201
|
Hatano H, Goda T, Matsumoto A, Miyahara Y. Induced Proton Dynamics on Semiconductor Surfaces for Sensing Tight Junction Formation Enhanced by an Extracellular Matrix and Drug. ACS Sens 2019; 4:3195-3202. [PMID: 31763825 DOI: 10.1021/acssensors.9b01635] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In the fields of tissue engineering and drug discovery, confirming the formation and maturation of epithelial cell tight junctions (TJs), which are necessary for blocking pathogenic invasion and absorption of nutrients and ions, at a high spatiotemporal resolution is essential. We previously developed a system of monitoring pH perturbation induced by weak acid exposure to cells cultured on an ion-sensitive field-effect transistor that enables a sensitive and specific detection of biomembrane injuries and TJ breakdowns caused by external stimuli such as nanomaterials and cytotoxins. In this study, we monitor time-lapse changes in the paracellular diffusion of growing epithelial cell monolayers using the pH perturbation assay as well as conventional permeability and trans-epithelial electrical resistance assays. The effects of the extracellular matrix and a TJ potentiator (KN-93) on epithelial TJ formation are evaluated. TJ formations were promoted on the substrate coated with Matrigel more than on the one coated with poly(l-lysine). KN-93 accelerated TJ formations in a dose-dependent manner. The pH perturbation assay denoted a longer incubation time for the completion of TJ formation compared with the conventional assays under the same conditions. Importantly, the pH perturbation assay is able to rigorously evaluate TJ formation, as the assay uses protons as the smallest indicator for detecting paracellular gaps, and the pH perturbation is specific to TJ alterations. These features for in vitro TJ evaluation using proton dynamics are advantageous for applications in tissue engineering and drug development.
Collapse
Affiliation(s)
- Hiroaki Hatano
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Tatsuro Goda
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
- Nano Innovation Institute, Inner Mongolia University for Nationalities, No. 22 Huoline Street, Tongliao, Inner Mongolia 028000, P. R. China
| | - Akira Matsumoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
- Kanagawa Institute of Industrial Science and Technology, 705-1 Shimoimaizumi, Ebina, Kanagawa 243-0435, Japan
| | - Yuji Miyahara
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| |
Collapse
|
202
|
Early life nutrition influences susceptibility to chronic inflammatory colitis in later life. Sci Rep 2019; 9:18111. [PMID: 31792267 PMCID: PMC6889478 DOI: 10.1038/s41598-019-54308-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 11/08/2019] [Indexed: 12/21/2022] Open
Abstract
The first thousand days of life are a critical time of development in humans during which the risk profile for diseases in later life can be modified. Nevertheless, long-term consequences of early environment on susceptibility to intestinal diseases have not yet been assessed. Using a mouse model of postnatal growth restriction (PNGR), we showed that early life nutrition influences intestinal maturation and gut health in later life. PNGR induced an alteration of the intestinal barrier in pups at weaning, resulting in increased intestinal permeability, and affected gut bacterial colonization. Specifically, pups with PNGR harbored a decreased bacterial diversity, higher Enterococcus spp., Staphylococcus spp., and Escherichia-Shigella spp., and lower Odoribacter spp. and several members of the Lachnospiraceae family. The lack of an efficient intestinal barrier in early life and the dysbiosis induced by PNGR were associated with a higher susceptibility to chronic colitis in adulthood.
Collapse
|
203
|
Jin C, Zeng Z, Wang C, Luo T, Wang S, Zhou J, Ni Y, Fu Z, Jin Y. Insights into a Possible Mechanism Underlying the Connection of Carbendazim-Induced Lipid Metabolism Disorder and Gut Microbiota Dysbiosis in Mice. Toxicol Sci 2019; 166:382-393. [PMID: 30496565 DOI: 10.1093/toxsci/kfy205] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Carbendazim (CBZ), a systemic, broad-spectrum benzimidazole fungicide, is widely used to control fungal diseases and has been regarded as an endocrine disruptor that causes mammalian toxicity in different target organs. Here, we discovered that chronic administrations of CBZ at 0.2, 1, and 5 mg/kg body weight for 14 weeks not only changed the composition of gut microbiota but also induced significant increases in body, liver, and epididymal fat weight in mice. At the biochemical level, the serum triglyceride (TG) and glucose levels also increased after CBZ exposure. Moreover, the level of serum lipoprotein lipase (LPL), which plays an important role in fatty acid release from TG, was decreased significantly. For gut microbiota, 16S rRNA gene sequencing and real-time qPCR revealed that CBZ exposure significantly perturbed the mice gut microbiome, and gas chromatography found that the production of short-chain fatty acids were altered. Moreover, CBZ exposure increased the absorption of exogenous TG in the mice intestine and inhibited the TG consumption, eventually leading the serum triglyceride to maintain higher levels. The increase of lipid absorption in the intestine direct caused hyperlipidemia and the multi-tissue inflammatory response. In response to the rise of lipid in blood, the body maintains the balance of lipid metabolism in mice by reducing lipid synthesis in the liver and increasing lipid storage in the fat. Chronic CBZ exposure induced the gut microbiota dysbiosis and disturbed lipid metabolism, which promoted the intestinal absorption of excess triglyceride and caused multiple tissue inflammatory responses in mice.
Collapse
Affiliation(s)
- Cuiyuan Jin
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zhaoyang Zeng
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Caiyun Wang
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Ting Luo
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Siyu Wang
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jicong Zhou
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yingchun Ni
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zhengwei Fu
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yuanxiang Jin
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
204
|
Duan X, Tian G, Chen D, Huang L, Zhang D, Zheng P, Mao X, Yu J, He J, Huang Z, Yu B. Mannan oligosaccharide supplementation in diets of sow and (or) their offspring improved immunity and regulated intestinal bacteria in piglet1. J Anim Sci 2019; 97:4548-4556. [PMID: 31603198 PMCID: PMC6827392 DOI: 10.1093/jas/skz318] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/04/2019] [Indexed: 01/02/2023] Open
Abstract
The objectives of the current study were to explore the effects of mannan oligosaccharide (MOS) supplementation in the diets of sow and (or) their offspring on intestinal bacteria, intestinal and systemic inflammation in the piglet. A total of 60 multiparous sows (4 ± 1 parity; Landrace × Yorkshire) were fed either control diet (sCON, n = 30) or a diet containing 400 mg kg-1 MOS (sMOS, n = 30) from day 86 of gestation until weaning (day 20 of postpartum). On day 7 of age, offspring (Duroc × Landrace Yorkshire) were assigned within sow treatments and fed control diet (pCON) or diet containing 800 mg kg-1 MOS (pMOS) for 28 d (end at 35 d of age), resulting in four piglet diet groups (n = 15 litters per diet group): sCON-pCON, sCON-pMOS, sMOS-pCON, and sMOS-pMOS. Results found that piglet diet MOS increased or tend to increase Lactobacillus amount in the ileum digesta (P < 0.01) and jejunum digesta (P = 0.07), respectively; while tend to decrease Escherichia coli amount in jejunum digesta (P =0.06) and cecum digesta (P = 0.08). Both sow and piglet diets add MOS (sMOS-pMOS) increased Lactobacillus amount but decreased E. coli amount in jejunum digesta (P < 0.05) compared with the sCON-pCON diet group. In addition, sow diet MOS (rather than piglet diet MOS) increased sIgA content in piglet jejunum mucosa compared with control (P = 0.04). Sow diet MOS decreased toll-like receptor 2 (TLR2), toll-like receptor 4 (TLR4), and interleukin 8 (IL-8) mRNA levels (P < 0.05) and tended to decrease nuclear factor-κB p65 (NF-κB p65) mRNA level (P = 0.07) in piglet intestinal lymphatic. The interaction effects between sow and piglet diets were found on the mRNA levels of NF- κB p65 (P = 0.03) and IL-8 (P = 0.02) in piglet jejunum. Finally, the sow diet MOS decreased proinflammatory cytokines IL-2 (P < 0.01) and IL-4 (P < 0.01) concentrations in piglet serum. Piglets diet MOS decreased the contents of IL-2 (P = 0.03), IL-4 (P = 0.01) and interferon (IFN)-γ (P < 0.01) while increased anti-inflammatory cytokine IL-10 (P < 0.01) content in serum. The interaction effects between sows and piglet diets on IL-4 (P = 0.02), IL-10 (P < 0.01), and IFN-γ (P = 0.08) were observed. In conclusion, sow and/or piglet diet MOS could improve intestinal microbiota, enhance intestinal mucosal immune competence, and suppress intestinal and systemic inflammation in the piglet.
Collapse
Affiliation(s)
- Xudong Duan
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Gang Tian
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Linhui Huang
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Dan Zhang
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agriculture University, Yucheng District, Yaan, Sichuan, China
| |
Collapse
|
205
|
Feng L, Chen S, Zhang L, Qu W, Chen Z. Bisphenol A increases intestinal permeability through disrupting intestinal barrier function in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 254:112960. [PMID: 31394344 DOI: 10.1016/j.envpol.2019.112960] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/20/2019] [Accepted: 07/25/2019] [Indexed: 05/28/2023]
Abstract
That an alteration of the intestinal permeability is associated with gut barrier function has been increasingly evident, which plays an important role in human and animal health. Bisphenol A (BPA), an industrial compound used worldwide, has recently been classified as an environmental pollutant. One of our earlier studies has demonstrated that BPA disrupts the intestinal barrier function by inducing apoptosis and inhibiting cell proliferation in the human colonic epithelial cells line. In this study, we investigated the effects of dietary BPA uptake on the colonic barrier function in mice, as well as the intestinal permeability. Dietary BPA uptake was observed to destroy the morphology of the colonic epithelium and increase the pathology score. The levels of endotoxin, diamine peroxidase, D-lactate, and zonulin were found to have been significantly elevated in both plasma and colonic mucosa. A decline in the number of intestinal goblet cells and in mucin 2 gene expression was observed in the mice belonging to the BPA group. The results of immunohistochemistry revealed that the expression of tight junction proteins (ZO-1, occludin, and claudin-1) in colonic epithelium of BPA mice decreased significantly, and their gene abundance was also inhibited. Moreover, dietary BPA uptake was also found to have significantly reduced colonic microbial diversity and altered microbial structural composition. The functional profiles of colonic bacterial community exhibited adverse effects of dietary BPA intake on the endocrine and digestive systems, as well as the transport and catabolism functions. Collectively, our study highlighted that dietary BPA increased the colonic permeability, and this effect was closely related to the disruption of intestinal chemistry and physical and biological barrier functions.
Collapse
Affiliation(s)
- Ling Feng
- Jiangyin Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangyin 214400, Jiangsu, China.
| | - Sijin Chen
- Department of Pharmacy, The Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin, Jiangsu 214400, People's Republic of China.
| | - Lijin Zhang
- Department of Urinary Surgery, The Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin, Jiangsu 214400, People's Republic of China.
| | - Wei Qu
- Department of Pharmacy, The Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin, Jiangsu 214400, People's Republic of China.
| | - Zhigao Chen
- Department of Pharmacy, The Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin, Jiangsu 214400, People's Republic of China.
| |
Collapse
|
206
|
Misra P, Singh S. Site specific microbiome of Leishmania parasite and its cross-talk with immune milieu. Immunol Lett 2019; 216:79-88. [PMID: 31678358 DOI: 10.1016/j.imlet.2019.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/17/2019] [Accepted: 10/02/2019] [Indexed: 12/12/2022]
Abstract
Microbiota consists of commensal, symbiotic and pathogenic microorganisms found in all multicellular organisms. These micro-organisms are found in or on many parts of the body, including the intestinal tract, skin, mouth, and the reproductive tract. This review focuses on interplay of site specific microbiota, vector microbiota along with immune response and severity of Leishmaniasis. Herein, we have reviewed and summarized the counter effect of microbiome post infection with the Leishmania parasite. We have studied skin microbiome along with the gut microbiome of sand-fly which is the vector for transmission of this disease. Our major focus was to understand the skin and gut microbiome during Leishmania infection,their interaction and effect on immunological responses generated during the infection.Moreover, systems biology approach is envisioned to enumerate bacterial species in skin microbiota and Phlebotmus gut microbiota during Leishmania infection.
