1
|
Lima JC, Brito RMDM, Pereira LC, Pereira NDS, Nascimento MSL, de Melo AL, Guedes PMM. Innate immune receptors are differentially expressed in mice during experimental Schistosoma mansoni early infection. Mem Inst Oswaldo Cruz 2024; 119:e240013. [PMID: 38896633 PMCID: PMC11182339 DOI: 10.1590/0074-02760240013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/09/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND The impact of Schistosoma mansoni infection over the immune response and the mechanisms involved in pathogenesis are not yet completely understood. OBJECTIVES This study aimed to evaluate the expression of innate immune receptors in three distinct mouse lineages (BALB/c, C57BL/6 and Swiss) during experimental S. mansoni infection with LE strain. METHODS The parasite burden, intestinal tissue oogram and presence of hepatic granulomas were evaluated at 7- and 12-weeks post infection (wpi). The mRNA expression for innate Toll-like receptors, Nod-like receptors, their adaptor molecules, and cytokines were determined at 2, 7 and 12 wpi in the hepatic tissue by real-time quantitative polymerase chain reaction (qPCR). FINDINGS Swiss mice showed 100% of survival, had lower parasite burden and intestinal eggs, while infected BALB/c and C57BL/6 presented 80% and 90% of survival, respectively, higher parasite burden and intestinal eggs. The three mouse lineages displayed distinct patterns in the expression of innate immune receptors, their adaptor molecules and cytokines, at 2 and 7 wpi. MAIN CONCLUSIONS Our results suggest that the pathogenesis of S. mansoni infection is related to a dynamic early activation of innate immunity receptors and cytokines important for the control of developing worms.
Collapse
Affiliation(s)
- Janete Cunha Lima
- Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Biologia Parasitária, Natal, RN, Brasil
| | | | - Luanderson Cardoso Pereira
- Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Biologia Parasitária, Natal, RN, Brasil
| | - Nathalie de Sena Pereira
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Natal, RN, Brasil
| | | | - Alan Lane de Melo
- Universidade Federal de Minas Gerais, Departamento de Parasitologia, Belo Horizonte, MG, Brasil
| | - Paulo Marcos Matta Guedes
- Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Biologia Parasitária, Natal, RN, Brasil
| |
Collapse
|
2
|
Lekki-Jóźwiak J, Bąska P. The Roles of Various Immune Cell Populations in Immune Response against Helminths. Int J Mol Sci 2023; 25:420. [PMID: 38203591 PMCID: PMC10778651 DOI: 10.3390/ijms25010420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Helminths are multicellular parasites that are a substantial problem for both human and veterinary medicine. According to estimates, 1.5 billion people suffer from their infection, resulting in decreased life quality and burdens for healthcare systems. On the other hand, these infections may alleviate autoimmune diseases and allergy symptoms. The immune system is programmed to combat infections; nevertheless, its effector mechanisms may result in immunopathologies and exacerbate clinical symptoms. This review summarizes the role of the immune response against worms, with an emphasis on the Th2 response, which is a hallmark of helminth infections. We characterize non-immune cells (enteric tuft cells-ETCs) responsible for detecting parasites, as well as the role of hematopoietic-derived cells (macrophages, basophils, eosinophils, neutrophils, innate lymphoid cells group 2-ILC2s, mast cells, T cells, and B cells) in initiating and sustaining the immune response, as well as the functions they play in granulomas. The aim of this paper is to review the existing knowledge regarding the immune response against helminths, to attempt to decipher the interactions between cells engaged in the response, and to indicate the gaps in the current knowledge.
Collapse
Affiliation(s)
- Janina Lekki-Jóźwiak
- Division of Parasitology and Parasitic Diseases, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|
3
|
Guo N, Liu Y, Hao Q, Sun M, Li F. A Mannose Receptor from Litopenaeus vannamei Involved in Innate Immunity by Pathogen Recognition and Inflammation Regulation. Int J Mol Sci 2023; 24:10665. [PMID: 37445842 DOI: 10.3390/ijms241310665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Mannose receptor, as a member of the C-type lectin superfamily, is a non-canonical pattern recognition receptor that can internalize pathogen-associated ligands and activate intracellular signaling. Here, a mannose receptor gene, LvMR, was identified from the Pacific white shrimp Litopenaeus vannamei. LvMR encoded a signal peptide, a fibronectin type II (FN II) domain, and two carbohydrate-recognition domains (CRDs) with special EPS and FND motifs. LvMR transcripts were mainly detected in the hepatopancreas, and presented a time-dependent response after pathogen challenge. The recombinant LvMR (rLvMR) could bind to various PAMPs and agglutinate microorganisms in a Ca2+-dependent manner with strong binding ability to D-mannose and N-acetyl sugars. The knockdown of LvMR enhanced the expression of most NF-κB pathway genes, inflammation and redox genes, while it had no obvious effect on the transcription of most phagocytosis genes. Moreover, the knockdown of LvMR caused an increase in reactive oxygen species (ROS) content and inducible nitric oxide synthase (iNOS) activity in the hepatopancreas after Vibrio parahaemolyticus infection. All these results indicate that LvMR might perform as a PRR in immune recognition and a negative regulator of inflammation during bacterial infection.
Collapse
Affiliation(s)
- Na Guo
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Yuan Liu
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
| | - Qiang Hao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Mingzhe Sun
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
| | - Fuhua Li
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
4
|
Chayé MAM, Gasan TA, Ozir-Fazalalikhan A, Scheenstra MR, Zawistowska-Deniziak A, van Hengel ORJ, Gentenaar M, Manurung MD, Harvey MR, Codée JDC, Chiodo F, Heijke AM, Kalinowska A, van Diepen A, Hensbergen PJ, Yazdanbakhsh M, Guigas B, Hokke CH, Smits HH. Schistosoma mansoni egg-derived thioredoxin and Sm14 drive the development of IL-10 producing regulatory B cells. PLoS Negl Trop Dis 2023; 17:e0011344. [PMID: 37363916 DOI: 10.1371/journal.pntd.0011344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 05/02/2023] [Indexed: 06/28/2023] Open
Abstract
During chronic schistosome infections, a complex regulatory network is induced to regulate the host immune system, in which IL-10-producing regulatory B (Breg) cells play a significant role. Schistosoma mansoni soluble egg antigens (SEA) are bound and internalized by B cells and induce both human and mouse IL-10 producing Breg cells. To identify Breg-inducing proteins in SEA, we fractionated SEA by size exclusion chromatography and found 6 fractions able to induce IL-10 production by B cells (out of 18) in the high, medium and low molecular weight (MW) range. The high MW fractions were rich in heavily glycosylated molecules, including multi-fucosylated proteins. Using SEA glycoproteins purified by affinity chromatography and synthetic glycans coupled to gold nanoparticles, we investigated the role of these glycan structures in inducing IL-10 production by B cells. Then, we performed proteomics analysis on active low MW fractions and identified a number of proteins with putative immunomodulatory properties, notably thioredoxin (SmTrx1) and the fatty acid binding protein Sm14. Subsequent splenic murine B cell stimulations and hock immunizations with recombinant SmTrx1 and Sm14 showed their ability to dose-dependently induce IL-10 production by B cells both in vitro and in vivo. Identification of unique Breg cells-inducing molecules may pave the way to innovative therapeutic strategies for inflammatory and auto-immune diseases.
Collapse
Affiliation(s)
- Mathilde A M Chayé
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas A Gasan
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Maaike R Scheenstra
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anna Zawistowska-Deniziak
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Parasitology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Oscar R J van Hengel
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Max Gentenaar
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mikhael D Manurung
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael R Harvey
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Fabrizio Chiodo
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
- Italian National Research Council, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Anouk M Heijke
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alicja Kalinowska
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
- Museum and Institute of Zoology, Polish Academy of Sciences, Warsaw, Poland
| | - Angela van Diepen
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul J Hensbergen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
5
|
Eswar K, Mukherjee S, Ganesan P, Kumar Rengan A. Immunomodulatory Natural Polysaccharides: An Overview of the Mechanisms Involved. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2023.111935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
6
|
Dibo N, Liu X, Chang Y, Huang S, Wu X. Pattern recognition receptor signaling and innate immune responses to schistosome infection. Front Cell Infect Microbiol 2022; 12:1040270. [PMID: 36339337 PMCID: PMC9633954 DOI: 10.3389/fcimb.2022.1040270] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/03/2022] [Indexed: 08/22/2023] Open
Abstract
Schistosomiasis remains to be a significant public health problem in tropical and subtropical regions. Despite remarkable progress that has been made in the control of the disease over the past decades, its elimination remains a daunting challenge in many countries. This disease is an inflammatory response-driven, and the positive outcome after infection depends on the regulation of immune responses that efficiently clear worms and allow protective immunity to develop. The innate immune responses play a critical role in host defense against schistosome infection and pathogenesis. Initial pro-inflammatory responses are essential for clearing invading parasites by promoting appropriate cell-mediated and humoral immunity. However, elevated and prolonged inflammatory responses against the eggs trapped in the host tissues contribute to disease progression. A better understanding of the molecular mechanisms of innate immune responses is important for developing effective therapies and vaccines. Here, we update the recent advances in the definitive host innate immune response to schistosome infection, especially highlighting the critical roles of pattern recognition receptors and cytokines. The considerations for further research are also provided.
Collapse
Affiliation(s)
- Nouhoum Dibo
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Xianshu Liu
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Yunfeng Chang
- Department of Forensic Medicine Science, Xiangya School of Basic Medicine, Central South University, Yueyang, China
| | - Shuaiqin Huang
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
- Hunan Provincial Key Lab of Immunology and Transmission Control on Schistosomiasis, Hunan Provincial Institute of Schistosomiasis Control, Yueyang, China
| | - Xiang Wu
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
- Hunan Provincial Key Lab of Immunology and Transmission Control on Schistosomiasis, Hunan Provincial Institute of Schistosomiasis Control, Yueyang, China
| |
Collapse
|
7
|
Li H, Qiu D, Yuan Y, Wang X, Wu F, Yang H, Wang S, Ma M, Qian Y, Zhan B, Yang X. Trichinella spiralis cystatin alleviates polymicrobial sepsis through activating regulatory macrophages. Int Immunopharmacol 2022; 109:108907. [PMID: 35691271 DOI: 10.1016/j.intimp.2022.108907] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Sepsis is a life-threateningorgandysfunction caused by the cytokine storm induced by the severe bacterial infection. Excessive inflammatory responses are responsible for the lethal organ damage during the early stage of sepsis. Helminth infection and helminth-derived proteins have been identified to have the ability to immunomodulate the host immune system by reducing inflammation against inflammatory diseases. Trichinella spiralis cystatin (Ts-Cys) is a cysteine protease inhibitor with strong immunomodulatory functions on host immune system. Our previous studies have shown that excretory-secretory proteins of T. spiralis reduced sepsis-induced inflammation and Ts-Cys was able to inhibit macrophages to produce inflammatory cytokines. Whether Ts-Cys has a therapeutic effect on polymicrobial sepsis and related immunological mechanism are not yet known. METHODS Sepsis was induced in BALB/c mice using cecal ligation and puncture (CLP), followed by intraperitoneal injection of 15 µg recombinant Ts-Cys (rTs-Cys). The therapeutic effect of rTs-Cys on sepsis was evaluated by observing the 72-hour survival rates of CLP-induced septic mice and the acute injury of lung and kidney through measuring the wet/dry weight ratio of lung, the levels of blood urea nitrogen (BUN) and creatinine (Cr) in sera and the tissue section pathology. The potential underlying mechanism was investigated using mouse bone marrow-derived macrophages (BMDMs) by observing the effect of rTs-Cys on LPS-stimulated macrophage polarization. The expression of genes associated with macrophage polarization in BMDMs and tissues of septic mice was measured by Western Blotting and qPCR. RESULTS In this study, we demonstrated the treatment with rTs-Cys alleviated CLP-induced sepsis in mice with significantly reduced pathological injury in vital organs of lung and kidney and reduced mortality of septic mice. The further study identified that treatment with rTs-Cys promoted macrophage polarization from classically activated macrophage (M1) to alternatively activated macrophage (M2) phenotype via inhibiting TLR2/MyD88 signal pathway and increasing expression of mannose receptor (MR), inhibited pro-inflammatory cytokines (TNF-α, IL-6 and IL-1β) and increased regulatory anti-inflammatory cytokines (IL-10 and TGF-β) in sera and tissues (lung and kidney) of mice with polymicrobial sepsis. CONCLUSIONS Our results demonstrated that rTs-Cys had a therapeutic effect on sepsis through activating regulatory macrophages possibly via suppressing TLR2/MyD88 signal pathway. We also identified that rTs-Cys-induced M2 macrophage differentiation was associated with increased expression of MR on the surface of macrophages. Our results underscored the importance of MR in regulating macrophages during the treatment with rTs-Cys, providing another immunological mechanism in which helminths and their derived proteins modulate the host immune system. The findings in this study suggest that rTs-Cys is a potential therapeutic agent for the prevention and treatment of sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Huihui Li
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Dapeng Qiu
- Department of Orthopedics, Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuan Yuan
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Xiaoli Wang
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Fengjiao Wu
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Huijuan Yang
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shuying Wang
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Mengxi Ma
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Yayun Qian
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiaodi Yang
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
8
|
Yin X, Bai H, Mu L, Chen N, Qi W, Huang Y, Xu H, Jian J, Wang A, Ye J. Expression and functional characterization of the mannose receptor (MR) from Nile tilapia (Oreochromis niloticus) in response to bacterial infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 126:104257. [PMID: 34530040 DOI: 10.1016/j.dci.2021.104257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/11/2021] [Accepted: 09/11/2021] [Indexed: 06/13/2023]
Abstract
Mannose receptor (MR) as a member of the pattern recognition receptors (PRRs) plays an important role in the immune response. In mammals, the role of MR in the regulation of phagocytosis is clarified; however, its contribution to opsonize phagocytosis remains unclear in bony fish. In this study, the expression pattern of Nile tilapia mannose receptor gene (OnMR) was investigated and its regulation of the phagocytosis of monocytes/macrophages to pathogenic bacteria was identified. The full-length of OnMR open reading frame is 4314 bp, encoding a peptide containing 1437 amino acid residues. The deduced amino acid sequence revealed that OnMR contained a cysteine-rich domain, a fibronectin type II domain, multiple C-type lectin-like domains, a transmembrane domain and a short cytoplasmic domain. Tissue distribution analysis showed the OnMR transcripts was widely distribute in the ten detected tissues, and highly expressed in head kidney, hind kidney, intestine and spleen. After S. agalactiae and A. hydrophila infection, the expression of OnMR in head kidney and spleen increased significantly. Moreover, the expression of OnMR in MO/Mø were also upregulated post the infection of bacteria and mannose solutions in vitro. This suggested that MR, as a mannose receptor on macrophage surface, could respond strongly to the stimulation of pathogenic bacteria. In addition, the (r)OnMR protein could effectively bind and agglutinate S. agalactiae and A. hydrophila, and regulate the phagocytic ability of monocytes/macrophages to pathogenic bacteria. These results suggest that OnMR is involved in response against bacterial infection in Nile tilapia, and this study will help us better understand the function of MR in teleost fish.
