1
|
Qin P, Li Y, Su Y, Wang Z, Wu R, Liang X, Zeng Y, Guo P, Yu Z, Huang X, Yang H, Zeng Z, Zhao X, Gong S, Han J, Chen Z, Xiao W, Chen A. Bifidobacterium adolescentis-derived hypaphorine alleviates acetaminophen hepatotoxicity by promoting hepatic Cry1 expression. J Transl Med 2024; 22:525. [PMID: 38822329 PMCID: PMC11143572 DOI: 10.1186/s12967-024-05312-6] [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: 01/09/2024] [Accepted: 05/16/2024] [Indexed: 06/02/2024] Open
Abstract
Acetaminophen (APAP)-induced liver injury (AILI) is a pressing public health concern. Although evidence suggests that Bifidobacterium adolescentis (B. adolescentis) can be used to treat liver disease, it is unclear if it can prevent AILI. In this report, we prove that B. adolescentis significantly attenuated AILI in mice, as demonstrated through biochemical analysis, histopathology, and enzyme-linked immunosorbent assays. Based on untargeted metabolomics and in vitro cultures, we found that B. adolescentis generates microbial metabolite hypaphorine. Functionally, hypaphorine inhibits the inflammatory response and hepatic oxidative stress to alleviate AILI in mice. Transcriptomic analysis indicates that Cry1 expression is increased in APAP-treated mice after hypaphorine treatment. Overexpression of Cry1 by its stabilizer KL001 effectively mitigates liver damage arising from oxidative stress in APAP-treated mice. Using the gene expression omnibus (GEO) database, we verified that Cry1 gene expression was also decreased in patients with APAP-induced acute liver failure. In conclusion, this study demonstrates that B. adolescentis inhibits APAP-induced liver injury by generating hypaphorine, which subsequently upregulates Cry1 to decrease inflammation and oxidative stress.
Collapse
Affiliation(s)
- Ping Qin
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yanru Li
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- School of Nursing, Southern Medical University, Guangzhou, 510515, China
| | - Yangjing Su
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ze Wang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510665, China
| | - Rong Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoqi Liang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yunong Zeng
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Peiheng Guo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhichao Yu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xintao Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Hong Yang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510665, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Shenhai Gong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jiaochan Han
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Wei Xiao
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Ali Chen
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Bao H, Wang Y, Xiong H, Xia Y, Cui Z, Liu L. Mechanism of Iron Ion Homeostasis in Intestinal Immunity and Gut Microbiota Remodeling. Int J Mol Sci 2024; 25:727. [PMID: 38255801 PMCID: PMC10815743 DOI: 10.3390/ijms25020727] [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: 12/05/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Iron is a vital trace element that plays an important role in humans and other organisms. It plays an active role in the growth, development, and reproduction of bacteria, such as Bifidobacteria. Iron deficiency or excess can negatively affect bacterial hosts. Studies have reported a major role of iron in the human intestine, which is necessary for maintaining body homeostasis and intestinal barrier function. Organisms can maintain their normal activities and regulate some cancer cells in the body by regulating iron excretion and iron-dependent ferroptosis. In addition, iron can modify the interaction between hosts and microorganisms by altering their growth and virulence or by affecting the immune system of the host. Lactic acid bacteria such as Lactobacillus acidophilus (L. acidophilus), Lactobacillus rhamnosus (L. rhamnosus), and Lactobacillus casei (L. casei) were reported to increase trace elements, protect the host intestinal barrier, mitigate intestinal inflammation, and regulate immune function. This review article focuses on the two aspects of the iron and gut and generally summarizes the mechanistic role of iron ions in intestinal immunity and the remodeling of gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | - Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (H.B.); (Y.W.); (H.X.); (Y.X.)
| | - Lingbin Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (H.B.); (Y.W.); (H.X.); (Y.X.)
| |
Collapse
|
3
|
Shayya NW, Bandick R, Busmann LV, Mousavi S, Bereswill S, Heimesaat MM. Metabolomic signatures of intestinal colonization resistance against Campylobacter jejuni in mice. Front Microbiol 2023; 14:1331114. [PMID: 38164399 PMCID: PMC10757985 DOI: 10.3389/fmicb.2023.1331114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Campylobacter jejuni stands out as one of the leading causes of bacterial enteritis. In contrast to humans, specific pathogen-free (SPF) laboratory mice display strict intestinal colonization resistance (CR) against C. jejuni, orchestrated by the specific murine intestinal microbiota, as shown by fecal microbiota transplantation (FMT) earlier. Methods Murine infection models, comprising SPF, SAB, hma, and mma mice were employed. FMT and microbiota depletion were confirmed by culture and culture-independent analyses. Targeted metabolome analyses of fecal samples provided insights into the associated metabolomic signatures. Results In comparison to hma mice, the murine intestinal microbiota of mma and SPF mice (with CR against C. jejuni) contained significantly elevated numbers of lactobacilli, and Mouse Intestinal Bacteroides, whereas numbers of enterobacteria, enterococci, and Clostridium coccoides group were reduced. Targeted metabolome analysis revealed that fecal samples from mice with CR contained increased levels of secondary bile acids and fatty acids with known antimicrobial activities, but reduced concentrations of amino acids essential for C. jejuni growth as compared to control animals without CR. Discussion The findings highlight the role of microbiota-mediated nutrient competition and antibacterial activities of intestinal metabolites in driving murine CR against C. jejuni. The study underscores the complex dynamics of host-microbiota-pathogen interactions and sets the stage for further investigations into the mechanisms driving CR against enteric infections.