Collapse
Affiliation(s)
- Pragya Misra
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Shailza Singh
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India.
| |
Collapse
|
207
|
An infection of Enterobacter ludwigii affects development and causes age-dependent neurodegeneration in Drosophila melanogaster. INVERTEBRATE NEUROSCIENCE 2019; 19:13. [PMID: 31641932 DOI: 10.1007/s10158-019-0233-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023]
Abstract
The effects of teeth-blackening bacteria Enterobacter ludwigii on the physiological system were investigated using the model organism Drosophila melanogaster. The bacteria were mixed with the fly food, and its effect was checked on the growth, development and behaviour of Drosophila. Microbes generate reactive oxygen species (ROS) within the haemolymph of the larvae once it enters into the body. The increased amount of ROS was evidenced by the NBT assay and using 2',7'-dichlorofluorescin diacetate dye, which indicates the mitochondrial ROS. The increased amount of ROS resulted in a number of abnormal nuclei within the gut. Besides that larvae walking became sluggish in comparison with wild type although the larvae crawling path did not change much. Flies hatched from the infectious larvae have the posterior scutellar bristle absent from the thorax and abnormal mechanosensory hairs in the eye, and they undergo time-dependent neurodegeneration as evidenced by the geotrophic and phototrophic assays. To decipher the mechanism of neurodegeneration, flies were checked for the presence of four important bioamines: tyramine, cadaverine, putrescine and histamine. Out of these four, histamine was found to be absent in infected flies. Histamine is a key molecule required for the functioning of the photoreceptor as well as mechanoreceptors. The mechanism via which mouth infectious bacteria E. ludwigii can affect the development and cause age-dependent neurodegeneration is explained in this paper.
Collapse
|
208
|
Burns G, Pryor J, Holtmann G, Walker MM, Talley NJ, Keely S. Immune Activation in Functional Gastrointestinal Disorders. Gastroenterol Hepatol (N Y) 2019; 15:539-548. [PMID: 31802978 PMCID: PMC6883739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
There is growing appreciation that functional gastrointestinal disorders (FGIDs) such as functional dyspepsia and irritable bowel syndrome are heterogeneous conditions linked by subtle inflammation within the gastrointestinal (GI) tract. The literature suggests that while the symptoms of these diseases may manifest with similar clinical presentations, there are significant differences in triggers and disease severity among patients classified into the same subtype. It is hypothesized that the subtle inflammation observed in these patients is related to an imbalance in GI homeostasis. Disruption of the delicate homeostatic balance within the GI tract can result from any number or combination of factors, including dysbiosis, loss of barrier integrity, genetic predisposition, or immune responses to dietary or luminal antigens. This article discusses the interplay between the immune system, microbiota, and luminal environment in FGIDs. In addition, the article proposes emerging immune pathways, including those involving T-helper type 17 response and innate lymphoid cells, as potential regulators of the subtle inflammation characteristic of FGIDs that warrant investigation in future studies.
Collapse
Affiliation(s)
- Grace Burns
- Ms Burns is a PhD graduate student, Ms Pryor is an undergraduate research student
- Dr Walker is a professor of anatomical pathology
- Dr Talley is a laureate professor
- Dr Keely is an associate professor in the Priority Research Centre for Digestive Health and Neurogastroenterology in the Faculty of Health and Medicine at the University of Newcastle in Callaghan, New South Wales, Australia, as well as in the Hunter Medical Research Institute in New Lambton Heights, New South Wales, Australia
- Dr Holtmann is director of gastroenterology and hepatology at the Princess Alexandra Hospital in Brisbane, Queensland, Australia and a professor in the Faculty of Medicine at the University of Queensland in Woolloongabba, Queensland, Australia
| | - Jennifer Pryor
- Ms Burns is a PhD graduate student, Ms Pryor is an undergraduate research student
- Dr Walker is a professor of anatomical pathology
- Dr Talley is a laureate professor
- Dr Keely is an associate professor in the Priority Research Centre for Digestive Health and Neurogastroenterology in the Faculty of Health and Medicine at the University of Newcastle in Callaghan, New South Wales, Australia, as well as in the Hunter Medical Research Institute in New Lambton Heights, New South Wales, Australia
- Dr Holtmann is director of gastroenterology and hepatology at the Princess Alexandra Hospital in Brisbane, Queensland, Australia and a professor in the Faculty of Medicine at the University of Queensland in Woolloongabba, Queensland, Australia
| | - Gerald Holtmann
- Ms Burns is a PhD graduate student, Ms Pryor is an undergraduate research student
- Dr Walker is a professor of anatomical pathology
- Dr Talley is a laureate professor
- Dr Keely is an associate professor in the Priority Research Centre for Digestive Health and Neurogastroenterology in the Faculty of Health and Medicine at the University of Newcastle in Callaghan, New South Wales, Australia, as well as in the Hunter Medical Research Institute in New Lambton Heights, New South Wales, Australia
- Dr Holtmann is director of gastroenterology and hepatology at the Princess Alexandra Hospital in Brisbane, Queensland, Australia and a professor in the Faculty of Medicine at the University of Queensland in Woolloongabba, Queensland, Australia
| | - Marjorie M Walker
- Ms Burns is a PhD graduate student, Ms Pryor is an undergraduate research student
- Dr Walker is a professor of anatomical pathology
- Dr Talley is a laureate professor
- Dr Keely is an associate professor in the Priority Research Centre for Digestive Health and Neurogastroenterology in the Faculty of Health and Medicine at the University of Newcastle in Callaghan, New South Wales, Australia, as well as in the Hunter Medical Research Institute in New Lambton Heights, New South Wales, Australia
- Dr Holtmann is director of gastroenterology and hepatology at the Princess Alexandra Hospital in Brisbane, Queensland, Australia and a professor in the Faculty of Medicine at the University of Queensland in Woolloongabba, Queensland, Australia
| | - Nicholas J Talley
- Ms Burns is a PhD graduate student, Ms Pryor is an undergraduate research student
- Dr Walker is a professor of anatomical pathology
- Dr Talley is a laureate professor
- Dr Keely is an associate professor in the Priority Research Centre for Digestive Health and Neurogastroenterology in the Faculty of Health and Medicine at the University of Newcastle in Callaghan, New South Wales, Australia, as well as in the Hunter Medical Research Institute in New Lambton Heights, New South Wales, Australia
- Dr Holtmann is director of gastroenterology and hepatology at the Princess Alexandra Hospital in Brisbane, Queensland, Australia and a professor in the Faculty of Medicine at the University of Queensland in Woolloongabba, Queensland, Australia
| | - Simon Keely
- Ms Burns is a PhD graduate student, Ms Pryor is an undergraduate research student
- Dr Walker is a professor of anatomical pathology
- Dr Talley is a laureate professor
- Dr Keely is an associate professor in the Priority Research Centre for Digestive Health and Neurogastroenterology in the Faculty of Health and Medicine at the University of Newcastle in Callaghan, New South Wales, Australia, as well as in the Hunter Medical Research Institute in New Lambton Heights, New South Wales, Australia
- Dr Holtmann is director of gastroenterology and hepatology at the Princess Alexandra Hospital in Brisbane, Queensland, Australia and a professor in the Faculty of Medicine at the University of Queensland in Woolloongabba, Queensland, Australia
| |
Collapse
|
209
|
Pan Z, Yuan X, Tu W, Fu Z, Jin Y. Subchronic exposure of environmentally relevant concentrations of F-53B in mice resulted in gut barrier dysfunction and colonic inflammation in a sex-independent manner. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 253:268-277. [PMID: 31319243 DOI: 10.1016/j.envpol.2019.07.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/25/2019] [Accepted: 07/04/2019] [Indexed: 06/10/2023]
Abstract
F-53B (6:2 chlorinated polyfluorinated ether sulfonate) is currently recognized as a safe alternative to long-chain PFASs in China. However, an increasing number of studies have recently authenticated its biotoxicological effects. In this study, for evaluating the gut toxicity of F-53B in mammals, both female and male mice were orally exposed to 0, 1, 3, or 10 μg/L F-53B for 10 weeks. Our results showed that F-53B significantly accumulated in the colon, ileum and serum when exposed to 10 μg/L F-53B for 10 weeks. F-53B exposure not only increased the transcriptional levels of ion transport-related genes but could also interact with the CFTR protein directly. Interestingly, subchronic F-53B exposure also increased the transcription of mucus secretion-related genes, but the protein level of Muc2 decreased after F-53B exposure, indicating that there was a compensatory phenomenon after mucus barrier injury. Furthermore, F-53B exposure also induced colonic inflammation associated with gut microbiota dysbiosis in the colon. Taken together, our results indicated that the potential gut toxicity of F-53B and almost all of the changed parameters were significantly affected in both female and male mice, suggesting that F-53B could disturb the gut barrier without sex dependence in mice.
Collapse
Affiliation(s)
- Zihong Pan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Xianling Yuan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Wenqing Tu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang, 330029, China.
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China.
| |
Collapse
|
210
|
Tao S, Bai Y, Li T, Li N, Wang J. Original low birth weight deteriorates the hindgut epithelial barrier function in pigs at the growing stage. FASEB J 2019; 33:9897-9912. [PMID: 31170357 DOI: 10.1096/fj.201900204rr] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
The deteriorative effect of low birth weight (LBW) on the mucosal permeability of the small intestine in piglets has been widely confirmed. However, whether the hindgut epithelial barrier function in LBW pigs is deteriorated during the growing stage is still unclear. Our study investigated differences in the hindgut epithelial barrier function between LBW and normal birth weight pigs during the growing stage (d 90). Our data demonstrated that the hindgut epithelium of LBW pigs has a high histopathological score, accompanied by decreased antioxidant capacity and increased apoptosis rate, as well as elevated expression and activity of caspase-3. In addition, the number of intestinal goblet cells and gene expression of mucin 2 were significantly down-regulated in LBW pigs. The expression of tight junction proteins (ZO-1 and occludin) was markedly inhibited by the LBW. The mRNA abundance of inflammatory cytokines such as TNF-α, IL-1β, and IL-8 was significantly increased in the hindgut mucosa of LBW pigs. Furthermore, our data revealed that LBW altered the intestinal microbial community in the hindgut mucosa of pigs. Collectively, these finding add to our understanding of the mechanisms responsible for hindgut epithelial barrier dysfunction in LBW pigs during the growing stage and facilitate the development of nutritional intervention strategies.-Tao, S., Bai, Y., Li, T., Li, N., Wang, J. Original low birth weight deteriorates the hindgut epithelial barrier function in pigs at the growing stage.
