1
|
Ahn SY, Joo HG, Ko EJ. Lactobacillus johnsonii JERA01 activates macrophages and increases Th-1 T cell population in mouse small intestine. PLoS One 2025; 20:e0320946. [PMID: 40273138 PMCID: PMC12021164 DOI: 10.1371/journal.pone.0320946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/26/2025] [Indexed: 04/26/2025] Open
Abstract
Lactobacillus johnsonii is a commensal bacterium isolated from the vaginal and gastrointestinal tracts of vertebrate hosts, including humans. It is a potential anti-inflammatory bacterium. As reported in many animal studies, L. johnsonii supplementation reduces inflammation in the intestine and enhances the epithelial barrier. However, in this study, we observed immunostimulatory effects of heat-killed L. johnsonii JERA01 (LJ) supplementation on antigen-presenting cells, such as dendritic cells and macrophages, in mice. LJ pretreatment increased the expression of maturation markers and TNF-α, IL-6, IL-12p40 production in bone marrow-derived dendritic cells and macrophages (BMDCs and BMDMs). Co-culture of LJ-pretreated BMDCs or BMDMs with lymphocytes enhanced IFN-γ production in vitro. Oral LJ (108 CFU) supplementation induced macrophage infiltration into the peritoneal cavity and Peyer's patch at 12-h after administration, resulting in an increase in the population of IFN-γ-producing T cells in the Peyer's patch. Our investigation revealed the effects of LJ, which activates macrophages and increases the Th-1 T cell population in the intestine, implying the possibility of using L. johnsonii as an immune stimulator.
Collapse
Affiliation(s)
- So Yeon Ahn
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Hong-Gu Joo
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Eun-Ju Ko
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
2
|
Zhang S, Zhong R, Zhou M, Li K, Lv H, Wang H, Xu Y, Liu D, Ma Q, Chen L, Zhang H. Mechanisms of Baicalin Alleviates Intestinal Inflammation: Role of M1 Macrophage Polarization and Lactobacillus amylovorus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415948. [PMID: 40200426 DOI: 10.1002/advs.202415948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/18/2025] [Indexed: 04/10/2025]
Abstract
Baicalin has been widely used for its anti-inflammatory pharmacological properties, yet its effects on bacterial intestinal inflammation and the mechanisms remain unclear. This study revealed that baicalin alleviates bacterial intestinal inflammation through regulating macrophage polarization and increasing Lactobacillus amylovorus abundance in colon. Specifically, transcriptomic analysis showed that baicalin restored Escherichia coli-induced genes expression changes including T helper cell 17 differentiation-related genes, macrophage polarization related genes, and TLR/IRF/STAT signaling pathway. Subsequent microbial and non-targeted metabolomic analysis revealed that these changes may be related to the enhancement of Lactobacillus amylovorus and the upregulation of its metabolites including chrysin, lactic acid, and indoles. Furthermore, whole-genome sequencing of Lactobacillus amylovorus provided insights into its functional potential and metabolic annotations. Lactobacillus amylovorus supplementation alleviates Escherichia coli-induced intestinal inflammation in mice and similarly inhibited M1 macrophage polarization through TLR4/IRF/STAT pathway. Additionally, baicalin, Lactobacillus amylovorus, or chrysin alone could regulate macrophage polarization, highlighting their independent anti-inflammatory potential. Notably, this study revealed that baicalin alleviates intestinal inflammation through TLR4/IRF/STAT pathway and increasing Lactobacillus amylovorus abundance and the synthesis of chrysin. These findings provide new insights into the therapeutic potential of baicalin and Lactobacillus amylovorus in preventing and treating intestinal inflammation, offering key targets for future interventions.
Collapse
Affiliation(s)
- Shunfen Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal Nutrition and Feed Science of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal Nutrition and Feed Science of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Miao Zhou
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
| | - Kai Li
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal Nutrition and Feed Science of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Huiyuan Lv
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Huixin Wang
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal Nutrition and Feed Science of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Ye Xu
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal Nutrition and Feed Science of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Dadan Liu
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal Nutrition and Feed Science of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Qiugang Ma
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal Nutrition and Feed Science of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal Nutrition and Feed Science of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| |
Collapse
|
3
|
Lam LY, Liang TR, Wu WJ, Lam HYP. Intestinal Lactobacillus johnsonii protects against neuroangiostrongyliasis in BALB/c mice through modulation of immune response. PLoS Negl Trop Dis 2025; 19:e0012977. [PMID: 40198714 PMCID: PMC11978024 DOI: 10.1371/journal.pntd.0012977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025] Open
Abstract
Neuroangiostrongyliasis is characterized by eosinophilic meningoencephalitis with a robust onset of severe neurological symptoms, by which immunological factors and peripheral metabolites have been postulated to affect the course of the disease. The gut-brain axis provides a bidirectional communication between the gut and the central nervous system, and therefore, understanding the gut microbiome may provide us with a deeper insight into the pathogenesis of angiostrongyliasis. Using 16S rRNA sequencing, we identified an increase in the abundance of different Lactobacillus species in Angiostrongylus cantonensis-infected mice, which was correlated to the disease severity. However, attempts to inoculate L. johnsonii into A. cantonensis-infected mice surprisingly revealed an improvement in neuroinflammation and prolonged survival. RNA sequencing suggested an immune-modulatory effect of L. johnsonii, which was confirmed by ELISA, showing increased levels of IL-10 and reduced levels of IL-2, IL-4, IL-5, and MCP-1 in the brain. Nevertheless, L. johnsonii-associated improvements were not associated with microbiome-related metabolites, as UHPLC-MS/MS analysis revealed no change in short-chain fatty acids, tryptophan metabolites, and bile acids. Our results suggest that while intestinal L. johnsonii appears to be linked to the progression of neuroangiostrongyliasis, these bacteria are likely attempting to modulate the dysregulated immune response to combat the disease. This is one of the first studies to investigate the gut microbiome in mice with A. cantonensis infection, which extends our knowledge from the microbiome-point-of-view of the pathogenesis of angiostrongyliasis and how the body defends against A. cantonensis. This work also extends to possible treatment approaches using L. johnsonii as probiotics.
Collapse
Affiliation(s)
- Long Yin Lam
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Ting-Ruei Liang
- PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien, Taiwan
| | - Wen-Jui Wu
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
| | - Ho Yin Pekkle Lam
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
4
|
Zhang X, Yin H, Yang X, Kang J, Sui N. Therapeutic Mechanism of Zhuyang Tongbian Decoction in Treating Functional Constipation: Insights from a Pilot Study Utilizing 16S rRNA Sequencing, Metagenomics, and Metabolomics. Int J Gen Med 2025; 18:1007-1022. [PMID: 40026814 PMCID: PMC11871934 DOI: 10.2147/ijgm.s509592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose To explore the mechanism of Zhuyang Tongbian Decoction (ZTD) in treating functional constipation (FC) by observing its effects on intestinal flora composition, the metabolic function of gut microbiota, fecal short-chain fatty acid (SCFA) levels, and serum concentrations of TLR4, NF-κB, TNF-α, and IL-6 in patients with FC. Patients and Methods 40 patients with FC were randomly divided into the control group and the treatment group, 20 cases in each group. And 20 healthy volunteers were recruited during the same period. The control group was administered lactulose, while the treatment group was treated with ZTD. 16s RNA sequencing technology was used to compare the changes in the structure and diversity of the intestinal flora of patients before and after treatment. Changes in the levels of SCFAs in faeces and the levels of TLR4, NF-κB, TNF-α and IL-6 in serum were analysed. Metagenomics sequencing assessed microbiota metabolic functions. Results The treatment group showed a significant increase in the relative abundance of beneficial bacteria, including Bifidobacterium, Lactobacillus, and Faecalibacterium_prausnitzii (P < 0.05), whereas Desulfobacterota and Ruminococcus were significantly reduced (P < 0.05). Notably, fecal acetic and propionic acid levels were significantly higher in the treatment group (P < 0.05). Serum biomarkers TLR4, NF-κB, TNF-α, and IL-6 decreased significantly (P < 0.05). Metagenomics sequencing showed that Carbohydrate metabolism, Metabolism of cofactors and vitamins, and C5- Branched dibasic acid metabolism were significantly increased in functional abundance (P < 0.05). Conclusion ZTD notably improves intestinal flora composition and gut microbiota metabolic function, regulates SCFA levels, and reduces inflammation markers in FC patients. The strain Faecalibacterium_prausnitzii shows significant potential in regulation of intestinal inflammation and may play a crucial role in the treatment efficacy of ZTD for FC.
Collapse
Affiliation(s)
- Xuan Zhang
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, People’s Republic of China
| | - Hang Yin
- Scientific Education Section, The Third Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, People’s Republic of China
| | - Xu Yang
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning Province, People’s Republic of China
| | - Jie Kang
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, People’s Republic of China
| | - Nan Sui
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, People’s Republic of China
- Department of Anorectal Diseases, The Third Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, People’s Republic of China
| |
Collapse
|
5
|
Nguyen HD, Vu GH, Kim WK. The molecular mechanisms of steroid hormone effects on cognitive function. Arch Gerontol Geriatr 2025; 129:105684. [PMID: 39549628 DOI: 10.1016/j.archger.2024.105684] [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: 08/13/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
OBJECTIVE There is a lack of information on the molecular mechanisms by which steroid hormones (testosterone, estrogen, and progesterone) regulate cognitive impairment. Thus, we aimed to identify the protective effects of steroid hormones on cognitive function. METHODS We analyzed the literature on the molecular mechanisms, biological activities, physicochemical properties, and pharmacokinetics of steroid hormones. RESULTS Steroid hormones can protect against cognitive impairment by regulating key genes (INS, TNF, STAT3, ESR1). Specific microRNAs, namely hsa-miR-335-5p, hsa-miR-16-5p, and hsa-miR-26b-5p, along with transcription factors NFKB1, PPARG, NR3C1, GATA2, EGR1, ATF3, and CEBPA, play a significant role in this protective mechanism. The involvement in cognitive processes, regulation of phosphorylation, neuronal apoptosis, and signaling pathways related to Alzheimer's disease significantly influence the protein-protein interaction network underlying these effects. Additionally, steroid hormones exhibit anti-hypercholesterolemic properties, anti-inflammatory activity, antitoxic properties, and function as inhibitors of acetylcholine neuromuscular transmission. They also hold promise as therapeutic agents for the treatment of dementia. Promising therapeutic interventions for cognitive impairment include the use of miRNA sponges targeting hsa-miR-16-5p, along with the administration of capsaicin, minocycline, dopamine, sertraline, and minaprine. The gut microbiota species Lactobacillus amylovorus, Paraprevotella clara, Libanicoccus massiliensis, Prevotella oris, Turicibacter sanguinis, and Dubosiella newyorkensis were identified as significant contributors to cognitive impairment and altered levels of steroid hormones. CONCLUSION Steroid hormones are promising compounds for improving cognitive function. Further research is needed to validate these findings through focused investigations into apoptosis, regulation of neuronal cell death, miRNA sponges, interactions with gut microbiota, and the potential efficacy of pharmaceutical agents.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Division of microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA.
| | - Giang Huong Vu
- Department of Public Health, Hong Bang Health Center, Hai Phong, Vietnam
| | - Woong-Ki Kim
- Division of microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA.
| |
Collapse
|
6
|
Li J, Shi M, Wang Y, Liu J, Liu S, Kang W, Liu X, Chen X, Huang K, Liu Y. Probiotic-derived extracellular vesicles alleviate AFB1-induced intestinal injury by modulating the gut microbiota and AHR activation. J Nanobiotechnology 2024; 22:697. [PMID: 39529091 PMCID: PMC11555919 DOI: 10.1186/s12951-024-02979-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Aflatoxin B1 (AFB1) is a mycotoxin that widely found in the environment and mouldy foods. AFB1 initially targets the intestine, and AFB1-induced intestinal injury cannot be ignored. Lactobacillus amylovorus (LA), a predominant species of Lactobacillus, plays a role in carbohydrate metabolism. Extracellular vesicles (EVs), small lipid membrane vesicles, are widely involved in diverse cellular processes. However, the mechanism by which Lactobacillus amylovorus-QC1H-derived EVs (LA.EVs) protect against AFB1-induced intestinal injury remains unclear. RESULTS In our study, a new strain named Lactobacillus amylovorus-QC1H (LA-QC1H) was isolated from pig faeces. Then, EVs derived from LA-QC1H were extracted via ultracentrifugation. Our results showed that LA.EVs significantly alleviated AFB1-induced intestinal injury by inhibiting the production of proinflammatory cytokines, decreasing intestinal permeability and increasing the expression of tight junction proteins. Moreover, 16 S rRNA analysis revealed that LA.EVs modulated AFB1-induced gut dysbiosis in mice. However, LA.EVs did not exert beneficial effects in antibiotic-treated mice. LA.EVs treatment increased intestinal levels of indole-3-acetic acid (IAA) and activated intestinal aryl hydrocarbon receptor (AHR)/interleukin-22 (IL-22) signalling in AFB1-exposed mice. Inhibition of intestinal AHR signalling markedly weakened the protective effect of LA.EVs in AFB1-exposed mice. CONCLUSIONS LA.EVs alleviated AFB1-induced intestinal injury by modulating the gut microbiota, activating the intestinal AHR/IL-22 signalling, reducing the inflammatory response and promoting intestinal barrier repair in mice.
Collapse
Affiliation(s)
- Jinyan Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Mengdie Shi
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yubo Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jinyan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Shuiping Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Weili Kang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Xianjiao Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Xingxiang Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yunhuan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, China.
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu, China.
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China.
| |
Collapse
|
7
|
Yoshikawa S, Itaya K, Hoshina R, Tashiro Y, Suda W, Cho Y, Matsuura M, Shindo C, Ito T, Hattori M, Miyamoto H, Kodama H. Thermophile-fermented feed modulates the gut microbiota related to lactate metabolism in pigs. J Appl Microbiol 2024; 135:lxae254. [PMID: 39333026 DOI: 10.1093/jambio/lxae254] [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: 04/16/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024]
Abstract
AIMS Extracts of fermented feed obtained via fermentation of marine animal resources with thermophilic Bacillaceae bacteria increase the fecundity of livestock. The intestinal bacterial profiles in response to long-term administration of this extract to pigs were investigated. METHODS AND RESULTS Half of a swine farm was supplied with potable water containing an extract of fermented feed for more than 2 years, whereas the other half was supplied with potable water without the extract. Feces from 6-month-old pigs rearing in these two areas were collected. 16S rRNA gene sequencing and isolation of lactic acid bacteria revealed an increase in the D/L-lactate-producing bacterium, Lactobacillus amylovorus, and a decrease in several members of Clostridiales following administration of fermented feed. A lactate-utilizing bacterium, Megasphaera elsdenii, was more abundant in the feces of pigs in the fermented feed group. All representative isolates of M. elsdenii showed rapid utilization of D-lactate relative to L-lactate, and butyrate and valerate were the main products. CONCLUSION The probiotic effect of fermented feed is associated with the modulation of lactate metabolism in the digestive organs of pigs.