Collapse
Affiliation(s)
- Xiaoxue Yin
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China.
| | - Hao Bai
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Liangliang Mu
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China.
| | - Nuo Chen
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Weiwei Qi
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Yu Huang
- Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Heyi Xu
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Jichang Jian
- Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Anli Wang
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Jianmin Ye
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China.
| |
Collapse
|
9
|
Li K, Li R, Zou P, Li L, Wang H, Kong D, Zheng G, Li LL. Glycopeptide-nanotransforrs eyedrops with enhanced permeability and retention for preventing fundus neovascularization. Biomaterials 2022; 281:121361. [PMID: 34991034 DOI: 10.1016/j.biomaterials.2021.121361] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/19/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023]
Abstract
Efficient and non-invasive drug delivery to the fundus has always been a medical difficulty. Here, a co-assembled glycopeptide nanotransforrs (GPNTs) named MRP@DOX as a drug delivery system is reported. The MRP@DOX co-assemble nanoparticles consisting of glycopeptide, cationic peptide, and doxorubicin (DOX). The nanoparticles are positively charged with the nano-size, which can be induced transformation by legumain cleavage. Once administrate to the eyes, MRP@DOX has a high penetration through the ocular surface to specifically targets M2 macrophages in the fundus. Then, the mannose receptor mediates phagocytosis and intracellular highly expressed legumain induces its nanofibrous transformation, which contributes to a 44.7% DOX retention in cells at 24 h than that of the non-transformed controls (MAP@DOX: 5.1%). The nanofiber transformation provides an inhibition of exocytosis, which explains the higher retention of the delivered drug. In the mouse OIR model, MRP@DOX completely restores the physiological angiogenesis and reduces pathological neovascularization. Pathological neovascularization branches and cell nuclei that break through the inner limiting membrane are reduced by 55% and 72%, respectively, which are 25% and 20% less than those in the non-transformed controls. In addition, MRP@DOX also has good histocompatibility, which provides a possible strategy for non-invasive treatment of fundus diseases in the future.
Collapse
Affiliation(s)
- Ke Li
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Hennan, 450052, China; Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Hennan, 450052, China
| | - Ruxiang Li
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Pengfei Zou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Li Li
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Hennan, 450052, China
| | - Huajun Wang
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Hennan, 450052, China
| | - Deqian Kong
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Hennan, 450052, China; Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Hennan, 450052, China
| | - Guangying Zheng
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Hennan, 450052, China.
| | - Li-Li Li
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing, 100190, China.
| |
Collapse
|
10
|
Feng Y, Xie H, Shi F, Chen D, Xie A, Li J, Fang C, Wei H, Huang H, Pan X, Tang X, Huang J. Roles of TLR7 in Schistosoma japonicum Infection-Induced Hepatic Pathological Changes in C57BL/6 Mice. Front Cell Infect Microbiol 2021; 11:754299. [PMID: 34692568 PMCID: PMC8531751 DOI: 10.3389/fcimb.2021.754299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022] Open
Abstract
S. japonicum infection can induce granulomatous inflammation in the liver of the host. Granulomatous inflammation limits the spread of infection and plays a role in host protection. Toll-like receptor 7 (TLR7) is an endosomal TLR that recognizes single-stranded RNA (ssRNA). In this study, the role of TLR7 in S. japonicum infection-induced hepatitis was investigated in both normal and TLR7 knockout (KO) C57BL/6 mice. The results indicated that TLR7 KO could aggravate S. japonicum infection-induced damage in the body, with less granuloma formation in the tissue, lower WBCs in blood, and decreased ALT and AST in the serum. Then, the expression of TLR7 was detected in isolated hepatic lymphocytes. The results indicated that the percentage of TLR7+ cells was increased in the infected mice. Hepatic macrophages, DCs, and B cells could express TLR7, and most of the TLR7-expressing cells in the liver of infected mice were macrophages. The percentage of TLR7-expressing macrophages was also increased after infection. Moreover, macrophages, T cells, and B cells showed significant changes in the counts, activation-associated molecule expression, and cytokine secretion between S. japonicum-infected WT and TLR7 KO mice. Altogether, this study indicated that TLR7 could delay the progression of S. japonicum infection-induced hepatitis mainly through macrophages. DCs, B cells, and T cells were involved in the TLR7-mediated immune response.
Collapse
Affiliation(s)
- Yuanfa Feng
- Department of Infectious Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Hongyan Xie
- China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Feihu Shi
- Department of Infectious Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dianhui Chen
- Department of Infectious Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Anqi Xie
- China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Jiajie Li
- China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Chao Fang
- China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Haixia Wei
- China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - He Huang
- China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Xingfei Pan
- Department of Infectious Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoping Tang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jun Huang
- China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
11
|
Ouyang A, Wang H, Su J, Liu X. Mannose Receptor Mediates the Activation of Chitooligosaccharides on Blunt Snout Bream ( Megalobrama amblycephala) Macrophages. Front Immunol 2021; 12:686846. [PMID: 34408745 PMCID: PMC8365301 DOI: 10.3389/fimmu.2021.686846] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/16/2021] [Indexed: 01/23/2023] Open
Abstract
Chitooligosaccharide (COS) is an important immune enhancer and has been proven to have a variety of biological activities. Our previous research has established an M1 polarization mode by COS in blunt snout bream (Megalobrama amblycephala) macrophages, but the mechanism of COS activation of blunt snout bream macrophages remains unclear. In this study, we further explored the internalization mechanism and signal transduction pathway of chitooligosaccharide hexamer (COS6) in blunt snout bream macrophages. The results showed that mannose receptor C-type lectin-like domain 4-8 of M. amblycephala (MaMR CTLD4-8) could recognize and bind to COS6 and mediate COS6 into macrophages by both clathrin-dependent and caveolin-dependent pathways. In the inflammatory response of macrophages activated by COS6, the gene expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and nitric oxide synthase 2 (NOS2) was significantly inhibited after MaMR CTLD4-8-specific antibody blockade. However, even if it was blocked, the expression of these inflammation-related genes was still relatively upregulated, which suggested that there are other receptors involved in immune regulation. Further studies indicated that MaMR CTLD4-8 and Toll-like receptor 4 (TLR4) cooperated to regulate the pro-inflammatory response of macrophages caused by COS6. Taken together, these results revealed that mannose receptor (MR) CTLD4-8 is indispensable in the process of recognition, binding, internalization, and immunoregulation of COS in macrophages of blunt snout bream.
Collapse
Affiliation(s)
- Aotian Ouyang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Huabing Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jianguo Su
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Xiaoling Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| |
Collapse
|
12
|
Hinshaw DC, Hanna A, Lama-Sherpa T, Metge B, Kammerud SC, Benavides GA, Kumar A, Alsheikh HA, Mota M, Chen D, Ballinger SW, Rathmell JC, Ponnazhagan S, Darley-Usmar V, Samant RS, Shevde LA. Hedgehog signaling regulates metabolism and polarization of mammary tumor-associated macrophages. Cancer Res 2021; 81:5425-5437. [PMID: 34289986 DOI: 10.1158/0008-5472.can-20-1723] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 05/03/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022]
Abstract
Elevated infiltration of immunosuppressive alternatively polarized (M2) macrophages is associated with poor prognosis in cancer patients. The tumor microenvironment remarkably orchestrates molecular mechanisms that program these macrophages. Here we identify a novel role for oncogenic Hedgehog (Hh) signaling in programming signature metabolic circuitries that regulate alternative polarization of tumor-associated macrophages. Two immunocompetent orthotopic mouse models of mammary tumors were used to test the effect of inhibiting Hh signaling on tumor-associated macrophages. Treatment with the pharmacological Hh inhibitor Vismodegib induced a significant shift in the profile of tumor-infiltrating macrophages. Mass spectrometry-based metabolomic analysis showed Hh inhibition induced significant alterations in metabolic processes, including metabolic sensing, mitochondrial adaptations, and lipid metabolism. In particular, inhibition of Hh in M2 macrophages reduced flux through the UDP-GlcNAc biosynthesis pathway. Consequently, O-GlcNAc-modification of STAT6 decreased, mitigating the immune suppressive program of M2 macrophages, and the metabolically demanding M2 macrophages shifted their metabolism and bioenergetics from fatty acid oxidation to glycolysis. M2 macrophages enriched from Vismodegib-treated mammary tumors showed characteristically decreased O-GlcNAcylation and altered mitochondrial dynamics. These Hh-inhibited macrophages are reminiscent of inflammatory (M1) macrophages, phenotypically characterized by fragmented mitochondria. This is the first report highlighting the relevance of Hh signaling in controlling a complex metabolic network in immune cells. These data describe a novel immunometabolic function of Hh signaling that can be clinically exploited.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Atul Kumar
- Department of Pathology, University of Alabama at Birmingham
| | | | - Mateus Mota
- Department of Pathology, University of Alabama at Birmingham
| | - Dongquan Chen
- Division of Preventive Medicine, University of Alabama at Birmingham
| | | | - Jeffrey C Rathmell
- Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
| | | | | | | | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham
| |
Collapse
|
13
|
Miles S, Velasco-de-Andrés M, Lozano F, Mourglia-Ettlin G. Interactome analysis of CD5 and CD6 ectodomains with tegumental antigens from the helminth parasite Echinococcus granulosus sensu lato. Int J Biol Macromol 2020; 164:3718-3728. [PMID: 32891642 DOI: 10.1016/j.ijbiomac.2020.08.219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 01/29/2023]
Abstract
Echinococcus granulosus sensu lato (s.l.) is a cestode parasite affecting both human and livestock health. Recombinant ectodomains of human scavenger receptors CD5 (rshCD5) and CD6 (rshCD6) were previously reported to bind its tegumental antigens and to exert prophylactic effects in a murine model of infection. Although the properties of mammalian scavenger receptors include the binding to diverse pathogen-derived structures, their interaction with helminth parasites has been scarcely explored. Therefore, we report here a search for CD5 and CD6 interactors within E. granulosus s.l. antigens. Mass spectrometry analysis of pull-downs from soluble tegumental components with biotinylated rshCD5 and rshCD6 resulted in 17 and 11 overrepresented interactors, respectively, 8 of which were shared. The interactors included previously reported protective molecules against E. granulosus s.l. and/or other helminths. Similar studies performed with 11-mer peptides mapping to each of the three extracellular scavenger domains of CD5 and CD6 allowed an estimated molecular topology of the interactions. In conclusion, the fact that most helminth interactors identified for rshCD5 and rshCD6 were already reported as vaccine candidates or pharmacological targets against different helminthiases, supports the view that their beneficial effects in experimental infection results from binding to multiple relevant tegumental antigens.
