301
|
Luo M, Li D, Wang Z, Guo W, Kang L, Zhou S. Juvenile hormone differentially regulates two Grp78 genes encoding protein chaperones required for insect fat body cell homeostasis and vitellogenesis. J Biol Chem 2017; 292:8823-8834. [PMID: 28356351 DOI: 10.1074/jbc.m117.780957] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/27/2017] [Indexed: 11/06/2022] Open
Abstract
Juvenile hormone (JH) has a well known role in stimulating insect vitellogenesis (i.e. yolk deposition) and oocyte maturation, but the molecular mechanisms of JH action in insect reproduction are unclear. The 78-kDa glucose-regulated protein (Grp78) is a heat shock protein 70-kDa family member and one of the most abundant chaperones in the endoplasmic reticulum (ER) where it helps fold newly synthesized peptides. Because of its prominent role in protein folding, and also ER stress, we hypothesized that Grp78 might be involved in fat body cell homeostasis and vitellogenesis and a regulatory target of JH. We report here that the migratory locust Locusta migratoria possesses two Grp78 genes that are differentially regulated by JH. We found that Grp78-1 is regulated by JH through Mcm4/7-dependent DNA replication and polyploidization, whereas Grp78-2 expression is directly activated by the JH-receptor complex comprising methoprene-tolerant and Taiman proteins. Interestingly, Grp78-2 expression in the fat body is about 10-fold higher than that of Grp78-1 Knockdown of either Grp78-1 or Grp78-2 significantly reduced levels of vitellogenin (Vg) protein, accompanied by retarded maturation of oocytes. Depletion of both Grp78-1 and Grp78-2 resulted in ER stress and apoptosis in the fat body and in severely defective Vg synthesis and oocyte maturation. These results indicate a crucial role of Grp78 in JH-dependent vitellogenesis and egg production. The presence and differential regulation of two Grp78 genes in L. migratoria likely help accelerate the production of this chaperone in the fat body to facilitate folding of massively synthesized Vg and other proteins.
Collapse
Affiliation(s)
- Maowu Luo
- From the State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101.,the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dong Li
- the State Key laboratory of Cotton Biology, Institute of Plant Stress Biology, School of Life Sciences, Henan University, Kaifeng, Henan 475004, and
| | - Zhiming Wang
- From the State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101
| | - Wei Guo
- From the State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101
| | - Le Kang
- From the State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101,
| | - Shutang Zhou
- the State Key laboratory of Cotton Biology, Institute of Plant Stress Biology, School of Life Sciences, Henan University, Kaifeng, Henan 475004, and
| |
Collapse
|
302
|
Kramer CD, Genco CA. Microbiota, Immune Subversion, and Chronic Inflammation. Front Immunol 2017; 8:255. [PMID: 28348558 PMCID: PMC5346547 DOI: 10.3389/fimmu.2017.00255] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/21/2017] [Indexed: 12/12/2022] Open
Abstract
Several host-adapted pathogens and commensals have evolved mechanisms to evade the host innate immune system inducing a state of low-grade inflammation. Epidemiological studies have also documented the association of a subset of these microorganisms with chronic inflammatory disorders. In this review, we summarize recent studies demonstrating the role of the microbiota in chronic inflammatory diseases and discuss how specific microorganisms subvert or inhibit protective signaling normally induced by toll-like receptors (TLRs). We highlight our work on the oral pathogen Porphyromonas gingivalis and discuss the role of microbial modulation of lipid A structures in evasion of TLR4 signaling and resulting systemic immunopathology associated with atherosclerosis. P. gingivalis intrinsically expresses underacylated lipid A moieties and can modify the phosphorylation of lipid A, leading to altered TLR4 signaling. Using P. gingivalis mutant strains expressing distinct lipid A moieties, we demonstrated that expression of antagonist lipid A was associated with P. gingivalis-mediated systemic inflammation and immunopathology, whereas strains expressing agonist lipid A exhibited modest systemic inflammation. Likewise, mice deficient in TLR4 were more susceptible to vascular inflammation after oral infection with P. gingivalis wild-type strain compared to mice possessing functional TLR4. Collectively, our studies support a role for P. gingivalis-mediated dysregulation of innate and adaptive responses resulting in immunopathology and systemic inflammation. We propose that anti-TLR4 interventions must be designed with caution, given the balance between the protective and destructive roles of TLR signaling in response to microbiota and associated immunopathologies.
Collapse
Affiliation(s)
- Carolyn D Kramer
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine , Boston, MA , USA
| | - Caroline Attardo Genco
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine , Boston, MA , USA
| |
Collapse
|
303
|
Talarico ST, Santos FE, Brandt KG, Martinez MB, Taddei CR. Anaerobic bacteria in the intestinal microbiota of Brazilian children. Clinics (Sao Paulo) 2017; 72:154-160. [PMID: 28355361 PMCID: PMC5348578 DOI: 10.6061/clinics/2017(03)05] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 12/20/2016] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE: Changes in the neonatal gut environment allow for the colonization of the mucin layer and lumen by anaerobic bacteria. The aim of the present study was to evaluate Bifidobacterium, Lactobacillus and Lactococcus colonization through the first year of life in a group of 12 Brazilian infants and to correlate these data with the levels of Escherichia coli. The presence of anaerobic members of the adult intestinal microbiota, including Eubacterium limosum and Faecalibacterium prausnitzii, was also evaluated. METHODS: Fecal samples were collected during the first year of life, and 16S rRNA from anaerobic and facultative bacteria was detected by real-time PCR. RESULTS: Bifidobacterium was present at the highest levels at all of the studied time points, followed by E. coli and Lactobacillus. E. limosum was rarely detected, and F. prausnitzii was detected only in the samples from the latest time points. CONCLUSION: These results are consistent with reports throughout the world on the community structure of the intestinal microbiota in infants fed a milk diet. Our findings also provide evidence for the influence of the environment on intestinal colonization due to the high abundance of E. coli. The presence of important anaerobic genera was observed in Brazilian infants living at a low socioeconomic level, a result that has already been well established for infants living in developed countries.
Collapse
Affiliation(s)
- Silvia T Talarico
- Universidade de São Paulo, Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas e Toxicológicas, São Paulo/SP, Brazil
| | - Florenza E Santos
- Universidade de São Paulo, Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas e Toxicológicas, São Paulo/SP, Brazil
| | | | - Marina B Martinez
- Universidade de São Paulo, Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas e Toxicológicas, São Paulo/SP, Brazil
| | - Carla R Taddei
- Universidade de São Paulo, Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas e Toxicológicas, São Paulo/SP, Brazil
- Universidade de São Paulo, Escola de Artes, Ciências e Humanidades, São Paulo/SP, Brazil
- *Corresponding author. E-mail:
| |
Collapse
|
304
|
Mayerhofer R, Fröhlich EE, Reichmann F, Farzi A, Kogelnik N, Fröhlich E, Sattler W, Holzer P. Diverse action of lipoteichoic acid and lipopolysaccharide on neuroinflammation, blood-brain barrier disruption, and anxiety in mice. Brain Behav Immun 2017; 60:174-187. [PMID: 27751870 PMCID: PMC5419569 DOI: 10.1016/j.bbi.2016.10.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/15/2016] [Accepted: 10/13/2016] [Indexed: 12/29/2022] Open
Abstract
Microbial metabolites are known to affect immune system, brain, and behavior via activation of pattern recognition receptors such as Toll-like receptor 4 (TLR4). Unlike the effect of the TLR4 agonist lipopolysaccharide (LPS), the role of other TLR agonists in immune-brain communication is insufficiently understood. We therefore hypothesized that the TLR2 agonist lipoteichoic acid (LTA) causes immune activation in the periphery and brain, stimulates the hypothalamic-pituitary-adrenal (HPA) axis and has an adverse effect on blood-brain barrier (BBB) and emotional behavior. Since LTA preparations may be contaminated by LPS, an extract of LTA (LTAextract), purified LTA (LTApure), and pure LPS (LPSultrapure) were compared with each other in their effects on molecular and behavioral parameters 3h after intraperitoneal (i.p.) injection to male C57BL/6N mice. The LTAextract (20mg/kg) induced anxiety-related behavior in the open field test, enhanced the circulating levels of particular cytokines and the cerebral expression of cytokine mRNA, and blunted the cerebral expression of tight junction protein mRNA. A dose of LPSultrapure matching the amount of endotoxin/LPS contaminating the LTAextract reproduced several of the molecular and behavioral effects of LTAextract. LTApure (20mg/kg) increased plasma levels of tumor necrosis factor-α (TNF-α), interleukin-6 and interferon-γ, and enhanced the transcription of TNF-α, interleukin-1β and other cytokines in the amygdala and prefrontal cortex. These neuroinflammatory effects of LTApure were associated with transcriptional down-regulation of tight junction-associated proteins (claudin 5, occludin) in the brain. LTApure also enhanced circulating corticosterone, but failed to alter locomotor and anxiety-related behavior in the open field test. These data disclose that TLR2 agonism by LTA causes peripheral immune activation and initiates neuroinflammatory processes in the brain that are associated with down-regulation of BBB components and activation of the HPA axis, although emotional behavior (anxiety) is not affected. The results obtained with an LTA preparation contaminated with LPS hint at a facilitatory interaction between TLR2 and TLR4, the adverse impact of which on long-term neuroinflammation, disruption of the BBB and mental health warrants further analysis.
Collapse
Affiliation(s)
- Raphaela Mayerhofer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Esther E Fröhlich
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Nora Kogelnik
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21/III, 8010 Graz, Austria
| | - Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Stiftingtalstrasse 24/1, 8010 Graz, Austria
| | - Wolfgang Sattler
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Harrachgasse 21/III, 8010 Graz, Austria
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria; BioTechMed-Graz, Mozartgasse 12, 8010 Graz, Austria.
| |
Collapse
|
305
|
Graves CL, Li J, LaPato M, Shapiro MR, Glover SC, Wallet MA, Wallet SM. Intestinal Epithelial Cell Regulation of Adaptive Immune Dysfunction in Human Type 1 Diabetes. Front Immunol 2017; 7:679. [PMID: 28119693 PMCID: PMC5222791 DOI: 10.3389/fimmu.2016.00679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 12/21/2016] [Indexed: 01/29/2023] Open
Abstract
Environmental factors contribute to the initiation, progression, and maintenance of type 1 diabetes (T1D), although a single environmental trigger for disease has not been identified. Studies have documented the contribution of immunity within the gastrointestinal tract (GI) to the expression of autoimmunity at distal sites. Intestinal epithelial cells (IECs) regulate local and systemic immunologic homeostasis through physical and biochemical interactions with innate and adaptive immune populations. We hypothesize that a loss in the tolerance-inducing nature of the GI tract occurs within T1D and is due to altered IECs' innate immune function. As a first step in addressing this hypothesis, we contrasted the global immune microenvironment within the GI tract of individuals with T1D as well as evaluated the IEC-specific effects on adaptive immune cell phenotypes. The soluble and cellular immune microenvironment within the duodenum, the soluble mediator profile of primary IECs derived from the same duodenal tissues, and the effect of the primary IECs' soluble mediator profile on T-cell expansion and polarization were evaluated. Higher levels of IL-17C and beta-defensin 2 (BD-2) mRNA in the T1D-duodenum were observed. Higher frequencies of type 1 innate lymphoid cells (ILC1) and CD8+CXCR3+ T-cells (Tc1) were also observed in T1D-duodenal tissues, concomitant with lower frequencies of type 3 ILC (ILC3) and CD8+CCR6+ T-cells (Tc17). Higher levels of proinflammatory mediators (IL-17C and BD-2) in the absence of similar changes in mediators associated with homeostasis (interleukin 10 and thymic stromal lymphopoietin) were also observed in T1D-derived primary IEC cultures. T1D-derived IEC culture supernatants induced more robust CD8+ T-cell proliferation along with enhanced polarization of Tc1 populations, at the expense of Tc17 polarization, as well as the expansion of CXCR3+CCR6+/- Tregs, indicative of a Th1-like and less regulatory phenotype. These data demonstrate a proinflammatory microenvironment of the T1D-duodenum, whereby IECs have the potential to contribute to the expansion and polarization of innate and adaptive immune cells. Although these data do not discern whether these observations are not simply a consequence of T1D, the data indicate that the T1D-GI tract has the capacity to foster a permissive environment under which autoreactive T-cells could be expanded and polarized.
