1
|
Liu D, Han X, Zou W, Yang Z, Peng J, Li Y, Liu Y, Jia M, Liu W, Li H, Zhou Q, Tan Z, Zhang J. Probiotics Combined with Metformin Improves Sperm Parameters in Obese Male Mice through Modulation of Intestinal Microbiota Equilibrium. Reprod Sci 2025; 32:116-130. [PMID: 39623123 DOI: 10.1007/s43032-024-01748-9] [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: 06/13/2024] [Accepted: 11/02/2024] [Indexed: 01/15/2025]
Abstract
The decline in sperm parameters among obese males has attracted significant scholarly interest. The intestinal microbiota plays a crucial role in obesity, and investigating the intestinal-reproductive axis may offer a novel molecular approach to addressing the decline in male sperm parameters caused by obesity. To clarify whether probiotics, either alone or in conjunction with metformin, can enhance sperm parameters in obese male mice and assess the underlying mechanisms involved. 6-week-old male mice were constructed as obese models. Probiotics and metformin were used as intervention conditions. Changes in inflammatory factors and ROS content were detected by ELISA, morphological changes in testicular and colon tissues were observed by H&E staining, changes in intestinal microbiota abundance were detected by 16SrRNA gene sequencing, and changes in metabolites such as blood glucose, blood lipids, and lipopolysaccharide were detected by biochemical testing to investigate the mechanism of probiotics, metformin, and their combination to ameliorate reproductive impairment in obese male mice. Our results revealed that high-fat diet would result in reduced testicular spermatogenic tubule hierarchy, decreased spermatogenic cell counts, decreased sperm concentration and motility, and altered abundance of intestinal microbiota, whereas the combination of probiotics and metformin could restore high-fat-mediated pathophysiological alterations thereby ameliorating spermatogenic disorders in mice. The combination of probiotics and metformin can attenuate inflammation and oxidative stress, while enhancing androgen production to improve testicular spermatogenic function by re-construction intestinal microbiota equilibrium in HFD mice.
Collapse
Affiliation(s)
- Dan Liu
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Xiaolong Han
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenda Zou
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Zhenyu Yang
- Department of Pathology, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Juan Peng
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Yuli Liu
- Precision Medicine Center, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Man Jia
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Weijun Liu
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Qianyin Zhou
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Zhirong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China.
| |
Collapse
|
2
|
Zhao Y, Lyu G. Fetal echogenic bowel may be related to intestinal microbiota: A prospective cohort study. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024; 52:1338-1345. [PMID: 39212092 DOI: 10.1002/jcu.23794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE The purpose of the current study was to determine the difference in intestinal microbiota after delivery between healthy fetuses and fetuses with hyperechogenic bowel during the second trimester and the relationship between fetal echogenic bowel and microbiota. METHODS Fourteen healthy fetuses (control group), 13 fetuses with echogenic bowel (EB group), and seven fetuses with echogenic bowel and other abnormalities (C-EB group) were selected. The first meconium after delivery was collected for 16S rRNA sequencing. RESULTS A total of 1 222 131 high-quality sequences were generated after sequencing optimization of all samples. Each sample contained an average of 35 945 high-quality sequences and 2036 operational taxonomic units (OTUs). There was no significant difference in the Shannon, Simpson index among the three groups. At the genus level, the abundance of Escherichia coli/Shigella in the EB and C-EB groups was significantly lower than the control group, while the abundance of Staphylococcus, Methylobactrium, and Curvibacter in the EB group was significantly higher than the other groups. There was a difference in abundance of Gammaproteobacteria, Fusobacteria, Enterobacteriaceae, and E. coli in the EB and C-EB groups. CONCLUSIONS The formation of echogenic bowel may be related to the microbiota.
Collapse
Affiliation(s)
- Yanping Zhao
- Department of Medical Imaging, Quanzhou Medical College, Quanzhou, China
| | - Guorong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
3
|
Shi YB, Dong HL, Chang WK, Zhao Y, Jin HJ, Li JK, Yan S. Genetic evidence for a causal link between gut microbiota and arterial embolism and thrombosis: a two-sample Mendelian randomization study. Front Microbiol 2024; 15:1396699. [PMID: 38957618 PMCID: PMC11217536 DOI: 10.3389/fmicb.2024.1396699] [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: 03/08/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024] Open
Abstract
Background Previous research has hinted at a crucial link between gut microbiota and arterial embolism and thrombosis, yet the causal relationship remains enigmatic. To gain a deeper understanding, we aimed to comprehensively explore the causal relationship and elucidate the impact of the gut microbiota on the risk through a two-sample Mendelian randomization (MR) study. Methods Genetic instrumental variables for gut microbiota were identified from a genome-wide association study (GWAS) of 18,340 participants. Summary statistics for IBS were drawn from a GWAS including 1,076 cases and 381,997 controls. We used the inverse-variance weighted (IVW) method as the primary analysis. To test the robustness of our results, we further performed the weighted median method, MR-Egger regression, and MR pleiotropy residual sum and outlier test. Results We identified three bacterial traits that were associated with the risk of arterial embolism and thrombosis: odds ratio (OR): 1.58, 95% confidence interval (CI): 1.08-2.31, p = 0.017 for genus Catenibacterium; OR: 0.64, 95% CI: 0.42-0.96, p = 0.031 for genus Dialister; and OR: 2.08, 95% CI: 1.25-3.47, p = 0.005 for genus Odoribacter. The results of sensitivity analyses for these bacterial traits were consistent (P<0.05). Conclusion Our systematic analyses provided evidence to support a potential causal relationship between several gut microbiota taxa and the risk of arterial embolism and thrombosis. More studies are required to show how the gut microbiota affects the development of arterial embolism and thrombosis.
Collapse
Affiliation(s)
- Yong-Bin Shi
- Department of Vascular Surgery, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hong-Lin Dong
- Department of Vascular Surgery, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Wen-Kai Chang
- Department of Vascular Surgery, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan Zhao
- Department of Vascular Surgery, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hai-Jiang Jin
- Department of Vascular Surgery, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jun-Kai Li
- Department of Vascular Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Sheng Yan
- Department of Vascular Surgery, Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
4
|
Wu J, Xu X, Duan J, Chai Y, Song J, Gong D, Wang B, Hu Y, Han T, Ding Y, Liu Y, Li J, Cao X. EFHD2 suppresses intestinal inflammation by blocking intestinal epithelial cell TNFR1 internalization and cell death. Nat Commun 2024; 15:1282. [PMID: 38346956 PMCID: PMC10861516 DOI: 10.1038/s41467-024-45539-x] [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: 04/13/2023] [Accepted: 01/23/2024] [Indexed: 02/15/2024] Open
Abstract
TNF acts as one pathogenic driver for inducing intestinal epithelial cell (IEC) death and substantial intestinal inflammation. How the IEC death is regulated to physiologically prevent intestinal inflammation needs further investigation. Here, we report that EF-hand domain-containing protein D2 (EFHD2), highly expressed in normal intestine tissues but decreased in intestinal biopsy samples of ulcerative colitis patients, protects intestinal epithelium from TNF-induced IEC apoptosis. EFHD2 inhibits TNF-induced apoptosis in primary IECs and intestinal organoids (enteroids). Mice deficient of Efhd2 in IECs exhibit excessive IEC death and exacerbated experimental colitis. Mechanistically, EFHD2 interacts with Cofilin and suppresses Cofilin phosphorylation, thus blocking TNF receptor I (TNFR1) internalization to inhibit IEC apoptosis and consequently protecting intestine from inflammation. Our findings deepen the understanding of EFHD2 as the key regulator of membrane receptor trafficking, providing insight into death receptor signals and autoinflammatory diseases.
Collapse
Affiliation(s)
- Jiacheng Wu
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Xiaoqing Xu
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jiaqi Duan
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yangyang Chai
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jiaying Song
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Dongsheng Gong
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Bingjing Wang
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Ye Hu
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Taotao Han
- Department of Gastroenterology, Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuanyuan Ding
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, 215123, China
| | - Yin Liu
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jingnan Li
- Department of Gastroenterology, Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Xuetao Cao
- Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
5
|
Yoon JW, Shin S, Park J, Lee BR, Lee SI. TLR/MyD88-Mediated Inflammation Induced in Porcine Intestinal Epithelial Cells by Ochratoxin A Affects Intestinal Barrier Function. TOXICS 2023; 11:toxics11050437. [PMID: 37235251 DOI: 10.3390/toxics11050437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023]
Abstract
The intestinal epithelium performs vital functions such as nutrient absorption and acting as an intestinal barrier to maintain the host's homeostasis. Mycotoxin, which affects the processing and storage of animal feedstuff, is a problematic pollutant in farming products. Ochratoxin A generated by Aspergillus and Penicillium fungi causes inflammation, intestinal dysfunction, decline in growth, and reduced intake in porcine and other livestock. Despite these ongoing problems, OTA-related studies in intestinal epithelium are lacking. This study aimed to demonstrate that OTA regulates TLR/MyD88 signaling in IPEC-J2 cells and induces barrier function impairment through tight junction reduction. We measured expression of TLR/MyD88 signaling-related mRNAs and proteins. The indicator of intestinal barrier integrity was confirmed through immunofluorescence and transepithelial electrical resistance. Additionally, we confirmed whether inflammatory cytokines and barrier function were affected by MyD88 inhibition. MyD88 inhibition alleviated inflammatory cytokine levels, tight junction reduction, and damage to barrier function due to OTA. These results indicate that OTA induces TLR/MyD88 signaling-related genes and impairs tight junctions and intestinal barrier function in IPEC-J2 cells. MyD88 regulation in OTA-treated IPEC-J2 cells mitigates the tight junction and intestinal barrier function impairments. Our findings provide a molecular understanding of OTA toxicity in porcine intestinal epithelial cells.
Collapse
Affiliation(s)
- Jung Woong Yoon
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju-si 37224, Republic of Korea
| | - Sangsu Shin
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju-si 37224, Republic of Korea
- Research Center for Horse Industry, Kyungpook National University, Sangju-si 37224, Republic of Korea
- Department of Animal Biotechnology, Kyungpook National University, Sangju-si 37224, Republic of Korea
| | - JeongWoong Park
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju-si 37224, Republic of Korea
- Research Center for Horse Industry, Kyungpook National University, Sangju-si 37224, Republic of Korea
| | - Bo Ram Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Sang In Lee
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju-si 37224, Republic of Korea
- Research Center for Horse Industry, Kyungpook National University, Sangju-si 37224, Republic of Korea
- Department of Animal Biotechnology, Kyungpook National University, Sangju-si 37224, Republic of Korea
| |
Collapse
|
6
|
He E, Quan W, Luo J, Liu C, Zheng W, Shen Q. Absorption and Transport Mechanism of Red Meat-Derived N-glycolylneuraminic Acid and Its Damage to Intestinal Barrier Function through the NF-κB Signaling Pathway. Toxins (Basel) 2023; 15:toxins15020132. [PMID: 36828446 PMCID: PMC9966629 DOI: 10.3390/toxins15020132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
N-glycolylneuraminic acid (Neu5Gc) is a specific factor in red meat that induces intestinal disease. Our aim was to investigate the effect of Neu5Gc on the intestinal barrier as well as its mechanism of endocytosis and exocytosis. Ten specific inhibitors were used to explore the mechanism of Neu5Gc endocytosis and exocytosis by Caco-2 cells. Amiloride hydrochloride and cytochalasin D had the strongest inhibitory effect on the endocytosis of Neu5Gc. Sodium azide, dynasore, chlorpromazine hydrochloride, and nystatin also inhibited Neu5Gc endocytosis. Dynasore exhibited a stronger inhibitory effect than that of chlorpromazine hydrochloride or nystatin alone. Exocytosis inhibitors, including nocodazole, brefeldin A, monensin, and bafilomycin A, inhibited the transmembrane transport of Neu5Gc. Monensin promoted the exocytosis of Neu5Gc from Caco-2 cells. In another experiment, we observed no significant inhibitory effects of monensin and brefeldin A. Dietary concentrations of Neu5Gc induced prominent damage to intestinal tight junction proteins zonula occludens-1 (ZO-1), occludin, and claudin-1 and promoted the phosphorylation of IκB-α and P65 to activate the canonical Nuclear Factor kappa-B (NF-κB) pathway. Neu5Gc increased the RNA levels of pro-inflammatory factors IL-1β, IL-6, and TNF-α and inhibited those of anti-inflammatory factors TGF-β and IL-10. BAY, an NF-κB signaling pathway inhibitor, attenuated these changes. Reductions in the levels of ZO-1, occludin, and claudin-1 were recovered in response to BAY. Our data reveal the endocytosis and exocytosis mechanism of Neu5Gc and prove that Neu5Gc can activate the canonical NF-κB signaling pathway, regulate the transcription of inflammatory factors, thereby damaging intestinal barrier function.