Collapse
Affiliation(s)
- Sebastián Miles
- Área Inmunología, Facultad de Química/Facultad de Ciencias, DEPBIO/IQB, Universidad de la República, Montevideo, Uruguay
| | - María Velasco-de-Andrés
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Servei d'Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain; Departament de Biomedicina, Universitat de Barcelona, Barcelona, Spain.
| | - Gustavo Mourglia-Ettlin
- Área Inmunología, Facultad de Química/Facultad de Ciencias, DEPBIO/IQB, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
14
|
Souza COS, Gardinassi LG, Rodrigues V, Faccioli LH. Monocyte and Macrophage-Mediated Pathology and Protective Immunity During Schistosomiasis. Front Microbiol 2020; 11:1973. [PMID: 32922381 PMCID: PMC7456899 DOI: 10.3389/fmicb.2020.01973] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Infection by Schistosoma parasites culminates in a chronic granulomatous disease characterized by intense tissue fibrosis. Along the course of schistosomiasis, diverse leukocytes are recruited for inflammatory foci. Innate immune cell accumulation in Th2-driven granulomas around Schistosoma eggs is associated with increased collagen deposition, while monocytes and macrophages exert critical roles during this process. Monocytes are recruited to damaged tissues from blood, produce TGF-β and differentiate into monocyte-derived macrophages (MDMs), which become alternatively activated by IL-4/IL-13 signaling via IL-4Rα (AAMs). AAMs are key players of tissue repair and wound healing in response to Schistosoma infection. Alternative activation of macrophages is characterized by the activation of STAT6 that coordinates the transcription of Arg1, Chi3l3, Relma, and Mrc1. In addition to these markers, monocyte-derived AAMs also express Raldh2 and Pdl2. AAMs produce high levels of IL-10 and TGF-β that minimizes tissue damage caused by Schistosoma egg accumulation in tissues. In this review, we provide support to previous findings about the host response to Schistosoma infection reusing public transcriptome data. Importantly, we discuss the role of monocytes and macrophages with emphasis on the mechanisms of alternative macrophage activation during schistosomiasis.
Collapse
Affiliation(s)
- Camila Oliveira Silva Souza
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Gustavo Gardinassi
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Vanderlei Rodrigues
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Lúcia Helena Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Coakley G, Harris NL. Interactions between macrophages and helminths. Parasite Immunol 2020; 42:e12717. [PMID: 32249432 DOI: 10.1111/pim.12717] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Macrophages, the major population of tissue-resident mononuclear phagocytes, contribute significantly to the immune response during helminth infection. Alternatively activated macrophages (AAM) are induced early in the anti-helminth response following tissue insult and parasite recognition, amplifying the early type 2 immune cascade initiated by epithelial cells and ILC2s, and subsequently driving parasite expulsion. AAM also contribute to functional alterations in tissues infiltrated with helminth larvae, mediating both tissue repair and inflammation. Their activation is amplified and occurs more rapidly following reinfection, where they can play a dual role in trapping tissue migratory larvae and preventing or resolving the associated inflammation and damage. In this review, we will address both the known and emerging roles of tissue macrophages during helminth infection, in addition to considering both outstanding research questions and new therapeutic strategies.
Collapse
Affiliation(s)
- Gillian Coakley
- Department of Immunology and Pathology, Central Clinical School, The Alfred Centre The Alfred Centre, Monash University, Melbourne, Victoria, Australia
| | - Nicola Laraine Harris
- Department of Immunology and Pathology, Central Clinical School, The Alfred Centre The Alfred Centre, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
16
|
Kuipers ME, Nolte-'t Hoen ENM, van der Ham AJ, Ozir-Fazalalikhan A, Nguyen DL, de Korne CM, Koning RI, Tomes JJ, Hoffmann KF, Smits HH, Hokke CH. DC-SIGN mediated internalisation of glycosylated extracellular vesicles from Schistosoma mansoni increases activation of monocyte-derived dendritic cells. J Extracell Vesicles 2020; 9:1753420. [PMID: 32489529 PMCID: PMC7241508 DOI: 10.1080/20013078.2020.1753420] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Helminths like Schistosoma mansoni release excretory/secretory (E/S) products that modulate host immunity to enable infection. Extracellular vesicles (EVs) are among these E/S products, yet molecular mechanisms and functionality of S. mansoni EV interaction with host immune cells is unknown. Here we demonstrate that EVs released by S. mansoni schistosomula are internalised by human monocyte-derived dendritic cells (moDCs). Importantly, we show that this uptake was mainly mediated via DC-SIGN (CD209). Blocking DC-SIGN almost completely abrogated EV uptake, while blocking mannose receptor (MR, CD206) or dendritic cell immunoreceptor (DCIR, CLEC4A) had no effect on EV uptake. Mass spectrometric analysis of EV glycans revealed the presence of surface N-glycans with terminal Galβ1-4(Fucα1-3)GlcNAc (LewisX) motifs, and a wide array of fucosylated lipid-linked glycans, including LewisX, a known ligand for DC-SIGN. Stimulation of moDCs with schistosomula EVs led to increased expression of costimulatory molecules CD86 and CD80 and regulatory surface marker PD-L1. Furthermore, schistosomula EVs increased expression of IL-12 and IL-10 by moDCs, which was partly dependent on the interaction with DC-SIGN. These results provide the first evidence that glycosylation of S. mansoni EVs facilitates the interaction with host immune cells and reveals a role for DC-SIGN and EV-associated glycoconjugates in parasite-induced immune modulation.
Collapse
Affiliation(s)
- Marije E Kuipers
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Esther N M Nolte-'t Hoen
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Alwin J van der Ham
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - D Linh Nguyen
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Clarize M de Korne
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Roman I Koning
- Department of Cell & Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - John J Tomes
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, UK
| | - Karl F Hoffmann
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, UK
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
17
|
Kaur D, Patiyal S, Sharma N, Usmani SS, Raghava GPS. PRRDB 2.0: a comprehensive database of pattern-recognition receptors and their ligands. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2020; 2019:5523871. [PMID: 31250014 PMCID: PMC6597477 DOI: 10.1093/database/baz076] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/19/2022]
Abstract
PRRDB 2.0 is an updated version of PRRDB that maintains comprehensive information about pattern-recognition receptors (PRRs) and their ligands. The current version of the database has ~2700 entries, which are nearly five times of the previous version. It contains extensive information about 467 unique PRRs and 827 pathogens-associated molecular patterns (PAMPs), manually extracted from ~600 research articles. It possesses information about PRRs and PAMPs that has been extracted manually from research articles and public databases. Each entry provides comprehensive details about PRRs and PAMPs that includes their name, sequence, origin, source, type, etc. We have provided internal and external links to various databases/resources (like Swiss-Prot, PubChem) to obtain further information about PRRs and their ligands. This database also provides links to ~4500 experimentally determined structures in the protein data bank of various PRRs and their complexes. In addition, 110 PRRs with unknown structures have also been predicted, which are important in order to understand the structure-function relationship between receptors and their ligands. Numerous web-based tools have been integrated into PRRDB 2.0 to facilitate users to perform different tasks like (i) extensive searching of the database; (ii) browsing or categorization of data based on receptors, ligands, source, etc. and (iii) similarity search using BLAST and Smith-Waterman algorithm.
Collapse
Affiliation(s)
- Dilraj Kaur
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi 110020, India
| | - Sumeet Patiyal
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi 110020, India
| | - Neelam Sharma
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi 110020, India
| | - Salman Sadullah Usmani
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi 110020, India.,Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh, Chandigarh 160036, India
| | - Gajendra P S Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, New Delhi 110020, India
| |
Collapse
|
18
|
Systemically Administered Plant Recombinant Holo-Intrinsic Factor Targets the Liver and is not Affected by Endogenous B12 levels. Sci Rep 2019; 9:12269. [PMID: 31439908 PMCID: PMC6706418 DOI: 10.1038/s41598-019-48555-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 08/06/2019] [Indexed: 01/09/2023] Open
Abstract
Precision targeting imaging agents and/or treatment agents to select cells or organs in the body remains a significant need and is an area of intense research. It has been hypothesized that the vitamin B12 (B12) dietary pathway, or components thereof, may be exploitable in this area. The question of whether gastric Intrinsic factor (IF), critical for B12 absorption in the GI tract via the cubilin receptor, could be used as a targeting moiety for the cubilin receptor systemically, has not been investigated. Cubilin is the only known receptor for holo-IF and is found primarily in the kidney and ear (outside of the ileum of the GI) offering significant scope for specific targeting. We utilized plant derived human gastric IF in fluorescent cell and PET based in vivo imaging and biodistribution studies and demonstrated that plant derived IF primarily targets the liver, likely a consequence of the unique glycosylation profile of the IF, and is not affected by endogenous B12 levels.
Collapse
|
19
|
Jürgensen HJ, Silva LM, Krigslund O, van Putten S, Madsen DH, Behrendt N, Engelholm LH, Bugge TH. CCL2/MCP-1 signaling drives extracellular matrix turnover by diverse macrophage subsets. Matrix Biol Plus 2019; 1:100003. [PMID: 33543002 PMCID: PMC7852312 DOI: 10.1016/j.mbplus.2019.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
Macrophage plasticity, cellular origin, and phenotypic heterogeneity are perpetual challenges for studies addressing the biology of this pivotal immune cell in development, homeostasis, and tissue remodeling/repair. Consequently, a myriad of macrophage subtypes has been described in these contexts. To facilitate the identification of functional macrophage subtypes in vivo, here we used a flow cytometry-based assay that allows for detailed phenotyping of macrophages engaged in extracellular matrix (ECM) degradation. Of the five macrophage subtypes identified in the remodeling dermis by using this assay, collagen degradation was primarily executed by Ly6C−CCR2+ and Ly6C−CCR2low macrophages via mannose receptor-dependent collagen endocytosis, while Ly6C+CCR2+ macrophages were the dominant fibrin-endocytosing cells. Unexpectedly, the CCL2/MCP1-CCR2 signaling axis was critical for both collagen and fibrin degradation, while collagen degradation was independent of IL-4Ra signaling. Furthermore, the cytokine GM-CSF selectively enhanced collagen degradation by Ly6C+CCR2+ macrophages. This study reveals distinct subsets of macrophages engaged in ECM turnover and identifies novel wound healing-associated functions for CCL2 and GM-CSF inflammatory cytokines. Phenotypically diverse subsets of dermal macrophages undertake the degradation of extracellular matrix C-C motif chemokine Ligand 2 (CCL2) signaling is critical for macrophage-mediated endocytosis of collagen and fibrin. Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) and Interleukin (IL)-13 stimulate collagen endocytosis. The wound healing-associated IL4-IL4 Receptor a (IL4Ra) signaling is dispensable for collagen endocytosis by macrophages. The mannose receptor is the principal endocytic collagen receptor utilized by resident dermal macrophages.
Collapse
Key Words
- AF, Alexa Fluor
- CCL2/MCP-1, chemokine (C-C motif) ligand 2/monocyte chemoattractant protein 1
- CCR2, C-C chemokine receptor type 2
- CEMS, collagen-endocytosing macrophages
- Collagen degradation
- ECM, extracellular matrix
- Extracellular matrix endocytosis
- FEMS, fibrin-endocytosing macrophages
- FMO, fluorescence minus one
- Fibrin degradation
- GM-CSF, Granulocyte Macrophage-Colony Stimulating Factor
- GM-CSFR, GM-CSF Receptor
- IL, Interleukin
- IL4Ra, IL4 Receptor a
- Interleukin-13
- M-CSF, Macrophage-Colony Stimulating Factor
- MR, mannose receptor/CD206
- Mannose receptor/CD206
- Plg, plasminogen
- RFP, red fluorescent protein
- uPARAP, urokinase plasminogen activator receptor associated protein/Endo180
- uPARAP/Endo180
Collapse
Affiliation(s)
- Henrik J. Jürgensen
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Lakmali M. Silva
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
- Oral Inflammation and Immunity Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Oliver Krigslund
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Sander van Putten
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Daniel H. Madsen
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
- Center for Cancer Immune Therapy (CCIT), Department of Haematology, Herlev Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
- Department of Oncology, Herlev Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| | - Niels Behrendt
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Lars H. Engelholm
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Thomas H. Bugge
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
- Corresponding author at: Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 211, Bethesda, MD 20892, USA.