Collapse
Affiliation(s)
- Shota Yoshikawa
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
| | - Kaede Itaya
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
| | - Ryo Hoshina
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
| | - Yukihiro Tashiro
- Institute of Advanced Study, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
- Laboratory of Functional Food Design, Department of Functional Metabolic Design, Bio-Architecture Center, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
| | - Wataru Suda
- RIKEN Center for Integrative Medical Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| | - Yuichiro Cho
- Department of Anatomy and Physiological Science, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Makiko Matsuura
- Sermas Co., Ltd., 4-3-5 Onitaka, Ichikawa City, Chiba 272-0015, Japan
| | - Chie Shindo
- RIKEN Center for Integrative Medical Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| | - Toshiyuki Ito
- Keiyo Gas Energy Solution Co. Ltd., 4-3-5 Onitaka, Ichikawa City, Chiba 272-0015, Japan
| | - Masahira Hattori
- RIKEN Center for Integrative Medical Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
- School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Hirokuni Miyamoto
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
- RIKEN Center for Integrative Medical Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
- Sermas Co., Ltd., 4-3-5 Onitaka, Ichikawa City, Chiba 272-0015, Japan
- Japan Eco-science (Nikkan Kagaku) Co. Ltd., 11-1-211 Shiomigaokacho, Chiba City, Chiba 260-0034, Japan
| | - Hiroaki Kodama
- Graduate School of Horticulture, Chiba University, 648 Matsudo, Chiba 271-8510, Japan
| |
Collapse
|
8
|
Cheng Y, Xiao X, Fu J, Zong X, Lu Z, Wang Y. Escherichia coli K88 activates NLRP3 inflammasome-mediated pyroptosis in vitro and in vivo. Biochem Biophys Rep 2024; 38:101665. [PMID: 38419757 PMCID: PMC10900769 DOI: 10.1016/j.bbrep.2024.101665] [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: 04/17/2023] [Revised: 11/20/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Pyroptosis induced by lipopolysaccharide (LPS) has an obvious impact on intestinal inflammation and immune regulation. Enterotoxigenic Escherichia coli (ETEC) K88 has been proved to induce inflammatory responses in several models, but whether E. coli K88 participates in the same process of pyroptotic cell death as LPS remains to be identified. We conducted a pilot experiment to confirm that E. coli K88, instead of Escherichia coli O157 and Salmonella typhimurium, promotes the secretion of interleukin-1 beta (IL-1β) and interleukin-18 (IL-18) in macrophages. Further experiments were carried out to dissect the molecular mechanism both in vitro and in vivo. The Enzyme-Linked Immunosorbent Assay (ELISA) results suggested that E. coli K88 treatment increased the expression of pro-inflammatory cytokines IL-18 and IL-1β in both C57BL/6 mice and the supernatant of J774A.1 cells. Intestinal morphology observations revealed that E. coli K88 treatment mainly induced inflammation in the colon. Real-time PCR and Western blot analysis showed that the mRNA and protein expressions of pyroptosis-related factors, such as NLRP3, ASC, and Caspase1, were significantly upregulated by E. coli K88 treatment. The RNA-seq results confirmed that the effect was associated with the activation of NLRP3, ASC, Caspase1, GSDMD, IL-18, and IL-1β, and might also be related to inflammatory bowel disease and the tumor necrosis factor pathway. The pyroptosis-activated effect of E. coli K88 was significantly blocked by NLRP3 siRNA. Our data suggested that E. coli K88 caused inflammation by triggering pyroptosis, which provides a theoretical basis for the prevention and treatment of ETEC in intestinal infection.
Collapse
Affiliation(s)
- Yuanzhi Cheng
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| | - Xiao Xiao
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang A&F University, Hangzhou, 311300, China
| | - Jie Fu
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| | - Xin Zong
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| | - Zeqing Lu
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| | - Yizhen Wang
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| |
Collapse
|
9
|
Wu Y, Liu X, Zou Y, Zhang X, Wang Z, Hu J, Han D, Zhao J, Dai Z, Wang J. Lactobacillus amylovorus Promotes Lactose Utilization in Small Intestine and Enhances Intestinal Barrier Function in Intrauterine Growth Restricted Piglets. J Nutr 2024; 154:535-542. [PMID: 38072153 DOI: 10.1016/j.tjnut.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/28/2023] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) resulted in high mortality and many physiological defects of piglets, causing huge economic loss in the swine industry. Lactobacillus amylovorus (L. amylovorus) was identified as one of the main differential bacteria between IUGR and normal piglets. However, the effects of L. amylovorus on the growth performance and intestinal health in IUGR piglets remained unclear. OBJECTIVES This study aimed to investigate the promoting effects of L. amylovorus Mafic1501, a new strain isolated from normal piglets, on the growth performance and intestinal barrier functions in IUGR piglets. METHODS Newborn mice or piglets were assigned into 3 groups: CON (normal birth weight, control), IUGR (low birth weight), and IUGR+L. amy (low birth weight), administered with sterile saline or L. amylovorus Mafic1501, respectively. Growth performance, lactose content in the digesta, intestinal lactose transporter, and barrier function parameters were profiled. IPEC-J2 cells were cultured to verify the effects of L. amylovorus Mafic1501 on lactose utilization and intestinal barrier functions. RESULTS L. amylovorus Mafic1501 elevated body weight and average daily gain of IUGR mice and piglets (P < 0.05). The lactose content in the ileum was decreased, whereas gene expression of glucose transporter 2 (GLUT2) was increased by L. amylovorus Mafic1501 in IUGR piglets during suckling period (P < 0.05). Besides, L. amylovorus Mafic1501 promoted intestinal barrier functions by increasing the villus height and relative gene expressions of tight junctions (P < 0.05). L. amylovorus Mafic1501 and its culture supernatant decreased the lactose level in the medium and upregulated gene expressions of transporter GLUT2 and tight junction protein Claudin-1 of IPEC-J2 cells (P < 0.05). CONCLUSION L. amylovorus Mafic1501 improved the growth performance of IUGR piglets by promoting the lactose utilization in small intestine and enhancing intestinal barrier functions. Our results provided the new evidence of L. amylovorus Mafic1501 for its application in the swine industry.
Collapse
Affiliation(s)
- Yujun Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiaoyi Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Youwei Zou
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhenyu Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jie Hu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| |
Collapse
|
10
|
Adejumo SA, Oli AN, ROWAIYE AB, IGBOKWE NH, EZEJIEGU CK, YAHAYA ZS. Immunomodulatory Benefits of Probiotic Bacteria: A Review of Evidence. OBM GENETICS 2023; 07:1-73. [DOI: 10.21926/obm.genet.2304206] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Over the past few decades, probiotics have emerged as a viable medical tool for preventing and/or treating diseases. This narrative review provides recent findings on Probiotics and their benefits on the host immune system. It also highlights the specific mechanisms through which probiotics mediate those benefits. The study also explores the topical or systemic probiotic administration method. Authors screened databases like Google Scholar, Web of Science, PubMed, Scopus, and China National Knowledge Infrastructure database, using various keyword combinations such as: “probiotic” AND “Immunomodulation” OR “probiotic” AND “Immunoregulation” OR “probiotic” AND “Immunostimulation”, for relevant literature written in English only. The review shows that probiotics can regulate the host immune system, including regulating T cells, dendritic cells, intestinal epithelial cells, and several signal pathways, and confer health benefits. Although several clinical trials also revealed the prospects and efficacy of probiotics as immunomodulators and treatment of diseases, there is a need for thorough future investigations on the effectiveness of specific strains of probiotics involved in immunomodulation.
Collapse
|
11
|
Wang W, Peng Y, Nie Y, Wang Y, Wang C, Huang B. Dietary supplementation with Acremonium terricola culture alters the gut microbial structure and improves the growth performance, antioxidant status, and immune function of weaning piglets. BMC Vet Res 2023; 19:258. [PMID: 38053083 DOI: 10.1186/s12917-023-03778-y] [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: 11/27/2022] [Accepted: 10/06/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Acremonium terricola is used in the feed of dairy animals to promote growth and control diseases. However, the effects of dietary supplementation with A. terricola on the gut microbial structure of weaning piglets remain poorly understood. Therefore, in this study, we investigated the effects of dietary supplementation with A. terricola culture (ATC) on the growth performance, antioxidant status, immunity, and gut environment of weaning piglets. Sixty piglets were fed a basal diet supplemented with 1 g ATC/kg of basal diet (experimental group). Another 60 piglets did not receive ATC (control group). The intervention lasted for 20 days. RESULTS The experimental group had higher daily weight gain and feed efficiency than did the control group. Significant increases were noted in the levels of serum insulin (P = 0.0018), insulin-like growth factor (P = 0.0018), triiodothyronine (P = 0.0031), immunoglobulin A (P < 0.0001), immunoglobulin M (P = 0.001), immunoglobulin G (P = 0.0001), and interferon γ (P < 0.0001) in the experimental group compared with the levels in the control group. Furthermore, ATC supplementation significantly reduced (P < 0.05) the relative abundance of Shuttleworthia, Succinivibrio, Roseburia, Ruminococcus, and Paludibacter but increased that of Phascolarctobacterium, Megasphaera, Faecalibacterium, and Prevotella in the experimental group compared with that in the control group. Notably, ATC supplementation significantly increased the relative abundance of Faecalibacterium prausnitzii (P < 0.05), which is involved in anti-inflammatory activities, gut barrier enhancement, and butyrate production. CONCLUSIONS Dietary supplementation with ATC may improve the growth performance, antioxidant status, immunity, and fecal microflora of weaning pigs.
Collapse
Affiliation(s)
- Wei Wang
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, 230036, China
- Jiangxi Zhengbang Academy of Agricultural Sciences, Nanchang, 330000, China
| | - Yizhu Peng
- Jiangxi Zhengbang Academy of Agricultural Sciences, Nanchang, 330000, China
| | - Yong Nie
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, 230036, China
- School of Civil Engineering and Architecture, Anhui University of Technology, Ma'anshan, 243002, China
| | - Yulong Wang
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, 230036, China
| | - Chuang Wang
- Jiangxi Zhengbang Academy of Agricultural Sciences, Nanchang, 330000, China.
| | - Bo Huang
- Anhui Provincial Key Laboratory of Microbial Pest Control, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
12
|
Middelkoop A, Segarra S, Molist F. Porcine digestible peptides as alternative protein source in weaner diets: effects on performance and systemic cytokine profile in pigs followed from weaning to slaughter. Animal 2023; 17:100998. [PMID: 37897869 DOI: 10.1016/j.animal.2023.100998] [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: 04/12/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 10/30/2023] Open
Abstract
Porcine digestible peptides (PDP) are high-quality hydrolysed proteins obtained from porcine intestinal mucosa as a by-product of the heparin manufacturing process. PDP contain bioactive peptides and are used as alternative protein sources in several animal species, including pigs. We aimed to explore the (carry-over) effects of feeding PDP to weaned piglets on performance and systemic cytokine levels of pigs followed until slaughter. A total of 192 piglets were allocated to one of two dietary treatments: control (CON) or PDP weaner diets. PDP was included at 5.0% until day 13 post-weaning at the expense of skimmed milk powder and partial replacement of soybean meal, and at 2.5% between days 13 and 34 post-weaning at the expense of soy protein concentrate. Grower-finishers were fed commercial diets according to a 3-phase feeding scheme until slaughter, when carcass traits were determined. Six pigs were housed per weaner pen and eight per grower-finisher pen with 16 and 10 pens per treatment, respectively. Pigs were weighed at the start and at the end of each phase, and feed intake was recorded. Faecal consistency was recorded twice a week in the weaner facility. Ten pigs per treatment were sampled for blood at days 13, 34 and 69 post-weaning. We found that PDP-fed piglets had a higher feed intake in the first two weeks post-weaning compared to CON-fed piglets (+32 g/pig per day; P = 0.02). Moreover, piglets in the PDP group showed improved feed conversion between days 13 and 34 versus the CON group (1.36 vs 1.43; P = 0.03). Piglets that were fed with PDP in the weaner diets tended to grow faster in the grower-finisher period (+32 g/pig per day; P = 0.07), tended to reach slaughter age earlier (129.9 vs 131.5 days; P = 0.07) and had a lower dressing percentage at slaughter (76.3 vs 76.7%, P = 0.045) than piglets previously fed with CON. Additionally, PDP-fed piglets showed higher serum levels of pro-inflammatory cytokines interleukin (IL)-12 (P = 0.02), tumour necrosis factor-alpha (P = 0.02), interferon-gamma (P = 0.03) and IL-8 (at day 34 post-weaning, P = 0.06) as well as anti-inflammatory cytokines transforming growth factor-beta (P = 0.02), IL-4 (P = 0.04) and IL-10 (at day 34 post-weaning, P = 0.02). No significant differences among dietary treatments were observed regarding faecal consistency of weaned piglets and carcass weight, lean meat percentage, muscle depth, and back fat thickness at slaughter. We conclude that feeding PDP, as an alternative to conventional milk and soy protein sources, showed positive effects on pig performance, not only during the provisioning period but also thereafter into the grower-finisher phase.