Collapse
Affiliation(s)
- Christina L. Graves
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| | - Jian Li
- Department of Gastroenterology, Hepatology, and Nutrition, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Melissa LaPato
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| | - Melanie R. Shapiro
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| | - Sarah C. Glover
- Department of Gastroenterology, Hepatology, and Nutrition, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Mark A. Wallet
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Shannon M. Wallet
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| |
Collapse
|
306
|
Jeong JH, Kim K, Lim D, Kim KH, Kim HS, Lee S, Song JH, Moon BG, Choy HE, Park SC. Microvasculature remodeling in the mouse lower gut during inflammaging. Sci Rep 2017; 7:39848. [PMID: 28045067 PMCID: PMC5206655 DOI: 10.1038/srep39848] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/28/2016] [Indexed: 12/03/2022] Open
Abstract
Inflammaging is defined as low-grade, chronic, systemic inflammation in aging, in the absence of overt infection. Age-associated deterioration of gastrointestinal function could be ascribed to the inflammaging, although evidence is yet to emerge. Here we show that microvessels in aging mouse intestine were progressively deprived of supportive structures, microvessel-associated pericytes and adherens junction protein vascular endothelial (VE)-cadherin, and became leaky. This alteration was ascribed to up-regulation of angiopoetin-2 in microvascular endothelial cells. Up-regulation of the angiopoietin-2 was by TNF-α, originated from M2-like residential CD206+ macrophages, proportion of which increases as animal ages. It was concluded that antigenic burdens encountered in intestine throughout life create the condition of chronic stage of inflammation, which accumulates M2-like macrophages expressing TNF-α. The TNF-α induces vascular leakage to facilitate recruitment of immune cells into intestine under the chronic inflammatory setting.
Collapse
Affiliation(s)
- Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Republic of Korea.,Department of Molecular Medicine(BK21plus), Chonnam National University Graduate School, Republic of Korea
| | - KwangSoo Kim
- Department of Microbiology, Chonnam National University Medical School, Republic of Korea.,Department of Molecular Medicine(BK21plus), Chonnam National University Graduate School, Republic of Korea
| | - Daejin Lim
- Department of Microbiology, Chonnam National University Medical School, Republic of Korea.,Department of Molecular Medicine(BK21plus), Chonnam National University Graduate School, Republic of Korea
| | - Kun-Hee Kim
- Department of Microbiology, Chonnam National University Medical School, Republic of Korea.,Department of Molecular Medicine(BK21plus), Chonnam National University Graduate School, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Republic of Korea
| | - Sungsu Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Joo-Hye Song
- Well Aging Research Center, SAIT. Suwon, Gyeonggido, Republic of Korea
| | - Byoung-Gon Moon
- Well Aging Research Center, SAIT. Suwon, Gyeonggido, Republic of Korea
| | - Hyon E Choy
- Department of Microbiology, Chonnam National University Medical School, Republic of Korea.,Department of Molecular Medicine(BK21plus), Chonnam National University Graduate School, Republic of Korea
| | - Sang Chul Park
- Well Aging Research Center, SAIT. Suwon, Gyeonggido, Republic of Korea.,Department of New Biology, Well Aging Research Center, DGIST, Daegu, Republic of Korea
| |
Collapse
|
307
|
Abstract
The lining of the gastrointestinal tract needs to be easily accessible to nutrients and, at the same time, defend against pathogens and chemical challenges. This lining is the largest and most vulnerable surface that faces the outside world. To manage the dual problems of effective nutrient conversion and defence, the gut lining has a sophisticated system for detection of individual chemical entities, pathogenic organisms and their products, and physico-chemical properties of its contents. Detection is through specific receptors that signal to the gut endocrine system, the nervous system, the immune system and local tissue defence systems. These effectors, in turn, modify digestive functions and contribute to tissue defence. Receptors for nutrients include taste receptors for sweet, bitter and savoury, free fatty acid receptors, peptide and phytochemical receptors, that are primarily located on enteroendocrine cells. Hormones released by enteroendocrine cells act locally, through the circulation and via the nervous system, to optimise digestion and mucosal health. Pathogen detection is both through antigen presentation to T-cells and through pattern-recognition receptors (PRRs). Activation of PRRs triggers local tissue defence, for example, by causing release of antimicrobials from Paneth cells. Toxic chemicals, including plant toxins, are sensed and then avoided, expelled or metabolised. It continues to be a major challenge to develop a comprehensive understanding of the integrated responses of the gastrointestinal tract to its luminal contents.
Collapse
|
308
|
Kotani T, Murata Y, Saito Y, Matozaki T. Future therapeutic potential of SAP-1 in inflammatory bowel diseases. Expert Rev Gastroenterol Hepatol 2016; 10:1313-1315. [PMID: 27705005 DOI: 10.1080/17474124.2016.1245144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Takenori Kotani
- a Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology , Kobe University Graduate School of Medicine , Kobe , Japan
| | - Yoji Murata
- a Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology , Kobe University Graduate School of Medicine , Kobe , Japan
| | - Yasuyuki Saito
- a Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology , Kobe University Graduate School of Medicine , Kobe , Japan
| | - Takashi Matozaki
- a Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology , Kobe University Graduate School of Medicine , Kobe , Japan
| |
Collapse
|
309
|
Hu J, Nie Y, Chen J, Zhang Y, Wang Z, Fan Q, Yan X. Gradual Changes of Gut Microbiota in Weaned Miniature Piglets. Front Microbiol 2016; 7:1727. [PMID: 27853453 PMCID: PMC5090779 DOI: 10.3389/fmicb.2016.01727] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/17/2016] [Indexed: 12/26/2022] Open
Abstract
Colonization of gut microbiota in mammals during the early life is vital to host health. The miniature piglet has recently been considered as an optimal infant model. However, less is known about the development of gut microbiota in miniature piglets. Here, this study was conducted to explore how the gut microbiota develops in weaned Congjiang miniature piglets. In contrast to the relatively stabilized gut fungal community, gut bacterial community showed a marked drop in alpha diversity, accompanied by significant alterations in taxonomic compositions. The relative abundances of 24 bacterial genera significantly declined, whereas the relative abundances of 7 bacterial genera (Fibrobacter, Collinsella, Roseburia, Prevotella, Dorea, Howardella, and Blautia) significantly increased with the age of weaned piglets. Fungal taxonomic analysis showed that the relative abundances of two genera (Kazachstania and Aureobasidium) significantly decreased, whereas the relative abundances of four genera (Aspergillus, Cladosporium, Simplicillium, and Candida) significantly increased as the piglets aged. Kazachstania telluris was the signature species predominated in gut fungal communities of weaned miniature piglets. The functional maturation of the gut bacterial community was characterized by the significantly increased digestive system, glycan biosynthesis and metabolism, and vitamin B biosynthesis as the piglets aged. These findings suggest that marked gut microbial changes in Congjiang miniature piglets may contribute to understand the potential gut microbiota development of weaned infants.
Collapse
Affiliation(s)
- Jun Hu
- Department of Animal Nutrition and Feed Science, College of Animal Sciences and Technology, Huazhong Agricultural UniversityWuhan, China; The Cooperative Innovation Center for Sustainable Pig ProductionWuhan, China
| | - Yangfan Nie
- Department of Animal Nutrition and Feed Science, College of Animal Sciences and Technology, Huazhong Agricultural UniversityWuhan, China; The Cooperative Innovation Center for Sustainable Pig ProductionWuhan, China
| | - Jianwei Chen
- Institute of Marine Omics Research, Beijing Genomics Institute-Shenzhen Shenzhen, China
| | - Yong Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education/Guizhou University Guiyang, China
| | - Zhichang Wang
- Department of Animal Nutrition and Feed Science, College of Animal Sciences and Technology, Huazhong Agricultural UniversityWuhan, China; The Cooperative Innovation Center for Sustainable Pig ProductionWuhan, China
| | - Qiwen Fan
- Department of Animal Nutrition and Feed Science, College of Animal Sciences and Technology, Huazhong Agricultural UniversityWuhan, China; The Cooperative Innovation Center for Sustainable Pig ProductionWuhan, China
| | - Xianghua Yan
- Department of Animal Nutrition and Feed Science, College of Animal Sciences and Technology, Huazhong Agricultural UniversityWuhan, China; The Cooperative Innovation Center for Sustainable Pig ProductionWuhan, China
| |
Collapse
|
310
|
Sato H, Zhang LS, Martinez K, Chang EB, Yang Q, Wang F, Howles PN, Hokari R, Miura S, Tso P. Antibiotics Suppress Activation of Intestinal Mucosal Mast Cells and Reduce Dietary Lipid Absorption in Sprague-Dawley Rats. Gastroenterology 2016; 151:923-932. [PMID: 27436071 PMCID: PMC5391873 DOI: 10.1053/j.gastro.2016.07.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/28/2016] [Accepted: 07/08/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS The gut microbiota affects intestinal permeability and mucosal mast cells (MMCs) responses. Activation of MMCs has been associated with absorption of dietary fat. We investigated whether the gut microbiota contributes to the fat-induced activation of MMCs in rats, and how antibiotics might affect this process. METHODS Adult male Sprague-Dawley rats were given streptomycin and penicillin for 4 days (n = 6-8) to reduce the abundance of their gut flora, or normal drinking water (controls, n = 6-8). They underwent lymph fistula surgery and after an overnight recovery were given an intraduodenal bolus of intralipid. We collected intestinal tissues and lymph fluid and assessed activation of MMCs, intestinal permeability, and fat transport parameters. RESULTS Compared with controls, intestinal lymph from rats given antibiotics had reduced levels of mucosal mast cell protease II (produced by MMCs) and decreased activity of diamine oxidase (produced by enterocytes) (P < .05). Rats given antibiotics had reduced intestinal permeability in response to dietary lipid compared with controls (P < .01). Unexpectedly, antibiotics also reduced lymphatic transport of triacylglycerol and phospholipid (P < .01), concomitant with decreased levels of mucosal apolipoproteins B, A-I, and A-IV (P < .01). No differences were found in intestinal motility or luminal pancreatic lipase activity between rats given antibiotics and controls. These effects were not seen with an acute dose of antibiotics or 4 weeks after the antibiotic regimen ended. CONCLUSIONS The intestinal microbiota appears to activate MMCs after the ingestion of fat in rats; this contributes to fat-induced intestinal permeability. We found that the gut microbiome promotes absorption of lipid, probably by intestinal production of apolipoproteins and secretion of chylomicrons.