| |
Collapse
|
20
|
Mourglia-Ettlin G, Miles S, Velasco-De-Andrés M, Armiger-Borràs N, Cucher M, Dematteis S, Lozano F. The ectodomains of the lymphocyte scavenger receptors CD5 and CD6 interact with tegumental antigens from Echinococcus granulosus sensu lato and protect mice against secondary cystic echinococcosis. PLoS Negl Trop Dis 2018; 12:e0006891. [PMID: 30500820 PMCID: PMC6267981 DOI: 10.1371/journal.pntd.0006891] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022] Open
Abstract
Background Scavenger Receptors (SRs) from the host’s innate immune system are known to bind multiple ligands to promote the removal of non-self or altered-self targets. CD5 and CD6 are two highly homologous class I SRs mainly expressed on all T cells and the B1a cell subset, and involved in the fine tuning of activation and differentiation signals delivered by the antigen-specific receptors (TCR and BCR, respectively), to which they physically associate. Additionally, CD5 and CD6 have been shown to interact with and sense the presence of conserved pathogen-associated structures from bacteria, fungi and/or viruses. Methodology/Principal findings We report herein the interaction of CD5 and CD6 lymphocyte surface receptors with Echinococcus granulosus sensu lato (s.l.). Binding studies show that both soluble and membrane-bound forms of CD5 and CD6 bind to intact viable protoscoleces from E. granulosus s.l. through recognition of metaperiodate-resistant tegumental components. Proteomic analyses allowed identification of thioredoxin peroxidase for CD5, and peptidyl-prolyl cis-trans isomerase (cyclophilin) and endophilin B1 (antigen P-29) for CD6, as their potential interactors. Further in vitro assays demonstrate that membrane-bound or soluble CD5 and CD6 forms differentially modulate the pro- and anti-inflammatory cytokine release induced following peritoneal cells exposure to E. granulosus s.l. tegumental components. Importantly, prophylactic infusion of soluble CD5 or CD6 significantly ameliorated the infection outcome in the mouse model of secondary cystic echinococcosis. Conclusions/Significance Taken together, the results expand the pathogen binding properties of CD5 and CD6 and provide novel evidence for their therapeutic potential in human cystic echinococcosis. Scavenger Receptors (SRs) are constituents of host’s innate immune system able to sense and remove altered-self and/or pathogen components. Data on their interaction with helminth parasites is scarce. In this work, we describe that CD5 and CD6 -two lymphoid SRs previously reported to interact with conserved structures from bacteria, fungi and viruses- recognize tegumental components in the cestode parasite Echinococcus granulosus sensu lato (s.l.). Moreover, both receptors differentially modulate the cytokine release by host cells exposed to E. granulosus s.l. tegumental components. Importantly, the infusion of soluble forms of CD5 or CD6 improve infection outcomes in a murine model of secondary cystic echinococcosis. In summary, our results expand the pathogen binding properties of CD5 and CD6 and suggest their therapeutic potential against helminth infections.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/metabolism
- CD5 Antigens/genetics
- CD5 Antigens/metabolism
- Echinococcosis/genetics
- Echinococcosis/metabolism
- Echinococcosis/parasitology
- Echinococcus granulosus/genetics
- Echinococcus granulosus/metabolism
- Female
- Helminth Proteins/genetics
- Helminth Proteins/metabolism
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Protein Binding
- Proteomics
- Receptors, Scavenger/genetics
- Receptors, Scavenger/metabolism
- T-Lymphocytes/metabolism
- T-Lymphocytes/parasitology
Collapse
Affiliation(s)
- Gustavo Mourglia-Ettlin
- Área Inmunología, Facultad de Química/Facultad de Ciencias, DEPBIO/IQB, Universidad de la República, Montevideo, Uruguay
- * E-mail: (GM-E); (FL)
| | - Sebastián Miles
- Área Inmunología, Facultad de Química/Facultad de Ciencias, DEPBIO/IQB, Universidad de la República, Montevideo, Uruguay
| | - María Velasco-De-Andrés
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Noelia Armiger-Borràs
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marcela Cucher
- Instituto de Investigaciones en Microbiología y Parasitología Médica, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sylvia Dematteis
- Área Inmunología, Facultad de Química/Facultad de Ciencias, DEPBIO/IQB, Universidad de la República, Montevideo, Uruguay
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Servei d’Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
- Departament de Biomedicina, Universitat de Barcelona, Barcelona, Spain
- * E-mail: (GM-E); (FL)
| |
Collapse
|
21
|
Winkel BMF, Dalenberg MR, de Korne CM, Feijt C, Langenberg MCC, Pelgrom L, Ganesh MS, Yazdanbakhsh M, Smits HH, de Jong EC, Everts B, van Leeuwen FWB, Hokke CH, Roestenberg M. Early Induction of Human Regulatory Dermal Antigen Presenting Cells by Skin-Penetrating Schistosoma Mansoni Cercariae. Front Immunol 2018; 9:2510. [PMID: 30429854 PMCID: PMC6220649 DOI: 10.3389/fimmu.2018.02510] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022] Open
Abstract
Following initial invasion of Schistosoma mansoni cercariae, schistosomula reside in the skin for several days during which they can interact with the dermal immune system. While murine experiments have indicated that exposure to radiation-attenuated (RA) cercariae can generate protective immunity which is initiated in the skin stage, contrasting non-attenuated cercariae, such data is missing for the human model. Since murine skin does not form a reliable marker for immune responses in human skin, we used human skin explants to study the interaction with non-attenuated and RA cercariae with dermal innate antigen presenting cells (APCs) and the subsequent immunological responses. We exposed human skin explants to cercariae and visualized their invasion in real time (initial 30 min) using novel imaging technologies. Subsequently, we studied dermal immune responses and found an enhanced production of regulatory cytokine interleukin (IL)-10, pro-inflammatory cytokine IL-6 and macrophage inflammatory protein (MIP)-1α within 3 days of exposure. Analysis of dermal dendritic cells (DDCs) for their phenotype revealed an increased expression of immune modulators programmed death ligand (PD-L) 1 and 2, and increased IL-10 production. Ex vivo primed DDCs suppress Th1 polarization of naïve T-cells and increase T-cell IL-10 production, indicating their regulatory potential. These immune responses were absent or decreased after exposure to RA parasites. Using transwells, we show that direct contact between APCs and cercariae is required to induce their regulatory phenotype. To the best of our knowledge this is the first study that attempts to provide insight in the human dermal S. mansoni cercariae invasion and subsequent immune responses comparing non-attenuated with RA parasites. We reveal that cercariae induce a predominantly regulatory immune response whereas RA cercariae fail to achieve this. This initial understanding of the dermal immune suppressive capacity of S. mansoni cercariae in humans provides a first step toward the development of an effective schistosome vaccine.
Collapse
Affiliation(s)
- Béatrice M F Winkel
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Mirjam R Dalenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Clarize M de Korne
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Carola Feijt
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Leonard Pelgrom
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Munisha S Ganesh
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Esther C de Jong
- Department of Cell Biology and Histology, Academic Medical Center, Amsterdam, Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Fijs W B van Leeuwen
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
22
|
Zakeri A, Hansen EP, Andersen SD, Williams AR, Nejsum P. Immunomodulation by Helminths: Intracellular Pathways and Extracellular Vesicles. Front Immunol 2018; 9:2349. [PMID: 30369927 PMCID: PMC6194161 DOI: 10.3389/fimmu.2018.02349] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022] Open
Abstract
Helminth parasites are masters at manipulating host immune responses, using an array of sophisticated mechanisms. One of the major mechanisms enabling helminths to establish chronic infections is the targeting of pattern recognition receptors (PRRs) including toll-like receptors, C-type lectin receptors, and the inflammasome. Given the critical role of these receptors and their intracellular pathways in regulating innate inflammatory responses, and also directing adaptive immunity toward Th1 and Th2 responses, recognition of the pathways triggered and/or modulated by helminths and their products will provide detailed insights about how helminths are able to establish an immunoregulatory environment. However, helminths also target PRRs-independent mechanisms (and most likely other yet unknown mechanisms and pathways) underpinning the battery of different molecules helminths produce. Herein, the current knowledge on intracellular pathways in antigen presenting cells activated by helminth-derived biomolecules is reviewed. Furthermore, we discuss the importance of helminth-derived vesicles as a less-appreciated components released during infection, their role in activating these host intracellular pathways, and their implication in the development of new therapeutic approaches for inflammatory diseases and the possibility of designing a new generation of vaccines.
Collapse
Affiliation(s)
- Amin Zakeri
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Eline P. Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Sidsel D. Andersen
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Andrew R. Williams
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Peter Nejsum
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
23
|
Knuhr K, Langhans K, Nyenhuis S, Viertmann K, Kildemoes AMO, Doenhoff MJ, Haas H, Schramm G. Schistosoma mansoni Egg-Released IPSE/alpha-1 Dampens Inflammatory Cytokine Responses via Basophil Interleukin (IL)-4 and IL-13. Front Immunol 2018; 9:2293. [PMID: 30364177 PMCID: PMC6191518 DOI: 10.3389/fimmu.2018.02293] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/17/2018] [Indexed: 12/21/2022] Open
Abstract
Schistosomes control inflammation in their hosts via highly effective mechanisms such as induction of Tregs, Bregs, and alternatively activated macrophages (AAMs). Notably, IPSE/alpha-1, the major secretory product from Schistosoma mansoni eggs, triggers basophils to release interleukin (IL)-4 and IL-13. Both cytokines are essential for AAM induction, suggesting an important role for IPSE/alpha-1 in inflammation control. Here, we show by in vitro co-culture experiments that IPSE/alpha-1-induced basophil IL-4/IL-13 inhibited pro-inflammatory cytokine release from human LPS-activated monocytes. This effect was cell/cell contact-independent but dependent on IL-4, since it was abrogated in the presence of anti-IL-4 antibodies. Importantly, the IPSE/alpha-1-induced IL-4/IL-13 release from basophils was amplified in the presence of LPS. Moreover, monocytes co-cultured in the presence of LPS with IPSE/alpha-1-stimulated basophils adopted an AAM-like phenotype as assessed by elevated expression of CD206 and CD209. The putative in vivo relevance of these findings was supported by immunohistological staining of S. mansoni-infected murine tissue revealing close physical contact between IPSE/alpha-1 and basophils in schistosome egg granulomas. Taken together, we found that IPSE/alpha-1 dampens inflammatory cytokine responses by triggering basophil IL-4/IL-13, in particular in the context of TLR activation, thereby turning inflammatory monocytes into anti-inflammatory AAMs. This might represent a mechanism used by schistosomes to control inflammation in the host.
Collapse
Affiliation(s)
- Katrin Knuhr
- Experimental Pneumology, Research Center Borstel, Airway Research Center North, Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Kristina Langhans
- Experimental Pneumology, Research Center Borstel, Airway Research Center North, Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Sandra Nyenhuis
- Experimental Pneumology, Research Center Borstel, Airway Research Center North, Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Kerstin Viertmann
- Experimental Pneumology, Research Center Borstel, Airway Research Center North, Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Anna M Overgaard Kildemoes
- Section for Parasitology and Aquatic Pathobiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Doenhoff
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Gabriele Schramm
- Experimental Pneumology, Research Center Borstel, Airway Research Center North, Member of the German Center for Lung Research (DZL), Borstel, Germany
| |
Collapse
|
24
|
Macrophage Activation and Functions during Helminth Infection: Recent Advances from the Laboratory Mouse. J Immunol Res 2018; 2018:2790627. [PMID: 30057915 PMCID: PMC6051086 DOI: 10.1155/2018/2790627] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 05/23/2018] [Indexed: 12/20/2022] Open
Abstract
Macrophages are highly plastic innate immune cells that adopt an important diversity of phenotypes in response to environmental cues. Helminth infections induce strong type 2 cell-mediated immune responses, characterized among other things by production of high levels of interleukin- (IL-) 4 and IL-13. Alternative activation of macrophages by IL-4 in vitro was described as an opposite phenotype of classically activated macrophages, but the in vivo reality is much more complex. Their exact activation state as well as the role of these cells and associated molecules in type 2 immune responses remains to be fully understood. We can take advantage of a variety of helminth models available, each of which have their own feature including life cycle, site of infection, or pathological mechanisms influencing macrophage biology. Here, we reviewed the recent advances from the laboratory mouse about macrophage origin, polarization, activation, and effector functions during parasitic helminth infection.
Collapse
|
25
|
Clonorchis sinensis adult-derived proteins elicit Th2 immune responses by regulating dendritic cells via mannose receptor. PLoS Negl Trop Dis 2018; 12:e0006251. [PMID: 29505573 PMCID: PMC5854424 DOI: 10.1371/journal.pntd.0006251] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 03/15/2018] [Accepted: 01/18/2018] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Clonorchis sinensis (C. sinensis) is the most widespread human liver fluke in East Asia including China and Korea. Clonorchiasis as a neglected tropical zoonosis, leads to serious economic and public health burden in China. There are considerable evidences for an etiological relation between chronic clonorchiasis and liver fibrosis in human beings. Liver fibrosis is a highly conserved and over-protected response to hepatic tissue injury. Immune cells including CD4+ T cell as well as dendritic cell (DC), and pro-fibrogenic cytokines like interleukin 4 (IL-4), IL-13 have been identified as vital manipulators in liver fibrogenesis. Our previous studies had a mere glimpse of T helper type 2 (Th2) dominant immune responses as key players in liver fibrosis induced by C. sinensis infection, but little is known about the involved mechanisms in this pathological process. METHODOLOGY/PRINCIPAL FINDINGS By flow cytometry (FACS), adult-derived total proteins of C. sinensis (CsTPs) down-regulated the expression of surface markers CD80, CD86 and major histocompatibility complex class II (MHC-II) on lipopolysaccharide (LPS) induced DC. ELISA results demonstrated that CsTPs inhibited IL-12p70 release from LPS-treated bone marrow-derived dendritic cells (BMDC). IL-10 level increased in a time-dependent manner in LPS-treated BMDCs after incubation with CsTPs. CD4+ T cells incubated with LPS-treated BMDCs plus CsTPs could significantly elevate IL-4 level by ELISA. Meanwhile, elevated expression of pro-fibrogenic mediators including IL-13 and IL-4 were detected in a co-culture system of LPS-activated BMDCs and naive T cells containing CsTPs. In vivo, CsTPs-immunized mice enhanced expression of type 2 cytokines IL-13, IL-10 and IL-4 in both splenocytes and hepatic tissue. Exposure of BMDCs to CsTPs activated expression of mannose receptor (MR) but not toll like receptor 2 (TLR2), TLR4, C-type lectin receptor DC-SIGN and Dectin-2 on the cell surface by RT-PCR and FACS. Blockade of MR almost completely reversed the capacity of CsTPs to suppress LPS-induced BMDCs surface markers CD80, CD86 and MHC-II expression, and further made these BMDCs fail to induce a Th2-skewed response as well as Th2 cell-associated cytokines IL-13 and IL-4 release in vitro. CONCLUSIONS/SIGNIFICANCE Collectively, we validated that CsTPs could suppress the maturation of BMDCs in the presence of LPS via binding MR, and showed that the CsTPs-pulsed BMDCs actively polarized naive T helper cells to Th2 cells though the production of IL-10 instead of IL-12. CsTPs endowed host with the capacity to facilitate Th2 cytokines production including IL-13 and IL-4 in vitro and vivo. The study might provide useful information for developing potential therapeutic targets against the disease.