Collapse
Affiliation(s)
- A Middelkoop
- R&D, Schothorst Feed Research B.V, PO Box 533, 8200 AM Lelystad, The Netherlands.
| | - S Segarra
- R&D Animal Health, Bioiberica S.A.U, 08950 Esplugues de Llobregat, Spain
| | - F Molist
- R&D, Schothorst Feed Research B.V, PO Box 533, 8200 AM Lelystad, The Netherlands
| |
Collapse
|
13
|
Splichalova A, Kindlova Z, Killer J, Neuzil Bunesova V, Vlkova E, Valaskova B, Pechar R, Polakova K, Splichal I. Commensal Bacteria Impact on Intestinal Toll-like Receptor Signaling in Salmonella-Challenged Gnotobiotic Piglets. Pathogens 2023; 12:1293. [PMID: 38003758 PMCID: PMC10675043 DOI: 10.3390/pathogens12111293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Gnotobiotic (GN) animals with simple and defined microbiota can help to elucidate host-pathogen interferences. Hysterectomy-derived germ-free (GF) minipigs were associated at 4 and 24 h post-hysterectomy with porcine commensal mucinolytic Bifidobacterium boum RP36 (RP36) strain or non-mucinolytic strain RP37 (RP37) or at 4 h post-hysterectomy with Lactobacillus amylovorus (LA). One-week-old GN minipigs were infected with Salmonella Typhimurium LT2 strain (LT2). We monitored histological changes in the ileum, mRNA expression of Toll-like receptors (TLRs) 2, 4, and 9 and their related molecules lipopolysaccharide-binding protein (LBP), coreceptors MD-2 and CD14, adaptor proteins MyD88 and TRIF, and receptor for advanced glycation end products (RAGE) in the ileum and colon. LT2 significantly induced expression of TLR2, TLR4, MyD88, LBP, MD-2, and CD14 in the ileum and TLR4, MyD88, TRIF, LBP, and CD14 in the colon. The LT2 infection also significantly increased plasmatic levels of inflammatory markers interleukin (IL)-6 and IL-12/23p40. The previous colonization with RP37 alleviated damage of the ileum caused by the Salmonella infection, and RP37 and LA downregulated plasmatic levels of IL-6. A defined oligo-microbiota composed of bacterial species with selected properties should probably be more effective in downregulating inflammatory response than single bacteria.
Collapse
Affiliation(s)
- Alla Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| | - Zdislava Kindlova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| | - Jiri Killer
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic; (J.K.); (V.N.B.); (E.V.); (R.P.)
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Vera Neuzil Bunesova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic; (J.K.); (V.N.B.); (E.V.); (R.P.)
| | - Eva Vlkova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic; (J.K.); (V.N.B.); (E.V.); (R.P.)
| | - Barbora Valaskova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| | - Radko Pechar
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic; (J.K.); (V.N.B.); (E.V.); (R.P.)
- Department of Research, Food Research Institute Prague, 102 00 Prague, Czech Republic
| | - Katerina Polakova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| | - Igor Splichal
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| |
Collapse
|
14
|
Fagan MM, Welch CB, Scheulin KM, Sneed SE, Jeon JH, Golan ME, Cheek SR, Barany DA, Oeltzschner G, Callaway TR, Zhao Q, Park HJ, Lourenco JM, Duberstein KJ, West FD. Fecal microbial transplantation limits neural injury severity and functional deficits in a pediatric piglet traumatic brain injury model. Front Neurosci 2023; 17:1249539. [PMID: 37841685 PMCID: PMC10568032 DOI: 10.3389/fnins.2023.1249539] [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: 06/28/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Pediatric traumatic brain injury (TBI) is a leading cause of death and disability in children. Due to bidirectional communication between the brain and gut microbial population, introduction of key gut bacteria may mitigate critical TBI-induced secondary injury cascades, thus lessening neural damage and improving functional outcomes. The objective of this study was to determine the efficacy of a daily fecal microbial transplant (FMT) to alleviate neural injury severity, prevent gut dysbiosis, and improve functional recovery post TBI in a translational pediatric piglet model. Male piglets at 4-weeks of age were randomly assigned to Sham + saline, TBI + saline, or TBI + FMT treatment groups. A moderate/severe TBI was induced by controlled cortical impact and Sham pigs underwent craniectomy surgery only. FMT or saline were administered by oral gavage daily for 7 days. MRI was performed 1 day (1D) and 7 days (7D) post TBI. Fecal and cecal samples were collected for 16S rRNA gene sequencing. Ipsilateral brain and ileum tissue samples were collected for histological assessment. Gait and behavior testing were conducted at multiple timepoints. MRI showed that FMT treated animals demonstrated decreased lesion volume and hemorrhage volume at 7D post TBI as compared to 1D post TBI. Histological analysis revealed improved neuron and oligodendrocyte survival and restored ileum tissue morphology at 7D post TBI in FMT treated animals. Microbiome analysis indicated decreased dysbiosis in FMT treated animals with an increase in multiple probiotic Lactobacilli species, associated with anti-inflammatory therapeutic effects, in the cecum of the FMT treated animals, while non-treated TBI animals showed an increase in pathogenic bacteria, associated with inflammation and disease such in feces. FMT mediated enhanced cellular and tissue recovery resulted in improved motor function including stride and step length and voluntary motor activity in FMT treated animals. Here we report for the first time in a highly translatable pediatric piglet TBI model, the potential of FMT treatment to significantly limit cellular and tissue damage leading to improved functional outcomes following a TBI.
Collapse
Affiliation(s)
- Madison M. Fagan
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Biomedical and Health Sciences Institute, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Christina B. Welch
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Kelly M. Scheulin
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Biomedical and Health Sciences Institute, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Sydney E. Sneed
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Julie H. Jeon
- Department of Nutritional Sciences, College of Family and Consumer Sciences, University of Georgia, Athens, GA, United States
| | - Morgane E. Golan
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Biomedical and Health Sciences Institute, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Savannah R. Cheek
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Deborah A. Barany
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Kinesiology, College of Education, University of Georgia, Athens, GA, United States
| | - Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Todd R. Callaway
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Qun Zhao
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Physics and Astronomy, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
| | - Hea Jin Park
- Department of Nutritional Sciences, College of Family and Consumer Sciences, University of Georgia, Athens, GA, United States
| | - Jeferson M. Lourenco
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Kylee J. Duberstein
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Biomedical and Health Sciences Institute, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Franklin D. West
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Biomedical and Health Sciences Institute, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| |
Collapse
|
15
|
Kiernan DP, O’Doherty JV, Sweeney T. The Effect of Maternal Probiotic or Synbiotic Supplementation on Sow and Offspring Gastrointestinal Microbiota, Health, and Performance. Animals (Basel) 2023; 13:2996. [PMID: 37835602 PMCID: PMC10571980 DOI: 10.3390/ani13192996] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
The increasing prevalence of antimicrobial-resistant pathogens has prompted the reduction in antibiotic and antimicrobial use in commercial pig production. This has led to increased research efforts to identify alternative dietary interventions to support the health and development of the pig. The crucial role of the GIT microbiota in animal health and performance is becoming increasingly evident. Hence, promoting an improved GIT microbiota, particularly the pioneer microbiota in the young pig, is a fundamental focus. Recent research has indicated that the sow's GIT microbiota is a significant contributor to the development of the offspring's microbiota. Thus, dietary manipulation of the sow's microbiota with probiotics or synbiotics, before farrowing and during lactation, is a compelling area of exploration. This review aims to identify the potential health benefits of maternal probiotic or synbiotic supplementation to both the sow and her offspring and to explore their possible modes of action. Finally, the results of maternal sow probiotic and synbiotic supplementation studies are collated and summarized. Maternal probiotic or synbiotic supplementation offers an effective strategy to modulate the sow's microbiota and thereby enhance the formation of a health-promoting pioneer microbiota in the offspring. In addition, this strategy can potentially reduce oxidative stress and inflammation in the sow and her offspring, enhance the immune potential of the milk, the immune system development in the offspring, and the sow's feed intake during lactation. Although many studies have used probiotics in the maternal sow diet, the most effective probiotic or probiotic blends remain unclear. To this extent, further direct comparative investigations using different probiotics are warranted to advance the current understanding in this area. Moreover, the number of investigations supplementing synbiotics in the maternal sow diet is limited and is an area where further exploration is warranted.
Collapse
Affiliation(s)
- Dillon P. Kiernan
- School of Veterinary Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
| | - John V. O’Doherty
- School of Agriculture and Food Science, University College Dublin, D04 C1P1 Dublin, Ireland;
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
| |
Collapse
|
16
|
Steigleder KM, Pascoal LB, Siqueira NSN, Simino LADP, Ayrizono MDLS, Ferreira MM, Fagundes JJ, Azevedo ATD, Torsoni AS, Leal RF. Mathematical Models Including microRNA Levels of Mesenteric Adipose Tissue May Predict Postoperative Relapse in Crohn's Disease Patients. GASTRO HEP ADVANCES 2023; 3:17-30. [PMID: 39132178 PMCID: PMC11307883 DOI: 10.1016/j.gastha.2023.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/21/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims Recent evidence suggests that the mesenteric adipose tissue (MAT) near the affected intestine may play a role in Crohn's disease (CD) pathophysiology. Modulation of several transcripts has already been identified in the MAT of CD in the literature. Therefore, our aim was to validate the microRNA (miRNA) transcript levels and their target genes in the MAT of active CD patients and correlate them with clinical and epidemiological data. Methods Samples from the MAT of surgical specimens from 25 active CD patients were obtained. The control group comprised fifteen patients who underwent surgery for other diseases, except inflammatory bowel diseases. Transcriptional levels of miRNA and their target genes were assessed by quantitative real-time polymerase chain reaction. The correlation between transcripts and clinical characteristics was obtained using multiple linear regression. The mathematical models (M) underwent a statistical filter to ensure robustness and reliability (P value < .05; adjusted R-squared (Rˆ2)> .99; correct predictions of more than 60%). Results miRNA-650 and miRNA-29c were upregulated in the MAT of CD compared to the control group (P < .0001 and P = .0032, respectively), besides presenting decreased levels of their target genes. Two were target genes of the miRNA-650: glutamine-fructose-6-phosphate transaminase 2 (P = .012) and aldehyde dehydrogenase 4 family (P = .0035); and 4 were targets of the miRNA-29c: cell death-inducing DFFA-like effector c (P = .001), E2F transcription factor-1 (P = .007), hypoxia-inducible factor 3 subunit alpha (P = .0029), and pyruvate dehydrogenase kinase 4 (P = .0054). We found 2 M with statistical strength and robustness. The performance test identified one model with 100% accuracy for predicting the month of recurrence and determining patients with less risk of early relapse after surgery. Conclusion We demonstrate that miRNA-650 and miRNA-29c and some of their target genes, besides clinical and epidemiological variables, may be useful in a model to predict when disease relapse may occur in CD patients who underwent surgery. These findings constitute a potential tool to guide postoperative clinical management.
Collapse
Affiliation(s)
- Karine Mariane Steigleder
- Inflammatory Bowel Diseases Research Laboratory, Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Lívia Bitencourt Pascoal
- Inflammatory Bowel Diseases Research Laboratory, Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Natália Souza Nunes Siqueira
- Inflammatory Bowel Diseases Research Laboratory, Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Laís Angélica de Paula Simino
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas (Unicamp), Limeira, São Paulo, Brazil
| | - Maria de Lourdes Setsuko Ayrizono
- Inflammatory Bowel Diseases Research Laboratory, Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Marciane Milanski Ferreira
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas (Unicamp), Limeira, São Paulo, Brazil
| | - João José Fagundes
- Inflammatory Bowel Diseases Research Laboratory, Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Aníbal Tavares de Azevedo
- Simulation Laboratory, School of Applied Sciences, University of Campinas (Unicamp), Limeira, Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas (Unicamp), Limeira, São Paulo, Brazil
| | - Raquel Franco Leal
- Inflammatory Bowel Diseases Research Laboratory, Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| |
Collapse
|
17
|
Ismael M, Qayyum N, Gu Y, Zhezhe Y, Cui Y, Zhang Y, Lü X. Protective effect of plantaricin bio-LP1 bacteriocin on multidrug-resistance Escherichia Coli infection by alleviate the inflammation and modulate of gut-microbiota in BALB/c mice model. Int J Biol Macromol 2023; 246:125700. [PMID: 37414312 DOI: 10.1016/j.ijbiomac.2023.125700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
The rapid spread of multidrug-resistant pathogens with the low efficacy of common antibiotics for humans and animals in its clinical therapeutics are a global health concern. Therefore, there is a need to develop new treatment strategies to control them clinically. The study aimed to evaluate the effects of Plantaricin Bio-LP1 bacteriocin produced from Lactiplantibacillus plantarum NWAFU-BIO-BS29 to alleviate the inflammation caused by multidrug-resistance Escherichia Coli (MDR-E. coli) infection in BALB/c mice-model. The focus was given on aspects linked to the mechanism of the immune response. Results indicated that Bio-LP1 had highly promising effects on partially ameliorating MDR-E. coli infection by reducing the inflammatory response through inhibiting the overexpression of proinflammatory-cytokines such as secretion of tumor necrosis factor (TNF-α) and interleukin (IL-6 and IL-β) and strongly regulated theTLR4 signaling-pathway. Additionally, avoided the villous destruct, colon length shortening, loss of intestinal barrier integrity, and increased disease activity index. Furthermore, significantly increased the relative abundance of beneficial-intestinal-bacteria including Ligilactobacillus, Enterorhabdus, Pervotellaceae, etc. Finally, improved the intestinal mucosal barrier to alleviate the pathological damages and promote the production of short-chain fatty acids (SCFAs) a source of energy for the proliferation. In conclusion, plantaricin Bio-LP1 bacteriocin can be considered a safe alternative to antibiotics against MDR-E. coli-induced intestinal inflammation.
Collapse
Affiliation(s)
- Mohamedelfatieh Ismael
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; Sudanese Standard and Metrology Organization, Khartoum, 13573, Sudan
| | - Nageena Qayyum
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Yaxin Gu
- College of Food Science, China Agricultural University, Beijing, China
| | - Yu Zhezhe
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Yanlong Cui
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Yu Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
18
|
Du W, Wang X, Hu M, Hou J, Du Y, Si W, Yang L, Xu L, Xu Q. Modulating gastrointestinal microbiota to alleviate diarrhea in calves. Front Microbiol 2023; 14:1181545. [PMID: 37362944 PMCID: PMC10286795 DOI: 10.3389/fmicb.2023.1181545] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
The calf stage is a critical period for the development of heifers. Newborn calves have low gastrointestinal barrier function and immunity before weaning, making them highly susceptible to infection by various intestinal pathogens. Diarrhea in calves poses a significant threat to the health of young ruminants and may cause serious economic losses to livestock farms. Antibiotics are commonly used to treat diarrhea and promote calf growth, leading to bacterial resistance and increasing antibiotic residues in meat. Therefore, finding new technologies to improve the diarrhea of newborn calves is a challenge for livestock production and public health. The operation of the gut microbiota in the early stages after birth is crucial for optimizing immune function and body growth. Microbiota colonization of newborn animals is crucial for healthy development. Early intervention of the calf gastrointestinal microbiota, such as oral probiotics, fecal microbiota transplantation and rumen microbiota transplantation can effectively relieve calf diarrhea. This review focuses on the role and mechanisms of oral probiotics such as Lactobacillus, Bifidobacterium and Faecalibacterium in relieving calf diarrhea. The aim is to develop appropriate antibiotic alternatives to improve calf health in a sustainable and responsible manner, while addressing public health issues related to the use of antibiotics in livestock.