Collapse
|
7
|
Mei Z, Yuan J, Li D. Biological activity of galacto-oligosaccharides: A review. Front Microbiol 2022; 13:993052. [PMID: 36147858 PMCID: PMC9485631 DOI: 10.3389/fmicb.2022.993052] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Galacto-oligosaccharides (GOS) are oligosaccharides formed by β-galactosidase transgalactosylation. GOS is an indigestible food component that can pass through the upper gastrointestinal tract relatively intact and ferment in the colon to produce short-chain fatty acids (SCFAs) that further regulate the body’s intestinal flora. GOS and other prebiotics are increasingly recognized as useful food tools for regulating the balance of colonic microbiota-human health. GOS performed well compared to other oligosaccharides in regulating gut microbiota, body immunity, and food function. This review summarizes the sources, classification, preparation methods, and biological activities of GOS, focusing on the introduction and summary of the effects of GOS on ulcerative colitis (UC), to gain a comprehensive understanding of the application of GOS.
Collapse
Affiliation(s)
- Zhaojun Mei
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People’s Hospital, Luzhou, China
| | - Jiaqin Yuan
- Department of Orthopedics, The Second People’s Hospital of Yibin, Yibin, China
| | - Dandan Li
- University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Dandan Li,
| |
Collapse
|
8
|
Yoon JW, Lee SI. Gene expression profiling after ochratoxin A treatment in small intestinal epithelial cells from pigs. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:842-853. [PMID: 36287785 PMCID: PMC9574622 DOI: 10.5187/jast.2022.e49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 01/24/2023]
Abstract
Ochratoxin A (OTA) is a well-known mycotoxin that causes disease through the ingestion of contaminated food or feed, for example, in the porcine industry. The intestinal epithelium acts as the first barrier against food contamination. We conducted a study on the exposure of the porcine intestinal epithelium to OTA. We used the intestinal porcine epithelial cell line IPEC-J2 as an in vitro model to evaluate the altered molecular mechanisms following OTA exposure. Gene expression profiling revealed that OTA upregulated 782 genes and downregulated 896, totalling 1678 differentially expressed genes. Furthermore, immunofluorescence, quantitative real-time polymerase chain reaction, and western blotting confirmed that OTA damages the tight junction protein ZO-1. Moreover, OTA activated the expression of inflammatory genes (IL-6, IL-8, IL-10, NF-kB, TLR4, and TNF-α). In summary, this study confirmed that OTA alters various molecular mechanisms and has several adverse effects on IPEC-J2 cells.
Collapse
Affiliation(s)
- Jung Woong Yoon
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea
| | - Sang In Lee
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea,Corresponding author: Sang In Lee,
Department of Animal Science and Biotechnology, Kyungpook National University,
Sangju 37224, Korea. Tel: +82-54-530-1943, E-mail:
| |
Collapse
|
9
|
Abstract
Akkermansia muciniphila is a commensal bacterium using mucin as its sole carbon and nitrogen source. A. muciniphila is a promising candidate for next-generation probiotics to prevent inflammatory and metabolic disorders, including diabetes and obesity, and to increase the response to cancer immunotherapy. In this study, a comparative pan-genome analysis was conducted to investigate the genomic diversity and evolutionary relationships between complete genomes of 27 A. muciniphila strains, including KGMB strains isolated from healthy Koreans. The analysis showed that A. muciniphila strains formed two clades of group A and B in a phylogenetic tree constructed using 1,219 orthologous single-copy core genes. Interestingly, group A comprised of strains from human feces in Korea, whereas most of group B comprised strains from human feces in Europe and China, and from mouse feces. As group A and B branched, mucin hydrolysis played an important role in the stability of the core genome and drove evolution in the direction of defense against invading pathogens, survival in, and colonization in the mucus layer. In addition, WapA and anSME, which function in competition and post-translational modification of sulfatase, respectively, have been a particularly important selective pressure in the evolution of group A. KGMB strains in group A with anSME gene showed sulfatase activity, but KCTC 15667T in group B without anSME did not. Our findings revealed that KGMB strains evolved to gain an edge in the competition with other gut bacteria by increasing the utilization of sulfated mucin, which will allow it to become highly colonized in the gut environment.
Collapse
Affiliation(s)
- Ji-Sun Kim
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Se Won Kang
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Seung-Hwan Park
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Jung-Sook Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea,Department of Environmental Biotechnology, University of Science and Technology, Yuseong-gu, Republic of Korea,CONTACT Jung-Sook Lee Korean Collection for Type Cultures,Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si56212Republic of Korea
| |
Collapse
|
10
|
Conte M, Nigro F, Porpora M, Bellomo C, Furone F, Budelli AL, Nigro R, Barone MV, Nanayakkara M. Gliadin Peptide P31-43 Induces mTOR/NFkβ Activation and Reduces Autophagy: The Role of Lactobacillus paracasei CBA L74 Postbiotc. Int J Mol Sci 2022; 23:ijms23073655. [PMID: 35409015 PMCID: PMC8999065 DOI: 10.3390/ijms23073655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022] Open
Abstract
Celiac disease (CD) is an autoimmune disease characterized by an altered immune response stimulated by gliadin peptides that are not digested and cause damage to the intestinal mucosa. The aim of this study was to investigate whether the postbiotic Lactobacillus paracasei (LP) could prevent the action of gliadin peptides on mTOR, autophagy, and the inflammatory response. Most of the experiments performed were conducted on intestinal epithelial cells Caco-2 treated with a peptic-tryptic digest of gliadin (PTG) and P31-43. Furthermore, we pretreated the Caco-2 with the postbiotic LP before treatment with the previously described stimuli. In both cases, we evaluated the levels of pmTOR, p70S6k, and p4EBP-1 for the mTOR pathway, pNFkβ, and pERK for inflammation and LC 3 and p62 for autophagy. For autophagy, we also used immunofluorescence analysis. Using intestinal organoids derivate from celiac (CD) patients, we analyzed the effect of gliadin after postbiotic pretreatment with LP on inflammation marker NFkβ. Through these experiments, we showed that gliadin peptides are able to induce the increase of the inflammatory response in a more complex model of intestinal epithelial cells. LP postbiotic was able to induce autophagy in Caco-2 cells and prevent gliadin effects. In conclusion, postbiotic pretreatment with LP could be considered for in vivo clinical trials.
Collapse
Affiliation(s)
- Mariangela Conte
- ELFID (European Laboratory for the Investigation of Food Induced Diseases), Department of Translational Medical Science, Section of Paediatrics, University Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.C.); (M.P.); (C.B.); (F.F.); (M.N.)
| | - Federica Nigro
- I.T.P. Innovation and Technology Provider s.r.l., Via Bisignano a Chiaia 68, 80121 Naples, Italy;
| | - Monia Porpora
- ELFID (European Laboratory for the Investigation of Food Induced Diseases), Department of Translational Medical Science, Section of Paediatrics, University Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.C.); (M.P.); (C.B.); (F.F.); (M.N.)
| | - Claudia Bellomo
- ELFID (European Laboratory for the Investigation of Food Induced Diseases), Department of Translational Medical Science, Section of Paediatrics, University Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.C.); (M.P.); (C.B.); (F.F.); (M.N.)
| | - Francesca Furone
- ELFID (European Laboratory for the Investigation of Food Induced Diseases), Department of Translational Medical Science, Section of Paediatrics, University Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.C.); (M.P.); (C.B.); (F.F.); (M.N.)
| | - Andrea Luigi Budelli
- DICMAPI, University of Naples Federico II, 80125 Naples, Italy; (A.L.B.); (R.N.)
| | - Roberto Nigro
- DICMAPI, University of Naples Federico II, 80125 Naples, Italy; (A.L.B.); (R.N.)
| | - Maria Vittoria Barone
- ELFID (European Laboratory for the Investigation of Food Induced Diseases), Department of Translational Medical Science, Section of Paediatrics, University Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.C.); (M.P.); (C.B.); (F.F.); (M.N.)
- Correspondence: ; Tel.: +39-0817464568
| | - Merlin Nanayakkara
- ELFID (European Laboratory for the Investigation of Food Induced Diseases), Department of Translational Medical Science, Section of Paediatrics, University Federico II, Via S. Pansini 5, 80131 Naples, Italy; (M.C.); (M.P.); (C.B.); (F.F.); (M.N.)
| |
Collapse
|
11
|
Matsumoto Y, Koga H, Takahashi M, Suda K, Ochi T, Seo S, Miyano G, Miyake Y, Nakajima H, Yoshida S, Mikami T, Okazaki T, Hattori N, Yamataka A, Nakamura T. Defined serum-free culture of human infant small intestinal organoids with predetermined doses of Wnt3a and R-spondin1 from surgical specimens. Pediatr Surg Int 2021; 37:1543-1554. [PMID: 34216241 DOI: 10.1007/s00383-021-04957-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE Refinement of organoid technology is important for studying physiology and disease of the intestine. We aimed to optimize defined serum-free conditions for human infant small intestinal (SI) organoid culture with predetermined doses of Wnt3a and Rspo1 from surgical specimens. We further assessed whether intestinal specimens could be stored before use as a source of organoids. METHODS Different doses of Wnt3a and Rspo1 in a serum-free medium were tested to establish a condition in which surgically resected SI cells grew as organoids over multiple passages. The expression of marker genes for stem and differentiated cells was assessed by quantitative polymerase chain reaction. We also investigated the organoid-forming efficiency of cells in degenerating intestines stored at 4 °C for various intervals post-resection. RESULTS We determined the doses of Wnt3a and Rspo1 required for the continuous growth of infant SI organoids with multi-differentiation potential. We revealed that, despite the time-dependent loss of stem cells, tissues stored for up to 2 days preserved cells capable of generating amplifiable organoids. CONCLUSION SI cells can be grown as organoids under defined conditions. This could provide a reproducible and customizable method of using surgical specimens for the study of intestinal maturation and their relevance to pediatric diseases.
