P, Berlec A. Secretion and surface display of binders of IL-23/IL-17 cytokines and their receptors in Lactococcus lactis as a therapeutic approach against inflammation. Eur J Pharm Sci 2023; 190:106568. [PMID: 37619953 DOI: 10.1016/j.ejps.2023.106568] [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: 05/16/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
The cytokine IL-23 activates the IL-23 receptor (IL-23R) and stimulates the differentiation of naïve T helper (Th) cells into a Th17 cell population that secretes inflammatory cytokines and chemokines. This IL-23/Th17 proinflammatory axis drives inflammation in Crohn's disease and ulcerative colitis and represents a therapeutic target of monoclonal antibodies. Non-immunoglobulin binding proteins based on the Streptococcus albumin-binding domain (ABD) provide a small protein alternative to monoclonal antibodies. They can be readily expressed in bacteria. Lactococcus lactis is a safe lactic acid bacterium that has previously been engineered as a vector for the delivery of recombinant therapeutic proteins to mucosal surfaces. Here, L. lactis was engineered to display or secrete ABD-variants against the IL-17 receptor (IL-17R). Its expression and functionality were confirmed with flow cytometry using specific antibody and recombinant IL-17R, respectively. In addition, L. lactis were engineered into multifunctional bacteria that simultaneously express two binders from pNBBX plasmid. First, binders of IL-17R were combined with binder of IL-17. Second, binders of IL-23R were combined with binders of IL-23. The dual functionality of the bacteria was confirmed by flow cytometry using corresponding targets, namely the recombinant receptors IL-17R and IL-23R or the p19 subunit of IL-23. Binding of IL-17 was confirmed by ELISA. With the latter, 97% of IL-17 was removed from solution by 2 × 109 recombinant bacteria. Moreover, multifunctional bacteria targeting IL-17/IL-17R prevented IL-17A-mediated activation of downstream signaling pathways in HEK-Blue IL-17 cell model. Thus, we have developed several multifunctional L. lactis capable of targeting multiple factors of the IL-23/Th17 proinflammatory axis. This represents a novel therapeutic strategy with synergistic potential for the treatment of intestinal inflammations.
Collapse
Affiliation(s)
- Tina Vida Plavec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, Ljubljana SI-1000, Slovenia; Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana SI-1000, Slovenia
| | - Kaja Klemenčič
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, Ljubljana SI-1000, Slovenia
| | - Milan Kuchař
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, v. v. i., Průmyslová 595, Vestec 252 50, Czech Republic
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, v. v. i., Průmyslová 595, Vestec 252 50, Czech Republic
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, Ljubljana SI-1000, Slovenia; Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana SI-1000, Slovenia.
| |
Collapse
|
7
|
Alves JL, Lemos L, Rodrigues NM, Pereira VB, Barros PAV, Canesso MCC, Guimarães MAF, Cara DC, Miyoshi A, Azevedo VA, Maioli TU, Gomes-Santos AC, Faria AMC. Immunomodulatory effects of different strains of Lactococcus lactis in DSS-induced colitis. Braz J Microbiol 2023; 54:1203-1215. [PMID: 36821043 PMCID: PMC10234881 DOI: 10.1007/s42770-023-00928-0] [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: 12/12/2021] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are gastrointestinal disorders characterized by a breakdown in intestinal homeostasis by inflammatory immune responses to luminal antigens. Novel strategies for ameliorating IBD have been proposed in many studies using animal models. Our group has demonstrated that administration of Lactococcus lactis NCDO 2118 can improve clinical parameters of colitis induced by oral administration of dextran sulphate sodium (DSS). However, it is not clear whether other strains of L. lactis can yield the same effect. The objective of present study was to analyze the effects of three different L. lactis strains (NCDO2118, IL1403 and MG1363) in the development of DSS-induced colitis in C57BL/6 mice. Acute colitis was induced in C57/BL6 mice by the administration of 2% DSS during 7 consecutive days. Body weight loss and shortening of colon length were observed in DSS-treated mice, and none of L. lactis strains had an impact in these clinical signs of colitis. On the other hand, all strains improved the global macroscopical disease index and prevented goblet cells depletion as well as the increase of intestinal permeability. TNF-α production was reduced in gut mucosa of L. lactis DSS-treated mice indicating a modulation of a critical pro-inflammatory response by all strains tested. However, only L. lactis NCDO2118 and MG1363 induced a higher frequency of CD11c+CD11b-CD103+ tolerogenic dendritic cells in lymphoid organs of mice at steady state. We conclude that all tested strains of L. lactis improved the clinical scores and parameters of colitis, which confirm their anti-inflammatory properties in this model of colitis.
Collapse
Affiliation(s)
- Juliana Lima Alves
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| | - Luisa Lemos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Nubia Morais Rodrigues
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Vanessa Bastos Pereira
- Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Ecologia E Evolução, Belo Horizonte, MG, Brazil
| | - Patrícia A Vieira Barros
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Maria Cecília Campos Canesso
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Mauro A F Guimarães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Denise Carmona Cara
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Anderson Miyoshi
- Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Ecologia E Evolução, Belo Horizonte, MG, Brazil
| | - Vasco Ariston Azevedo
- Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Ecologia E Evolução, Belo Horizonte, MG, Brazil
| | - Tatiani Uceli Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ana Cristina Gomes-Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
8
|
Ning L, Ye N, Ye B, Miao Z, Cao T, Lu W, Xu D, Tan C, Xu Y, Yan J. Qingre Xingyu recipe exerts inhibiting effects on ulcerative colitis development by inhibiting TNFα/NLRP3/Caspase-1/IL-1β pathway and macrophage M1 polarization. Cell Death Discov 2023; 9:84. [PMID: 36890151 PMCID: PMC9995513 DOI: 10.1038/s41420-023-01361-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 03/10/2023] Open
Abstract
As a chronic inflammatory bowel disease, ulcerative colitis (UC) imposes a significant burden on public healthcare worldwide due to its increasing morbidity. Chinese medicines are regarded as potent therapeutic agents for UC treatment with minimal side effects. In the present study, we sought to determine the novel role of a traditional medicine Qingre Xingyu (QRXY) recipe in the development of UC and aimed to contribute to the currently available knowledge about UC by exploring the downstream mechanism of QRXY recipe in UC. Mouse models of UC were established by injections with dextran sulphate sodium (DSS), where the expression of tumor necrosis factor-alpha (TNFα), NLR family pyrin domain containing 3 (NLRP3), and interleukin-1β (IL-1β) was determined followed by an analysis of their interactions. The DSS-treated NLRP3 knockout (-/-) Caco-2 cell model was successfully constructed. The in vitro and in vivo effects of the QRXY recipe on UC were investigated with the determination of disease activity index (DAI), histopathological scores, transepithelial electrical resistance, FITC-dextran, as well as cell proliferation and apoptosis. In vivo and in vitro experiments indicated that the QRXY recipe reduced the degree of intestinal mucosal injury of UC mice and functional damage of DSS-induced Caco-2 cells by inhibition of the TNFα/NLRP3/caspase-1/IL-1β pathway and M1 polarization of macrophages, and TNFα overexpression or NLRP3 knockdown could counterweigh the therapeutic effects of QRXY recipe. To conclude, our study elicited that QRXY inhibited the expression of TNFα and inactivated the NLRP3/Caspase-1/IL-1β pathway, thereby alleviating intestinal mucosal injury and relieving UC in mice.