Collapse
|
4
|
Viswanath K, Hayes M, Avni D. Inflammatory bowel disease - A peek into the bacterial community shift and algae-based ‘biotic’ approach to combat the disease. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2022.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
5
|
Rodríguez-Sorrento A, Castillejos L, López-Colom P, Cifuentes-Orjuela G, Moreno-Muñoz JA, Martín-Orúe SM. Assessment of the Effects of the Synbiotic Combination of Bifidobacterium longum subsp. infantis CECT 7210 and Oligofructose-Enriched Inulin Against Digestive Bacterial Infections in a Piglet Model. Front Microbiol 2022; 13:831737. [PMID: 35350617 PMCID: PMC8957890 DOI: 10.3389/fmicb.2022.831737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/11/2022] [Indexed: 11/24/2022] Open
Abstract
The use of bifidobacteria as probiotics has proven to be beneficial in gastroenteric infections. Furthermore, prebiotics such as inulin can enhance the survival and growth of these bacteria. Two trials were performed to evaluate the effects of the administration of Bifidobacterium longum subsp. infantis CECT 7210 and oligofructose-enriched inulin against Salmonella enterica serovar Typhimurium or enterotoxigenic Escherichia coli (ETEC) F4. A total of 72 (Salmonella trial) and 96 (ETEC F4 trial) weaned piglets were used in a 2 × 2 design (with or without synbiotic, inoculated or not with the pathogen). After adaptation, animals were orally inoculated. Performance and clinical signs were evaluated. On days 4 and 8 (Salmonella trial) and 3 and 7 (ETEC F4 trial) post-inoculation (PI), one animal per pen was euthanized. Blood, digestive content and tissue samples were collected and microbiological counts, fermentation products, serum inflammatory markers and ileum histomorphometry analysis were performed. Both challenges had an impact on faecal consistency (p < 0.001), including the faecal shedding of Salmonella and increased numbers of enterobacteria and coliforms. The synbiotic administration did not have any effect on pathogen loads but induced changes in the fermentation profile, such as increased valeric acid in both trials as well as decreased acetic acid, except for Salmonella-challenged animals. The effect on propionate varied among trials, increasing in challenged synbiotic-treated pigs and decreasing in non-challenged ones in the Salmonella trial (P interaction = 0.013), while the opposed occurred in the ETEC F4 trial (P interaction = 0.013). The administration of the synbiotic increased intraepithelial lymphocytes (IEL; p = 0.039) on day 8 PI in the Salmonella trial and a similar trend occurred in non-challenged pigs in the ETEC F4 trial (P interaction = 0.086). The results did not provide evidence of reduced pathogen load with the synbiotic, although a modulation in fermentative activity could be identified depending on the challenge. Consistent increases were found in IEL, suggesting that this synbiotic combination has some immunomodulatory properties.
Collapse
Affiliation(s)
- Agustina Rodríguez-Sorrento
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i Dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lorena Castillejos
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i Dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Paola López-Colom
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i Dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Departamento de Producción Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Agraria del Ecuador (UAE), Guayaquil, Ecuador
| | | | | | - Susana M Martín-Orúe
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i Dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
6
|
Ruiz MJ, Sirini NE, Signorini ML, Etcheverría A, Zbrun MV, Soto LP, Zimmermann JA, Frizzo LS. Protective effect of Lactiplantibacillus plantarum LP5 in a murine model of colonisation by Campylobacter coli DSPV458. Benef Microbes 2021; 12:553-565. [PMID: 34590533 DOI: 10.3920/bm2021.0010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Thermotolerant Campylobacter species are the leading cause of foodborne bacterial diarrheal disease worldwide. Campylobacter coli, abundant in pigs and pork products, have been identified as a source of human infection. In this study, we propose the use of Lactiplantibacillus plantarum LP5 as a probiotic to reduce colonisation of this intestinal pathogen in a murine colonisation model of C. coli DSPV458. Six-week-old adult female Balb/cCmedc mice were housed in groups: Control, Campy and Pro-Campy. Control and Pro-Campy groups received antibiotics for 5 days and the Campy group for 12 days. Pro-Campy group was inoculated for 7 days with 8.78 log10 cfu total of L. plantarum LP5 suspended in De Man, Rogosa and Sharpe broth. All groups were inoculated with 6.72 log10 cfu of C. coli DSPV458 suspended in brain heart infusion broth. L. plantarum LP5 was recovered only in the Pro- Campy group. C. coli DSPV458 was recovered at higher levels in the Control and Campy groups. The differences with the Pro-Campy group were significant. As regards faeces, Control and Campy groups reached 7.41 and 7.84 log10 cfu/g, respectively, and the Pro-Campy group only 4.62 log10 cfu/g. In the caecum, Control and Campy groups reached 8.01 and 9.26 log10cfu/g, respectively, and the Pro-Campy group only 4.51 log10 cfu/g. In the ileum, Control and Campy groups reached 3.43 and 3.26 log10 cfu/g, respectively, and the Pro-Campy group did not show detectable levels. The reduction of C. coli DSPV458 in the Pro-Campy group compared to the Control group in faeces, caecum and ileum was 99.55, 99.98 and 100%, respectively. Animals were maintained under normal health conditions, and haematological parameters were within the standard values for Balb/cCmedc. The incorporation of a probiotic generated a protective effect in the mice colonisation model. The protective effect would also apply to intestinal colonisation by indigenous enterobacteria. Therefore, the strategy used in this study is of great importance to understand the protection mechanisms in a murine model, as well as its application in food-producing animals.
Collapse
Affiliation(s)
- M J Ruiz
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,Department of Animal Health and Preventive Medicine, Faculty of Veterinary Sciences, National University of the Center of the Province of Buenos Aires, Tandil, Argentina
| | - N E Sirini
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| | - M L Signorini
- Department of Public Health, Faculty of Veterinary Science, Litoral National University, Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,National Council of Scientific and Technical Research, National Institute of Agricultural Technology EEA Rafaela, Ruta 34 Km 227, 2300 Rafaela, Province of Santa Fe, Argentina
| | - A Etcheverría
- Department of Animal Health and Preventive Medicine, Faculty of Veterinary Sciences, National University of the Center of the Province of Buenos Aires, Tandil, Argentina
| | - M V Zbrun
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| | - L P Soto
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| | - J A Zimmermann
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| | - L S Frizzo
- Laboratory of Food Analysis 'Rodolfo Oscar Dalla Santina', Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral - National Council of Scientific and Technical Research (UNL/CONICET), Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Kreder 2805, 3080 Esperanza, Province of Santa Fe, Argentina
| |
Collapse
|
7
|
Ruiz MJ, Soto LP, Sirini NE, Werning ML, Olivero CR, Zimmermann JA, Zbrun MV, Acosta FF, Signorini ML, Frizzo LS. Murine colonization model by Campylobacter coli DSPV458. J Appl Microbiol 2021; 132:1457-1466. [PMID: 34465011 DOI: 10.1111/jam.15272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/11/2021] [Accepted: 08/24/2021] [Indexed: 11/29/2022]
Abstract
AIMS To generate a murine experimental model of colonization by Campylobacter coli DSPV458. METHODS AND RESULTS Twelve adult Balb/cCmedc female mice were housed in a treated group (T-G) and a control group (C-G) for 4 weeks. Both experimental groups received antibiotics for 5 days during the first week. The T-G was administered with 6.68log10 CFU of C. coli DSPV458 by oesophageal gavage. Necropsies were performed weekly to evaluate translocation and intestinal colonization in the spleen and liver and in the ileum and cecum respectively. Samples were cultured to quantify intestinal microbiota members. Faeces were cultured weekly for a C. coli DSPV458 count. Campylobacter coli DSPV458 was isolated from all the inoculated mice. The recovered level of C. coli DSPV458 was, on average, 6.9 log10 CFUg-1 , 8.0 log10 CFUg-1 and 1.6 log10 CFUg-1 in faeces, cecum and ileum respectively. Colonization by C. coli DSPV458 does not alter the normal clinical and physiological status. CONCLUSIONS Campylobacter coli DSPV458 does not have an invasive capacity, and the model is suitable for evaluating strategies to reduce intestinal loads. SIGNIFICANCE AND IMPACT OF STUDY Farm animals have an important impact on thermotolerant Campylobacter transmission to humans. Extremely few colonization models by C. coli have been reported to date. In food-producing animals, infection is mild or absent and thermotolerant Campylobacter colonize the intestines of animals. Colonization models are specific models that do not cause infection as they do not generally result in diarrhoea or other signs of disease. Therefore, this model will allow to evaluate the evolution of colonization by thermotolerant Campylobacter and the alternative tools development to antibiotics that limit their colonization in food-producing animals.