Collapse
Affiliation(s)
- Yuka Matsumoto
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroyuki Koga
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Mirei Takahashi
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kazuto Suda
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takanori Ochi
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Shogo Seo
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Go Miyano
- Department of Pediatric Surgery, Juntendo Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi, Chiba, 279-0021, Japan
| | - Yuichiro Miyake
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hideaki Nakajima
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Shiho Yoshida
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takafumi Mikami
- Department of Pediatric Surgery, Juntendo Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi, Chiba, 279-0021, Japan
| | - Tadaharu Okazaki
- Department of Pediatric Surgery, Juntendo Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi, Chiba, 279-0021, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Atsuyuki Yamataka
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Tetsuya Nakamura
- Department of Research and Development for Organoids, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
12
|
Rodrigues LE, Kishibe MM, Keller R, Caetano HRDS, Rufino MN, Sanches ODC, Giometti IC, Giuffrida R, Bremer-Neto H. Prebiotics mannan-oligosaccharides accelerate sexual maturity in rats: A randomized preclinical study. Vet World 2021; 14:1210-1219. [PMID: 34220123 PMCID: PMC8243662 DOI: 10.14202/vetworld.2021.1210-1219] [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: 12/18/2020] [Accepted: 03/24/2021] [Indexed: 12/20/2022] Open
Abstract
Background and Aim: The prebiotics, mannan-oligosaccharides (MOS), demonstrate the ability to increase probiotic microorganisms and fixation and removal of pathogens associated with chronic systemic inflammation in the digestive system. Inflammatory processes play an important role in modulating the brain-intestinal axis, including maintaining male reproductive function and spermatogenesis and regulating stress. The aim of the present study was to evaluate the action of MOS on testosterone and corticosterone concentrations and the reproductive system development of rats in the growth phase as an animal model. Materials and Methods: In total, 128 male rats were used, randomly divided into four experimental groups (n=32): Control; MOS 1; MOS 2; and MOS 3. From each group, eight animals were sacrificed in four experimental moments (14, 28, 42, and 56 days, respectively, moments 1, 2, 3, and 4) and hormonal measurements and histological evaluations were performed. Results: The results revealed the effect of diet, MOS, and timing on testicle weight (p<0.05). At moments 3 and 4, the groups supplemented with MOS showed higher concentrations of testosterone and decreased corticosterone levels throughout the experimental period. Groups supplemented with MOS showed an increase in the frequency of relative sperm and sperm scores. The radii of the seminiferous tubules presented a significant statistical effect of the diet, moments, and diet + moment interaction. Conclusion: It was concluded that the three different MOS prebiotics brought forward sexual maturity.
Collapse
Affiliation(s)
- Luiz Eduardo Rodrigues
- Department of Functional Sciences, Laboratory of Physiology and Biophysics, Faculty of Medicine, Universidade do Oeste Paulista, Presidente Prudente, São Paulo, Brazil
| | - Milena Miyoshi Kishibe
- Department of Functional Sciences, Laboratory of Physiology and Biophysics, Faculty of Medicine, Universidade do Oeste Paulista, Presidente Prudente, São Paulo, Brazil
| | - Rogeria Keller
- Department of Functional Sciences, Laboratory of Microbiology, Faculty of Biological Sciences, Universidade do Oeste Paulista, Presidente Prudente, São Paulo, Brazil
| | - Heliard Rodrigues Dos Santos Caetano
- Department of Functional Sciences, Laboratory of Physiology, Faculty of Physiotherapy, Universidade do Oeste Paulista, Presidente Prudente, São Paulo, Brazil
| | - Marcos Natal Rufino
- Department of Functional Sciences, Laboratory of Physiology, Faculty of Medicine, Universidade do Oeste Paulista, Presidente Prudente, São Paulo, Brazil
| | | | - Ines Cristina Giometti
- Department of Reproduction, Faculty of Veterinary Medicine, Universidade do Oeste Paulista, Presidente Prudente, São Paulo, Brazil
| | - Rogério Giuffrida
- Department of Statistics, Faculty of Veterinary Medicine, Universidade do Oeste Paulista, São Paulo, Brazil
| | - Hermann Bremer-Neto
- Department of Functional Sciences, Laboratory of Physiology and Biophysics, Faculty of Medicine, Universidade do Oeste Paulista, Presidente Prudente, São Paulo, Brazil
| |
Collapse
|
13
|
Andersen MCE, Johansen MW, Nissen T, Nexoe AB, Madsen GI, Sorensen GL, Holmskov U, Schlosser A, Moeller JB, Husby S, Rathe M. FIBCD1 ameliorates weight loss in chemotherapy-induced murine mucositis. Support Care Cancer 2021; 29:2415-2421. [PMID: 32918133 DOI: 10.1007/s00520-020-05762-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/08/2020] [Indexed: 11/30/2022]
Abstract
PURPOSE Chemotherapy-induced gastrointestinal toxicity is a common adverse event during chemotherapeutic treatment. No uniformly applicable strategies exist to predict, prevent, or treat gastrointestinal toxicity. Thus, a goal of mucositis research is to identify targets for therapeutic interventions and individualized risk prediction. Fibrinogen C domain containing 1 (FIBCD1) is a transmembrane protein expressed in human intestinal epithelial cells with functions in the innate immune system. Previous observations have shown that FIBCD1 ameliorates dextran sulfate sodium (DSS)-induced intestinal inflammation in vivo. We evaluated the effect of FIBCD1 in a murine model of chemotherapy-induced gastrointestinal toxicity and inflammation. METHODS Transgenic (Tg) mice overexpressing FIBCD1 in the intestinal epithelium (Fibcd1Tg) and wild-type (WT) littermates (C57BL/6N) were randomized to receive an intraperitoneal injection of doxorubicin 20 mg/kg or saline and were terminated 2 or 7 days after the injection. Gastrointestinal toxicity was evaluated by weight change, intestinal length, villus height/crypt depth, and histological mucositis score. Expression of inflammatory markers (IL-6, IL-1β, and Tnfα) was measured by quantitative real-time PCR in intestinal tissue samples. RESULTS Following doxorubicin treatment, WT mice exhibited an increased weight loss compared with Tg littermates (p < 0.001). No differences between genotypes were seen in mucositis score, intestinal length, villus height/crypt depth, or IL-6, IL-1β, and Tnfα expression. CONCLUSION Our findings suggest that FIBCD1 could ameliorate chemotherapy-induced gastrointestinal toxicity by reducing weight loss; however, the mechanism of this possible protective effect remains to be defined warranting additional investigations.
Collapse
Affiliation(s)
- Maria C E Andersen
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Sdr. Boulevard 29, DK-5000, Odense C, Denmark
- Department of Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Malene W Johansen
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Sdr. Boulevard 29, DK-5000, Odense C, Denmark
- Department of Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Thomas Nissen
- Department of Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Anders B Nexoe
- Department of Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Gunvor I Madsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Grith L Sorensen
- Department of Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Uffe Holmskov
- Department of Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Anders Schlosser
- Department of Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Jesper B Moeller
- Department of Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Steffen Husby
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Sdr. Boulevard 29, DK-5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mathias Rathe
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Sdr. Boulevard 29, DK-5000, Odense C, Denmark.
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
14
|
Microbiota-mediated protection against antibiotic-resistant pathogens. Genes Immun 2021; 22:255-267. [PMID: 33947987 PMCID: PMC8497270 DOI: 10.1038/s41435-021-00129-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/19/2021] [Accepted: 04/09/2021] [Indexed: 02/03/2023]
Abstract
Colonization by the microbiota provides one of our most effective barriers against infection by pathogenic microbes. The microbiota protects against infection by priming immune defenses, by metabolic exclusion of pathogens from their preferred niches, and through direct antimicrobial antagonism. Disruption of the microbiota, especially by antibiotics, is a major risk factor for bacterial pathogen colonization. Restoration of the microbiota through microbiota transplantation has been shown to be an effective way to reduce pathogen burden in the intestine but comes with a number of drawbacks, including the possibility of transferring other pathogens into the host, lack of standardization, and potential disruption to host metabolism. More refined methods to exploit the power of the microbiota would allow us to utilize its protective power without the drawbacks of fecal microbiota transplantation. To achieve this requires detailed understanding of which members of the microbiota protect against specific pathogens and the mechanistic basis for their effects. In this review, we will discuss the clinical and experimental evidence that has begun to reveal which members of the microbiota protect against some of the most troublesome antibiotic-resistant pathogens: Klebsiella pneumoniae, vancomycin-resistant enterococci, and Clostridioides difficile.
Collapse
|
15
|
Akkermansia muciniphila uses human milk oligosaccharides to thrive in the early life conditions in vitro. Sci Rep 2020; 10:14330. [PMID: 32868839 PMCID: PMC7459334 DOI: 10.1038/s41598-020-71113-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/10/2020] [Indexed: 02/07/2023] Open
Abstract
Akkermansia muciniphila is a well-studied anaerobic bacterium specialized in mucus degradation and associated with human health. Because of the structural resemblance of mucus glycans and free human milk oligosaccharides (HMOs), we studied the ability of A. muciniphila to utilize human milk oligosaccharides. We found that A. muciniphila was able to grow on human milk and degrade HMOs. Analyses of the proteome of A. muciniphila indicated that key-glycan degrading enzymes were expressed when the bacterium was grown on human milk. Our results display the functionality of the key-glycan degrading enzymes (α-l-fucosidases, β-galactosidases, exo-α-sialidases and β-acetylhexosaminidases) to degrade the HMO-structures 2′-FL, LNT, lactose, and LNT2. The hydrolysation of the host-derived glycan structures allows A. muciniphila to promote syntrophy with other beneficial bacteria, contributing in that way to a microbial ecological network in the gut. Thus, the capacity of A. muciniphila to utilize human milk will enable its survival in the early life intestine and colonization of the mucosal layer in early life, warranting later life mucosal and metabolic health.
Collapse
|
16
|
Stoffel MA, Acevedo-Whitehouse K, Morales-Durán N, Grosser S, Chakarov N, Krüger O, Nichols HJ, Elorriaga-Verplancken FR, Hoffman JI. Early sexual dimorphism in the developing gut microbiome of northern elephant seals. Mol Ecol 2020; 29:2109-2122. [PMID: 32060961 DOI: 10.1111/mec.15385] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/13/2022]
Abstract
The gut microbiome is an integral part of a species' ecology, but we know little about how host characteristics impact its development in wild populations. Here, we explored the role of such intrinsic factors in shaping the gut microbiome of northern elephant seals (Mirounga angustirostris) during a critical developmental window of 6 weeks after weaning, when the pups stay ashore without feeding. We found substantial sex differences in the early-life gut microbiome, even though males and females could not yet be distinguished morphologically. Sex and age both explained around 15% of the variation in gut microbial beta diversity, while microbial communities sampled from the same individual showed high levels of similarity across time, explaining another 40% of the variation. Only a small proportion of the variation in beta diversity was explained by health status, assessed by full blood counts, but clinically healthy individuals had a greater microbial alpha diversity than their clinically abnormal peers. Across the post-weaning period, the northern elephant seal gut microbiome was highly dynamic. We found evidence for several colonization and extinction events as well as a decline in Bacteroides and an increase in Prevotella, a pattern that has previously been associated with the transition from nursing to solid food. Lastly, we show that genetic relatedness was correlated with gut microbiome similarity in males but not females, again reflecting early sex differences. Our study represents a naturally diet-controlled and longitudinal investigation of how intrinsic factors shape the early gut microbiome in a species with extreme sex differences in morphology and life history.
Collapse
Affiliation(s)
- Martin A Stoffel
- Department of Animal Behaviour, Bielefeld University, Bielefeld, Germany.,School of Natural Sciences and Psychology, Faculty of Science, Liverpool John Moores University, Liverpool, UK.,Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, UK
| | - Karina Acevedo-Whitehouse
- Unit for Basic and Applied Microbiology, School of Natural Sciences, Autonomous University of Queretaro, Queretaro, México.,The Marine Mammal Center, Sausalito, CA, USA
| | - Nami Morales-Durán
- Unit for Basic and Applied Microbiology, School of Natural Sciences, Autonomous University of Queretaro, Queretaro, México
| | - Stefanie Grosser
- Division of Evolutionary Biology, Faculty of Biology, LMU Munich, Planegg-Martinsried, Germany
| | - Nayden Chakarov
- Department of Animal Behaviour, Bielefeld University, Bielefeld, Germany
| | - Oliver Krüger
- Department of Animal Behaviour, Bielefeld University, Bielefeld, Germany
| | - Hazel J Nichols
- Department of Animal Behaviour, Bielefeld University, Bielefeld, Germany.,Department of Biosciences, College of Science, Swansea University, Swansea, UK
| | - Fernando R Elorriaga-Verplancken
- Departamento de Pesquerías y Biología Marina, Centro Interdisciplinario de Ciencias Marinas (CICIMAR-IPN), Instituto Politécnico Nacional, La Paz, Mexico
| | - Joseph I Hoffman
- Department of Animal Behaviour, Bielefeld University, Bielefeld, Germany.,British Antarctic Survey, Cambridge, UK
| |
Collapse
|
17
|
The Gut Microbiota in Cardiovascular Disease and Arterial Thrombosis. Microorganisms 2019; 7:microorganisms7120691. [PMID: 31847071 PMCID: PMC6956001 DOI: 10.3390/microorganisms7120691] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 12/16/2022] Open
Abstract
The gut microbiota has emerged as a contributing factor in the development of atherosclerosis and arterial thrombosis. Metabolites from the gut microbiota, such as trimethylamine N-oxide and short chain fatty acids, were identified as messengers that induce cell type-specific signaling mechanisms and immune reactions in the host vasculature, impacting the development of cardiovascular diseases. In addition, microbial-associated molecular patterns drive atherogenesis and the microbiota was recently demonstrated to promote arterial thrombosis through Toll-like receptor signaling. Furthermore, by the use of germ-free mouse models, the presence of a gut microbiota was shown to influence the synthesis of endothelial adhesion molecules. Hence, the gut microbiota is increasingly being recognized as an influencing factor of arterial thrombosis and attempts of dietary pre- or probiotic modulation of the commensal microbiota, to reduce cardiovascular risk, are becoming increasingly significant.