Collapse
Affiliation(s)
- Shiyu Tao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yu Bai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tiantian Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Na Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
211
|
Na YR, Stakenborg M, Seok SH, Matteoli G. Macrophages in intestinal inflammation and resolution: a potential therapeutic target in IBD. Nat Rev Gastroenterol Hepatol 2019; 16:531-543. [PMID: 31312042 DOI: 10.1038/s41575-019-0172-4] [Citation(s) in RCA: 576] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2019] [Indexed: 02/07/2023]
Abstract
Macrophages are the gatekeepers of intestinal immune homeostasis as they discriminate between innocuous antigens and potential pathogens to maintain oral tolerance. However, in individuals with a genetic and environmental predisposition, regulation of intestinal immunity is impaired, leading to chronic relapsing immune activation and pathologies of the gastrointestinal tract, such as IBD. As evidence suggests a causal link between defects in the resolution of intestinal inflammation and altered monocyte-macrophage differentiation in patients with IBD, macrophages have been considered as a novel potential target to develop new treatment approaches. This Review discusses the molecular and cellular mechanisms involved in the differentiation and function of intestinal macrophages in homeostasis and inflammation, and their role in resolving the inflammatory process. Understanding the molecular pathways involved in the specification of intestinal macrophages might lead to a new class of targets that promote remission in patients with IBD.
Collapse
Affiliation(s)
- Yi Rang Na
- Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University Medical College, Seoul, South Korea
| | - Michelle Stakenborg
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Seung Hyeok Seok
- Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University Medical College, Seoul, South Korea.
| | - Gianluca Matteoli
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.
| |
Collapse
|
212
|
Abstract
This review chapter describes the current knowledge about the nature of pericytes in the gut, their interaction with endothelial cells in blood vessels, and their pathophysiological functions in the setting of chronic liver disease. In particular, it focuses on the role of these vascular cell types and related molecular signaling pathways in pathological angiogenesis associated with liver disease and in the establishment of the gut-vascular barrier and the potential implications in liver disease through the gut-liver axis.
Collapse
|
213
|
Wang C, Zhang Y, Deng M, Wang X, Tu W, Fu Z, Jin Y. Bioaccumulation in the gut and liver causes gut barrier dysfunction and hepatic metabolism disorder in mice after exposure to low doses of OBS. ENVIRONMENT INTERNATIONAL 2019; 129:279-290. [PMID: 31146162 DOI: 10.1016/j.envint.2019.05.056] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 05/23/2023]
Abstract
The compound sodium ρ-perfluorous nonenoxybenzene sulfonate (OBS), a new kind of perfluoroalkyl and polyfluoroalkyl compound, is a surfactant for increasing oil production, and it has been widely detected in various organisms. Because of its wide use, OBS is detectable in the environment. However, knowledge about the biological toxicity of OBS to animals is very limited. Here, male mice were exposed to 0, 0.1, 1 or 10 μg/L of OBS for 6 weeks via drinking water. It was demonstrated that OBS was highly bioaccumulated both in the liver and gut in the mice after low doses of OBS exposure. Curiously, a low dose of OBS exposure also caused gut barrier dysfunction by decreasing mucus secretion and altering Ionic transport in the gut via the CFTR pathway. In addition, liver function was influenced by OBS at both the histopathological and physiological levels. Hepatic transcriptomics and metabolomics analysis showed a total of 1157 genes, and multiple metabolites changed significantly in the livers of mice exposed to low-dose OBS for 6 weeks. The functions of these changed genes and metabolites are tightly related to glycolysis, fatty acid synthesis, fatty acid transport, and β-oxidation. All these results indicate that the liver and gut are important target tissues for OBS exposure. Importantly, it is possible that high levels of bioaccumulation of OBS in the gut and liver might directly cause gut barrier dysfunction and hepatic metabolism disorder in mice.
Collapse
Affiliation(s)
- Caiyun Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yi Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Mi Deng
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330029, China
| | - Xia Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Wenqing Tu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330029, China.
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
214
|
Markota A, Metzger R, Heiseke AF, Jandl L, Dursun E, Eisenächer K, Reindl W, Haller D, Krug AB. Comparison of iron-reduced and iron-supplemented semisynthetic diets in T cell transfer colitis. PLoS One 2019; 14:e0218332. [PMID: 31276514 PMCID: PMC6611680 DOI: 10.1371/journal.pone.0218332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022] Open
Abstract
Clinical observations in inflammatory bowel disease patients and experimental studies in rodents suggest that iron in the intestinal lumen derived from iron-rich food or oral iron supplementation could exacerbate inflammation and that iron depletion from the diet could be protective. To test the hypothesis that dietary iron reduction is protective against colitis development, the impact of iron reduction in the diet below 10 mg/kg on the course of CD4+ CD62L+ T cell transfer colitis was investigated in adult C57BL/6 mice. Weight loss as well as clinical and histological signs of inflammation were comparable between mice pretreated with semisynthetic diets with either < 10mg/kg iron content or supplemented with 180 mg/kg iron in the form of ferrous sulfate or hemin. Accumulation and activation of Ly6Chigh monocytes, changes in dendritic cell subset composition and induction of proinflammatory Th1/Th17 cells in the inflamed colon were not affected by the iron content of the diets. Thus, dietary iron reduction did not protect adult mice against severe intestinal inflammation in T cell transfer induced colitis.
Collapse
Affiliation(s)
- Anamarija Markota
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Rebecca Metzger
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Alexander F. Heiseke
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Lisa Jandl
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Ezgi Dursun
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Katharina Eisenächer
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Wolfgang Reindl
- Klinikum Mannheim, II. Medizinische Klinik, Mannheim, Germany
| | - Dirk Haller
- Chair for Nutrition and Immunology, Technical University Munich, Freising, Germany
| | - Anne B. Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
- * E-mail:
| |
Collapse
|
215
|
Nie Y, Hu J, Hou Q, Zheng W, Zhang X, Yang T, Ma L, Yan X. Lactobacillus frumenti improves antioxidant capacity via nitric oxide synthase 1 in intestinal epithelial cells. FASEB J 2019; 33:10705-10716. [PMID: 31262191 DOI: 10.1096/fj.201900253rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Oxidative damages have adverse effects on mammals. Growing studies have focused on exploring new antioxidants. Here, we report that Lactobacillus frumenti increases the total antioxidation capacity activities and decreases the total reactive oxygen species levels in porcine intestinal epithelial cells. Comparative proteomics revealed that expressions of peroxiredoxin 2, isocitrate dehydrogenase 1, NAD(P)H dehydrogenase quinone 1, antioxidant protein 1, and metallothionein-2A, which are associated with antioxidant defense system, were significantly increased with L. frumenti treatment. In germ-free mice, L. frumenti treatment also remarkably improves the intestinal antioxidant capacity. We further illustrated that nitric oxide production-mediated by nitric oxide synthase 1 activation is essential for L. frumenti-induced improvements in intestinal epithelial antioxidant capacity and barrier function. This study suggested that L. frumenti may be a potential probiotic used to prevent oxidative stress-induced aging and diseases in mammals.-Nie, Y., Hu, J., Hou, Q., Zheng, W., Zhang, X., Yang, T., Ma, L., Yan, X. Lactobacillus frumenti improves antioxidant capacity via nitric oxide synthase 1 in intestinal epithelial cells.
Collapse
Affiliation(s)
- Yangfan Nie
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China.,National Center for International Research on Animal Genetics, Breeding and Reproduction, Wuhan, China
| | - Jun Hu
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China.,National Center for International Research on Animal Genetics, Breeding and Reproduction, Wuhan, China
| | - Qiliang Hou
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China.,National Center for International Research on Animal Genetics, Breeding and Reproduction, Wuhan, China
| | - Wenyong Zheng
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China.,National Center for International Research on Animal Genetics, Breeding and Reproduction, Wuhan, China
| | - Xianghua Zhang
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China.,National Center for International Research on Animal Genetics, Breeding and Reproduction, Wuhan, China
| | - Tao Yang
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China.,National Center for International Research on Animal Genetics, Breeding and Reproduction, Wuhan, China
| | - Libao Ma
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China.,National Center for International Research on Animal Genetics, Breeding and Reproduction, Wuhan, China
| | - Xianghua Yan
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China.,National Center for International Research on Animal Genetics, Breeding and Reproduction, Wuhan, China
| |
Collapse
|
216
|
Probiotic Mixture VSL#3 Alleviates Dextran Sulfate Sodium-induced Colitis in Mice by Downregulating T Follicular Helper Cells. Curr Med Sci 2019; 39:371-378. [DOI: 10.1007/s11596-019-2045-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 01/23/2019] [Indexed: 01/01/2023]
|
217
|
Huang C, Yao R, Zhu Z, Pang D, Cao X, Feng B, Paulsen BS, Li L, Yin Z, Chen X, Jia R, Song X, Ye G, Luo Q, Chen Z, Zou Y. A pectic polysaccharide from water decoction of Xinjiang Lycium barbarum fruit protects against intestinal endoplasmic reticulum stress. Int J Biol Macromol 2019; 130:508-514. [PMID: 30826406 DOI: 10.1016/j.ijbiomac.2019.02.157] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/16/2019] [Accepted: 02/27/2019] [Indexed: 02/05/2023]
Abstract
Neutral polysaccharides from Ningxia L. barbarum fruit have been reported with immunomodulatory and antioxidative biological activities. Few studies on pectic polysaccharides have been reported, especially not from the Xinjiang L. barbarum. In the present study, a pectic polysaccharide, XLBP-I-I, was obtained from water decoction of Xinjiang L. barbarum using anion exchange chromatography and gel filtration. The results from methanolysis, methylation, FT-IR and NMR experiments indicated that XLBP-I-I was a typical pectic polysaccharide. In vitro assay showed that XLBP-I-I could reduce the ER stress and UPR in tunicamycin insult IPEC-J2 cells, and further protect IPEC-J2 cells against apoptosis induced by ER stress. These results reveal a new perspective for pectic L. barbarum polysaccharides on intestine ER stress, and this elicited interests for its further applications.
Collapse
Affiliation(s)
- Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Ruyu Yao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, PR China
| | - Zhongkai Zhu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Dejiang Pang
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Xiyue Cao
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, PR China
| | | | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Xingfu Chen
- Key Laboratory of Crop Ecophysiology and Farming System in Southwest China, Ministry of Agriculture, College of Agronomy, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Renrong Jia
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Gang Ye
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China.
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, PR China.
| |
Collapse
|
218
|
Phillipson M, Kubes P. The Healing Power of Neutrophils. Trends Immunol 2019; 40:635-647. [PMID: 31160208 DOI: 10.1016/j.it.2019.05.001] [Citation(s) in RCA: 312] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/01/2019] [Accepted: 05/01/2019] [Indexed: 11/28/2022]
Abstract
Neutrophils promptly accumulate in large numbers at sites of tissue injury. Injuries to the skin or mucosae disrupt barriers against the external environment, and the bactericidal actions of neutrophils are important in preventing microbial invasion. Neutrophils have also been associated with exacerbated inflammation, for example in non-healing wounds or in conditions such as inflammatory bowel disease (IBD). However, additional neutrophil functions important for angiogenesis and tissue restoration have been uncovered in models of sterile and ischemic injury, as well as in tumors. These functions are also relevant in healing skin and mucosal wounds, and can be impaired in conditions associated with non-healing wounds, such as diabetes. Here, we discuss our current understanding of neutrophil contributions to healing, and how the latter can be compromised in disease.
