151
|
Bleuler-Martinez S, Stutz K, Sieber R, Collot M, Mallet JM, Hengartner M, Schubert M, Varrot A, Künzler M. Dimerization of the fungal defense lectin CCL2 is essential for its toxicity against nematodes. Glycobiology 2017; 27:486-500. [PMID: 27980000 DOI: 10.1093/glycob/cww113] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 11/09/2016] [Indexed: 11/12/2022] Open
Abstract
Lectins are used as defense effector proteins against predators, parasites and pathogens by animal, plant and fungal innate defense systems. These proteins bind to specific glycoepitopes on the cell surfaces and thereby interfere with the proper cellular functions of the various antagonists. The exact cellular toxicity mechanism is in many cases unclear. Lectin CCL2 of the mushroom Coprinopsis cinerea was previously shown to be toxic for Caenorhabditis elegans and Drosophila melanogaster. This toxicity is dependent on a single, high-affinity binding site for the trisaccharide GlcNAc(Fucα1,3)β1,4GlcNAc, which is a hallmark of nematode and insect N-glycan cores. The carbohydrate-binding site is located at an unusual position on the protein surface when compared to other β-trefoil lectins. Here, we show that CCL2 forms a compact dimer in solution and in crystals. Substitution of two amino acid residues at the dimer interface, R18A and F133A, interfered with dimerization of CCL2 and reduced toxicity but left carbohydrate-binding unaffected. These results, together with the positioning of the two carbohydrate-binding sites on the surface of the protein dimer, suggest that crosslinking of N-glycoproteins on the surface of intestinal cells of invertebrates is a crucial step in the mechanism of CCL2-mediated toxicity. Comparisons of the number and positioning of carbohydrate-binding sites among different dimerizing fungal β-trefoil lectins revealed a considerable variability in the carbohydrate-binding patterns of these proteins, which are likely to correlate with their respective functions.
Collapse
Affiliation(s)
| | - Katrin Stutz
- Institute of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Ramon Sieber
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Mayeul Collot
- Laboratoire des Biomolécules, UPMC Université Paris 06, Ecole Normale Supérieure, Paris, France
| | - Jean-Maurice Mallet
- Laboratoire des Biomolécules, UPMC Université Paris 06, Ecole Normale Supérieure, Paris, France
| | - Michael Hengartner
- Institute of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Mario Schubert
- Institute of Molecular Biology and Biophysics, ETH Zürich, Schafmattstr. 20, 8093 Zürich, Switzerland.,Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, 5020 Salzburg, Austria
| | - Annabelle Varrot
- CERMAV, UPR5301, CNRS and Université Grenoble Alpes, 38041 Grenoble, France
| | - Markus Künzler
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| |
Collapse
|
152
|
Selber-Hnatiw S, Rukundo B, Ahmadi M, Akoubi H, Al-Bizri H, Aliu AF, Ambeaghen TU, Avetisyan L, Bahar I, Baird A, Begum F, Ben Soussan H, Blondeau-Éthier V, Bordaries R, Bramwell H, Briggs A, Bui R, Carnevale M, Chancharoen M, Chevassus T, Choi JH, Coulombe K, Couvrette F, D'Abreau S, Davies M, Desbiens MP, Di Maulo T, Di Paolo SA, Do Ponte S, Dos Santos Ribeiro P, Dubuc-Kanary LA, Duncan PK, Dupuis F, El-Nounou S, Eyangos CN, Ferguson NK, Flores-Chinchilla NR, Fotakis T, Gado Oumarou H D M, Georgiev M, Ghiassy S, Glibetic N, Grégoire Bouchard J, Hassan T, Huseen I, Ibuna Quilatan MF, Iozzo T, Islam S, Jaunky DB, Jeyasegaram A, Johnston MA, Kahler MR, Kaler K, Kamani C, Karimian Rad H, Konidis E, Konieczny F, Kurianowicz S, Lamothe P, Legros K, Leroux S, Li J, Lozano Rodriguez ME, Luponio-Yoffe S, Maalouf Y, Mantha J, McCormick M, Mondragon P, Narayana T, Neretin E, Nguyen TTT, Niu I, Nkemazem RB, O'Donovan M, Oueis M, Paquette S, Patel N, Pecsi E, Peters J, Pettorelli A, Poirier C, Pompa VR, Rajen H, Ralph RO, Rosales-Vasquez J, Rubinshtein D, Sakr S, Sebai MS, Serravalle L, Sidibe F, Sinnathurai A, Soho D, Sundarakrishnan A, Svistkova V, Ugbeye TE, Vasconcelos MS, Vincelli M, Voitovich O, Vrabel P, Wang L, Wasfi M, Zha CY, Gamberi C. Human Gut Microbiota: Toward an Ecology of Disease. Front Microbiol 2017; 8:1265. [PMID: 28769880 PMCID: PMC5511848 DOI: 10.3389/fmicb.2017.01265] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/23/2017] [Indexed: 12/17/2022] Open
Abstract
Composed of trillions of individual microbes, the human gut microbiota has adapted to the uniquely diverse environments found in the human intestine. Quickly responding to the variances in the ingested food, the microbiota interacts with the host via reciprocal biochemical signaling to coordinate the exchange of nutrients and proper immune function. Host and microbiota function as a unit which guards its balance against invasion by potential pathogens and which undergoes natural selection. Disturbance of the microbiota composition, or dysbiosis, is often associated with human disease, indicating that, while there seems to be no unique optimal composition of the gut microbiota, a balanced community is crucial for human health. Emerging knowledge of the ecology of the microbiota-host synergy will have an impact on how we implement antibiotic treatment in therapeutics and prophylaxis and how we will consider alternative strategies of global remodeling of the microbiota such as fecal transplants. Here we examine the microbiota-human host relationship from the perspective of the microbial community dynamics.
Collapse
Affiliation(s)
| | - Belise Rukundo
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Masoumeh Ahmadi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Hayfa Akoubi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Hend Al-Bizri
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Adelekan F Aliu
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Lilit Avetisyan
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Irmak Bahar
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Alexandra Baird
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Fatema Begum
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | - Helene Bramwell
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Alicia Briggs
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Richard Bui
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Talia Chevassus
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Jin H Choi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Karyne Coulombe
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Meghan Davies
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Tamara Di Maulo
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | - Paola K Duncan
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Sara El-Nounou
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | - Tanya Fotakis
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Metodi Georgiev
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | - Tazkia Hassan
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Iman Huseen
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Tania Iozzo
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Safina Islam
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Dilan B Jaunky
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | - Cedric Kamani
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Filip Konieczny
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Karina Legros
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Jun Li
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Yara Maalouf
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Jessica Mantha
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | - Thi T T Nguyen
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Ian Niu
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | - Matthew Oueis
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Nehal Patel
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Emily Pecsi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Jackie Peters
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | | | | | | | - Surya Sakr
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Lisa Serravalle
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Fily Sidibe
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | - Dominique Soho
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | | | | | | | | | | | - Olga Voitovich
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Pamela Vrabel
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Lu Wang
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Maryse Wasfi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Cong Y Zha
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| | - Chiara Gamberi
- Department of Biology, Concordia UniversityMontréal, QC, Canada
| |
Collapse
|
153
|
Bierschenk D, Boucher D, Schroder K. Salmonella- induced inflammasome activation in humans. Mol Immunol 2017; 86:38-43. [DOI: 10.1016/j.molimm.2016.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 11/01/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022]
|
154
|
Okumura R, Takeda K. Roles of intestinal epithelial cells in the maintenance of gut homeostasis. Exp Mol Med 2017; 49:e338. [PMID: 28546564 PMCID: PMC5454438 DOI: 10.1038/emm.2017.20] [Citation(s) in RCA: 425] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/02/2017] [Indexed: 12/15/2022] Open
Abstract
The intestine is a unique organ inhabited by a tremendous number of microorganisms. Intestinal epithelial cells greatly contribute to the maintenance of the symbiotic relationship between gut microbiota and the host by constructing mucosal barriers, secreting various immunological mediators and delivering bacterial antigens. Mucosal barriers, including physical barriers and chemical barriers, spatially segregate gut microbiota and the host immune system to avoid unnecessary immune responses to gut microbes, leading to the intestinal inflammation. In addition, various immunological mediators, including cytokines and chemokines, secreted from intestinal epithelial cells stimulated by gut microbiota modulate host immune responses, maintaining a well-balanced relationship between gut microbes and the host immune system. Therefore, impairment of the innate immune functions of intestinal epithelial cells is associated with intestinal inflammation.
Collapse
Affiliation(s)
- Ryu Okumura
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
155
|
Recognition of microbial glycans by soluble human lectins. Curr Opin Struct Biol 2017; 44:168-178. [PMID: 28482337 DOI: 10.1016/j.sbi.2017.04.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/31/2017] [Accepted: 04/05/2017] [Indexed: 01/28/2023]
Abstract
Human innate immune lectins that recognize microbial glycans can conduct microbial surveillance and thereby help prevent infection. Structural analysis of soluble lectins has provided invaluable insight into how these proteins recognize their cognate carbohydrate ligands and how this recognition gives rise to biological function. In this opinion, we cover the structural features of lectins that allow them to mediate microbial recognition, highlighting examples from the collectin, Reg protein, galectin, pentraxin, ficolin and intelectin families. These analyses reveal how some lectins (e.g., human intelectin-1) can recognize glycan epitopes that are remarkably diverse, yet still differentiate between mammalian and microbial glycans. We additionally discuss strategies to identify lectins that recognize microbial glycans and highlight tools that facilitate these discovery efforts.
Collapse
|
156
|
Innate immunity mediated longevity and longevity induced by germ cell removal converge on the C-type lectin domain protein IRG-7. PLoS Genet 2017; 13:e1006577. [PMID: 28196094 PMCID: PMC5308781 DOI: 10.1371/journal.pgen.1006577] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 01/10/2017] [Indexed: 01/22/2023] Open
Abstract
In C. elegans, removal of the germline triggers molecular events in the neighboring intestine, which sends an anti-aging signal to the rest of the animal. In this study, we identified an innate immunity related gene, named irg-7, as a novel mediator of longevity in germlineless animals. We consider irg-7 to be an integral downstream component of the germline longevity pathway because its expression increases upon germ cell removal and its depletion interferes with the activation of the longevity-promoting transcription factors DAF-16 and DAF-12 in germlineless animals. Furthermore, irg-7 activation by itself sensitizes the animals' innate immune response and extends the lifespan of animals exposed to live bacteria. This lifespan-extending pathogen resistance relies on the somatic gonad as well as on many genes previously associated with the reproductive longevity pathway. This suggests that these genes are also relevant in animals with an intact gonad, and can affect their resistance to pathogens. Altogether, this study demonstrates the tight association between germline homeostasis and the immune response of animals, and raises the possibility that the reproductive system can act as a signaling center to divert resources towards defending against putative pathogen attacks.