Collapse
|
18
|
Wang H, Zhai N, Chen Y, Fu C, Huang K. OTA induces intestinal epithelial barrier dysfunction and tight junction disruption in IPEC-J2 cells through ROS/Ca 2+-mediated MLCK activation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 242:106-112. [PMID: 29966834 DOI: 10.1016/j.envpol.2018.06.062] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/23/2018] [Accepted: 06/20/2018] [Indexed: 06/08/2023]
Abstract
Ochratoxin A (OTA) is a frequent contaminant of feed and food worldwide. The toxicity of OTA on intestinal barrier was investigated in porcine intestinal epithelial cells (IPEC-J2). We observed that OTA induced intestinal barrier dysfunction as indicated by the reduction in transepithelial electrical resistance (TEER) and elevation in paracellular permeability to 4 kDa dextran. The barrier dysfunction was accompanied with tight junction disruption including a down-regulation in ZO-1 expression and redistribution of Occludin and ZO-1. Moreover, OTA exposure increased reactive oxygen species (ROS) generation, elevated the intracellular calcium level ([Ca2+]c) and activated myosin light chain kinase (MLCK). Simultaneously, NAC, a ROS scavenger, blocked OTA-induced ROS generation, [Ca2+]c elevation, barrier dysfunction and tight junction disruption, suggesting that OTA-induced ROS generation may act as a trigger. Next, we found that OTA-induced MLCK activation was inhibited by BAPTA-AM, the cytosolic Ca2+ chelator, demonstrating that OTA-induced MLCK activation is dependent on [Ca2+]c elevation. Furthermore, inhibition of MLCK with ML-7 or inhibition of [Ca2+]c elevation with BAPTA-AM markedly prevented OTA-induced barrier dysfunction and tight junction disruption. Taken together, our results indicated that OTA induces ROS generation, and then elevates the [Ca2+]c and MLCK activity in turn, which finally induces barrier dysfunction and disrupts tight junction in IPEC-J2 cell monolayers.
Collapse
Affiliation(s)
- Hong Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Nianhui Zhai
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Ying Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Chongyang Fu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
19
|
Bar Shira E, Friedman A. Innate immune functions of avian intestinal epithelial cells: Response to bacterial stimuli and localization of responding cells in the developing avian digestive tract. PLoS One 2018; 13:e0200393. [PMID: 29979771 PMCID: PMC6034880 DOI: 10.1371/journal.pone.0200393] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022] Open
Abstract
Intestinal epithelial cells are multi-tasked cells that participate in digestion and absorption as well as in protection of the digestive tract. While information on the physiology and immune functions of intestinal epithelial cells in mammals is abundant, little is known of their immune function in birds and other species. Our main objectives were to study the development of anti-bacterial innate immune functions in the rapidly developing gut of the pre- and post-hatch chick and to determine the functional diversity of epithelial cells. After establishing primary intestinal epithelial cell cultures, we demonstrated their capacity to uptake and process bacteria. The response to bacterial products, LPS and LTA, induced expression of pro-inflammatory cytokine genes (IL-6, IL-18) as well as the expression of the acute phase proteins avidin, lysozyme and the secretory component derived from the polymeric immunoglobulin receptor. These proteins were then localized in gut sections, and the goblet cell was shown to store avidin, lysozyme as well as secretory component. Lysozyme staining was also located in a novel rod-shaped intestinal cell, situated at different loci along the villus, thus deviating from the classical Paneth cell in the mammal, that is restricted to crypts. Thus, in the chicken, the intestinal epithelium, and particularly goblet cells, are committed to innate immune protection. The unique role of the goblet cell in chicken intestinal immunity, as well as the unique distribution of lysozyme-positive cells highlight alternative solutions of gut protection in the bird.
Collapse
Affiliation(s)
- Enav Bar Shira
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Aharon Friedman
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- * E-mail:
| |
Collapse
|
20
|
Harbeson D, Ben-Othman R, Amenyogbe N, Kollmann TR. Outgrowing the Immaturity Myth: The Cost of Defending From Neonatal Infectious Disease. Front Immunol 2018; 9:1077. [PMID: 29896192 PMCID: PMC5986917 DOI: 10.3389/fimmu.2018.01077] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/30/2018] [Indexed: 12/28/2022] Open
Abstract
Newborns suffer high rates of mortality due to infectious disease-this has been generally regarded to be the result of an "immature" immune system with a diminished disease-fighting capacity. However, the immaturity dogma fails to explain (i) greater pro-inflammatory responses than adults in vivo and (ii) the ability of neonates to survive a significantly higher blood pathogen burden than of adults. To reconcile the apparent contradiction of clinical susceptibility to disease and the host immune response findings when contrasting newborn to adult, it will be essential to capture the entirety of available host-defense strategies at the newborn's disposal. Adults focus heavily on the disease resistance approach: pathogen reduction and elimination. Newborn hyperactive innate immunity, sensitivity to immunopathology, and the energetic requirements of growth and development (immune and energy costs), however, preclude them from having an adult-like resistance response. Instead, newborns also may avail themselves of disease tolerance (minimizing immunopathology without reducing pathogen load), as a disease tolerance approach provides a counterbalance to the dangers of a heightened innate immunity and has lower-associated immune costs. Further, disease tolerance allows for the establishment of a commensal bacterial community without mounting an unnecessarily dangerous immune resistance response. Since disease tolerance has its own associated costs (immune suppression leading to unchecked pathogen proliferation), it is the maintenance of homeostasis between disease tolerance and disease resistance that is critical to safe and effective defense against infections in early life. This paradigm is consistent with nearly all of the existing evidence.
Collapse
Affiliation(s)
- Danny Harbeson
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rym Ben-Othman
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Nelly Amenyogbe
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tobias R. Kollmann
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Vaziri F, Tarashi S, Fateh A, Siadat SD. New insights of Helicobacter pylori host-pathogen interactions: The triangle of virulence factors, epigenetic modifications and non-coding RNAs. World J Clin Cases 2018; 6:64-73. [PMID: 29774218 PMCID: PMC5955730 DOI: 10.12998/wjcc.v6.i5.64] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 02/09/2018] [Accepted: 03/07/2018] [Indexed: 02/05/2023] Open
Abstract
Helicobacter pylori (H. pylori) is a model organism for understanding host-pathogen interactions and infection-mediated carcinogenesis. Gastric cancer and H. pylori colonization indicates the strong correlation. The progression and exacerbation of H. pylori infection are influenced by some factors of pathogen and host. Several virulence factors involved in the proper adherence and attenuation of immune defense to contribute the risk of emerging gastric cancer, therefore analysis of them is very important. H. pylori also modulates inflammatory and autophagy process to intensify its pathogenicity. From the host regard, different genetic factors particularly affect the development of gastric cancer. Indeed, epigenetic modifications, MicroRNA and long non-coding RNA received more attention. Generally, various factors related to pathogen and host that modulate gastric cancer development in response to H. pylori need more attention due to develop an efficacious therapeutic intervention. Therefore, this paper will present a brief overview of host-pathogen interaction especially emphases on bacterial virulence factors, interruption of host cellular signaling, the role of epigenetic modifications and non-coding RNAs.
Collapse
Affiliation(s)
- Farzam Vaziri
- Microbiology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Samira Tarashi
- Microbiology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Abolfazl Fateh
- Microbiology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Seyed Davar Siadat
- Microbiology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran 1316943551, Iran
| |
Collapse
|
22
|
Nawaz A, Bakhsh Javaid A, Irshad S, Hoseinifar SH, Xiong H. The functionality of prebiotics as immunostimulant: Evidences from trials on terrestrial and aquatic animals. FISH & SHELLFISH IMMUNOLOGY 2018; 76:272-278. [PMID: 29510254 DOI: 10.1016/j.fsi.2018.03.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/21/2018] [Accepted: 03/02/2018] [Indexed: 05/24/2023]
Abstract
The gut immune system is, the main option for maintaining host's health, affected by numerous factors comprising dietary constituents and commensal bacteria. These dietary components that affect the intestinal immunity and considered as an alternative of antibiotics are called immunosaccharides. Fructooligosaccharide (FOS), Galactooligosaccharide (GOS), inulin, dietary carbohydrates, and xylooligosaccharide (XOS) are among the most studied prebiotics in human as well as in aquaculture. Although prebiotics and probiotics have revealed potential as treatment for numerous illnesses in both human and fish, a comprehensive understanding of the molecular mechanism behind direct and indirect effect on the intestinal immune response will help more and perhaps extra effective therapy intended for ailments. This review covers the most newly deep-rooted scientific outcomes about the direct and indirect mechanism through which these dietetic strategies can affect intestinal immunity of terrestrial and aquatic animals. Prebiotics exert an influence on gut immune system via the increase in lysozyme and phagocytic activity, macrophage activation and stimulation of monocyte-derived dendritic cells. Furthermore, these functional molecules also enhance epithelial barrier function, beneficial gut microbial population, and production of intermediate metabolites for example short chain fatty acids (SCFAs) that assist in balancing the immune system. Moreover, emphasis will be sited on the relationship among food/feed, the microbiota, and the gut immune system. In conclusion, further studies are nonetheless essential to confirm the direct effect of prebiotics on immune response.
Collapse
Affiliation(s)
- Asad Nawaz
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Allah Bakhsh Javaid
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Sana Irshad
- School of Environmental Studies, China University of Geosciences, Wuhan 430070, China
| | - Seyed Hossein Hoseinifar
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Hanguo Xiong
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
23
|
Action and function of Akkermansia muciniphila in microbiome ecology, health and disease. Best Pract Res Clin Gastroenterol 2017; 31:637-642. [PMID: 29566906 DOI: 10.1016/j.bpg.2017.10.001] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/22/2017] [Accepted: 10/10/2017] [Indexed: 01/31/2023]
Abstract
The discovery of Akkermansia muciniphila has opened new avenues for the use of this abundant intestinal symbiont in next generation therapeutic products, as well as targeting microbiota dynamics. A. muciniphila is known to colonize the mucosal layer of the human intestine where it triggers both host metabolic and immune responses. A. muciniphila is particularly effective in increasing mucus thickness and increasing gut barrier function. As a result host metabolic markers ameliorate. The mechanism of host regulation is thought to involve the outer membrane composition, including the type IV pili of A. muciniphila, that directly signal to host immune receptors. At the same time the metabolic activity of A. muciniphila leads to the production of short chain fatty acids that are beneficial to the host and microbiota members. This contributes to host-microbiota and microbe-microbe syntrophy The mucolytic activity and metabolite production make A. muciniphila a key species in the mucus layer, stimulating beneficial mucosal microbial networks. This well studied member of the microbiota has been studied in three aspects that will be further described in this review: i) A. muciniphila characteristics and mucin adaptation, ii) its role as key species in the mucosal microbiome, and iii) its role in host health.