Collapse
|
26
|
Coakley G, McCaskill JL, Borger JG, Simbari F, Robertson E, Millar M, Harcus Y, McSorley HJ, Maizels RM, Buck AH. Extracellular Vesicles from a Helminth Parasite Suppress Macrophage Activation and Constitute an Effective Vaccine for Protective Immunity. Cell Rep 2018; 19:1545-1557. [PMID: 28538175 PMCID: PMC5457486 DOI: 10.1016/j.celrep.2017.05.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/30/2017] [Accepted: 04/27/2017] [Indexed: 12/04/2022] Open
Abstract
Recent studies have demonstrated that many parasites release extracellular vesicles (EVs), yet little is known about the specific interactions of EVs with immune cells or their functions during infection. We show that EVs secreted by the gastrointestinal nematode Heligmosomoides polygyrus are internalized by macrophages and modulate their activation. EV internalization causes downregulation of type 1 and type 2 immune-response-associated molecules (IL-6 and TNF, and Ym1 and RELMα) and inhibits expression of the IL-33 receptor subunit ST2. Co-incubation with EV antibodies abrogated suppression of alternative activation and was associated with increased co-localization of the EVs with lysosomes. Furthermore, mice vaccinated with EV-alum generated protective immunity against larval challenge, highlighting an important role in vivo. In contrast, ST2-deficient mice are highly susceptible to infection, and they are unable to clear parasites following EV vaccination. Hence, macrophage activation and the IL-33 pathway are targeted by H. polygyrus EVs, while neutralization of EV function facilitates parasite expulsion. EVs from a nematode parasite suppress type 1 and type 2 activation of macrophages Antibodies block EV function and increase their co-localization with the lysosome in macrophages EV vaccination generates strong antibody responses and protective immunity against infection EVs target both the IL-33 pathway and macrophage activation to counter parasite expulsion
Collapse
Affiliation(s)
- Gillian Coakley
- Institute of Immunology and Infection Research and Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Jana L McCaskill
- Institute of Immunology and Infection Research and Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Jessica G Borger
- Institute of Immunology and Infection Research and Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Fabio Simbari
- Institute of Immunology and Infection Research and Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Elaine Robertson
- Institute of Immunology and Infection Research and Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Marissa Millar
- Institute of Immunology and Infection Research and Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Yvonne Harcus
- Institute of Immunology and Infection Research and Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Henry J McSorley
- Centre for Inflammation Research, University of Edinburgh, The Queens Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Rick M Maizels
- Wellcome Centre for Molecular Parasitology, Institute for Infection, Immunity and Inflammation, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK.
| | - Amy H Buck
- Institute of Immunology and Infection Research and Centre for Immunity, Infection & Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| |
Collapse
|
27
|
van Die I, Cummings RD. The Mannose Receptor in Regulation of Helminth-Mediated Host Immunity. Front Immunol 2017; 8:1677. [PMID: 29238348 PMCID: PMC5712593 DOI: 10.3389/fimmu.2017.01677] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/15/2017] [Indexed: 12/31/2022] Open
Abstract
Infection with parasitic helminths affects humanity and animal welfare. Parasitic helminths have the capacity to modulate host immune responses to promote their survival in infected hosts, often for a long time leading to chronic infections. In contrast to many infectious microbes, however, the helminths are able to induce immune responses that show positive bystander effects such as the protection to several immune disorders, including multiple sclerosis, inflammatory bowel disease, and allergies. They generally promote the generation of a tolerogenic immune microenvironment including the induction of type 2 (Th2) responses and a sub-population of alternatively activated macrophages. It is proposed that this anti-inflammatory response enables helminths to survive in their hosts and protects the host from excessive pathology arising from infection with these large pathogens. In any case, there is an urgent need to enhance understanding of how helminths beneficially modulate inflammatory reactions, to identify the molecules involved and to promote approaches to exploit this knowledge for future therapeutic interventions. Evidence is increasing that C-type lectins play an important role in driving helminth-mediated immune responses. C-type lectins belong to a large family of calcium-dependent receptors with broad glycan specificity. They are abundantly present on immune cells, such as dendritic cells and macrophages, which are essential in shaping host immune responses. Here, we will focus on the role of the C-type lectin macrophage mannose receptor (MR) in helminth-host interactions, which is a critically understudied area in the field of helminth immunobiology. We give an overview of the structural aspects of the MR including its glycan specificity, and the functional implications of the MR in helminth-host interactions focusing on a few selected helminth species.
Collapse
Affiliation(s)
- Irma van Die
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
28
|
Han Y, Wang HQ, Fu R, Qu W, Ruan EB, Wang XM, Wang GJ, Wu YH, Liu H, Song J, Guan J, Xing LM, Li LJ, Jiang HJ, Liu H, Wang YH, Liu CY, Zhang W, Shao ZH. [Characteristic and function of peripheral blood mononuclear cells-induced macrophages in patients with myelodysplastic syndrome]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 38:706-709. [PMID: 28954351 PMCID: PMC7348254 DOI: 10.3760/cma.j.issn.0253-2727.2017.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Indexed: 11/22/2022]
Abstract
Objective: To explore characteristic and function of peripheral blood mononuclear cells (PBMNC) -induced macrophages in patients with myelodysplastic syndrome (MDS) to couple with its progression. Methods: A total of 24 MDS patients (11 low-risk patients and 13 high-risk group patients) referred to Department of Hematology of Tianjin Medical University General Hospital and normal controls were enrolled from September 2014 to December 2015. PBMNC was stimulated with GM-CSF to transform to macrophages. The morphology of macrophages was observed by microscope. The quantity of macrophages, CD206 and SIRPα on surface of macrophages were detected by flow cytometry. The phagocytic function of macrophages was analyzed by fluorescence microscopy and flow cytometry. Results: The morphology of macrophages from MDS patients was abnormal. The percentage of transformed macrophages was (5.17±3.47) % in patients with MDS, which was lower than that in controls significantly[ (66.18±13.43) %, t=3.529, P=0.001]. The expression of CD206 on macrophages from MDS patients was significantly lower than that of controls[ (9.73±2.59) % vs (51.15±10.82) %, t=4.551, P<0.001]. The SIRPα level of macrophages from MDS patients was significantly lower than that of controls [ (0.51±0.09) % vs (0.77±0.06) %, t=2.102, P=0.043]. The phagocytic index and the percentage of phagocytic of macrophages from MDS patients were significantly lower than those of macrophages from normal controls[0.45±0.08 vs 0.92±0.07, t=-6.253, P=0.008; (23.69±3.22) % vs (42.75±2.13) %, t=-6.982, P=0.006 respectively]by flow cytometry. The phagocytic index of MDS patients was significantly lower than that of controls (0.24±0.04 vs 0.48±0.96, t=3.464, P=0.001) by fluorescence microscopy. Conclusion: The quantity, recognization receptors and phagocytosis of PBMNC-induced macrophages decreased in MDS patients.
Collapse
Affiliation(s)
- Y Han
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Řimnáčová J, Mikeš L, Turjanicová L, Bulantová J, Horák P. Changes in surface glycosylation and glycocalyx shedding in Trichobilharzia regenti (Schistosomatidae) during the transformation of cercaria to schistosomulum. PLoS One 2017; 12:e0173217. [PMID: 28296924 PMCID: PMC5351870 DOI: 10.1371/journal.pone.0173217] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/17/2017] [Indexed: 11/25/2022] Open
Abstract
The invasive larvae (cercariae) of schistosomes penetrate the skin of their definitive hosts. During the invasion, they undergo dramatic ultrastructural and physiological transitions. These changes result in the development of the subsequent stage, schistosomulum, which migrates through host tissues in close contact with host's immune system. One of the striking changes in the transforming cercariae is the shedding of their thick tegumental glycocalyx, which represents an immunoattractive structure; therefore its removal helps cercariae to avoid immune attack. A set of commercial fluorescently labeled lectin probes, their saccharide inhibitors and monoclonal antibodies against the trisaccharide Lewis-X antigen (LeX, CD15) were used to characterize changes in the surface saccharide composition of the neuropathogenic avian schistosome Trichobilharzia regenti during the transformation of cercariae to schistosomula, both in vitro and in vivo. The effect of various lectins on glycocalyx shedding was evaluated microscopically. The involvement of peptidases and their inhibitors on the shedding of glycocalyx was investigated using T. regenti recombinant cathepsin B2 and a set of peptidase inhibitors. The surface glycocalyx of T. regenti cercariae was rich in fucose and mannose/glucose residues. After the transformation of cercariae in vitro or in vivo within their specific duck host, reduction and vanishing of these epitopes was observed, and galactose/N-acetylgalactosamine emerged. The presence of LeX was not observed on the cercariae, but the antigen was gradually expressed from the anterior part of the body in the developing schistosomula. Some lectins which bind to the cercarial surface also induced secretion from the acetabular penetration glands. Seven lectins induced the shedding of glycocalyx by cercariae, among which five bound strongly to cercarial surface; the effect could be blocked by saccharide inhibitors. Mannose-binding protein, part of the lectin pathway of the complement system, also bound to cercariae and schistosomula, but had little effect on glycocalyx shedding. Our study did not confirm the involvement of proteolysis in glycocalyx shedding.
Collapse
Affiliation(s)
- Jana Řimnáčová
- Department of Parasitology, Faculty of Science, Charles University, Viničná 7, Prague 2, Czech Republic
| | - Libor Mikeš
- Department of Parasitology, Faculty of Science, Charles University, Viničná 7, Prague 2, Czech Republic
| | - Libuše Turjanicová
- Department of Parasitology, Faculty of Science, Charles University, Viničná 7, Prague 2, Czech Republic
| | - Jana Bulantová
- Department of Parasitology, Faculty of Science, Charles University, Viničná 7, Prague 2, Czech Republic
| | - Petr Horák
- Department of Parasitology, Faculty of Science, Charles University, Viničná 7, Prague 2, Czech Republic
| |
Collapse
|
30
|
Aira N, Andersson AM, Singh SK, McKay DM, Blomgran R. Species dependent impact of helminth-derived antigens on human macrophages infected with Mycobacterium tuberculosis: Direct effect on the innate anti-mycobacterial response. PLoS Negl Trop Dis 2017; 11:e0005390. [PMID: 28192437 PMCID: PMC5325601 DOI: 10.1371/journal.pntd.0005390] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 02/24/2017] [Accepted: 02/06/2017] [Indexed: 02/05/2023] Open
Abstract
Background In countries with a high prevalence of tuberculosis there is high coincident of helminth infections that might worsen disease outcome. While Mycobacterium tuberculosis (Mtb) gives rise to a pro-inflammatory Th1 response, a Th2 response is typical of helminth infections. A strong Th2 response has been associated with decreased protection against tuberculosis. Principal findings We investigated the direct effect of helminth-derived antigens on human macrophages, hypothesizing that helminths would render macrophages less capable of controlling Mtb. Measuring cytokine output, macrophage surface markers with flow cytometry, and assessing bacterial replication and phagosomal maturation revealed that antigens from different species of helminth directly affect macrophage responses to Mtb. Antigens from the tapeworm Hymenolepis diminuta and the nematode Trichuris muris caused an anti-inflammatory response with M2-type polarization, reduced macrophage phagosome maturation and ability to activate T cells, along with increased Mtb burden, especially in T. muris exposed cells which also induced the highest IL-10 production upon co-infection. However, antigens from the trematode Schistosoma mansoni had the opposite effect causing a decrease in IL-10 production, M1-type polarization and increased control of Mtb. Conclusion We conclude that, independent of any adaptive immune response, infection with helminth parasites, in a species-specific manner can influence the outcome of tuberculosis by either enhancing or diminishing the bactericidal function of macrophages. The innate immune system is the first response against invading pathogens like the bacterium Mycobacterium tuberculosis (Mtb) or parasitic worms (helminths). The adaptive immune response takes over after being primed by the innate immune response. Infection with Mycobacterium tuberculosis typically gives rise to a pro-inflammatory T-helper(Th)-1 response while helminths promote a Th2 response which is needed to combat the infection. Co-infection with both of these pathogens could lead to reduced immunity contributing to worsening of tuberculosis due to an increased Th2 response caused by helminths. We found that antigens from different helminth species (a nematode, a cestode and a trematode) caused different responses towards Mtb in macrophages. Depending on the helminth species, the macrophages can be more or less capable of combating Mtb infection and priming the adaptive immune response, which in turn would influence the outcome of tuberculosis. Thus, exposure to helminth antigens, in a species-dependent manner, could lead to a better control of Mtb infection or worsening of tuberculosis.