Collapse
|
157
|
Carvalho RD, Breyner N, Menezes-Garcia Z, Rodrigues NM, Lemos L, Maioli TU, da Gloria Souza D, Carmona D, de Faria AMC, Langella P, Chatel JM, Bermúdez-Humarán LG, Figueiredo HCP, Azevedo V, de Azevedo MS. Secretion of biologically active pancreatitis-associated protein I (PAP) by genetically modified dairy Lactococcus lactis NZ9000 in the prevention of intestinal mucositis. Microb Cell Fact 2017; 16:27. [PMID: 28193209 PMCID: PMC5307810 DOI: 10.1186/s12934-017-0624-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022] Open
Abstract
Background Mucositis is one of the most relevant gastrointestinal inflammatory conditions in humans, generated by the use of chemotherapy drugs, such as 5-fluoracil (5-FU). 5-FU-induced mucositis affects 80% of patients undergoing oncological treatment causing mucosal gut dysfunctions and great discomfort. As current therapy drugs presents limitations in alleviating mucositis symptoms, alternative strategies are being pursued. Recent studies have shown that the antimicrobial pancreatitis-associated protein (PAP) has a protective role in intestinal inflammatory processes. Indeed, it was demonstrated that a recombinant strain of Lactococcus lactis expressing human PAP (LL-PAP) could prevent and improve murine DNBS-induced colitis, an inflammatory bowel disease (IBD) that causes severe inflammation of the colon. Hence, in this study we sought to evaluate the protective effects of LL-PAP on 5-FU-induced experimental mucositis in BALB/c mice as a novel approach to treat the disease. Results Our results show that non-recombinant L. lactis NZ9000 have antagonistic activity, in vitro, against the enteroinvasive gastrointestinal pathogen L. monocytogenes and confirmed PAP inhibitory effect against Opportunistic E. faecalis. Moreover, L. lactis was able to prevent histological damage, reduce neutrophil and eosinophil infiltration and secretory Immunoglobulin-A in mice injected with 5-FU. Recombinant lactococci carrying antimicrobial PAP did not improve those markers of inflammation, although its expression was associated with villous architecture preservation and increased secretory granules density inside Paneth cells in response to 5-FU inflammation. Conclusions We have demonstrated for the first time that L. lactis NZ9000 by itself, is able to prevent 5-FU-induced intestinal inflammation in BALB/c mice. Moreover, PAP delivered by recombinant L. lactis strain showed additional protective effects in mice epithelium, revealing to be a promising strategy to treat intestinal mucositis.
Collapse
Affiliation(s)
- Rodrigo D Carvalho
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Natalia Breyner
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Zelia Menezes-Garcia
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Nubia M Rodrigues
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Luisa Lemos
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Tatiane U Maioli
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Danielle da Gloria Souza
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil.,Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Denise Carmona
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Ana M C de Faria
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Philippe Langella
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Jean-Marc Chatel
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Luis G Bermúdez-Humarán
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Henrique C P Figueiredo
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Vasco Azevedo
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Marcela S de Azevedo
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
158
|
Chairatana P, Nolan EM. Defensins, lectins, mucins, and secretory immunoglobulin A: microbe-binding biomolecules that contribute to mucosal immunity in the human gut. Crit Rev Biochem Mol Biol 2017; 52:45-56. [PMID: 27841019 PMCID: PMC5233583 DOI: 10.1080/10409238.2016.1243654] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/19/2016] [Accepted: 09/28/2016] [Indexed: 02/08/2023]
Abstract
In the intestine, the mucosal immune system plays essential roles in maintaining homeostasis between the host and microorganisms, and protecting the host from pathogenic invaders. Epithelial cells produce and release a variety of biomolecules into the mucosa and lumen that contribute to immunity. In this review, we focus on a subset of these remarkable host-defense factors - enteric α-defensins, select lectins, mucins, and secretory immunoglobulin A - that have the capacity to bind microbes and thereby contribute to barrier function in the human gut. We provide an overview of the intestinal epithelium, describe specialized secretory cells named Paneth cells, and summarize our current understanding of the biophysical and functional properties of these select microbe-binding biomolecules. We intend for this compilation to complement prior reviews on intestinal host-defense factors, highlight recent advances in the field, and motivate investigations that further illuminate molecular mechanisms as well as the interplay between these molecules and microbes.
Collapse
Affiliation(s)
- Phoom Chairatana
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
159
|
Shi N, Li N, Duan X, Niu H. Interaction between the gut microbiome and mucosal immune system. Mil Med Res 2017; 4:14. [PMID: 28465831 PMCID: PMC5408367 DOI: 10.1186/s40779-017-0122-9] [Citation(s) in RCA: 338] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 04/10/2017] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota, the largest symbiotic ecosystem with the host, has been shown to play important roles in maintaining intestinal homeostasis. Dysbiosis of the gut microbiome is caused by the imbalance between the commensal and pathogenic microbiomes. The commensal microbiome regulates the maturation of the mucosal immune system, while the pathogenic microbiome causes immunity dysfunction, resulting in disease development. The gut mucosal immune system, which consists of lymph nodes, lamina propria and epithelial cells, constitutes a protective barrier for the integrity of the intestinal tract. The composition of the gut microbiota is under the surveillance of the normal mucosal immune system. Inflammation, which is caused by abnormal immune responses, influences the balance of the gut microbiome, resulting in intestinal diseases. In this review, we briefly outlined the interaction between the gut microbiota and the immune system and provided a reference for future studies.
Collapse
Affiliation(s)
- Na Shi
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical Collage (PUMC), Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, 100021 China
| | - Na Li
- Department of Rheumatology, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006 China
| | - Xinwang Duan
- Department of Rheumatology, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006 China
| | - Haitao Niu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Center, Peking Union Medical Collage (PUMC), Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing, 100021 China
| |
Collapse
|
160
|
Muramoto K. Lectins as Bioactive Proteins in Foods and Feeds. FOOD SCIENCE AND TECHNOLOGY RESEARCH 2017. [DOI: 10.3136/fstr.23.487] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
161
|
Valeri M, Raffatellu M. Cytokines IL-17 and IL-22 in the host response to infection. Pathog Dis 2016; 74:ftw111. [PMID: 27915228 PMCID: PMC5975231 DOI: 10.1093/femspd/ftw111] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/13/2016] [Accepted: 11/17/2016] [Indexed: 12/12/2022] Open
Abstract
Cytokines IL-17 and IL-22 play pivotal roles in host defense against microbes and in the development of chronic inflammatory diseases. These cytokines are produced by cells that are often located in epithelial barriers, including subsets of T cells and innate lymphoid cells. In general, IL-17 and IL-22 can be characterized as important cytokines in the rapid response to infectious agents, both by recruiting neutrophils and by inducing the production of antimicrobial peptides. Although each cytokine induces an innate immune response in epithelial cells, their functional spectra are generally distinct: IL-17 mainly induces an inflammatory tissue response and is involved in the pathogenesis of several autoimmune diseases, whereas IL-22 is largely protective and regenerative. In this review, we compare IL-17 and IL-22, describing overlaps and differences in their cellular sources as well as their regulation, signaling, biological functions and roles during disease, with a focus on the contribution of these cytokines to the gut mucosal barrier during bacterial infection.
Collapse
Affiliation(s)
- Maria Valeri
- Department of Microbiology and Molecular Genetics, University of California Irvine School of Medicine, Irvine, CA 92697-4025, USA
- Institute for Immunology, University of California Irvine School of Medicine, Irvine, CA 92697-4025, USA
| | - Manuela Raffatellu
- Department of Microbiology and Molecular Genetics, University of California Irvine School of Medicine, Irvine, CA 92697-4025, USA
- Institute for Immunology, University of California Irvine School of Medicine, Irvine, CA 92697-4025, USA
| |
Collapse
|
162
|
Wippler J, Kleiner M, Lott C, Gruhl A, Abraham PE, Giannone RJ, Young JC, Hettich RL, Dubilier N. Transcriptomic and proteomic insights into innate immunity and adaptations to a symbiotic lifestyle in the gutless marine worm Olavius algarvensis. BMC Genomics 2016; 17:942. [PMID: 27871231 PMCID: PMC5117596 DOI: 10.1186/s12864-016-3293-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
Background The gutless marine worm Olavius algarvensis has a completely reduced digestive and excretory system, and lives in an obligate nutritional symbiosis with bacterial symbionts. While considerable knowledge has been gained of the symbionts, the host has remained largely unstudied. Here, we generated transcriptomes and proteomes of O. algarvensis to better understand how this annelid worm gains nutrition from its symbionts, how it adapted physiologically to a symbiotic lifestyle, and how its innate immune system recognizes and responds to its symbiotic microbiota. Results Key adaptations to the symbiosis include (i) the expression of gut-specific digestive enzymes despite the absence of a gut, most likely for the digestion of symbionts in the host's epidermal cells; (ii) a modified hemoglobin that may bind hydrogen sulfide produced by two of the worm’s symbionts; and (iii) the expression of a very abundant protein for oxygen storage, hemerythrin, that could provide oxygen to the symbionts and the host under anoxic conditions. Additionally, we identified a large repertoire of proteins involved in interactions between the worm's innate immune system and its symbiotic microbiota, such as peptidoglycan recognition proteins, lectins, fibrinogen-related proteins, Toll and scavenger receptors, and antimicrobial proteins. Conclusions We show how this worm, over the course of evolutionary time, has modified widely-used proteins and changed their expression patterns in adaptation to its symbiotic lifestyle and describe expressed components of the innate immune system in a marine oligochaete. Our results provide further support for the recent realization that animals have evolved within the context of their associations with microbes and that their adaptive responses to symbiotic microbiota have led to biological innovations. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3293-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juliane Wippler
- Symbiosis Department, Max Planck Institute for Marine Microbiology, Celsiusstr. 1, D-28359, Bremen, Germany. .,Symbiosis Department, Max Planck Institute for Marine Microbiology, Celsiusstr. 1, D-28359, Bremen, Germany.