Collapse
Affiliation(s)
- Hirokazu Sato
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio; Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Linda S Zhang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kristina Martinez
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois
| | - Eugene B Chang
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois
| | - Qing Yang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Fei Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Philip N Howles
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ryota Hokari
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Soichiro Miura
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
311
|
Abstract
The notion that the colon's deep crypt pockets provide a protected location that shields stem cells from potentially toxic substances is widely accepted. In this issue of Cell, Kaiko et al. reveal how a metabolite abundantly produced by the gut microbiota can inhibit stem cell proliferation but is blocked from doing so by crypt architecture.
Collapse
Affiliation(s)
- Hugo J Snippert
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands.
| |
Collapse
|
312
|
Parasar B, Chang PV. Chemical optogenetic modulation of inflammation and immunity. Chem Sci 2016; 8:1450-1453. [PMID: 28451285 PMCID: PMC5390787 DOI: 10.1039/c6sc03702j] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/19/2016] [Indexed: 01/09/2023] Open
Abstract
The immune system is an essential component of host defense against pathogens and is largely mediated by inflammatory molecules produced by immune cells, such as macrophages. These inflammatory mediators are regulated at the transcriptional level by chromatin-modifying enzymes including histone deacetylases (HDACs). Here we describe a strategy to regulate inflammation and immunity with photocontrolled HDAC inhibitors, which can be selectively delivered to target cells by UV irradiation to minimize off-target effects. We strategically photocaged the active moiety of an HDAC inhibitor and showed that mild UV irradiation leads to the selective release of the inhibitor in a spatiotemporal manner. This methodology was used to decrease the amount of pro-inflammatory mediators produced by a subpopulation of macrophages. Our approach could ultimately be used to control inflammation in vivo as a therapeutic for inflammatory diseases, while minimizing off-target effects to healthy tissues.
Collapse
Affiliation(s)
- Bibudha Parasar
- Department of Chemistry and Chemical Biology , Cornell University , Ithaca , NY 14853 , USA
| | - Pamela V Chang
- Department of Microbiology and Immunology , Cornell University , 930 Campus Road, VMC C4-185 , Ithaca , NY 14853 , USA .
| |
Collapse
|
313
|
Qin Z, Wan JJ, Sun Y, Wu T, Wang PY, Du P, Su DF, Yang Y, Liu X. Nicotine protects against DSS colitis through regulating microRNA-124 and STAT3. J Mol Med (Berl) 2016; 95:221-233. [PMID: 27709266 DOI: 10.1007/s00109-016-1473-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 07/19/2016] [Accepted: 09/08/2016] [Indexed: 01/15/2023]
Abstract
Although it is generally believed that nicotine accounts for the beneficial effect of smoking on ulcerative colitis, the underlying mechanisms remain not well understood. Our previous finding that nicotine inhibits inflammatory responses through inducing miR-124 prompted us to ask whether the miRNA is involved in the protective action of nicotine against UC. Our present study found that miR-124 expression is upregulated in colon tissues from UC patients and DSS colitis mice. Nicotine treatment further augmented miR-124 expression in lymphocytes isolated from human ulcerative colonic mucosa and ulcerative colon tissues from DSS mice, both in infiltrated lymphocytes and epithelial cells. Moreover, knockdown of miR-124 significantly diminished the beneficial effect of nicotine on murine colitis and IL-6-treated Caco-2 colon epithelial cells. Further analysis indicated that nicotine inhibited STAT3 activation in vivo and in IL-6 treated Caco-2 cells and Jurkat human T lymphocytes, in which miR-124 knockdown led to increased activation of STAT3. Blocking STAT3 activity alone is beneficial for DSS colitis and also abolished nicotine's protective effect in this model. These data indicate that nicotine exerts its protective action in UC through inducing miR-124 and inhibiting STAT3, and suggest that the miR-124/STAT3 system is a potential target for the therapeutic intervention of UC. KEY MESSAGE Nicotine upregulates miR-124 expression in ulcerative colon tissues and cells. MiR-124 is required for the protective role of nicotine in DSS colitis mice and epithelial cells. The protective effect of nicotine in murine DSS colitis depends on blocking STAT3 activation. MiR-124 mediates the inhibitory role of nicotine on STAT3/p-STAT3. Targeting miR-124 and STAT3 represents a novel approach for treating ulcerative colitis.
Collapse
Affiliation(s)
- Zhen Qin
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Jing-Jing Wan
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Yang Sun
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Tingyu Wu
- Department of Colorectal Surgery, Xinhua hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200095, People's Republic of China
| | - Peng-Yuan Wang
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Peng Du
- Department of Colorectal Surgery, Xinhua hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200095, People's Republic of China
| | - Ding-Feng Su
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, People's Republic of China.
| | - Yili Yang
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, 215123, People's Republic of China.
| | - Xia Liu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
314
|
Targeting the complex interactions between microbiota, host epithelial and immune cells in inflammatory bowel disease. Pharmacol Res 2016; 113:574-584. [PMID: 27702681 DOI: 10.1016/j.phrs.2016.09.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 09/30/2016] [Accepted: 09/30/2016] [Indexed: 12/18/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disorder that includes two distinct disease categories: ulcerative colitis and Crohn's disease. Epidemiological, genetic, and experimental studies have revealed many important aspects of IBD. Genetic susceptibility, inappropriate immune responses, environmental changes, and intestinal microbiota are all associated with the development of IBD. However, the exact mechanisms of the disease and the interactions among these pathogenic factors are largely unknown. Here we introduce recent findings from experimental colitis models that investigated the interactions between host genetic susceptibility and gut microbiota. In addition, we discuss new strategies for the treatment of IBD, focusing on the complex interactions between microbiota and host epithelial and immune cells.
Collapse
|
315
|
Jin C, Zeng Z, Fu Z, Jin Y. Oral imazalil exposure induces gut microbiota dysbiosis and colonic inflammation in mice. CHEMOSPHERE 2016; 160:349-58. [PMID: 27393971 DOI: 10.1016/j.chemosphere.2016.06.105] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 05/27/2023]
Abstract
The fungicide imazalil (IMZ) is used extensively in vegetable and fruit plantations and as a post-harvest treatment to avoid rot. Here, we revealed that ingestion of 25, 50 and 100 mg IMZ kg(-1) body weight for 28 d induced gut microbiota dysbiosis and colonic inflammation in mice. The relative abundance of Bacteroidetes, Firmicutes and Actinobacteria in the cecal contents decreased significantly after exposure to 100 mg kg(-1) IMZ for 28 d. In feces, the relative abundance in Bacteroidetes, Firmicutes and Actinobacteria decreased significantly after being exposed to 100 mg kg(-1) IMZ for 1, 14 and 7 d, respectively. High throughput sequencing of the V3-V4 region of the bacterial 16S rRNA gene revealed a significant reduction in the richness and diversity of microbiota in cecal contents and feces of IMZ-treated mice. Operational taxonomic units (OTUs) analysis identified 49.3% of OTUs changed in cecal contents, while 55.6% of OTUs changed in the feces after IMZ exposure. Overall, at the phylum level, the relative abundance of Firmicutes, Proteobacteria and Actinobacteria increased and that of Bacteroidetes decreased in IMZ-treated groups. At the genus level, the abundance of Lactobacillus and Bifidobacterium decreased while those of Deltaproteobacteria and Desulfovibrio increased in response to IMZ exposure. In addition, it was observed that IMZ exposure could induce colonic inflammation characterized by infiltration of inflammatory cells, elevated levels of lipocalin-2 (lcn-2) in the feces, and increased mRNA levels of Tnf-α, IL-1β, IL-22 and IFN-γ in the colon. Our findings strongly suggest that ingestion of IMZ has some risks to human health.
Collapse
Affiliation(s)
- Cuiyuan Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhaoyang Zeng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, 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
|
316
|
Zaiss MM, Harris NL. Interactions between the intestinal microbiome and helminth parasites. Parasite Immunol 2016; 38:5-11. [PMID: 26345715 PMCID: PMC5019230 DOI: 10.1111/pim.12274] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/01/2015] [Indexed: 12/14/2022]
Abstract
Throughout evolution, both helminths and bacteria have inhabited our intestines. As intestinal helminths and bacteria inhabit the same environmental niche, it is likely that these organisms interact with, and impact on, each other. In addition, intestinal helminths are well known to alter intestinal physiology, permeability, mucous secretion and the production of antimicrobial peptides – all of which may impact on bacterial survival and spatial organization. Yet despite rapid advances in our understanding of host–intestinal bacteria interactions, the impact of helminths on this relationship has remained largely unexplored. Moreover, although intestinal helminths are generally accepted to possess potent immuno‐modulatory activity, it is unknown whether this capacity requires interactions with intestinal bacteria. We propose that this ‘ménage à trois’ situation is likely to have exerted a strong selective pressure on the development of our metabolic and immune systems. Whilst such pressures remain in developing countries, the eradication of helminths in industrialized countries has shifted this evolutionary balance, possibly underlying the increased development of chronic inflammatory diseases. Thus, helminth–bacteria interactions may represent a key determinant of healthy homoeostasis.
Collapse
Affiliation(s)
- M M Zaiss
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - N L Harris
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
317
|
Cheng L, Jin H, Qiang Y, Wu S, Yan C, Han M, Xiao T, Yan N, An H, Zhou X, Shao Q, Xia S. High fat diet exacerbates dextran sulfate sodium induced colitis through disturbing mucosal dendritic cell homeostasis. Int Immunopharmacol 2016; 40:1-10. [PMID: 27567245 DOI: 10.1016/j.intimp.2016.08.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 02/06/2023]
Abstract
Epidemiological studies have shown that fat rich western diet contributes to the high incidence of inflammatory bowel disease (IBD). Moreover, accumulated data indicated that fat dietary factor might promote the change of the composition and metabolism in commensal flora. But, the exact mechanisms for fatty diet in gut inflammation are not well demonstrated. In this study, we found that high fat diet (HFD) promoted inflammation and exacerbated the disease severity of dextran sulfate sodium (DSS) induced colitis in mice. Compared with low fat diet (LFD)/DSS mice, shorter colon length, more epithelial loss and crypt destruction and more Gr-1+ myeloid inflammatory cells infiltration in colons were observed in HFD/DSS cohorts. Interestingly, such HFD mediated inflammation accompanied with the dys-regulation of hematopoiesis, and more hematopoiesis stem and progenitor cells were detected in colon and spleen. We further analyzed the effects of HFD and DSS treatment on mucosal DC subsets, and found that DSS treatment in LFD mice mainly dramatically increased the percentage of CD11c+CD103-CD11b+ DCs in lamina propria (LP). While, in HFD/DSS mice, HFD pre-treatment not only increased the percentage of CD11c+CD103-CD11b+ DCs, but also decreased CD11c+CD103+CD11b+ in both LP and mesenteric lymph nodes (MLN) in mice with colitis. This disequilibrium of mucosal dendritic cells in HFD/DSS mice may depend on the reduced levels of buytrate and retinoic acid. Thus, this study declared the effects of HFD on gut microenviroment, and further indicated its potential role in the development of DSS induced colitis.