Collapse
|
19
|
Lombardi F, Augello FR, Palumbo P, Bonfili L, Artone S, Altamura S, Sheldon JM, Latella G, Cifone MG, Eleuteri AM, Cinque B. Bacterial Lysate from the Multi-Strain Probiotic SLAB51 Triggers Adaptative Responses to Hypoxia in Human Caco-2 Intestinal Epithelial Cells under Normoxic Conditions and Attenuates LPS-Induced Inflammatory Response. Int J Mol Sci 2023; 24:ijms24098134. [PMID: 37175841 PMCID: PMC10179068 DOI: 10.3390/ijms24098134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α), a central player in maintaining gut-microbiota homeostasis, plays a pivotal role in inducing adaptive mechanisms to hypoxia and is negatively regulated by prolyl hydroxylase 2 (PHD2). HIF-1α is stabilized through PI3K/AKT signaling regardless of oxygen levels. Considering the crucial role of the HIF pathway in intestinal mucosal physiology and its relationships with gut microbiota, this study aimed to evaluate the ability of the lysate from the multi-strain probiotic formulation SLAB51 to affect the HIF pathway in a model of in vitro human intestinal epithelium (intestinal epithelial cells, IECs) and to protect from lipopolysaccharide (LPS) challenge. The exposure of IECs to SLAB51 lysate under normoxic conditions led to a dose-dependent increase in HIF-1α protein levels, which was associated with higher glycolytic metabolism and L-lactate production. Probiotic lysate significantly reduced PHD2 levels and HIF-1α hydroxylation, thus leading to HIF-1α stabilization. The ability of SLAB51 lysate to increase HIF-1α levels was also associated with the activation of the PI3K/AKT pathway and with the inhibition of NF-κB, nitric oxide synthase 2 (NOS2), and IL-1β increase elicited by LPS treatment. Our results suggest that the probiotic treatment, by stabilizing HIF-1α, can protect from an LPS-induced inflammatory response through a mechanism involving PI3K/AKT signaling.
Collapse
Affiliation(s)
- Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | | | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Serena Artone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Jenna Marie Sheldon
- Dr. Kiran C Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314-7796, USA
| | - Giovanni Latella
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
20
|
Comparative Metagenomics and Metabolomes Reveals Abnormal Metabolism Activity Is Associated with Gut Microbiota in Alzheimer's Disease Mice. Int J Mol Sci 2022; 23:ijms231911560. [PMID: 36232865 PMCID: PMC9569518 DOI: 10.3390/ijms231911560] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
A common symptom in Alzheimer's disease (AD) is cognitive decline, of which the potential pathogenesis remains unclear. In order to understand the mechanism of gut microbiota in AD, it is necessary to clarify the relationship between gut microbiota and metabolites. Behavioral tests, pathological examination, metagenomics, and metabolomics were applied to analyze the difference of gut microbiota and metabolome between APPswe/PS1ΔE9 (PAP) mice with cognitive decline and age-matched controls, and their possible correlations. Our results showed that PAP mice and health mice had different structures of the bacterial communities in the gut. The abundances and diversities of the bacterial communities in health mice were higher than in PAP mice by metagenomics analysis. The abundances of Libanicoccus massiliensis, Paraprevotella clara, and Lactobacillus amylovorus were significantly increased in PAP mice, while the abundances of Turicibacter sanguinis, Dubosiella newyorkensis, and Prevotella oris were greatly reduced. Furthermore, PAP mice possessed peculiar metabolic phenotypes in stool, serum, and hippocampus relative to WT mice, as is demonstrated by alterations in neurotransmitters metabolism, lipid metabolism, aromatic amino acids metabolism, energy metabolism, vitamin digestion and absorption, and bile metabolism. Microbiota-host metabolic correlation analysis suggests that abnormal metabolism in stool, serum, and hippocampus of PAP mice may be modulated by the gut microbiota, especially T. sanguinis, D. newyorkensis, and P. oris. Therefore, abnormal metabolism activity is associated with gut microbiota in Alzheimer's disease mice. Our results imply that modifying host metabolism through targeting gut microbiota may be a novel and viable strategy for the prevention and treatment of AD in the future.
Collapse
|
21
|
Effects of Compound Mycotoxin Detoxifier on Alleviating Aflatoxin B 1-Induced Inflammatory Responses in Intestine, Liver and Kidney of Broilers. Toxins (Basel) 2022; 14:toxins14100665. [PMID: 36287934 PMCID: PMC9609892 DOI: 10.3390/toxins14100665] [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: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
In order to alleviate the toxic effects of aflatoxins B1 (AFB1) on inflammatory responses in the intestine, liver, and kidney of broilers, the aflatoxin B1-degrading enzyme, montmorillonite, and compound probiotics were selected and combined to make a triple-action compound mycotoxin detoxifier (CMD). The feeding experiment was divided into two stages. In the early feeding stage (1−21 day), a total of 200 one-day-old Ross broilers were randomly divided into four groups; in the later feeding stage (22−42 day), 160 broilers aged at 22 days were assigned to four groups: Group A: basal diet (4.31 μg/kg AFB1); Group B: basal diet with 40 μg/kg AFB1; Group C: Group A plus 1.5 g/kg CMD; Group D: Group B plus 1.5 g/kg CMD. After the feeding experiment, the intestine, liver, and kidney tissues of the broilers were selected to investigate the molecular mechanism for CMD to alleviate the tissue damages. Analyses of mRNA abundances and western blotting (WB) of inflammatory factors, as well as immunohistochemical (IHC) staining of intestine, liver, and kidney tissues showed that AFB1 aggravated the inflammatory responses through NF-κB and TN-α signaling pathways via TLR pattern receptors, while the addition of CMD significantly inhibited the inflammatory responses. Phylogenetic investigation showed that AFB1 significantly increased interleukin-1 receptor-associated kinase (IRAK-1) and mitogen-activated protein kinase (MAPK) activities (p < 0.05), which were restored to normal levels by CMD addition, indicating that CMD could alleviate cell inflammatory damages induced by AFB1.
Collapse
|
22
|
Smith EM, Grassel CL, Papadimas A, Foulke-Abel J, Barry EM. The role of CFA/I in adherence and toxin delivery by ETEC expressing multiple colonization factors in the human enteroid model. PLoS Negl Trop Dis 2022; 16:e0010638. [PMID: 35881640 PMCID: PMC9355178 DOI: 10.1371/journal.pntd.0010638] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/05/2022] [Accepted: 07/07/2022] [Indexed: 01/10/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a primary causative agent of diarrhea in travelers and young children in low-to-middle-income countries (LMICs). ETEC adhere to intestinal epithelia via colonization factors (CFs) and secrete heat-stable toxin (ST) and/or heat-labile toxin (LT), causing dysregulated cellular ion transport and water secretion. ETEC isolates often harbor genes encoding more than one CF that are targets as vaccine antigens. CFA/I is a major CF that is associated with ETEC that causes moderate-to-severe diarrhea and plays an important role in pathogenesis. The Global Enteric Multicenter Study finding that 78% of CFA/I-expressing ETEC also encode the minor CF CS21 prompted investigation of the combined role of these two CFs. Western blots and electron microscopy demonstrated growth media-dependent and strain-dependent differences in CFA/I and CS21 expression. The critical role of CFA/I in adherence by ETEC strains expressing CFA/I and CS21 was demonstrated using the human enteroid model and a series of CFA/I- and CS21-specific mutants. Furthermore, only anti-CFA/I antibodies inhibited adherence by global ETEC isolates expressing CFA/I and CS21. Delivery of ST and resulting cGMP secretion was measured in supernatants from infected enteroid monolayers, and strain-specific ST delivery and time-dependent cGMP production was observed. Interestingly, cGMP levels were similar across wildtype and CF-deficient strains, reflecting a limitation of this static aerobic infection model. Despite adherence by ETEC and delivery of ST, the enteroid monolayer integrity was not disrupted, as shown by the lack of decrease in transepithelial electrical resistance and the lack of IL-8 cytokines produced during infection. Taken together, these data demonstrate that targeting CFA/I in global clinical CFA/I-CS21 strains is sufficient for adherence inhibition, supporting a vaccine strategy that focuses on blocking major CFs. In addition, the human enteroid model has significant utility for the study of ETEC pathogenesis and evaluation of vaccine-induced functional antibody responses.
Collapse
Affiliation(s)
- Emily M. Smith
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Christen L. Grassel
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Antonia Papadimas
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jennifer Foulke-Abel
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Eileen M. Barry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
23
|
Wu S, Liu M, Chen H, Song Q, Wu Z, Dai Z. Tryptophan regulates bile and nitrogen metabolism in two pig gut lactobacilli species in vitro based on metabolomics study. Amino Acids 2022; 54:1421-1435. [PMID: 35838843 DOI: 10.1007/s00726-022-03179-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 06/06/2022] [Indexed: 02/07/2023]
Abstract
Research has demonstrated that tryptophan (Trp) regulated the composition and metabolism of the gut microbiota. However, the detailed mode of action of Trp on the metabolism of intestinal commensal lactobacilli has not been well characterized. This study aimed to compare the effects of Trp concentration (0.2, 0.4, 0.6 mmol/L) in the media on the metabolism of Lactobacillus amylovorus and Limosilactobacillus mucosae isolated from the small intestine of piglets in vitro by high-performance liquid chromatography and metabolomics study. Results showed that increased Trp concentration increased (P < 0.05) net utilization of lysine, methionine, tryptophan, asparagine/aspartate, glutamine/glutamate, however, increased net production of glycine and taurine in Lac. amylovorus. In contrast, increased Trp concentration decreased (P < 0.05) net utilization of leucine, phenylalanine, and serine and increased (P < 0.05) net utilization of arginine and net production of ornithine and glycine in Lim. mucosae. Targeted metabolomics analysis showed that increased Trp concentration promoted (P < 0.05) the production of indole-3-lactic acid and 3-indoleacetic acid in the two lactobacilli strains. Increased concentration of Trp increased (P < 0.01) glycochenodeoxycholic acid metabolism in Lim. mucosae and glycocholic acid and taurocholic acid metabolism in Lac. amylovorus. Untargeted metabolomics analysis showed that metabolic pathways related to phenylalanine and tryptophan metabolism, and nicotinate and nicotinamide metabolism were regulated by Trp in Lim. mucosae. These findings will help develop new biomarkers and dietary strategies to maintain the functionality of the gut microbiota aiming at improving the nutrition and health of both humans and animals.
Collapse
Affiliation(s)
- Shizhe Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Moyan Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hui Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Qingqing Song
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
24
|
Amin H, Šantl-Temkiv T, Cramer C, Vestergaard DV, Holst GJ, Elholm G, Finster K, Bertelsen RJ, Schlünssen V, Sigsgaard T, Marshall IPG. Cow Farmers’ Homes Host More Diverse Airborne Bacterial Communities Than Pig Farmers’ Homes and Suburban Homes. Front Microbiol 2022; 13:883991. [PMID: 35847077 PMCID: PMC9278274 DOI: 10.3389/fmicb.2022.883991] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/05/2022] [Indexed: 01/04/2023] Open
Abstract
Living on a farm has been linked to a lower risk of immunoregulatory disorders, such as asthma, allergy, and inflammatory bowel disease. It is hypothesized that a decrease in the diversity and composition of indoor microbial communities is a sensible explanation for the upsurge in immunoregulatory diseases, with airborne bacteria contributing to this protective effect. However, the composition of this potentially beneficial microbial community in various farm and suburban indoor environments is still to be characterized. We collected settled airborne dust from stables and the associated farmers’ homes and from suburban homes using electrostatic dust collectors (EDCs) over a period of 14 days. Then, quantitative PCR (qPCR) was used to assess bacterial abundance. The V3–V4 region of the bacterial 16S rRNA gene was amplified and sequenced using Ilumina MiSeq in order to assess microbial diversity. The Divisive Amplicon Denoising Algorithm (DADA2) algorithm was used for the inference of amplicon sequence variants from amplicon data. Airborne bacteria were significantly more abundant in farmers’ indoor environments than in suburban homes (p < 0.001). Cow farmers’ homes had significantly higher bacterial diversity than pig farmers’ and suburban homes (p < 0.001). Bacterial taxa, such as Firmicutes, Prevotellaceae, Lachnospiraceae, and Lactobacillus were significantly more abundant in farmers’ homes than suburban homes, and the same was true for beneficial intestinal bacterial species, such as Lactobacillus amylovorus, Eubacterium hallii, and Faecalibacterium prausnitzii. Furthermore, we found a higher similarity between bacterial communities in individual farmers’ homes and their associated cow stables than for pig stables. Our findings contribute with important knowledge on bacterial composition, abundance, and diversity in different environments, which is highly valuable in the discussion on how microbial exposure may contribute to the development of immune-mediated diseases in both children and adults.
Collapse
Affiliation(s)
- Hesham Amin
- Department of Clinical Science, University of Bergen, Bergen, Norway
- *Correspondence: Hesham Amin,
| | - Tina Šantl-Temkiv
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus, Denmark
| | - Christine Cramer
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
- Department of Occupational Medicine, Danish Ramazzini Center, Aarhus University Hospital, Aarhus, Denmark
| | - Ditte V. Vestergaard
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus, Denmark
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
| | - Gitte J. Holst
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
| | - Grethe Elholm
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
| | - Kai Finster
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus, Denmark
| | | | - Vivi Schlünssen
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
- The National Research Center for the Working Environment, Copenhagen, Denmark
| | - Torben Sigsgaard
- Department of Public Health, Environment, Work, and Health, Danish Ramazzini Center, Aarhus University, Aarhus, Denmark
| | - Ian P. G. Marshall
- Section for Microbiology, Department of Biology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
25
|
Ram AK, Vairappan B, Srinivas BH. Nimbolide attenuates gut dysbiosis and prevents bacterial translocation by improving intestinal barrier integrity and ameliorating inflammation in hepatocellular carcinoma. Phytother Res 2022; 36:2143-2160. [PMID: 35229912 DOI: 10.1002/ptr.7434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/04/2022] [Accepted: 02/16/2022] [Indexed: 09/25/2023]
Abstract
Gut microbiota imbalance plays a key pathological role in hepatocellular carcinoma (HCC) progression; however, the mechanism is poorly understood. We previously showed nimbolide impede tumor development by improving hepatic tight junction (TJ) proteins expression and attenuating inflammation in HCC mice. Here, we aimed to study the role of nimbolide in regulating gut microbiota imbalance and bacterial translocation (BT) through modulating intestinal TJ proteins in an experimental hepatocarcinogenesis. Nimbolide (6 mg/kg) was administered orally for 4 weeks following induction of HCC in mice at the 28th week. Nimbolide treatment attenuated the gut microbiota imbalance by decreasing 16 s rRNA levels of Escherichia coli, Enterococcus, Bacteroides and increasing Bifidobacterium, and Lactobacillus in the intestinal tissue, which was otherwise altered in HCC mice. Furthermore, nimbolide improved intestinal barrier integrity in HCC mice by upregulating TJ proteins such as occludin and ZO-1 expression and subsequently prevented hepatic BT and decreased BT markers such as LBP, sCD14, and procalcitonin in the plasma of HCC mice. Moreover, nimbolide ameliorated intestinal and hepatic inflammation by downregulating TLR4, MyD88, and NF-κB protein expression in HCC mice. Thus, nimbolide represents a novel therapeutic drug for HCC treatment by targeting the gut-liver axis, which plays an imperative role in HCC pathogenesis.