Collapse
Affiliation(s)
- M J Ruiz
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Animal Health and Preventive Medicine, Faculty of Veterinary Sciences, National University of the Center of the Province of Buenos Aires, Argentina
| | - L P Soto
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina
| | - N E Sirini
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina
| | - M L Werning
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina
| | - C R Olivero
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina
| | - J A Zimmermann
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina
| | - M V Zbrun
- Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina.,National Council of Scientific and Technical Research, National Institute of Agricultural Technology EEA Rafaela, Rafaela, Province of Santa Fe, Argentina
| | - F F Acosta
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina
| | - M L Signorini
- Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina.,National Council of Scientific and Technical Research, National Institute of Agricultural Technology EEA Rafaela, Rafaela, Province of Santa Fe, Argentina
| | - L S Frizzo
- Laboratory of Food Analysis "Rodolfo Oscar DALLA SANTINA", Institute of Veterinary Science (ICiVet Litoral), National University of the Litoral-National Council of Scientific and Technical Research (UNL/CONICET), Esperanza, Province of Santa Fe, Argentina.,Department of Public Health, Faculty of Veterinary Science, Litoral National University, Esperanza, Province of Santa Fe, Argentina
| |
Collapse
|
8
|
Jin X, He Y, Zhou Y, Chen X, Lee YK, Zhao J, Zhang H, Chen W, Wang G. Lactic acid bacteria that activate immune gene expression in Caenorhabditis elegans can antagonise Campylobacter jejuni infection in nematodes, chickens and mice. BMC Microbiol 2021; 21:169. [PMID: 34090326 PMCID: PMC8180125 DOI: 10.1186/s12866-021-02226-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 05/13/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Campylobacter jejuni is the major micro-bacillary pathogen responsible for human coloenteritis. Lactic acid bacteria (LAB) have been shown to protect against Campylobacter infection. However, LAB with a good ability to inhibit the growth of C. jejuni in vitro are less effective in animals and animal models, and have the disadvantages of high cost, a long cycle, cumbersome operation and insignificant immune response indicators. Caenorhabditis elegans is increasingly used to screen probiotics for their anti-pathogenic properties. However, no research on the use of C. elegans to screen for probiotic candidates antagonistic to C. jejuni has been conducted to date. RESULTS This study established a lifespan model of C. elegans, enabling the preselection of LAB to counter C. jejuni infection. A potential protective mechanism of LAB was identified. Some distinct LAB species offered a high level of protection to C. elegans against C. jejuni. The LAB strains with a high protection rate reduced the load of C. jejuni in C. elegans. The transcription of antibacterial peptide genes, MAPK and Daf-16 signalling pathway-related genes was elevated using the LAB isolates with a high protection rate. The reliability of the lifespan model of C. elegans was verified using mice and chickens infected with C. jejuni. CONCLUSIONS The results showed that different LAB had different abilities to protect C. elegans against C. jejuni. C. elegans provides a reliable model for researchers to screen for LAB that are antagonistic to C. jejuni on a large scale.
Collapse
Affiliation(s)
- Xing Jin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Yufeng He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Yonghua Zhou
- Key Laboratory of National Health and Family Planning Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, P. R. China
| | - Xiaohua Chen
- College of Life Sciences and Environment, Hengyang Normal University, Hengyang, 421008, P. R. China
| | - Yuan-Kun Lee
- Department of Microbiology & Immunology, National University of Singapore, Singapore, 117597, Singapore
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, 214122, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, P. R. China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, 214122, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, P. R. China
- Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, 100048, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China.
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, 214122, P. R. China.
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, P. R. China.
| |
Collapse
|
9
|
Iqbal Z, Ahmed S, Tabassum N, Bhattacharya R, Bose D. Role of probiotics in prevention and treatment of enteric infections: a comprehensive review. 3 Biotech 2021; 11:242. [PMID: 33968585 PMCID: PMC8079594 DOI: 10.1007/s13205-021-02796-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/15/2021] [Indexed: 12/16/2022] Open
Abstract
Microorganisms that inhabits human digestive tract affect global health and enteric disorders. Previous studies have documented the effectiveness and mode of action of probiotics and classified as human-friendly biota and a competitor to enteric pathogens. Statistical studies reported more than 1.5 billion cases of gastrointestinal infections caused by enteric pathogens and their long-term exposure can lead to mental retardation, temporary or permanent physical weakness, and leaving the patient susceptible for opportunistic pathogens, which can cause fatality. We reviewed previous literature providing evidence about therapeutic approaches regarding probiotics to cure enteric infections efficiently by producing inhibitory substances, immune system modulation, improved barrier function. The therapeutic effects of probiotics have shown success against many foodborne pathogens and their therapeutic effectiveness has been exponentially increased using genetically engineered probiotics. The bioengineered probiotic strains are expected to provide a better and alternative approach than traditional antibiotic therapy against enteric pathogens, but the novelty of these strains also raise doubts about the possible untapped side effects, for which there is a need for further studies to eliminate the concerns relating to the use and safety of probiotics. Many such developments and optimization of the classical techniques will revolutionize the treatments for enteric infections.