| | - Manuel Kleiner
- Symbiosis Department, Max Planck Institute for Marine Microbiology, Celsiusstr. 1, D-28359, Bremen, Germany. .,Energy Bioengineering and Geomicrobiology Research Group, University of Calgary, Calgary, T2N 1N4, AB, Canada.
| | - Christian Lott
- Symbiosis Department, Max Planck Institute for Marine Microbiology, Celsiusstr. 1, D-28359, Bremen, Germany.,HYDRA Institute for Marine Sciences, Elba Field Station, Via del Forno 80, 57034, Campo nell' Elba, (LI), Italy
| | - Alexander Gruhl
- Symbiosis Department, Max Planck Institute for Marine Microbiology, Celsiusstr. 1, D-28359, Bremen, Germany
| | - Paul E Abraham
- Oak Ridge National Laboratory, Chemical Sciences Division, Oak Ridge, Tennessee, 1 Bethel Valley Rd, Oak Ridge, TN, 37831, USA
| | - Richard J Giannone
- Oak Ridge National Laboratory, Chemical Sciences Division, Oak Ridge, Tennessee, 1 Bethel Valley Rd, Oak Ridge, TN, 37831, USA
| | - Jacque C Young
- Oak Ridge National Laboratory, Chemical Sciences Division, Oak Ridge, Tennessee, 1 Bethel Valley Rd, Oak Ridge, TN, 37831, USA.,Present Address: Saul Ewing LLP, 1500 Market Street, 37th Floor, Philadelphia, PA, 19102-2186, USA
| | - Robert L Hettich
- Oak Ridge National Laboratory, Chemical Sciences Division, Oak Ridge, Tennessee, 1 Bethel Valley Rd, Oak Ridge, TN, 37831, USA
| | - Nicole Dubilier
- Symbiosis Department, Max Planck Institute for Marine Microbiology, Celsiusstr. 1, D-28359, Bremen, Germany
| |
Collapse
|
163
|
Chairatana P, Nolan EM. Defensins, lectins, mucins, and secretory immunoglobulin A: microbe-binding biomolecules that contribute to mucosal immunity in the human gut. Crit Rev Biochem Mol Biol 2016. [PMID: 27841019 DOI: 10,1080/10409238.2016.124365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the intestine, the mucosal immune system plays essential roles in maintaining homeostasis between the host and microorganisms, and protecting the host from pathogenic invaders. Epithelial cells produce and release a variety of biomolecules into the mucosa and lumen that contribute to immunity. In this review, we focus on a subset of these remarkable host-defense factors - enteric α-defensins, select lectins, mucins, and secretory immunoglobulin A - that have the capacity to bind microbes and thereby contribute to barrier function in the human gut. We provide an overview of the intestinal epithelium, describe specialized secretory cells named Paneth cells, and summarize our current understanding of the biophysical and functional properties of these select microbe-binding biomolecules. We intend for this compilation to complement prior reviews on intestinal host-defense factors, highlight recent advances in the field, and motivate investigations that further illuminate molecular mechanisms as well as the interplay between these molecules and microbes.
Collapse
Affiliation(s)
- Phoom Chairatana
- a Department of Chemistry , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Elizabeth M Nolan
- a Department of Chemistry , Massachusetts Institute of Technology , Cambridge , MA , USA
| |
Collapse
|
164
|
Dang L, Van Damme EJM. Genome-wide identification and domain organization of lectin domains in cucumber. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2016; 108:165-176. [PMID: 27434144 DOI: 10.1016/j.plaphy.2016.07.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/04/2016] [Accepted: 07/09/2016] [Indexed: 05/21/2023]
Abstract
Lectins are ubiquitous proteins in plants and play important roles in a diverse set of biological processes, such as plant defense and cell signaling. Despite the availability of the Cucumis sativus L. genome sequence since 2009, little is known with respect to the occurrence of lectins in cucumber. In this study, a total of 146 putative lectin genes belonging to 10 different lectin families were identified and localized in the cucumber genome. Domain architecture analysis revealed that most of these lectin gene sequences contain multiple domains, where lectin domains are linked with other domains, as such creating chimeric lectin sequences encoding proteins with dual activities. This study provides an overview of lectin motifs in cucumber and will help to understand their potential biological role(s).
Collapse
Affiliation(s)
- Liuyi Dang
- Laboratory of Biochemistry and Glycobiology, Department of Molecular Biotechnology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium.
| | - Els J M Van Damme
- Laboratory of Biochemistry and Glycobiology, Department of Molecular Biotechnology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium.
| |
Collapse
|
165
|
Yang C, Jiang M, Wu F, Yu L, Tian J, Liu W, Lu X, Wen H. Identification of a C-type lectin from tilapia (Oreochromis niloticus) and its functional characterization under low-temperature stress. FISH & SHELLFISH IMMUNOLOGY 2016; 58:631-640. [PMID: 27717900 DOI: 10.1016/j.fsi.2016.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/29/2016] [Accepted: 10/01/2016] [Indexed: 06/06/2023]
Abstract
C-type lectin, which plays an important role in fish innate immunity, was cloned from tilapia and its functional characterization under low-temperature stress is reported. Its ORF is 453 bp, encoding 150 amino acids, and has a 5'UTR of 83 bp, a 3'UTR of 559 bp, and a poly (A) tail. The tilapia C-type lectin genomic DNA was acquired with a length of 5714 bp, containing six exons and five introns. Its promoter sequence was cloned and has a length of 2251 bp. The highest promoter activity occurs in the regulatory region (-900 bp to -450 bp). A hemagglutination assay of recombinant tilapia C-type lectin protein showed positive hemagglutination of rabbit and tilapia erythrocytes. RT-qPCR and western blot assays showed that its expression in the liver, spleen, and intestine were clearly affected by low-temperature stress. Thus, tilapia C-type lectin appear to be affected by abiotic stress, as well as by biological stress.
Collapse
Affiliation(s)
- ChangGeng Yang
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Ming Jiang
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Fan Wu
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Lijuan Yu
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Juan Tian
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Wei Liu
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Xing Lu
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Hua Wen
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China.
| |
Collapse
|
166
|
Wang L, Fouts DE, Stärkel P, Hartmann P, Chen P, Llorente C, DePew J, Moncera K, Ho SB, Brenner DA, Hooper LV, Schnabl B. Intestinal REG3 Lectins Protect against Alcoholic Steatohepatitis by Reducing Mucosa-Associated Microbiota and Preventing Bacterial Translocation. Cell Host Microbe 2016; 19:227-39. [PMID: 26867181 DOI: 10.1016/j.chom.2016.01.003] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 12/14/2015] [Accepted: 01/13/2016] [Indexed: 02/07/2023]
Abstract
Approximately half of all deaths from liver cirrhosis, the tenth leading cause of mortality in the United States, are related to alcohol use. Chronic alcohol consumption is accompanied by intestinal dysbiosis and bacterial overgrowth, yet little is known about the factors that alter the microbial composition or their contribution to liver disease. We previously associated chronic alcohol consumption with lower intestinal levels of the antimicrobial-regenerating islet-derived (REG)-3 lectins. Here, we demonstrate that intestinal deficiency in REG3B or REG3G increases numbers of mucosa-associated bacteria and enhances bacterial translocation to the mesenteric lymph nodes and liver, promoting the progression of ethanol-induced fatty liver disease toward steatohepatitis. Overexpression of Reg3g in intestinal epithelial cells restricts bacterial colonization of mucosal surfaces, reduces bacterial translocation, and protects mice from alcohol-induced steatohepatitis. Thus, alcohol appears to impair control of the mucosa-associated microbiota, and subsequent breach of the mucosal barrier facilitates progression of alcoholic liver disease.
Collapse
Affiliation(s)
- Lirui Wang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | - Peter Stärkel
- St. Luc University Hospital, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Phillipp Hartmann
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Peng Chen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | | | - Samuel B Ho
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - David A Brenner
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Lora V Hooper
- Howard Hughes Medical Institute; Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA.
| |
Collapse
|
167
|
Malanovic N, Lohner K. Antimicrobial Peptides Targeting Gram-Positive Bacteria. Pharmaceuticals (Basel) 2016; 9:E59. [PMID: 27657092 PMCID: PMC5039512 DOI: 10.3390/ph9030059] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/07/2016] [Accepted: 09/13/2016] [Indexed: 01/01/2023] Open
Abstract
Antimicrobial peptides (AMPs) have remarkably different structures as well as biological activity profiles, whereupon most of these peptides are supposed to kill bacteria via membrane damage. In order to understand their molecular mechanism and target cell specificity for Gram-positive bacteria, it is essential to consider the architecture of their cell envelopes. Before AMPs can interact with the cytoplasmic membrane of Gram-positive bacteria, they have to traverse the cell wall composed of wall- and lipoteichoic acids and peptidoglycan. While interaction of AMPs with peptidoglycan might rather facilitate penetration, interaction with anionic teichoic acids may act as either a trap for AMPs or a ladder for a route to the cytoplasmic membrane. Interaction with the cytoplasmic membrane frequently leads to lipid segregation affecting membrane domain organization, which affects membrane permeability, inhibits cell division processes or leads to delocalization of essential peripheral membrane proteins. Further, precursors of cell wall components, especially the highly conserved lipid II, are directly targeted by AMPs. Thereby, the peptides do not inhibit peptidoglycan synthesis via binding to proteins like common antibiotics, but form a complex with the precursor molecule, which in addition can promote pore formation and membrane disruption. Thus, the multifaceted mode of actions will make AMPs superior to antibiotics that act only on one specific target.
Collapse
Affiliation(s)
- Nermina Malanovic
- Institute of Molecular Biosciences, Biophysics Division, University of Graz, NAWI Graz, Austria.
| | - Karl Lohner
- Institute of Molecular Biosciences, Biophysics Division, University of Graz, NAWI Graz, Austria.