Collapse
Affiliation(s)
- Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Paul Kubes
- Snyder Institute of Infection, Immunity, and Inflammation, University of Calgary, Calgary, Alberta, T2N 4N1, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
219
|
Adams S, Xiangjie K, Hailong J, Guixin Q, Sossah FL, Dongsheng C. Prebiotic effects of alfalfa ( Medicago sativa) fiber on cecal bacterial composition, short-chain fatty acids, and diarrhea incidence in weaning piglets. RSC Adv 2019; 9:13586-13599. [PMID: 35519545 PMCID: PMC9063875 DOI: 10.1039/c9ra01251f] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/10/2019] [Indexed: 12/16/2022] Open
Abstract
Dietary alfalfa fiber (AF) is conceived to modulate gut microbial richness and diversity to improve the health and growth of weaning piglets. The objective of this study was to evaluate the prebiotic effects of AF on diarrhea incidence, the production of short-chain fatty acids (SCFAs), and microbiota composition in weaning piglets. This study utilized 100 crossbred piglets (Duroc × Landrace × Yorkshire) with a body weight of 8.42 ± 1.88 kg randomly assigned to the following treatments: 0.00% AF meal (A), 6.00% of AF meal (B), 12.00% AF meal (C), and 18.00% AF meal (D). The cecum samples were used to determine microbial community composition and diversity through high-throughput 16S rDNA sequencing. The results of this study show that the lowest average daily gain (ADG) was observed in treatment D, and the highest ADG was recorded in treatment C. However there was no significant difference between the treatment groups and the control. The average daily feed intake (ADFI) was significantly higher in treatment C compared to the other treatments. The feed conversion ratio was high in the control group compared to the AF treated groups. The highest diarrhea incidence was observed in treatment A and the lowest diarrhea incidence was observed in treatment C and D. The highest acetate and propionate levels were observed in treatment B, but there was no significant difference between the treatment groups and the control. The supplementation of AF significantly increased the butyrate level in treatment D compared with treatments A and B but was not significantly different from treatment C. The Observed_species richness and Simpson diversity values of the cecum bacterial composition in the AF fed piglets were higher than the control. In addition, the Chao 1 richness and Shannon diversity increased with an increase in AF supplementation, reaching a plateau at treatment B and C, then decreasing at treatment D. The Bacteroidetes, Firmicutes, Tenericutes, Proteobacteria, Cyanobacteria, Spirochaetae, Actinobacteria, Fibrobacteres, Saccharibacteria, Synergistetes, Chlamydiae, Elusimicrobia, Deferribacteres, Fusobacteria, and others were relatively abundant in all treatments. The Bacteroidetes and Firmicutes were the dominant phyla, accounting for 98% of all reads. AF treatment decreased the Bacteroidetes phylum and increased the Firmicutes phylum compared with treatment A. Therefore, the dietary inclusion of AF may decrease diarrhea incidence, increase cecal bacterial composition and richness, and consequently improve the growth performance of weaning piglets.
Collapse
Affiliation(s)
- Seidu Adams
- College of Animal Science and Technology, Jilin Agricultural University Changchun 130118 China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University Changchun 130118 China
| | - Kong Xiangjie
- College of Animal Science and Technology, Jilin Agricultural University Changchun 130118 China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University Changchun 130118 China
| | - Jiang Hailong
- College of Animal Science and Technology, Jilin Agricultural University Changchun 130118 China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University Changchun 130118 China
- Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University Changchun 130118 China
| | - Qin Guixin
- College of Animal Science and Technology, Jilin Agricultural University Changchun 130118 China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University Changchun 130118 China
- Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University Changchun 130118 China
| | | | - Che Dongsheng
- College of Animal Science and Technology, Jilin Agricultural University Changchun 130118 China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University Changchun 130118 China
- Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University Changchun 130118 China
| |
Collapse
|
220
|
Ali Mubaraki M, Ahmad M, Hafiz TA, Marie MA. The therapeutic prospect of crosstalk between prokaryotic and eukaryotic organisms in the human gut. FEMS Microbiol Ecol 2019; 94:4966977. [PMID: 29796663 DOI: 10.1093/femsec/fiy065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/09/2018] [Indexed: 12/19/2022] Open
Abstract
The peaceful phenomenon of the co-evolution between the prokaryotes (microbiota) and the eukaryotes (parasites including protozoa and helminths) in the animal gut has drawn the researchers' attention. Importantly, exploring the potential of helminths for therapeutic uses was one of the reasons behind understanding the physiological and immunological crosstalk existing between them. Here we discuss the interactive immunological associations of helminths and microbial responses individually and in combination with their hosts. Considering that there is probably crosstalk between eukaryotic organisms like helminths and protozoa with their host's gut microbiota, in this review we searched the literature identifying the privileged and favourable relationship generated between them in the host. Understanding the possibilities of the role of helminths along with gut microbiota as a black box would certainly help decode the therapeutic intrusion with helminths in experimental clinical trials, and a successful trial could be used to consider possible future and safe treatments for various immune-inflammatory diseases in humans.
Collapse
Affiliation(s)
- Murad Ali Mubaraki
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud University, Saudi Arabia
| | - Mohammad Ahmad
- Medical Surgical Nursing Department, College of Nursing, King Saud University, Saudi Arabia
| | - Taghreed A Hafiz
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud University, Saudi Arabia
| | - Mohammed A Marie
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud University, Saudi Arabia
| |
Collapse
|
221
|
Xavier R, Mazzei R, Pérez-Losada M, Rosado D, Santos JL, Veríssimo A, Soares MC. A Risky Business? Habitat and Social Behavior Impact Skin and Gut Microbiomes in Caribbean Cleaning Gobies. Front Microbiol 2019; 10:716. [PMID: 31024495 PMCID: PMC6467100 DOI: 10.3389/fmicb.2019.00716] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/21/2019] [Indexed: 11/21/2022] Open
Abstract
The broadstripe cleaning goby Elacatinus prochilos has two alternative ecotypes: sponge-dwellers, which live in large groups and feed mainly upon nematode parasites; and coral-dwellers, that live in small groups or in solitude and behave as cleaners. Recent studies focusing on the skin and gut microbiomes of tropical fish showed that microbial communities are influenced mainly by diet and host species. Here, we compare the skin and gut microbiomes of the Caribbean broadstripe cleaning goby E. prochilos alternative ecotypes (cleaners and non-cleaners) from Barbados and predict that different habitat use and behavior (cleaning vs. non-cleaning) will translate in different bacterial profiles between the two ecotypes. We found significant differences in both alpha- and beta-diversity of skin and gut microbiomes belonging to different ecotypes. Importantly, the skin microbiome of obligate cleaners showed greater intra-sample diversity and harbored a significantly higher prevalence of potential fish pathogens. Likewise, potential pathogens were also more prevalent in the gut of obligate cleaners. We suggest that habitat use, diet, but also direct contact with potential diseased clientele during cleaning, could be the cause for these patterns.
Collapse
Affiliation(s)
- Raquel Xavier
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos da Universidade do Porto, Porto, Portugal
| | - Renata Mazzei
- Laboratoire d’Eco-Ethologie, Institut de Biologie, Université de Neuchâtel, Neuchâtel, Switzerland
| | - Marcos Pérez-Losada
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos da Universidade do Porto, Porto, Portugal
- Computational Biology Institute, Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, The George Washington University, Ashburn, VA, United States
| | - Daniela Rosado
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos da Universidade do Porto, Porto, Portugal
| | - Joana L. Santos
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos da Universidade do Porto, Porto, Portugal
| | - Ana Veríssimo
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos da Universidade do Porto, Porto, Portugal
| | - Marta C. Soares
- CIBIO/InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos da Universidade do Porto, Porto, Portugal
| |
Collapse
|
222
|
Kim WH, Lillehoj HS. Immunity, immunomodulation, and antibiotic alternatives to maximize the genetic potential of poultry for growth and disease response. Anim Feed Sci Technol 2019. [DOI: 10.1016/j.anifeedsci.2018.09.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
223
|
Sun M, Ma N, He T, Johnston LJ, Ma X. Tryptophan (Trp) modulates gut homeostasis via aryl hydrocarbon receptor (AhR). Crit Rev Food Sci Nutr 2019; 60:1760-1768. [PMID: 30924357 DOI: 10.1080/10408398.2019.1598334] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The intestinal homeostasis is an orchestrated dynamic equilibrium state composed of the coexistence and interactions among the nutrients, microbial flora, and immune system. The intestinal balance disorder can trigger a series of diseases, such as inflammatory bowel disease (IBD). Many of tryptophan (Trp) metabolites, such as kynurenine and indole, generated under a series of endogenous enzymes or microbial metabolism, have been reported enable to bind and activate the aryl hydrocarbon receptor (AhR), this series of process is termed the Trp-AhR pathway. The activated Trp-AhR pathway can induce the expression of downstream cytokines such as interleukin-22 (IL-22) and interleukin-17 (IL-17), thereby regulating the intestinal homeostasis. This review highlights the advance of Trp-AhR pathway in the regulation of intestinal homeostasis and provides some insights for the clinical strategies that expect to effectively prevent and treat gut diseases via intervening the Trp-AhR pathway.
Collapse
Affiliation(s)
- Meige Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lee J Johnston
- Swine Nutrition and Production, West Central Research and Outreach Center, University of Minnesota, Morris, MN, USA
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
- Department of Internal Medicine Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
224
|
Saito K, Kuroda K, Suzuki R, Kino Y, Sekine T, Shinoda H, Yamashiro H, Fukuda T, Kobayashi J, Abe Y, Nishimura J, Urushihara Y, Yoneyama H, Fukumoto M, Isogai E. Intestinal Bacteria as Powerful Trapping Lifeforms for the Elimination of Radioactive Cesium. Front Vet Sci 2019; 6:70. [PMID: 30915344 PMCID: PMC6422879 DOI: 10.3389/fvets.2019.00070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/18/2019] [Indexed: 11/13/2022] Open
Abstract
In March 2011, an accident at the Fukushima Daiichi Nuclear Power Plant led to major problems, including the release of radionuclides such as Cesium (Cs)-137 into the environment. Ever since this accident, Cs-137 in foods has become a serious problem. In this study, we determined the concentration of Cs-137 in the feces, urine, and ruminal contents of cattle and demonstrated the possibility of its elimination from the body by intestinal bacteria. The results revealed a high Cs-137 concentration in the feces; in fact, this concentration was higher than that in skeletal muscles and other samples from several animals. Furthermore, intestinal bacteria were able to trap Cs-137, showing an uptake ratio within the range of 38–81% in vitro. This uptake appeared to be mediated through the sodium–potassium (Na+-K+) ion pump in the bacterial cell membrane. This inference was drawn based on the fact that the uptake ratio of Cs-137 was decreased in media with high potassium concentration. In addition, it was demonstrated that intestinal bacteria hindered the trapping of Cs-137 by the animal. Cattle feces showed high concentration of Cs-137 and intestinal bacteria trapped Cs-137. This study is the first report showing that intestinal bacteria contribute to the elimination of Cs-137 from the body.