Collapse
|
24
|
Duval M, Cossart P, Lebreton A. Mammalian microRNAs and long noncoding RNAs in the host-bacterial pathogen crosstalk. Semin Cell Dev Biol 2017; 65:11-19. [PMID: 27381344 PMCID: PMC7089780 DOI: 10.1016/j.semcdb.2016.06.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/30/2016] [Accepted: 06/01/2016] [Indexed: 12/20/2022]
Abstract
Gene expression regulation is a critical question in host-pathogen interactions, and RNAs act as key players in this process. In this review, we focus on the mammalian RNA response to bacterial infection, with a special interest on microRNAs and long non-coding RNAs. We discuss the role of cellular miRNAs in immunity, the implication of circulating miRNAs as well as the influence of the microbiome on the miRNA response. We also review how pathogens counteract the host miRNA expression. Interestingly, bacterial non-coding RNAs regulate host gene expression and conversely eukaryotic miRNAs may regulate bacterial gene expression. Overall, the characterization of RNA regulatory networks represents an emerging theme in the field of host pathogen interactions.
Collapse
Affiliation(s)
- Mélodie Duval
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, USC2020, 75015 Paris, France
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, USC2020, 75015 Paris, France.
| | - Alice Lebreton
- École Normale Supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'École Normale Supérieure (IBENS), Équipe Infection et Devenir de l'ARN, 75005 Paris, France; INRA, IBENS, 75005 Paris, France.
| |
Collapse
|
25
|
Xu L, Fan Q, Zhuang Y, Wang Q, Gao Y, Wang C. Bacillus Coagulans Enhance the Immune Function of the Intestinal Mucosa of Yellow Broilers. ACTA ACUST UNITED AC 2017. [DOI: 10.1590/1806-9061-2015-0180] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- L Xu
- Fujian Agriculture and Forestry University, China
| | - Q Fan
- Fujian Agriculture and Forestry University, China
| | - Y Zhuang
- Fujian Agriculture and Forestry University, China; Fujian Agriculture and Forestry University, China
| | - Q Wang
- Fujian Agriculture and Forestry University, China; Fujian Agriculture and Forestry University, China
| | - Y Gao
- Fujian Agriculture and Forestry University, China
| | - C Wang
- Fujian Agriculture and Forestry University, China
| |
Collapse
|
26
|
Gravato-Nobre MJ, Vaz F, Filipe S, Chalmers R, Hodgkin J. The Invertebrate Lysozyme Effector ILYS-3 Is Systemically Activated in Response to Danger Signals and Confers Antimicrobial Protection in C. elegans. PLoS Pathog 2016; 12:e1005826. [PMID: 27525822 PMCID: PMC4985157 DOI: 10.1371/journal.ppat.1005826] [Citation(s) in RCA: 27] [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: 03/04/2016] [Accepted: 07/25/2016] [Indexed: 12/17/2022] Open
Abstract
Little is known about the relative contributions and importance of antibacterial effectors in the nematode C. elegans, despite extensive work on the innate immune responses in this organism. We report an investigation of the expression, function and regulation of the six ilys (invertebrate-type lysozyme) genes of C. elegans. These genes exhibited a surprising variety of tissue-specific expression patterns and responses to starvation or bacterial infection. The most strongly expressed, ilys-3, was investigated in detail. ILYS-3 protein was expressed constitutively in the pharynx and coelomocytes, and dynamically in the intestine. Analysis of mutants showed that ILYS-3 was required for pharyngeal grinding (disruption of bacterial cells) during normal growth and consequently it contributes to longevity, as well as being protective against bacterial pathogens. Both starvation and challenge with Gram-positive pathogens resulted in ERK-MAPK-dependent up-regulation of ilys-3 in the intestine. The intestinal induction by pathogens, but not starvation, was found to be dependent on MPK-1 activity in the pharynx rather than in the intestine, demonstrating unexpected communication between these two tissues. The coelomocyte expression appeared to contribute little to normal growth or immunity. Recombinant ILYS-3 protein was found to exhibit appropriate lytic activity against Gram-positive cell wall material. Innate immune defenses against bacterial pathogenesis depend on the activation of antibacterial factors. We examined the expression and relative importance of a gene family encoding six invertebrate-type lysozymes in the much-studied nematode C. elegans. The ilys genes exhibit distinct patterns of tissue-specific expression and response to pathogenic challenge and/or starvation. The most abundantly expressed, ilys-3, exhibits constitutive pharyngeal expression, which we show is essential for efficient disruption of bacteria under non-pathogenic growth conditions, and consequently it contributes to normal longevity. ilys-3 is also strongly up-regulated in intestinal cells after starvation or exposure to Gram-positive pathogens such as Microbacterium nematophilum and acts as a ‘slow-effector’ in limiting pathogenic damage from intestinal infections. We show that this induction by pathogens depends on the action of an ERK-MAPK cascade, which acts in pharyngeal rather than intestinal cells; this implies communication between pharynx and intestine. Tagged ILYS-3 protein was detected mainly in recycling endosomes of intestinal cells and in the intestinal lumen after starvation. ILYS-3 was also expressed in coelomocytes (scavenger cells) but we found that these cells make little or no contribution to defense. We examined the enzymatic properties of recombinant ILYS-3 protein, finding that it has lytic activity against M. nematophilum cell-walls.
Collapse
Affiliation(s)
| | - Filipa Vaz
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Sergio Filipe
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Ronald Chalmers
- Laboratory of Bacterial Cell Surfaces and Pathogenesis, Instituto de Tecnologia Química e Biológica and Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Jonathan Hodgkin
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Stanifer ML, Rippert A, Kazakov A, Willemsen J, Bucher D, Bender S, Bartenschlager R, Binder M, Boulant S. Reovirus intermediate subviral particles constitute a strategy to infect intestinal epithelial cells by exploiting TGF-β dependent pro-survival signaling. Cell Microbiol 2016; 18:1831-1845. [PMID: 27279006 DOI: 10.1111/cmi.12626] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/02/2016] [Accepted: 06/02/2016] [Indexed: 12/24/2022]
Abstract
Intestinal epithelial cells (IECs) constitute the primary barrier that separates us from the outside environment. These cells, lining the surface of the intestinal tract, represent a major challenge that enteric pathogens have to face. How IECs respond to viral infection and whether enteric viruses have developed strategies to subvert IECs innate immune response remains poorly characterized. Using mammalian reovirus (MRV) as a model enteric virus, we found that the intermediate subviral particles (ISVPs), which are formed in the gut during the natural course of infection by proteolytic digestion of the reovirus virion, trigger reduced innate antiviral immune response in IECs. On the contrary, infection of IECs by virions induces a strong antiviral immune response that leads to cellular death. Additionally, we determined that virions can be sensed by both TLR and RLR pathways while ISVPs are sensed by RLR pathways only. Interestingly, we found that ISVP infected cells secrete TGF-β acting as a pro-survival factor that protects IECs against virion induced cellular death. We propose that ISVPs represent a reovirus strategy to initiate primary infection of the gut by subverting IECs innate immune system and by counteracting cellular-death pathways.
Collapse
Affiliation(s)
- Megan L Stanifer
- Schaller research group at CellNetworks, Department of Infectious Diseases, Virology, Heidelberg University, Germany
| | - Anja Rippert
- Schaller research group at CellNetworks, Department of Infectious Diseases, Virology, Heidelberg University, Germany
| | - Alexander Kazakov
- Schaller research group at CellNetworks, Department of Infectious Diseases, Virology, Heidelberg University, Germany
| | - Joschka Willemsen
- Research Group 'Dynamics of early viral infection and the innate antiviral response'.,Division Virus-associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Delia Bucher
- Schaller research group at CellNetworks, Department of Infectious Diseases, Virology, Heidelberg University, Germany
| | - Silke Bender
- Division Virus-associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralf Bartenschlager
- Division Virus-associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marco Binder
- Research Group 'Dynamics of early viral infection and the innate antiviral response'.,Division Virus-associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steeve Boulant
- Schaller research group at CellNetworks, Department of Infectious Diseases, Virology, Heidelberg University, Germany.,Research Group 'Cellular polarity and viral infection' (F140), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
28
|
Tremellen K. Gut Endotoxin Leading to a Decline IN Gonadal function (GELDING) - a novel theory for the development of late onset hypogonadism in obese men. Basic Clin Androl 2016; 26:7. [PMID: 27340554 PMCID: PMC4918028 DOI: 10.1186/s12610-016-0034-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/24/2016] [Indexed: 12/25/2022] Open
Abstract
Obesity is an increasing public health problem, with two-thirds of the adult population in many Western countries now being either overweight or obese. Male obesity is associated with late onset hypogonadism, a condition characterised by decreased serum testosterone, sperm quality plus diminished fertility and quality of life. In this paper we propose a novel theory underlying the development of obesity related hypogonadism- the GELDING theory (Gut Endotoxin Leading to a Decline IN Gonadal function). Several observational studies have previously reported an association between obesity related hypogonadism (low testosterone) and systemic inflammation. However, for the first time we postulate that the trans-mucosal passage of bacterial lipopolysaccharide (LPS) from the gut lumen into the circulation is a key inflammatory trigger underlying male hypogonadism. Obesity and a high fat/high calorie diet are both reported to result in changes to gut bacteria and intestinal wall permeability, leading to the passage of bacterial endotoxin (lipopolysaccharide- LPS) from within the gut lumen into the circulation (metabolic endotoxaemia), where it initiates systemic inflammation. Endotoxin is known to reduce testosterone production by the testis, both by direct inhibition of Leydig cell steroidogenic pathways and indirectly by reducing pituitary LH drive, thereby also leading to a decline in sperm production. In this paper we also highlight the novel evolutionary benefits of the GELDING theory. Testosterone is known to be a powerful immune-suppressive, decreasing a man's ability to fight infection. Therefore we postulate that the male reproductive axis has evolved the capacity to lower testosterone production during times of infection and resulting endotoxin exposure, decreasing the immunosuppressive influence of testosterone, in turn enhancing the ability to fight infection. While this response is adaptive in times of sepsis, it becomes maladaptive in the setting of "non-infectious" obesity related metabolic endotoxaemia.
Collapse
Affiliation(s)
- Kelton Tremellen
- Department of Obstetrics, Gynaecology and Reproductive Medicine, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
29
|
Newburg DS, Ko JS, Leone S, Nanthakumar NN. Human Milk Oligosaccharides and Synthetic Galactosyloligosaccharides Contain 3'-, 4-, and 6'-Galactosyllactose and Attenuate Inflammation in Human T84, NCM-460, and H4 Cells and Intestinal Tissue Ex Vivo. J Nutr 2016; 146:358-67. [PMID: 26701795 PMCID: PMC4725434 DOI: 10.3945/jn.115.220749] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 11/16/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The immature intestinal mucosa responds excessively to inflammatory insult, but human milk protects infants from intestinal inflammation. The ability of galactosyllactoses [galactosyloligosaccharides (GOS)], newly found in human milk oligosaccharides (HMOS), to suppress inflammation was not known. OBJECTIVE The objective was to test whether GOS can directly attenuate inflammation and to explore the components of immune signaling modulated by GOS. METHODS Galactosyllactose composition was measured in sequential human milk samples from days 1 through 21 of lactation and in random colostrum samples from 38 mothers. Immature [human normal fetal intestinal epithelial cell (H4)] and mature [human metastatic colonic epithelial cell (T84) and human normal colon mucosal epithelial cell (NCM-460)] enterocyte cell lines were treated with the pro-inflammatory molecules tumor necrosis factor-α (TNF-α) or interleukin-1β (IL-1β) or infected with Salmonella or Listeria. The inflammatory response was measured as induction of IL-8, monocyte chemoattractant protein 1 (MCP-1), or macrophage inflammatory protein-3α (MIP-3α) protein by ELISA and mRNA by quantitative reverse transcriptase-polymerase chain reaction. The ability of HMOS or synthetic GOS to attenuate this inflammation was tested in vitro and in immature human intestinal tissue ex vivo. RESULTS The 3 galactosyllactoses (3'-GL, 4-GL, and 6'-GL) expressed in colostrum rapidly declined over early lactation (P < 0.05). In H4 cells, HMOS attenuated TNF-α- and IL-1β-induced expression of IL-8, MIP-3α, and MCP-1 to 48-51% and pathogen-induced IL-8 and MCP-1 to 26-30% of positive controls (P < 0.001). GOS reduced TNF-α- and IL-1β-induced inflammatory responses to 25-26% and pathogen-induced IL-8 and MCP-1 to 36-39% of positive controls (P < 0.001). GOS and HMOS mitigated nuclear translocation of nuclear transcription factor κB (NF-κB) p65. HMOS quenched the inflammatory response to Salmonella infection by immature human intestinal tissue ex vivo to 26% and by GOS to 50% of infected controls (P < 0.01). CONCLUSION Galactosyllactose attenuated NF-κB inflammatory signaling in human intestinal epithelial cells and in human immature intestine. Thus, galactosyllactoses are strong physiologic anti-inflammatory agents in human colostrum and early milk, contributing to innate immune modulation. The potential clinical utility of galactosyllactose warrants investigation.