Collapse
Affiliation(s)
- Liqin Ning
- Chinese Medicine Master Studio, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Ningyuan Ye
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Bai Ye
- Department of Gastroenterology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Zhiwei Miao
- Department of Gastroenterology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215600, P. R. China
| | - Tingting Cao
- Department of Gastroenterology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Weimin Lu
- Department of Internal Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Danhua Xu
- Chinese Medicine Master Studio, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Chang Tan
- Chinese Medicine Master Studio, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Yi Xu
- Department of Gastroenterology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China.
| | - Jing Yan
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China.
| |
Collapse
|
9
|
Goggans ML, Bilbrey EA, Quiroz-Moreno CD, Francis DM, Jacobi SK, Kovac J, Cooperstone JL. Short-Term Tomato Consumption Alters the Pig Gut Microbiome toward a More Favorable Profile. Microbiol Spectr 2022; 10:e0250622. [PMID: 36346230 PMCID: PMC9769997 DOI: 10.1128/spectrum.02506-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
Diets rich in fruits and vegetables have been shown to exert positive effects on the gut microbiome. However, little is known about the specific effect of individual fruits or vegetables on gut microbe profiles. This study aims to elucidate the effects of tomato consumption on the gut microbiome, as tomatoes account for 22% of vegetable consumption in Western diets, and their consumption has been associated with positive health outcomes. Using piglets as a physiologically relevant model of human metabolism, 20 animals were assigned to either a control or a tomato powder-supplemented diet (both macronutrient matched and isocaloric) for 14 days. The microbiome was sampled rectally at three time points: day 0 (baseline), day 7 (midpoint), and day 14 (end of study). DNA was sequenced using shotgun metagenomics, and reads were annotated using MG-RAST. There were no differences in body weight or feed intake between our two treatment groups. There was a microbial shift which included a higher ratio of Bacteroidota to Bacillota (formerly known as Bacteroidetes and Firmicutes, respectively) and higher alpha-diversity in tomato-fed animals, indicating a shift to a more desirable phenotype. Analyses at both the phylum and genus levels showed global microbiome profile changes (permutational multivariate analysis of variance [PERMANOVA], P ≤ 0.05) over time but not with tomato consumption. These data suggest that short-term tomato consumption can beneficially influence the gut microbial profile, warranting further investigation in humans. IMPORTANCE The composition of the microorganisms in the gut is a contributor to overall health, prompting the development of strategies to alter the microbiome composition. Studies have investigated the role of the diet on the microbiome, as it is a major modifiable risk factor contributing to health; however, little is known about the causal effects of consumption of specific foods on the gut microbiota. A more complete understanding of how individual foods impact the microbiome will enable more evidence-based dietary recommendations for long-term health. Tomatoes are of interest as the most consumed nonstarchy vegetable and a common source of nutrients and phytochemicals across the world. This study aimed to elucidate the effect of short-term tomato consumption on the microbiome, using piglets as a physiologically relevant model to humans. We found that tomato consumption can positively affect the gut microbial profile, which warrants further investigation in humans.
Collapse
Affiliation(s)
- Mallory L. Goggans
- Food Science and Technology, The Ohio State University, Columbus, Ohio, USA
| | - Emma A. Bilbrey
- Horticulture and Crop Science, The Ohio State University, Columbus, Ohio, USA
| | | | - David M. Francis
- Horticulture and Crop Science, The Ohio State University, Wooster, Ohio, USA
| | | | - Jasna Kovac
- Food Science, The Pennsylvania State University, University Park, Pennsylvania, USA
- Microbiome Center, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Jessica L. Cooperstone
- Food Science and Technology, The Ohio State University, Columbus, Ohio, USA
- Horticulture and Crop Science, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
10
|
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]
|
11
|
Zahirović A, Berlec A. Targeting IL-6 by engineered Lactococcus lactis via surface-displayed affibody. Microb Cell Fact 2022; 21:143. [PMID: 35842694 PMCID: PMC9287920 DOI: 10.1186/s12934-022-01873-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dysregulated production of interleukin (IL)-6 is implicated in the pathology of inflammatory bowel disease (IBD). Neutralization of IL-6 in the gut by safe probiotic bacteria may help alleviate intestinal inflammation. Here, we developed Lactococcus lactis with potent and selective IL-6 binding activity by displaying IL-6-specific affibody on its surface. RESULTS Anti-IL-6 affibody (designated as ZIL) was expressed in fusion with lactococcal secretion peptide Usp45 and anchoring protein AcmA. A high amount of ZIL fusion protein was detected on bacterial surface, and its functionality was validated by confocal microscopy and flow cytometry. Removal of IL-6 from the surrounding medium by the engineered L. lactis was evaluated using enzyme-linked immunosorbent assay. ZIL-displaying L. lactis sequestered recombinant human IL-6 from the solution in a concentration-dependent manner by up to 99% and showed no binding to other pro-inflammatory cytokines, thus proving to be highly specific for IL-6. The removal was equally efficient across different IL-6 concentrations (150-1200 pg/mL) that were found to be clinically relevant in IBD patients. The ability of engineered bacteria to capture IL-6 from cell culture supernatant was assessed using immunostimulated human monocytic cell lines (THP-1 and U-937) differentiated into macrophage-like cells. ZIL-displaying L. lactis reduced the content of IL-6 in the supernatants of both cell lines in a concentration-dependent manner by up to 94%. Dose response analysis showed that bacterial cell concentrations of 107 and 109 CFU/mL (colony forming units per mL) were required for half-maximal removal of recombinant and macrophage-derived IL-6, respectively. CONCLUSION The ability of ZIL-displaying L. lactis to bind pathological concentrations of IL-6 at common bacterial doses suggests physiological significance.
Collapse
Affiliation(s)
- Abida Zahirović
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia. .,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
12
|
Laroute V, Beaufrand C, Gomes P, Nouaille S, Tondereau V, Daveran-Mingot ML, Theodorou V, Eutamene H, Mercier-Bonin M, Cocaign-Bousquet M. Lactococcus lactis NCDO2118 exerts visceral antinociceptive properties in rat via GABA production in the gastro-intestinal tract. eLife 2022; 11:77100. [PMID: 35727704 PMCID: PMC9213000 DOI: 10.7554/elife.77100] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 06/01/2022] [Indexed: 12/01/2022] Open
Abstract
Gut disorders associated to irritable bowel syndrome (IBS) are combined with anxiety and depression. Evidence suggests that microbially produced neuroactive molecules, like γ-aminobutyric acid (GABA), can modulate the gut-brain axis. Two natural strains of Lactococcus lactis and one mutant were characterized in vitro for their GABA production and tested in vivo in rat by oral gavage for their antinociceptive properties. L. lactis NCDO2118 significantly reduced visceral hypersensitivity induced by stress due to its glutamate decarboxylase (GAD) activity. L. lactis NCDO2727 with similar genes for GABA metabolism but no detectable GAD activity had no in vivo effect, as well as the NCDO2118 ΔgadB mutant. The antinociceptive effect observed for the NCDO2118 strain was mediated by the production of GABA in the gastro-intestinal tract and blocked by GABAB receptor antagonist. Only minor changes in the faecal microbiota composition were observed after the L. lactis NCDO2118 treatment. These findings reveal the crucial role of the microbial GAD activity of L. lactis NCDO2118 to deliver GABA into the gastro-intestinal tract for exerting antinociceptive properties in vivo and open avenues for this GRAS (Generally Recognized As safe) bacterium in the management of visceral pain and anxious profile of IBS patients.