Collapse
Affiliation(s)
- Kazuki Saito
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kengo Kuroda
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Rie Suzuki
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yasushi Kino
- Department of Chemistry, Tohoku University, Sendai, Japan
| | - Tsutomu Sekine
- Center for the Advancement of Higher Education, Tohoku University, Sendai, Japan
| | - Hisashi Shinoda
- Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Hideaki Yamashiro
- Graduate School of Science and Technology, Niigata University, Niigata, Japan
| | - Tomokazu Fukuda
- Faculty of Science and Engineering, Iwate University, Morioka, Japan
| | - Jin Kobayashi
- School of Food, Agricultural and Environmental Sciences, Miyagi University, Sendai, Japan
| | - Yasuyuki Abe
- Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Junko Nishimura
- Department of Biotechnology and Environmental Engineering, Faculty of Engineering, Hachinohe Institute of Technology, Hachinohe, Japan
| | - Yusuke Urushihara
- Department of Radiation Biology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hiroshi Yoneyama
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | | | - Emiko Isogai
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
225
|
Zhao Z, Qu W, Wang K, Chen S, Zhang L, Wu D, Chen Z. Bisphenol A inhibits mucin 2 secretion in intestinal goblet cells through mitochondrial dysfunction and oxidative stress. Biomed Pharmacother 2019; 111:901-908. [DOI: 10.1016/j.biopha.2019.01.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 01/14/2023] Open
|
226
|
Huang A, Cai R, Wang Q, Shi L, Li C, Yan H. Dynamic Change of Gut Microbiota During Porcine Epidemic Diarrhea Virus Infection in Suckling Piglets. Front Microbiol 2019; 10:322. [PMID: 30858839 PMCID: PMC6397872 DOI: 10.3389/fmicb.2019.00322] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/07/2019] [Indexed: 12/15/2022] Open
Abstract
Porcine epidemic diarrhea (PED) is a disease that has a devastating effect on livestock. Currently, most studies are focused on comparing gut microbiota of healthy piglets and piglets with PED, resulting in gut microbial populations related to dynamic change in diarrheal piglets being poorly understood. The current study analyzed the characteristics of gut microbiota in porcine epidemic diarrhea virus (PEDV)-infected piglets during the suckling transition stage. Fresh fecal samples were collected from 1 to 3-week-old healthy piglets (n = 20) and PEDV infected piglets (n = 18) from the same swine farm. Total DNA was extracted from each sample and the V3-V4 hypervariable region of the 16S rRNA gene was amplified and sequenced using the Illumina MiSeq platform. Statistically significant differences were observed in bacterial diversity and richness between the healthy and diarrheal piglets. Principal coordinates analysis (PCoA) showed structural segregation between diseased and healthy groups, as well as among 3 different age groups. The abundance of Escherichia-Shigella, Enterococcus, Fusobacterium, and Veillonella increased due to dysbiosis induced by PEDV infection. Notably, there was a remarkable age-related increase in Fusobacterium and Veillonella in diarrheal piglets. Certain SCFA-producing bacteria, such as Ruminococcaceae_UCG-002, Butyricimonas, and Alistipes, were shared by all healthy piglets, but were not identified in various age groups of diarrheal piglets. In addition, significant differences were observed between clusters of orthologous groups (COG) functional categories of healthy and PEDV-infected piglets. Our findings demonstrated that PEDV infection caused severe perturbations in porcine gut microbiota. Therefore, regulating gut microbiota in an age-related manner may be a promising method for the prevention or treatment of PEDV.
Collapse
Affiliation(s)
- Anni Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Rujian Cai
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
| | - Qun Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Lei Shi
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China
| | - Chunling Li
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Guangzhou, China.,Guangdong Open Laboratory of Veterinary Public Health, Guangzhou, China
| | - He Yan
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
227
|
Haas OA. Primary Immunodeficiency and Cancer Predisposition Revisited: Embedding Two Closely Related Concepts Into an Integrative Conceptual Framework. Front Immunol 2019; 9:3136. [PMID: 30809233 PMCID: PMC6379258 DOI: 10.3389/fimmu.2018.03136] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022] Open
Abstract
Common understanding suggests that the normal function of a "healthy" immune system safe-guards and protects against the development of malignancies, whereas a genetically impaired one might increase the likelihood of their manifestation. This view is primarily based on and apparently supported by an increased incidence of such diseases in patients with specific forms of immunodeficiencies that are caused by high penetrant gene defects. As I will review and discuss herein, such constellations merely represent the tip of an iceberg. The overall situation is by far more varied and complex, especially if one takes into account the growing difficulties to define what actually constitutes an immunodeficiency and what defines a cancer predisposition. The enormous advances in genome sequencing, in bioinformatic analyses and in the functional in vitro and in vivo assessment of novel findings together with the availability of large databases provide us with a wealth of information that steadily increases the number of sequence variants that concur with clinically more or less recognizable immunological problems and their consequences. Since many of the newly identified hard-core defects are exceedingly rare, their tumor predisposing effect is difficult to ascertain. The analyses of large data sets, on the other hand, continuously supply us with low penetrant variants that, at least in statistical terms, are clearly tumor predisposing, although their specific relevance for the respective carriers still needs to be carefully assessed on an individual basis. Finally, defects and variants that affect the same gene families and pathways in both a constitutional and somatic setting underscore the fact that immunodeficiencies and cancer predisposition can be viewed as two closely related errors of development. Depending on the particular genetic and/or environmental context as well as the respective stage of development, the same changes can have either a neutral, predisposing and, in some instances, even a protective effect. To understand the interaction between the immune system, be it "normal" or "deficient" and tumor predisposition and development on a systemic level, one therefore needs to focus on the structure and dynamic functional organization of the entire immune system rather than on its isolated individual components alone.
Collapse
Affiliation(s)
- Oskar A. Haas
- Department of Clinical Genetics, Children's Cancer Research Institute, Vienna, Austria
| |
Collapse
|
228
|
Yao Q, Yang H, Wang X, Wang H. Effects of hexavalent chromium on intestinal histology and microbiota in Bufo gargarizans tadpoles. CHEMOSPHERE 2019; 216:313-323. [PMID: 30384300 DOI: 10.1016/j.chemosphere.2018.10.147] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 06/08/2023]
Abstract
Chromium is detrimental heavy metal pollutants and can enter and affect aquatic organisms. In our experiment, B. gargarizans embryos were chronically exposed in different concentrations of Cr (VI) (0, 13, 104, and 416 μg Cr6+ L-1) until reached Gosner stage 38. We measured morphological parameters of the body and intestine of B. gargarizans tadpoles, and examined alteration of intestinal tissue. Furthermore, we analyzed the intestinal microbial community of B. gargarizans tadpoles using 16S rRNA gene amplicon sequencing. Our research demonstrated that Cr (VI) exposure caused alteration of intestinal tissue structure in 416 μg Cr6+ L-1 treatment groups. Total body length, body wet weight, intestinal length and wet weight of B. gargarizans tadpoles were significantly declined at 416 μg Cr6+ L-1. In addition, 16S rRNA gene sequencing revealed that Cr (VI) exposure significantly altered the intestinal microbiota diversity and composition, and perturbed the community structure of the microbiota. As for the intestinal microbiota, at the phylum level, Fusobacteria significantly changed in all Cr (VI) treated groups. Saccharibacteria and TM6_Dependentiae were detected only in the high dose exposure groups. At the genus level, Aeromonas was significantly decreased in Cr (VI) treated groups. According to the results of functional prediction, Cr (VI) exposure affected metabolism and increased risk of disease by inducing the alterations of intestinal microbiota structure. Taken together, the present study provide a new framework elucidating the toxic effects Cr (VI) exposure on B. gargarizans tadpoles associated with intestinal histology and microbiota.
Collapse
Affiliation(s)
- Qiong Yao
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China
| | - Hongyu Yang
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China
| | - Xianchan Wang
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China
| | - Hongyuan Wang
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China.
| |
Collapse
|
229
|
Embregts CWE, Reyes-Lopez F, Pall AC, Stratmann A, Tort L, Lorenzen N, Engell-Sorensen K, Wiegertjes GF, Forlenza M, Sunyer JO, Parra D. Pichia pastoris yeast as a vehicle for oral vaccination of larval and adult teleosts. FISH & SHELLFISH IMMUNOLOGY 2019; 85:52-60. [PMID: 30016686 DOI: 10.1016/j.fsi.2018.07.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 06/08/2023]
Abstract
Oral vaccination is of major interest because it can be used for mass vaccination of fish of various size and age. Given that their administration is relatively easy and stress-free, oral vaccines have both economic and animal welfare benefits. Yet, mostly due to their limited efficacy, only very few oral vaccines are available to aquaculture industry. Here we present a method for oral vaccine delivery based on the yeast Pichia pastoris. We could express a model antigen, green fluorescent protein (GFP), in this yeast and subsequently show delivery of the GFP protein to the intestine of juvenile flounder or adult carp and trout. We tested this approach in several commercially-relevant fish species, from juvenile to adult stage. To test the oral delivery of antigen to larval fish, the GFP-expressing Pichia pastoris was first fed to planktonic crustacean Daphnia or rotifers that served as 'bioencapsulation vehicles' and afterwards, fed to flounder larvae. Again, we could show delivery of intact GFP protein to the intestine. In rainbow trout, the orally-administered GFP-expressing yeast elicited a rapid local innate immune response in the intestine and a subsequent systemic response in the spleen. Our results show that Pichia pastoris is a good vehicle for oral antigen delivery and that it can be used in non-encapsulated form for older fish or in bioencapsulated form for larval fish. We discuss the immunomodulatory properties of the yeast itself, and its potential to enhance local immune responses and act as an adjuvant.
Collapse
Affiliation(s)
- Carmen W E Embregts
- Cell Biology and Immunology Group, Department of Animal Sciences, Wageningen University & Research, 6708 WD, Wageningen, the Netherlands
| | - Felipe Reyes-Lopez
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Adina C Pall
- Fishlab, Terp Skovvej 107B, DK 8270, Højbjerg, Denmark
| | - Ansgar Stratmann
- W42 Industrial Biotechnology GmbH, BMZ Dortmund, Otto-Hahn-Straße 15, D-44227, Dortmund, Germany
| | - Luis Tort
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Niels Lorenzen
- Department of Animal Science, Aarhus University, Aarhus, Denmark; Technical University of Denmark, Denmark
| | | | - Geert F Wiegertjes
- Cell Biology and Immunology Group, Department of Animal Sciences, Wageningen University & Research, 6708 WD, Wageningen, the Netherlands; Aquaculture and Fisheries Group, Department of Animal Sciences, Wageningen University & Research, 6708 WD, Wageningen, the Netherlands
| | - Maria Forlenza
- Cell Biology and Immunology Group, Department of Animal Sciences, Wageningen University & Research, 6708 WD, Wageningen, the Netherlands
| | - J Oriol Sunyer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David Parra
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
| |
Collapse
|
230
|
Lavoie S, Conway KL, Lassen KG, Jijon HB, Pan H, Chun E, Michaud M, Lang JK, Gallini Comeau CA, Dreyfuss JM, Glickman JN, Vlamakis H, Ananthakrishnan A, Kostic A, Garrett WS, Xavier RJ. The Crohn's disease polymorphism, ATG16L1 T300A, alters the gut microbiota and enhances the local Th1/Th17 response. eLife 2019; 8:39982. [PMID: 30666959 PMCID: PMC6342529 DOI: 10.7554/elife.39982] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 12/20/2018] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel disease (IBD) is driven by dysfunction between host genetics, the microbiota, and immune system. Knowledge gaps remain regarding how IBD genetic risk loci drive gut microbiota changes. The Crohn's disease risk allele ATG16L1 T300A results in abnormal Paneth cells due to decreased selective autophagy, increased cytokine release, and decreased intracellular bacterial clearance. To unravel the effects of ATG16L1 T300A on the microbiota and immune system, we employed a gnotobiotic model using human fecal transfers into ATG16L1 T300A knock-in mice. We observed increases in Bacteroides ovatus and Th1 and Th17 cells in ATG16L1 T300A mice. Association of altered Schaedler flora mice with B. ovatus specifically increased Th17 cells selectively in ATG16L1 T300A knock-in mice. Changes occur before disease onset, suggesting that ATG16L1 T300A contributes to dysbiosis and immune infiltration prior to disease symptoms. Our work provides insight for future studies on IBD subtypes, IBD patient treatment and diagnostics.
