1
|
Inclan-Rico JM, Napuri CM, Lin C, Hung LY, Ferguson AA, Liu X, Wu Q, Pastore CF, Stephenson A, Femoe UM, Musaigwa F, Rossi HL, Freedman BD, Reed DR, Macháček T, Horák P, Abdus-Saboor I, Luo W, Herbert DR. MrgprA3 neurons drive cutaneous immunity against helminths through selective control of myeloid-derived IL-33. Nat Immunol 2024; 25:2068-2084. [PMID: 39354200 DOI: 10.1038/s41590-024-01982-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 09/11/2024] [Indexed: 10/03/2024]
Abstract
Skin uses interdependent cellular networks for barrier integrity and host immunity, but most underlying mechanisms remain obscure. Herein, we demonstrate that the human parasitic helminth Schistosoma mansoni inhibited pruritus evoked by itch-sensing afferents bearing the Mas-related G-protein-coupled receptor A3 (MrgprA3) in mice. MrgprA3 neurons controlled interleukin (IL)-17+ γδ T cell expansion, epidermal hyperplasia and host resistance against S. mansoni through shaping cytokine expression in cutaneous antigen-presenting cells. MrgprA3 neuron activation downregulated IL-33 but induced IL-1β and tumor necrosis factor in macrophages and type 2 conventional dendritic cells partially through the neuropeptide calcitonin gene-related peptide. Macrophages exposed to MrgprA3-derived secretions or bearing cell-intrinsic IL-33 deletion showed increased chromatin accessibility at multiple inflammatory cytokine loci, promoting IL-17/IL-23-dependent changes to the epidermis and anti-helminth resistance. This study reveals a previously unrecognized intercellular communication mechanism wherein itch-inducing MrgprA3 neurons initiate host immunity against skin-invasive parasites by directing cytokine expression patterns in myeloid antigen-presenting cell subsets.
Collapse
Affiliation(s)
- Juan M Inclan-Rico
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Camila M Napuri
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cailu Lin
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Li-Yin Hung
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Annabel A Ferguson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaohong Liu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qinxue Wu
- Department of Neuroscience, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher F Pastore
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adriana Stephenson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ulrich M Femoe
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fungai Musaigwa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather L Rossi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Tomáš Macháček
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Horák
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Ishmail Abdus-Saboor
- Department of Biological Sciences, Zuckerman Mind, Brain, Behavior Institute, Columbia University, New York, NY, USA
| | - Wenqin Luo
- Department of Neuroscience, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Inclan-Rico JM, Napuri CM, Lin C, Hung LY, Ferguson AA, Wu Q, Pastore CF, Stephenson A, Femoe UM, Rossi HL, Reed DR, Luo W, Abdus-Saboor I, Herbert DR. "MrgprA3 neurons selectively control myeloid-derived cytokines for IL-17 dependent cutaneous immunity". RESEARCH SQUARE 2023:rs.3.rs-3644984. [PMID: 38076920 PMCID: PMC10705600 DOI: 10.21203/rs.3.rs-3644984/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Skin employs interdependent cellular networks to facilitate barrier integrity and host immunity through ill-defined mechanisms. This study demonstrates that manipulation of itch-sensing neurons bearing the Mas-related G protein-coupled receptor A3 (MrgprA3) drives IL-17+ γδ T cell expansion, epidermal thickening, and resistance to the human pathogen Schistosoma mansoni through mechanisms that require myeloid antigen presenting cells (APC). Activated MrgprA3 neurons instruct myeloid APCs to downregulate interleukin 33 (IL-33) and up-regulate TNFα partially through the neuropeptide calcitonin gene related peptide (CGRP). Strikingly, cell-intrinsic deletion of IL-33 in myeloid APC basally alters chromatin accessibility at inflammatory cytokine loci and promotes IL-17/23-dependent epidermal thickening, keratinocyte hyperplasia, and resistance to helminth infection. Our findings reveal a previously undescribed mechanism of intercellular cross-talk wherein "itch" neuron activation reshapes myeloid cytokine expression patterns to alter skin composition for cutaneous immunity against invasive pathogens.
Collapse
Affiliation(s)
- Juan M. Inclan-Rico
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Camila M. Napuri
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cailu Lin
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Li-Yin Hung
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Annabel A. Ferguson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Qinxue Wu
- Department of Neuroscience, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher F. Pastore
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adriana Stephenson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ulrich M. Femoe
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Heather L. Rossi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Wenqin Luo
- Department of Neuroscience, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Regenerative Medicine, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ishmail Abdus-Saboor
- Department of Biological Sciences, Zuckerman Mind, Brain, Behavior Institute, Columbia University, New York, New York, USA
| | - De’Broski R. Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Licá ICL, Frazão GCCG, Nogueira RA, Lira MGS, dos Santos VAF, Rodrigues JGM, Miranda GS, Carvalho RC, Silva LA, Guerra RNM, Nascimento FRF. Immunological mechanisms involved in macrophage activation and polarization in schistosomiasis. Parasitology 2023; 150:401-415. [PMID: 36601859 PMCID: PMC10089811 DOI: 10.1017/s0031182023000021] [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: 05/18/2022] [Revised: 12/21/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023]
Abstract
Human schistosomiasis is caused by helminths of the genus Schistosoma. Macrophages play a crucial role in the immune regulation of this disease. These cells acquire different phenotypes depending on the type of stimulus they receive. M1 macrophages can be ‘classically activated’ and can display a proinflammatory phenotype. M2 or ‘alternatively activated’ macrophages are considered anti-inflammatory cells. Despite the relevance of macrophages in controlling infections, the role of the functional types of these cells in schistosomiasis is unclear. This review highlights different molecules and/or macrophage activation and polarization pathways during Schistosoma mansoni and Schistosoma japonicum infection. This review is based on original and review articles obtained through searches in major databases, including Scopus, Google Scholar, ACS, PubMed, Wiley, Scielo, Web of Science, LILACS and ScienceDirect. Our findings emphasize the importance of S. mansoni and S. japonicum antigens in macrophage polarization, as they exert immunomodulatory effects in different stages of the disease and are therefore important as therapeutic targets for schistosomiasis and in vaccine development. A combination of different antigens can provide greater protection, as it possibly stimulates an adequate immune response for an M1 or M2 profile and leads to host resistance; however, this warrants in vitro and in vivo studies.