Collapse
Affiliation(s)
- Lu Cheng
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Clinical Laboratory, The Seventh People's Hospital of Changzhou, Jiangsu 213011, China
| | - Huimin Jin
- Institute of Clinic Laboratory Diagnosis, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yetao Qiang
- Institute of Clinic Laboratory Diagnosis, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Shuiyun Wu
- Institute of Clinic Laboratory Diagnosis, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Cheng Yan
- Institute of Clinic Laboratory Diagnosis, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Mutian Han
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Tengfei Xiao
- Institute of Clinic Laboratory Diagnosis, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Nannan Yan
- Institute of Clinic Laboratory Diagnosis, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Huazhang An
- Institute of Cancer, Second Military Medical University, Shanghai 200433, China
| | - Xiaoming Zhou
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Qixiang Shao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Institute of Clinic Laboratory Diagnosis, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Institute of Clinic Laboratory Diagnosis, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
318
|
Bernier-Latmani J, Petrova TV. High-resolution 3D analysis of mouse small-intestinal stroma. Nat Protoc 2016; 11:1617-29. [PMID: 27560169 DOI: 10.1038/nprot.2016.092] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Here we detail a protocol for whole-mount immunostaining of mouse small-intestinal villi that can be used to generate high-resolution 3D images of all gut cell types, including blood and lymphatic vessel cells, neurons, smooth muscle cells, fibroblasts and immune cells. The procedure describes perfusion, fixation, dissection, immunostaining, mounting, clearing, confocal imaging and quantification, using intestinal vasculature as an example. As intestinal epithelial cells prevent visualization with some antibodies, we also provide an optional protocol to remove these cells before fixation. In contrast to alternative current techniques, our protocol enables the entire villus to be visualized with increased spatial resolution of cell location, morphology and cell-cell interactions, thus allowing for easy quantification of phenotypes. The technique, which takes 7 d from mouse dissection to microscopic examination, will be useful for researchers who are interested in most aspects of intestinal biology, including mucosal immunology, infection, nutrition, cancer biology and intestinal microbiota.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences. Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
319
|
Malnutrition Is Associated with Protection from Rotavirus Diarrhea: Evidence from a Longitudinal Birth Cohort Study in Bangladesh. J Clin Microbiol 2016; 54:2568-74. [PMID: 27510830 PMCID: PMC5035411 DOI: 10.1128/jcm.00916-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/02/2016] [Indexed: 01/05/2023] Open
Abstract
Rotavirus is a leading cause of dehydrating diarrhea and death among infants and children globally, particularly in communities of the developing world. While numerous studies have described the complex relationships among infectious diarrhea, growth faltering, and poverty, the impact of nutritional status on susceptibility to rotavirus diarrhea is not well understood. In a longitudinal study conducted over the first 3 years of life among 626 slum-dwelling infants enrolled at birth in Dhaka, Bangladesh, we observed that common measures of healthy growth and development were positively associated with a risk of symptomatic rotavirus infection. This finding runs counter to the idea that improving childhood nutrition will implicitly decrease the incidence of symptomatic infection by enteric pathogens. As childhood nutrition improves worldwide, rotavirus infection may remain a public health challenge, making universal vaccination of even greater importance.
Collapse
|
320
|
Outer Membrane Vesicles from the Probiotic Escherichia coli Nissle 1917 and the Commensal ECOR12 Enter Intestinal Epithelial Cells via Clathrin-Dependent Endocytosis and Elicit Differential Effects on DNA Damage. PLoS One 2016; 11:e0160374. [PMID: 27487076 PMCID: PMC4972321 DOI: 10.1371/journal.pone.0160374] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/18/2016] [Indexed: 01/26/2023] Open
Abstract
Interactions between intestinal microbiota and the human host are complex. The gut mucosal surface is covered by a mucin layer that prevents bacteria from accessing the epithelial cells. Thus, the crosstalk between microbiota and the host mainly rely on secreted factors that can go through the mucus layer and reach the epithelium. In this context, vesicles released by commensal strains are seen as key players in signaling processes in the intestinal mucosa. Studies with Gram-negative pathogens showed that outer membrane vesicles (OMVs) are internalized into the host cell by endocytosis, but the entry mechanism for microbiota-derived vesicles is unknown. Escherichia coli strains are found as part of normal human gut microbiota. In this work, we elucidate the pathway that mediate internalization of OMVs from the probiotic E.coli Nissle 1917 (EcN) and the commensal ECOR12 strains in several human intestinal epithelial cell lines. Time course measurement of fluorescence and microscopy analysis performed with rhodamine B-R18-labeled OMVs in the presence of endocytosis inhibitors showed that OMVs from these strains enter epithelial cells via clathrin-mediated endocytosis. Vesicles use the same endocytosis pathway in polarized epithelial monolayers. Internalized OMVs are sorted to lysosomal compartments as shown by their colocalization with clathrin and specific markers of endosomes and lysosomes. OMVs from both strains did not affect cell viability, but reduce proliferation of HT-29 cells. Labeling of 8-oxo-dG adducts in DNA revealed that neither OMVs from EcN nor from ECOR12 promoted oxidative DNA damage. In contrast, flow cytometry analysis of phosphorylated γH2AX evidenced that OMVs from the probiotic EcN significantly produced more double strand breaks in DNA than ECOR12 OMVs. The EcN genotoxic effects have been attributed to the synthesis of colibactin. However, it is not known how colibactin is exported and delivered into host cells. Whether colibactin is secreted via OMVs is an open question that needs further study.
Collapse
|
321
|
Tao S, Luo Y, Bin He, Liu J, Qian X, Ni Y, Zhao R. Paraoxonase 2 modulates a proapoptotic function in LS174T cells in response to quorum sensing molecule N-(3-oxododecanoyl)-L-homoserine lactone. Sci Rep 2016; 6:28778. [PMID: 27364593 PMCID: PMC4929476 DOI: 10.1038/srep28778] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/08/2016] [Indexed: 12/14/2022] Open
Abstract
A mucus layer coats the gastrointestinal tract and serves as the first line of intestinal defense against infection. N-acyl-homoserine lactone (AHL) quorum-sensing molecules produced by gram-negative bacteria in the gut can influence the homeostasis of intestinal epithelium. In this study, we investigated the effects of two representative long- and short-chain AHLs, N-3-(oxododecanoyl)-homoserine lactone (C12-HSL) and N-butyryl homoserine lactone (C4-HSL), on cell viability and mucus secretion in LS174T cells. C12-HSL but not C4-HSL significantly decreased cell viability by inducing mitochondrial dysfunction and activating cell apoptosis which led to a decrease in mucin expression. Pretreatment with lipid raft disruptor (Methyl-β-cyclodextrin, MβCD) and oxidative stress inhibitor (N-acetyl-L-cysteine, NAC) slightly rescued the viability of cells damaged by C12-HSL exposure, while the paraoxonase 2 (PON2) inhibitor (Triazolo[4,3-a]quinolone, TQ416) significantly affected recovering cells viability and mucin secretion. When LS174T cells were treated with C12-HSL and TQ416 simultaneously, TQ416 showed the maximal positive effect on cells viability. However, if cells were first treated with C12-HSL for 40 mins, and then TQ46 was added, the TQ416 had no effect on cell viability. These results suggest that the C12-HSL-acid process acts at an early step to activate apoptosis as part of C12-HSL’s effect on intestinal mucus barrier function.
Collapse
Affiliation(s)
- Shiyu Tao
- Key Laboratory of Animal Physiology &Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yanwen Luo
- Key Laboratory of Animal Physiology &Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Bin He
- Key Laboratory of Animal Physiology &Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jie Liu
- Key Laboratory of Animal Physiology &Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Xi Qian
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT 05452, USA
| | - Yingdong Ni
- Key Laboratory of Animal Physiology &Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology &Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| |
Collapse
|
322
|
Fábrega MJ, Aguilera L, Giménez R, Varela E, Alexandra Cañas M, Antolín M, Badía J, Baldomà L. Activation of Immune and Defense Responses in the Intestinal Mucosa by Outer Membrane Vesicles of Commensal and Probiotic Escherichia coli Strains. Front Microbiol 2016; 7:705. [PMID: 27242727 PMCID: PMC4863414 DOI: 10.3389/fmicb.2016.00705] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/28/2016] [Indexed: 12/23/2022] Open
Abstract
The influence of microbiota in human health is well-known. Imbalances in microbiome structure have been linked to several diseases. Modulation of microbiota composition through probiotic therapy is an attempt to harness the beneficial effects of commensal microbiota. Although, there is wide knowledge of the responses induced by gut microbiota, the microbial factors that mediate these effects are not well-known. Gram-negative bacteria release outer membrane vesicles (OMVs) as a secretion mechanism of microbial factors, which have an important role in intercellular communication. Here, we investigated whether OMVs from the probiotic Escherichia coli strain Nissle 1917 (EcN) or the commensal E. coli strain ECOR12 trigger immune responses in various cellular models: (i) peripheral blood mononuclear cells (PBMCs) as a model of intestinal barrier disruption, (ii) apical stimulation of Caco-2/PMBCs co-culture as a model of intact intestinal mucosa, and (iii) colonic mucosa explants as an ex vivo model. Stimulations with bacterial lysates were also performed. Whereas, both OMVs and lysates activated expression and secretion of several cytokines and chemokines in PBMCs, only OMVs induced basolateral secretion and mRNA upregulation of these mediators in the co-culture model. We provide evidence that OMVs are internalized in polarized Caco-2 cells. The activated epithelial cells elicit a response in the underlying immunocompetent cells. The OMVs effects were corroborated in the ex vivo model. This experimental study shows that OMVs are an effective strategy used by beneficial gut bacteria to communicate with and modulate host responses, activating signaling events through the intestinal epithelial barrier.