Collapse
Affiliation(s)
- Sydney Lavoie
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, United States.,Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, United States
| | - Kara L Conway
- Gastrointestinal Unit, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, United States
| | - Kara G Lassen
- Broad Institute of Harvard and MIT, Cambridge, United States.,Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, United States
| | - Humberto B Jijon
- Gastrointestinal Unit, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, United States
| | - Hui Pan
- Joslin Diabetes Center, Boston, United States
| | - Eunyoung Chun
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, United States.,Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, United States
| | - Monia Michaud
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, United States.,Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, United States
| | - Jessica K Lang
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, United States.,Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, United States
| | - Carey Ann Gallini Comeau
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, United States.,Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, United States
| | | | - Jonathan N Glickman
- Department of Pathology, Harvard Medical School, Boston, United States.,Beth Israel Deaconess Medical Center, Boston, United States
| | - Hera Vlamakis
- Broad Institute of Harvard and MIT, Cambridge, United States
| | - Ashwin Ananthakrishnan
- Gastrointestinal Unit, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, United States
| | - Aleksander Kostic
- Joslin Diabetes Center, Boston, United States.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, United States.,Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, United States.,Broad Institute of Harvard and MIT, Cambridge, United States.,Department and Division of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, United States
| | - Ramnik J Xavier
- Gastrointestinal Unit, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, United States.,Broad Institute of Harvard and MIT, Cambridge, United States
| |
Collapse
|
231
|
Roles of Autophagy-Related Genes in the Pathogenesis of Inflammatory Bowel Disease. Cells 2019; 8:cells8010077. [PMID: 30669622 PMCID: PMC6356351 DOI: 10.3390/cells8010077] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 12/13/2022] Open
Abstract
Autophagy is an intracellular catabolic process that is essential for a variety of cellular responses. Due to its role in the maintenance of biological homeostasis in conditions of stress, dysregulation or disruption of autophagy may be linked to human diseases such as inflammatory bowel disease (IBD). IBD is a complicated inflammatory colitis disorder; Crohn’s disease and ulcerative colitis are the principal types. Genetic studies have shown the clinical relevance of several autophagy-related genes (ATGs) in the pathogenesis of IBD. Additionally, recent studies using conditional knockout mice have led to a comprehensive understanding of ATGs that affect intestinal inflammation, Paneth cell abnormality and enteric pathogenic infection during colitis. In this review, we discuss the various ATGs involved in macroautophagy and selective autophagy, including ATG16L1, IRGM, LRRK2, ATG7, p62, optineurin and TFEB in the maintenance of intestinal homeostasis. Although advances have been made regarding the involvement of ATGs in maintaining intestinal homeostasis, determining the precise contribution of autophagy has remained elusive. Recent efforts based on direct targeting of ATGs and autophagy will further facilitate the development of new therapeutic opportunities for IBD.
Collapse
|
232
|
Suh SS, Hong JM, Kim EJ, Jung SW, Chae H, Kim JE, Kim JH, Kim IC, Kim S. Antarctic freshwater microalga, Chloromonas reticulata, suppresses inflammation and carcinogenesis. Int J Med Sci 2019; 16:189-197. [PMID: 30745798 PMCID: PMC6367532 DOI: 10.7150/ijms.30647] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammation triggered by the innate immune system is a strategy to protect organisms from the risk of environmental infection. However, it has recently become clear that inflammation can cause a variety of human diseases, including cancer. In this study, we investigated the effects of an ethanol extract of the Antarctic freshwater microalgae, Chloromonas reticulata (ETCH), on inflammation and carcinogenesis in RAW 264.7 macrophages and HCT116 human colon cancer cells, respectively. ETCH exhibited significant anti-inflammatory activity through the dose-dependent modulation of major inflammatory markers such as COX-2, IL-6, iNOS, TNF-α, and NO production. For example, ETCH reduced LPS-induced upregulation of COX-2, IL-6, iNOS, and TNF- alpha mRNA levels, leading to a significant decrease in the levels of LPS-stimulated NO and IL-6 as well as TNF-alpha products. In contract, ETCH exhibited dose-dependent cytotoxic activity against HCT116 cells, yielding a profound reduction in the proliferation of the cancer cells. Furthermore, ETCH induced G2 phase cell cycle arrest by transcriptionally regulating of genes involved in G2 / M transition including p21 (CDKN1A), cyclin B1 (CCNB1), and CDK1; CDKN1A mRNA levels were upregulated in response to ETCH, whereas CCNB1 and CDK1 were downregulated. This study reports for the first time anti-inflammatory and anti-cancer effects of, C. reticulata and provides new insights into the molecular mechanisms of the linkage between inflammation and cancer.
Collapse
Affiliation(s)
- Sung-Suk Suh
- Department of Bioscience, Mokpo National University, Muan 58554, Republic of Korea
| | - Ju-Mi Hong
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, 21990, Republic of Korea
| | - Eun Jae Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, 21990, Republic of Korea
| | - Seung Won Jung
- South Sea Environment Research Department, Korea Institute of Ocean Science and Technology, Geoje, 656-830, Republic of Korea
| | - Hyunsik Chae
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, 21990, Republic of Korea
| | - Jung Eun Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, 21990, Republic of Korea.,Department of Pharmacy, Graduate School, Sungkyunkwan University, Suwan 16419, Republic of Korea
| | - Ji Hee Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, 21990, Republic of Korea.,Department of Polar Sciences, University of Science and Technology, Incheon 21990, Republic of Korea
| | - Il-Chan Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, 21990, Republic of Korea.,Department of Polar Sciences, University of Science and Technology, Incheon 21990, Republic of Korea
| | - Sanghee Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, 21990, Republic of Korea.,Department of Polar Sciences, University of Science and Technology, Incheon 21990, Republic of Korea
| |
Collapse
|
233
|
Peng Y, Li H, Chen D. Silencing astrocyte elevated gene-1 attenuates lipopolysaccharide-induced inflammation and mucosal barrier injury in NCM460 cells by suppressing the activation of NLRP3 inflammasome. Cell Biol Int 2018; 43:56-64. [PMID: 30489008 DOI: 10.1002/cbin.11078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/24/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Peng
- Emergency Department; Jingmen No. 1 People's Hospital; No. 168 Xiangshan Road Jingmen Hubei Province 448000 China
| | - Hongyan Li
- Department of Thyroid and Breast Surgery; Jingmen No. 1 People's Hospital; No. 168 Xiangshan Road Jingmen Hubei Province 448000 China
| | - Dandan Chen
- Department of General Surgery; Jingmen No. 1 People's Hospital; No. 168 Xiangshan Road Jingmen Hubei Province 448000 China
| |
Collapse
|
234
|
Iida T, Yokoyama Y, Wagatsuma K, Hirayama D, Nakase H. Impact of Autophagy of Innate Immune Cells on Inflammatory Bowel Disease. Cells 2018; 8:cells8010007. [PMID: 30583538 PMCID: PMC6356773 DOI: 10.3390/cells8010007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 12/13/2022] Open
Abstract
Autophagy, an intracellular degradation mechanism, has many immunological functions and is a constitutive process necessary for maintaining cellular homeostasis and organ structure. One of the functions of autophagy is to control the innate immune response. Many studies conducted in recent years have revealed the contribution of autophagy to the innate immune response, and relationships between this process and various diseases have been reported. Inflammatory bowel disease is an intractable disorder with unknown etiology; however, immunological abnormalities in the intestines are known to be involved in the pathology of inflammatory bowel disease, as is dysfunction of autophagy. In Crohn's disease, many associations with autophagy-related genes, such as ATG16L1, IRGM, NOD2, and others, have been reported. Abnormalities in the ATG16L1 gene, in particular, have been reported to cause autophagic dysfunction, resulting in enhanced production of inflammatory cytokines by macrophages as well as abnormal function of Paneth cells, which are important in intestinal innate immunity. In this review, we provide an overview of the autophagy mechanism in innate immune cells in inflammatory bowel disease.
Collapse
Affiliation(s)
- Tomoya Iida
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| | - Yoshihiro Yokoyama
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| | - Kohei Wagatsuma
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| | - Daisuke Hirayama
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo 060-8543, Japan.
| |
Collapse
|
235
|
Zhou L, Sonnenberg GF. Essential immunologic orchestrators of intestinal homeostasis. Sci Immunol 2018; 3:3/20/eaao1605. [PMID: 29440266 DOI: 10.1126/sciimmunol.aao1605] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/21/2017] [Indexed: 12/14/2022]
Abstract
Over the past 25 years, substantial advances have been made in our understanding of the cellular and molecular pathways that are essential to maintain a state of health in the mammalian gastrointestinal tract, an organ that is densely colonized by both immune cells and trillions of microbes. Seminal studies in the 1990s identified that several cytokines, antigen-presentation molecules, and components of the T cell receptor were necessary to prevent the development of spontaneous intestinal inflammation in mice. Subsequent research revealed that these pathways orchestrate beneficial interactions with intestinal microbes, involve complex communication between innate and adaptive immune cells, and can be dysregulated in human inflammatory bowel disease. Here, we discuss how these early findings set the stage for numerous other advances and shaped our current knowledge of host-microbiota interactions and intestinal homeostasis in mammals. It is expected that continued investigation of these areas will define previously unknown immunologic mechanisms of tolerance and inflammation in the intestine that can be exploited to benefit human health.
Collapse
Affiliation(s)
- Lei Zhou
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA. .,Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| |
Collapse
|
236
|
Arenas‐Padilla M, Duarte‐Gutiérrez J, Mata‐Haro V. Bifidobacterium animalis ssp. lactis Bb12 induces IL-10 through cell membrane-associated components via TLR2 in swine. J Appl Microbiol 2018; 125:1881-1889. [PMID: 30106205 PMCID: PMC7166459 DOI: 10.1111/jam.14069] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/06/2018] [Accepted: 08/09/2018] [Indexed: 01/22/2023]
Abstract
AIM To investigate the role of Toll-like receptor 2 (TLR2) in interleukin-10 (IL-10) production induced by Bifidobacterium animalis ssp. lactis Bb12 (Bb12) in swine immune cells. METHODS AND RESULTS Blood-monocytes and cells from mesenteric lymph nodes were obtained from pigs and cultured with live Bb12 for 4 and 12 h. Transcript levels of IL-10 and TLR2 were analysed. Furthermore, TLR2 was blocked to determine its participation in IL-10 production. TLR2 blockade was achieved with neutralizing antibodies, followed by stimulation with Bb12. Bifidobacteria induced IL-10 production in both swine monocytes and mesenteric cells. Monocytes with TLR2 blockade had a decrease in IL-10 transcripts, while mesenteric cells did not. Bacterial cell wall components were responsible for Bb12-induced IL-10 production since no IL-10 was detected in the culture supernatant. CONCLUSIONS We demonstrated that IL-10 production is largely mediated through the recognition of Bb12 structures by TLR2, as bacterial metabolites in the culture supernatant failed to induce IL-10 expression. SIGNIFICANCE AND IMPACT OF THE STUDY The present study provides evidence for the potential use of Bb12 in the swine industry; these bacteria can also be used as additional method to treat intestinal inflammation and enhance intestinal health in pigs.