Collapse
Affiliation(s)
- David S Newburg
- Program in Glycobiology, Department of Biology, Boston College, Chestnut Hill, MA;
| | - Jae Sung Ko
- Department of Pediatrics, Seoul National University Children’s Hospital, Jongno-gu, Seoul, Korea; and
| | - Serena Leone
- Program in Glycobiology, Department of Biology, Boston College, Chestnut Hill, MA
| | - N Nanda Nanthakumar
- Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA
| |
Collapse
|
30
|
Addis MF, Tanca A, Uzzau S, Oikonomou G, Bicalho RC, Moroni P. The bovine milk microbiota: insights and perspectives from -omics studies. MOLECULAR BIOSYSTEMS 2016; 12:2359-72. [DOI: 10.1039/c6mb00217j] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent findings and future perspectives of -omics studies on the bovine milk microbiota, focusing on its impact on animal health.
Collapse
Affiliation(s)
- M. F. Addis
- Porto Conte Ricerche
- SP 55 Porto Conte/Capo Caccia
- 07041 Alghero
- Italy
| | - A. Tanca
- Porto Conte Ricerche
- SP 55 Porto Conte/Capo Caccia
- 07041 Alghero
- Italy
| | - S. Uzzau
- Porto Conte Ricerche
- SP 55 Porto Conte/Capo Caccia
- 07041 Alghero
- Italy
- Università degli Studi di Sassari
| | - G. Oikonomou
- Epidemiology and Population Health
- Institute of Infection and Global Health
- University of Liverpool
- Liverpool
- UK
| | - R. C. Bicalho
- Cornell University
- Department of Population Medicine and Diagnostic Sciences
- College of Veterinary Medicine
- Ithaca
- USA
| | - P. Moroni
- Cornell University
- Department of Population Medicine and Diagnostic Sciences
- College of Veterinary Medicine
- Ithaca
- USA
| |
Collapse
|
31
|
Liu YW, Fu TY, Peng WS, Chen YH, Cao YM, Chen CC, Hung WL, Tsai YC. Evaluation of the potential anti-allergic effects of heat-inactivated Lactobacillus paracasei V0151 in vitro, ex vivo, and in vivo. Benef Microbes 2015; 6:697-705. [PMID: 26192907 DOI: 10.3920/bm2014.0159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The efficacy of Lactobacillus paracasei V0151 (V0151), isolated from the faeces of a child, to modulate immune responses was investigated. In RAW 264.7 cells expressing an inducible nitric oxide synthase (iNOS)-directed luciferase gene, heat-inactivated V0151 stimulated iNOS expression followed by nitric oxide production. V0151 significantly elevated interferon gamma, interleukin (IL)-10, tumour necrosis factor alpha, and IL-1β production in human peripheral blood mononuclear cells. In splenocytes isolated from ovalbumin (OVA)-sensitised BALB/c mice treated with OVA and V0151 at different bacterium-to-cell ratios (1:1, 10:1, and 20:1) for 96 h, IL-2, IL-4, IL-5, and IL-13 production was dose-dependently downregulated, whereas IL-12 was dose-dependently upregulated. Collectively, our findings indicate that V0151 might regulate pro-inflammatory factors in macrophages and splenocytes. Furthermore, the T helper 1/T helper 2 (Th1/Th2) balance was also skewed toward Th1 dominance through the elevation of Th1 cytokine production.
Collapse
Affiliation(s)
- Y W Liu
- 1 Institute of Biochemistry and Molecular Biology, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, BeiTou Dist., Taipei 11221, Taiwan, R.O.C
| | - T Y Fu
- 1 Institute of Biochemistry and Molecular Biology, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, BeiTou Dist., Taipei 11221, Taiwan, R.O.C.,2 Probiotics Research Center, National Yang-Ming University, No. 155, Li-Nong St., Section 2, BeiTou Dist., Taipei 11221, Taiwan, R.O.C
| | - W S Peng
- 1 Institute of Biochemistry and Molecular Biology, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, BeiTou Dist., Taipei 11221, Taiwan, R.O.C.,2 Probiotics Research Center, National Yang-Ming University, No. 155, Li-Nong St., Section 2, BeiTou Dist., Taipei 11221, Taiwan, R.O.C
| | - Y H Chen
- 1 Institute of Biochemistry and Molecular Biology, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, BeiTou Dist., Taipei 11221, Taiwan, R.O.C.,2 Probiotics Research Center, National Yang-Ming University, No. 155, Li-Nong St., Section 2, BeiTou Dist., Taipei 11221, Taiwan, R.O.C
| | - Y M Cao
- 3 Want Want China Holdings Ltd., No.1088, East Hond Song Rd., Shanghai 201103, China P.R
| | - C C Chen
- 3 Want Want China Holdings Ltd., No.1088, East Hond Song Rd., Shanghai 201103, China P.R
| | - W L Hung
- 3 Want Want China Holdings Ltd., No.1088, East Hond Song Rd., Shanghai 201103, China P.R
| | - Y C Tsai
- 1 Institute of Biochemistry and Molecular Biology, National Yang-Ming University, No. 155, Li-Nong St., Sec. 2, BeiTou Dist., Taipei 11221, Taiwan, R.O.C.,2 Probiotics Research Center, National Yang-Ming University, No. 155, Li-Nong St., Section 2, BeiTou Dist., Taipei 11221, Taiwan, R.O.C
| |
Collapse
|
32
|
Sellin ME, Maslowski KM, Maloy KJ, Hardt WD. Inflammasomes of the intestinal epithelium. Trends Immunol 2015; 36:442-50. [PMID: 26166583 DOI: 10.1016/j.it.2015.06.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 06/16/2015] [Accepted: 06/16/2015] [Indexed: 02/08/2023]
Abstract
While the functional importance of inflammasomes in blood-derived cell types is well established, it remains poorly understood how inflammasomes in nonhematopoietic cells contribute to mucosal immunity. Recent studies have revealed functional roles of inflammasomes - particularly NAIP/NLRC4, NLRP6, and noncanonical caspase-4 (caspase-11) - within epithelial cells of the gut in mucosal immune defense, inflammation, and tumorigenesis. Here, we review and discuss these findings in the broader context of tissue compartment-specific mucosal immunity. We propose several models whereby activities of the intestinal epithelial inflammasomes converge on mechanisms to remove compromised epithelial cells, maintain host-microbiota mutualism, and communicate with immune cells of the underlying lamina propria.
Collapse
Affiliation(s)
- Mikael E Sellin
- Institute of Microbiology, ETH Zürich, 8093 Zürich, Switzerland.
| | - Kendle M Maslowski
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | | |
Collapse
|
33
|
Leibelt S, Friede ME, Rohe C, Gütle D, Rutkowski E, Weigert A, Kveberg L, Vaage JT, Hornef MW, Steinle A. Dedicated immunosensing of the mouse intestinal epithelium facilitated by a pair of genetically coupled lectin-like receptors. Mucosal Immunol 2015; 8:232-42. [PMID: 24985083 DOI: 10.1038/mi.2014.60] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 06/03/2014] [Indexed: 02/04/2023]
Abstract
The integrity of the intestinal epithelium is constantly surveyed by a peculiar subset of innate-like T lymphocytes embedded in the epithelial cell layer, hence called intestinal intraepithelial lymphocytes (IELs). IELs are thought to act as "first-line" sentinels sensing the state of adjacent epithelial cells via both T-cell receptors and auxiliary receptors. Auxiliary receptors modulating IEL activity include C-type lectin-like receptors encoded in the natural killer gene complex such as NKG2D. Here, we report that the CTLR Nkrp1g is expressed by a subpopulation of mouse CD103(+) IELs allowing immunosensing of the intestinal epithelium through ligation of the genetically coupled CTLR Clr-f that is almost exclusively expressed on differentiated intestinal epithelial cells (IECs). Most of these Nkrp1g-expressing IELs exhibit a γδTCR(bright)Nkg2a(-) phenotype and are intimately associated with the intestinal epithelium. As Clr-f expression strongly inhibits effector functions of Nkrp1g-expressing cells and is upregulated upon poly(I:C) challenge, Clr-f molecules may quench reactivity of these IELs towards the epithelial barrier that is constantly provoked by microbial and antigenic stimuli. Altogether, we here newly characterize a genetically linked C-type lectin-like receptor/ligand pair with a highly restricted tissue expression that apparently evolved to allow for a dedicated immunosurveillance of the mouse intestinal epithelium.
Collapse
Affiliation(s)
- S Leibelt
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - M E Friede
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - C Rohe
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - D Gütle
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - E Rutkowski
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - A Weigert
- Institute for Biochemistry I, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - L Kveberg
- Department of Immunology, Oslo University Hospital, Rikshospitalet and University of Oslo, Oslo, Norway
| | - J T Vaage
- Department of Immunology, Oslo University Hospital, Rikshospitalet and University of Oslo, Oslo, Norway
| | - M W Hornef
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - A Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
34
|
Wan LYM, Chen ZJ, Shah NP, El-Nezami H. Modulation of Intestinal Epithelial Defense Responses by Probiotic Bacteria. Crit Rev Food Sci Nutr 2015; 56:2628-41. [DOI: 10.1080/10408398.2014.905450] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
35
|
Walker WA, Iyengar RS. Breast milk, microbiota, and intestinal immune homeostasis. Pediatr Res 2015; 77:220-8. [PMID: 25310762 DOI: 10.1038/pr.2014.160] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 07/22/2014] [Indexed: 12/22/2022]
Abstract
Newborns adjust to the extrauterine environment by developing intestinal immune homeostasis. Appropriate initial bacterial colonization is necessary for adequate intestinal immune development. An environmental determinant of adequate colonization is breast milk. Although the full-term infant is developmentally capable of mounting an immune response, the effector immune component requires bacterial stimulation. Breast milk stimulates the proliferation of a well-balanced and diverse microbiota, which initially influences a switch from an intrauterine TH2 predominant to a TH1/TH2 balanced response and with activation of T-regulatory cells by breast milk-stimulated specific organisms (Bifidobacteria, Lactobacillus, and Bacteroides). As an example of its effect, oligosaccharides in breast milk are fermented by colonic bacteria producing an acid milieu for bacterial proliferation. In addition, short-chain fatty acids in breast milk activate receptors on T-reg cells and bacterial genes, which preferentially mediate intestinal tight junction expression and anti-inflammation. Other components of breast milk (defensins, lactoferrin, etc.) inhibit pathogens and further contribute to microbiota composition. The breast milk influence on initial intestinal microbiota also prevents expression of immune-mediated diseases (asthma, inflammatory bowel disease, type 1 diabetes) later in life through a balanced initial immune response, underscoring the necessity of breastfeeding as the first source of nutrition.