Collapse
Affiliation(s)
- Valérie Laroute
- Toulouse Biotechnology Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Catherine Beaufrand
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Pedro Gomes
- Toulouse Biotechnology Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.,Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Sébastien Nouaille
- Toulouse Biotechnology Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Valérie Tondereau
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | | | - Vassilia Theodorou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Hélène Eutamene
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Muriel Mercier-Bonin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Muriel Cocaign-Bousquet
- Toulouse Biotechnology Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| |
Collapse
|
13
|
Boodhoo N, Shojadoost B, Alizadeh M, Kulkarni RR, Sharif S. Ex Vivo Differential Responsiveness to Clostridium perfringens and Lactococcus lactis by Avian Small Intestine Macrophages and T Cells. Front Immunol 2022; 13:807343. [PMID: 35222386 PMCID: PMC8863843 DOI: 10.3389/fimmu.2022.807343] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Tissue resident immune system cells in the chicken intestine play a significant role in the protection against pathogens. However, very little is known about these cells. The current study was conducted to further characterize chicken intestinal immune system cells. Furthermore, this study aimed to assess the immune modulatory action of a highly virulent Clostridium perfringens, a commonly found chicken intestinal microbe, in comparison with a non-commensal, Lactococcus lactis, on intestine-derived immune system cells. The results demonstrated varying distribution of innate and adaptive immune cells along the avian gut-associated lymphoid tissue (GALT) in the duodenum, jejunum, ileum, and cecal tonsils. In addition, steady-state and tissue-specific presence of CD25+ cells among αβ and γδ T-cell subsets was assessed along the intestine. Ex vivo stimulation with C. perfringens or L. lactis resulted in a significant increase in the frequency of CD25+ T cells (γδ and αβ T cells). In addition, significantly more cell death was observed in ex vivo stimulation with C. perfringens, which was indirectly correlated with a decrease in macrophage activation based on nitric oxide (NO) production with no effect on lymphoid cell responsiveness as per intracellular interferon (IFN)-gamma (γ) staining. Ex vivo stimulation with L. lactis activated γδ T cells and αβ T cells, based on intracellular IFN-γ staining, while it had limited effect on macrophages. However, the ability of γδ and αβ T cells to produce IFN-γ and the ability of macrophages production of NO was rescued in the presence of L. lactis. These results demonstrate the potential application of L. lactis, as a probiotic, against virulent C. perfringens infection in chicken.
Collapse
Affiliation(s)
- Nitish Boodhoo
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Bahram Shojadoost
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Mohammadali Alizadeh
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Raveendra R Kulkarni
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
14
|
Zahirović A, Plavec TV, Berlec A. Dual Functionalized Lactococcus lactis Shows Tumor Antigen Targeting and Cytokine Binding in Vitro. Front Bioeng Biotechnol 2022; 10:822823. [PMID: 35155394 PMCID: PMC8826564 DOI: 10.3389/fbioe.2022.822823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/10/2022] [Indexed: 01/19/2023] Open
Abstract
Pro-inflammatory cytokines play an important role in the development and progression of colorectal cancer (CRC). Tumor-targeting bacteria that can capture pro-inflammatory cytokines in the tumor microenvironment and thus block their tumor-promoting effects might provide clinical benefits in inflammation-associated CRC. The aim of this study was to develop bacteria with dual functionality for selective delivery of cytokine-binding proteins to the tumor by targeting specific receptors on cancer cells. We engineered a model lactic acid bacterium, Lactococcus lactis, to co-display on its surface a protein ligand for tumor antigens (EpCAM-binding affitin; HER2-binding affibody) and a ligand for pro-inflammatory cytokines (IL-8-binding evasin; IL-6-binding affibody). Genes that encoded protein binders were cloned into a lactococcal dual promoter plasmid, and protein co-expression was confirmed by Western blotting. To assess the removal of IL-8 and IL-6 by the engineered bacteria, we established inflammatory cell models by stimulating cytokine secretion in human colon adenocarcinoma cells (Caco-2; HT-29) and monocyte-like cells (THP-1; U-937). The engineered L. lactis removed considerable amounts of IL-8 from the supernatant of Caco-2 and HT-29 cells, and depleted IL-6 from the supernatant of THP-1 and U-937 cells as determined by ELISA. The tumor targeting properties of the engineered bacteria were evaluated in human embryonic kidney epithelial cells HEK293 transfected to overexpress EpCAM or HER2 receptors. Fluorescence microscopy revealed that the engineered L. lactis specifically adhered to transfected HEK293 cells, where the EpCAM-targeting bacteria exhibited greater adhesion efficiency than the HER2-targeting bacteria. These results confirm the concept that L. lactis can be efficiently modified to display two proteins simultaneously on their surface: a tumor antigen binder and a cytokine binder. Both proteins remain biologically active and provide the bacteria with tumor antigen targeting and cytokine binding ability.
Collapse
Affiliation(s)
- Abida Zahirović
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Tina Vida Plavec
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Aleš Berlec,
| |
Collapse
|
15
|
Fu Y, Liu S, Li M, Ren F, Wang Y, Chang Z. IL-17RD/sef exacerbates experimental mouse colitis and inflammation-associated tumorigenesis by regulating the proportion of T cell subsets. FEBS Lett 2021; 596:427-436. [PMID: 34939667 DOI: 10.1002/1873-3468.14266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 11/09/2022]
Abstract
T helper cells, especially Th1 and Th17 cells, were reported to play a pivotal role in the pathogenesis of inflammatory bowel disease (IBD). However, the underlying factors regulating T cell functions in IBD progression remain to be fully elucidated. Here, we revealed that IL-17RD/Sef exacerbates DSS-induced colitis by regulating the balance of T cell subsets and their secretion of associated cytokines. We also observed that IL-17RD/Sef promotes colitis-associated tumorigenesis and negatively correlates with survival in both mouse and colorectal cancer patients. Our results suggested that IL-17RD/Sef functions as a regulator of T cell subsets to promote the inflammatory responses in the pathogenesis of IBD and colitis-associated colon cancer.
Collapse
Affiliation(s)
- Yanxia Fu
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, 100069, China.,State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Sihan Liu
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Mengdi Li
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Fangli Ren
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Yinyin Wang
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Zhijie Chang
- State Key Laboratory of Membrane Biology, School of Medicine, Institute of Precision Medicine, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
16
|
Barnabei L, Laplantine E, Mbongo W, Rieux-Laucat F, Weil R. NF-κB: At the Borders of Autoimmunity and Inflammation. Front Immunol 2021; 12:716469. [PMID: 34434197 PMCID: PMC8381650 DOI: 10.3389/fimmu.2021.716469] [Citation(s) in RCA: 348] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/22/2021] [Indexed: 12/18/2022] Open
Abstract
The transcription factor NF-κB regulates multiple aspects of innate and adaptive immune functions and serves as a pivotal mediator of inflammatory response. In the first part of this review, we discuss the NF-κB inducers, signaling pathways, and regulators involved in immune homeostasis as well as detail the importance of post-translational regulation by ubiquitination in NF-κB function. We also indicate the stages of central and peripheral tolerance where NF-κB plays a fundamental role. With respect to central tolerance, we detail how NF-κB regulates medullary thymic epithelial cell (mTEC) development, homeostasis, and function. Moreover, we elaborate on its role in the migration of double-positive (DP) thymocytes from the thymic cortex to the medulla. With respect to peripheral tolerance, we outline how NF-κB contributes to the inactivation and destruction of autoreactive T and B lymphocytes as well as the differentiation of CD4+-T cell subsets that are implicated in immune tolerance. In the latter half of the review, we describe the contribution of NF-κB to the pathogenesis of autoimmunity and autoinflammation. The recent discovery of mutations involving components of the pathway has both deepened our understanding of autoimmune disease and informed new therapeutic approaches to treat these illnesses.