Collapse
Affiliation(s)
- Zunaira Iqbal
- Department of Microbiology, University of Central Punjab, Johar Town, 1-Khayaban-e-Jinnah Road, Lahore, Pakistan
| | - Shahzaib Ahmed
- Department of Biotechnology, University of Central Punjab, Johar Town, 1-Khayaban-e-Jinnah Road, Lahore, Pakistan
| | - Natasha Tabassum
- Department of Biotechnology, University of Central Punjab, Johar Town, 1-Khayaban-e-Jinnah Road, Lahore, Pakistan
| | - Riya Bhattacharya
- Faculty of Applied Sciences and Biotechnology, School of Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh India
| | - Debajyoti Bose
- Faculty of Applied Sciences and Biotechnology, School of Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh India
| |
Collapse
|
10
|
Vendrik KEW, Ooijevaar RE, de Jong PRC, Laman JD, van Oosten BW, van Hilten JJ, Ducarmon QR, Keller JJ, Kuijper EJ, Contarino MF. Fecal Microbiota Transplantation in Neurological Disorders. Front Cell Infect Microbiol 2020; 10:98. [PMID: 32266160 PMCID: PMC7105733 DOI: 10.3389/fcimb.2020.00098] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Several studies suggested an important role of the gut microbiota in the pathophysiology of neurological disorders, implying that alteration of the gut microbiota might serve as a treatment strategy. Fecal microbiota transplantation (FMT) is currently the most effective gut microbiota intervention and an accepted treatment for recurrent Clostridioides difficile infections. To evaluate indications of FMT for patients with neurological disorders, we summarized the available literature on FMT. In addition, we provide suggestions for future directions. Methods: In July 2019, five main databases were searched for studies and case descriptions on FMT in neurological disorders in humans or animal models. In addition, the ClinicalTrials.gov website was consulted for registered planned and ongoing trials. Results: Of 541 identified studies, 34 were included in the analysis. Clinical trials with FMT have been performed in patients with autism spectrum disorder and showed beneficial effects on neurological symptoms. For multiple sclerosis and Parkinson's disease, several animal studies suggested a positive effect of FMT, supported by some human case reports. For epilepsy, Tourette syndrome, and diabetic neuropathy some studies suggested a beneficial effect of FMT, but evidence was restricted to case reports and limited numbers of animal studies. For stroke, Alzheimer's disease and Guillain-Barré syndrome only studies with animal models were identified. These studies suggested a potential beneficial effect of healthy donor FMT. In contrast, one study with an animal model for stroke showed increased mortality after FMT. For Guillain-Barré only one study was identified. Whether positive findings from animal studies can be confirmed in the treatment of human diseases awaits to be seen. Several trials with FMT as treatment for the above mentioned neurological disorders are planned or ongoing, as well as for amyotrophic lateral sclerosis. Conclusions: Preliminary literature suggests that FMT may be a promising treatment option for several neurological disorders. However, available evidence is still scanty and some contrasting results were observed. A limited number of studies in humans have been performed or are ongoing, while for some disorders only animal experiments have been conducted. Large double-blinded randomized controlled trials are needed to further elucidate the effect of FMT in neurological disorders.
Collapse
Affiliation(s)
- Karuna E W Vendrik
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands.,Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Centre for Infectious Disease Control, National Institute for Public Health and the Environment (Rijksinstituut voor Volksgezondheid en Milieu, RIVM), Bilthoven, Netherlands
| | - Rogier E Ooijevaar
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Department of Gastroenterology, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, Netherlands
| | - Pieter R C de Jong
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - Jon D Laman
- Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, Groningen, Netherlands
| | - Bob W van Oosten
- Department of Neurology, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, Netherlands
| | | | - Quinten R Ducarmon
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands.,Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
| | - Josbert J Keller
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, Netherlands.,Department of Gastroenterology, Haaglanden Medical Center, The Hague, Netherlands
| | - Eduard J Kuijper
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands.,Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, Netherlands.,Centre for Infectious Disease Control, National Institute for Public Health and the Environment (Rijksinstituut voor Volksgezondheid en Milieu, RIVM), Bilthoven, Netherlands.,Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Fiorella Contarino
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands.,Department of Neurology, Haga Teaching Hospital, The Hague, Netherlands
| |
Collapse
|
11
|
Bereswill S, Ekmekciu I, Escher U, Fiebiger U, Stingl K, Heimesaat MM. Lactobacillus johnsonii ameliorates intestinal, extra-intestinal and systemic pro-inflammatory immune responses following murine Campylobacter jejuni infection. Sci Rep 2017; 7:2138. [PMID: 28522817 PMCID: PMC5437126 DOI: 10.1038/s41598-017-02436-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/11/2017] [Indexed: 12/23/2022] Open
Abstract
Campylobacter jejuni infections are progressively increasing worldwide. Probiotic treatment might open novel therapeutic or even prophylactic approaches to combat campylobacteriosis. In the present study secondary abiotic mice were generated by broad-spectrum antibiotic treatment and perorally reassociated with a commensal murine Lactobacillus johnsonii strain either 14 days before (i.e. prophylactic regimen) or 7 days after (i.e. therapeutic regimen) peroral C. jejuni strain 81-176 infection. Following peroral reassociation both C. jejuni and L. johnsonii were able to stably colonize the murine intestinal tract. Neither therapeutic nor prophylactic L. johnsonii application, however, could decrease intestinal C. jejuni burdens. Notably, C. jejuni induced colonic apoptosis could be ameliorated by prophylactic L. johnsonii treatment, whereas co-administration of L. johnsonii impacted adaptive (i.e. T and B lymphocytes, regulatory T cells), but not innate (i.e. macrophages and monocytes) immune cell responses in the intestinal tract. Strikingly, C. jejuni induced intestinal, extra-intestinal and systemic secretion of pro-inflammatory mediators (such as IL-6, MCP-1, TNF and nitric oxide) could be alleviated by peroral L. johnsonii challenge. In conclusion, immunomodulatory probiotic species might offer valuable strategies for prophylaxis and/or treatment of C. jejuni induced intestinal, extra-intestinal as well as systemic pro-inflammatory immune responses in vivo.