Collapse
Affiliation(s)
- Amit Kumar Ram
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Balasubramaniyan Vairappan
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Bheemanathi Hanuman Srinivas
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| |
Collapse
|
26
|
Roselli M, Maruszak A, Grimaldi R, Harthoorn L, Finamore A. Galactooligosaccharide Treatment Alleviates DSS-Induced Colonic Inflammation in Caco-2 Cell Model. Front Nutr 2022; 9:862974. [PMID: 35495925 PMCID: PMC9047546 DOI: 10.3389/fnut.2022.862974] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/04/2022] [Indexed: 12/19/2022] Open
Abstract
The biological activities of dietary bioactive polysaccharides have been largely explored. Studies on the immunomodulating effects of oligosaccharides and polysaccharides have shown that they are able to modulate innate immunity. Prebiotics are a class of poorly digested carbohydrates that are mainly produced from dietary fibers, which are carbohydrate polymers with ten or more monomeric units as defined by the Codex Alimentarius Commission in 2009. Considering the capacity of prebiotics in reducing gut inflammation, the aim of this study was to investigate the anti-inflammatory activity of galactooligosaccharide (Bimuno® GOS) in an in vitro model of ulcerative colitis (UC)-like inflamed intestinal cells. Differentiated Caco-2 cells were exposed to 2 % dextran-sulfate-sodium salt (DSS) to induce inflammation, and then with different concentrations of Bimuno GOS (1-1,000 μg/ml). Cell monolayer permeability, tight- and adherent junction protein distribution, pro-inflammatory cytokine secretion, and NF-kB cascade were assessed. Bimuno GOS at different concentrations, while not affecting cell monolayer permeability, was shown to counteract UC-like intestinal inflammatory responses and damages induced by DSS. Indeed, Bimuno GOS was able to counteract the detrimental effects of DSS on cell permeability, determined by transepithelial electrical resistance, phenol red apparent permeability, and tight- and adherent junction protein distribution. Furthermore, Bimuno GOS inhibited the DSS-induced NF-kB nuclear translocation and pro-inflammatory cytokine secretion. Further analyses showed that Bimuno GOS was able to revert the expression levels of most of the proteins involved in the NF-kB cascade to control levels. Thus, the prebiotic Bimuno GOS can be a safe and effective way to modulate the gut inflammatory state through NF-kB pathway modulation, and could possibly further improve efficacy in inducing remission of UC.
Collapse
Affiliation(s)
- Marianna Roselli
- Research Centre for Food and Nutrition, CREA (Consiglio per la ricerca in agricoltura e l'analisi dell'economia agraria), Rome, Italy
| | | | | | | | - Alberto Finamore
- Research Centre for Food and Nutrition, CREA (Consiglio per la ricerca in agricoltura e l'analisi dell'economia agraria), Rome, Italy
| |
Collapse
|
27
|
Luo Y, Liu Y, Li H, Zhao Y, Wright ADG, Cai J, Tian G, Mao X. Differential Effect of Dietary Fibers in Intestinal Health of Growing Pigs: Outcomes in the Gut Microbiota and Immune-Related Indexes. Front Microbiol 2022; 13:843045. [PMID: 35273589 PMCID: PMC8902361 DOI: 10.3389/fmicb.2022.843045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
Although dietary fibers (DFs) have been shown to improve intestinal health in pigs, it is unclear whether this improvement varies according to the type/source of DF. In the current study, we investigated the impact of dietary supplement (15%) of pea-hull fiber (PF), oat bran (OB), and their mixture (MIX, PF, and OB each accounted for 7.5%) in the growth performance as well as intestinal barrier and immunity-related indexes in growing pigs. Twenty-four cross-bred pigs (32.42 ± 1.95 kg) were divided into four groups: CON (basal diet with no additional DF), PF, OB, and MIX. After 56 days of feeding, we found that the growth performance of PF pigs was decreased (p < 0.05) compared with pigs in other groups. Results of real-time polymerase chain reaction and Western blot showed that the improvement of immune-related indexes (e.g., interleukin 10 [IL-10]) in OB and MIX pigs mainly presented in the ileum, whereas the improvement of intestinal barrier–related indexes (e.g., MUC1 and MUC2) mainly presented in the colon. Whether in the ileum or colon, such improvement of immune function may be dependent on NOD rather than TLR-associated pathways. Amplicon sequencing results showed that PF and MIX pigs shared a similar bacterial community, such as lower abundance of ileal Clostridiaceae and colonic Streptoccocus than that of CON pigs (p < 0.05). Our results indicate that OB and MIX, rather than PF, benefit the intestinal health in growing pigs, and multiple-sourced DF may reduce the adverse effect of single-soured DF on the growth performance and gut microbiota in pigs.
Collapse
Affiliation(s)
- Yuheng Luo
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition, Sichuan Agricultural University, Chengdu, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yang Liu
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition, Sichuan Agricultural University, Chengdu, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Hua Li
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition, Sichuan Agricultural University, Chengdu, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yao Zhao
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition, Sichuan Agricultural University, Chengdu, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | | | - Jingyi Cai
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition, Sichuan Agricultural University, Chengdu, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Gang Tian
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition, Sichuan Agricultural University, Chengdu, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xiangbing Mao
- Key Laboratory of Animal Disease-Resistant Nutrition, Ministry of Education, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-Resistant Nutrition, Sichuan Agricultural University, Chengdu, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
28
|
Soltan MA, Behairy MY, Abdelkader MS, Albogami S, Fayad E, Eid RA, Darwish KM, Elhady SS, Lotfy AM, Alaa Eldeen M. In silico Designing of an Epitope-Based Vaccine Against Common E. coli Pathotypes. Front Med (Lausanne) 2022; 9:829467. [PMID: 35308494 PMCID: PMC8931290 DOI: 10.3389/fmed.2022.829467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/21/2022] [Indexed: 12/20/2022] Open
Abstract
Escherichia coli (E. coli) is a Gram-negative bacterium that belongs to the family Enterobacteriaceae. While E. coli can stay as an innocuous resident in the digestive tract, it can cause a group of symptoms ranging from diarrhea to live threatening complications. Due to the increased rate of antibiotic resistance worldwide, the development of an effective vaccine against E. coli pathotypes is a major health priority. In this study, a reverse vaccinology approach along with immunoinformatics has been applied for the detection of potential antigens to develop an effective vaccine. Based on our screening of 5,155 proteins, we identified lipopolysaccharide assembly protein (LptD) and outer membrane protein assembly factor (BamA) as vaccine candidates for the current study. The conservancy of these proteins in the main E. coli pathotypes was assessed through BLASTp to make sure that the designed vaccine will be protective against major E. coli pathotypes. The multitope vaccine was constructed using cytotoxic T lymphocyte (CTL), helper T lymphocyte (HTL), and B cell lymphocyte (BCL) epitopes with suitable linkers and adjuvant. Following that, it was analyzed computationally where it was found to be antigenic, soluble, stable, and non-allergen. Additionally, the adopted docking study, as well as all-atom molecular dynamics simulation, illustrated the promising predicted affinity and free binding energy of this constructed vaccine against the human Toll-like receptor-4 (hTLR-4) dimeric state. In this regard, wet lab studies are required to prove the efficacy of the potential vaccine construct that demonstrated promising results through computational validation.
Collapse
Affiliation(s)
- Mohamed A. Soltan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Ismailia, Egypt
| | - Mohammed Y. Behairy
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Mennatallah S. Abdelkader
- Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Eman Fayad
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Refaat A. Eid
- Department of Pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Khaled M. Darwish
- Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed M. Lotfy
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Muhammad Alaa Eldeen
- Division of Cell Biology, Histology and Genetics, Department of Zoology, Faculty of Science, Zagazig University, Zagazig, Egypt
| |
Collapse
|
29
|
Helmy YA, Kassem II, Rajashekara G. Immuno-modulatory effect of probiotic E. coli Nissle 1917 in polarized human colonic cells against Campylobacter jejuni infection. Gut Microbes 2022; 13:1-16. [PMID: 33382951 PMCID: PMC7781529 DOI: 10.1080/19490976.2020.1857514] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Campylobacter jejuni is among the leading causes of bacterial foodborne illness. Poultry is the major reservoir and source of human campylobacteriosis. Currently, there is no effective and practical method to decrease C. jejuni colonization in chickens or to reduce human infections. Additionally, antibiotic-resistant infections pose a serious public health concern; therefore, antibiotic-alternative approaches are needed to reduce transmission of C. jejuni including resistant bacteria from chickens to humans. Here, we evaluated the effect of E. coli Nissle 1917 (EcN) on innate responses of polarized HT-29 cells and consequently on C. jejuni 81176 infections in HT-29 cells. Pre-treatment of HT-29 cells with EcN for 4 h had a significant effect on the invasion of different C. jejuni strains (2 h post-infection) (P < .05) and no intracellular C. jejuni (24 h post-infection) were recovered. To further understand how EcN mediates its impact on C. jejuni's survival inside the cells, we used Human Antibacterial RT2 ProfilerTM PCR arrays to profile gene expression in HT-29 cells after treatment with EcN with or without C. jejuni 81-176 infection. Our results suggest that pre-treatment of the HT-29 cells with EcN induced the anti-inflammatory cytokines and activated the anti-apoptotic Akt signaling which likely to protect the cells against the proinflammatory and apoptosis responses induced by C. jejuni. EcN also positively affected the expression of genes involved in cellular maintenance, growth, development, and proliferation. Further, EcN modulated the expression of genes involved in protective innate immunity, such as TLRs, ERK1/2, p38 MAPK, Ap1, JNK, IL1B, IL17A, and NF-κB signaling.
Collapse
Affiliation(s)
- Yosra A. Helmy
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, the Ohio State University, Wooster, Ohio, USA
| | - Issmat I. Kassem
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, the Ohio State University, Wooster, Ohio, USA,College of Agricultural and Environmental Sciences, Center for Food Safety, University of Georgia, Griffin, Georgia, USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, the Ohio State University, Wooster, Ohio, USA,Corresponding author Gireesh Rajashekara Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH44691, USA
| |
Collapse
|
30
|
Kaur H, Ali SA. Probiotics and gut microbiota: mechanistic insights into gut immune homeostasis through TLR pathway regulation. Food Funct 2022; 13:7423-7447. [DOI: 10.1039/d2fo00911k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Consumption of probiotics as a useful functional food improves the host's wellbeing, and, when paired with prebiotics (indigestible dietary fibre/carbohydrate), often benefits the host through anaerobic fermentation.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-NDRI, 132001, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
31
|
Liu G, Gu K, Wang F, Jia G, Zhao H, Chen X, Wu C, Zhang R, Tian G, Cai J, Tang J, Wang J. Tryptophan Ameliorates Barrier Integrity and Alleviates the Inflammatory Response to Enterotoxigenic Escherichia coli K88 Through the CaSR/Rac1/PLC-γ1 Signaling Pathway in Porcine Intestinal Epithelial Cells. Front Immunol 2021; 12:748497. [PMID: 34745120 PMCID: PMC8566706 DOI: 10.3389/fimmu.2021.748497] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022] Open
Abstract
Background Impaired intestinal barrier integrity plays a crucial role in the development of many diseases such as obesity, inflammatory bowel disease, and type 2 diabetes. Thus, protecting the intestinal barrier from pathological disruption is of great significance. Tryptophan can increase gut barrier integrity, enhance intestinal absorption, and decrease intestinal inflammation. However, the mechanism of tryptophan in decreasing intestinal barrier damage and inflammatory response remains largely unknown. The objective of this study was to test the hypothesis that tryptophan can enhance intestinal epithelial barrier integrity and decrease inflammatory response mediated by the calcium-sensing receptor (CaSR)/Ras-related C3 botulinum toxin substrate 1 (Rac1)/phospholipase Cγ1 (PLC-γ1) signaling pathway. Methods IPEC-J2 cells were treated with or without enterotoxigenic Escherichia coli (ETEC) K88 in the absence or presence of tryptophan, CaSR inhibitor (NPS-2143), wild-type CaSR overexpression (pcDNA3.1-CaSR-WT), Rac1-siRNA, and PLC-γ1-siRNA. Results The results showed that ETEC K88 decreased the protein concentration of occludin, zonula occludens-1 (ZO-1), claudin-1, CaSR, total Rac1, Rho family member 1 of porcine GTP-binding protein (GTP-rac1), phosphorylated phospholipase Cγ1 (p-PLC-γ1), and inositol triphosphate (IP3); suppressed the transepithelial electrical resistance (TEER); and enhanced the permeability of FITC-dextran compared with the control group. Compared with the control group, 0.7 mM tryptophan increased the protein concentration of CaSR, total Rac1, GTP-rac1, p-PLC-γ1, ZO-1, claudin-1, occludin, and IP3; elevated the TEER; and decreased the permeability of FITC-dextran and contents of interleukin-8 (IL-8) and TNF-α. However, 0.7 mM tryptophan+ETEC K88 reversed the effects induced by 0.7 mM tryptophan alone. Rac1-siRNA+tryptophan+ETEC K88 or PLC-γ1-siRNA+tryptophan+ETEC K88 reduced the TEER, increased the permeability of FITC-dextran, and improved the contents of IL-8 and TNF-α compared with tryptophan+ETEC K88. NPS2143+tryptophan+ETEC K88 decreased the TEER and the protein concentration of CaSR, total Rac1, GTP-rac1, p-PLC-γ1, ZO-1, claudin-1, occludin, and IP3; increased the permeability of FITC-dextran; and improved the contents of IL-8 and TNF-α compared with tryptophan+ETEC K88. pcDNA3.1-CaSR-WT+Rac1-siRNA+ETEC K88 and pcDNA3.1-CaSR-WT+PLC-γ1-siRNA+ETEC K88 decreased the TEER and enhanced the permeability in porcine intestine epithelial cells compared with pcDNA3.1-CaSR-WT+ETEC K88. Conclusion Tryptophan can improve intestinal epithelial barrier integrity and decrease inflammatory response through the CaSR/Rac1/PLC-γ1 signaling pathway.