Collapse
Affiliation(s)
- M. Arenas‐Padilla
- Department of Food Science, Microbiology and ImmunologyCentro de Investigación en Alimentación y Desarrollo, A. C.HermosilloMéxico
| | - J.L. Duarte‐Gutiérrez
- Department of Food Science, Microbiology and ImmunologyCentro de Investigación en Alimentación y Desarrollo, A. C.HermosilloMéxico
| | - V. Mata‐Haro
- Department of Food Science, Microbiology and ImmunologyCentro de Investigación en Alimentación y Desarrollo, A. C.HermosilloMéxico
| |
Collapse
|
237
|
Zhang Q, Li C, Niu X, Zhang Z, Li F, Li F. An intensive milk replacer feeding program benefits immune response and intestinal microbiota of lambs during weaning. BMC Vet Res 2018; 14:366. [PMID: 30477479 PMCID: PMC6258415 DOI: 10.1186/s12917-018-1691-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/09/2018] [Indexed: 12/31/2022] Open
Abstract
Background Pre-weaning milk replacer (MR) feeding program is a key factor affecting the health and welfare of lambs during their weaning. Weaning stress is well known as an inducement that negatively impacts the immune system of young ruminants, whose physiological and immune state is closely linked to the community of microbiota in their intestines. This study had two objectives: 1) To evaluate the innate immune response to weaning stress at both the physiological and molecular level; 2) To investigate changes to the jejunal chyme and mucosal adhesive microbiota between the control and high plane of MR groups. Results In this experiment, the plasma concentrations of cortisol, norepinephrine (NE) and tumor necrosis factor-α (TNFα) were higher in the C than the H group (P < 0.05), as was the expression of pro-inflammatory cytokines such as TNFα and CXCL8 (P < 0.05) in plasma. In jejunal tissue, the expression of TLR4 and TNFα were also higher in the C group (P < 0.01); histopathology showed the H group had lower lymphocyte infiltration. In the C group, however, major pathological changes were associated with extensive infiltration of lymphocytes, eosinophils, and neutrophils. Principal component analysis indicated the lamb immune response was influenced by weaning stress and modulated by the MR treatments. 16S-rRNA sequencing was used to evaluate jejunal mucosa and chyme bacterial diversity and composition. The C group’s chyme had a greater alpha index (ACE: P = 0.095; Chao1: P = 0.085) than H group. In jejunal mucosa, the relative abundance of Plesiomonas was 4-fold higher (P = 0.017) in the C than the H group. Conclusions This study’s results revealed that weaning stress induced alterations to the lambs’ immune system that lasted beyond the 21 d measured, and that a long-term inflammatory response effect was evidenced by changes in their hematological and expressed pro-inflammatory cytokines. Pre-weaning with a differing MR allowance resulted in complicated biological responses and compositional changes to the lambs’ jejunal microbiota. Clearly, an intensive MR feeding program induced a milder immunity response and lower relative abundance of pathogenic bacteria when compared with the traditional feeding program.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Grassland Agro-ecosystems; Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs; College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Chong Li
- State Key Laboratory of Grassland Agro-ecosystems; Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs; College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China.,College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiaolin Niu
- State Key Laboratory of Grassland Agro-ecosystems; Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs; College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Zhian Zhang
- State Key Laboratory of Grassland Agro-ecosystems; Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs; College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Fadi Li
- State Key Laboratory of Grassland Agro-ecosystems; Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs; College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China.
| | - Fei Li
- State Key Laboratory of Grassland Agro-ecosystems; Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs; College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China.
| |
Collapse
|
238
|
Li Y, Lu X, Wu H, Xia M, Hou Q, Hu W, Li T, Wu L, Yu Q. The effect of dietary supplementation of low crude protein on intestinal morphology in pigs. Res Vet Sci 2018; 122:15-21. [PMID: 30447500 DOI: 10.1016/j.rvsc.2018.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 10/30/2018] [Accepted: 11/11/2018] [Indexed: 12/20/2022]
Abstract
To explore the effects of reducing the Cp levels on intestinal barrier function, low Cp (LP) and NRC standard Cp (NP) diets were fed to pigs from 45 to 160 days, and in vitro experiments were performed using monolayers of IPEC-J2 cells. The number of goblet cells, expression of proteins related to cell junction, amino acid transport, glucose transport, transepithelial electrical resistance (TEER), dextran permeability, and IL-6 secretion level were detected in pigs. The results demonstrated that a moderate reduction of Cp levels did not affect intestinal morphology, as demonstrated by a normal villi height, crypt depth and normal numbers of goblet cells. The maintenance of the intestinal structure obtained with LP was also confirmed by stable mRNA expression levels of muc2 and E-cadherin in the jejunum. We also found that LP did not affect the protein expression of cationic amino acid transporter 1 (CAT-1) and alanine serine cysteine transporter 1 (ASCT1) from 45 to 160 days. Moreover, the excitatory amino acid transporter 3 (EAAT3), sodium-glucose cotransporter 1 (SGLT1) and glucose transporter (GLUT2) protein expression levels in the jejunum were significantly increased at a certain age during the rearing period. Furthermore, we also demonstrated that a reduction in protein concentration up to 15% in the cultural medium of IPEC-J2 cells did not impact the mucosal barrier function. This study demonstrated that a moderate reduction of the protein level did not affect intestinal mucosal barrier function and morphology in the jejunum.
Collapse
Affiliation(s)
- Yunyun Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China
| | - Xiaoxi Lu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China
| | - Haiqin Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China
| | - Mi Xia
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China
| | - Qihang Hou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China
| | - Weiwei Hu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China
| | - Tiejun Li
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, PR China
| | - Li Wu
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, PR China
| | - Qinghua Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China.
| |
Collapse
|
239
|
Iatsenko I, Boquete JP, Lemaitre B. Microbiota-Derived Lactate Activates Production of Reactive Oxygen Species by the Intestinal NADPH Oxidase Nox and Shortens Drosophila Lifespan. Immunity 2018; 49:929-942.e5. [DOI: 10.1016/j.immuni.2018.09.017] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/01/2018] [Accepted: 09/24/2018] [Indexed: 12/22/2022]
|
240
|
Aynacıoğlu AŞ, Bilir A, Tuna MY. Involvement of midkine in autoimmune and autoinflammatory diseases. Mod Rheumatol 2018; 29:567-571. [DOI: 10.1080/14397595.2018.1523701] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Ahmet Şükrü Aynacıoğlu
- Department of Medical Pharmacology, Istanbul Aydin University Medical Faculty, Istanbul, Turkey
| | - Ayhan Bilir
- Department of Histology and Embryology, Istanbul Aydin University Medical Faculty, Istanbul, Turkey
| | - Mehmet Yakup Tuna
- Department of Anatomy, Istanbul Aydin University Medical Faculty, Istanbul, Turkey
| |
Collapse
|
241
|
Liu L, Zeng D, Yang M, Wen B, Lai J, Zhou Y, Sun H, Xiong L, Wang J, Lin Y, Pan K, Jing B, Wang P, Ni X. Probiotic Clostridium butyricum Improves the Growth Performance, Immune Function, and Gut Microbiota of Weaning Rex Rabbits. Probiotics Antimicrob Proteins 2018; 11:1278-1292. [DOI: 10.1007/s12602-018-9476-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
242
|
Kuntal BK, Chandrakar P, Sadhu S, Mande SS. 'NetShift': a methodology for understanding 'driver microbes' from healthy and disease microbiome datasets. ISME JOURNAL 2018; 13:442-454. [PMID: 30287886 DOI: 10.1038/s41396-018-0291-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/05/2018] [Accepted: 09/14/2018] [Indexed: 12/12/2022]
Abstract
The combined effect of mutual association within the co-inhabiting microbes in human body is known to play a major role in determining health status of individuals. The differential taxonomic abundance between healthy and disease are often used to identify microbial markers. However, in order to make a microbial community based inference, it is important not only to consider microbial abundances, but also to quantify the changes observed among inter microbial associations. In the present study, we introduce a method called 'NetShift' to quantify rewiring and community changes in microbial association networks between healthy and disease. Additionally, we devise a score to identify important microbial taxa which serve as 'drivers' from the healthy to disease. We demonstrate the validity of our score on a number of scenarios and apply our methodology on two real world metagenomic datasets. The 'NetShift' methodology is also implemented as a web-based application available at https://web.rniapps.net/netshift.
Collapse
Affiliation(s)
- Bhusan K Kuntal
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., 54-B Hadapsar Industrial Estate, Pune, 411 013, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-National Chemical Laboratory Campus, Pune, 411 008, India
| | - Pranjal Chandrakar
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., 54-B Hadapsar Industrial Estate, Pune, 411 013, India.,Decision Sciences, Indian Institute of Management Bangalore, Bannerghatta Road, Bengaluru, Karnataka, 560076, India
| | - Sudipta Sadhu
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., 54-B Hadapsar Industrial Estate, Pune, 411 013, India
| | - Sharmila S Mande
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., 54-B Hadapsar Industrial Estate, Pune, 411 013, India.
| |
Collapse
|
243
|
Watanabe K, Petri WA. Learning from the research on amebiasis and gut microbiome: Is stimulation by gut flora essential for effective neutrophil mediated protection from external pathogens? Gut Microbes 2018; 10:100-104. [PMID: 30252579 PMCID: PMC6363069 DOI: 10.1080/19490976.2018.1479626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 05/14/2018] [Indexed: 02/03/2023] Open
Abstract
Amebiasis, caused by intestinal infection with Entamoeba histolytica, is one of the leading causes of parasite infection-related mortality and morbidity globally. Although its pathogenesis, including determinant factors of infection outcome, remains unclear, recent clinical data indicate that the gut microbiome plays a role in determining the severity of amebiasis. Recently, we investigated the effects of the gut microbiome on neutrophil mediated protection from E. histolytica infection using a mouse model. We identified that surface expression of CXCR2 on neutrophils was diminished in mice with dysbiosis, which resulted in decreased neutrophil recruitment to the infection site, allowing more aggressive intestinal tissue damage by E. histolytica. Our results indicated that oxidase activity during E. histolytica infection was also diminished after dysbiosis, consistent with the results from prior research. Thus, the gut microbiome plays an important role in regulating neutrophil phenotype when fighting against external pathogens.