- BioTechMed Graz, Humboldtstrasse 50/III, 8010 Graz, Austria.
| |
Collapse
|
168
|
Cao J, Lv Y. Evolutionary analysis of the jacalin-related lectin family genes in 11 fishes. FISH & SHELLFISH IMMUNOLOGY 2016; 56:543-553. [PMID: 27514782 DOI: 10.1016/j.fsi.2016.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/12/2016] [Accepted: 08/06/2016] [Indexed: 06/06/2023]
Abstract
Jacalin-related lectins are a type of carbohydrate-binding proteins, which are distributed across a wide variety of organisms and involved in some important biological processes. The evolution of this gene family in fishes is unknown. Here, 47 putative jacalin genes in 11 fish species were identified and divided into 4 groups through phylogenetic analysis. Conserved gene organization and motif distribution existed in each group, suggesting their functional conservation. Some fishes have eleven jacalin genes, while others have only one or zero gene in their genomes, suggesting dynamic changes in the number of jacalin genes during the evolution of fishes. Intragenic recombination played a key role in the evolution of jacalin genes. Synteny analyses of jacalin genes in some fishes implied conserved and dynamic evolution characteristics of this gene family and related genome segments. Moreover, a few functional divergence sites were identified within each group pairs. Divergent expression profiles of the zebra fish jacalin genes were further investigated in different stresses. The results provided a foundation for exploring the characterization of the jacalin genes in fishes and will offer insights for additional functional studies.
Collapse
Affiliation(s)
- Jun Cao
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| | - Yueqing Lv
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
169
|
The goblet cell-derived mediator RELM-β drives spontaneous colitis in Muc2-deficient mice by promoting commensal microbial dysbiosis. Mucosal Immunol 2016; 9:1218-33. [PMID: 26813339 DOI: 10.1038/mi.2015.140] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 12/13/2015] [Indexed: 02/04/2023]
Abstract
Intestinal goblet cells are potentially key players in controlling susceptibility to ulcerative colitis (UC). Although impaired mucin (Muc2) production by goblet cells increases microbial stimulation of the colonic mucosa, goblet cells secrete other mediators that may influence or promote UC development. Correspondingly, Muc2-deficient ((-/-)) mice develop spontaneous colitis, concurrent with the dramatic upregulation of the goblet cell mediator, resistin-like molecule-beta (RELM-β). Testing RELM-β's role, we generated Muc2(-/-)/Retnlb(-/-) mice, finding that RELM-β deficiency significantly attenuated colitis development and symptoms compared with Muc2(-/-) mice. RELM-β expression in Muc2(-/-) mice strongly induced the production/secretion of the antimicrobial lectin RegIIIβ, that exerted its microbicidal effect predominantly on Gram-positive Lactobacillus species. Compared with Muc2(-/-)/Retnlb(-/-) mice, this worsened intestinal microbial dysbiosis with a selective loss of colonic Lactobacilli spp. in Muc2(-/-) mice. Orally replenishing Muc2(-/-) mice with murine Lactobacillus spp., but not with a probiotic formulation containing several human Lactobacillus spp. (VSL#3), ameliorated their spontaneous colitis in concert with increased production of short-chain fatty acids. These studies demonstrate that the goblet cell mediator RELM-β drives colitis in Muc2(-/-) mice by depleting protective commensal microbes. The ability of selective commensal microbial replacement to ameliorate colitis suggests that personalized bacterial therapy may prove beneficial for treatment of UC.
Collapse
|
170
|
Zhang W, Liu J, Shi H, Yang K, Wang P, Wang G, Liu N, Wang H, Ji J, Chu PK. Communication between nitric oxide synthase and positively-charged surface and bone formation promotion. Colloids Surf B Biointerfaces 2016; 148:354-362. [PMID: 27619187 DOI: 10.1016/j.colsurfb.2016.08.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/30/2016] [Accepted: 08/27/2016] [Indexed: 12/11/2022]
Abstract
Despite the effects on physiology of bone marrow mesenchymal stem cells (BMSCs) and bone tissue, biological signal communication between bone implants and them is seldom employed as a guidance to create an osteo-inductive interface. Herein, the positively-charged surface is constructed on bone implant from the perspective of mediation of nitric oxide synthase (NOS) expression to signal BMSCs osteo-differentiation. In vitro and in vivo results indicate that the proper surface potential on the positively-charged surface affects NOS to express a high level of inducible nitric oxide synthase (iNOS) in three NOS isoforms of the contacted BMSCs, upregulates their osteogenetic expression, and ultimately foster new bone growth. However, an excessively high surface potential produces substantial immunomodulatory effects thereby offsetting the aforementioned advantages. This study demonstrates that fine-tuning of the positively-charged surface and proper utilization of the communication between NOS and bone implants promote bone formation.
Collapse
Affiliation(s)
- Wei Zhang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Jun Liu
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Haigang Shi
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Kun Yang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Pingli Wang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Gexia Wang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Na Liu
- Stomatology Department of the General Hospital of Chinese PLA, 28 FuXing Road, Beijing 100853, China
| | - Huaiyu Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Junhui Ji
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Paul K Chu
- Department of Physics & Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| |
Collapse
|
171
|
Fischbach MA, Segre JA. Signaling in Host-Associated Microbial Communities. Cell 2016; 164:1288-1300. [PMID: 26967294 DOI: 10.1016/j.cell.2016.02.037] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Indexed: 12/14/2022]
Abstract
Human-associated microbiota form and stabilize communities based on interspecies interactions. We review how these microbe-microbe and microbe-host interactions are communicated to shape communities over a human's lifespan, including periods of health and disease. Modeling and dissecting signaling in host-associated communities is crucial to understand their function and will open the door to therapies that prevent or correct microbial community dysfunction to promote health and treat disease.
Collapse
Affiliation(s)
- Michael A Fischbach
- Department of Bioengineering and Therapeutic Sciences and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Julia A Segre
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
172
|
Kitayama Y, Fukui H, Hara K, Eda H, Kodani M, Yang M, Sun C, Yamagishi H, Tomita T, Oshima T, Watari J, Takasawa S, Miwa H. Role of regenerating gene I in claudin expression and barrier function in the small intestine. Transl Res 2016; 173:92-100. [PMID: 27055226 DOI: 10.1016/j.trsl.2016.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 02/17/2016] [Accepted: 03/12/2016] [Indexed: 01/01/2023]
Abstract
We have recently shown that loss of the regenerating gene (Reg) I causes susceptibility to nonsteroidal anti-inflammatory drug-induced gastrointestinal damage. However, the mechanism by which Reg I plays a protective role against this pathophysiological condition is unclear. Here, we investigated whether Reg I plays roles in the induction of tight junction proteins and mucosal barrier function in the small intestine. The small-intestinal permeability was evaluated in Reg I-deficient mice by FITC-dextran and transepithelial electrical resistance (TEER) assay. The effect of REG Iα on TEER, claudins expression, and intracellular signaling was examined using Caco2 cells in vitro. Small-intestinal expression of claudins 3 and 4 was investigated in Reg I-deficient mice in vivo. REG I deficiency significantly decreased the expression of claudin 3 in the small-intestinal epithelium. When mice were treated with indomethacin, the serum level of FITC-dextran in Reg I knockout mice was significantly higher than that in wild-type (WT) mice. The level of small-intestinal TEER was significantly decreased in Reg I knockout mice compared with WT mice under normal condition. REG Iα stimulation significantly enhanced the level of TEER in Caco2 cells. Treatment with REG Iα enhanced the expression of claudins 3 and 4 and promoted Sp1, Akt, and ERK phosphorylation in Caco2 cells, whereas these effects were attenuated by treatment with anti-REG Iα antibody. Reg I may play a role in the maintenance of mucosal barrier function by inducing tight junction proteins such as claudins 3 and 4.
Collapse
Affiliation(s)
- Yoshitaka Kitayama
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hirokazu Fukui
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan.
| | - Ken Hara
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hirotsugu Eda
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Mio Kodani
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Mo Yang
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan; Department of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Sun
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan; Department of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Hidetsugu Yamagishi
- Department of Surgical and Molecular Pathology, Dokkyo University School of Medicine, Tochigi, Japan
| | - Toshihiko Tomita
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tadayuki Oshima
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Jiro Watari
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, Kashihara, Japan
| | - Hiroto Miwa
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
173
|
Argudo D, Bethel NP, Marcoline FV, Grabe M. Continuum descriptions of membranes and their interaction with proteins: Towards chemically accurate models. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1858:1619-34. [PMID: 26853937 PMCID: PMC4877259 DOI: 10.1016/j.bbamem.2016.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 01/30/2016] [Accepted: 02/01/2016] [Indexed: 01/21/2023]
Abstract
Biological membranes deform in response to resident proteins leading to a coupling between membrane shape and protein localization. Additionally, the membrane influences the function of membrane proteins. Here we review contributions to this field from continuum elastic membrane models focusing on the class of models that couple the protein to the membrane. While it has been argued that continuum models cannot reproduce the distortions observed in fully-atomistic molecular dynamics simulations, we suggest that this failure can be overcome by using chemically accurate representations of the protein. We outline our recent advances along these lines with our hybrid continuum-atomistic model, and we show the model is in excellent agreement with fully-atomistic simulations of the nhTMEM16 lipid scramblase. We believe that the speed and accuracy of continuum-atomistic methodologies will make it possible to simulate large scale, slow biological processes, such as membrane morphological changes, that are currently beyond the scope of other computational approaches. This article is part of a Special Issue entitled: Membrane Proteins edited by J.C. Gumbart and Sergei Noskov.