Collapse
Affiliation(s)
- Stefan Bereswill
- Department of Microbiology and Hygiene, Charité - University Medicine, Berlin, Germany
| | - Ira Ekmekciu
- Department of Microbiology and Hygiene, Charité - University Medicine, Berlin, Germany
| | - Ulrike Escher
- Department of Microbiology and Hygiene, Charité - University Medicine, Berlin, Germany
| | - Ulrike Fiebiger
- Department of Microbiology and Hygiene, Charité - University Medicine, Berlin, Germany
| | - Kerstin Stingl
- Federal Institute for Risk Assessment (BfR), Department of Biological Safety, National Reference Laboratory for Campylobacter, Berlin, Germany
| | - Markus M Heimesaat
- Department of Microbiology and Hygiene, Charité - University Medicine, Berlin, Germany.
| |
Collapse
|
12
|
Ekmekciu I, von Klitzing E, Fiebiger U, Neumann C, Bacher P, Scheffold A, Bereswill S, Heimesaat MM. The Probiotic Compound VSL#3 Modulates Mucosal, Peripheral, and Systemic Immunity Following Murine Broad-Spectrum Antibiotic Treatment. Front Cell Infect Microbiol 2017; 7:167. [PMID: 28529928 PMCID: PMC5418240 DOI: 10.3389/fcimb.2017.00167] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/18/2017] [Indexed: 12/16/2022] Open
Abstract
There is compelling evidence linking the commensal intestinal microbiota with host health and, in turn, antibiotic induced perturbations of microbiota composition with distinct pathologies. Despite the attractiveness of probiotic therapy as a tool to beneficially alter the intestinal microbiota, its immunological effects are still incompletely understood. The aim of the present study was to assess the efficacy of the probiotic formulation VSL#3 consisting of eight distinct bacterial species (including Streptococcus thermophilus, Bifidobacterium breve, B. longum, B. infantis, Lactobacillus acidophilus, L. plantarum, L. paracasei, and L. delbrueckii subsp. Bulgaricus) in reversing immunological effects of microbiota depletion as compared to reassociation with a complex murine microbiota. To address this, conventional mice were subjected to broad-spectrum antibiotic therapy for 8 weeks and perorally reassociated with either VSL#3 bacteria or a complex murine microbiota. VSL#3 recolonization resulted in restored CD4+ and CD8+ cell numbers in the small and large intestinal lamina propria as well as in B220+ cell numbers in the former, whereas probiotic intervention was not sufficient to reverse the antibiotic induced changes of respective cell populations in the spleen. However, VSL#3 application was as efficient as complex microbiota reassociation to attenuate the frequencies of regulatory T cells, activated dendritic cells and memory/effector T cells in the small intestine, colon, mesenteric lymph nodes, and spleen. Whereas broad-spectrum antibiotic treatment resulted in decreased production of cytokines such as IFN-γ, IL-17, IL-22, and IL-10 by CD4+ cells in respective immunological compartments, VSL#3 recolonization was sufficient to completely recover the expression of the anti-inflammatory cytokine IL-10 without affecting pro-inflammatory mediators. In summary, the probiotic compound VSL#3 has an extensive impact on mucosal, peripheral, and systemic innate as well as adaptive immunity, exerting beneficial anti-inflammatory effects in intestinal as well as systemic compartments. Hence, VSL#3 might be considered a therapeutic immunomodulatory tool following antibiotic therapy.
Collapse
Affiliation(s)
- Ira Ekmekciu
- Gastrointestinal Microbiology Research Group, Department of Microbiology and Hygiene, Institute for Microbiology and Hygiene, Charité - University MedicineBerlin, Germany
| | - Eliane von Klitzing
- Gastrointestinal Microbiology Research Group, Department of Microbiology and Hygiene, Institute for Microbiology and Hygiene, Charité - University MedicineBerlin, Germany
| | - Ulrike Fiebiger
- Gastrointestinal Microbiology Research Group, Department of Microbiology and Hygiene, Institute for Microbiology and Hygiene, Charité - University MedicineBerlin, Germany
| | - Christian Neumann
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - University MedicineBerlin, Germany.,German Rheumatism Research Center, Leibniz AssociationBerlin, Germany
| | - Petra Bacher
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - University MedicineBerlin, Germany
| | - Alexander Scheffold
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - University MedicineBerlin, Germany.,German Rheumatism Research Center, Leibniz AssociationBerlin, Germany
| | - Stefan Bereswill
- Gastrointestinal Microbiology Research Group, Department of Microbiology and Hygiene, Institute for Microbiology and Hygiene, Charité - University MedicineBerlin, Germany
| | - Markus M Heimesaat
- Gastrointestinal Microbiology Research Group, Department of Microbiology and Hygiene, Institute for Microbiology and Hygiene, Charité - University MedicineBerlin, Germany
| |
Collapse
|
13
|
Ekmekciu I, Fiebiger U, Stingl K, Bereswill S, Heimesaat MM. Amelioration of intestinal and systemic sequelae of murine Campylobacter jejuni infection by probiotic VSL#3 treatment. Gut Pathog 2017; 9:17. [PMID: 28413453 PMCID: PMC5387377 DOI: 10.1186/s13099-017-0168-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/05/2017] [Indexed: 12/16/2022] Open
Abstract
Background The incidence of human Campylobacter jejuni infections is progressively increasing worldwide. Probiotic compounds might open up valuable tools to decrease pathogen burden and subsequent pro-inflammatory immune responses, but in vivo data are scarce. Methods and results Secondary abiotic mice generated by broad-spectrum antibiotic treatment were perorally challenged with the commercial probiotic compound VSL#3 consisting of Streptococcus thermophilus, Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus paracasei, and Lactobacillus delbrueckii ssp. bulgaricus) either 5 days before (i.e. prophylactic regimen) or after (i.e. therapeutic regimen) peroral C. jejuni strain 81–176 infection, and analyzed 3 weeks following the initial bacterial re-association. Upon challenge, mice were colonized with the probiotic bacteria and/or C. jejuni at comparable intestinal loads, but co-colonization did not result in reduction of the pathogen burden. Remarkably, prophylactic as well as therapeutic VSL#3 treatment of C. jejuni infected mice ameliorated intestinal apoptosis and pro-inflammatory immune responses as indicated by lower numbers of innate and adaptive immune cell populations in the murine colon upon probiotic prophylaxis or treatment and reduced colonic concentrations of pro-inflammatory mediators including IL-6 and MCP-1. Importantly, concentrations of anti-inflammatory mediators such as IL-10 were significantly elevated in the colon of probiotics treated mice as compared to untreated controls. Strikingly, prophylactic VSL#3 treatment attenuated C. jejuni induced systemic pro-inflammatory responses as indicated by less TNF and IL-12p70 secretion in the spleen of VSL#3 pre-treated as compared to non-treated mice. Conclusion Administration of probiotic formulations such as VSL#3 might open up valuable strategies for prophylaxis and/or treatment of C. jejuni induced intestinal and systemic sequelae in vivo by the suppression of pro-inflammatory and induction of anti-inflammatory responses.