Collapse
Affiliation(s)
- Laura Barnabei
- INSERM UMR 1163, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Emmanuel Laplantine
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| | - William Mbongo
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| | - Frédéric Rieux-Laucat
- INSERM UMR 1163, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Robert Weil
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| |
Collapse
|
17
|
The Anti-Tumor Effect of Lactococcus lactis Bacteria-Secreting Human Soluble TRAIL Can Be Enhanced by Metformin Both In Vitro and In Vivo in a Mouse Model of Human Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13123004. [PMID: 34203951 PMCID: PMC8232584 DOI: 10.3390/cancers13123004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/25/2021] [Accepted: 06/11/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Colorectal cancer (CRC) is a major cause of morbidity and mortality in Europe, and accounts for over 10% of all cancer-related deaths worldwide. These indicate an urgent need for novel therapeutic options in CRC. Here, we analysed if genetically modified non-pathogenic Lactococcus lactis bacteria can be used for local delivery of human recombinant Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) and induction of tumor cells death in vitro and in vivo in CRC mouse model. We showed that modified L. lactis bacteria were able to secrete biologically active human soluble TRAIL (L. lactis(hsTRAIL+)), which selectively eliminated human CRC cells in vitro, and was further strengthened by metformin (MetF). Our results from in vitro studies were confirmed in vivo using subcutaneous NOD-SCID mouse model of human CRC. The data showed a significant reduction of the tumor growth by intratumor injection of L. lactis(hsTRAIL+) bacteria producing hsTRAIL. This effect could be further enhanced by oral administration of MetF. Abstract Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) induces apoptosis of many cancer cells, including CRC cells, being non-harmful for normal ones. However, recombinant form of human TRAIL failed in clinical trial when administered intravenously. To assess the importance of TRAIL in CRC patients, new form of TRAIL delivery would be required. Here we used genetically modified, non-pathogenic Lactococcus lactis bacteria as a vehicle for local delivery of human soluble TRAIL (hsTRAIL) in CRC. Operating under the Nisin Controlled Gene Expression System (NICE), the modified bacteria (L. lactis(hsTRAIL+)) were able to induce cell death of HCT116 and SW480 human cancer cells and reduce the growth of HCT116-tumor spheres in vitro. This effect was cancer cell specific as the cells of normal colon epithelium (FHC cells) were not affected by hsTRAIL-producing bacteria. Metformin (MetF), 5-fluorouracil (5-FU) and irinotecan (CPT-11) enhanced the anti-tumor actions of hsTRAIL in vitro. In the NOD-SCID mouse model, treatment of subcutaneous HCT116-tumors with L. lactis(hsTRAIL+) bacteria given intratumorally, significantly reduced the tumor growth. This anti-tumor activity of hsTRAIL in vivo was further enhanced by oral administration of MetF. These findings indicate that L. lactis bacteria could be suitable for local delivery of biologically active human proteins. At the same time, we documented that anti-tumor activity of hsTRAIL in experimental therapy of CRC can be further enhanced by MetF given orally, opening a venue for alternative CRC-treatment strategies.
Collapse
|
18
|
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
|
19
|
Kim JM, Kim DH, Park HJ, Ma HW, Park IS, Son M, Ro SY, Hong S, Han HK, Lim SJ, Kim SW, Cheon JH. Nanocomposites-based targeted oral drug delivery systems with infliximab in a murine colitis model. J Nanobiotechnology 2020; 18:133. [PMID: 32933548 PMCID: PMC7493402 DOI: 10.1186/s12951-020-00693-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/09/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Infliximab (IFX), a TNF-α blocking chimeric monoclonal antibody, induces clinical response and mucosal healing in patients with inflammatory bowel disease (IBD). However, systemic administration of this agent causes unwanted side effects. Oral delivery of antibody therapeutics might be an effective treatment strategy for IBD compared to intravenous administration. RESULTS All three carriers had a high encapsulation efficiency, narrow size distribution, and minimal systemic exposure. There was a higher interaction between nanocomposite carriers and monocytes compared to lymphocytes in the PBMC of IBD patients. Orally administered nanocomposite carriers targeted to inflamed colitis minimized systemic exposure. All IFX delivery formulations with nanocomposite carriers had a significantly less colitis-induced body weight loss, colon shortening and histomorphological score, compared to the DSS-treated group. AC-IFX-L and EAC-IFX-L groups showed significantly higher improvement of the disease activity index, compared to the DSS-treated group. In addition, AC-IFX-L and EAC-IFX-L alleviated pro-inflammatory cytokine expressions (Tnfa, Il1b, and Il17). CONCLUSION We present orally administered antibody delivery systems which improved efficacy in murine colitis while reducing systemic exposure. These oral delivery systems suggest a promising therapeutic approach for treating IBD.
Collapse
Affiliation(s)
- Jung Min Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Da Hye Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Jeong Park
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
| | - Hyun Woo Ma
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - I Seul Park
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Mijeong Son
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - So Youn Ro
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
| | - Seokmann Hong
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, 05006, Republic of Korea
| | - Hyo Kyung Han
- College of Pharmacy, Dongguk University-Seoul, Dongguk‑ro‑32, Ilsan‑donggu, Goyang, South Korea
| | - Soo Jeong Lim
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
20
|
Komaki S, Haque A, Miyazaki H, Matsumoto T, Nakamura S. Unexpected effect of probiotics by Lactococcus lactis subsp. lactis against colitis induced by dextran sulfate sodium in mice. J Infect Chemother 2020; 26:549-553. [PMID: 32122783 DOI: 10.1016/j.jiac.2020.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/15/2020] [Accepted: 01/22/2020] [Indexed: 02/08/2023]
Abstract
Ulcerative colitis (UC) is a representative intestinal chronic inflammatory disease whose incidence is rapidly increasing worldwide. It was previously shown that some specific probiotics help to guard against UC. In this study, we analyzed the effect of Lactococcus lactis subsp. lactis JCM5805 (L. lactis), which has been put to practical use as a probiotic, on the pathogenesis of UC using a dextran sulfate sodium-induced colitis mouse model. Survival rate, length, and histopathological parameters of the colon were elucidated. Further, the concentrations of inflammatory cytokines in serum were measured. As a result, the oral administration of high-dose L. lactis showed significant decreases in survival rate and colon length. Histopathological analysis showed that a bleeding appearance was observed in the L. lactis group, and the histology scores in the L. lactis group were significantly higher than those in the normal saline group. Furthermore, the levels of interferon gamma, tumor necrosis factor alpha, and interleukin-6 were significantly elevated in the L. lactis group. These results support that high-dose administration of L. lactis deteriorates intestinal inflammation and suggest that the careful selection of probiotics strains and administration dose is important for improving colitis including UC.