Collapse
Affiliation(s)
- Irlla Correia Lima Licá
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Gleycka Cristine Carvalho Gomes Frazão
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Ranielly Araujo Nogueira
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Maria Gabriela Sampaio Lira
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Vitor Augusto Ferreira dos Santos
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - João Gustavo Mendes Rodrigues
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Guilherme Silva Miranda
- Department of Biology, Federal Institute of Education, Science and Technology of Maranhão, São Raimundo das Mangabeiras, Brazil
| | - Rafael Cardoso Carvalho
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Lucilene Amorim Silva
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Rosane Nassar Meireles Guerra
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Flávia Raquel Fernandes Nascimento
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| |
Collapse
|
4
|
da Silva JV, Moreira CC, Montija EDA, Feitosa KA, Correia RDO, Domingues NLDC, Soares EG, Allegretti SM, Afonso A, Anibal FDF. Schiff bases complexed with iron and their relation with the life cycle and infection by Schistosoma mansoni. Front Immunol 2022; 13:1021768. [PMID: 36618401 PMCID: PMC9811594 DOI: 10.3389/fimmu.2022.1021768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction The trematode Schistosoma mansoni causes schistosomiasis, and this parasite's life cycle depends on the mollusk Biomphalaria glabrata. The most effective treatment for infected people is administering a single dose of Praziquantel. However, there are naturally resistant to treatment. This work has developed, considering this parasite's complex life cycle. Methods The synthetics compound were evaluated: i) during the infection of B. glabrata, ii) during the infection of BALB/c mice, and iii) during the treatment of mice infected with S. mansoni. Results and Discussion For the first objective, snails infected with miracidia treated with compounds C1 and C3 at concentrations of 25% IC50 and 50% IC50, after 80 days of infection, released fewer cercariae than the infected group without treatment. For the second objective, compounds C1 and C3 did not show significant results in the infected group without treatment. For the third objective, the mice treated with C3 and C1 reduced the global and differential cell count. The results suggest that although the evaluated compounds do not present schistosomicidal properties when placed in cercariae suspension, they can stimulate an immune reaction in snails and decrease mice's inflammatory response. In general, we can conclude that compound C1 and C3 has an anti-schistosomicidal effect both in the larval phase (miracidia) and in the adult form of the parasite.
Collapse
Affiliation(s)
- Juliana Virginio da Silva
- Departamento de Morfologia e Patologia (DMP), Laboratório de Inflamação e Doenças Infecciosas (LIDI), Universidade Federal de São Carlos (UFSCar), São Paulo, Brazil,*Correspondence: Juliana Virginio da Silva,
| | - Carla Cristina Moreira
- Departamento de Morfologia e Patologia (DMP), Laboratório de Inflamação e Doenças Infecciosas (LIDI), Universidade Federal de São Carlos (UFSCar), São Paulo, Brazil
| | - Elisandra de Almeida Montija
- Departamento de Morfologia e Patologia (DMP), Laboratório de Inflamação e Doenças Infecciosas (LIDI), Universidade Federal de São Carlos (UFSCar), São Paulo, Brazil
| | - Karina Alves Feitosa
- Departamento de Morfologia e Patologia (DMP), Laboratório de Inflamação e Doenças Infecciosas (LIDI), Universidade Federal de São Carlos (UFSCar), São Paulo, Brazil
| | - Ricardo de Oliveira Correia
- Departamento de Morfologia e Patologia (DMP), Laboratório de Inflamação e Doenças Infecciosas (LIDI), Universidade Federal de São Carlos (UFSCar), São Paulo, Brazil
| | - Nelson Luis de Campos Domingues
- Laboratório de catálise orgânica e biocatálise, Universidade Federal da Grande Dourados, Dourados, Mato Grosso do Sul, Brazil
| | - Edson Garcia Soares
- Departamento de Patologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Silmara Marques Allegretti
- Departamento De Biologia Animal, Instituto de BiologiaEstadual de Campinas, Universidade, Campinas, São Paulo, Brazil
| | - Ana Afonso
- Departamento de Morfologia e Patologia (DMP), Laboratório de Inflamação e Doenças Infecciosas (LIDI), Universidade Federal de São Carlos (UFSCar), São Paulo, Brazil,Global Health and Tropical Medicine (GHTM), Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisboa, Portugal,Instituto de Química de São Carlos (IQSC), Universidade de São Paulo (USP), São Paulo, Brazil,Instituto Nacional de Investigação Agrária e Veterinária, I.P., (INIAV), Laboratório de Parasitologia, Oeiras, Portugal,Laboratório de Parasitologia, Quantoom’s Bioscience, Nivelles, Bélgica, Belgium
| | - Fernanda de Freitas Anibal
- Departamento de Morfologia e Patologia (DMP), Laboratório de Inflamação e Doenças Infecciosas (LIDI), Universidade Federal de São Carlos (UFSCar), São Paulo, Brazil
| |
Collapse
|
5
|
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
|
6
|