Collapse
Affiliation(s)
- Ira Ekmekciu
- Department of Microbiology and Hygiene, Charité-University Medicine Berlin, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Ulrike Fiebiger
- Department of Microbiology and Hygiene, Charité-University Medicine Berlin, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Kerstin Stingl
- Department of Biological Safety, Federal Institute for Risk Assessment (BfR), National Reference Laboratory for Campylobacter, Berlin, Germany
| | - Stefan Bereswill
- Department of Microbiology and Hygiene, Charité-University Medicine Berlin, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Markus M Heimesaat
- Department of Microbiology and Hygiene, Charité-University Medicine Berlin, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| |
Collapse
|
14
|
Wagner RD, Johnson SJ. Probiotic bacteria prevent Salmonella - induced suppression of lymphoproliferation in mice by an immunomodulatory mechanism. BMC Microbiol 2017; 17:77. [PMID: 28356067 PMCID: PMC5372341 DOI: 10.1186/s12866-017-0990-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/23/2017] [Indexed: 11/25/2022] Open
Abstract
Background Salmonella enterica infections often exhibit a form of immune evasion. We previously observed that probiotic bacteria could prevent inhibition of lymphoproliferation and apoptosis responses of T cells associated with S. enterica infections in orally challenged mice. Results In this study, changes in expression of genes related to lymphocyte activation in mucosa-associated lymphoid tissues (MALT) of mice orally infected with S. enterica with and without treatment with probiotic bacteria were evaluated. Probiotic bacteria increased expression of mRNA for clusters of differentiation antigen 2 (Cd2), protein tyrosine phosphatase receptor type C (Ptprc), and Toll-like receptor 6 (Tlr6) genes related to T and B cell activation in mouse intestinal tissue. The probiotic bacteria were also associated with reduced mRNA expression of a group of genes (RelB, Myd88, Iκκa, Jun, Irak2) related to nuclear factor of kappa light chains enhancer in B cells (NF-κB) signal transduction pathway-regulated cytokine responses. Probiotic bacteria were also associated with reduced mRNA expression of apoptotic genes (Casp2, Casp12, Dad1, Akt1, Bad) that suggest high avidity lymphocyte sparing. Reduced CD2 immunostaining in mesenteric lymph nodes (MLN) was suggestive of reduced lymphocyte activation in probiotic-treated mice. Reduced immunostaining of TLR6 in MALT of probiotic-treated, S. enterica-infected mice suggests that diminished innate immune sensitivity to S. enterica antigens is associated with preventing lymphocyte deletion. Conclusions The results of this study are consistent with prevention of S. enterica-induced deletion of lymphocytes by the influence of probiotic bacteria in mucosal lymphoid tissues of mice.
Collapse
Affiliation(s)
- R Doug Wagner
- Microbiology Division, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA.
| | - Shemedia J Johnson
- Microbiology Division, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| |
Collapse
|
15
|
Saint-Cyr MJ, Guyard-Nicodème M, Messaoudi S, Chemaly M, Cappelier JM, Dousset X, Haddad N. Recent Advances in Screening of Anti-Campylobacter Activity in Probiotics for Use in Poultry. Front Microbiol 2016; 7:553. [PMID: 27303366 PMCID: PMC4885830 DOI: 10.3389/fmicb.2016.00553] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/04/2016] [Indexed: 12/17/2022] Open
Abstract
Campylobacteriosis is the most common cause of bacterial gastroenteritis worldwide. Campylobacter species involved in this infection usually include the thermotolerant species Campylobacter jejuni. The major reservoir for C. jejuni leading to human infections is commercial broiler chickens. Poultry flocks are frequently colonized by C. jejuni without any apparent symptoms. Risk assessment analyses have identified the handling and consumption of poultry meat as one of the most important sources of human campylobacteriosis, so elimination of Campylobacter in the poultry reservoir is a crucial step in the control of this foodborne infection. To date, the use of probiotics has demonstrated promising results to reduce Campylobacter colonization. This review provides recent insights into methods used for probiotic screening to reduce the prevalence and colonization of Campylobacter at the farm level. Different eukaryotic epithelial cell lines are employed to screen probiotics with an anti-Campylobacter activity and yield useful information about the inhibition mechanism involved. These in vitro virulence models involve only human intestinal or cervical cell lines whereas the use of avian cell lines could be a preliminary step to investigate mechanisms of C. jejuni colonization in poultry in the presence of probiotics. In addition, in vivo trials to evaluate the effect of probiotics on Campylobacter colonization are conducted, taking into account the complexity introduced by the host, the feed, and the microbiota. However, the heterogeneity of the protocols used and the short time duration of the experiments lead to results that are difficult to compare and draw conclusions at the slaughter-age of broilers. Nevertheless, the combined approach using complementary in vitro and in vivo tools (cell cultures and animal experiments) leads to a better characterization of probiotic strains and could be employed to assess reduced Campylobacter spp. colonization in chickens if some parameters are optimized.