Collapse
Affiliation(s)
- Shinichirou Komaki
- Department of Microbiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan.
| | - Anwarul Haque
- International University of Health and Welfare, School of Medicine, Kozunomori 4-3, Narita City, Chiba, 286-8686, Japan
| | - Haruko Miyazaki
- Department of Microbiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Tetsuya Matsumoto
- International University of Health and Welfare, School of Medicine, Kozunomori 4-3, Narita City, Chiba, 286-8686, Japan
| | - Shigeki Nakamura
- Department of Microbiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| |
Collapse
|
21
|
Zhu C, Song W, Tao Z, Liu H, Zhang S, Xu W, Li H. Analysis of microbial diversity and composition in small intestine during different development times in ducks. Poult Sci 2020; 99:1096-1106. [PMID: 32029146 PMCID: PMC7587750 DOI: 10.1016/j.psj.2019.12.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Indexed: 01/09/2023] Open
Abstract
We studied the microbial profiles of the duodenum, jejunum, and ileum during different developmental stages in the duck using high-throughput sequencing of the bacterial 16S rRNA gene. We also investigated the differences in the microbiota in the duodenum, jejunum, and ileum at different developmental times. A correlation analysis was performed between the most abundant bacterial genera and the development of the small intestine. An analysis of alpha diversity indicated different species richness and bacterial diversity in the different small intestinal segments and at different development times. A beta diversity analysis indicated differences in the bacterial community compositions across time. In a weighted UniFrac principal coordinates analysis, the samples clustered into two categories, 2 to 4 wk and 6 to 10 wk, in the duodenum, jejunum, and ileum. Our results show that the small intestine is predominantly populated by the phyla Firmicutes, Bacteroidetes, and Proteobacteria throughout the developmental stages of the duck. The duodenum, jejunum, and ileum shared most of the bacterial phyla and genera present, although they showed significant differences in their relative abundances in the intestinal segments and developmental stages. They shared different bacterial taxa during development times and among different segments when the intergroup differences were analyzed. The genera Bacillus, Corynebacterium 1, Lactococcus, Sphingomonas, and Haliangium correlated moderately positively with the increase in bodyweight and the lengths and weights of the duodenum, jejunum, and ileum, and these genera may be considered important markers when assessing the heath of the intestinal microbiota in ducks. This study provides a foundation upon which to extend our knowledge of the diversity and composition of the duck microbiota and a basis for further studies of the management of the small intestinal microbiota and improvements in the health and production of ducks.
Collapse
Affiliation(s)
- Chunhong Zhu
- Jiangsu Institute of Poultry Science, Jiangsu Yangzhou, 225125, China.
| | - Weitao Song
- Jiangsu Institute of Poultry Science, Jiangsu Yangzhou, 225125, China
| | - Zhiyun Tao
- Jiangsu Institute of Poultry Science, Jiangsu Yangzhou, 225125, China
| | - Hongxiang Liu
- Jiangsu Institute of Poultry Science, Jiangsu Yangzhou, 225125, China
| | - Shuangjie Zhang
- Jiangsu Institute of Poultry Science, Jiangsu Yangzhou, 225125, China
| | - Wenjuan Xu
- Jiangsu Institute of Poultry Science, Jiangsu Yangzhou, 225125, China
| | - Huifang Li
- Jiangsu Institute of Poultry Science, Jiangsu Yangzhou, 225125, China.
| |
Collapse
|
22
|
Wang C, Li W, Wang H, Ma Y, Zhao X, Zhang X, Yang H, Qian J, Li J. Saccharomyces boulardii alleviates ulcerative colitis carcinogenesis in mice by reducing TNF-α and IL-6 levels and functions and by rebalancing intestinal microbiota. BMC Microbiol 2019; 19:246. [PMID: 31694526 PMCID: PMC6836350 DOI: 10.1186/s12866-019-1610-8] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/01/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND AIMS To explore the inhibition mechanism of Saccharomyces boulardii (S. boulardii) on ulcerative colitis (UC) carcinogenesis. METHODS C57BL/6 mice were treated with azoxymethane and dextran sulfate sodium (AOM/DSS) to develop a UC carcinogenesis model. The treatment group was lavaged with S. boulardii (5 × 107 CFU/d) for 12 weeks. The mice were sacrificed and the tumor load in the treatment group was compared with that of a control group. The levels of TNF-α and IL-6 in colon tissue were measured by enzyme-linked immunosorbent assays. The influence of S. boulardii on TNF-α and IL-6 regulation was also investigated using different colon cell lines. Differences in intestinal microbiota in both stool and intestinal mucosa samples were assessed using 16S rDNA sequencing. RESULTS S. boulardii treatment reduced AOM/DSS-induced UC carcinogenesis in mice, as indicated by the reduced tumor load and reduced TNF-α and IL-6 levels in vivo, as well its effects on TNF-α and IL-6 activities in vitro. Significant changes in both fecal and mucosal microbiota were observed among the control, the AOM/DSS treated, and AOM/DSS plus S. boulardii treated groups. For fecal microbiota, the AOM/DSS treated group was lower in Lactobacillus, but higher in Oscillibacter and Lachnoclostridium than the control group. After intervention with S. boulardii, the percentage of Bacillus and Lactococcus increased, but Lachnoclostridium, Oscillibacter, Bacteroides, and Pseudomonas decreased. For the intestinal mucosal microbiota, the AOM/DSS treated group was lower in Bifidobacterium and Ruminococcaceae_UCG-014 and higher in Alloprevotella than the control group. After S. boulardii exposure, the percentage contributions of Lachnoclostridium and Lachnospiraceae_NK4A136 increased. CONCLUSIONS S. boulardii effectively reduced UC carcinogenesis in an AOM/DSS induced mice model. This positive result can likely be attributed to the reduction of TNF-α and IL-6 levels or the blockade of their function combined with alterations to the intestinal microbiota.
Collapse
Affiliation(s)
- Chunsaier Wang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenbin Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hongying Wang
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yiming Ma
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinhua Zhao
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xudong Zhang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New Lambton Heights, New South Wales, Australia
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jiaming Qian
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
23
|
Plavec TV, Štrukelj B, Berlec A. Screening for New Surface Anchoring Domains for Lactococcus lactis. Front Microbiol 2019; 10:1879. [PMID: 31456787 PMCID: PMC6700490 DOI: 10.3389/fmicb.2019.01879] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022] Open
Abstract
The display of recombinant proteins on bacterial surfaces is a developing research area with a wide range of potential biotechnological applications. The lactic acid bacterium Lactococcus lactis is an attractive host for such surface display, and a promising vector for in vivo delivery of bioactive proteins. Surface-displayed recombinant proteins are usually anchored to the bacterial cell wall through anchoring domains. Here, we investigated alternatives to the commonly applied lactococcal lysine motif (LysM)-containing surface anchoring domain, the C-terminus of AcmA (cAcmA). We screened 15 anchoring domains of lactococcal or phage origins that belong to the Pfam categories LPXTG, LysM, CW_1, Cpl-7, WxL, SH3, and ChW, which can provide non-covalent or covalent binding to the cell wall. LPXTG, LysM, the duplicated CW_1 and SH3 domains promoted significant surface display of two model proteins, B domain and DARPin I07, although the display achieved was lower than that for the reference anchoring domain, cAcmA. On the other hand, the ChW-containing anchoring domain of the lactococcal phage AM12 endolysin (cAM12) demonstrated surface display comparable to that of cAcmA. The anchoring ability of cAM12 was confirmed by enabling non-covalent heterologous anchoring of the B domain on wild-type bacteria, as well as anchoring of CXCL8-binding evasin-3, which provided potential therapeutic applicability; both were displayed to an extent comparable to that of cAcmA. We have thereby demonstrated the effective use of different protein anchoring domains in L. lactis, with ChW-containing cAM12 the most promising alternative to the established approaches for surface display on L. lactis.