Ho CH, Cheng CH, Huang TW, Peng SY, Lee KM, Cheng PC. Switched phenotypes of macrophages during the different stages of Schistosoma japonicum infection influenced the subsequent trends of immune responses. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2021; 55:503-526. [PMID: 34330662 DOI: 10.1016/j.jmii.2021.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/28/2021] [Accepted: 06/24/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Macrophages play crucial roles in immune responses during the course of schistosomal infections. METHODS We currently investigated influence of immunocompetent changes in macrophages via microarray-based analysis, mRNA expression analysis, detection of serum cytokines, and subsequent evaluation of the immune phenotypes following the differentiation of infection-induced lymphocytes in a unique T1/T2 double-transgenic mouse model. RESULTS The gradual upregulation of genes encoding YM1, YM2, and interleukin (IL)-4/IL-13 receptors in infected mice indicated the role of type 2 alternatively activated macrophages (M2, AAMφs) in immune responses after Schistosoma japonicum egg production. FACS analysis showed that surface markers MHC class II (IA/IE) and CD8α+ of the macrophages also exhibited a dramatic change at the various time points before and after egg-production. The transgenic mouse experiments further demonstrated that the shifting of macrophage phenotypes influenced the percentage of helper T (Th)-2 cells, which was observed to be higher than that of Th1 cells, which increased only at 3 and 5 weeks post-infection. The differentiation of effector B cells showed a similar but more significant trend toward type-2 immunity. CONCLUSION These results suggest that the infection of mice with S. japonicum resulted in a final Th2- and Be2-skewed immune response. This may be due to phenotypic changes in the macrophages. The influence of alternatively activated macrophages was also activated by S. japonicum egg production. This study elucidated the existence of variations in immune mechanisms at the schistosome infection stages.
Collapse
Affiliation(s)
- Chen-Hsun Ho
- Division of Urology, Department of Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chia-Hsiung Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Wen Huang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Yi Peng
- Department of Biochemistry, College of Medicine, Tzu Chi University, Hualien, Taiwan.
| | - Kin-Mu Lee
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Po-Ching Cheng
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Center for International Tropical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
7
|
Miranda GS, Resende SD, Cardoso DT, Camelo GMA, Silva JKAO, de Castro VN, Geiger SM, Carneiro M, Negrão-Corrêa D. Previous History of American Tegumentary Leishmaniasis Alters Susceptibility and Immune Response Against Schistosoma mansoni Infection in Humans. Front Immunol 2021; 12:630934. [PMID: 33777015 PMCID: PMC7990892 DOI: 10.3389/fimmu.2021.630934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/18/2021] [Indexed: 01/21/2023] Open
Abstract
Schistosomiasis and Leishmaniasis are chronic parasitic diseases with high prevalence in some tropical regions and, due to their wide distribution, a risk of co-infections is present in some areas. Nevertheless, the impact of this interaction on human populations is still poorly understood. Thus, the current study evaluated the effect of previous American Tegumentary Leishmaniasis (ATL) on the susceptibility and immune response to Schistosoma mansoni infection in residents from a rural community in Northern of Minas Gerais state, Brazil, an area endemic for both parasitic infections. The participants answered a socioeconomic questionnaire and provided stool and blood samples for parasitological and immunological evaluations. Stool samples were examined by a combination of parasitological techniques to identify helminth infections, especially S. mansoni eggs. Blood samples were used for hemograms and to measure the serum levels of cytokines and chemokines. Reports on previous ATL were obtained through interviews, clinical evaluation forms, and medical records. S. mansoni infection was the most prevalent parasitic infection in the study population (46%), and the majority of the infected individuals had a very low parasite burden. In the same population, 93 individuals (36.2%) reported previous ATL, and the prevalence of S. mansoni infection among these individuals was significantly higher than among individuals with no ATL history. A multiple logistic regression model revealed that S. mansoni infection was positively associated with higher levels of CCL3 and CCL17, and a higher frequency of IL-17 responders. Moreover, this model demonstrated that individuals with an ATL history had a 2-fold higher probability to be infected with S. mansoni (OR = 2.0; 95% CI 1.04–3.68). Among S. mansoni-infected individuals, the logistic regression demonstrated that a previous ATL history was negatively associated with the frequency of IL-17 responders and CXCL10 higher responders, but positively associated with higher IL-27 responders. Altogether, our data suggest that previous ATL may alter the susceptibility and the immune response in S. mansoni-infected individuals, which may likely affect the outcome of schistosomiasis and the severity of the disease in humans.