Collapse
Affiliation(s)
| | - Muriel Guyard-Nicodème
- Hygiene and Quality of Poultry and Pork Products Unit, Ploufragan/Plouzané Laboratory, ANSES, Université Bretagne LoirePloufragan, France
| | - Soumaya Messaoudi
- SECALIM Unit UMR1014, Oniris, INRA, Université Bretagne LoireNantes, France
| | - Marianne Chemaly
- Hygiene and Quality of Poultry and Pork Products Unit, Ploufragan/Plouzané Laboratory, ANSES, Université Bretagne LoirePloufragan, France
| | | | - Xavier Dousset
- SECALIM Unit UMR1014, Oniris, INRA, Université Bretagne LoireNantes, France
| | - Nabila Haddad
- SECALIM Unit UMR1014, Oniris, INRA, Université Bretagne LoireNantes, France
| |
Collapse
|
16
|
Saxena A. Probiotics as a Potential Alternative for Relieving Peripheral Neuropathies: a Case for Guillain-Barré Syndrome. Front Microbiol 2016; 6:1497. [PMID: 26779152 PMCID: PMC4703756 DOI: 10.3389/fmicb.2015.01497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/11/2015] [Indexed: 01/29/2023] Open
Affiliation(s)
- Abhishek Saxena
- Department of Biotechnology, TERI University New Delhi, India
| |
Collapse
|
17
|
Cosby DE, Cox NA, Harrison MA, Wilson JL, Buhr RJ, Fedorka-Cray PJ. Salmonella and antimicrobial resistance in broilers: A review. J APPL POULTRY RES 2015. [DOI: 10.3382/japr/pfv038] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
18
|
The role of probiotics in the inhibition of Campylobacter jejuni colonization and virulence attenuation. Eur J Clin Microbiol Infect Dis 2015; 34:1503-13. [PMID: 25934376 DOI: 10.1007/s10096-015-2392-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/19/2015] [Indexed: 01/30/2023]
Abstract
Campylobacter jejuni is one of the most common bacterial causes of human gastroenterocolitis worldwide, leading to diarrhea and other serious post-infectious complications. Probiotics form an attractive alternative intervention strategy for most of the enteric infections. However, the role of probiotics in C. jejuni infections requires detailed investigations in order to delineate the probiotic strains that are effective against C. jejuni. Although there are several biological mechanisms involved in the inhibition of pathogenic bacterial growth, the strains of probiotics and their mechanisms of actions through which they combat C. jejuni invasion have not been studied in greater detail. This mini review details the factors that are involved in the colonization and establishment of C. jejuni infection, with special reference to chickens, the natural host of C. jejuni, and the studies that have investigated the effect of different probiotic strains against C. jejuni colonization and growth. This review has collated the studies conducted using probiotics to inhibit C. jejuni colonization and growth to date to provide a collective knowledge about the role of probiotics as an alternative intervention strategy for campylobacteriosis.
Collapse
|
19
|
Wang G, Zhao Y, Tian F, Jin X, Chen H, Liu X, Zhang Q, Zhao J, Chen Y, Zhang H, Chen W. Screening of adhesive lactobacilli with antagonistic activity against Campylobacter jejuni. Food Control 2014. [DOI: 10.1016/j.foodcont.2014.03.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
20
|
Amalaradjou MAR, Bhunia AK. Bioengineered probiotics, a strategic approach to control enteric infections. Bioengineered 2013; 4:379-87. [PMID: 23327986 PMCID: PMC3937199 DOI: 10.4161/bioe.23574] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/09/2013] [Accepted: 01/10/2013] [Indexed: 01/19/2023] Open
Abstract
Enteric infections account for high morbidity and mortality and are considered to be the fifth leading cause of death at all ages worldwide. Seventy percent of all enteric infections are foodborne. Thus significant efforts have been directed toward the detection, control and prevention of foodborne diseases. Many antimicrobials including antibiotics have been used for their control and prevention. However, probiotics offer a potential alternative intervention strategy owing to their general health beneficial properties and inhibitory effects against foodborne pathogens. Often, antimicrobial probiotic action is non-specific and non-discriminatory or may be ineffective. In such cases, bioengineered probiotics expressing foreign gene products to achieve specific function is highly desirable. In this review we summarize the strategic development of recombinant bioengineered probiotics to control enteric infections, and to examine how scientific advancements in the human microbiome and their immunomodulatory effects help develop such novel and safe bioengineered probiotics.
Collapse
Affiliation(s)
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory; Department of Food Science; Purdue University; West Lafayette, IN USA
- Department of Comparative Pathobiology; Purdue University; West Lafayette, IN USA
| |
Collapse
|
21
|
Anti-inflammatory effect of lactobacilli bacteria on HepG2 cells is through cross-regulation of TLR4 and NOD2 signalling. J Funct Foods 2013. [DOI: 10.1016/j.jff.2013.01.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
22
|
Ganan M, Martinez-Rodriguez AJ, Carrascosa A, Vesterlund S, Salminen S, Satokari R. Interaction ofCampylobacterspp. and Human Probiotics in Chicken Intestinal Mucus. Zoonoses Public Health 2012; 60:141-8. [DOI: 10.1111/j.1863-2378.2012.01510.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
23
|
|
24
|
Amalaradjou MAR, Bhunia AK. Modern approaches in probiotics research to control foodborne pathogens. ADVANCES IN FOOD AND NUTRITION RESEARCH 2012; 67:185-239. [PMID: 23034117 PMCID: PMC7150249 DOI: 10.1016/b978-0-12-394598-3.00005-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Foodborne illness is a serious public health concern. There are over 200 known microbial, chemical, and physical agents that are known to cause foodborne illness. Efforts are made for improved detection, control and prevention of foodborne pathogen in food, and pathogen associated diseases in the host. Several commonly used approaches to control foodborne pathogens include antibiotics, natural antimicrobials, bacteriophages, bacteriocins, ionizing radiations, and heat. In addition, probiotics offer a potential intervention strategy for the prevention and control of foodborne infections. This review focuses on the use of probiotics and bioengineered probiotics to control foodborne pathogens, their antimicrobial actions, and their delivery strategies. Although probiotics have been demonstrated to be effective in antagonizing foodborne pathogens, challenges exist in the characterization and elucidation of underlying molecular mechanisms of action and in the development of potential delivery strategies that could maintain the viability and functionality of the probiotic in the target organ.
Collapse
|
25
|
Claes IJJ, De Keersmaecker SCJ, Vanderleyden J, Lebeer S. Lessons from probiotic-host interaction studies in murine models of experimental colitis. Mol Nutr Food Res 2011; 55:1441-53. [PMID: 21796777 DOI: 10.1002/mnfr.201100139] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 04/17/2011] [Accepted: 05/11/2011] [Indexed: 12/19/2022]
Abstract
In inflammatory bowel diseases (IBD), it is known that besides genetic and environmental factors (e.g. diet, drugs, stress), the microbiota play an important role in the pathogenesis. Patients with IBD have an altered microbiota (dysbiosis) and therefore, probiotics, defined as 'live micro-organisms that when administered in adequate amounts can confer a health benefit on the host', have been suggested as nutritional supplements to restore these imbalances. The best response on probiotics among the different types of IBD appears to be in the case of ulcerative colitis. Although probiotics show promise in IBD in both clinical and animal studies, further mechanistic studies are necessary to optimize the use of probiotics as supporting therapy in IBD. Murine models of experimental colitis have been used for decades to study this pathology, and these models have been proven useful to search for new therapeutic approaches. The purpose of this review is to summarize probiotic-host interaction studies in murine models of experimental colitis and to evaluate how these models can further help in understanding these complex interactions. Unraveling the molecular mechanisms behind the beneficial effects will assist in better and possibly more efficient probiotic formulations.