Collapse
Affiliation(s)
- W Allan Walker
- Department of Pediatrics, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts
| | - Rajashri Shuba Iyengar
- Department of Pediatrics, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
36
|
Guo X, Tanaka Y, Kondo M. Thymic precursors of TCRαβ(+)CD8αα(+) intraepithelial lymphocytes are negative for CD103. Immunol Lett 2014; 163:40-8. [PMID: 25448708 DOI: 10.1016/j.imlet.2014.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/27/2014] [Accepted: 11/12/2014] [Indexed: 12/26/2022]
Abstract
CD5(+)TCRαβ(+) cells in CD4(-)CD8(-) double negative (DN) thymocytes are generally regarded as the thymic precursors of TCRαβ(+)CD8αα(+) intestinal intraepithelial lymphocytes (IELs). However, this population is not homogenous and can be subdivided based on the expression of cell surface markers such as CD103. In this study, we aimed to define a cell population that is enriched in thymic IEL precursors. Here we report that only CD103(-) but not CD103(+)cells in the CD5(+)TCRαβ(+) DN thymocyte population can give rise to TCRαβ(+)CD8αα(+) IELs or IEL-like cells in in vivo injections and in vitro cultures, respectively. In addition, we demonstrate that IL-15 stimulation alone is sufficient for upregulation of CD8αα in CD103(-)CD5(+)TCRαβ(+) DN thymocytes. We also found that the CD103(-)CD5(+)TCRαβ(+) DN population can be further separated into two fractions: CD69(-/lo) and CD69(+). Of these two fractions, only CD69(-/lo) cells can give rise to CD8αα IEL-like cells in the presence of IL-15 in in vitro cultures. Based on these results, we conclude that a CD69(-/lo)CD103(-)CD5(+)TCRαβ(+) DN population is highly enriched in thymic IEL precursors.
Collapse
Affiliation(s)
- Xianghua Guo
- Department of Molecular Immunology, Toho University School of Medicine, Tokyo, Japan; Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Yuriko Tanaka
- Department of Molecular Immunology, Toho University School of Medicine, Tokyo, Japan
| | - Motonari Kondo
- Department of Molecular Immunology, Toho University School of Medicine, Tokyo, Japan.
| |
Collapse
|
37
|
Dishaw LJ, Cannon JP, Litman GW, Parker W. Immune-directed support of rich microbial communities in the gut has ancient roots. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 47:36-51. [PMID: 24984114 PMCID: PMC4146740 DOI: 10.1016/j.dci.2014.06.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 05/30/2014] [Accepted: 06/21/2014] [Indexed: 05/12/2023]
Abstract
The animal gut serves as a primary location for the complex host-microbe interplay that is essential for homeostasis and may also reflect the types of ancient selective pressures that spawned the emergence of immunity in metazoans. In this review, we present a phylogenetic survey of gut host-microbe interactions and suggest that host defense systems arose not only to protect tissue directly from pathogenic attack but also to actively support growth of specific communities of mutualists. This functional dichotomy resulted in the evolution of immune systems much more tuned for harmonious existence with microbes than previously thought, existing as dynamic but primarily cooperative entities in the present day. We further present the protochordate Ciona intestinalis as a promising model for studying gut host-bacterial dialogue. The taxonomic position, gut physiology and experimental tractability of Ciona offer unique advantages in dissecting host-microbe interplay and can complement studies in other model systems.
Collapse
Affiliation(s)
- Larry J Dishaw
- Department of Pediatrics, University of South Florida Morsani College of Medicine, USF/ACH Children's Research Institute, 140 7th Avenue South, St. Petersburg, FL 33701, USA.
| | - John P Cannon
- Department of Pediatrics, University of South Florida Morsani College of Medicine, USF/ACH Children's Research Institute, 140 7th Avenue South, St. Petersburg, FL 33701, USA
| | - Gary W Litman
- Department of Pediatrics, University of South Florida Morsani College of Medicine, USF/ACH Children's Research Institute, 140 7th Avenue South, St. Petersburg, FL 33701, USA; Department of Molecular Genetics, All Children's Hospital-Johns Hopkins Medicine, 501 6th Avenue South, St. Petersburg, FL 33701, USA
| | - William Parker
- Department of Surgery, Duke University Medical Center, Box 2605, Durham, NC 27710, USA
| |
Collapse
|
38
|
Abstract
The innate immune response is of prime importance in the immediate recognition and elimination of invading micro-organisms. However, deregulation of this system is clearly associated with the pathogenesis of a wide range of inflammatory diseases. Innate immunity consists of a humoral and a cellular branch, which are closely interacting. An additional level of control is found at the level of neuronal reflexes that can fine-tune these immunological mechanisms.
Collapse
|
39
|
Boudry G, Hamilton M. 35. Milk formula and intestinal barrier function. HUMAN HEALTH HANDBOOKS 2014. [DOI: 10.3920/978-90-8686-223-8_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
40
|
Cooperativity among secretory IgA, the polymeric immunoglobulin receptor, and the gut microbiota promotes host-microbial mutualism. Immunol Lett 2014; 162:10-21. [PMID: 24877874 DOI: 10.1016/j.imlet.2014.05.008] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 05/06/2014] [Accepted: 05/17/2014] [Indexed: 01/01/2023]
Abstract
Secretory IgA (SIgA) antibodies in the intestinal tract form the first line of antigen-specific immune defense, preventing access of pathogens as well as commensal microbes to the body proper. SIgA is transported into external secretions by the polymeric immunoglobulin receptor (pIgR). Evidence is reported here that the gut microbiota regulates production of SIgA and pIgR, which act together to regulate the composition and activity of the microbiota. SIgA in the intestinal mucus layer helps to maintain spatial segregation between the microbiota and the epithelial surface without compromising the metabolic activity of the microbes. Products shed by members of the microbial community promote production of SIgA and pIgR by activating pattern recognition receptors on host epithelial and immune cells. Maternal SIgA in breast milk provides protection to newborn mammals until the developing intestinal immune system begins to produce its own SIgA. Disruption of the SIgA-pIgR-microbial triad can increase the risk of infectious, allergic and inflammatory diseases of the intestine.
Collapse
|
41
|
Abstract
The colon serves as the habitat for trillions of microbes, which it must maintain, regulate, and sequester. This is managed by what is termed the mucosal barrier. The mucosal barrier separates the gut flora from the host tissues; regulates the absorption of water, electrolytes, minerals, and vitamins; and facilitates host-flora interactions. Colonic homeostasis depends on a complex interaction between the microflora and the mucosal epithelium, immune system, vasculature, stroma, and nervous system. Disruptions in the colonic microenvironment such as changes in microbial composition, epithelial cell function/proliferation/differentiation, mucus production/makeup, immune function, diet, motility, or blood flow may have substantial local and systemic consequences. Understanding the complex activities of the colon in health and disease is important in drug development, as xenobiotics can impact all segments of the colon. Direct and indirect effects of pharmaceuticals on intestinal function can produce adverse findings in laboratory animals and humans and can negatively impact drug development. This review will discuss normal colon homeostasis with examples, where applicable, of xenobiotics that disrupt normal function.
Collapse
Affiliation(s)
- Rani S Sellers
- 1Albert Einstein College of Medicine, Bronx, New York, USA
| | | |
Collapse
|
42
|
Vieira AT, Teixeira MM, Martins FS. The role of probiotics and prebiotics in inducing gut immunity. Front Immunol 2013; 4:445. [PMID: 24376446 PMCID: PMC3859913 DOI: 10.3389/fimmu.2013.00445] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/26/2013] [Indexed: 12/13/2022] Open
Abstract
The gut immune system is influenced by many factors, including dietary components and commensal bacteria. Nutrients that affect gut immunity and strategies that restore a healthy gut microbial community by affecting the microbial composition are being developed as new therapeutic approaches to treat several inflammatory diseases. Although probiotics (live microorganisms) and prebiotics (food components) have shown promise as treatments for several diseases in both clinical and animal studies, an understanding of the molecular mechanisms behind the direct and indirect effects on the gut immune response will facilitate better and possibly more efficient therapy for diseases. In this review, we will first describe the concept of prebiotics, probiotics, and symbiotics and cover the most recently well-established scientific findings regarding the direct and indirect mechanisms by which these dietary approaches can influence gut immunity. Emphasis will be placed on the relationship of diet, the microbiota, and the gut immune system. Second, we will highlight recent results from our group, which suggest a new dietary manipulation that includes the use of nutrient products (organic selenium and Lithothamnium muelleri) and probiotics (Saccharomyces boulardii UFMG 905 and Bifidobacterium sp.) that can stimulate and manipulate the gut immune response, inducing intestinal homeostasis. Furthermore, the purpose of this review is to discuss and translate all of this knowledge into therapeutic strategies and into treatment for extra-intestinal compartment pathologies. We will conclude by discussing perspectives and molecular advances regarding the use of prebiotics or probiotics as new therapeutic strategies that manipulate the microbial composition and the gut immune responses of the host.
Collapse
Affiliation(s)
- Angélica T Vieira
- Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil
| | - Mauro M Teixeira
- Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil
| | - Flaviano S Martins
- Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil
| |
Collapse
|
43
|
The intestinal microbiota interferes with the microRNA response upon oral Listeria infection. mBio 2013; 4:e00707-13. [PMID: 24327339 PMCID: PMC3870255 DOI: 10.1128/mbio.00707-13] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The intestinal tract is the largest reservoir of microbes in the human body. The intestinal microbiota is thought to be able to modulate alterations of the gut induced by enteropathogens, thereby maintaining homeostasis. Listeria monocytogenes is the agent of listeriosis, an infection transmitted to humans upon ingestion of contaminated food. Crossing of the intestinal barrier is a critical step of the infection before dissemination into deeper organs. Here, we investigated the role of the intestinal microbiota in the regulation of host protein-coding genes and microRNA (miRNA or miR) expression during Listeria infection. We first established the intestinal miRNA signatures corresponding to the 10 most highly expressed miRNAs in the murine ileum of conventional and germfree mice, noninfected and infected with Listeria. Next, we identified 6 miRNAs whose expression decreased upon Listeria infection in conventional mice. Strikingly, five of these miRNA expression variations (in miR-143, miR-148a, miR-200b, miR-200c, and miR-378) were dependent on the presence of the microbiota. In addition, as is already known, protein-coding genes were highly affected by infection in both conventional and germfree mice. By crossing bioinformatically the predicted targets of the miRNAs to our whole-genome transcriptomic data, we revealed an miRNA-mRNA network that suggested miRNA-mediated global regulation during intestinal infection. Other recent studies have revealed an miRNA response to either bacterial pathogens or commensal bacteria. In contrast, our work provides an unprecedented insight into the impact of the intestinal microbiota on host transcriptional reprogramming during infection by a human pathogen. While the crucial role of miRNAs in regulating the host response to bacterial infection is increasingly recognized, the involvement of the intestinal microbiota in the regulation of miRNA expression has not been explored in detail. Here, we investigated the impact of the intestinal microbiota on the regulation of protein-coding genes and miRNA expression in a host infected by L. monocytogenes, a food-borne pathogen. We show that the microbiota interferes with the microRNA response upon oral Listeria infection and identify several protein-coding target genes whose expression correlates inversely with that of the miRNA. Further investigations of the regulatory networks involving miR-143, miR-148a, miR-200b, miR-200c, and miR-378 will provide new insights into the impact of the intestinal microbiota on the host upon bacterial infection.