Collapse
Affiliation(s)
- Guilherme Silva Miranda
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Biology, Department of Biology, Institute of Education, Science and Technology of Maranhão, São Raimundo das Mangabeiras, Brazil
| | - Samira Diniz Resende
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Diogo Tavares Cardoso
- Laboratory of Intestinal Helminthiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Genil Mororó Araújo Camelo
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jeferson Kelvin Alves Oliveira Silva
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Normandio de Castro
- Laboratory of Intestinal Helminthiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Stefan Michael Geiger
- Laboratory of Intestinal Helminthiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mariângela Carneiro
- Laboratory of Epidemiology of Infectious and Parasitic Diseases, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Deborah Negrão-Corrêa
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
8
|
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
|
9
|
Shiels J, Cwiklinski K, Alvarado R, Thivierge K, Cotton S, Gonzales Santana B, To J, Donnelly S, Taggart CC, Weldon S, Dalton JP. Schistosoma mansoni immunomodulatory molecule Sm16/SPO-1/SmSLP is a member of the trematode-specific helminth defence molecules (HDMs). PLoS Negl Trop Dis 2020; 14:e0008470. [PMID: 32644998 PMCID: PMC7373315 DOI: 10.1371/journal.pntd.0008470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/21/2020] [Accepted: 06/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Sm16, also known as SPO-1 and SmSLP, is a low molecular weight protein (~16kDa) secreted by the digenean trematode parasite Schistosoma mansoni, one of the main causative agents of human schistosomiasis. The molecule is secreted from the acetabular gland of the cercariae during skin invasion and is believed to perform an immune-suppressive function to protect the invading parasite from innate immune cell attack. METHODOLOGY/PRINCIPAL FINDINGS We show that Sm16 homologues of the Schistosomatoidea family are phylogenetically related to the helminth defence molecule (HDM) family of immunomodulatory peptides first described in Fasciola hepatica. Interrogation of 69 helminths genomes demonstrates that HDMs are exclusive to trematode species. Structural analyses of Sm16 shows that it consists predominantly of an amphipathic alpha-helix, much like other HDMs. In S. mansoni, Sm16 is highly expressed in the cercariae and eggs but not in adult worms, suggesting that the molecule is of importance not only during skin invasion but also in the pro-inflammatory response to eggs in the liver tissues. Recombinant Sm16 and a synthetic form, Sm16 (34-117), bind to macrophages and are internalised into the endosomal/lysosomal system. Sm16 (34-117) elicited a weak pro-inflammatory response in macrophages in vitro but also suppressed the production of bacterial lipopolysaccharide (LPS)-induced inflammatory cytokines. Evaluation of the transcriptome of human macrophages treated with a synthetic Sm16 (34-117) demonstrates that the peptide exerts significant immunomodulatory effects alone, as well as in the presence of LPS. Pathways most significantly influenced by Sm16 (34-117) were those involving transcription factors peroxisome proliferator-activated receptor (PPAR) and liver X receptors/retinoid X receptor (LXR/RXR) which are intricately involved in regulating the cellular metabolism of macrophages (fatty acid, cholesterol and glucose homeostasis) and are central to inflammatory responses. CONCLUSIONS/SIGNIFICANCE These results offer new insights into the structure and function of a well-known immunomodulatory molecule, Sm16, and places it within a wider family of trematode-specific small molecule HDM immune-modulators with immuno-biotherapeutic possibilities.
Collapse
Affiliation(s)
- Jenna Shiels
- School of Biological Sciences, Queen’s University Belfast, Northern Ireland
- Airway Innate Immunity Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine (WWIEM), School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Northern Ireland
| | - Krystyna Cwiklinski
- School of Biological Sciences, Queen’s University Belfast, Northern Ireland
- Center of One Health (COH) and Ryan Institute, School of Natural Science, National University of Ireland Galway, Galway, Ireland
| | - Raquel Alvarado
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW, Australia
| | - Karine Thivierge
- Institute of Parasitology, McGill University, Montreal, Quebec, Canada
| | - Sophie Cotton
- Institute of Parasitology, McGill University, Montreal, Quebec, Canada
| | | | - Joyce To
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW, Australia
| | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, The University of Technology Sydney, Ultimo, NSW, Australia
| | - Clifford C. Taggart
- Airway Innate Immunity Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine (WWIEM), School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Northern Ireland
| | - Sinead Weldon
- Airway Innate Immunity Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine (WWIEM), School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Northern Ireland
| | - John P. Dalton
- School of Biological Sciences, Queen’s University Belfast, Northern Ireland
- Center of One Health (COH) and Ryan Institute, School of Natural Science, National University of Ireland Galway, Galway, Ireland
- Institute of Parasitology, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
10
|
Angeles JMM, Mercado VJP, Rivera PT. Behind Enemy Lines: Immunomodulatory Armamentarium of the Schistosome Parasite. Front Immunol 2020; 11:1018. [PMID: 32582161 PMCID: PMC7295904 DOI: 10.3389/fimmu.2020.01018] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
The deeply rooted, intricate relationship between the Schistosoma parasite and the human host has enabled the parasite to successfully survive within the host and surreptitiously evade the host's immune attacks. The parasite has developed a variety of strategies in its immunomodulatory armamentarium to promote infection without getting harmed or killed in the battlefield of immune responses. These include the production of immunomodulatory molecules, alteration of membranes, and the promotion of granuloma formation. Schistosomiasis thus serves as a paradigm for understanding the Th2 immune responses seen in various helminthiases. This review therefore aims to summarize the immunomodulatory mechanisms of the schistosome parasites to survive inside the host. Understanding these immunomodulatory strategies not only provides information on parasite-host interactions, but also forms the basis in the development of novel drugs and vaccines against the schistosome infection, as well as various types of autoimmune and inflammatory conditions.