Collapse
Affiliation(s)
- Ingmar J J Claes
- Centre of Microbial and Plant Genetics, K.U. Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
26
|
Xue H, Sawyer MB, Wischmeyer PE, Baracos VE. Nutrition modulation of gastrointestinal toxicity related to cancer chemotherapy: from preclinical findings to clinical strategy. JPEN J Parenter Enteral Nutr 2011; 35:74-90. [PMID: 21224434 DOI: 10.1177/0148607110377338] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chemotherapy-induced gut toxicity is a major dose-limiting toxicity for many anticancer drugs. Gastrointestinal (GI) complications compromise the efficacy of chemotherapy, promote overall malnutrition, aggravate cancer cachexia, and may contribute to worsened prognosis. The GI tract is an attractive target for nutrition modulation, owing to its direct exposure to the diet, participation in uptake and metabolism of nutrients, high rate of cell turnover, and plasticity to nutrition stimuli. Glutamine, ω-3 polyunsaturated fatty acids, and probiotics/prebiotics are therapeutic factors that potentially modulate GI toxicity related to cancer treatments. Preclinical and clinical evidence are reviewed to critically define plausible benefits of these factors and their potential development into adjuncts to cancer chemotherapy. Mechanisms underlying the action of these nutrients are being unraveled in the laboratory. Optimal strategies to translate these findings into clinical care still remain to be elucidated. Key questions that remain to be answered include the following: which nutrient or combination of nutrients is selected for which patient and chemotherapy regimen? What mechanisms are responsible for modulation, and how are nutrient(s) administered in a clinically optimal manner? Research exploring interactions between different nutrients in GI protection is ongoing and demands further understanding. How nutrition preparations given to chemotherapy-treated patients are formulated in terms of component selection and dose optimization should be carefully studied and justified.
Collapse
Affiliation(s)
- Hongyu Xue
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.
| | | | | | | |
Collapse
|
27
|
Probiotic colonization of the adherent mucus layer of HT29MTXE12 cells attenuates Campylobacter jejuni virulence properties. Infect Immun 2010; 78:2812-22. [PMID: 20308300 DOI: 10.1128/iai.01249-09] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The HT29MTXE12 (E12) cell line harbors an adherent mucus layer, providing a novel technique to model mucosal infection in vitro. In this study, we have characterized the interaction of Campylobacter jejuni with the E12 cell line and exploited its unique mucus layer to examine the potential efficacy of probiotic treatment to attenuate C. jejuni virulence properties. C. jejuni 81-176 colonized and reproduced in E12 mucus. Adhesion to and internalization of C. jejuni were enhanced in E12 cells harboring mucus compared to parental cells without mucus. Translocation of C. jejuni occurred at early time points following infection. C. jejuni aligned with tight junctions and colocalized with the tight junction protein occludin, suggesting a paracellular route of translocation. Probiotic strains Lactobacillus rhamnosus R0011, Lactobacillus helveticus R0052, Lactobacillus salivarius AH102, Bifidobacterium longum AH1205, a commercial combination of L. rhamnosus R0011 and L. helveticus R0052 (Lacidofil), and a cocktail consisting of L. rhamnosus, L. helveticus, and L. salivarius (RhHeSa) colonized E12 mucus and bound to underlying cells. Probiotics attenuated C. jejuni association with and internalization into E12 cells and translocation to the basolateral medium of transwells. Live bacteria and prolonged precolonization of E12 cells with probiotics were necessary for probiotic action. These results demonstrate the potential for E12 cells as a model of mucosal pathogenesis and provide a rationale for the further investigation of probiotics as prophylaxis against human campylobacteriosis.
Collapse
|
28
|
Wohlgemuth S, Loh G, Blaut M. Recent developments and perspectives in the investigation of probiotic effects. Int J Med Microbiol 2009; 300:3-10. [PMID: 19783478 DOI: 10.1016/j.ijmm.2009.08.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The quest for a better understanding of how probiotics work has boosted an enormous interest in the molecular processes underlying host-microbe interactions. This review covers recent developments and perspectives in the study of probiotic mechanisms.
Collapse
Affiliation(s)
- Steffen Wohlgemuth
- German Institute of Human Nutrition Potsdam-Rehbrücke, Department of Gastrointestinal Microbiology, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
| | | | | |
Collapse
|
29
|
Bernardeau M, Gueguen M, Smith DGE, Corona-Barrera E, Vernoux JP. In vitro antagonistic activities of Lactobacillus spp. against Brachyspira hyodysenteriae and Brachyspira pilosicoli. Vet Microbiol 2009; 138:184-90. [PMID: 19356863 DOI: 10.1016/j.vetmic.2009.03.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Revised: 03/04/2009] [Accepted: 03/06/2009] [Indexed: 11/24/2022]
Abstract
The sensitivity of Brachyspira hyodysenteriae and Brachyspira pilosicoli, respectively the causative agents of Swine Dysentery and Porcine Intestinal Spirochaetosis to two probiotic Lactobacillus strains, L. rhamnosus CNCM-I-3698 and L. farciminis CNCM-I-3699 was studied through viability, motility and coaggregation assays. The cell-free supernatant of these lactobacilli contains lactic acid, that is stressful for Brachyspira (leading to the formation of spherical bodies), and lethal. It was demonstrated for the first time the in vitro coaggregation properties of two probiotic Lactobacillus strains (active or heat-treated) with two pathogenic strains of Brachyspira, leading to (1) trapping of spirochaetal cells in a physical network as demonstrated by SEM; (2) inhibition of the motility of Brachyspira. Such in vitro studies should encourage in vivo studies in animal model to evaluate the potential of the use of probiotic lactobacilli through a feeding strategy for the prevention of B. hyodysenteriae and B. pilosicoli.
Collapse
Affiliation(s)
- Marion Bernardeau
- Unité de recherche des Microorganismes d'Intérêt Laitier et Alimentaire, IFR 146 ICORE, Université de Caen Basse-Normandie, Esplanade de la Paix, 14032 Caen, France
| | | | | | | | | |
Collapse
|