Collapse
Affiliation(s)
- David Argudo
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States
| | - Neville P Bethel
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States
| | - Frank V Marcoline
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States
| | - Michael Grabe
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, United States.
| |
Collapse
|
174
|
Malanovic N, Lohner K. Gram-positive bacterial cell envelopes: The impact on the activity of antimicrobial peptides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:936-46. [DOI: 10.1016/j.bbamem.2015.11.004] [Citation(s) in RCA: 249] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 12/21/2022]
|
175
|
Maeda H, Miyata T, Kusakisako K, Galay RL, Talactac MR, Umemiya-Shirafuji R, Mochizuki M, Fujisaki K, Tanaka T. A novel C-type lectin with triple carbohydrate recognition domains has critical roles for the hard tick Haemaphysalis longicornis against Gram-negative bacteria. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 57:38-47. [PMID: 26724379 DOI: 10.1016/j.dci.2015.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/17/2015] [Accepted: 12/17/2015] [Indexed: 06/05/2023]
Abstract
C-type lectins (CLecs) play an important role in innate immunity against invaders. In this study, a novel CLec was identified from Haemaphysalis longicornis ticks (HlCLec). HlCLec contains a signal peptide and a transmembrane region. Interestingly, HlCLec possesses three dissimilar carbohydrate recognition domains (CRDs). Each CRD contains the mutated motif of Ca(2+)-binding site 2. HlCLec mRNA was up-regulated during blood feeding, and had highest expression in the midgut and ovary. HlCLec localization was also confirmed by immunofluorescent antibody test (IFAT). HlCLec was found on the cell membrane and basal lamina of midgut and ovary. In addition, the recombinant HlCLec and individual CRDs demonstrated direct binding activity to Escherichia coli and Staphylococcus aureus; however, no growth inhibition activity was observed. Furthermore, E. coli injection after silencing of HlCLec caused drastic reduction in survival rate of ticks. These results strongly suggest the key role of HlCLec in tick innate immunity against Gram-negative bacteria.
Collapse
Affiliation(s)
- Hiroki Maeda
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan
| | - Takeshi Miyata
- Division of Molecular Functions of Food, Department of Biochemistry and Biotechnology, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan
| | - Kodai Kusakisako
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan
| | - Remil Linggatong Galay
- Department of Veterinary Paraclinical Sciences, College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños, Laguna 4031, Philippines
| | - Melbourne Rio Talactac
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan
| | - Rika Umemiya-Shirafuji
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| | - Masami Mochizuki
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan
| | - Kozo Fujisaki
- Zen-noh Institute of Animal Health, Ooja, Sakura, Chiba 285-0043, Japan
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima 890-0065, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan.
| |
Collapse
|
176
|
Ramanan D, Cadwell K. Intrinsic Defense Mechanisms of the Intestinal Epithelium. Cell Host Microbe 2016; 19:434-41. [PMID: 27049583 DOI: 10.1016/j.chom.2016.03.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The intestinal epithelium is a single cell layer that facilitates the absorption of nutrients but also provides a tight barrier to prevent pathogen invasion and dissemination of commensal microbes. Specialized epithelial cells of the gastrointestinal tract achieve this frontline defense by working in concert with lymphoid, myeloid, and stromal cells to secrete an array of factors that limit direct contact between the epithelium and infectious agents. The importance of these mechanisms is underscored by the ability of enteric pathogens to target these mechanisms to achieve invasion and dissemination. This review highlights recent advances in our understanding of these intricate molecular and cellular mechanisms adopted by these cells to promote spatial segregation and barrier maintenance.
Collapse
Affiliation(s)
- Deepshika Ramanan
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Sackler Institute of Graduate Biomedical Sciences, New York University School of Medicine, New York, NY 10016, USA
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
177
|
Lypd8 promotes the segregation of flagellated microbiota and colonic epithelia. Nature 2016; 532:117-21. [PMID: 27027293 DOI: 10.1038/nature17406] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 02/05/2016] [Indexed: 12/26/2022]
Abstract
Colonic epithelial cells are covered by thick inner and outer mucus layers. The inner mucus layer is free of commensal microbiota, which contributes to the maintenance of gut homeostasis. In the small intestine, molecules critical for prevention of bacterial invasion into epithelia such as Paneth-cell-derived anti-microbial peptides and regenerating islet-derived 3 (RegIII) family proteins have been identified. Although there are mucus layers providing physical barriers against the large number of microbiota present in the large intestine, the mechanisms that separate bacteria and colonic epithelia are not fully elucidated. Here we show that Ly6/PLAUR domain containing 8 (Lypd8) protein prevents flagellated microbiota invading the colonic epithelia in mice. Lypd8, selectively expressed in epithelial cells at the uppermost layer of the large intestinal gland, was secreted into the lumen and bound flagellated bacteria including Proteus mirabilis. In the absence of Lypd8, bacteria were present in the inner mucus layer and many flagellated bacteria invaded epithelia. Lypd8(-/-) mice were highly sensitive to intestinal inflammation induced by dextran sulfate sodium (DSS). Antibiotic elimination of Gram-negative flagellated bacteria restored the bacterial-free state of the inner mucus layer and ameliorated DSS-induced intestinal inflammation in Lypd8(-/-) mice. Lypd8 bound to flagella and suppressed motility of flagellated bacteria. Thus, Lypd8 mediates segregation of intestinal bacteria and epithelial cells in the colon to preserve intestinal homeostasis.
Collapse
|
178
|
Schwartzman JA, Ruby EG. Stress as a Normal Cue in the Symbiotic Environment. Trends Microbiol 2016; 24:414-424. [PMID: 27004825 DOI: 10.1016/j.tim.2016.02.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/19/2016] [Accepted: 02/22/2016] [Indexed: 02/06/2023]
Abstract
All multicellular hosts form associations with groups of microorganisms. These microbial communities can be taxonomically diverse and dynamic, and their persistence is due to robust, and sometimes coevolved, host-microbe and microbe-microbe interactions. Chemical and physical sources of stress are prominently situated in this molecular exchange, as cues for cellular responses in symbiotic microbes. Stress in the symbiotic environment may arise from three sources: host tissues, microbe-induced immune responses, or other microbes in the host environment. The responses of microbes to these stresses can be general or highly specialized, and collectively may contribute to the stability of the symbiotic system. In this review, we highlight recent work that emphasizes the role of stress as a cue in the symbiotic environment of plants and animals.
Collapse
Affiliation(s)
- Julia A Schwartzman
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Edward G Ruby
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, WI, USA; Kewalo Marine Laboratory, University of Hawaii, Manoa, Honolulu, HI, USA
| |
Collapse
|
179
|
Zhang N, He QS. Commensal Microbiome Promotes Resistance to Local and Systemic Infections. Chin Med J (Engl) 2016; 128:2250-5. [PMID: 26265621 PMCID: PMC4717980 DOI: 10.4103/0366-6999.162502] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objective: In this review, to illustrate the resistance mechanism for pathogen insult, we discussed the role of the intestinal microbiome in promoting resistance to local gastrointestinal tract infections and to respiratory tract infections. Data Sources: The review was based on data obtained from the published research articles. Study Selection: A total of 49 original articles were selected in accordance with our main objective to illustrate the resistance mechanism(s) by which commensal microbiota can contribute to host defense against local and systemic infections. Results: Diverse microorganisms colonize human environmentally exposed surfaces such as skin, respiratory tract, and gastrointestinal tract. Co-evolution has resulted in these microbes with extensive and diverse impacts on multiple aspects of host biological functions. During the last decade, high-throughput sequencing technology developed has been applied to study commensal microbiota and their impact on host biological functions. By using pathogen recognition receptors pathway and nucleotide binding oligomerization domain-like receptors pathway, the commensal microbiome promotes resistance to local and systemic infections, respectively. To protect against the local infections, the microbiome functions contain the following: The competing for sites of colonization, direct production of inhibition molecules or depletion of nutrients needed for pathogens, and priming immune defenses against pathogen insult. At the same time, with the purpose to maintain homeostasis, the commensal bacteria can program systemic signals toward not only local tissue but also distal tissue to modify their function for infections accordingly. Conclusions: Commensal bacteria play an essential role in protecting against infections, shaping and regulating immune responses, and maintaining host immune homeostasis.
Collapse
Affiliation(s)
| | - Qiu-Shui He
- Department of Medical Microbiology and Research Centre of Microbiome, Capital Medical University, Beijing 100069, China; Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland,
| |
Collapse
|
180
|
Longman RS, Littman DR. The functional impact of the intestinal microbiome on mucosal immunity and systemic autoimmunity. Curr Opin Rheumatol 2016; 27:381-7. [PMID: 26002030 DOI: 10.1097/bor.0000000000000190] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review will highlight recent advances functionally linking the gut microbiome with mucosal and systemic immune cell activation underlying autoimmunity. RECENT FINDINGS Dynamic interactions between the gut microbiome and environmental cues (including diet and medicines) shape the effector potential of the microbial organ. Key bacteria and viruses have emerged that, in defined microenvironments, play a critical role in regulating effector lymphocyte functions. The coordinated interactions between these different microbial kingdoms - including bacteria, helminths, and viruses (termed transkingdom interactions) - play a key role in shaping immunity. Emerging strategies to identify immunologically relevant microbes with the potential to regulate immune cell functions both at mucosal sites and systemically will likely define diagnostic and therapeutic targets. SUMMARY The microbiome constitutes a critical microbial organ with coordinated interactions that shape host immunity.
Collapse
Affiliation(s)
- Randy S Longman
- Department of Medicine, Jill Roberts Institute for IBD Research, Weill Cornell Medical College, New York, New York, USA
| | | |
Collapse
|
181
|
Xie Y, Yang L. Calcium and Magnesium Ions Are Membrane-Active against Stationary-Phase Staphylococcus aureus with High Specificity. Sci Rep 2016; 6:20628. [PMID: 26865182 PMCID: PMC4749956 DOI: 10.1038/srep20628] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/06/2016] [Indexed: 11/09/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is notorious for its ability to acquire antibiotic-resistance, and antibiotic-resistant S. aureus has become a wide-spread cause of high mortality rate. Novel antimicrobials capable of eradicating S. aureus cells including antibiotic-resistant ones are thus highly desired. Membrane-active bactericides and species-specific antimicrobials are two promising sources of novel anti-infective agents for fighting against bacterial antibiotic-resistance. We herein show that Ca2+ and Mg2+, two alkaline-earth-metal ions physiologically essential for diverse living organisms, both disrupt model S. aureus membranes and kill stationary-phase S. aureus cells, indicative of membrane-activity. In contrast to S. aureus, Escherichia coli and Bacillus subtilis exhibit unaffected survival after similar treatment with these two cations, indicative of species-specific activity against S. aureus. Moreover, neither Ca2+ nor Mg2+ lyses mouse red blood cells, indicative of hemo-compatibility. This works suggests that Ca2+ and Mg2+ may have implications in targeted eradication of S. aureus pathogen including the antibiotic-resistant ones.