Collapse
Affiliation(s)
- Jose Ma M Angeles
- Department of Parasitology, College of Public Health, University of the Philippines Manila, Manila, Philippines
| | - Van Jerwin P Mercado
- Department of Parasitology, College of Public Health, University of the Philippines Manila, Manila, Philippines
| | - Pilarita T Rivera
- Department of Parasitology, College of Public Health, University of the Philippines Manila, Manila, Philippines
| |
Collapse
|
11
|
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
|
12
|
A Solid-in-Oil Nanodispersion System for Transcutaneous Immunotherapy of Cow's Milk Allergies. Pharmaceutics 2020; 12:pharmaceutics12030205. [PMID: 32120968 PMCID: PMC7150982 DOI: 10.3390/pharmaceutics12030205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 01/08/2023] Open
Abstract
An allergy to cow’s milk proteins is the most common food allergy in infants and toddlers. Conventional oral immunotherapy for cow’s milk allergies requires hospital admission due to the risk of severe allergic reactions, including anaphylaxis. Therefore, a simpler and safer immunotherapeutic method is desirable. We examined transcutaneous immunotherapy with a solid-in-oil (S/O) system. In the S/O system, nano-sized particles of proteins are dispersed in an oil-vehicle with the assistance of nonionic surfactants. In the present study, the S/O system enhanced the skin permeation of the allergen molecule β-lactoglobulin (BLG), as compared with a control PBS solution. The patches containing BLG in the S/O nanodispersion skewed the immune response in the allergy model mice toward T helper type 1 immunity, indicating the amelioration of allergic symptoms. This effect was more pronounced when the immunomodulator resiquimod (R-848) was included in the S/O system.
Collapse
|
13
|
Egesa M, Lubyayi L, Tukahebwa EM, Bagaya BS, Chalmers IW, Wilson S, Hokke CH, Hoffmann KF, Dunne DW, Yazdanbakhsh M, Labuda LA, Cose S. Schistosoma mansoni schistosomula antigens induce Th1/Pro-inflammatory cytokine responses. Parasite Immunol 2018; 40:e12592. [PMID: 30239006 PMCID: PMC6492251 DOI: 10.1111/pim.12592] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/15/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022]
Abstract
Larvae of Schistosoma (schistosomula) are highly susceptible to host immune responses and are attractive prophylactic vaccine targets, although cellular immune responses against schistosomula antigens in endemic human populations are not well characterized. We collected blood and stool from 54 Schistosoma mansoni-infected Ugandans, isolated peripheral blood mononuclear cells and stimulated them for 24 hours with schistosome adult worm and soluble egg antigens (AWA and SEA), along with schistosomula recombinant proteins rSmKK7, Lymphocyte Antigen 6 isoforms (rSmLy6A and rSmLy6B), tetraspanin isoforms (rSmTSP6 and rSmTSP7). Cytokines, chemokines and growth factors were measured in the culture supernatants using a multiplex luminex assay, and infection intensity was determined before and at 1 year after praziquantel (PZQ) treatment using the Kato-Katz method. Cellular responses were grouped and the relationship between groups of correlated cellular responses and infection intensity before and after PZQ treatment was investigated. AWA and SEA induced mainly Th2 responses. In contrast, rSmLy6B, rSmTSP6 and rSmTSP7 induced Th1/pro-inflammatory responses. While recombinant antigens rSmKK7 and rSmLy6A did not induce a Th1/pro-inflammatory response, they had an association with pre-treatment infection intensity after adjusting for age and sex. Testing more schistosomula antigens using this approach could provide immune-epidemiology identifiers necessary for prioritizing next generation schistosomiasis vaccine candidates.
Collapse
Affiliation(s)
- Moses Egesa
- Department of Medical MicrobiologySchool of Biomedical SciencesMakerere University College of Health SciencesKampalaUganda
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research UnitEntebbeUganda
| | - Lawrence Lubyayi
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research UnitEntebbeUganda
| | | | - Bernard S. Bagaya
- Department of Immunology and Molecular BiologySchool of Biomedical SciencesMakerere University College of Health SciencesKampalaUganda
| | - Iain W. Chalmers
- Institute of Biological, Environmental & Rural SciencesAberystwyth UniversityAberystwythUK
| | - Shona Wilson
- Department of PathologyUniversity of CambridgeCambridgeUK
| | - Cornelis H. Hokke
- Department of ParasitologyLeiden University Medical CenterLeidenThe Netherlands
| | - Karl F. Hoffmann
- Institute of Biological, Environmental & Rural SciencesAberystwyth UniversityAberystwythUK
| | - David W. Dunne
- Department of PathologyUniversity of CambridgeCambridgeUK
| | - Maria Yazdanbakhsh
- Department of ParasitologyLeiden University Medical CenterLeidenThe Netherlands
| | - Lucja A. Labuda
- Department of ParasitologyLeiden University Medical CenterLeidenThe Netherlands
| | - Stephen Cose
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research UnitEntebbeUganda
- Department of Clinical ResearchLondon School of Hygiene & Tropical MedicineLondonUK
| |
Collapse
|
14
|
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
|
15
|
Jiang X, Xia S, He X, Ma H, Feng Y, Liu Z, Wang W, Tian M, Chen H, Peng F, Wang L, Zhao P, Ge J, Liu D. Targeting peptide‐enhanced antibody and CD11c+dendritic cells to inclusion bodies expressing protective antigen against ETEC in mice. FASEB J 2018; 33:2836-2847. [DOI: 10.1096/fj.201800289rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Xinpeng Jiang
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
- Postdoctoral WorkstationHeilongjiang Academy of Agricultural SciencesHarbinChina
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - Shuang Xia