Collapse
Affiliation(s)
- Tina Vida Plavec
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Borut Štrukelj
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
24
|
Xu C, Qiao L, Ma L, Yan S, Guo Y, Dou X, Zhang B, Roman A. Biosynthesis of Polysaccharides-Capped Selenium Nanoparticles Using Lactococcus lactis NZ9000 and Their Antioxidant and Anti-inflammatory Activities. Front Microbiol 2019; 10:1632. [PMID: 31402902 PMCID: PMC6676592 DOI: 10.3389/fmicb.2019.01632] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/02/2019] [Indexed: 11/18/2022] Open
Abstract
Lactococcus lactis (L. lactis) NZ9000, which has been genetically modified, is the most commonly used host strain for nisin regulated gene expression. Selenium (Se) is an essential trace element in the diet of humans and animals important for the maintenance of health and growth. Biosynthesized Se nanoparticles (SeNPs) that use microorganisms as a vehicle are uniquely advantages in terms of low costs, low toxicity and high bioavailability. This study was aimed at preparing novel functionalized SeNPs by L. lactis NZ9000 through eco-friendly and economic biotechnology methods. Moreover, its physicochemical characteristics, antioxidant and anti-inflammatory activities were investigated. L. lactis NZ9000 synthesized elemental red SeNPs when co-cultivated with sodium selenite under anaerobic conditions. Biosynthesized SeNPs by L. lactis NZ9000 were mainly capped with polysaccharides and significantly alleviated the increase of malondialdehyde (MDA) concentration, the decrease of glutathione peroxidase (GPx) and total superoxide dismutase (T-SOD) activity in porcine intestinal epithelial cells (IPEC-J2) challenged by hydrogen peroxide (H2O2). SeNPs also prevented the H2O2-caused reduction of transepithelial electrical resistance (TEER) and the increase of FITC-Dextran fluxes across IPEC-J2. Moreover, SeNPs attenuated the increase of reactive oxygen species (ROS), the reduction of adenosine triphosphate (ATP) and the mitochondrial membrane potential (MMP) and maintained intestinal epithelial permeability in IPEC-J2 cells exposed to H2O2. In addition, SeNPs pretreatment alleviated the cytotoxicity of Enterotoxigenic Escherichia coli (ETEC) K88 on IPEC-J2 cells and maintained the intestinal epithelial barrier integrity by up-regulating the expression of Occludin and Claudin-1 and modulating inflammatory cytokines. Biosynthesized SeNPs by L. lactis NZ9000 are a promising selenium supplement with antioxidant and anti-inflammatory activities.
Collapse
Affiliation(s)
- Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Lei Qiao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Li Ma
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Shuqi Yan
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yu Guo
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xina Dou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Baohua Zhang
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Alexandra Roman
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
25
|
Chiabai MJ, Almeida JF, de Azevedo MGD, Fernandes SS, Pereira VB, de Castro RJA, Jerônimo MS, Sousa IG, de Souza Vianna LM, Miyoshi A, Bocca AL, Maranhão AQ, Brigido MM. Mucosal delivery of Lactococcus lactis carrying an anti-TNF scFv expression vector ameliorates experimental colitis in mice. BMC Biotechnol 2019; 19:38. [PMID: 31238939 PMCID: PMC6593574 DOI: 10.1186/s12896-019-0518-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/25/2019] [Indexed: 02/06/2023] Open
Abstract
Background Anti-Tumor Necrosis Factor-alpha therapy has become clinically important for treating inflammatory bowel disease. However, the use of conventional immunotherapy requires a systemic exposure of patients and collateral side effects. Lactic acid bacteria have been shown to be effective as mucosal delivering system for cytokine and single domain antibodies, and it is amenable to clinical purposes. Therefore, lactic acid bacteria may function as vehicles for delivery of therapeutic antibodies molecules to the gastrointestinal tract restricting the pharmacological effect towards the gut. Here, we use the mucosal delivery of Lactococcus lactis carrying an anti-TNFα scFv expression plasmid on a DSS-induced colitis model in mice. Results Experimental colitis was induced with DSS administered in drinking water. L. lactis carrying the scFv expression vector was introduced by gavage. After four days of treatment, animals showed a significant improvement in histological score and disease activity index compared to those of untreated animals. Moreover, treated mice display IL-6, IL17A, IL1β, IL10 and FOXP3 mRNA levels similar to health control mice. Therefore, morphological and molecular markers suggest amelioration of the experimentally induced colitis. Conclusion These results provide evidence for the use of this alternative system for delivering therapeutic biopharmaceuticals in loco for treating inflammatory bowel disease, paving the way for a novel low-cost and site-specific biotechnological route for the treatment of inflammatory disorders. Electronic supplementary material The online version of this article (10.1186/s12896-019-0518-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maria José Chiabai
- Laboratório de Imunologia Molecular, Departamento de Biologia Molecular, Universidade de Brasília, Brasília, Distrito Federal, Brazil
| | - Juliana Franco Almeida
- Centro de Biotecnologia, Departamento de Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | | | - Suelen Soares Fernandes
- Laboratório de Imunologia Molecular, Departamento de Biologia Molecular, Universidade de Brasília, Brasília, Distrito Federal, Brazil
| | - Vanessa Bastos Pereira
- Laboratório de Tecnologia Genética, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Raffael Júnio Araújo de Castro
- Laboratório de Imunologia Aplicada, Departamento de Biologia Celular, Universidade de Brasília, Brasília, Distrito Federal, Brazil
| | - Márcio Sousa Jerônimo
- Laboratório de Imunologia Aplicada, Departamento de Biologia Celular, Universidade de Brasília, Brasília, Distrito Federal, Brazil
| | - Isabel Garcia Sousa
- Laboratório de Imunologia Molecular, Departamento de Biologia Molecular, Universidade de Brasília, Brasília, Distrito Federal, Brazil
| | | | - Anderson Miyoshi
- Laboratório de Tecnologia Genética, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anamelia Lorenzetti Bocca
- Laboratório de Imunologia Aplicada, Departamento de Biologia Celular, Universidade de Brasília, Brasília, Distrito Federal, Brazil
| | - Andrea Queiroz Maranhão
- Laboratório de Imunologia Molecular, Departamento de Biologia Molecular, Universidade de Brasília, Brasília, Distrito Federal, Brazil.,Instituto Nacional de Investigação em Imunologia, INCTii, Brasília, Distrito Federal, Brazil
| | - Marcelo Macedo Brigido
- Laboratório de Imunologia Molecular, Departamento de Biologia Molecular, Universidade de Brasília, Brasília, Distrito Federal, Brazil. .,Instituto Nacional de Investigação em Imunologia, INCTii, Brasília, Distrito Federal, Brazil.
| |
Collapse
|
26
|
Engineered Lactococcus lactis Secreting IL-23 Receptor-Targeted REX Protein Blockers for Modulation of IL-23/Th17-Mediated Inflammation. Microorganisms 2019; 7:microorganisms7050152. [PMID: 31137908 PMCID: PMC6560508 DOI: 10.3390/microorganisms7050152] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/09/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
Lactococcus lactis, a probiotic bacterium of food origin, has recently been demonstrated as a suitable strain for the production and in vivo delivery of therapeutically important proteins into the gut. We aimed to engineer recombinant L. lactis cells producing/secreting REX binding proteins that have been described as IL-23 receptor (IL-23R) blockers and IL-23R antagonists suppressing the secretion of cytokine IL-17A, a pivotal step in the T-helper Th17-mediated pro-inflammatory cascade, as well as in the development of autoimmune diseases, including inflammatory bowel disease (IBD). To reach this goal, we introduced cDNA sequences coding for REX009, REX115, and REX125 proteins into plasmid vectors carrying a Usp45 secretion signal, a FLAG tag sequence consensus, and a LysM-containing cA surface anchor (AcmA), thus allowing cell-surface peptidoglycan anchoring. These plasmids, or their non-FLAG/non-AcmA versions, were introduced into L. lactis host cells, thus generating unique recombinant L. lactis-REX strains. We demonstrate that all three REX proteins are expressed in L. lactis cells and are efficiently displayed on the bacterial surface, as tested by flow cytometry using an anti-FLAG antibody conjugate. Upon 10-fold concentration of the conditioned media, a REX125 secretory variant can be detected by Western blotting. To confirm that the FLAG/non-FLAG REX proteins displayed by L. lactis retain their binding specificity, cell-surface interactions of REX proteins with an IL-23R-IgG chimera were demonstrated by flow cytometry. In addition, statistically significant binding of secreted REX009 and REX115 proteins to bacterially produced, soluble human IL-23R was confirmed by ELISA. We conclude that REX-secreting L. lactis strains were engineered that might serve as IL-23/IL-23R blockers in an experimentally induced mouse model of colitis.