Collapse
Affiliation(s)
- Naomi Aira
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Anna-Maria Andersson
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Susmita K. Singh
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Derek M. McKay
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Disease, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert Blomgran
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
31
|
Monocyte-Derived Macrophages Are Impaired in Myelodysplastic Syndrome. J Immunol Res 2016; 2016:5479013. [PMID: 28074192 PMCID: PMC5198175 DOI: 10.1155/2016/5479013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/23/2016] [Indexed: 11/17/2022] Open
Abstract
Background. The myelodysplastic syndrome (MDS) comprises a group of clonal hematopoietic stem cell diseases characterized by cytopenia, dysplasia in one or more of the major myeloid lineages, ineffective hematopoiesis, and increased risk of development of acute myeloid leukemia (AML). Macrophages are innate immune cells that ingest and degrade abnormal cells, debris, and foreign material and orchestrate inflammatory processes. We analyzed the role of macrophages from MDS patients in vitro. Methods. Macrophages were induced from peripheral blood of patients with MDS via granulocyte macrophage colony-stimulating factor (GM-CSF). Phagocytic capacity of macrophages was measured with carboxyfluorescein succinimidyl ester and fluorescent microspheres. CD206 and signal regulatory protein alpha (SIRPα) on macrophages were detected by flow cytometry. Inducible nitric oxide synthase (iNOS) was measured by ELISA method. Results. Compared with normal control group, the number of monocytes increased in MDS patients. However, the monocytes showed impaired ability to induce macrophages and the number of macrophages induced from MDS samples was lower. Further, we demonstrated that the ex vivo phagocytic function of macrophages from MDS patients was impaired and levels of reorganization receptors CD206 and SIRPα were lower. Levels of iNOS secreted by macrophages in MDS were increased. Conclusions. Monocyte-derived macrophages are impaired in myelodysplastic syndromes.
Collapse
|
32
|
Abdel Aziz N, Tallima H, Hafez EA, El Ridi R. Papain-Based Vaccination Modulates Schistosoma mansoni Infection-Induced Cytokine Signals. Scand J Immunol 2016; 83:128-38. [PMID: 26603950 DOI: 10.1111/sji.12399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/09/2015] [Indexed: 01/07/2023]
Abstract
We have previously shown that immunization of outbred rodents with cysteine peptidases-based vaccine elicited type 2-biased immune responses associated with consistent and reproducible protection against challenge Schistosoma mansoni. We herein start to elucidate the molecular basis of C57BL/6 mouse resistance to S. mansoni following treatment with the cysteine peptidase, papain. We evaluated the early cytokine signals delivered by epidermal, dermal, and draining lymph node cells of naïve, and S. mansoni -infected mice treated 1 day earlier with 0 or 50 μg papain, or immunized twice with papain only (10 μg/mouse), papain-free recombinant S. mansoni glyceraldehyde 3-phosphate dehydrogenase and 2-Cys peroxiredoxin peptide (10 and 15 μg/mouse, respectively = antigen Mix), or papain-adjuvanted antigen Mix. Schistosoma mansoni infection induced epidermal and lymph node cells to release type 1, type 2 and type 17 cytokines, known to counteract each other. Expectedly, humoral immune responses were negligible until patency. Papain pretreatment or papain-based vaccination diminished or shut off S. mansoni infection early induction of type 1, type 17 and type 2 cytokines except for thymic stromal lymphopoietin and programmed the immune system towards a polarized type 2 immune milieu, associated with highly significant (P < 0.005 - <0.0001) resistance to S. mansoni infection.
Collapse
Affiliation(s)
- N Abdel Aziz
- Chemistry Department, Cairo University, Cairo, Egypt
| | - H Tallima
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - E A Hafez
- Chemistry Department, Cairo University, Cairo, Egypt
| | - R El Ridi
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| |
Collapse
|
33
|
Yamaguchi R, Yamamoto T, Sakamoto A, Ishimaru Y, Narahara S, Sugiuchi H, Yamaguchi Y. Roles of myeloperoxidase and GAPDH in interferon-gamma production of GM-CSF-dependent macrophages. Heliyon 2016; 2:e00080. [PMID: 27441256 PMCID: PMC4945965 DOI: 10.1016/j.heliyon.2016.e00080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 02/02/2016] [Accepted: 02/17/2016] [Indexed: 01/12/2023] Open
Abstract
Interferon (IFN)-gamma is highly expressed in atherosclerotic lesions and may have an important role in atherogenesis. Myeloperoxidase (MPO), the most abundant protein in neutrophils, is a marker of plaque vulnerability and a possible bridge between inflammation and cardiovascular disease. Granulocyte-macrophage colony-stimulating factor (GM-CSF) has also been implicated in the pathogenesis of atherosclerosis. The present study investigated the role of neutrophil activation in atherosclerosis. Adherent macrophages were obtained from primary cultures of human mononuclear cells. Expression of IFN-gamma protein by GM-CSF-dependent-macrophages was investigated by enzyme-linked immunosorbent assay after stimulation with MPO. GM-CSF enhanced macrophage expression of the mannose receptor (CD206), which is involved in MPO uptake. MPO increased IFN-gamma production by GM-CSF-dependent macrophages in a concentration-dependent manner. Pretreatment of macrophages with small interfering RNA (siRNA) for CD206 or extracellular signal-regulated kinase (ERK)-2 attenuated IFN-gamma production, while siRNA for ERK-1 did not. GAPDH is known to bind to adenylate/uridylate (AU)-rich elements of RNA and may influence IFN-gamma protein expression by binding to the AU-rich element of IFN-gamma mRNA. Interestingly, pretreatment with siRNA for GAPDH significantly reduced IFN-gamma production by macrophages, while it did not affect TF protein expression. In conclusion, MPO upregulates IFN-gamma production by GM-CSF-dependent-macrophages via the CD206/ERK-2 signaling pathway, while silencing GAPDH reduces IFN-gamma production.
Collapse
Affiliation(s)
- Rui Yamaguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
- Graduate School of Medical Science, Kumamoto University Medical School, Kumamoto, Japan
| | - Takatoshi Yamamoto
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Arisa Sakamoto
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Yasuji Ishimaru
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Shinji Narahara
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Hiroyuki Sugiuchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Yasuo Yamaguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
- Corresponding author at: Graduate School of Medical Science, Kumamoto Health Science University, Kitaku Izumi-machi 325, Kumamoto 861–5598, Japan.
| |
Collapse
|
34
|
Rodríguez E, Noya V, Cervi L, Chiribao ML, Brossard N, Chiale C, Carmona C, Giacomini C, Freire T. Glycans from Fasciola hepatica Modulate the Host Immune Response and TLR-Induced Maturation of Dendritic Cells. PLoS Negl Trop Dis 2015; 9:e0004234. [PMID: 26720149 PMCID: PMC4697847 DOI: 10.1371/journal.pntd.0004234] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 10/23/2015] [Indexed: 11/19/2022] Open
Abstract
Helminths express various carbohydrate-containing glycoconjugates on their surface, and they release glycan-rich excretion/secretion products that can be very important in their life cycles, infection and pathology. Recent evidence suggests that parasite glycoconjugates could play a role in the evasion of the immune response, leading to a modified Th2-polarized immune response that favors parasite survival in the host. Nevertheless, there is limited information about the nature or function of glycans produced by the trematode Fasciola hepatica, the causative agent of fasciolosis. In this paper, we investigate whether glycosylated molecules from F. hepatica participate in the modulation of host immunity. We also focus on dendritic cells, since they are an important target of immune-modulation by helminths, affecting their activity or function. Our results indicate that glycans from F. hepatica promote the production of IL-4 and IL-10, suppressing IFNγ production. During infection, this parasite is able to induce a semi-mature phenotype of DCs expressing low levels of MHCII and secrete IL-10. Furthermore, we show that parasite glycoconjugates mediate the modulation of LPS-induced maturation of DCs since their oxidation restores the capacity of LPS-treated DCs to secrete high levels of the pro-inflammatory cytokines IL-6 and IL-12/23p40 and low levels of the anti-inflammatory cytokine IL-10. Inhibition assays using carbohydrates suggest that the immune-modulation is mediated, at least in part, by the recognition of a mannose specific-CLR that signals by recruiting the phosphatase Php2. The results presented here contribute to the understanding of the role of parasite glycosylated molecules in the modulation of the host immunity and might be useful in the design of vaccines against fasciolosis.
Collapse
Affiliation(s)
- Ernesto Rodríguez
- Laboratory of Immunomodulation and Vaccine Development, Departamento de Inmunobiología, Facultad de Medicina, UdelaR, Montevideo, Uruguay
| | - Verónica Noya
- Laboratory of Immunomodulation and Vaccine Development, Departamento de Inmunobiología, Facultad de Medicina, UdelaR, Montevideo, Uruguay
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, CIBICI-CONICET, Córdoba, Argentina
| | | | - Natalie Brossard
- Laboratory of Immunomodulation and Vaccine Development, Departamento de Inmunobiología, Facultad de Medicina, UdelaR, Montevideo, Uruguay
| | - Carolina Chiale
- Laboratory of Immunomodulation and Vaccine Development, Departamento de Inmunobiología, Facultad de Medicina, UdelaR, Montevideo, Uruguay
| | - Carlos Carmona
- Unidad de Biología Parasitaria, Departamento de Biología Celular y Molecular, Instituto de Higiene, Facultad de Ciencias, UdelaR, Montevideo, Uruguay
| | - Cecilia Giacomini
- Cátedra de Bioquímica, Departamento de Biociencias, Facultad de Química, UdelaR, Montevideo, Uruguay
| | - Teresa Freire
- Laboratory of Immunomodulation and Vaccine Development, Departamento de Inmunobiología, Facultad de Medicina, UdelaR, Montevideo, Uruguay
- * E-mail:
| |
Collapse
|
35
|
Smit CH, Homann A, van Hensbergen VP, Schramm G, Haas H, van Diepen A, Hokke CH. Surface expression patterns of defined glycan antigens change duringSchistosoma mansonicercarial transformation and development of schistosomula. Glycobiology 2015; 25:1465-79. [DOI: 10.1093/glycob/cwv066] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/12/2015] [Indexed: 01/28/2023] Open
|
36
|
Sanin DE, Prendergast CT, Mountford AP. IL-10 Production in Macrophages Is Regulated by a TLR-Driven CREB-Mediated Mechanism That Is Linked to Genes Involved in Cell Metabolism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:1218-32. [PMID: 26116503 PMCID: PMC4505959 DOI: 10.4049/jimmunol.1500146] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/27/2015] [Indexed: 12/14/2022]
Abstract
IL-10 is produced by macrophages in diverse immune settings and is critical in limiting immune-mediated pathology. In helminth infections, macrophages are an important source of IL-10; however, the molecular mechanism underpinning production of IL-10 by these cells is poorly characterized. In this study, bone marrow-derived macrophages exposed to excretory/secretory products released by Schistosoma mansoni cercariae rapidly produce IL-10 as a result of MyD88-mediated activation of MEK/ERK/RSK and p38. The phosphorylation of these kinases was triggered by TLR2 and TLR4 and converged on activation of the transcription factor CREB. Following phosphorylation, CREB is recruited to a novel regulatory element in the Il10 promoter and is also responsible for regulating a network of genes involved in metabolic processes, such as glycolysis, the tricarboxylic acid cycle, and oxidative phosphorylation. Moreover, skin-resident tissue macrophages, which encounter S. mansoni excretory/secretory products during infection, are the first monocytes to produce IL-10 in vivo early postinfection with S. mansoni cercariae. The early and rapid release of IL-10 by these cells has the potential to condition the dermal microenvironment encountered by immune cells recruited to this infection site, and we propose a mechanism by which CREB regulates the production of IL-10 by macrophages in the skin, but also has a major effect on their metabolic state.
Collapse
Affiliation(s)
- David E Sanin
- Department of Biology, Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| | - Catriona T Prendergast
- Department of Biology, Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| | - Adrian P Mountford
- Department of Biology, Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| |
Collapse
|
37
|
Schistosoma mansoni Soluble Egg Antigens Induce Expression of the Negative Regulators SOCS1 and SHP1 in Human Dendritic Cells via Interaction with the Mannose Receptor. PLoS One 2015; 10:e0124089. [PMID: 25897665 PMCID: PMC4405200 DOI: 10.1371/journal.pone.0124089] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/25/2015] [Indexed: 12/31/2022] Open
Abstract
Schistosomiasis is a common debilitating human parasitic disease in (sub)tropical areas, however, schistosome infections can also protect against a variety of inflammatory diseases. This has raised broad interest in the mechanisms by which Schistosoma modulate the immune system into an anti-inflammatory and regulatory state. Human dendritic cells (DCs) show many phenotypic changes upon contact with Schistosoma mansoni soluble egg antigens (SEA). We here show that oxidation of SEA glycans, but not heat-denaturation, abrogates the capacity of SEA to suppress both LPS-induced cytokine secretion and DC proliferation, indicating an important role of SEA glycans in these processes. Remarkably, interaction of SEA glycans with DCs results in a strongly increased expression of Suppressor Of Cytokine Signalling1 (SOCS1) and SH2-containing protein tyrosine Phosphatase-1 (SHP1), important negative regulators of TLR4 signalling. In addition, SEA induces the secretion of transforming growth factor β (TGF-β), and the surface expression of the costimulatory molecules Programmed Death Ligand-1 (PD-L1) and OX40 ligand (OX40L), which are known phenotypic markers for the capacity of DCs to polarize naïve T cells into Th2/Treg cell subsets. Inhibition of mannose receptor (MR)-mediated internalization of SEA into DCs by blocking with allyl α-D-mannoside or anti-MR antibodies, significantly reduced SOCS1 and SHP1 expression. In conclusion, we demonstrate that SEA glycans are essential for induction of enhanced SOCS1 and SHP1 levels in DCs via the MR. Our data provide novel mechanistic evidence for the potential of S. mansoni SEA glycans to modulate human DCs, which may contribute to the capacity of SEA to down-regulate inflammatory responses.