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - Xinmiao He
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Hong Ma
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Yanzhong Feng
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Ziguang Liu
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Wentao Wang
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Ming Tian
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
- Postdoctoral WorkstationHeilongjiang Academy of Agricultural SciencesHarbinChina
| | - Heshu Chen
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Fugang Peng
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Liang Wang
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| | - Peng Zhao
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - Junwei Ge
- Department of Preventive Veterinary MedicineCollege of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
| | - Di Liu
- Key Laboratory of Combining Farming and Animal HusbandryMinistry of AgricultureAnimal Husbandry Research InstituteHarbinChina
| |
Collapse
|
16
|
Sombetzki M, Koslowski N, Rabes A, Seneberg S, Winkelmann F, Fritzsche C, Loebermann M, Reisinger EC. Host Defense Versus Immunosuppression: Unisexual Infection With Male or Female Schistosoma mansoni Differentially Impacts the Immune Response Against Invading Cercariae. Front Immunol 2018; 9:861. [PMID: 29743881 PMCID: PMC5930291 DOI: 10.3389/fimmu.2018.00861] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/06/2018] [Indexed: 12/31/2022] Open
Abstract
Infection with the intravascular diecious trematode Schistosoma spp. remains a serious tropical disease and public health problem in the developing world, affecting over 258 million people worldwide. During chronic Schistosoma mansoni infection, complex immune responses to tissue-entrapped parasite eggs provoke granulomatous inflammation which leads to serious damage of the liver and intestine. The suppression of protective host immune mechanisms by helminths promotes parasite survival and benefits the host by reducing tissue damage. However, immune-suppressive cytokines may reduce vaccine-induced immune responses. By combining a single-sex infection system with a murine air pouch model, we were able to demonstrate that male and female schistosomes play opposing roles in modulating the host’s immune response. Female schistosomes suppress early innate immune responses to invading cercariae in the skin and upregulate anergy-associated genes. In contrast, male schistosomes trigger strong innate immune reactions which lead to a reduction in worm and egg burden in the liver. Our data suggest that the female worm is a neglected player in the dampening of the host’s immune defense system and is therefore a promising target for new immune modulatory therapies.
Collapse
Affiliation(s)
- Martina Sombetzki
- Division of Tropical Medicine and Infectious Diseases, Center of Internal Medicine II, University Medical Center Rostock, Rostock, Germany
| | - Nicole Koslowski
- Division of Tropical Medicine and Infectious Diseases, Center of Internal Medicine II, University Medical Center Rostock, Rostock, Germany
| | - Anne Rabes
- Division of Tropical Medicine and Infectious Diseases, Center of Internal Medicine II, University Medical Center Rostock, Rostock, Germany
| | - Sonja Seneberg
- Division of Tropical Medicine and Infectious Diseases, Center of Internal Medicine II, University Medical Center Rostock, Rostock, Germany
| | - Franziska Winkelmann
- Division of Tropical Medicine and Infectious Diseases, Center of Internal Medicine II, University Medical Center Rostock, Rostock, Germany
| | - Carlos Fritzsche
- Division of Tropical Medicine and Infectious Diseases, Center of Internal Medicine II, University Medical Center Rostock, Rostock, Germany
| | - Micha Loebermann
- Division of Tropical Medicine and Infectious Diseases, Center of Internal Medicine II, University Medical Center Rostock, Rostock, Germany
| | - Emil C Reisinger
- Division of Tropical Medicine and Infectious Diseases, Center of Internal Medicine II, University Medical Center Rostock, Rostock, Germany
| |
Collapse
|
17
|
Prendergast CT, Sanin DE, Mountford AP. CD4 T-cell hyporesponsiveness induced by schistosome larvae is not dependent upon eosinophils but may involve connective tissue mast cells. Parasite Immunol 2016; 38:81-92. [PMID: 26679416 PMCID: PMC4744672 DOI: 10.1111/pim.12300] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/07/2015] [Indexed: 12/12/2022]
Abstract
In areas endemic for schistosomiasis, people can often be in contact with contaminated water resulting in repeated exposures to infective Schistosoma mansoni cercariae. Using a murine model, repeated infections result in IL‐10‐dependent CD4+ T‐cell hyporesponsiveness in the skin‐draining lymph nodes (sdLN), which could be caused by an abundance of eosinophils and connective tissue mast cells at the skin infection site. Here, we show that whilst the absence of eosinophils did not have a significant effect on cytokine production, MHC‐II+ cells were more numerous in the dermal cell exudate population. Nevertheless, the absence of dermal eosinophils did not lead to an increase in the responsiveness of CD4+ T cells in the sdLN, revealing that eosinophils in repeatedly exposed skin did not impact on the development of CD4+ T‐cell hyporesponsiveness. On the other hand, the absence of connective tissue mast cells led to a reduction in dermal IL‐10 and to an increase in the number of MHC‐II+ cells infiltrating the skin. There was also a small but significant alleviation of hyporesponsiveness in the sdLN, suggesting that mast cells may have a role in regulating immune responses after repeated exposures of the skin to S. mansoni cercariae.