Collapse
|
27
|
He Z, Zhou Q, Wen K, Wu B, Sun X, Wang X, Chen Y. Huangkui Lianchang Decoction Ameliorates DSS-Induced Ulcerative Colitis in Mice by Inhibiting the NF-kappaB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:1040847. [PMID: 31093294 PMCID: PMC6481129 DOI: 10.1155/2019/1040847] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/20/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The nuclear factor kappa beta (NF-κB) signaling pathway plays an important role in ulcerative colitis (UC). Huangkui Lianchang decoction (HLD) is an effective traditional Chinese medicinal compound used in the treatment of UC. HLD has good effects in the clinic, but the mechanism by which HLD acts is unclear. This study aims to reveal the exact molecular mechanism of HLD in the treatment of UC. METHODS Mouse ulcerative colitis was induced by dextran sulfate sodium (DSS) and treated with HLD. Intestinal damage was assessed by disease activity index (DAI), colon macroscopic lesion scores, and histological scores. Interleukin (IL)-6, tumor necrosis factor (TNF)-α, and IL-1β were detected in colon tissue using ELISA. Myeloperoxidase (MPO) and superoxide dismutase (SOD) activities in the colonic mucosa were measured. The levels of IL-6, inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) in the colon were determined by real-time quantitative polymerase chain reaction (qPCR). The expression of NF-κB, IκBα, and p-IκBα in the colon was measured by Western blot. RESULTS After treatment with HLD, the DAI scores, macroscopic lesion scores, and histological scores decreased, and the levels of inflammatory cytokines related to the NF-κB signaling pathway, such as IL-6, TNF-α, and IL-1β, as well as those of iNOS and COX-2, were reduced; at the same time, colonic pathological damage was alleviated, and the MPO and SOD activities decreased. Western blot confirmed that HLD can inhibit the NF-κB signaling pathway in DSS-induced ulcerative colitis. CONCLUSION HLD can alleviate the inflammation caused by ulcerative colitis. In particular, high doses of HLD can significantly alleviate intestinal inflammation and have comparable efficacy to Mesalazine. We propose that the anti-inflammatory activity of HLD on DSS-induced colitis in mice may involve the inhibition of the NF-κB pathway.
Collapse
Affiliation(s)
- Zongqi He
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210046, China
- Department of Colorectal Surgery, Suzhou Hospital Affiliated with Nanjing University of Chinese Medicine, Suzhou 215009, China
| | - Qing Zhou
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Ke Wen
- Department of Colorectal Surgery, Suzhou Hospital Affiliated with Nanjing University of Chinese Medicine, Suzhou 215009, China
| | - Bensheng Wu
- Department of Colorectal Surgery, Suzhou Hospital Affiliated with Nanjing University of Chinese Medicine, Suzhou 215009, China
| | - Xueliang Sun
- Department of Colorectal Surgery, Suzhou Hospital Affiliated with Nanjing University of Chinese Medicine, Suzhou 215009, China
| | - Xiaopeng Wang
- Department of Colorectal Surgery, Suzhou Hospital Affiliated with Nanjing University of Chinese Medicine, Suzhou 215009, China
| | - Yugen Chen
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210046, China
| |
Collapse
|
28
|
Carvalho R, Vaz A, Pereira FL, Dorella F, Aguiar E, Chatel JM, Bermudez L, Langella P, Fernandes G, Figueiredo H, Goes-Neto A, Azevedo V. Gut microbiome modulation during treatment of mucositis with the dairy bacterium Lactococcus lactis and recombinant strain secreting human antimicrobial PAP. Sci Rep 2018; 8:15072. [PMID: 30305667 PMCID: PMC6180057 DOI: 10.1038/s41598-018-33469-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/20/2018] [Indexed: 12/19/2022] Open
Abstract
Mucositis is an inflammatory condition of the gut, caused by an adverse effect of chemotherapy drugs, such as 5-fluorouracil (5-FU). In an attempt to develop alternative treatments for the disease, several research groups have proposed the use of probiotics, in particular, Lactic Acid Bacteria (LAB). In this context, the use of recombinant LAB, for delivering anti-inflammatory compounds has also been explored. In previous work, we demonstrated that either Lactococcus lactis NZ9000 or a recombinant strain expressing an antimicrobial peptide involved in human gut homeostasis, the Pancreatitis-associated Protein (PAP), could ameliorate 5-FU-induced mucositis in mice. However, the impact of these strains on the gut microbiota still needs to be elucidated. Therefore, in the present study, we aimed to characterize the effects of both Lactococci strains in the gut microbiome of mice through a 16 S rRNA gene sequencing metagenomic approach. Our data show 5-FU caused a significant decrease in protective bacteria and increase of several bacteria associated with pro-inflammatory traits. The Lactococci strains were shown to reduce several potential opportunistic microbes, while PAP delivery was able to suppress the growth of Enterobacteriaceae during inflammation. We conclude the strain secreting antimicrobial PAP was more effective in the control of 5-FU-dysbiosis.
Collapse
Affiliation(s)
- Rodrigo Carvalho
- Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, MG, Brazil.
| | - Aline Vaz
- Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, MG, Brazil
| | | | - Fernanda Dorella
- Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, MG, Brazil
| | - Eric Aguiar
- Federal University of Bahia (UFBA), Salvador, BA, Brazil
| | - Jean-Marc Chatel
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Luis Bermudez
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Philippe Langella
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Gabriel Fernandes
- Fiocruz - Centro de Pesquisa Renê Rachou, Belo Horizonte, MG, Brazil
| | | | | | - Vasco Azevedo
- Federal University of Minas Gerais (UFMG-ICB), Belo Horizonte, MG, Brazil
| |
Collapse
|
29
|
Wang CSE, Li WB, Wang HY, Ma YM, Zhao XH, Yang H, Qian JM, Li JN. VSL#3 can prevent ulcerative colitis-associated carcinogenesis in mice. World J Gastroenterol 2018; 24:4254-4262. [PMID: 30310258 PMCID: PMC6175759 DOI: 10.3748/wjg.v24.i37.4254] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/07/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effects of VSL#3 on tumor formation, and fecal and intestinal mucosal microbiota in azoxymethane/dextran sulfate sodium (AOM/DSS) induced mice model.
METHODS C57BL/6 mice were administered AOM/DSS to develop the ulcerative colitis (UC) carcinogenesis model. Mice were treated with 5-ASA (75 mg/kg/d), VSL#3 (1.5 × 109 CFU/d), or 5-ASA combined with VSL#3 by gavage from the day of AOM injection for three months (five days/week). The tumor load was compared in each group, and tumor necrosis factor (TNF-α) and interleukin (IL)-6 levels were evaluated in colon tissue. The stool and intestinal mucosa samples were collected to analyze the differences in the intestinal microbiota by 16s rDNA sequencing method.