Collapse
|
38
|
Smit CH, van Diepen A, Nguyen DL, Wuhrer M, Hoffmann KF, Deelder AM, Hokke CH. Glycomic Analysis of Life Stages of the Human Parasite Schistosoma mansoni Reveals Developmental Expression Profiles of Functional and Antigenic Glycan Motifs. Mol Cell Proteomics 2015; 14:1750-69. [PMID: 25883177 PMCID: PMC4587318 DOI: 10.1074/mcp.m115.048280] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 11/30/2022] Open
Abstract
Glycans present on glycoproteins and glycolipids of the major human parasite Schistosoma mansoni induce innate as well as adaptive immune responses in the host. To be able to study the molecular characteristics of schistosome infections it is therefore required to determine the expression profiles of glycans and antigenic glycan-motifs during a range of critical stages of the complex schistosome lifecycle. We performed a longitudinal profiling study covering schistosome glycosylation throughout worm- and egg-development using a mass spectrometry-based glycomics approach. Our study revealed that during worm development N-glycans with Galβ1–4(Fucα1–3)GlcNAc (LeX) and core-xylose motifs were rapidly lost after cercariae to schistosomula transformation, whereas GalNAcβ1–4GlcNAc (LDN)-motifs gradually became abundant and predominated in adult worms. LeX-motifs were present on glycolipids up to 2 weeks of schistosomula development, whereas glycolipids with mono- and multifucosylated LDN-motifs remained present up to the adult worm stage. In contrast, expression of complex O-glycans diminished to undetectable levels within days after transformation. During egg development, a rich diversity of N-glycans with fucosylated motifs was expressed, but with α3-core fucose and a high degree of multifucosylated antennae only in mature eggs and miracidia. N-glycan antennae were exclusively LDN-based in miracidia. O-glycans in the mature eggs were also diverse and contained LeX- and multifucosylated LDN, but none of these were associated with miracidia in which we detected only the Galβ1–3(Galβ1–6)GalNAc core glycan. Immature eggs also exhibited short O-glycan core structures only, suggesting that complex fucosylated O-glycans of schistosome eggs are derived primarily from glycoproteins produced by the subshell envelope in the developed egg. Lipid glycans with multifucosylated GlcNAc repeats were present throughout egg development, but with the longer highly fucosylated stretches enriched in mature eggs and miracidia. This global analysis of the developing schistosome's glycome provides new insights into how stage-specifically expressed glycans may contribute to different aspects of schistosome-host interactions.
Collapse
Affiliation(s)
- Cornelis H Smit
- From the ‡Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Angela van Diepen
- From the ‡Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - D Linh Nguyen
- From the ‡Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Manfred Wuhrer
- §Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Karl F Hoffmann
- ¶Institute of Biological Environmental and Rural Sciences (IBERS), Aberystwyth University, Penglais Campus, Aberystwyth SY23 3FG, United Kingdom
| | - André M Deelder
- §Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Cornelis H Hokke
- From the ‡Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| |
Collapse
|
39
|
Biedroń R, Konopiński MK, Marcinkiewicz J, Józefowski S. Oxidation by neutrophils-derived HOCl increases immunogenicity of proteins by converting them into ligands of several endocytic receptors involved in antigen uptake by dendritic cells and macrophages. PLoS One 2015; 10:e0123293. [PMID: 25849867 PMCID: PMC4388828 DOI: 10.1371/journal.pone.0123293] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/26/2015] [Indexed: 11/21/2022] Open
Abstract
The initiation of adaptive immune responses to protein antigens has to be preceded by their uptake by antigen presenting cells and intracellular proteolytic processing. Paradoxically, endocytic receptors involved in antigen uptake do not bind the majority of proteins, which may be the main reason why purified proteins stimulate at most weak immune responses. A shared feature of different types of adjuvants, capable of boosting immunogenicity of protein vaccines, is their ability to induce acute inflammation, characterized by early influx of activated neutrophils. Neutrophils are also rapidly recruited to sites of tissue injury or infection. These cells are the source of potent oxidants, including hypochlorous acid (HOCl), causing oxidation of proteins present in inflammatory foci. We demonstrate that oxidation of proteins by endogenous, neutrophils-derived HOCl increases their immunogenicity. Upon oxidation, different, randomly chosen simple proteins (yeast alcohol dehydrogenase, human and bovine serum albumin) and glycoproteins (human apo-transferrin, ovalbumin) gain the ability to bind with high affinity to several endocytic receptors on antigen presenting cells, which seems to be the major mechanism of their increased immunogenicity. The mannose receptor (CD206), scavenger receptors A (CD204) and CD36 were responsible for the uptake and presentation of HOCl-modified proteins by murine dendritic cells and macrophages. Other scavenger receptors, SREC-I and LOX-1, as well as RAGE were also able to bind HOCl-modified proteins, but they did not contribute significantly to these ligands uptake by dendritic cells because they were either not expressed or exhibited preference for more heavily oxidised proteins. Our results indicate that oxidation by neutrophils-derived HOCl may be a physiological mechanism of conferring immunogenicity on proteins which in their native forms do not bind to endocytic receptors. This mechanism might enable the immune system to detect infections caused by pathogens not recognized by pattern recognition receptors.
Collapse
Affiliation(s)
- Rafał Biedroń
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | | | - Janusz Marcinkiewicz
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
| | - Szczepan Józefowski
- Department of Immunology, Jagiellonian University Medical College, Cracow, Poland
- * E-mail:
| |
Collapse
|
40
|
Zhao X, Liu L, Hegazy AM, Wang H, Li J, Zheng F, Zhou Y, Wang W, Li J, Liu X, Lin L. Mannose receptor mediated phagocytosis of bacteria in macrophages of blunt snout bream (Megalobrama amblycephala) in a Ca(2+)-dependent manner. FISH & SHELLFISH IMMUNOLOGY 2015; 43:357-363. [PMID: 25583544 DOI: 10.1016/j.fsi.2015.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/30/2014] [Accepted: 01/02/2015] [Indexed: 06/04/2023]
Abstract
Mannose receptor (MR) is an important pattern-recognition receptor in macrophages and plays a critical role in immune responses. It is has been reported that mammalian macrophages are able to engulf a wide range of microorganisms mediated by Ca(2+)-dependent MR binding to terminal mannose residues which are frequently found on the pathogen surfaces. However, little is known about the MR-mediated phagocytosis in macrophages of fish. In this report, the distributions of MR in the macrophage and head kidney tissue from blunt snout bream were examined using MaMR specific antibody generated in our lab. Mannan and MaMR specific antibody inhibition experiments results collectively showed that MR was involved in the GFP-expressed E. coli engulfed in the macrophages, resulting in respiratory burst, nitric oxide production as well as inflammatory cytokines secretion, and the MaMR-mediated phagocytosis was Ca(2+)-dependent. These results will shed a new light on the immune functions of teleost MRs.
Collapse
Affiliation(s)
- Xiaoheng Zhao
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, Hubei 430070, China
| | - Lichun Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Abeer M Hegazy
- Central Laboratory for Environmental Quality Monitoring (CLEQM), National Water Research Center (NWRC), 13621, Egypt
| | - Hong Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jie Li
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Feifei Zheng
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yang Zhou
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, Hubei 430070, China
| | - Weimin Wang
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, Hubei 430070, China
| | - Jun Li
- School of Biological Sciences, Lake Superior State University, Sault Ste. Marie 49783, MI, USA
| | - Xiaoling Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, Hubei 430070, China.
| | - Li Lin
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, Hubei 430070, China; Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, Hubei 430070, China.
| |
Collapse
|
41
|
CD4+ T cell hyporesponsiveness after repeated exposure to Schistosoma mansoni larvae is dependent upon interleukin-10. Infect Immun 2015; 83:1418-30. [PMID: 25624353 PMCID: PMC4363412 DOI: 10.1128/iai.02831-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The effect that multiple percutaneous exposures to Schistosoma larvae has on the development of early CD4+ lymphocyte reactivity is unclear, yet it is important in the context of humans living in areas where schistosomiasis is endemic. In a murine model of multiple infections, we show that exposure of mice to repeated doses (4×) of Schistosoma mansoni cercariae, compared to a single dose (1×), results in CD4+ T cell hyporesponsiveness within the skin-draining lymph nodes (sdLN), manifested as reduced CD4+ cell proliferation and cytokine production. FoxP3+ CD4+ regulatory T cells were present in similar numbers in the sdLN of 4× and 1× mice and thus are unlikely to have a role in effecting hyporesponsiveness. Moreover, anergy of the CD4+ cell population from 4× mice was slight, as proliferation was only partly circumvented through the in vitro addition of exogenous interleukin-2 (IL-2), and the in vivo blockade of the regulatory molecule PD1 had a minimal effect on restoring responsiveness. In contrast, IL-10 was observed to be critical in mediating hyporesponsiveness, as CD4+ cells from the sdLN of 4× mice deficient for IL-10 were readily able to proliferate, unlike those from 4× wild-type cohorts. CD4+ cells from the sdLN of 4× mice exhibited higher levels of apoptosis and cell death, but in the absence of IL-10, there was significantly less cell death. Combined, our data show that IL-10 is a key factor in the development of CD4+ T cell hyporesponsiveness after repeated parasite exposure involving CD4+ cell apoptosis.
Collapse
|
42
|
The macrophage galactose-type lectin-1 (MGL1) recognizes Taenia crassiceps antigens, triggers intracellular signaling, and is critical for resistance to this infection. BIOMED RESEARCH INTERNATIONAL 2015; 2015:615865. [PMID: 25664320 PMCID: PMC4312580 DOI: 10.1155/2015/615865] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 01/01/2023]
Abstract
C-type lectins are multifunctional sugar-binding molecules expressed on dendritic cells (DCs) and macrophages that internalize antigens for processing and presentation. Macrophage galactose-type lectin 1 (MGL1) recognizes glycoconjugates expressing Lewis X structures which contain galactose residues, and it is selectively expressed on immature DCs and macrophages. Helminth parasites contain large amounts of glycosylated components, which play a role in the immune regulation induced by such infections. Macrophages from MGL1−/− mice showed less binding ability toward parasite antigens than their wild-type (WT) counterparts. Exposure of WT macrophages to T. crassiceps antigens triggered tyrosine phosphorylation signaling activity, which was diminished in MGL1−/− macrophages. Following T. crassiceps infection, MGL1−/− mice failed to produce significant levels of inflammatory cytokines early in the infection compared to WT mice. In contrast, MGL1−/− mice developed a Th2-dominant immune response that was associated with significantly higher parasite loads, whereas WT mice were resistant. Flow cytometry and RT-PCR analyses showed overexpression of the mannose receptors, IL-4Rα, PDL2, arginase-1, Ym1, and RELM-α on MGL1−/− macrophages. These studies indicate that MGL1 is involved in T. crassiceps recognition and subsequent innate immune activation and resistance.