Collapse
Affiliation(s)
- Guangmang Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Ke Gu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Fang Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Gang Jia
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Hua Zhao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Caimei Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Ruinan Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Gang Tian
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Jingyi Cai
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Jiayong Tang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, China
- Key Laboratory of Animal Disease-Resistant Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Chengdu, China
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
32
|
Effects of Live and Pasteurized Forms of Akkermansia from the Human Gut on Obesity and Metabolic Dysregulation. Microorganisms 2021; 9:microorganisms9102039. [PMID: 34683361 PMCID: PMC8538271 DOI: 10.3390/microorganisms9102039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/14/2021] [Accepted: 09/18/2021] [Indexed: 01/08/2023] Open
Abstract
Akkermansia muciniphila (A. muciniphila) is a promising probiotic candidate owing to its health-promoting properties. A previous study reported that the pasteurized form of A. muciniphila strains isolated from human stool samples had a beneficial impact on high-fat diet-induced obese mice. On the other hand, the differences in the probiotic effects between live and pasteurized A. muciniphila on the metabolism and immune system of the host are still inconclusive. This study examines the differences between the live and pasteurized forms of A. muciniphila strains on the lipid and glucose metabolism and on regulating the inflammatory immune responses using a HFD-fed obese mouse model. The animals were administered the live and pasteurized forms of two A. muciniphila strains five times per week for the entire study period of 12 weeks. Both forms of the bacterial strains improved the HFD-induced obesity and metabolic dysregulation in the mice by preventing body-weight gains after one week. In addition, they cause a decrease in the weights of the major adipose tissues, adipogenesis/lipogenesis and serum TC levels, improvement in glucose homeostasis and suppression of inflammatory insults. Furthermore, these treatments restored the damaged gut architecture and integrity and improved the hepatic structure and function in HFD-induced animals. On the other hand, for both bacterial strains, the pasteurized form was more potent in improving glucose tolerance than the live form. Moreover, specific A. muciniphila preparations with either live or pasteurized bacteria decreased the number and population (%) of splenic Treg cells (CD4+ Foxp3+) significantly in the HFD-fed animals, further supporting the anti-inflammatory properties of these bacteria.
Collapse
|
33
|
Gupta T, Kaur H, Kapila S, Kapila R. Potential probiotic Lacticaseibacillus rhamnosus MTCC-5897 attenuates Escherichia coli induced inflammatory response in intestinal cells. Arch Microbiol 2021; 203:5703-5713. [PMID: 34476513 DOI: 10.1007/s00203-021-02541-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/31/2021] [Accepted: 08/18/2021] [Indexed: 12/31/2022]
Abstract
Probiotics are microbes having tremendous potential to prevent gastrointestinal disorders. In current investigation, immunomodulatory action of probiotic Lacticaseibacillus rhamnosus MTCC-5897 was studied during exclusion, competition and displacement of Escherichia coli on intestinal epithelial (Caco-2) cells. The incubation of intestinal cells with Escherichia coli, enhanced downstream signalling and activated nuclear factor kappa B (NF-κB). This significantly increased (p < 0.01) the pro-inflammatory cytokines (IL-8, TNF-α, IFN-ϒ) expression. While, incubation of epithelial cells with Lacticaseibacillus rhamnosus during exclusion and competition with Escherichia coli, counteracted these enhanced expressions. The immunomodulatory feature of Lacticaseibacillus rhamnosus was also highlighted with increased (p < 0.05) transcription of toll-like receptor-2 (TLR-2) and single Ig IL-1-related receptor (SIGIRR) along with diminished expression of TLR-4. Likewise, attenuation (p < 0.05) of E. coli-mediated enhanced nuclear translocation of NF-κB p-65 subunit by Lacticaseibacillus rhamnosus during exclusion was confirmed with western blotting. Thus, present finding establishes the prophylactic potential of Lacticaseibacillus rhamnosus against exclusion of Escherichia coli in intestinal cells.
Collapse
Affiliation(s)
- Taruna Gupta
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Harpreet Kaur
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Suman Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Rajeev Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India.
| |
Collapse
|
34
|
Vadopalas L, Zokaityte E, Zavistanaviciute P, Gruzauskas R, Starkute V, Mockus E, Klementaviciute J, Ruzauskas M, Lele V, Cernauskas D, Klupsaite D, Dauksiene A, Sederevicius A, Badaras S, Bartkiene E. Supplement Based on Fermented Milk Permeate for Feeding Newborn Calves: Influence on Blood, Growth Performance, and Faecal Parameters, including Microbiota, Volatile Compounds, and Fatty and Organic Acid Profiles. Animals (Basel) 2021; 11:ani11092544. [PMID: 34573514 PMCID: PMC8466287 DOI: 10.3390/ani11092544] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/17/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Gastrointestinal infections and diarrhoea are the main health issues in young calves. The application of microbial products containing probiotics and prebiotics may lead to better management of the gut microbiome and improved calf health. After fermentation with selected lactic acid bacterial strains, milk permeate (a dairy industry by-product) contains lactic acid bacteria and prebiotics, both of which possess viable antimicrobial properties. We hypothesised that fermented milk permeate could be a prospective feed supplement for newborn calves. A 14-day experiment was conducted in which a group of newborn calves were given a supplement of milk permeate fermented with Lactobacillus uvarum LUHS245. A significantly higher count of lactic acid bacteria, a lower total count of enterobacteria, a higher species variety, and greater concentrations of both propionic acid and dry matter were found in the faeces of the calves fed with fermented milk permeate compared with a control group. Most of the fatty acids and volatile compounds in the faeces differed significantly between the two groups. The results suggest that supplementing the calves’ feed with fermented milk permeate has a positive effect on certain health parameters but no influence on blood parameters and growth performance. Abstract The aim of this study was to assess the effect of a feed supplement, namely milk permeate (MP) fermented with Lactobacillus uvarum LUHS245, on the newborn calves’ growth performance and blood and faecal parameters, including microbiota and volatile compound and fatty acid profiles. Ten female Holstein calves in the control group (CON group) were fed with a standard milk replacer diet and colostrum only, from day 2 to 14 of life, while 10 calves of the treated group (MP group) were fed with the same diet supplemented with 50 mL of the fermented MP. After 14 days, there were no significant differences between the groups in blood parameters, growth performance, or faecal pH. There was a significantly higher percentage of live lactic acid bacteria (by 17.02%), a lower percentage of enterobacteria (by 10.38%), a higher overall number of probiotic bacteria, a 1.7-fold higher species variety, and a higher content of dry matter in the faeces of the MP group (p < 0.05). The fatty acid and volatile compound profiles differed significantly between the groups. The results suggest that supplementing calves’ feed with fermented milk permeate has a positive effect on certain health parameters but not on blood parameters or growth performance.
Collapse
Affiliation(s)
- Laurynas Vadopalas
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
| | - Egle Zokaityte
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary Medicine, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania
| | - Paulina Zavistanaviciute
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary Medicine, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania
| | - Romas Gruzauskas
- Department of Food Science and Technology, Kaunas University of Technology, Radvilenu Rd. 19, LT-50254 Kaunas, Lithuania;
| | - Vytaute Starkute
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary Medicine, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania
| | - Ernestas Mockus
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
| | - Jolita Klementaviciute
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
| | - Modestas Ruzauskas
- Faculty of Veterinary, Institute of Microbiology and Virology, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania;
- Department of Anatomy and Physiology, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania;
| | - Vita Lele
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary Medicine, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania
| | - Darius Cernauskas
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
| | - Dovile Klupsaite
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
| | - Agila Dauksiene
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
- Department of Anatomy and Physiology, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania;
| | - Antanas Sederevicius
- Department of Anatomy and Physiology, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania;
| | - Sarunas Badaras
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
| | - Elena Bartkiene
- Faculty of Animal Sciences, Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania; (L.V.); (E.Z.); (P.Z.); (V.S.); (E.M.); (J.K.); (V.L.); (D.C.); (D.K.); (A.D.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary Medicine, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania
- Correspondence: ; Tel.: +370-601-35837
| |
Collapse
|
35
|
Yang B, Huang Z, He Z, Yue Y, Zhou Y, Ross RP, Stanton C, Zhang H, Zhao J, Chen W. Protective effect of Bifidobacterium bifidum FSDJN7O5 and Bifidobacterium breve FHNFQ23M3 on diarrhea caused by enterotoxigenic Escherichia coli. Food Funct 2021; 12:7271-7282. [PMID: 34165468 DOI: 10.1039/d1fo00504a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the main bacterial cause of diarrhea among children in developing countries and of traveler's diarrhea. In this study, a mouse model was used to evaluate the effect of Bifidobacterium on alleviating diarrhea caused by ETEC. The results showed that B. breve FHNFQ23M3 and B. bifidum FSDJN7O5 could relieve the symptoms of diarrhea. Both strains significantly reduced the stool water content, restored the villi structure in the jejunum and ameliorated the fecal short-chain fatty acid (SCFA) content. In addition, B. breve FHNFQ23M3 restored body weight to the level before ETEC challenge and significantly reduced interferon-γ (IFN-γ), while B. bifidum FSDJN7O5 significantly improved interleukin (IL)-10. Furthermore, all the Bifidobacterium strains used in this study could significantly downregulate tumor necrosis factor-α (TNF-α) and restore the unbalanced gut microbiota, which had a high content of pathogenic Escherichia-Shigella and low content of Blautia and Clostridium innocuum groups due to ETEC. All the results proved that Bifidobacterium could be a potential probiotic for alleviating diarrhea from ETEC infection.
Collapse
Affiliation(s)
- Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Gresse R, Chaucheyras-Durand F, Garrido JJ, Denis S, Jiménez-Marín A, Beaumont M, Van de Wiele T, Forano E, Blanquet-Diot S. Pathogen Challenge and Dietary Shift Alter Microbiota Composition and Activity in a Mucin-Associated in vitro Model of the Piglet Colon (MPigut-IVM) Simulating Weaning Transition. Front Microbiol 2021; 12:703421. [PMID: 34349744 PMCID: PMC8328230 DOI: 10.3389/fmicb.2021.703421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the principal pathogen responsible for post-weaning diarrhea in newly weaned piglets. Expansion of ETEC at weaning is thought to be the consequence of various stress factors such as transient anorexia, dietary change or increase in intestinal inflammation and permeability, but the exact mechanisms remain to be elucidated. As the use of animal experiments raise more and more ethical concerns, we used a recently developed in vitro model of piglet colonic microbiome and mucobiome, the MPigut-IVM, to evaluate the effects of a simulated weaning transition and pathogen challenge at weaning. Our data suggested that the tested factors impacted the composition and functionality of the MPigut-IVM microbiota. The simulation of weaning transition led to an increase in relative abundance of the Prevotellaceae family which was further promoted by the presence of the ETEC strain. In contrast, several beneficial families such as Bacteroidiaceae or Ruminococcaceae and gut health related short chain fatty acids like butyrate or acetate were reduced upon simulated weaning. Moreover, the incubation of MPigut-IVM filtrated effluents with porcine intestinal cell cultures showed that ETEC challenge in the in vitro model led to an increased expression of pro-inflammatory genes by the porcine cells. This study provides insights about the etiology of a dysbiotic microbiota in post-weaning piglets.
Collapse
Affiliation(s)
- Raphaële Gresse
- INRAE, UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France.,Lallemand SAS, Blagnac, France
| | | | - Juan J Garrido
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain
| | - Sylvain Denis
- INRAE, UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Angeles Jiménez-Marín
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain
| | - Martin Beaumont
- GenPhySE, INRAE, ENVT, Université de Toulouse, Castanet-Tolosan, France
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Evelyne Forano
- INRAE, UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France
| | | |
Collapse
|
37
|
Liu L, Li Y, He Y, Wang Z, Zhao H, Jin X, Shi D, Wang X. Enterococcus faecium HDRsEf1 inhibits LPS-induced downregulation of ZO-1 expression via TLR2/4-mediated JNK/AP-1 signalling pathways. J Appl Microbiol 2021; 132:605-617. [PMID: 34062034 DOI: 10.1111/jam.15167] [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: 03/14/2021] [Revised: 05/14/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022]
Abstract
AIMS ZO-1 is a key regulatory tight junction protein that plays an important role in maintaining gastrointestinal health. In this study, we investigated the protective effect and regulation mechanism of the probiotic Enterococcus faecium HDRsEf1 on tight junction protein ZO-1 at the cellular and molecular levels. METHODS AND RESULTS We established LPS-induced intestinal epithelial cell injury model, and detected the protective effect of HDRsEf1 on ZO-1 in IPEC-J2 cells by Real-time PCR and Western blot. The results showed that HDRsEf1 inhibited the downregulation of ZO-1 expression induced by LPS. HDRsEf1 stabilized the destruction of the ZO-1 structure caused by LPS in an immunofluorescence assay. Through gene overexpression and siRNA interference tests, we found that transcription factor AP-1 inhibited the level of ZO-1 expression. Silencing experiment further supported that the protective effect of HDRSEF1 might mediated by suppression of LPS-provoked activation of ASK1/MKK7/JNK signalling pathways. In addition, HDRsEf1 could stabilize ZO-1 expression by increasing TLR2 expression and competing with LPS for the TLR4 binding site. More interestingly, we also found that HDRsEf1 could stabilize ZO-1 expression through inhibiting the production of TNF-α induced by LPS. CONCLUSIONS HDRsEf1 could protect the IPEC-J2 cell against LPS induced down-regulation of ZO-1 expression by inhibiting the activation of TLR2/4-mediated JNK-AP-1 and signalling cascade and the production of TNF-α. SIGNIFICANCE AND IMPACT OF THE STUDY This study can provide a theoretical basis for probiotics to regulate the expression of intestinal tight junction proteins, and supply technical support for probiotics to prevent and treat animal intestinal infectious diseases.