RESULTS VSL#3 significantly reduced the tumor load in AOM/DSS-induced mice model and decreased the level of TNF-α and IL-6 in colon tissue. The model group had a lower level of Lactobacillus and higher level of Oscillibacter and Lachnoclostridium in fecal microbiota than the control group. After the intervention with 5-ASA and VSL#3, Bacillus and Lactococcus were increased, while Lachnoclostridium and Oscillibacter were reduced. 5-ASA combined with VSL#3 increased the Lactobacillus and decreased the Oscillibacter. The intestinal mucosal microbiota analysis showed a lower level of Bifidobacterium and Ruminococcaceae_UCG-014 and higher level of Alloprevotella in the model group as compared to the control group. After supplementation with VSL#3, Bifidobacterium was increased. 5-ASA combined with VSL#3 increased the level of both Lachnoclostridium and Bifidobacterium.
CONCLUSION VSL#3 can prevent UC-associated carcinogenesis in mice, reduce the colonic mucosal inflammation levels, and rebalance the fecal and mucosal intestinal microbiota.
Collapse
Affiliation(s)
- Chun-Sai-Er Wang
- Department of Gastroenterology, PUMC Hospital, CAMS and PUMC, Beijing 100730, China
| | - Wen-Bin Li
- Department of Gastroenterology, PUMC Hospital, CAMS and PUMC, Beijing 100730, China
| | - Hong-Ying Wang
- National Cancer Center/Cancer Hospital, CAMS and PUMC, Beijing 100021, China
| | - Yi-Ming Ma
- National Cancer Center/Cancer Hospital, CAMS and PUMC, Beijing 100021, China
| | - Xin-Hua Zhao
- National Cancer Center/Cancer Hospital, CAMS and PUMC, Beijing 100021, China
| | - Hong Yang
- Department of Gastroenterology, PUMC Hospital, CAMS and PUMC, Beijing 100730, China
| | - Jia-Ming Qian
- Department of Gastroenterology, PUMC Hospital, CAMS and PUMC, Beijing 100730, China
| | - Jing-Nan Li
- Department of Gastroenterology, PUMC Hospital, CAMS and PUMC, Beijing 100730, China
| |
Collapse
|
30
|
p19-Targeting ILP Protein Blockers of IL-23/Th-17 Pro-Inflammatory Axis Displayed on Engineered Bacteria of Food Origin. Int J Mol Sci 2018; 19:ijms19071933. [PMID: 29966384 PMCID: PMC6073689 DOI: 10.3390/ijms19071933] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/23/2018] [Accepted: 06/29/2018] [Indexed: 12/31/2022] Open
Abstract
IL-23-mediated Th-17 cell activation and stimulation of IL-17-driven pro-inflammatory axis has been associated with autoimmunity disorders such as Inflammatory Bowel Disease (IBD) or Crohn’s Disease (CD). Recently we developed a unique class of IL-23-specific protein blockers, called ILP binding proteins that inhibit binding of IL-23 to its cognate cell-surface receptor (IL-23R) and exhibit immunosuppressive effect on human primary blood leukocytes ex vivo. In this study, we aimed to generate a recombinant Lactococcus lactis strain which could serve as in vivo producer/secretor of IL-23 protein blockers into the gut. To achieve this goal, we introduced ILP030, ILP317 and ILP323 cDNA sequences into expression plasmid vector containing USP45 secretion signal, FLAG sequence consensus and LysM-containing cA surface anchor (AcmA) ensuring cell-surface peptidoglycan anchoring. We demonstrate that all ILP variants are expressed in L. lactis cells, efficiently transported and secreted from the cell and displayed on the bacterial surface. The binding function of AcmA-immobilized ILP proteins is documented by interaction with a recombinant p19 protein, alpha subunit of human IL-23, which was assembled in the form of a fusion with Thioredoxin A. ILP317 variant exhibits the best binding to the human IL-23 cytokine, as demonstrated for particular L.lactis-ILP recombinant variants by Enzyme-Linked ImmunoSorbent Assay (ELISA). We conclude that novel recombinant ILP-secreting L. lactis strains were developed that might be useful for further in vivo studies of IL-23-mediated inflammation on animal model of experimentally-induced colitis.
Collapse
|
31
|
Škrlec K, Pucer Janež A, Rogelj B, Štrukelj B, Berlec A. Evasin-displaying lactic acid bacteria bind different chemokines and neutralize CXCL8 production in Caco-2 cells. Microb Biotechnol 2017; 10:1732-1743. [PMID: 28736998 PMCID: PMC5658612 DOI: 10.1111/1751-7915.12781] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/30/2017] [Accepted: 06/05/2017] [Indexed: 01/26/2023] Open
Abstract
Chemokines are key signals in the immune system and play an important role as proinflammatory mediators in the pathology of inflammatory bowel disease and colorectal cancer, making them an important target for therapy. Recombinant lactic acid bacteria (LAB) were engineered to bind CC and CXC chemokines by displaying chemokine‐binding proteins evasin‐1, evasin‐3 and evasin‐4 on their surface. Evasin genes were cloned into lactococcal surface display vector and overexpressed in L. lactis NZ9000 and NZ9000ΔhtrA in fusion with secretion signal and surface anchor. Evasin‐displaying bacteria removed from 15% to 90% of 11 different chemokines from the solution as determined with ELISA and Luminex multiplexing assays, whereby L. lactis NZ9000ΔhtrA proved more efficient. Lactobacillus salivarius ATCC 11741 was coated with L. . lactis‐expressed evasin fusion protein, and its ability to bind chemokines was also confirmed. Evasin‐3‐displaying L. lactis removed 76.0% of IL‐1β‐induced CXCL8 from the supernatant of Caco‐2 epithelial cells. It also prevented secretion of CXCL8 from Caco‐2 cells in a time‐dependent manner when added before induction with IL‐1β. Evasin‐displaying LAB have the ability to bind multiple chemokines simultaneously and exert synergistic activity. This innovative treatment approach therefore has the potential for mucosal therapy of inflammatory bowel disease or colorectal cancer.
Collapse
Affiliation(s)
- Katja Škrlec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia.,Graduate School of Biomedicine, Faculty of Medicine, University of Ljubljana, SI-1000, Ljubljana, Slovenia
| | - Anja Pucer Janež
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Rogelj
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia.,Biomedical Research Institute (BRIS), Puhova 10, SI-1000, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000, Ljubljana, Slovenia
| | - Borut Štrukelj
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia
| |
Collapse
|
32
|
Ståhl S, Gräslund T, Eriksson Karlström A, Frejd FY, Nygren PÅ, Löfblom J. Affibody Molecules in Biotechnological and Medical Applications. Trends Biotechnol 2017; 35:691-712. [PMID: 28514998 DOI: 10.1016/j.tibtech.2017.04.007] [Citation(s) in RCA: 273] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 01/08/2023]
Abstract
Affibody molecules are small (6.5-kDa) affinity proteins based on a three-helix bundle domain framework. Since their introduction 20 years ago as an alternative to antibodies for biotechnological applications, the first therapeutic affibody molecules have now entered clinical development and more than 400 studies have been published in which affibody molecules have been developed and used in a variety of contexts. In this review, we focus primarily on efforts over the past 5 years to explore the potential of affibody molecules for medical applications in oncology, neurodegenerative, and inflammation disorders, including molecular imaging, receptor signal blocking, and delivery of toxic payloads. In addition, we describe recent examples of biotechnological applications, in which affibody molecules have been exploited as modular affinity fusion partners.
Collapse
Affiliation(s)
- Stefan Ståhl
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden.
| | - Torbjörn Gräslund
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | | | - Fredrik Y Frejd
- Unit of Biomedical Radiation Sciences, Uppsala University, SE-751 85 Uppsala, Sweden; Affibody AB, Gunnar Asplunds Allé 24, SE-171 69 Solna, Sweden
| | - Per-Åke Nygren
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - John Löfblom
- Division of Protein Technology, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| |
Collapse
|