Collapse
Affiliation(s)
- María José Fábrega
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona Barcelona, Spain
| | - Laura Aguilera
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona Barcelona, Spain
| | - Rosa Giménez
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona Barcelona, Spain
| | - Encarna Varela
- Department of Gastroenterology, Digestive System Research Unit, Institut de Recerca Vall d'Hebron, CIBER EHD, Instituto de Salud Carlos III, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona Barcelona, Spain
| | - María Alexandra Cañas
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona Barcelona, Spain
| | - María Antolín
- Department of Gastroenterology, Digestive System Research Unit, Institut de Recerca Vall d'Hebron, CIBER EHD, Instituto de Salud Carlos III, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona Barcelona, Spain
| | - Josefa Badía
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona Barcelona, Spain
| | - Laura Baldomà
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona Barcelona, Spain
| |
Collapse
|
323
|
Zong C, Nie X, Zhang D, Ji Q, Qin Y, Wang L, Jiang D, Gong C, Liu Y, Zhou G. Up regulation of glyoxylate reductase/hydroxypyruvate reductase (GRHPR) is associated with intestinal epithelial cells apoptosis in TNBS-induced experimental colitis. Pathol Res Pract 2016; 212:365-71. [DOI: 10.1016/j.prp.2015.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 08/17/2015] [Accepted: 09/21/2015] [Indexed: 12/19/2022]
|
324
|
Genser L, Poitou C, Brot-Laroche É, Rousset M, Vaillant JC, Clément K, Thenet S, Leturque A. [Alteration of intestinal permeability: the missing link between gut microbiota modifications and inflammation in obesity?]. Med Sci (Paris) 2016; 32:461-469. [PMID: 27225918 DOI: 10.1051/medsci/20163205012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2023] Open
Abstract
The increasing incidence of obesity and associated metabolic complications is a worldwide public health issue. The role of the gut in the pathophysiology of obesity, with an important part for microbiota, is becoming obvious. In rodent models of diet-induced obesity, the modifications of gut microbiota are associated with an alteration of the intestinal permeability increasing the passage of food or bacterial antigens, which contribute to low-grade inflammation and insulin resistance. In human obesity, intestinal permeability modification, and its role in the crosstalk between gut microbiota changes and inflammation at systemic and tissular levels, are still poorly documented. Hence, further characterization of the triggering mechanisms of such inflammatory responses in obese subjects could enable the development of personalized intervention strategies that will help to reduce the risk of obesity-associated diseases.
Collapse
Affiliation(s)
- Laurent Genser
- Institut de cardiométabolisme et nutrition, ICAN, hôpital Pitié-Salpêtrière, F-75013, Paris, France - Sorbonne universités, université Paris 06, UMR-S 1166, Nutriomics Team, F-75013 Paris, France - Inserm, UMRS 1166, Nutriomics, F-75013 5Paris, France
| | - Christine Poitou
- Institut de cardiométabolisme et nutrition, ICAN, hôpital Pitié-Salpêtrière, F-75013, Paris, France - Sorbonne universités, université Paris 06, UMR-S 1166, Nutriomics Team, F-75013 Paris, France - Inserm, UMRS 1166, Nutriomics, F-75013 5Paris, France - Assistance-Publique Hôpitaux de Paris, groupe hospitalier Pitié-Salpêtrière, service de nutrition, 47-83, boulevard de l'Hôpital, F-75013 Paris, France
| | - Édith Brot-Laroche
- Institut de cardiométabolisme et nutrition, ICAN, hôpital Pitié-Salpêtrière, F-75013, Paris, France - Centre de recherche des Cordeliers, Inserm, UMPC université Paris 6, université Paris Descartes Paris 5, CNRS, EPHE laboratoire de pharmacologie cellulaire et moléculaire, 75006 Paris, France
| | - Monique Rousset
- Institut de cardiométabolisme et nutrition, ICAN, hôpital Pitié-Salpêtrière, F-75013, Paris, France - Centre de recherche des Cordeliers, Inserm, UMPC université Paris 6, université Paris Descartes Paris 5, CNRS, EPHE laboratoire de pharmacologie cellulaire et moléculaire, 75006 Paris, France
| | - Jean-Christophe Vaillant
- Assistance-Publique Hôpitaux de Paris, groupe hospitalier Pitié-Salpêtrière, service de chirurgie digestive, hépato-bilio-pancréatique, transplantation hépatique, F-75013 Paris, France
| | - Karine Clément
- Institut de cardiométabolisme et nutrition, ICAN, hôpital Pitié-Salpêtrière, F-75013, Paris, France - Sorbonne universités, université Paris 06, UMR-S 1166, Nutriomics Team, F-75013 Paris, France - Inserm, UMRS 1166, Nutriomics, F-75013 5Paris, France - Assistance-Publique Hôpitaux de Paris, groupe hospitalier Pitié-Salpêtrière, service de nutrition, 47-83, boulevard de l'Hôpital, F-75013 Paris, France
| | - Sophie Thenet
- Institut de cardiométabolisme et nutrition, ICAN, hôpital Pitié-Salpêtrière, F-75013, Paris, France - Centre de recherche des Cordeliers, Inserm, UMPC université Paris 6, université Paris Descartes Paris 5, CNRS, EPHE laboratoire de pharmacologie cellulaire et moléculaire, 75006 Paris, France
| | - Armelle Leturque
- Institut de cardiométabolisme et nutrition, ICAN, hôpital Pitié-Salpêtrière, F-75013, Paris, France - Centre de recherche des Cordeliers, Inserm, UMPC université Paris 6, université Paris Descartes Paris 5, CNRS, EPHE laboratoire de pharmacologie cellulaire et moléculaire, 75006 Paris, France
| |
Collapse
|
325
|
Fawley J, Gourlay DM. Intestinal alkaline phosphatase: a summary of its role in clinical disease. J Surg Res 2016; 202:225-34. [PMID: 27083970 PMCID: PMC4834149 DOI: 10.1016/j.jss.2015.12.008] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 11/07/2015] [Accepted: 12/08/2015] [Indexed: 12/19/2022]
Abstract
Over the past few years, there is increasing evidence implicating a novel role for Intestinal Alkaline Phosphatase (IAP) in mitigating inflammatory mediated disorders. IAP is an endogenous protein expressed by the intestinal epithelium that is believed to play a vital role in maintaining gut homeostasis. Loss of IAP expression or function is associated with increased intestinal inflammation, dysbiosis, bacterial translocation and subsequently systemic inflammation. As these events are a cornerstone of the pathophysiology of many diseases relevant to surgeons, we sought to review recent research in both animal and humans on IAP's physiologic function, mechanisms of action and current research in specific surgical diseases.
Collapse
Affiliation(s)
- Jason Fawley
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee; Department of Surgery, Division of Pediatric Surgery, Children's Hospital of Wisconsin, Milwaukee
| | - David M Gourlay
- Department of Surgery, Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee; Department of Surgery, Division of Pediatric Surgery, Children's Hospital of Wisconsin, Milwaukee.
| |
Collapse
|
326
|
Jovel J, Patterson J, Wang W, Hotte N, O'Keefe S, Mitchel T, Perry T, Kao D, Mason AL, Madsen KL, Wong GKS. Characterization of the Gut Microbiome Using 16S or Shotgun Metagenomics. Front Microbiol 2016; 7:459. [PMID: 27148170 PMCID: PMC4837688 DOI: 10.3389/fmicb.2016.00459] [Citation(s) in RCA: 540] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/21/2016] [Indexed: 02/06/2023] Open
Abstract
The advent of next generation sequencing (NGS) has enabled investigations of the gut microbiome with unprecedented resolution and throughput. This has stimulated the development of sophisticated bioinformatics tools to analyze the massive amounts of data generated. Researchers therefore need a clear understanding of the key concepts required for the design, execution and interpretation of NGS experiments on microbiomes. We conducted a literature review and used our own data to determine which approaches work best. The two main approaches for analyzing the microbiome, 16S ribosomal RNA (rRNA) gene amplicons and shotgun metagenomics, are illustrated with analyses of libraries designed to highlight their strengths and weaknesses. Several methods for taxonomic classification of bacterial sequences are discussed. We present simulations to assess the number of sequences that are required to perform reliable appraisals of bacterial community structure. To the extent that fluctuations in the diversity of gut bacterial populations correlate with health and disease, we emphasize various techniques for the analysis of bacterial communities within samples (α-diversity) and between samples (β-diversity). Finally, we demonstrate techniques to infer the metabolic capabilities of a bacteria community from these 16S and shotgun data.
Collapse
Affiliation(s)
- Juan Jovel
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Jordan Patterson
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Weiwei Wang
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Naomi Hotte
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Sandra O'Keefe
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Troy Mitchel
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Troy Perry
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Dina Kao
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Andrew L. Mason
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Karen L. Madsen
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
| | - Gane K.-S. Wong
- Department of Medicine, University of AlbertaEdmonton, AB, Canada
- Department of Biological Sciences, University of AlbertaEdmonton, AB, Canada
- BGI-ShenzhenShenzhen, China
| |
Collapse
|
327
|
Wen HZ, Hao WW, Liu YT, Shao LJ. Fecal microbiota transplantation for inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2016; 24:1545-1551. [DOI: 10.11569/wcjd.v24.i10.1545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fecal microbiota transplantation (FMT) for treatment of inflammatory bowel disease (IBD), a common digestive disease, has attracted great interest in recent years, mainly because of the high prevalence and recurrence of IBD. IBD is difficult to treat, and standard therapy does not always induce remission. FMT is an alternative approach that can induce remission in some patients with active IBD. However, many unanswered questions on the clinical application of FMT in IBD remain. This review discusses the efficacy, adverse events, and optional route of administration of FMT in IBD.
Collapse
|
328
|
Abstract
Research in the past decade has greatly expanded our understanding of the pathogenesis of inflammatory bowel disease, which includes Crohn's disease and ulcerative colitis. In addition to the sophisticated network of immune response, the epithelial layer lining the mucosa has emerged as an essential player in the development and persistence of intestinal inflammation. As the frontline of numerous environmental insults in the gut, the intestinal epithelial cells are subject to various cellular stresses. In eukaryotic cells, disturbance of endoplasmic reticulum homeostasis may lead to the accumulation of unfolded and misfolded proteins in the ER lumen, a condition called ER stress. This cellular process activates the unfolded protein response, which functions to enhance the ER protein folding capacity, alleviates the burden of protein synthesis and maturation, and activates ER-associated protein degradation. Paneth and goblet cells, 2 secretory epithelial populations in the gut, are particularly sensitive to ER stress on environmental or genetic disturbances. Recent studies suggested that epithelial ER stress may contribute to the pathogenesis of Crohn's disease and ulcerative colitis by compromising protein secretion, inducing epithelial cell apoptosis and activating proinflammatory response in the gut. Our knowledge of ER stress in intestinal epithelial function may open avenue to new inflammatory bowel disease therapies by targeting the ER protein folding homeostasis in the cells lining the intestinal mucosa.