Collapse
|
43
|
Sanin DE, Mountford AP. Sm16, a major component of Schistosoma mansoni cercarial excretory/secretory products, prevents macrophage classical activation and delays antigen processing. Parasit Vectors 2015; 8:1. [PMID: 25561160 PMCID: PMC4297449 DOI: 10.1186/s13071-014-0608-1] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 12/16/2014] [Indexed: 11/10/2022] Open
Abstract
Background Schistosoma mansoni cercariae penetrate the skin by releasing excretory/secretory (E/S) products known as 0-3hRP, which are associated with immune modulation through Toll like receptor (TLR) signalling. Furthermore, these secretions contain Sm16, which when given to cells as a recombinant protein inhibits human monocyte derived cytokine responses to TLR4 and TLR3 ligands. Nonetheless, the extent and mechanism(s) of these inhibitory effects remain largely uncharacterized. Methods Murine bone marrow derived macrophages were exposed to different fractions of 0-3hRP, obtained via ultracentrifugation, or recombinant Sm16. These cells were exposed to the parasite molecules in combination with different TLR ligands, or Interferon gamma, and tested for the production of the cytokines IL-10 and IL-12p40, and their ability to process antigen. Results The immunomodulatory function of 0-3hRP is enriched predominantly in the pellet fraction, which contains a greater proportion of Sm16, also corroborating the ability of recombinant Sm16 to inhibit macrophage activation in response to TLR ligands. We further demonstrate that Sm16 blocks classical activation of macrophages to LPS or IFN-γ stimulation in vitro, and that inhibition of macrophage classical activation is independent of TLR2 recognition. Finally we show that Sm16 shares the altered intracellular processing observed for 0-3hRP, and is able to delay antigen processing by macrophages. Conclusions Collectively, our findings show that Sm16 is a major component of S. mansoni cercarial E/S products, and is partly responsible for its immune-regulatory properties. Moreover, we propose that the mechanism employed by Sm16 to exert its inhibitory function is likely to be linked with alteration of endosomal trafficking and is not dependent on particular TLR receptors. Finally, we suggest that accumulation of Sm16 in the skin after percutaneous infection with S. mansoni cercariae could contribute to limiting dermal inflammation. Electronic supplementary material The online version of this article (doi:10.1186/s13071-014-0608-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David E Sanin
- Centre for Immunology and Infection, Department of Biology, University of York, York, UK.
| | - Adrian P Mountford
- Centre for Immunology and Infection, Department of Biology, University of York, York, UK.
| |
Collapse
|
44
|
Bourke CD, Prendergast CT, Sanin DE, Oulton TE, Hall RJ, Mountford AP. Epidermal keratinocytes initiate wound healing and pro-inflammatory immune responses following percutaneous schistosome infection. Int J Parasitol 2015; 45:215-24. [PMID: 25575749 PMCID: PMC4365920 DOI: 10.1016/j.ijpara.2014.11.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 11/17/2014] [Accepted: 11/18/2014] [Indexed: 10/28/2022]
Abstract
Keratinocytes constitute the majority of cells in the skin's epidermis, the first line of defence against percutaneous pathogens. Schistosome larvae (cercariae) actively penetrate the epidermis to establish infection, however the response of keratinocytes to invading cercariae has not been investigated. Here we address the hypothesis that cercariae activate epidermal keratinocytes to promote the development of a pro-inflammatory immune response in the skin. C57BL/6 mice were exposed to Schistosoma mansoni cercariae via each pinna and non-haematopoietic cells isolated from epidermal tissue were characterised for the presence of different keratinocyte sub-sets at 6, 24 and 96 h p.i. We identified an expansion of epidermal keratinocyte precursors (CD45(-), CD326(-), CD34(+)) within 24 h of infection relative to naïve animals. Following infection, cells within the precursor population displayed a more differentiated phenotype (α6integrin(-)) than in uninfected skin. Parallel immunohistochemical analysis of pinnae cryosections showed that this expansion corresponded to an increase in the intensity of CD34 staining, specifically in the basal bulge region of hair follicles of infected mice, and a higher frequency of keratinocyte Ki67(+) nuclei in both the hair follicle and interfollicular epidermis. Expression of pro-inflammatory cytokine and stress-associated keratin 6b genes was also transiently upregulated in the epidermal tissue of infected mice. In vitro exposure of keratinocyte precursors isolated from neonatal mouse skin to excretory/secretory antigens released by penetrating cercariae elicited IL-1α and IL-1β production, supporting a role for keratinocyte precursors in initiating cutaneous inflammatory immune responses. Together, these observations indicate that S.mansoni cercariae and their excretory/secretory products act directly upon epidermal keratinocytes, which respond by initiating barrier repair and pro-inflammatory mechanisms similar to those observed in epidermal wound healing.
Collapse
Affiliation(s)
- Claire D Bourke
- Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom.
| | | | - David E Sanin
- Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| | - Tate E Oulton
- Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| | - Rebecca J Hall
- Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| | - Adrian P Mountford
- Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| |
Collapse
|
45
|
Suabjakyong P, Nishimura K, Toida T, Van Griensven LJLD. Structural characterization and immunomodulatory effects of polysaccharides from Phellinus linteus and Phellinus igniarius on the IL-6/IL-10 cytokine balance of the mouse macrophage cell lines (RAW 264.7). Food Funct 2015; 6:2834-44. [DOI: 10.1039/c5fo00491h] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Phellinus linteus and igniarius (L.) Quel. have been used in traditional Asian medicine for over two centuries against a variety of diseases.
Collapse
Affiliation(s)
- Papawee Suabjakyong
- Department of Clinical and Analytical Biochemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | - Kazuhiro Nishimura
- Department of Clinical and Analytical Biochemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | - Toshihiko Toida
- Department of Clinical and Analytical Biochemistry
- Graduate School of Pharmaceutical Sciences
- Chiba University
- Chiba
- Japan
| | | |
Collapse
|
46
|
Knight M, Arican-Goktas HD, Ittiprasert W, Odoemelam EC, Miller AN, Bridger JM. Schistosomes and snails: a molecular encounter. Front Genet 2014; 5:230. [PMID: 25101114 PMCID: PMC4104801 DOI: 10.3389/fgene.2014.00230] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 06/30/2014] [Indexed: 11/13/2022] Open
Abstract
Biomphalaria glabrata snails play an integral role in the transmission of Schistosoma mansoni, the causative agent for human schistosomiasis in the Western hemisphere. For the past two decades, tremendous advances have been made in research aimed at elucidating the molecular basis of the snail/parasite interaction. The growing concern that there is no vaccine to prevent schistosomiasis and only one effective drug in existence provides the impetus to develop new control strategies based on eliminating schistosomes at the snail-stage of the life cycle. To elucidate why a given snail is not always compatible to each and every schistosome it encounters, B. glabrata that are either resistant or susceptible to a given strain of S. mansoni have been employed to track molecular mechanisms governing the snail/schistosome relationship. With such snails, genetic markers for resistance and susceptibility were identified. Additionally, differential gene expression studies have led to the identification of genes that underlie these phenotypes. Lately, the role of schistosomes in mediating non-random relocation of gene loci has been identified for the first time, making B. glabrata a model organism where chromatin regulation by changes in nuclear architecture, known as spatial epigenetics, orchestrated by a major human parasite can now be investigated. This review will highlight the progress that has been made in using molecular approaches to describe snail/schistosome compatibility issues. Uncovering the signaling networks triggered by schistosomes that provide the impulse to turn genes on and off in the snail host, thereby controlling the outcome of infection, could also yield new insights into anti-parasite mechanism(s) that operate in the human host as well.
Collapse
Affiliation(s)
- Matty Knight
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University Washington, DC, USA
| | | | | | - Edwin C Odoemelam
- Biosciences, Health Sciences and Social Care, Brunel University London London, UK
| | - André N Miller
- Schistosomiasis, Biomedical Research Institute Rockville, MD, USA
| | - Joanna M Bridger
- Biosciences, Health Sciences and Social Care, Brunel University London London, UK
| |
Collapse
|
47
|
Trichinella spiralis excretory-secretory products protect against polymicrobial sepsis by suppressing MyD88 via mannose receptor. BIOMED RESEARCH INTERNATIONAL 2014; 2014:898646. [PMID: 25054155 PMCID: PMC4098621 DOI: 10.1155/2014/898646] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/10/2014] [Accepted: 06/15/2014] [Indexed: 12/31/2022]
Abstract
Trichinella spiralis (T. spiralis) or its excretory-secretory products (TsES) protect hosts from autoimmune diseases, which depend on inducing host T helper (Th) 2 immune response and inhibiting inflammatory factors. Sepsis is a systemic inflammatory response syndrome (SIRS) evoked by infection. Little is known about the effects of helminths or their excretory-secretory products on sepsis. Here, we investigated the effects of TsES in a mice model of polymicrobial sepsis. TsES improved survival, reduced organ injury, and enhanced bacterial clearance in septic mice. To investigate the molecular mechanism, macrophages from septic patients or the control group were incubated with TsES. TsES reduced sepsis-inducing inflammatory cytokines mediated by Toll-like receptors (TLR) in vitro by suppressing TLR adaptor-transducer myeloid differentiation factor 88 (MyD88) and nuclear factor- (NF-)-κB. Furthermore, TsES upregulated mannose receptor (MR) expression during sepsis. MR blocking attenuated the effects of TsES on MyD88 and NF-κB expression. In vivo, MR RNAi reduced the survival rate of septic mice treated with TsES, suggesting that TsES-mediated protection against polymicrobial sepsis is dependent on MR. Thus, TsES administration might be a potential therapeutic strategy for treating sepsis.
Collapse
|
48
|
Turner JD, Bourke CD, Meurs L, Mbow M, Dièye TN, Mboup S, Polman K, Mountford AP. Circulating CD14brightCD16+ 'intermediate' monocytes exhibit enhanced parasite pattern recognition in human helminth infection. PLoS Negl Trop Dis 2014; 8:e2817. [PMID: 24762736 PMCID: PMC3998941 DOI: 10.1371/journal.pntd.0002817] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/11/2014] [Indexed: 12/17/2022] Open
Abstract
Circulating monocyte sub-sets have recently emerged as mediators of divergent immune functions during infectious disease but their role in helminth infection has not been investigated. In this study we evaluated whether ‘classical’ (CD14brightCD16−), ‘intermediate’ (CD14brightCD16+), and ‘non-classical’ (CD14dimCD16+) monocyte sub-sets from peripheral blood mononuclear cells varied in both abundance and ability to bind antigenic material amongst individuals living in a region of Northern Senegal which is co-endemic for Schistosoma mansoni and S. haematobium. Monocyte recognition of excretory/secretory (E/S) products released by skin-invasive cercariae, or eggs, of S. mansoni was assessed by flow cytometry and compared between S. mansoni mono-infected, S. mansoni and S. haematobium co-infected, and uninfected participants. Each of the three monocyte sub-sets in the different infection groups bound schistosome E/S material. However, ‘intermediate’ CD14brightCD16+ monocytes had a significantly enhanced ability to bind cercarial and egg E/S. Moreover, this elevation of ligand binding was particularly evident in co-infected participants. This is the first demonstration of modulated parasite pattern recognition in CD14brightCD16+ intermediate monocytes during helminth infection, which may have functional consequences for the ability of infected individuals to respond immunologically to infection. The parasite Schistosoma infects over 200 million people world-wide and can cause serious morbidity. Infection occurs following exposure to larvae (cercariae) which release excretory/secretory (E/S) material to aid their entry into exposed skin. Larvae mature into adult worms that produce hundreds of eggs per day which also release E/S material. Both sources of E/S material have the potential to stimulate the host’s innate immune system. Circulating monocytes are important cells that act as potential sentinels in the recognition of these E/S materials. Different sub-sets of human monocytes can be identified according to their expression of CD14 and CD16 but their role following infection with schistosome helminths has not been investigated. In the current study, three sub-sets (classical, intermediate and non-classical) were enumerated in individuals living in a region co-endemic for S. mansoni and S. haematobium. Although all three monocyte sub-sets bound to fluorescently-labelled schistosome E/S material, the intermediate sub-set had significantly enhanced ability to recognise cercarial and egg E/S in co-infected participants. This is the first demonstration that circulating human monocytes can recognize schistosome E/S antigens and that their ability to do so is modulated by infection which may affect the development of schistosome immunopathology and/or protective immunity.
Collapse
Affiliation(s)
- Joseph D Turner
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| | - Claire D Bourke
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| | - Lynn Meurs
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Moustapha Mbow
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium; Immunology Department of the Laboratory of Bacteriology and Virology of Aristide Le Dantec University Hospital, Dakar, Senegal
| | - Tandakha Ndiaye Dièye
- Immunology Department of the Laboratory of Bacteriology and Virology of Aristide Le Dantec University Hospital, Dakar, Senegal
| | - Souleymane Mboup
- Immunology Department of the Laboratory of Bacteriology and Virology of Aristide Le Dantec University Hospital, Dakar, Senegal
| | - Katja Polman
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Adrian P Mountford
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| |
Collapse
|
49
|
Du L, Wei H, Li L, Shan H, Yu Y, Wang Y, Zhang G. Regulation of recombinant Trichinella spiralis 53-kDa protein (rTsP53) on alternatively activated macrophages via STAT6 but not IL-4Rα in vitro. Cell Immunol 2014; 288:1-7. [DOI: 10.1016/j.cellimm.2014.01.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/16/2013] [Accepted: 01/28/2014] [Indexed: 12/26/2022]
|
50
|
Jang JC, Nair MG. Alternatively Activated Macrophages Revisited: New Insights into the Regulation of Immunity, Inflammation and Metabolic Function following Parasite Infection. ACTA ACUST UNITED AC 2014; 9:147-156. [PMID: 24772059 DOI: 10.2174/1573395509666131210232548] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The role of macrophages in homeostatic conditions and the immune system range from clearing debris to recognizing and killing pathogens. While classically activated macrophages (CAMacs) are induced by T helper type 1 (Th1) cytokines and exhibit microbicidal properties, Th2 cytokines promote alternative activation of macrophages (AAMacs). AAMacs contribute to the killing of helminth parasites and mediate additional host-protective processes such as regulating inflammation and wound healing. Yet, other parasites susceptible to Th1 type responses can exploit alternative activation of macrophages to diminish Th1 immune responses and prolong infection. In this review, we will delineate the factors that mediate alternative activation (e.g. Th2 cytokines and chitin) and the resulting downstream signaling events (e.g. STAT6 signaling). Next, the specific AAMac-derived factors (e.g. Arginase1) that contribute to resistance or susceptibility to parasitic infections will be summarized. Finally, we will conclude with the discussion of additional AAMac functions beyond immunity to parasites, including the regulation of inflammation, wound healing and the regulation of metabolic disorders.
Collapse
Affiliation(s)
- Jessica C Jang
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA
| |
Collapse
|