Collapse
Affiliation(s)
- Yuntao Xie
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui 230026 China.,Department of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026 China
| | - Lihua Yang
- CAS Key Laboratory of Soft Matter Chemistry, University of Science and Technology of China, Hefei, Anhui 230026 China.,Department of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026 China.,Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026 China
| |
Collapse
|
182
|
Okita Y, Shiono T, Yahagi A, Hamada S, Umemura M, Matsuzaki G. Interleukin-22-Induced Antimicrobial Phospholipase A2 Group IIA Mediates Protective Innate Immunity of Nonhematopoietic Cells against Listeria monocytogenes. Infect Immun 2016; 84:573-9. [PMID: 26644377 PMCID: PMC4730562 DOI: 10.1128/iai.01000-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/30/2015] [Indexed: 01/07/2023] Open
Abstract
Listeria monocytogenes is a bacterial pathogen which establishes intracellular parasitism in various cells, including macrophages and nonhematopoietic cells, such as hepatocytes. It has been reported that several proinflammatory cytokines have pivotal roles in innate protection against L. monocytogenes infection. We found that a proinflammatory cytokine, interleukin 22 (IL-22), was expressed by CD3(+) CD4(+) T cells at an early stage of L. monocytogenes infection in mice. To assess the influence of IL-22 on L. monocytogenes infection in hepatocytes, cells of a human hepatocellular carcinoma line, HepG2, were treated with IL-22 before L. monocytogenes infection in vitro. Gene expression analysis of the IL-22-treated HepG2 cells identified phospholipase A2 group IIA (PLA2G2A) as an upregulated antimicrobial molecule. Addition of recombinant PLA2G2A to the HepG2 culture significantly suppressed L. monocytogenes infection. Culture supernatant of the IL-22-treated HepG2 cells contained bactericidal activity against L. monocytogenes, and the activity was abrogated by a specific PLA2G2A inhibitor, demonstrating that HepG2 cells secreted PLA2G2A, which killed extracellular L. monocytogenes. Furthermore, colocalization of PLA2G2A and L. monocytogenes was detected in the IL-22-treated infected HepG2 cells, which suggests involvement of PLA2G2A in the mechanism of intracellular killing of L. monocytogenes by HepG2 cells. These results suggest that IL-22 induced at an early stage of L. monocytogenes infection enhances innate immunity against L. monocytogenes in the liver by stimulating hepatocytes to produce an antimicrobial molecule, PLA2G2A.
Collapse
Affiliation(s)
- Yamato Okita
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Takeru Shiono
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Ayano Yahagi
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Satoru Hamada
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Masayuki Umemura
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Goro Matsuzaki
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
183
|
Cao S, Su X, Zeng B, Yan H, Huang Y, Wang E, Yun H, Zhang Y, Liu F, Li W, Wei H, Che Y, Yang R. The Gut Epithelial Receptor LRRC19 Promotes the Recruitment of Immune Cells and Gut Inflammation. Cell Rep 2016; 14:695-707. [PMID: 26776522 PMCID: PMC4742566 DOI: 10.1016/j.celrep.2015.12.070] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/02/2015] [Accepted: 12/14/2015] [Indexed: 12/17/2022] Open
Abstract
Commensal microbes are necessary for a healthy gut immune system. However, the mechanism involving these microbes that establish and maintain gut immune responses is largely unknown. Here, we have found that the gut immune receptor leucine-rich repeat (LRR) C19 is involved in host-microbiota interactions. LRRC19 deficiency not only impairs the gut immune system but also reduces inflammatory responses in gut tissues. We demonstrate that the LRRC19-associated chemokines CCL6, CCL9, CXCL9, and CXCL10 play a critical role in immune cell recruitment and intestinal inflammation. The expression of these chemokines is associated with regenerating islet-derived (REG) protein-mediated microbiotas. We also found that the expression of REGs may be regulated by gut Lactobacillus through LRRC19-mediated activation of NF-κB. Therefore, our study establishes a regulatory axis of LRRC19, REGs, altered microbiotas, and chemokines for the recruitment of immune cells and the regulation of intestinal inflammation. The gut immune receptor LRRC19 is involved in host-microbiota interactions LRRC19-associated chemokines control immune cell recruitment and gut inflammation Chemokines are regulated by REG protein-mediated gut microbiotas Lactobacillus may modulate the expression of REG proteins through LRRC19
Collapse
Affiliation(s)
- Shuisong Cao
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaomin Su
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Benhua Zeng
- Department of Laboratory Animal Science, College of Basic Medicine Science, Third Military Medical University, Chongqing 404100, China
| | - Hui Yan
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Yugang Huang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Enlin Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Huan Yun
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Yuan Zhang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Feifei Liu
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Wenxia Li
- Department of Laboratory Animal Science, College of Basic Medicine Science, Third Military Medical University, Chongqing 404100, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medicine Science, Third Military Medical University, Chongqing 404100, China
| | - Yongzhe Che
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
184
|
OKUMURA R, TAKEDA K. Maintenance of gut homeostasis by the mucosal immune system. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2016; 92:423-435. [PMID: 27840390 PMCID: PMC5328791 DOI: 10.2183/pjab.92.423] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Inflammatory bowel diseases (IBD) are represented by ulcerative colitis (UC) and Crohn's disease (CD), both of which involve chronic intestinal inflammation. Recent evidence has indicated that gut immunological homeostasis is maintained by the interaction between host immunity and intestinal microbiota. A variety of innate immune cells promote or suppress T cell differentiation and activation in response to intestinal bacteria or their metabolites. Some commensal bacteria species or bacterial metabolites enhance or repress host immunity by inducing T helper (Th) 17 cells or regulatory T cells. Intestinal epithelial cells between host immune cells and intestinal microbiota contribute to the separation of these populations and modulate host immune responses to intestinal microbiota. Therefore, the imbalance between host immunity and intestinal microbiota caused by host genetic predisposition or abnormal environmental factors promote susceptibility to intestinal inflammation.
Collapse
Affiliation(s)
- Ryu OKUMURA
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Kiyoshi TAKEDA
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Correspondence should be addressed: K. Takeda, Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan (e-mail: )
| |
Collapse
|
185
|
Jia N, Liu N, Cheng W, Jiang YL, Sun H, Chen LL, Peng J, Zhang Y, Ding YH, Zhang ZH, Wang X, Cai G, Wang J, Dong MQ, Zhang Z, Wu H, Wang HW, Chen Y, Zhou CZ. Structural basis for receptor recognition and pore formation of a zebrafish aerolysin-like protein. EMBO Rep 2015; 17:235-48. [PMID: 26711430 DOI: 10.15252/embr.201540851] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/24/2015] [Indexed: 12/31/2022] Open
Abstract
Various aerolysin-like pore-forming proteins have been identified from bacteria to vertebrates. However, the mechanism of receptor recognition and/or pore formation of the eukaryotic members remains unknown. Here, we present the first crystal and electron microscopy structures of a vertebrate aerolysin-like protein from Danio rerio, termed Dln1, before and after pore formation. Each subunit of Dln1 dimer comprises a β-prism lectin module followed by an aerolysin module. Specific binding of the lectin module toward high-mannose glycans triggers drastic conformational changes of the aerolysin module in a pH-dependent manner, ultimately resulting in the formation of a membrane-bound octameric pore. Structural analyses combined with computational simulations and biochemical assays suggest a pore-forming process with an activation mechanism distinct from the previously characterized bacterial members. Moreover, Dln1 and its homologs are ubiquitously distributed in bony fishes and lamprey, suggesting a novel fish-specific defense molecule.
Collapse
Affiliation(s)
- Ning Jia
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Nan Liu
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Joint Center for Life Sciences, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Wang Cheng
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yong-Liang Jiang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Hui Sun
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Lan-Lan Chen
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Junhui Peng
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yonghui Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yue-He Ding
- National Institute of Biological Sciences, Beijing, China
| | - Zhi-Hui Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xuejuan Wang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Gang Cai
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Junfeng Wang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Meng-Qiu Dong
- National Institute of Biological Sciences, Beijing, China
| | - Zhiyong Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Hui Wu
- Departments of Microbiology and Pediatric Dentistry, Schools of Dentistry and Medicine University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hong-Wei Wang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Joint Center for Life Sciences, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuxing Chen
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Cong-Zhao Zhou
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, China
| |
Collapse
|
186
|
Spencer JD, Jackson AR, Li B, Ching CB, Vonau M, Easterling RS, Schwaderer AL, McHugh KM, Becknell B. Expression and Significance of the HIP/PAP and RegIIIγ Antimicrobial Peptides during Mammalian Urinary Tract Infection. PLoS One 2015; 10:e0144024. [PMID: 26658437 PMCID: PMC4675559 DOI: 10.1371/journal.pone.0144024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/11/2015] [Indexed: 12/22/2022] Open
Abstract
Recent evidence indicates that antimicrobial peptides (AMPs) serve key roles in defending the urinary tract against invading uropathogens. To date, the individual contribution of AMPs to urinary tract host defense is not well defined. In this study, we identified Regenerating islet-derived 3 gamma (RegIIIγ) as the most transcriptionally up-regulated AMP in murine bladder transcriptomes following uropathogenic Escherichia coli (UPEC) infection. We confirmed induction of RegIIIγ mRNA during cystitis and pyelonephritis by quantitative RT-PCR. Immunoblotting demonstrates increased bladder and urinary RegIIIγ protein levels following UPEC infection. Immunostaining localizes RegIIIγ protein to urothelial cells of infected bladders and kidneys. Human patients with UTI have increased urine concentrations of the orthologous Hepatocarcinoma-Intestine-Pancreas / Pancreatitis Associated Protein (HIP/PAP) compared to healthy controls. Recombinant RegIIIγ protein does not demonstrate bactericidal activity toward UPEC in vitro, but does kill Staphylococcus saprophyticus in a dose-dependent manner. Kidney and bladder tissue from RegIIIγ knockout mice and wild-type mice contain comparable bacterial burden following UPEC and Gram-positive UTI. Our results demonstrate that RegIIIγ and HIP/PAP expression is induced during human and murine UTI. However, their specific function in the urinary tract remains uncertain.