Collapse
|
329
|
Zhang W, Jin LH. Asparagus cochinchinensis Extract Alleviates Metal Ion-Induced Gut Injury in Drosophila: An In Silico Analysis of Potential Active Constituents. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:7603746. [PMID: 27123034 PMCID: PMC4830720 DOI: 10.1155/2016/7603746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/24/2016] [Accepted: 03/13/2016] [Indexed: 12/25/2022]
Abstract
Metal ions and sulfate are components of atmospheric pollutants that have diverse ways of entering the human body. We used Drosophila as a model to investigate the effect of Asparagus cochinchinensis (A. cochinchinensis) extracts on the gut and characterized gut homeostasis following the ingestion of metal ions (copper, zinc, and aluminum). In this study, we found that the aqueous A. cochinchinensis extract increased the survival rate, decreased epithelial cell death, and attenuated metal ion-induced gut morphological changes in flies following chronic exposure to metal ions. In addition, we screened out, by network pharmacology, six natural products (NPs) that could serve as putative active components of A. cochinchinensis that prevented gut injury. Altogether, the results of our study provide evidence that A. cochinchinensis might be an effective phytomedicine for the treatment of metal ion-induced gut injury.
Collapse
Affiliation(s)
- Weiyu Zhang
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Li Hua Jin
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| |
Collapse
|
330
|
Abrupt suspension of probiotics administration may increase host pathogen susceptibility by inducing gut dysbiosis. Sci Rep 2016; 6:23214. [PMID: 26983596 PMCID: PMC4794715 DOI: 10.1038/srep23214] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/29/2016] [Indexed: 01/14/2023] Open
Abstract
In this study, we investigated the risk associated with suspension of probiotics administration in tilapia, an animal model that may mimic immune-compromised conditions in humans. Tilapias were fed for 14 days using a probiotics-supplemented diet, followed by a three-day suspension of probiotics treatment and a subsequent challenge by Aeromonas hydrophila. Unexpectedly, the suspension of a probiotic strain Lactobacillus plantarum JCM1149 significantly triggered susceptibility of the host to A. hydrophila. We further observed that suspension of JCM1149 resulted in host gut microbiota dysbiosis and the subsequent disorder in the intestinal metabolites (bile acids, amino acids, and glucose) and damage in the intestinal epithelium, giving rise to a condition similar to antibiotics-induced gut dysbiosis, which collectively impaired tilapia’s gut health and resistance to pathogenic challenges. Additionally, we determined that JCM1149 adhered relatively poorly to tilapia intestinal mucosa and was rapidly released from the gastrointestinal tract (GIT) after suspension, with the rapid loss of probiotic strain probably being the direct cause of gut dysbiosis. Finally, three other probiotic Lactobacillus strains with low intestinal mucosa binding activity showed similar rapid loss phenotype following administration suspension, and induced higher host susceptibility to infection, indicating that the risk is a generic phenomenon in Lactobacillus.
Collapse
|
331
|
Durchschein F, Petritsch W, Hammer HF. Diet therapy for inflammatory bowel diseases: The established and the new. World J Gastroenterol 2016; 22:2179-2194. [PMID: 26900283 PMCID: PMC4734995 DOI: 10.3748/wjg.v22.i7.2179] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 11/10/2015] [Accepted: 12/30/2015] [Indexed: 02/06/2023] Open
Abstract
Although patients with inflammatory bowel diseases (IBD) have a strong interest in dietary modifications as part of their therapeutic management, dietary advice plays only a minor part in published guidelines. The scientific literature shows that dietary factors might influence the risk of developing IBD, that dysbiosis induced by nutrition contributes to the pathogenesis of IBD, and that diet may serve as a symptomatic treatment for irritable bowel syndrome-like symptoms in IBD. The role of nutrition in IBD is underscored by the effect of various dietary therapies. In paediatric patients with Crohn’s disease (CD) enteral nutrition (EN) reaches remission rates similar to steroids. In adult patients, however, EN is inferior to corticosteroids. EN is not effective in ulcerative colitis (UC). Total parenteral nutrition in IBD is not superior to steroids or EN. The use of specific probiotics in patients with IBD can be recommended only in special clinical situations. There is no evidence for efficacy of probiotics in CD. By contrast, studies in UC have shown a beneficial effect in selected patients. For patients with pouchitis, antibiotic treatment followed by probiotics, like VSL#3 or Lactobacillus GG, is effective. When probiotics are used, the risk of bacterial translocation and subsequent bacteremia has to be considered. More understanding of the normal intestinal microflora, and better characterization of probiotic strains at the phenotypic and genomic levels is needed as well as clarification of the mechanisms of action in different clinical settings. A FODMAP reduced diet may improve symptoms in IBD.
Collapse
|
332
|
Vaikunthanathan T, Safinia N, Lombardi G, Lechler RI. Microbiota, immunity and the liver. Immunol Lett 2016; 171:36-49. [PMID: 26835593 DOI: 10.1016/j.imlet.2016.01.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/24/2016] [Accepted: 01/27/2016] [Indexed: 12/18/2022]
Abstract
The gut harbors a complex community of over 100 trillion microbial cells known to exist in symbiotic harmony with the host influencing human physiology, metabolism, nutrition and immune function. It is now widely accepted that perturbations of this close partnership results in the pathogenesis of several major diseases with increasing evidence highlighting their role outside of the intestinal tract. The intimate proximity and circulatory loop of the liver and the gut has attracted significant attention regarding the role of the microbiota in the development and progression of liver disease. Here we give an overview of the interaction between the microbiota and the immune system and focus on their convincing role in both the propagation and treatment of liver disease.
Collapse
Affiliation(s)
- T Vaikunthanathan
- MRC Centre for Transplantation, Division of Transplantation Immunology & Mucosal Biology, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.
| | - N Safinia
- MRC Centre for Transplantation, Division of Transplantation Immunology & Mucosal Biology, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.
| | - G Lombardi
- MRC Centre for Transplantation, Division of Transplantation Immunology & Mucosal Biology, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.
| | - R I Lechler
- MRC Centre for Transplantation, Division of Transplantation Immunology & Mucosal Biology, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.
| |
Collapse
|
333
|
Oruc Z, Oblet C, Boumediene A, Druilhe A, Pascal V, Le Rumeur E, Cuvillier A, El Hamel C, Lecardeur S, Leanderson T, Morelle W, Demengeot J, Aldigier JC, Cogné M. IgA Structure Variations Associate with Immune Stimulations and IgA Mesangial Deposition. J Am Soc Nephrol 2016; 27:2748-61. [PMID: 26825533 DOI: 10.1681/asn.2015080911] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/17/2015] [Indexed: 11/03/2022] Open
Abstract
IgA1 mesangial deposition is the hallmark of IgA nephropathy and Henoch-Schönlein purpura, the onset of which often follows infections. Deposited IgA has been reported as polymeric, J chain associated, and often, hypogalactosylated but with no information concerning the influence of the IgA repertoire or the link between immune stimuli and IgA structure. We explored these issues in the α1KI mouse model, which produces polyclonal human IgA1 prone to mesangial deposition. Compared with mice challenged by a conventional environment, mice in a specific pathogen-free environment had less IgA deposition. However, serum IgA of specific pathogen-free mice showed more galactosylation and much lower polymerization. Notably, wild-type, α1KI, and even J chain-deficient mice showed increased polymeric serum IgA on exposure to pathogens. Strict germfree conditions delayed but did not completely prevent deposition; mice housed in these conditions had very low serum IgA levels and produced essentially monomeric IgA. Finally, comparing monoclonal IgA1 that had different variable regions and mesangial deposition patterns indicated that, independently of glycosylation and polymerization, deposition might also depend on IgA carrying specific variable domains. Together with IgA quantities and constant region post-translational modifications, repertoire changes during immune responses might, thus, modulate IgA propensity to deposition. These IgA features are not associated with circulating immune complexes and C3 deposition and are more pertinent to an initial IgA deposition step preceding overt clinical symptoms in patients.
Collapse
Affiliation(s)
- Zeliha Oruc
- Limoges University Hospital Dupuytren, Centre National de la Recherche Scientifique, Limoges University, Limoges, France
| | - Christelle Oblet
- Limoges University Hospital Dupuytren, Centre National de la Recherche Scientifique, Limoges University, Limoges, France
| | - Ahmed Boumediene
- Limoges University Hospital Dupuytren, Centre National de la Recherche Scientifique, Limoges University, Limoges, France
| | - Anne Druilhe
- Limoges University Hospital Dupuytren, Centre National de la Recherche Scientifique, Limoges University, Limoges, France
| | - Virginie Pascal
- Limoges University Hospital Dupuytren, Centre National de la Recherche Scientifique, Limoges University, Limoges, France
| | - Elisabeth Le Rumeur
- Genetics and Development Instittute, Rennes University, Centre National de la Recherche Scientifique, Rennes, France
| | | | - Chahrazed El Hamel
- Limoges University Hospital Dupuytren, Centre National de la Recherche Scientifique, Limoges University, Limoges, France
| | - Sandrine Lecardeur
- Limoges University Hospital Dupuytren, Centre National de la Recherche Scientifique, Limoges University, Limoges, France
| | | | - Willy Morelle
- Centre National de la Recherche Scientifique, Laboratory of Structural and Functional Glycobiology, University of Lille 1, France; and
| | | | - Jean-Claude Aldigier
- Limoges University Hospital Dupuytren, Centre National de la Recherche Scientifique, Limoges University, Limoges, France
| | - Michel Cogné
- Limoges University Hospital Dupuytren, Centre National de la Recherche Scientifique, Limoges University, Limoges, France;
| |
Collapse
|
334
|
Kim SH, Lee KA, Park DY, Jang IH, Lee WJ. Drosophila–Acetobacter as a Model System for Understanding Animal–Microbiota Interactions. ACETIC ACID BACTERIA 2016:143-158. [DOI: 10.1007/978-4-431-55933-7_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
335
|
Kayama H, Takeda K. Functions of innate immune cells and commensal bacteria in gut homeostasis. J Biochem 2015; 159:141-9. [PMID: 26615026 DOI: 10.1093/jb/mvv119] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/11/2015] [Indexed: 02/06/2023] Open
Abstract
The intestinal immune system remains unresponsive to beneficial microbes and dietary antigens while activating pro-inflammatory responses against pathogens for host defence. In intestinal mucosa, abnormal activation of innate immunity, which directs adaptive immune responses, causes the onset and/or progression of inflammatory bowel diseases. Thus, innate immunity is finely regulated in the gut. Multiple innate immune cell subsets have been identified in both murine and human intestinal lamina propria. Some innate immune cells play a key role in the maintenance of gut homeostasis by preventing inappropriate adaptive immune responses while others are associated with the pathogenesis of intestinal inflammation through development of Th1 and Th17 cells. In addition, intestinal microbiota and their metabolites contribute to the regulation of innate/adaptive immune responses. Accordingly, perturbation of microbiota composition can trigger intestinal inflammation by driving inappropriate immune responses.