Collapse
Affiliation(s)
- Lin Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Hubei Provincial Institute of Veterinary Drug Control, Wuhan, 430068, Hubei, China
| | - Yue Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Hubei Provincial Institute of Veterinary Drug Control, Wuhan, 430068, Hubei, China
| | - Yucheng He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Hubei Provincial Institute of Veterinary Drug Control, Wuhan, 430068, Hubei, China
| | - Zhaoyang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Hubei Provincial Institute of Veterinary Drug Control, Wuhan, 430068, Hubei, China
| | - Hongze Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Hubei Provincial Institute of Veterinary Drug Control, Wuhan, 430068, Hubei, China
| | - Xiue Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Hubei Provincial Institute of Veterinary Drug Control, Wuhan, 430068, Hubei, China
| | - Deshi Shi
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Hubei Provincial Institute of Veterinary Drug Control, Wuhan, 430068, Hubei, China
| | - Xiliang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Hubei Provincial Institute of Veterinary Drug Control, Wuhan, 430068, Hubei, China
| |
Collapse
|
38
|
Gupta T, Kaur H, Kapila S, Kapila R. Lactobacillus fermentum (MTCC-5898) alleviates Escherichia coli-induced inflammatory responses in intestinal epithelial cells by modulating immune genes and NF-κB signalling. J Appl Microbiol 2021; 131:3008-3017. [PMID: 33999475 DOI: 10.1111/jam.15153] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023]
Abstract
AIM Dietary intervention using probiotic bacteria has emerged as a promising preventive strategy in addressing foodborne infections or gastrointestinal disorders. This study investigated the immunomodulatory effects of Lactobacillus fermentum (MTCC-5898) on Escherichia coli-induced inflammatory responses in intestinal epithelial cells. METHODS AND RESULTS The immune response of intestinal cells (Caco-2) in the presence of probiotic Lact. fermentum was determined during exclusion, competition and displacement of E. coli as the inflammatory agent. To achieve this objective, transcriptional modulation of key immune-related genes (cytokines, pattern recognition receptors and NF-κB), release of cytokines and nuclear translocation of the NF-κB subunit p-65 were studied. Expression of pro-inflammatory cytokines IL-8, TNF-α, IFN-ϒ and IL-23 was high in E. coli-exposed intestinal cells. However, overexpression of these E. coli-induced pro-inflammatory cytokines was prevented by Lact. fermentum during exclusion and competition assays. It also modulated the transcriptional expression of regulatory cytokines (IL-10 and TGF-β), pattern recognition receptors (TLR-2 and TLR-4) and genes associated with master inflammatory regulators (NF-κB and SIGIRR) to reduce E. coli-induced inflammation. The protective effect of Lact. fermentum was further confirmed by suppression of nuclear translocation of cytoplasmic NF-κB subunit (p-65). CONCLUSION Lactobacillus fermentum alleviated E. coli-induced inflammatory responses by modulating the NF-κB signalling besides pro-inflammatory and regulatory cytokines expression. SIGNIFICANCE AND IMPACT OF THE STUDY Lactobacillus fermentum holds significant promise as a potent nutraceutical that prevents and manages inflammatory gut-associated dysfunctions.
Collapse
Affiliation(s)
- T Gupta
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, India
| | - H Kaur
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, India
| | - S Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, India
| | - R Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
39
|
Li X, Hu D, Tian Y, Song Y, Hou Y, Sun L, Zhang Y, Man C, Zhang W, Jiang Y. Protective effects of a novel Lactobacillus rhamnosus strain with probiotic characteristics against lipopolysaccharide-induced intestinal inflammation in vitro and in vivo. Food Funct 2021; 11:5799-5814. [PMID: 32568317 DOI: 10.1039/d0fo00308e] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lipopolysaccharides (LPS), a main component of the Gram-negative bacterial cell wall, can damage the epithelial wall barrier and induce chronic intestinal inflammation. The purpose of this study is to evaluate whether the novel L. rhamnosus could alleviate intestinal inflammation and damage induced by LPS and explore the possible underlying molecular mechanism. L. rhamnosus JL-1 was selected from five L. rhamnosus strains due to its strong adherence capacity to Caco-2 cells (92.89%) and it could survive in simulated gastrointestinal juices. Whole genome sequencing analysis showed that there were no translocation and inversion regions in the genome of L. rhamnosus JL-1 compared with L. rhamnosus GG. Comparative genomic analysis showed that there were encoding genes related to adhesion, acid resistance and bile salt resistance in the genome of L. rhamnosus JL-1. Both in vitro and in vivo experiments indicated that LPS challenge inhibited the mRNA and protein expression of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6). However, the mRNA and protein expressions of pro-inflammatory cytokines were inhibited by pre-treatment with L. rhamnosus JL-1 in a dose-dependent manner. The result of histopathology analysis of ileum showed that oral administration of L. rhamnosus JL-1 reduced pathological damage induced by LPS. Furthermore, it was revealed that L. rhamnosus JL-1 could inhibit the mRNA and protein expressions of TLR4 and NF-κB. These results strongly suggested that L. rhamnosus JL-1 relieved LPS-induced intestinal inflammation by inhibiting the TLR4/NF-κB signaling pathway. To sum up, L. rhamnosus JL-1 has a potential probiotic function and plays an important role in preventing LPS-induced intestinal inflammation and damage.
Collapse
Affiliation(s)
- Xuesong Li
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Dong Hu
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences, Shijiazhuang, 050051, China
| | - Yazhen Tian
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Yang Song
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Yichao Hou
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Linlin Sun
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Yu Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Wei Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
40
|
Raheem A, Liang L, Zhang G, Cui S. Modulatory Effects of Probiotics During Pathogenic Infections With Emphasis on Immune Regulation. Front Immunol 2021; 12:616713. [PMID: 33897683 PMCID: PMC8060567 DOI: 10.3389/fimmu.2021.616713] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
In order to inhibit pathogenic complications and to enhance animal and poultry growth, antibiotics have been extensively used for many years. Antibiotics applications not only affect target pathogens but also intestinal beneficially microbes, inducing long-lasting changes in intestinal microbiota associated with diseases. The application of antibiotics also has many other side effects like, intestinal barrier dysfunction, antibiotics residues in foodstuffs, nephropathy, allergy, bone marrow toxicity, mutagenicity, reproductive disorders, hepatotoxicity carcinogenicity, and antibiotic-resistant bacteria, which greatly compromise the efficacy of antibiotics. Thus, the development of new antibiotics is necessary, while the search for antibiotic alternatives continues. Probiotics are considered the ideal antibiotic substitute; in recent years, probiotic research concerning their application during pathogenic infections in humans, aquaculture, poultry, and livestock industry, with emphasis on modulating the immune system of the host, has been attracting considerable interest. Hence, the adverse effects of antibiotics and remedial effects of probiotics during infectious diseases have become central points of focus among researchers. Probiotics are live microorganisms, and when given in adequate quantities, confer good health effects to the host through different mechanisms. Among them, the regulation of host immune response during pathogenic infections is one of the most important mechanisms. A number of studies have investigated different aspects of probiotics. In this review, we mainly summarize recent discoveries and discuss two important aspects: (1) the application of probiotics during pathogenic infections; and (2) their modulatory effects on the immune response of the host during infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Abdul Raheem
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Lin Liang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Guangzhi Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Shangjin Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| |
Collapse
|
41
|
Dietary alternatives to in-feed antibiotics, gut barrier function and inflammation in piglets post-weaning: Where are we now? Anim Feed Sci Technol 2021. [DOI: 10.1016/j.anifeedsci.2021.114836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
42
|
Escherichia coli Nissle 1917 Enhances Innate and Adaptive Immune Responses in a Ciprofloxacin-Treated Defined-Microbiota Piglet Model of Human Rotavirus Infection. mSphere 2021; 6:6/2/e00074-21. [PMID: 33789939 PMCID: PMC8546683 DOI: 10.1128/msphere.00074-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human rotavirus (HRV) infection is a major cause of gastroenteritis in children worldwide. Broad-spectrum antibiotic-induced intestinal microbial imbalance and the ensuing immune-metabolic dysregulation contribute to the persistence of HRV diarrhea. Escherichia coli Nissle 1917 (EcN), a Gram-negative probiotic, was shown to be a potent immunostimulant and alleviated HRV-induced diarrhea in monocolonized gnotobiotic (Gn) piglets. Our goal was to determine how EcN modulates immune responses in ciprofloxacin (Cipro)-treated Gn piglets colonized with a defined commensal microbiota (DM) and challenged with virulent HRV (VirHRV). Cipro given in therapeutic doses for a short term reduced serum and intestinal total and HRV-specific antibody titers, while EcN treatment alleviated this effect. Similarly, EcN treatment increased the numbers of total immunoglobulin-secreting cells, HRV-specific antibody-secreting cells, activated antibody-forming cells, resting/memory antibody-forming B cells, and naive antibody-forming B cells in systemic and/or intestinal tissues. Decreased levels of proinflammatory but increased levels of immunoregulatory cytokines and increased frequencies of Toll-like receptor-expressing cells were evident in the EcN-treated VirHRV-challenged group. Moreover, EcN treatment increased the frequencies of T helper and T cytotoxic cells in systemic and/or intestinal tissues pre-VirHRV challenge and the frequencies of T helper cells, T cytotoxic cells, effector T cells, and T regulatory cells in systemic and/or intestinal tissues postchallenge. Moreover, EcN treatment increased the frequencies of systemic and mucosal conventional and plasmacytoid dendritic cells, respectively, and the frequencies of systemic natural killer cells. Our findings demonstrated that Cipro use altered immune responses of DM-colonized neonatal Gn pigs, while EcN supplementation rescued these immune parameters partially or completely. IMPORTANCE Rotavirus (RV) is a primary cause of malabsorptive diarrhea in children and is associated with significant morbidity and mortality, especially in developing countries. The use of antibiotics exacerbates intestinal microbial imbalance and results in the persistence of RV-induced diarrhea. Probiotics are now being used to treat enteric infections and ulcerative colitis. We showed previously that probiotics partially protected gnotobiotic (Gn) piglets against human RV (HRV) infection and decreased the severity of diarrhea by modulating immune responses. However, the interactions between antibiotic and probiotic treatments and HRV infection in the context of an established gut microbiota are poorly understood. In this study, we developed a Gn pig model to study antibiotic-probiotic-HRV interactions in the context of a defined commensal microbiota (DM) that mimics aspects of the infant gut microbiota. Our results provide valuable information that will contribute to the treatment of antibiotic- and/or HRV-induced diarrhea and may be applicable to other enteric infections in children.
Collapse
|
43
|
Yi Q, Liu J, Zhang Y, Qiao H, Chen F, Zhang S, Guan W. Anethole Attenuates Enterotoxigenic Escherichia coli-Induced Intestinal Barrier Disruption and Intestinal Inflammation via Modification of TLR Signaling and Intestinal Microbiota. Front Microbiol 2021; 12:647242. [PMID: 33841372 PMCID: PMC8027122 DOI: 10.3389/fmicb.2021.647242] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
This study aimed to investigate the effects of dietary anethole supplementation on the growth performance, intestinal barrier function, inflammatory response, and intestinal microbiota of piglets challenged with enterotoxigenic Escherichia coli K88. Thirty-six weaned piglets (24 ± 1 days old) were randomly allocated into four treatment groups: (1) sham challenge (CON); (2) Escherichia coli K88 challenge (ETEC); (3) Escherichia coli K88 challenge + antibiotics (ATB); and (4) Escherichia coli K88 challenge + anethole (AN). On day 12, the piglets in the ETEC, ATB, and AN group were challenged with 10 mL E. coli K88 (5 × 109 CFU/mL), whereas the piglets in the CON group were orally injected with 10 mL nutrient broth. On day 19, all the piglets were euthanized for sample collection. The results showed that the feed conversion ratio (FCR) was increased in the Escherichia coli K88-challenged piglets, which was reversed by the administration of antibiotics or anethole (P < 0.05). The duodenum and jejunum of the piglets in ETEC group exhibited greater villous atrophy and intestinal morphology disruption than those of the piglets in CON, ATB, and AN groups (P < 0.05). Administration of anethole protected intestinal barrier function and upregulated mucosal layer (mRNA expression of mucin-1 in the jejunum) and tight junction proteins (protein abundance of ZO-1 and Claudin-1 in the ileum) of the piglets challenged with Escherichia coli K88 (P < 0.05). In addition, administration of antibiotics or anethole numerically reduced the plasma concentrations of IL-1β and TNF-α (P < 0.1) and decreased the mRNA expression of TLR5, TLR9, MyD88, IL-1β, TNF-α, IL-6, and IL-10 in the jejunum of the piglets after challenge with Escherichia coli K88 (P < 0.05). Dietary anethole supplementation enriched the abundance of beneficial flora in the intestines of the piglets. In summary, anethole can improve the growth performance of weaned piglets infected by ETEC through attenuating intestinal barrier disruption and intestinal inflammation.
Collapse
Affiliation(s)
- Qingyuan Yi
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiaxin Liu
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yufeng Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Hanzhen Qiao
- College of Biological Engineering, Henan University of Technology, Zhengzhou, China
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| |
Collapse
|
44
|
Fu R, Liang C, Chen D, Yan H, Tian G, Zheng P, He J, Yu J, Mao X, Huang Z, Luo Y, Luo J, Yu B. Effects of dietary Bacillus coagulans and yeast hydrolysate supplementation on growth performance, immune response and intestinal barrier function in weaned piglets. J Anim Physiol Anim Nutr (Berl) 2021; 105:898-907. [PMID: 33715204 DOI: 10.1111/jpn.13529] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 09/10/2020] [Accepted: 02/20/2021] [Indexed: 12/25/2022]
Abstract
The present study investigated the effects of Bacillus coagulans and yeast hydrolysate supplementation on growth performance, immune response and intestinal barrier function of weaned piglets. Twenty-four weaned piglets with an average body weight (BW) of 6.89 ± 0.15 kg were divided into four diets for 28 days. The treatments were basal diet (control), basal diet supplemented with antibiotic (20 mg/kg colistin sulphate and 40 mg/kg bacitracin zinc, AT), probiotics (400 mg/kg Bacillus coagulans ≥5 × 109 CFU/g, BC) or yeast hydrolysate (5000 mg/kg yeast hydrolysate, YH). Average daily gain (ADG) and average daily feed intake (ADFI) were improved by AT and YH diets (p < 0.05), while BC diet only increased ADG (p < 0.05). The complement 3 (C3), lysozyme (LZM) and tumour necrosis factor-α (TNF-α) concentrations in serum were increased in BC diet (p < 0.05). Feeding AT and YH caused the increase of jejunal villus height (p < 0.05), and a higher ratio of villus height/crypt depth was observed in AT, BC and YH groups (p < 0.05). The mRNA expression of zonula occludens-1 (ZO-1) in jejunal mucosa was up-regulated by AT, BC and YH diets (p < 0.05). Dietary AT, BC or YH inclusion decreased the interleukin-1β (IL-1β) concentration and TNF-α mRNA expression (p < 0.05), and YH supplementation even down-regulated toll-like receptor 4 (TLR4) and CD14 expressions (p < 0.05). In summary, the dietary administration of BC or YH both improves growth performance through promoting the intestinal barrier function, indicating both of them can serve as potential alternatives to antibiotics growth promoters for the piglet production.