Collapse
Affiliation(s)
- C T Prendergast
- Centre for Immunology and Infection, Department of Biology, University of York, York, UK
| | - D E Sanin
- Centre for Immunology and Infection, Department of Biology, University of York, York, UK
| | - A P Mountford
- Centre for Immunology and Infection, Department of Biology, University of York, York, UK
| |
Collapse
|
18
|
Prendergast CT, Sanin DE, Mountford AP. Alternatively Activated Mononuclear Phagocytes from the Skin Site of Infection and the Impact of IL-4Rα Signalling on CD4+T Cell Survival in Draining Lymph Nodes after Repeated Exposure to Schistosoma mansoni Cercariae. PLoS Negl Trop Dis 2016; 10:e0004911. [PMID: 27505056 PMCID: PMC4978413 DOI: 10.1371/journal.pntd.0004911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/18/2016] [Indexed: 12/29/2022] Open
Abstract
In a murine model of repeated exposure of the skin to infective Schistosoma mansoni cercariae, events leading to the priming of CD4 cells in the skin draining lymph nodes were examined. The dermal exudate cell (DEC) population recovered from repeatedly (4x) exposed skin contained an influx of mononuclear phagocytes comprising three distinct populations according to their differential expression of F4/80 and MHC-II. As determined by gene expression analysis, all three DEC populations (F4/80-MHC-IIhigh, F4/80+MHC-IIhigh, F4/80+MHC-IIint) exhibited major up-regulation of genes associated with alternative activation. The gene encoding RELMα (hallmark of alternatively activated cells) was highly up-regulated in all three DEC populations. However, in 4x infected mice deficient in RELMα, there was no change in the extent of inflammation at the skin infection site compared to 4x infected wild-type cohorts, nor was there a difference in the abundance of different mononuclear phagocyte DEC populations. The absence of RELMα resulted in greater numbers of CD4+ cells in the skin draining lymph nodes (sdLN) of 4x infected mice, although they remained hypo-responsive. Using mice deficient for IL-4Rα, in which alternative activation is compromised, we show that after repeated schistosome infection, levels of regulatory IL-10 in the skin were reduced, accompanied by increased numbers of MHC-IIhigh cells and CD4+ T cells in the skin. There were also increased numbers of CD4+ T cells in the sdLN in the absence of IL-4Rα compared to cells from singly infected mice. Although their ability to proliferate was still compromised, increased cellularity of sdLN from 4x IL-4RαKO mice correlated with reduced expression of Fas/FasL, resulting in decreased apoptosis and cell death but increased numbers of viable CD4+ T cells. This study highlights a mechanism through which IL-4Rα may regulate the immune system through the induction of IL-10 and regulation of Fas/FasL mediated cell death.
Collapse
Affiliation(s)
- Catriona T. Prendergast
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| | - David E. Sanin
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| | - Adrian P. Mountford
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
- * E-mail:
| |
Collapse
|
19
|
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
|
20
|
Schistosoma mansoni Larvae Do Not Expand or Activate Foxp3+ Regulatory T Cells during Their Migratory Phase. Infect Immun 2015. [PMID: 26195548 PMCID: PMC4567639 DOI: 10.1128/iai.00408-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Foxp3+ regulatory T (Treg) cells play a key role in suppression of immune responses during parasitic helminth infection, both by controlling damaging immunopathology and by inhibiting protective immunity. During the patent phase of Schistosoma mansoni infection, Foxp3+ Treg cells are activated and suppress egg-elicited Th2 responses, but little is known of their induction and role during the early prepatent larval stage of infection. We quantified Foxp3+ Treg cell responses during the first 3 weeks of murine S. mansoni infection in C57BL/6 mice, a time when larval parasites migrate from the skin and transit the lungs en route to the hepatic and mesenteric vasculature. In contrast to other helminth infections, S. mansoni did not elicit a Foxp3+ Treg cell response during this early phase of infection. We found that the numbers and proportions of Foxp3+ Treg cells remained unchanged in the lungs, draining lymph nodes, and spleens of infected mice. There was no increase in the activation status of Foxp3+ Treg cells upon infection as assessed by their expression of CD25, Foxp3, and Helios. Furthermore, infection failed to induce Foxp3+ Treg cells to produce the suppressive cytokine interleukin 10 (IL-10). Instead, only CD4+ Foxp3− IL-4+ Th2 cells showed increased IL-10 production upon infection. These data indicate that Foxp3+ Treg cells do not play a prominent role in regulating immunity to S. mansoni larvae and that the character of the initial immune response invoked by S. mansoni parasites contrasts with the responses to other parasitic helminth infections that promote rapid Foxp3+ Treg cell responses.
Collapse
|
21
|
Sanin DE, Prendergast CT, Bourke CD, Mountford AP. Helminth Infection and Commensal Microbiota Drive Early IL-10 Production in the Skin by CD4+ T Cells That Are Functionally Suppressive. PLoS Pathog 2015; 11:e1004841. [PMID: 25974019 PMCID: PMC4431738 DOI: 10.1371/journal.ppat.1004841] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/30/2015] [Indexed: 12/12/2022] Open
Abstract
The skin provides an important first line of defence and immunological barrier to invasive pathogens, but immune responses must also be regulated to maintain barrier function and ensure tolerance of skin surface commensal organisms. In schistosomiasis-endemic regions, populations can experience repeated percutaneous exposure to schistosome larvae, however little is known about how repeated exposure to pathogens affects immune regulation in the skin. Here, using a murine model of repeated infection with Schistosoma mansoni larvae, we show that the skin infection site becomes rich in regulatory IL-10, whilst in its absence, inflammation, neutrophil recruitment, and local lymphocyte proliferation is increased. Whilst CD4+ T cells are the primary cellular source of regulatory IL-10, they expressed none of the markers conventionally associated with T regulatory (Treg) cells (i.e. FoxP3, Helios, Nrp1, CD223, or CD49b). Nevertheless, these IL-10+ CD4+ T cells in the skin from repeatedly infected mice are functionally suppressive as they reduced proliferation of responsive CD4+ T cells from the skin draining lymph node. Moreover, the skin of infected Rag-/- mice had impaired IL-10 production and increased neutrophil recruitment. Finally, we show that the mechanism behind IL-10 production by CD4+ T cells in the skin is due to a combination of an initial (day 1) response specific to skin commensal bacteria, and then over the following days schistosome-specific CD4+ T cell responses, which together contribute towards limiting inflammation and tissue damage following schistosome infection. We propose CD4+ T cells in the skin that do not express markers of conventional T regulatory cell populations have a significant role in immune regulation after repeated pathogen exposure and speculate that these cells may also help to maintain skin barrier function in the context of repeated percutaneous insult by other skin pathogens. The skin is a major barrier protecting the host from pathogen infection, but is also a site for immune regulation. Using a murine model of repeated percutaneous exposure to infectious Schistosoma mansoni cercariae, we show that, in the skin, CD4+ T cells that do not express markers of conventional regulatory T cells are the main early source of immunoregulatory IL-10 and are functionally suppressive of adaptive immune responses. We demonstrate that the production of regulatory IL-10 in the skin is greatly enhanced after repeated schistosome infection compared to levels present after a single infection and that it limits both neutrophil recruitment and local CD4+ T cell proliferation, thereby preventing excessive inflammation and tissue damage. Initially (day 1), IL-10 producing CD4+ T cells are reactive towards skin commensal bacteria, although over succeeding days they progressively become specific for schistosome antigens. Consequently, our findings highlight a role for early IL-10 produced by dermal CD4+ T cells to mediate immune regulation in advance of later stage chronic infection conventionally associated with the presence of IL-10. Our work provides a mechanistic insight into the triggers of early IL-10 production at barrier sites like the skin, and suggests how tolerance and pathogen clearance might be co-regulated early after exposure to infectious agents.