Collapse
Affiliation(s)
- Hisako Kayama
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine and Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine and Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
336
|
Loke P, Lim YAL. Helminths and the microbiota: parts of the hygiene hypothesis. Parasite Immunol 2015; 37:314-23. [PMID: 25869420 DOI: 10.1111/pim.12193] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/18/2015] [Indexed: 12/12/2022]
Abstract
In modern societies, diseases that are driven by dysregulated immune responses are increasing at an alarming pace, such as inflammatory bowel diseases and diabetes. There is an urgent need to understand these epidemiological trends, which are likely to be driven by the changing environment of the last few decades. There are complex interactions between human genetic factors and this changing environment that is leading to the increasing prevalence of metabolic and inflammatory diseases. Alterations to human gut bacterial communities (the microbiota) and lowered prevalence of helminth infections are potential environmental factors contributing to immune dysregulation. Helminths have co-evolved with the gut microbiota and their mammalian hosts. This three-way interaction is beginning to be characterized, and the knowledge gained may enable the design of new therapeutic strategies to treat metabolic and inflammatory diseases. However, these complex interactions need to be carefully investigated in the context of host genetic backgrounds to identify optimal treatment strategies. The complex nature of these interactions raises the possibility that only with highly personalized treatment, with knowledge of individual genetic and microbiota communities, will therapeutic interventions be successful for a majority of the individuals suffering from these complex diseases of immune dysregulation.
Collapse
Affiliation(s)
- P Loke
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | | |
Collapse
|
337
|
Bernier-Latmani J, Cisarovsky C, Demir CS, Bruand M, Jaquet M, Davanture S, Ragusa S, Siegert S, Dormond O, Benedito R, Radtke F, Luther SA, Petrova TV. DLL4 promotes continuous adult intestinal lacteal regeneration and dietary fat transport. J Clin Invest 2015; 125:4572-86. [PMID: 26529256 DOI: 10.1172/jci82045] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
The small intestine is a dynamic and complex organ that is characterized by constant epithelium turnover and crosstalk among various cell types and the microbiota. Lymphatic capillaries of the small intestine, called lacteals, play key roles in dietary fat absorption and the gut immune response; however, little is known about the molecular regulation of lacteal function. Here, we performed a high-resolution analysis of the small intestinal stroma and determined that lacteals reside in a permanent regenerative, proliferative state that is distinct from embryonic lymphangiogenesis or quiescent lymphatic vessels observed in other tissues. We further demonstrated that this continuous regeneration process is mediated by Notch signaling and that the expression of the Notch ligand delta-like 4 (DLL4) in lacteals requires activation of VEGFR3 and VEGFR2. Moreover, genetic inactivation of Dll4 in lymphatic endothelial cells led to lacteal regression and impaired dietary fat uptake. We propose that such a slow lymphatic regeneration mode is necessary to match a unique need of intestinal lymphatic vessels for both continuous maintenance, due to the constant exposure to dietary fat and mechanical strain, and efficient uptake of fat and immune cells. Our work reveals how lymphatic vessel responses are shaped by tissue specialization and uncover a role for continuous DLL4 signaling in the function of adult lymphatic vasculature.
Collapse
|
338
|
Reply to Keelan and Payne: Microbiota-related pathways for preterm birth. Proc Natl Acad Sci U S A 2015; 112:E6415. [PMID: 26515091 DOI: 10.1073/pnas.1517939112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
339
|
Tanaka K, Fujiya M, Konishi H, Ueno N, Kashima S, Sasajima J, Moriichi K, Ikuta K, Tanabe H, Kohgo Y. Probiotic-derived polyphosphate improves the intestinal barrier function through the caveolin-dependent endocytic pathway. Biochem Biophys Res Commun 2015; 467:541-8. [PMID: 26459590 DOI: 10.1016/j.bbrc.2015.09.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 12/15/2022]
Abstract
Probiotics exhibit beneficial functions for host homeostasis maintenance. We herein investigated the mechanism by which Lactobacillus brevis-derived poly P exhibited a beneficial function. Immunostaining indicated that poly P was captured in the plasma membrane via integrin β1 in Caco2/bbe cells. The uptake of poly P was reduced by the inhibition of integrin β1 as well as caveolin-1, a major component of lipid rafts. The function of poly P, including the induction of HSP27 and enhancement of the intestinal barrier function, was suppressed by the inhibition of caveolin-1, illustrating that the function of poly P was mediated by the endocytic pathway. High-throughput sequencing revealed that poly P induced tumor necrosis factor alpha-induced protein 3, which contributes to cytoprotection, including upregulation of the intestinal barrier function. The present study demonstrates a novel host-probiotic interaction through the uptake of bacterial substance into host cells, which is distinct from pattern recognition receptor pathways.
Collapse
Affiliation(s)
- Kazuyuki Tanaka
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Mikihiro Fujiya
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan.
| | - Hiroaki Konishi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Nobuhiro Ueno
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Shin Kashima
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Junpei Sasajima
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Kentaro Moriichi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Katsuya Ikuta
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Hiroki Tanabe
- Department of Gastroenterology, International Health and Science University Hospital, Japan
| | - Yutaka Kohgo
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan; Department of Gastroenterology, International Health and Science University Hospital, Japan
| |
Collapse
|
340
|
Apoptosis, Necrosis, and Necroptosis in the Gut and Intestinal Homeostasis. Mediators Inflamm 2015; 2015:250762. [PMID: 26483605 PMCID: PMC4592906 DOI: 10.1155/2015/250762] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 09/08/2015] [Indexed: 12/13/2022] Open
Abstract
Intestinal epithelial cells (IECs) form a physiochemical barrier that separates the intestinal lumen from the host's internal milieu and is critical for electrolyte passage, nutrient absorption, and interaction with commensal microbiota. Moreover, IECs are strongly involved in the intestinal mucosal inflammatory response as well as in mucosal innate and adaptive immune responses. Cell death in the intestinal barrier is finely controlled, since alterations may lead to severe disorders, including inflammatory diseases. The emerging picture indicates that intestinal epithelial cell death is strictly related to the maintenance of tissue homeostasis. This review is focused on previous reports on different forms of cell death in intestinal epithelium.
Collapse
|
341
|
Yu S, Hwang HE, Yun N, Goldenring JR, Nam KT. The mRNA and Protein Levels of Tubulin and β-Actin Are Greatly Reduced in the Proximal Duodenum of Mice Relative to the Rest of the Small Intestines. Dig Dis Sci 2015; 60:2670-6. [PMID: 25976623 DOI: 10.1007/s10620-015-3688-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/24/2015] [Indexed: 12/09/2022]
Abstract
To accurately quantify mRNA and protein levels, it is critical to choose appropriate internal standards. As the expression of housekeeping genes is assumed to remain constant, they are often employed to normalize signals to correct for sample-to-sample variations. However, recent studies have documented that β-actin and Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) expression levels change in response to various stimuli during proliferation, activation, and differentiation. We investigated levels of α-, β-, γ-tubulin, β-actin, and GAPDH vary across the gastrointestinal tract of mice. We found that different regions of the small intestines had dramatically different expression profiles, as measured by western blot, quantitative Reverse transcription polymerase chain reaction (RT-PCR), and immunohistochemical staining. These results revealed that the expression levels of tubulins and β-actin were dramatically lower in the proximal duodenum, relative to the rest of the small intestines. These varying levels of housekeeping genes may reflect differences in the activities of specialized tissues and suggest unique requirements for tubulins in these tissue types. We conclude that the use of a single housekeeping gene to normalize gene expression in the gastrointestinal tracts of mice may introduce errors, as measured differences in gene expression may reflect regulation of the internal control rather than the mRNA or protein under investigation.
Collapse
Affiliation(s)
- Sungsook Yu
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea
| | | | | | | | | |
Collapse
|
342
|
Sun M, He C, Cong Y, Liu Z. Regulatory immune cells in regulation of intestinal inflammatory response to microbiota. Mucosal Immunol 2015; 8:969-978. [PMID: 26080708 PMCID: PMC4540654 DOI: 10.1038/mi.2015.49] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/04/2015] [Indexed: 02/07/2023]
Abstract
The intestinal lumen harbors nearly 100 trillion commensal bacteria that exert crucial function for health. An elaborate balance between immune responses and tolerance to intestinal microbiota is required to maintain intestinal homeostasis. This process depends on diverse regulatory mechanisms, including both innate and adaptive immunity. Dysregulation of the homeostasis between intestinal immune systems and microbiota has been shown to be associated with the development of inflammatory bowel diseases (IBD) in genetically susceptible populations. In this review, we discuss the recent progress reported in studies of distinct types of regulatory immune cells in the gut, including intestinal intraepithelial lymphocytes, Foxp3(+) regulatory T cells, regulatory B cells, alternatively activated macrophages, dendritic cells, and innate lymphoid cells, and how dysfunction of this immune regulatory system contributes to intestinal diseases such as IBD. Moreover, we discuss the manipulation of these regulatory immune cells as a potential therapeutic method for management of intestinal inflammatory disorders.
Collapse
Affiliation(s)
- M Sun
- Department of Gastroenterology, Institute for Intestinal Diseases, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - C He
- Department of Gastroenterology, Institute for Intestinal Diseases, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Y Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Z Liu
- Department of Gastroenterology, Institute for Intestinal Diseases, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
343
|
Iyer A, Brown L, Whitehead JP, Prins JB, Fairlie DP. Nutrient and immune sensing are obligate pathways in metabolism, immunity, and disease. FASEB J 2015; 29:3612-25. [PMID: 26065858 DOI: 10.1096/fj.15-271155] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 06/02/2015] [Indexed: 12/13/2022]
Abstract
The growth and survival of multicellular organisms depend upon their abilities to acquire and metabolize nutrients, efficiently store and harness energy, and sense and fight infection. Systems for sensing and using nutrients have consequently coevolved alongside systems for sensing and responding to danger signals, including pathogens, and share many of the same cell signaling proteins and networks. Diets rich in carbohydrates and fats can overload these systems, leading to obesity, metabolic dysfunction, impaired immunity, and cardiovascular disease. Excessive nutrient intake promotes adiposity, typically altering adipocyte function and immune cell distribution, both of which trigger metabolic dysfunction. Here, we discuss novel mechanistic links between metabolism and immunity that underlie metabolic dysfunction in obesity. We aim to stimulate debate about how the endocrine and immune systems are connected through autocrine, paracrine, and neuroendocrine signaling in sophisticated networks that are only now beginning to be resolved. Understanding the expression and action of signaling proteins, together with modulating their receptors or pattern recognition using agonists or antagonists, will enable rational intervention in immunometabolism that may lead to novel treatments for obesity and metabolic dysfunction.