Collapse
Affiliation(s)
- Runqi Fu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Chan Liang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Hui Yan
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Gang Tian
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Xiangbing Mao
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Junqiu Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
45
|
Arsène MMJ, Davares AKL, Andreevna SL, Vladimirovich EA, Carime BZ, Marouf R, Khelifi I. The use of probiotics in animal feeding for safe production and as potential alternatives to antibiotics. Vet World 2021; 14:319-328. [PMID: 33776297 PMCID: PMC7994123 DOI: 10.14202/vetworld.2021.319-328] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022] Open
Abstract
Although the production of safe food for human consumption is the primary purpose for animal rearing, the environment and well-being of the animals must also be taken into consideration. Based on microbiological point of view, the production of healthy food from animals involves considering foodborne pathogens, on the one hand and on the other hand, the methods used to fight against germs during breeding. The conventional method to control or prevent bacterial infections in farming is the use antibiotics. However, the banning of these compounds as growth promoters caused many changes in animal breeding and their use has since been limited to the treatment and prevention of bacterial infections. In this function, their importance no longer needs to be demonstrated, but unfortunately, their excessive and abusive use have led to a double problem which can have harmful consequences on consumer health: Resistance to antibiotics and the presence of antibiotic residues in food. The use of probiotics appears to be a suitable alternative to overcome these problems because of their ability to modulate the immune system and intestinal microflora, and further considering their antagonistic role against certain pathogenic bacteria and their ability to play the role of growth factor (sometimes associated with prebiotics) when used as feed additives. This review aims to highlight some of the negative effects of the use of antibiotics in animal rearing as well as emphasize the current knowledge on the use of probiotics as a feed additive, their influence on animal production and their potential utility as an alternative to conventional antibiotics, particularly in poultry, pig, and fish farming.
Collapse
Affiliation(s)
- Mbarga M. J. Arsène
- Department of microbiology and virology, Institute of Medicine, RUDN University, Moscow, Russia
- Department of Food Sciences and Nutrition, National School of Agro-industrial Sciences, University of Ngaoundere, Cameroon
| | - Anyutoulou K. L. Davares
- Department of Food Sciences and Nutrition, National School of Agro-industrial Sciences, University of Ngaoundere, Cameroon
| | - Smolyakova L. Andreevna
- Department of microbiology and virology, Institute of Medicine, RUDN University, Moscow, Russia
| | | | - Bassa Z. Carime
- Department of Food Sciences and Nutrition, National School of Agro-industrial Sciences, University of Ngaoundere, Cameroon
| | - Razan Marouf
- Department of microbiology and virology, Institute of Medicine, RUDN University, Moscow, Russia
| | - Ibrahim Khelifi
- Department of microbiology and virology, Institute of Medicine, RUDN University, Moscow, Russia
| |
Collapse
|
46
|
Nguyen TH, Turek I, Meehan-Andrews T, Zacharias A, Irving H. Analysis of interleukin-1 receptor associated kinase-3 (IRAK3) function in modulating expression of inflammatory markers in cell culture models: A systematic review and meta-analysis. PLoS One 2020; 15:e0244570. [PMID: 33382782 PMCID: PMC7774834 DOI: 10.1371/journal.pone.0244570] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/13/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND IRAK3 is a critical modulator of inflammation in innate immunity. IRAK3 is associated with many inflammatory diseases, including sepsis, and is required in endotoxin tolerance to maintain homeostasis of inflammation. The impact of IRAK3 on inflammatory markers such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in cell culture models remains controversial. OBJECTIVE To analyse temporal effects of IRAK3 on inflammatory markers after one- or two-challenge interventions in cell culture models. METHODS A systematic search was performed to identify in vitro cell studies reporting outcome measures of expression of IRAK3 and inflammatory markers. Meta-analyses were performed where sufficient data were available. Comparisons of outcome measures were performed between different cell lines and human and mouse primary cells. RESULTS The literature search identified 7766 studies for screening. After screening titles, abstracts and full-texts, a total of 89 studies were included in the systematic review. CONCLUSIONS The review identifies significant effects of IRAK3 on decreasing NF-κB DNA binding activity in cell lines, TNF-α protein level at intermediate time intervals (4h-15h) in cell lines or at long term intervals (16h-48h) in mouse primary cells following one-challenge. The patterns of TNF-α protein expression in human cell lines and human primary cells in response to one-challenge are more similar than in mouse primary cells. Meta-analyses confirm a negative correlation between IRAK3 and inflammatory cytokine (IL-6 and TNF-α) expression after two-challenges.
Collapse
Affiliation(s)
- Trang Hong Nguyen
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Ilona Turek
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Terri Meehan-Andrews
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Anita Zacharias
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Helen Irving
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| |
Collapse
|
47
|
Dietary xylanase and live yeast supplementation influence intestinal bacterial populations and growth performance of piglets fed a sorghum-based diet. ACTA ACUST UNITED AC 2020; 6:457-466. [PMID: 33364462 PMCID: PMC7750802 DOI: 10.1016/j.aninu.2020.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 03/12/2020] [Accepted: 05/17/2020] [Indexed: 11/27/2022]
Abstract
This study was to evaluate the effect of xylanase supplementation and the addition of live yeast, Saccharomyces cerevisiae, on growth performance and intestinal microbiota in piglets. One hundred and eighty commercial crossbred 23-d-old piglets (PIC 417) were sorted by initial BW and allocated to 3 treatments: control (CTR) diet, CTR diet supplemented with xylanase at 16,000 birch xylan units/kg (XYL) and XYL diet supplemented with live yeast (2 × 1010 CFU/g) at 1 kg/t (XYL + LY). Each treatment had 10 replicates, with 6 animals each. A sorghum-based diet and water were available ad libitum for 42 d of the study. Average daily gain (ADG) and average daily feed intake (ADFI) were measured from 0 to 42 d (23- to 65-d-old) and feed conversion ratio (FCR) calculated. At the end of the study, bacterial identification through 16S rRNA (V3 to V4) sequencing of the ileal and caecal digesta from one piglet per replicate was performed. No treatment effects were observed on ADFI. Pigs offered the live yeast in addition to the xylanase had increased ADG compared with those supplemented with xylanase alone (XYL + LY vs. XYL; P = 0.655). FCR was improved with XYL and XYL + LY compared with CTR (P = 0.018). Clostridiaceae counts in the ileum tended to reduce by 10% with XYL and 14% with XYL + LY compared to CTR (P = 0.07). XYL and XYL + LY increased the counts of Lactobacillaceae in the caecum compared with CTR (P < 0.0001). Dietary supplementation of live yeast combined with xylanase improved growth performance and microbial balance of piglets during the nursery phase.
Collapse
|
48
|
Sun Z, Li H, Li Y, Qiao J. Lactobacillus salivarius, a Potential Probiotic to Improve the Health of LPS-Challenged Piglet Intestine by Alleviating Inflammation as Well as Oxidative Stress in a Dose-Dependent Manner During Weaning Transition. Front Vet Sci 2020; 7:547425. [PMID: 33392276 PMCID: PMC7772421 DOI: 10.3389/fvets.2020.547425] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 11/23/2020] [Indexed: 12/23/2022] Open
Abstract
Intestinal health is a critical issue for piglets during their weaning transition period. Previous reports have emphasized the promise of distinct probiotics in improving the enteric health. Here in this research, a newly isolated Lactobacillus salivarius strain was pretreated to Lipopolysaccharide (LPS)-challenged piglets and its association with integrity of the intestinal barrier coupled with effective dosage were expected to be signified. In the present study, 72 piglets (Landrace × Yorkshiere × Duroc) were randomly allotted to four groups, each group with six replicates. The subjects in the control group were provided with basal diet while those in other tested groups with extra 0.05, 0.1, and 0.2% L. salivarius, respectively. Fourteen days later, LPS was intraperitoneally injected and sodium pentobarbital was then delivered to euthanize those LPS-challenged piglets. An increase of average daily gain and body weight along with an apparent decline of diarrhea rate were observed in L. salivarius-treated groups. Both 0.1 and 0.2% L. salivarius supplement in total diet had the capability to markedly elevate levels of CAT, GSH-Px, SOD, anti-inflammatory cytokine from the serum as well as tight junction proteins (Claudin-1, Occludin, and ZO-1) extracted from intestine in LPS-challenged piglets. These changes were accompanied by the obvious downregulation of D-lactic acid, DAO, MDA and pro-inflammatory mediators in the serum, including IL-1β, IL-6, IFN-γ, and TNF-α. Meanwhile, the expression levels of TLR2 and TLR4 in spleen and mesenteric lymph nodes were significantly lower whereas the oxidation-related gene, ho-1 was up-regulated with L. salivarius administration. Our findings suggested that relatively high dose L. salivarius (0.1–0.2%) could regulate the progression of inflammatory response and oxidative stress when individuals were exposed to LPS, thus probably offering valuable assistance in restoring barrier function and improving overall performance.
Collapse
Affiliation(s)
- Zeyang Sun
- Tianjin Key Laboratory of Conservation and Utilization of Animal Diversity, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Haihua Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Yupeng Li
- College of Life Sciences, Tianjin Institute of Animal Husbandry and Veterinary Medicine, Tianjin, China
| | - Jiayun Qiao
- Tianjin Key Laboratory of Conservation and Utilization of Animal Diversity, College of Life Sciences, Tianjin Normal University, Tianjin, China
| |
Collapse
|
49
|
van Zyl WF, Deane SM, Dicks LM. Molecular insights into probiotic mechanisms of action employed against intestinal pathogenic bacteria. Gut Microbes 2020; 12:1831339. [PMID: 33112695 PMCID: PMC7595611 DOI: 10.1080/19490976.2020.1831339] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) diseases, and in particular those caused by bacterial infections, are a major cause of morbidity and mortality worldwide. Treatment is becoming increasingly difficult due to the increase in number of species that have developed resistance to antibiotics. Probiotic lactic acid bacteria (LAB) have considerable potential as alternatives to antibiotics, both in prophylactic and therapeutic applications. Several studies have documented a reduction, or prevention, of GI diseases by probiotic bacteria. Since the activities of probiotic bacteria are closely linked with conditions in the host's GI-tract (GIT) and changes in the population of enteric microorganisms, a deeper understanding of gut-microbial interactions is required in the selection of the most suitable probiotic. This necessitates a deeper understanding of the molecular capabilities of probiotic bacteria. In this review, we explore how probiotic microorganisms interact with enteric pathogens in the GIT. The significance of probiotic colonization and persistence in the GIT is also addressed.
Collapse
Affiliation(s)
- Winschau F. van Zyl
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Shelly M. Deane
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Leon M.T. Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa,CONTACT Leon M.T. Dicks; Department of Microbiology; Stellenbosch University, Stellenbosch7602, South Africa
| |
Collapse
|
50
|
Yang M, Bose S, Lim S, Seo J, Shin J, Lee D, Chung WH, Song EJ, Nam YD, Kim H. Beneficial Effects of Newly Isolated Akkermansia muciniphila Strains from the Human Gut on Obesity and Metabolic Dysregulation. Microorganisms 2020; 8:E1413. [PMID: 32937828 PMCID: PMC7564497 DOI: 10.3390/microorganisms8091413] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/25/2022] Open
Abstract
The identification of new probiotics with anti-obesity properties has attracted considerable interest. In the present study, the anti-obesity activities of Akkermansia muciniphila (A. muciniphila) strains isolated from human stool samples and their relationship with the gut microbiota were evaluated using a high fat-diet (HFD)-fed mice model. Three strains of A. muciniphila were chosen from 27 isolates selected based on their anti-lipogenic activity in 3T3-L1 cells. The anti-lipogenic, anti-adipogenic and anti-obesity properties of these three strains were evaluated further in HFD-induced obese mice. The animals were administered these strains six times per week for 12 weeks. The treatment improved the HFD-induced metabolic disorders in mice in terms of the prevention of body weight gain, caloric intake and reduction in the weights of the major adipose tissues and total fat. In addition, it improved glucose homeostasis and insulin sensitivity. These effects were also associated with the inhibition of low-grade intestinal inflammation and restoration of damaged gut integrity, prevention of liver steatosis and improvement of hepatic function. These results revealed a difference in the distribution pattern of the gut microbial communities between groups. Therefore, the gut microbial population modulation, at least in part, might contribute to the beneficial impact of the selected A. muciniphila strains against metabolic disorders.
Collapse
Affiliation(s)
- Meng Yang
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, 814 Siksa-dong, Ilsandong-gu, Goyang-si 10326, Korea; (M.Y.); (S.B.); (S.L.)
| | - Shambhunath Bose
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, 814 Siksa-dong, Ilsandong-gu, Goyang-si 10326, Korea; (M.Y.); (S.B.); (S.L.)
| | - Sookyoung Lim
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, 814 Siksa-dong, Ilsandong-gu, Goyang-si 10326, Korea; (M.Y.); (S.B.); (S.L.)
| | - JaeGu Seo
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si 10326, Korea; (J.S.); (J.S.); (D.L.)
| | - JooHyun Shin
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si 10326, Korea; (J.S.); (J.S.); (D.L.)
| | - Dokyung Lee
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si 10326, Korea; (J.S.); (J.S.); (D.L.)
| | - Won-Hyong Chung
- Research Group of Healthcare, Korea Food Research Institute, Wanju 55365, Korea;
| | - Eun-Ji Song
- Research Group of Gut Microbiome, Korea Food Research Institute, Wanju-gun 55365, Korea;
| | - Young-Do Nam
- Research Group of Gut Microbiome, Korea Food Research Institute, Wanju-gun 55365, Korea;
| | - Hojun Kim
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, 814 Siksa-dong, Ilsandong-gu, Goyang-si 10326, Korea; (M.Y.); (S.B.); (S.L.)
| |
Collapse
|