Collapse
|
44
|
Lamichhane A, Kiyono H, Kunisawa J. Nutritional components regulate the gut immune system and its association with intestinal immune disease development. J Gastroenterol Hepatol 2013; 28 Suppl 4:18-24. [PMID: 24251698 DOI: 10.1111/jgh.12259] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2013] [Indexed: 12/17/2022]
Abstract
The gut is equipped with a unique immune system for maintaining immunological homeostasis, and its functional immune disruption can result in the development of immune diseases such as food allergy and intestinal inflammation. Accumulating evidence has demonstrated that nutritional components play an important role in the regulation of gut immune responses and also in the development of intestinal immune diseases. In this review, we focus on the immunological functions of lipids, vitamins, and nucleotides in the regulation of the intestinal immune system and as potential targets for the control of intestinal immune diseases.
Collapse
Affiliation(s)
- Aayam Lamichhane
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan; Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan; Laboratory of Vaccine Materials, National Institute of Biomedical Innovation, Osaka, Japan
| | | | | |
Collapse
|
45
|
Turgeon N, Blais M, Gagné JM, Tardif V, Boudreau F, Perreault N, Asselin C. HDAC1 and HDAC2 restrain the intestinal inflammatory response by regulating intestinal epithelial cell differentiation. PLoS One 2013; 8:e73785. [PMID: 24040068 PMCID: PMC3764035 DOI: 10.1371/journal.pone.0073785] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/23/2013] [Indexed: 02/07/2023] Open
Abstract
Acetylation and deacetylation of histones and other proteins depends on histone acetyltransferases and histone deacetylases (HDACs) activities, leading to either positive or negative gene expression. HDAC inhibitors have uncovered a role for HDACs in proliferation, apoptosis and inflammation. However, little is known of the roles of specific HDACs in intestinal epithelial cells (IEC). We investigated the consequences of ablating both HDAC1 and HDAC2 in murine IECs. Floxed Hdac1 and Hdac2 homozygous mice were crossed with villin-Cre mice. Mice deficient in both IEC HDAC1 and HDAC2 weighed less and survived more than a year. Colon and small intestinal sections were stained with hematoxylin and eosin, or with Alcian blue and Periodic Acid Schiff for goblet cell identification. Tissue sections from mice injected with BrdU for 2 h, 14 h and 48 h were stained with anti-BrdU. To determine intestinal permeability, 4-kDa FITC-labeled dextran was given by gavage for 3 h. Microarray analysis was performed on total colon RNAs. Inflammatory and IEC-specific gene expression was assessed by Western blot or semi-quantitative RT-PCR and qPCR with respectively total colon protein and total colon RNAs. HDAC1 and HDAC2-deficient mice displayed: 1) increased migration and proliferation, with elevated cyclin D1 expression and phosphorylated S6 ribosomal protein, a downstream mTOR target; 2) tissue architecture defects with cell differentiation alterations, correlating with reduction of secretory Paneth and goblet cells in jejunum and goblet cells in colon, increased expression of enterocytic markers such as sucrase-isomaltase in the colon, increased expression of cleaved Notch1 and augmented intestinal permeability; 3) loss of tissue homeostasis, as evidenced by modifications of claudin 3 expression, caspase-3 cleavage and Stat3 phosphorylation; 4) chronic inflammation, as determined by inflammatory molecular expression signatures and altered inflammatory gene expression. Thus, epithelial HDAC1 and HDAC2 restrain the intestinal inflammatory response, by regulating intestinal epithelial cell proliferation and differentiation.
Collapse
Affiliation(s)
- Naomie Turgeon
- Département d’anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Mylène Blais
- Département d’anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Julie-Moore Gagné
- Département d’anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Véronique Tardif
- Département d’anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - François Boudreau
- Département d’anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Nathalie Perreault
- Département d’anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Claude Asselin
- Département d’anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, Québec, Canada
- * E-mail:
| |
Collapse
|
46
|
Staedel C, Darfeuille F. MicroRNAs and bacterial infection. Cell Microbiol 2013; 15:1496-507. [PMID: 23795564 DOI: 10.1111/cmi.12159] [Citation(s) in RCA: 165] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/14/2013] [Accepted: 06/17/2013] [Indexed: 12/13/2022]
Abstract
MicroRNAs, small non-coding RNAs expressed by eukaryotic cells, play pivotal roles in shaping cell differentiation and organism development. Deregulated microRNA expression is associated with several types of diseases including cancers, immune disorders and infection. Acting at the post-transcriptional level, miRNAs have expanded our understanding of the control of gene expression in regulatory networks involved in the adaptation to environmental situations such as biotic stress. It is increasingly clear that miRNAs are an important part of the host response to microbes. This review presents the current state of knowledge about the role of miRNAs in the response to both bacterial pathogens and commensal bacteria in human cells or animal experimental models. Some microRNAs, including miR-146, miR-155, miR-125, let-7 and miR-21, are commonly affected during bacterial infection and contribute to immune responses protecting the organism against overwhelmed inflammation. Cell-specific relationships between miRNAs and their targets are also engaged in the alterations induced by virulent bacteria in the proliferation/differentiation/apoptosis pathways of their host cells. In a separate role, miRNA modulation also represents a mechanism through which commensal bacteria impact the regulation of the barrier function and intestinal homeostasis.
Collapse
Affiliation(s)
- Cathy Staedel
- Univ. Bordeaux, ARNA Laboratory, F-33000, Bordeaux, France.
| | | |
Collapse
|
47
|
Obese humans with nonalcoholic fatty liver disease display alterations in fecal microbiota and volatile organic compounds. Clin Gastroenterol Hepatol 2013; 11:876-8. [PMID: 23628504 DOI: 10.1016/j.cgh.2013.04.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 04/08/2013] [Indexed: 02/07/2023]
|
48
|
Naydenov NG, Baranwal S, Khan S, Feygin A, Gupta P, Ivanov AI. Novel mechanism of cytokine-induced disruption of epithelial barriers: Janus kinase and protein kinase D-dependent downregulation of junction protein expression. Tissue Barriers 2013; 1:e25231. [PMID: 24665409 PMCID: PMC3783224 DOI: 10.4161/tisb.25231] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/01/2013] [Accepted: 05/31/2013] [Indexed: 12/13/2022] Open
Abstract
The ductal epithelium plays a key role in physiological secretion of pancreatic enzymes into the digestive system. Loss of barrier properties of the pancreatic duct may contribute to the development of pancreatitis and metastatic dissemination of pancreatic tumors. Proinflammatory cytokines are essential mediators of pancreatic inflammation and tumor progression; however, their effects on the integrity and barrier properties of the ductal epithelium have not been previously addressed. In the present study, we investigate mechanisms of cytokine-induced disassembly of tight junctions (TJs) and adherens junctions (AJs) in a model pancreatic epithelium. Exposure of HPAF-II human pancreatic epithelial cell monolayers to interferon (IFN)γ disrupted integrity and function of apical junctions as manifested by increased epithelial permeability and cytosolic translocation of AJ and TJ proteins. Tumor necrosis factor (TNF)α potentiated the effects of IFNγ on pancreatic epithelial junctions. The cytokine-induced increase in epithelial permeability and AJ/TJ disassembly was attenuated by pharmacological inhibition of Janus kinase (JAK) and protein kinase D (PKD). Loss of apical junctions in IFNγ/TNFα-treated HPAF-II cells was accompanied by JAK and PKD dependent decrease in expression of AJ (E-cadherin, p120 catenin) and TJ (occludin, ZO-1) proteins. Depletion of E-cadherin or p120 catenin recapitulated the effects of cytokines on HPAF-II cell permeability and junctions. Our data suggests that proinflammatory cytokines disrupt pancreatic epithelial barrier via expressional downregulation of key structural components of AJs and TJs. This mechanism is likely to be important for pancreatic inflammatory injury and tumorigenesis.
Collapse
Affiliation(s)
- Nayden G Naydenov
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA USA
| | - Somesh Baranwal
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA USA
| | - Shadab Khan
- Department of Medicine, University of Rochester School of Medicine, Rochester, NY USA
| | - Alex Feygin
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA USA
| | - Pooja Gupta
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA USA
| | - Andrei I Ivanov
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA USA; ; VCU Institute of Molecular Medicine; Virginia Commonwealth University School of Medicine; Richmond, VA USA ; VCU Massey Cancer Center; Virginia Commonwealth University School of Medicine; Richmond, VA USA
| |
Collapse
|
49
|
Current World Literature. Curr Opin Allergy Clin Immunol 2013. [DOI: 10.1097/aci.0b013e3283619e49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
50
|
Morrow AL, Lagomarcino AJ, Schibler KR, Taft DH, Yu Z, Wang B, Altaye M, Wagner M, Gevers D, Ward DV, Kennedy MA, Huttenhower C, Newburg DS. Early microbial and metabolomic signatures predict later onset of necrotizing enterocolitis in preterm infants. MICROBIOME 2013; 1:13. [PMID: 24450576 PMCID: PMC3971624 DOI: 10.1186/2049-2618-1-13] [Citation(s) in RCA: 263] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 03/18/2013] [Indexed: 05/21/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating intestinal disease that afflicts 10% of extremely preterm infants. The contribution of early intestinal colonization to NEC onset is not understood, and predictive biomarkers to guide prevention are lacking. We analyzed banked stool and urine samples collected prior to disease onset from infants <29 weeks gestational age, including 11 infants who developed NEC and 21 matched controls who survived free of NEC. Stool bacterial communities were profiled by 16S rRNA gene sequencing. Urinary metabolomic profiles were assessed by NMR. RESULTS During postnatal days 4 to 9, samples from infants who later developed NEC tended towards lower alpha diversity (Chao1 index, P = 0.086) and lacked Propionibacterium (P = 0.009) compared to controls. Furthermore, NEC was preceded by distinct forms of dysbiosis. During days 4 to 9, samples from four NEC cases were dominated by members of the Firmicutes (median relative abundance >99% versus <17% in the remaining NEC and controls, P < 0.001). During postnatal days 10 to 16, samples from the remaining NEC cases were dominated by Proteobacteria, specifically Enterobacteriaceae (median relative abundance >99% versus 38% in the other NEC cases and 84% in controls, P = 0.01). NEC preceded by Firmicutes dysbiosis occurred earlier (onset, days 7 to 21) than NEC preceded by Proteobacteria dysbiosis (onset, days 19 to 39). All NEC cases lacked Propionibacterium and were preceded by either Firmicutes (≥98% relative abundance, days 4 to 9) or Proteobacteria (≥90% relative abundance, days 10 to 16) dysbiosis, while only 25% of controls had this phenotype (predictive value 88%, P = 0.001). Analysis of days 4 to 9 urine samples found no metabolites associated with all NEC cases, but alanine was positively associated with NEC cases that were preceded by Firmicutes dysbiosis (P < 0.001) and histidine was inversely associated with NEC cases preceded by Proteobacteria dysbiosis (P = 0.013). A high urinary alanine:histidine ratio was associated with microbial characteristics (P < 0.001) and provided good prediction of overall NEC (predictive value 78%, P = 0.007). CONCLUSIONS Early dysbiosis is strongly involved in the pathobiology of NEC. These striking findings require validation in larger studies but indicate that early microbial and metabolomic signatures may provide highly predictive biomarkers of NEC.
Collapse
Affiliation(s)
- Ardythe L Morrow
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, MLC 7009, Cincinnati, OH 45229, USA
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Anne J Lagomarcino
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, MLC 7009, Cincinnati, OH 45229, USA
| | - Kurt R Schibler
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, MLC 7009, Cincinnati, OH 45229, USA
| | - Diana H Taft
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Department Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, MLC 7009, Cincinnati, OH 45229, USA
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Zhuoteng Yu
- Department of Biology, Boston College, Chestnut Hill, MA, USA
| | - Bo Wang
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, USA
| | - Mekibib Altaye
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael Wagner
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | | | - Michael A Kennedy
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, USA
| | | | - David S Newburg
- Department of Biology, Boston College, Chestnut Hill, MA, USA
| |
Collapse
|