Collapse
Affiliation(s)
- John David Spencer
- Division of Nephrology, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- Center for Clinical and Translational Research, Research Institute at Nationwide Children’s, Columbus, Ohio, United States of America
| | - Ashley R. Jackson
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Birong Li
- Center for Clinical and Translational Research, Research Institute at Nationwide Children’s, Columbus, Ohio, United States of America
| | - Christina B. Ching
- Division of Urology, Department of Surgery, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Martin Vonau
- Department of Pediatrics and Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | | | - Andrew L. Schwaderer
- Division of Nephrology, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- Center for Clinical and Translational Research, Research Institute at Nationwide Children’s, Columbus, Ohio, United States of America
| | - Kirk M. McHugh
- Department of Anatomy, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- Center for Molecular and Human Genetics, Research Institute at Nationwide Children’s, Columbus, Ohio, United States of America
| | - Brian Becknell
- Division of Nephrology, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- Center for Clinical and Translational Research, Research Institute at Nationwide Children’s, Columbus, Ohio, United States of America
| |
Collapse
|
187
|
Peraro MD, van der Goot FG. Pore-forming toxins: ancient, but never really out of fashion. Nat Rev Microbiol 2015; 14:77-92. [DOI: 10.1038/nrmicro.2015.3] [Citation(s) in RCA: 476] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
188
|
Hatakeyama T, Higashi E, Nakagawa H. cDNA cloning and expression of Contractin A, a phospholipase A2-like protein from the globiferous pedicellariae of the venomous sea urchin Toxopneustes pileolus. Toxicon 2015; 108:46-52. [DOI: 10.1016/j.toxicon.2015.09.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/21/2015] [Accepted: 09/29/2015] [Indexed: 11/25/2022]
|
189
|
Expression of HIP/PAP in hepatocellular carcinoma and effect of siRNA on migration and invasion in HCC cells. ASIAN PAC J TROP MED 2015; 8:848-54. [DOI: 10.1016/j.apjtm.2015.09.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/20/2015] [Accepted: 09/15/2015] [Indexed: 11/18/2022] Open
|
190
|
Nagae M, Yamaguchi Y. Sugar recognition and protein-protein interaction of mammalian lectins conferring diverse functions. Curr Opin Struct Biol 2015; 34:108-15. [PMID: 26418728 DOI: 10.1016/j.sbi.2015.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 08/06/2015] [Accepted: 08/10/2015] [Indexed: 11/24/2022]
Abstract
Recent advances in structural analyses of mammalian lectins reveal atomic-level details of their fine specificities toward diverse endogenous and exogenous glycans. Local variations on a common scaffold can enable certain lectins to recognize complex carbohydrate ligands including branched glycans and O-glycosylated peptides. Simultaneous recognition of both glycan and the aglycon moieties enhances the affinity and specificity of lectins such as CLEC-2 and PILRα. Attention has been paid to the roles of galectin and RegIII family of proteins in protein-protein interactions involved in critical biological functions including signal transduction and bactericidal pore formation.
Collapse
Affiliation(s)
- Masamichi Nagae
- Structural Glycobiology Team, Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN Global Research Cluster, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshiki Yamaguchi
- Structural Glycobiology Team, Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, RIKEN Global Research Cluster, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
191
|
Moniruzzaman M, Alam JM, Dohra H, Yamazaki M. Antimicrobial Peptide Lactoferricin B-Induced Rapid Leakage of Internal Contents from Single Giant Unilamellar Vesicles. Biochemistry 2015; 54:5802-14. [DOI: 10.1021/acs.biochem.5b00594] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Md. Moniruzzaman
- Integrated Bioscience Section, Graduate School of
Science and Technology, ‡Nanomaterials Research
Division, Research Institute of Electronics, §Instrumental Research Support Office,
Research Institute of Green Science and Technology, and ∥Department of Physics, Faculty of
Science, Shizuoka University, Shizuoka 422-8529, Japan
| | - Jahangir Md. Alam
- Integrated Bioscience Section, Graduate School of
Science and Technology, ‡Nanomaterials Research
Division, Research Institute of Electronics, §Instrumental Research Support Office,
Research Institute of Green Science and Technology, and ∥Department of Physics, Faculty of
Science, Shizuoka University, Shizuoka 422-8529, Japan
| | - Hideo Dohra
- Integrated Bioscience Section, Graduate School of
Science and Technology, ‡Nanomaterials Research
Division, Research Institute of Electronics, §Instrumental Research Support Office,
Research Institute of Green Science and Technology, and ∥Department of Physics, Faculty of
Science, Shizuoka University, Shizuoka 422-8529, Japan
| | - Masahito Yamazaki
- Integrated Bioscience Section, Graduate School of
Science and Technology, ‡Nanomaterials Research
Division, Research Institute of Electronics, §Instrumental Research Support Office,
Research Institute of Green Science and Technology, and ∥Department of Physics, Faculty of
Science, Shizuoka University, Shizuoka 422-8529, Japan
| |
Collapse
|
192
|
|
193
|
Mortzfeld BM, Urbanski S, Reitzel AM, Künzel S, Technau U, Fraune S. Response of bacterial colonization inNematostella vectensisto development, environment and biogeography. Environ Microbiol 2015; 18:1764-81. [DOI: 10.1111/1462-2920.12926] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Benedikt M. Mortzfeld
- Zoological Institute; Christian-Albrechts University Kiel; Olshausenstrasse 40 Kiel 24098 Germany
| | - Szymon Urbanski
- Zoological Institute; Christian-Albrechts University Kiel; Olshausenstrasse 40 Kiel 24098 Germany
| | - Adam M. Reitzel
- Department of Biological Sciences; The University of North Carolina at Charlotte; Woodward Hall 245 Charlotte NC 28223 USA
| | - Sven Künzel
- Max-Planck Institute for Evolutionary Biology; Plön 24306 Germany
| | - Ulrich Technau
- Department of Molecular Evolution and Development, Centre for Organismal Systems Biology, Faculty of Life Sciences; University of Vienna; Althanstrasse 14 Wien 1090 Austria
| | - Sebastian Fraune
- Zoological Institute; Christian-Albrechts University Kiel; Olshausenstrasse 40 Kiel 24098 Germany
| |
Collapse
|
194
|
Soler L, Miller I, Nöbauer K, Carpentier S, Niewold T. Identification of the major regenerative III protein (RegIII) in the porcine intestinal mucosa as RegIIIγ, not RegIIIα. Vet Immunol Immunopathol 2015; 167:51-6. [PMID: 26187439 DOI: 10.1016/j.vetimm.2015.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 07/01/2015] [Accepted: 07/03/2015] [Indexed: 12/13/2022]
Abstract
During the last years, an antimicrobial protein from the RegIII family has been consistently identified as one of the main up-regulated mRNA transcripts in the pig small intestinal mucosa during different infections such as enterotoxigenic Escherichia coli (ETEC). This transcript has been mainly referred to in the literature as pancreatitis-associated protein (PAP/RegIIIα). However, the identity of this transcript has not been confirmed, and no evidence of its expression at the protein level is available in the literature, because the absence of a specific antibody. In this study, we first unequivocally identified the PAP/RegIII family protein mainly expressed in ETEC infected pig intestine as RegIIIγ by 2D-DIGE and MALDI-TOF/TOF. This shows that the pig differs from species like human and mice in that RegIIIγ (and not RegIIIα) might be the major RegIII isotype during intestinal infection. Immunoblotting analysis with a specifically generated polyclonal rabbit antibody revealed that pig RegIIIγ is expressed throughout the intestinal tract, but most abundantly in the ileum. Although a higher abundance of mRNA was paralleled by higher protein abundance, a lack of linear relationship was found between RegIIIγ mRNA and protein abundances in the jejunal mucosa, the latter most pronounced in the case of natural infection. This may be related to the secretory nature of RegIIIγ. This would mean that the antimicrobial protein RegIIIγ is a good candidate as a non-invasive faecal intestinal health biomarker in swine.
Collapse
Affiliation(s)
- L Soler
- Livestock-Nutrition-Quality Division, Biosystems Department, Faculty of Biosciences Engineering, KU Leuven, Kasteelpark Arenberg 30, Heverlee 3001, Belgium
| | - I Miller
- Department of Biomedical Sciences, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria
| | - K Nöbauer
- VetCore, University of Veterinary Medicine, Veterinaerplatz 1, 1210 Vienna, Austria
| | - S Carpentier
- Division of Crop Biotechnics, O&N II Herestraat 49 - Box 901, Leuven 3000, Belgium
| | - T Niewold
- Livestock-Nutrition-Quality Division, Biosystems Department, Faculty of Biosciences Engineering, KU Leuven, Kasteelpark Arenberg 30, Heverlee 3001, Belgium.
| |
Collapse
|
195
|
Yang W, Dierking K, Esser D, Tholey A, Leippe M, Rosenstiel P, Schulenburg H. Overlapping and unique signatures in the proteomic and transcriptomic responses of the nematode Caenorhabditis elegans toward pathogenic Bacillus thuringiensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 51:1-9. [PMID: 25720978 DOI: 10.1016/j.dci.2015.02.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 06/04/2023]
Abstract
Pathogen infection can activate multiple signaling cascades that ultimately alter the abundance of molecules in cells. This change can be measured both at the transcript and protein level. Studies analyzing the immune response at both levels are, however, rare. Here, we compare transcriptome and proteome data generated after infection of the nematode and model organism Caenorhabditis elegans with the Gram-positive pathogen Bacillus thuringiensis. Our analysis revealed a high overlap between abundance changes of corresponding transcripts and gene products, especially for genes encoding C-type lectin domain-containing proteins, indicating their particular role in worm immunity. We additionally identified a unique signature at the proteome level, suggesting that the C. elegans response to infection is shaped by changes beyond transcription. Such effects appear to be influenced by AMP-activated protein kinases (AMPKs), which may thus represent previously unknown regulators of C. elegans immune defense.