Collapse
Affiliation(s)
- Koji Watanabe
- a AIDS Clinical Center , National Center for Global Health and Medicine , Tokyo , Japan
| | - William A Petri
- b Division of Infectious Diseases and International Health , University of Virginia , Charlottesville , VA , USA
| |
Collapse
|
244
|
Goltsman DSA, Sun CL, Proctor DM, DiGiulio DB, Robaczewska A, Thomas BC, Shaw GM, Stevenson DK, Holmes SP, Banfield JF, Relman DA. Metagenomic analysis with strain-level resolution reveals fine-scale variation in the human pregnancy microbiome. Genome Res 2018; 28:1467-1480. [PMID: 30232199 PMCID: PMC6169887 DOI: 10.1101/gr.236000.118] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 08/28/2018] [Indexed: 12/22/2022]
Abstract
Recent studies suggest that the microbiome has an impact on gestational health and outcome. However, characterization of the pregnancy-associated microbiome has largely relied on 16S rRNA gene amplicon-based surveys. Here, we describe an assembly-driven, metagenomics-based, longitudinal study of the vaginal, gut, and oral microbiomes in 292 samples from 10 subjects sampled every three weeks throughout pregnancy. Nonhuman sequences in the amount of 1.53 Gb were assembled into scaffolds, and functional genes were predicted for gene- and pathway-based analyses. Vaginal assemblies were binned into 97 draft quality genomes. Redundancy analysis (RDA) of microbial community composition at all three body sites revealed gestational age to be a significant source of variation in patterns of gene abundance. In addition, health complications were associated with variation in community functional gene composition in the mouth and gut. The diversity of Lactobacillus iners-dominated communities in the vagina, unlike most other vaginal community types, significantly increased with gestational age. The genomes of co-occurring Gardnerella vaginalis strains with predicted distinct functions were recovered in samples from two subjects. In seven subjects, gut samples contained strains of the same Lactobacillus species that dominated the vaginal community of that same subject and not other Lactobacillus species; however, these within-host strains were divergent. CRISPR spacer analysis suggested shared phage and plasmid populations across body sites and individuals. This work underscores the dynamic behavior of the microbiome during pregnancy and suggests the potential importance of understanding the sources of this behavior for fetal development and gestational outcome.
Collapse
Affiliation(s)
- Daniela S Aliaga Goltsman
- March of Dimes Prematurity Research Center at Stanford University, Stanford, California 94305, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Christine L Sun
- March of Dimes Prematurity Research Center at Stanford University, Stanford, California 94305, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Diana M Proctor
- Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, USA
| | - Daniel B DiGiulio
- March of Dimes Prematurity Research Center at Stanford University, Stanford, California 94305, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, USA
| | - Anna Robaczewska
- March of Dimes Prematurity Research Center at Stanford University, Stanford, California 94305, USA.,Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, USA
| | - Brian C Thomas
- Department of Earth and Planetary Science, University of California, Berkeley, California 94720, USA
| | - Gary M Shaw
- March of Dimes Prematurity Research Center at Stanford University, Stanford, California 94305, USA.,Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - David K Stevenson
- March of Dimes Prematurity Research Center at Stanford University, Stanford, California 94305, USA.,Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, California 94305, USA
| | - Jillian F Banfield
- Department of Earth and Planetary Science, University of California, Berkeley, California 94720, USA.,Earth and Environmental Science, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | - David A Relman
- March of Dimes Prematurity Research Center at Stanford University, Stanford, California 94305, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.,Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, USA
| |
Collapse
|
245
|
Brugman S, Ikeda-Ohtsubo W, Braber S, Folkerts G, Pieterse CMJ, Bakker PAHM. A Comparative Review on Microbiota Manipulation: Lessons From Fish, Plants, Livestock, and Human Research. Front Nutr 2018; 5:80. [PMID: 30234124 PMCID: PMC6134018 DOI: 10.3389/fnut.2018.00080] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
During recent years the impact of microbial communities on the health of their host (being plants, fish, and terrestrial animals including humans) has received increasing attention. The microbiota provides the host with nutrients, induces host immune development and metabolism, and protects the host against invading pathogens (1-6). Through millions of years of co-evolution bacteria and hosts have developed intimate relationships. Microbial colonization shapes the host immune system that in turn can shape the microbial composition (7-9). However, with the large scale use of antibiotics in agriculture and human medicine over the last decades an increase of diseases associated with so-called dysbiosis has emerged. Dysbiosis refers to either a disturbed microbial composition (outgrowth of possible pathogenic species) or a disturbed interaction between bacteria and the host (10). Instead of using more antibiotics to treat dysbiosis there is a need to develop alternative strategies to combat disturbed microbial control. To this end, we can learn from nature itself. For example, the plant root (or "rhizosphere") microbiome of sugar beet contains several bacterial species that suppress the fungal root pathogen Rhizoctonia solani, an economically important fungal pathogen of this crop (11). Likewise, commensal bacteria present on healthy human skin produce antimicrobial molecules that selectively kill skin pathogen Staphylococcus aureus. Interestingly, patients with atopic dermatitis (inflammation of the skin) lacked antimicrobial peptide secreting commensal skin bacteria (12). In this review, we will give an overview of microbial manipulation in fish, plants, and terrestrial animals including humans to uncover conserved mechanisms and learn how we might restore microbial balance increasing the resilience of the host species.
Collapse
Affiliation(s)
- Sylvia Brugman
- Cell Biology and Immunology Group, Animal Sciences Group, Wageningen University and Research, Wageningen, Netherlands
| | - Wakako Ikeda-Ohtsubo
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, Netherlands
| | - Corné M. J. Pieterse
- Plant-Microbe Interactions, Department of Biology, Science4Life, Utrecht University, Utrecht, Netherlands
| | - Peter A. H. M. Bakker
- Plant-Microbe Interactions, Department of Biology, Science4Life, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
246
|
Qu W, Zhao Z, Chen S, Zhang L, Wu D, Chen Z. Bisphenol A suppresses proliferation and induces apoptosis in colonic epithelial cells through mitochondrial and MAPK/AKT pathways. Life Sci 2018; 208:167-174. [DOI: 10.1016/j.lfs.2018.07.040] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 07/19/2018] [Indexed: 01/26/2023]
|
247
|
Li Y, Fu X, Ma X, Geng S, Jiang X, Huang Q, Hu C, Han X. Intestinal Microbiome-Metabolome Responses to Essential Oils in Piglets. Front Microbiol 2018; 9:1988. [PMID: 30210470 PMCID: PMC6120982 DOI: 10.3389/fmicb.2018.01988] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 08/07/2018] [Indexed: 12/29/2022] Open
Abstract
This study investigated the effects of dietary essential oils (EOs) on intestinal microbial composition and metabolic profiles in weaned piglets. The piglets were fed the same basal diet supplemented with EOs (EO) or without EOs (Con) in the current study. The results showed that the body weight gain was significantly increased, while the diarrhea incidence was significantly reduced in the EO group. In addition, EOs could modify the intestinal microbial composition of weaned piglets. The relative abundances of some beneficial bacterial species such as Bacilli, Lactobacillales, Streptococcaceae, and Veillonellaceae were significantly increased in the EO group. Metabolomics analysis indicated that protein biosynthesis, amino acid metabolism, and lipid metabolism were enriched in the EO group. And correlation analysis demonstrated that some gut bacterial genera were highly correlated with altered gut microbiota-related metabolites. Taken together, this study indicated that dietary EOs not only altered microbial composition and function but modulated the microbial metabolic profiles in the colon, which might help us understand EOs' beneficial effects on intestinal health of weaned piglets.
Collapse
Affiliation(s)
- Yuan Li
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiongfeng Fu
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xin Ma
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Shijie Geng
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xuemei Jiang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qichun Huang
- Department of Life Science, Longyan University, Longyan, China
| | - Caihong Hu
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xinyan Han
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
248
|
Dmitrieva O, Grivennikov SI. Microbiota and cancer: a complex equation with a lot of exciting unknowns. Semin Immunol 2018; 32:1-2. [PMID: 29078919 DOI: 10.1016/j.smim.2017.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Oxana Dmitrieva
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA; Personalized medicine and molecular immunology, National Research Center - Institute of Immunology Federal Medical-Biological Agency of Russia, Moscow, 115478, Russia
| | - Sergei I Grivennikov
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| |
Collapse
|
249
|
Cho A, Jantschitsch C, Knobler R. Extracorporeal Photopheresis-An Overview. Front Med (Lausanne) 2018; 5:236. [PMID: 30211164 PMCID: PMC6119964 DOI: 10.3389/fmed.2018.00236] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/03/2018] [Indexed: 12/23/2022] Open
Abstract
Extracorporeal photopheresis (ECP) has been in clinical use for over three decades after receiving FDA approval for the palliative treatment of the Sézary Syndrome variant of cutaneous T-cell lymphoma (CTCL) in 1988. After the first positive experiences with CTCL, additional indications have been successfully explored including areas such as graft-vs.-host disease (GVHD), scleroderma, and solid organ transplantation. The mechanism of action is still not fully resolved, but important steps in understanding ECP in recent years have been very informative. Originally, the primary hypothesis stated that psoralen and ultraviolet A (UVA) in combination induce apoptosis in the treated immune cells. This view shifted in favor of dendritic cell initiation, modification of the cytokine profile and stimulation of several T-cell lineages, in particular regulatory T-cells. A number of ECP guidelines have been produced to optimize treatment regimens in the clinical context. In CTCL, enough evidence is available for the use of ECP as a first line treatment for Sézary Syndrome (SS), but also as a second line or rescue treatment in therapy-refractory forms of mycosis fungoides (MF). ECP in the treatment of acute and chronic GVHD has shown promising results as second line therapy in steroid-refractory presentations. In solid organ transplantation, ECP has been used to increase tissue tolerance and decrease infections with opportunistic pathogens, attributed to the use of high doses of immunosuppressive medication. Infection with cytomegalovirus (CMV) remains a limiting factor affecting survival in solid organ transplantation and the role of ECP will be discussed in this review. A trend toward prophylactic use of ECP can be observed and may further contribute to improve the outcome in many patients. To further deepen our knowledge of ECP and thus facilitate its use in patients that potentially benefit most from it, future prospective randomized trials are urgently needed in this rapidly growing field. The aim of this review is to (1) introduce the method, (2) give an overview where ECP has shown promising effects and has become an essential part of treatment protocols, and (3) to give recommendations on how to proceed in numerous indications.
Collapse
Affiliation(s)
| | | | - Robert Knobler
- Department of Dermatology, Medizinische Universität Wien, Vienna, Austria
| |
Collapse
|
250
|
Chitin-based barrier immunity and its loss predated mucus-colonization by indigenous gut microbiota. Nat Commun 2018; 9:3402. [PMID: 30143642 PMCID: PMC6109156 DOI: 10.1038/s41467-018-05884-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/02/2018] [Indexed: 02/06/2023] Open
Abstract
Mammalian gut microbiota are integral to host health. However, how this association began remains unclear. We show that in basal chordates the gut space is radially compartmentalized into a luminal part where food microbes pass and an almost axenic peripheral part, defined by membranous delamination of the gut epithelium. While this membrane, framed with chitin nanofibers, structurally resembles invertebrate peritrophic membranes, proteome supports its affinity to mammalian mucus layers, where gut microbiota colonize. In ray-finned fish, intestines harbor indigenous microbes, but chitinous membranes segregate these luminal microbes from the surrounding mucus layer. These data suggest that chitin-based barrier immunity is an ancient system, the loss of which, at least in mammals, provided mucus layers as a novel niche for microbial colonization. These findings provide a missing link for intestinal immune systems in animals, revealing disparate mucosal environment in model organisms and highlighting the loss of a proven system as innovation. The coevolution of the animal gut mucosa and the gut microbiota is poorly understood. Here, Nakashima et al. identify intestinal chitinous membranes in basal chordates and ray-finned fish, and propose that the loss of this chitin barrier allowed mucus layers to become colonized by microbes in mammals.
Collapse
|