Collapse
Affiliation(s)
- Abishek Iyer
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - Lindsay Brown
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - Jonathan P Whitehead
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - Johannes B Prins
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - David P Fairlie
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| |
Collapse
|
344
|
Trejo-Vargas A, Hernández-Mercado E, Ordóñez-Razo RM, Lazzarini R, Arenas-Aranda DJ, Gutiérrez-Ruiz MC, Königsberg M, Luna-López A. Bik subcellular localization in response to oxidative stress induced by chemotherapy, in Two different breast cancer cell lines and a Non-tumorigenic epithelial cell line. J Appl Toxicol 2015; 35:1262-70. [PMID: 26059411 DOI: 10.1002/jat.3173] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 03/27/2015] [Accepted: 04/12/2015] [Indexed: 11/09/2022]
Abstract
Cancer chemotherapy remains one of the preferred therapeutic modalities against malignancies despite its damaging side effects. An expected outcome while utilizing chemotherapy is apoptosis induction. This is mainly regulated by a group of proteins known as the Bcl-2 family, usually found within the endoplasmic reticulum or the mitochondria. Recently, these proteins have been located in other sites and non-canonic functions have been unraveled. Bik is a pro-apoptotic protein, which becomes deregulated in cancer, and as apoptosis is associated with oxidative stress generation, our objective was to determine the subcellular localization of Bik either after a direct oxidative insult due to H2 O2 , or indirectly by cisplatin, an antineoplastic agent. Experiments were performed in two human transformed mammary gland cell lines MDA-MB-231 and MCF-7, and one non-tumorigenic epithelial cell line MCF-10A. Our results showed that in MCF-7, Bik is localized within the cytosol and that after oxidative stress treatment it translocates into the nucleus. However, in MDA-MB-231, Bik localizes in the nucleus and translocates to the cytosol. In MCF10A Bik did not change its cellular site after either treatment. Interestingly, MCF10A were more resistant to cisplatin than transformed cell lines. This is the first report showing that Bik is located in different cellular compartments depending on the cancer stage, and it has the ability to change its subcellular localization in response to oxidative stress. This is associated with increased sensitivity when exposed to toxic agents, thus rendering novel opportunities to study new therapeutic targets allowing the development of more active and less harmful agents.
Collapse
Affiliation(s)
- Aglaé Trejo-Vargas
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, México, D.F., México.,Unidad Médica en Genética Humana, Centro Médico Nacional Siglo XXI, México, D.F., Mexico.,Posgrado en Biologia Experimental
| | - Elisa Hernández-Mercado
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, México, D.F., México.,Unidad Médica en Genética Humana, Centro Médico Nacional Siglo XXI, México, D.F., Mexico
| | | | - Roberto Lazzarini
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, México, D.F., México
| | | | | | - Mina Königsberg
- Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, México, D.F., México
| | - Armando Luna-López
- Instituto Nacional de Geriatria, SSA, Posgrado en Biologia Experimental, UAMI, México, D.F., Mexico
| |
Collapse
|
345
|
The Wnt5a-Ror2 axis promotes the signaling circuit between interleukin-12 and interferon-γ in colitis. Sci Rep 2015; 5:10536. [PMID: 26030277 PMCID: PMC4450756 DOI: 10.1038/srep10536] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/23/2015] [Indexed: 01/30/2023] Open
Abstract
Wnt5a, which regulates various cellular functions in Wnt signaling, is involved in inflammatory responses, however the mechanism is not well understood. We examined the role of Wnt5a signaling in intestinal immunity using conditional knockout mice for Wnt5a and its receptor Ror2. Removing Wnt5a or Ror2 in adult mice suppressed dextran sodium sulfate (DSS)-induced colitis. It also attenuated the DSS-dependent increase in inflammatory cytokine production and decreased interferon-γ (IFN-γ)-producing CD4+ Th1 cell numbers in the colon. Wnt5a was highly expressed in stromal fibroblasts in ulcerative lesions in the DSS-treated mice and inflammatory bowel disease patients. Dendritic cells (DCs) isolated from the colon of Wnt5a and Ror2 deficient mice reduced the ability to differentiate naïve CD4+ T cells to IFN-γ-producing CD4+ Th1 cells. In vitro experiments demonstrated that the Wnt5a-Ror2 signaling axis augmented the DCs priming effect of IFN-γ, leading to enhanced lipopolysaccharide (LPS)-induced interleukin (IL)-12 expression. Taken together, these results suggest that Wnt5a promotes IFN-γ signaling, leading to IL-12 expression in DCs, and thereby inducing Th1 differentiation in colitis.
Collapse
|
346
|
Cheng P, Yao J, Wang C, Zhang L, Kong W. Molecular and cellular mechanisms of tight junction dysfunction in the irritable bowel syndrome. Mol Med Rep 2015; 12:3257-3264. [PMID: 25998845 PMCID: PMC4526093 DOI: 10.3892/mmr.2015.3808] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 03/16/2015] [Indexed: 12/24/2022] Open
Abstract
The pathophysiological mechanisms of the irritable bowel syndrome (IBS), one of the most prevalent gastrointestinal disorders, are complex and have not been fully elucidated. The present study aimed to investigate the molecular and cellular mechanisms of tight junction (TJ) dysfunction in IBS. Intestinal tissues of IBS and non‑IBS patients were examined to observe cellular changes by cell chemical tracer electron microscopy and transmission electron microscopy, and intestinal claudin‑1 protein was detected by immunohistochemistry, western blot analysis and fluorescence quantitative polymerase chain reaction. Compared with the control group, TJ broadening and the tracer extravasation phenomenon were observed in the diarrhea‑predominant IBS group, and a greater number of neuroendocrine cells and mast cells filled with high‑density particles in the endocrine package pulp as well as a certain extent of vacuolization were present. The expression of claudin‑1 in diarrhea‑predominant IBS patients was decreased, while it was increased in constipation‑predominant IBS patients. In conclusion, the results of the present study indicated that changes in cellular structure and claudin‑1 levels were associated with Tjs in IBS.
Collapse
Affiliation(s)
- Peng Cheng
- Department of Gastroenterology, First Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jianning Yao
- Department of Gastroenterology, First Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chunfeng Wang
- Department of Gastroenterology, First Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Lianfeng Zhang
- Department of Gastroenterology, First Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Wuming Kong
- Department of Gastroenterology, Affiliated Sixth People's Hospital of Shanghai Jiaotong University, Shanghai 201306, P.R. China
| |
Collapse
|
347
|
Wang L, Ryoo HD, Qi Y, Jasper H. PERK Limits Drosophila Lifespan by Promoting Intestinal Stem Cell Proliferation in Response to ER Stress. PLoS Genet 2015; 11:e1005220. [PMID: 25945494 PMCID: PMC4422665 DOI: 10.1371/journal.pgen.1005220] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 04/14/2015] [Indexed: 12/19/2022] Open
Abstract
Intestinal homeostasis requires precise control of intestinal stem cell (ISC) proliferation. In Drosophila, this control declines with age largely due to chronic activation of stress signaling and associated chronic inflammatory conditions. An important contributor to this condition is the age-associated increase in endoplasmic reticulum (ER) stress. Here we show that the PKR-like ER kinase (PERK) integrates both cell-autonomous and non-autonomous ER stress stimuli to induce ISC proliferation. In addition to responding to cell-intrinsic ER stress, PERK is also specifically activated in ISCs by JAK/Stat signaling in response to ER stress in neighboring cells. The activation of PERK is required for homeostatic regeneration, as well as for acute regenerative responses, yet the chronic engagement of this response becomes deleterious in aging flies. Accordingly, knocking down PERK in ISCs is sufficient to promote intestinal homeostasis and extend lifespan. Our studies highlight the significance of the PERK branch of the unfolded protein response of the ER (UPRER) in intestinal homeostasis and provide a viable strategy to improve organismal health- and lifespan. The long-term maintenance of tissue homeostasis in barrier epithelia requires precise coordination of cellular stress and inflammatory responses with regenerative processes. This coordination is lost with age, resulting in degenerative and proliferative diseases. The Unfolded Protein Response of the Endoplasmic Reticulum (UPRER) is emerging as a central regulator of tissue homeostasis in barrier epithelia. The UPRER adjusts the protein folding capacity of the ER in response to protein stress in stem cells and differentiated cells, and thus influences proliferative homeostasis, cell differentiation and epithelial inflammatory responses. How these responses are coordinated to maintain epithelial homeostasis in aging organisms remains unclear. In a previous study, we have found that the UPRER controls intestinal stem cell (ISC) proliferation in the Drosophila intestinal epithelium by influencing the intracellular redox state. How signaling through the canonical ER stress sensor PERK (PKR-like ER kinase) is integrated into this signaling network remained unclear. Here we show that PERK serves as a central regulator of ISC proliferation and tissue homeostasis in response ER stress. Strikingly, we find that within the intestinal epithelium, PERK is activated specifically in ISCs in response to both systemic and local ER stress, and is required for ISC proliferation under both homeostatic and stress conditions. We identify JAK/Stat signaling as an activator of PERK in ISCs in response to ER stress in neighboring cells, and find that the wide-spread age-associated increase in PERK activity in ISCs is a cause of age-related dysplasia in this tissue. Accordingly, limiting PERK activity in ISCs promotes homeostasis of the intestinal epithelium in old flies and extends lifespan.
Collapse
Affiliation(s)
- Lifen Wang
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Hyung Don Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
| | - Yanyan Qi
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Heinrich Jasper
- Buck Institute for Research on Aging, Novato, California, United States of America
- * E-mail:
| |
Collapse
|
348
|
Differential profiles of gastrointestinal proteins interacting with peptidoglycans from Lactobacillus plantarum and Staphylococcus aureus. Mol Immunol 2015; 65:77-85. [DOI: 10.1016/j.molimm.2015.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 12/24/2014] [Accepted: 01/08/2015] [Indexed: 12/17/2022]
|
349
|
Abstract
Inflammation has long been suspected to play a major role in the pathogenesis of cancer. Only recently, however, have some mechanisms of its tumor promoting effects become known. Microbes, both commensal and pathogenic, are critical regulators of the host immune system and, ultimately, of inflammation. Consequently, microbes have the potential power to influence tumor progression as well, through a wide variety of routes, including chronic activation of inflammation, alteration of tumor microenvironment, induction of genotoxic responses, and metabolism. In this review, we will provide a general overview of commensal microbiota, inflammation, and cancer, as well as how microbes fit into this emerging field.
Collapse
|
350
|
Oral tolerance failure upon neonatal gut colonization with Escherichia coli producing the genotoxin colibactin. Infect Immun 2015; 83:2420-9. [PMID: 25824839 DOI: 10.1128/iai.00064-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 03/23/2015] [Indexed: 01/06/2023] Open
Abstract
The intestinal barrier controls the balance between tolerance and immunity to luminal antigens. When this finely tuned equilibrium is deregulated, inflammatory disorders can occur. There is a concomitant increase, in urban populations of developed countries, of immune-mediated diseases along with a shift in Escherichia coli population from the declining phylogenetic group A to the newly dominant group B2, including commensal strains producing a genotoxin called colibactin that massively colonized the gut of neonates. Here, we showed that mother-to-offspring early gut colonization by colibactin-producing E. coli impairs intestinal permeability and enhances the transepithelial passage of luminal antigen, leading to an increased immune activation. Functionally, this was accompanied by a dramatic increase in local and systemic immune responses against a fed antigen, decreased regulatory T cell population, tolerogenic dendritic cells, and enhanced mucosal delayed-type hypersensitivity response. Conversely, the abolition of colibactin expression by mutagenesis abrogates the alteration of oral tolerance induced by neonatal colonization by E. coli. In conclusion, the vertical colonization by E. coli producing the genotoxin colibactin enhances intestinal translocation and subsequently alters oral tolerance. Thus, early colonization by E. coli from the newly dominant phylogenetic group B2, which produces colibactin, may represent a risk factor for the development of immune-mediated diseases.
Collapse
|