Collapse
Affiliation(s)
- Wentao Yang
- Evolutionary Ecology and Genetics, Zoological Institute, Christian-Albrechts University of Kiel, Germany
| | - Katja Dierking
- Evolutionary Ecology and Genetics, Zoological Institute, Christian-Albrechts University of Kiel, Germany
| | - Daniela Esser
- Institute of Clinical Molecular Biology, Christian-Albrechts University of Kiel, Germany
| | - Andreas Tholey
- Systematic Proteome Research and Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-University of Kiel, Germany
| | - Matthias Leippe
- Comparative Immunology, Zoological Institute, Christian-Albrechts University of Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts University of Kiel, Germany
| | - Hinrich Schulenburg
- Evolutionary Ecology and Genetics, Zoological Institute, Christian-Albrechts University of Kiel, Germany.
| |
Collapse
|
196
|
Hu K, Jiang Y, Xie Y, Liu H, Liu R, Zhao Z, Lai R, Yang L. Small-Anion Selective Transmembrane "Holes" Induced by an Antimicrobial Peptide Too Short to Span Membranes. J Phys Chem B 2015; 119:8553-60. [PMID: 26126210 DOI: 10.1021/acs.jpcb.5b03133] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Whereas many membrane-destabilization modes have been suggested for membrane-spanning antimicrobial peptides (AMPs), few are available for those too short to span membrane thickness. Here we show that ORB-1, a 15-residue disulfide-bridged AMP that is only ∼20 Å long even when fully stretched like a hairpin, may act by inducing small anion-selective transmembrane "holes" of negative mean curvature. In model membranes of Gram-negative bacteria, ORB-1 induces chloride transmembrane transport and formation of transmembrane channels of negative mean curvature, whereas the inactive analogue, ORB-N, does not, suggesting a correlation between antibacterial activity and ability to induce transmembrane channels. Given that ORB-N is the C-terminus amidated form of ORB-1, our results further suggest that formation of membrane-spanning dimers may be required to initiate the observed channel induction. Moreover, ORB-1 renders model bacterial membranes permeable to anions with effective hydration diameters of <1 nm (e.g., Cl(-) and NO3(-)), but not cations of similar sizes (e.g., H3O(+)), indicative of anion-selective transmembrane channels with an effective inner diameter of ≤1 nm. In addition, negative-intrinsic-curvature (NIC) lipids such as phosphoethanolamine (PE) may facilitate the membrane-destabilization process of ORB-1. Our findings may expand current understandings on how AMPs destabilize membranes and facilitate the pharmaceutical development of ORB-1.
Collapse
Affiliation(s)
| | | | | | | | - Rui Liu
- ‡Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | | | - Ren Lai
- ‡Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | | |
Collapse
|
197
|
Abstract
Body surfaces are colonized by resident microbes that are remarkably resilient to recurrent immune responses. In the latest issue of Science, Cullen et al. (2015) report that, contrary to prevailing assumptions, bacteria of the colonizing microbiota are resistant to antimicrobial peptides, and identify a common mechanism of resistance.
Collapse
Affiliation(s)
- Charles L Bevins
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA.
| | - Andreas J Bäumler
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
198
|
Hitting the sweet spot-glycans as targets of fungal defense effector proteins. Molecules 2015; 20:8144-67. [PMID: 25955890 PMCID: PMC6272156 DOI: 10.3390/molecules20058144] [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: 04/02/2015] [Revised: 04/29/2015] [Accepted: 04/30/2015] [Indexed: 11/16/2022] Open
Abstract
Organisms which rely solely on innate defense systems must combat a large number of antagonists with a comparably low number of defense effector molecules. As one solution of this problem, these organisms have evolved effector molecules targeting epitopes that are conserved between different antagonists of a specific taxon or, if possible, even of different taxa. In order to restrict the activity of the defense effector molecules to physiologically relevant taxa, these target epitopes should, on the other hand, be taxon-specific and easily accessible. Glycans fulfill all these requirements and are therefore a preferred target of defense effector molecules, in particular defense proteins. Here, we review this defense strategy using the example of the defense system of multicellular (filamentous) fungi against microbial competitors and animal predators.
Collapse
|
199
|
Moniaux N, Darnaud M, Garbin K, Dos Santos A, Guettier C, Samuel D, Amouyal G, Amouyal P, Bréchot C, Faivre J. The Reg3α (HIP/PAP) Lectin Suppresses Extracellular Oxidative Stress in a Murine Model of Acute Liver Failure. PLoS One 2015; 10:e0125584. [PMID: 25938566 PMCID: PMC4418718 DOI: 10.1371/journal.pone.0125584] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/23/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is a rapidly progressive heterogeneous illness with high mortality rate and no widely accessible cure. A promising drug candidate according to previous preclinical studies is the Reg3α (or HIP/PAP) lectin, which alleviates ALF through its free-radical scavenging activity. Here we study the therapeutic targets of Reg3α in order to gain information on the nature of the oxidative stress associated with ALF. METHODS Primary hepatocytes stressed with the reactive oxygen species (ROS) inducers TNFα and H2O2 were incubated with a recombinant Reg3α protein. ALF was induced in C57BL/6J mice by an anti-CD95 antibody. Livers and primary hepatocytes were harvested for deoxycholate separation of cellular and extracellular fractions, immunostaining, immunoprecipitation and malondialdehyde assays. Fibrin deposition was studied by immunofluorescence in frozen liver explants from patients with ALF. RESULTS Fibrin deposition occurs during experimental and clinical acute liver injuries. Reg3α bound the resulting transient fibrin network, accumulated in the inflammatory extracellular matrix (ECM), greatly reduced extracellular ROS levels, and improved cell viability. Hepatocyte treatment with ligands of death receptors, e.g. TNFα and Fas, resulted in a twofold increase of malondialdehyde (MDA) level in the deoxycholate-insoluble fractions. Reg3α treatment maintained MDA at a level similar to control cells and thereby increased hepatocyte survival by 35%. No antioxidant effect of Reg3α was noted in the deoxycholate-soluble fractions. Preventing fibrin network formation with heparin suppressed the prosurvival effect of Reg3α. CONCLUSIONS Reg3α is an ECM-targeted ROS scavenger that binds the fibrin scaffold resulting from hepatocyte death during ALF. ECM alteration is an important pathogenic factor of ALF and a relevant target for pharmacotherapy.
Collapse
Affiliation(s)
- Nicolas Moniaux
- INSERM, U1193, Centre Hépatobiliaire, Villejuif, F-94800, France
- Université Paris-Sud, Faculté de Médecine, Villejuif, F-94800, France
| | - Marion Darnaud
- INSERM, U1193, Centre Hépatobiliaire, Villejuif, F-94800, France
- Université Paris-Sud, Faculté de Médecine, Villejuif, F-94800, France
| | - Kévin Garbin
- INSERM, U1193, Centre Hépatobiliaire, Villejuif, F-94800, France
- Université Paris-Sud, Faculté de Médecine, Villejuif, F-94800, France
| | - Alexandre Dos Santos
- INSERM, U1193, Centre Hépatobiliaire, Villejuif, F-94800, France
- Université Paris-Sud, Faculté de Médecine, Villejuif, F-94800, France
| | - Catherine Guettier
- INSERM, U1193, Centre Hépatobiliaire, Villejuif, F-94800, France
- Université Paris-Sud, Faculté de Médecine, Villejuif, F-94800, France
- Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Universitaire Paul Brousse, Villejuif, F-94800, France
| | - Didier Samuel
- INSERM, U1193, Centre Hépatobiliaire, Villejuif, F-94800, France
- Université Paris-Sud, Faculté de Médecine, Villejuif, F-94800, France
- Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Universitaire Paul Brousse, Villejuif, F-94800, France
| | | | | | - Christian Bréchot
- INSERM, U1193, Centre Hépatobiliaire, Villejuif, F-94800, France
- Université Paris-Sud, Faculté de Médecine, Villejuif, F-94800, France
| | - Jamila Faivre
- INSERM, U1193, Centre Hépatobiliaire, Villejuif, F-94800, France
- Université Paris-Sud, Faculté de Médecine, Villejuif, F-94800, France
- Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Universitaire Paul Brousse, Villejuif, F-94800, France
| |
Collapse
|
200
|
Schroeder BO, Ehmann D, Precht JC, Castillo PA, Küchler R, Berger J, Schaller M, Stange EF, Wehkamp J. Paneth cell α-defensin 6 (HD-6) is an antimicrobial peptide. Mucosal Immunol 2015; 8:661-71. [PMID: 25354318 PMCID: PMC4424388 DOI: 10.1038/mi.2014.100] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 09/21/2014] [Indexed: 02/04/2023]
Abstract
Defensins protect human barriers from commensal and pathogenic microorganisms. Human α-defensin 6 (HD-6) is produced exclusively by small intestinal Paneth cells but, in contrast to other antimicrobial peptides (AMPs) for HD-6, no direct antibacterial killing activity has been detected so far. Herein, we systematically tested how environmental factors, like pH and reducing conditions, affect antimicrobial activity of different defensins against anaerobic bacteria of the human intestinal microbiota. Remarkably, by mimicking the intestinal milieu we detected for the first time antibacterial activity of HD-6. Activity was observed against anaerobic gut commensals but not against some pathogenic strains. Antibiotic activity was attributable to the reduced peptide and independent of free cysteines or a conserved histidine residue. Furthermore, the oxidoreductase thioredoxin, which is also expressed in Paneth cells, is able to reduce a truncated physiological variant of HD-6. Ultrastructural analyses revealed that reduced HD-6 causes disintegration of cytoplasmic structures and alterations in the bacterial cell envelope, while maintaining extracellular net-like structures. We conclude that HD-6 is an antimicrobial peptide. Our data suggest two distinct antimicrobial mechanisms by one peptide: HD-6 kills specific microbes depending on the local environmental conditions, whereas known microbial trapping by extracellular net structures is independent of the reducing milieu.
Collapse
Affiliation(s)
- B O Schroeder
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany,University of Tuebingen, Tuebingen, Germany,Department of Microbiology and Immunology, School of Medicine, University of California, Davis, California, USA
| | - D Ehmann
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany,University of Tuebingen, Tuebingen, Germany
| | - J C Precht
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany,University of Tuebingen, Tuebingen, Germany
| | - P A Castillo
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, California, USA
| | - R Küchler
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany,University of Tuebingen, Tuebingen, Germany
| | - J Berger
- Max-Planck-Institute for Developmental Biology, Electron Microscopy Unit, Tuebingen, Germany
| | - M Schaller
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - E F Stange
- Department of Internal Medicine I, Robert Bosch Hospital, Stuttgart, Germany
| | - J Wehkamp
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany,University of Tuebingen, Tuebingen, Germany,Department of Internal Medicine I, Robert Bosch Hospital, Stuttgart, Germany,Department of Internal Medicine I, University Hospital Tuebingen, Tuebingen, Germany,()
| |
Collapse
|