Collapse
Affiliation(s)
- David E. Sanin
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| | - Catriona T. Prendergast
- 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
| | - Adrian P. Mountford
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
- * E-mail:
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
El Ridi R, Tallima H. Why the radiation-attenuated cercarial immunization studies failed to guide the road for an effective schistosomiasis vaccine: A review. J Adv Res 2014; 6:255-67. [PMID: 26257924 PMCID: PMC4522536 DOI: 10.1016/j.jare.2014.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/05/2014] [Accepted: 10/09/2014] [Indexed: 12/31/2022] Open
Abstract
Schistosomiasis is a debilitating parasitic disease caused by platyhelminthes of the genus Schistosoma, notably Schistosoma mansoni, Schistosoma haematobium, and Schistosoma japonicum. Pioneer researchers used radiation-attenuated (RA) schistosome larvae to immunize laboratory rodent and non-human primate hosts. Significant and reproducible reduction in challenge worm burden varying from 30% to 90% was achieved, providing a sound proof that vaccination against this infection is feasible. Extensive histopathological, tissue mincing and incubation, autoradiographic tracking, parasitological, and immunological studies led to defining conditions and settings for achieving optimal protection and delineating the resistance underlying mechanisms. The present review aims to summarize these findings and draw the lessons that should have guided the development of an effective schistosomiasis vaccine.
Collapse
Affiliation(s)
- Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University, Cairo 12613, Egypt
| | - Hatem Tallima
- Zoology Department, Faculty of Science, Cairo University, Cairo 12613, Egypt
| |
Collapse
|
26
|
Ye L, Wen Z, Li Y, Chen B, Yu T, Liu L, Zhang J, Ma Y, Xiao S, Ding L, Li L, Huang Z. Interleukin-10 attenuation of collagen-induced arthritis is associated with suppression of interleukin-17 and retinoid-related orphan receptor γt production in macrophages and repression of classically activated macrophages. Arthritis Res Ther 2014; 16:R96. [PMID: 24742125 PMCID: PMC4060547 DOI: 10.1186/ar4544] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 04/04/2014] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Our objective in the present study was to determine the signaling pathway of interleukin 10 (IL-10) for modulating IL-17 expression in macrophages and the importance of this mediation in collagen-induced arthritis (CIA). METHODS IL-10-knockout (IL-10⁻/⁻) mice and wild-type (WT) mice were immunized with chicken type II collagen (CII) to induce arthritis. The expression levels of IL-17 and retinoid-related orphan receptor γt (RORγt) in macrophages and joint tissues of IL-10⁻/⁻ and WT mice were analyzed by enzyme-linked immunosorbent assay, quantitative RT-PCR (qRT-PCR) and Western blotting. The F4/80 macrophages and positive IL-17-producing macrophages in synovial tissues of the mice were determined by immunohistochemistry. The populations of classically activated macrophage (M1) and alternatively activated macrophage (M2) phenotypes were analyzed by flow cytometry. The expression of genes associated with M1 and M2 markers was analyzed by qRT-PCR. RESULTS Compared to WT mice, IL-10⁻/⁻ mice had exacerbated CIA development, which was associated with increased production of T helper 17 cell (Th17)/Th1 proinflammatory cytokines and CII-specific immunoglobulin G2a antibody after CII immunization. Macrophages in IL-10⁻/⁻ mice had increased amounts of IL-17 and RORγt compared with the amounts in WT mice with CIA. Immunofluorescence microscopy showed that the number of IL-17-producing macrophages in synovial tissues was significantly higher in IL-10⁻/⁻ mice than in WT mice. IL-10 deficiency might promote macrophage polarization toward the proinflammatory M1 phenotype, which contributes to the rheumatoid arthritis inflammation response. CONCLUSION IL-10 inhibits IL-17 and RORγt expression in macrophages and suppresses macrophages toward the proinflammatory M1 phenotype, which is important for the role of IL-10 in mediating the pathogenesis of CIA.
Collapse
|
27
|
Wang X, Dai Y, Zhao S, Tang J, Li H, Xing Y, Qu G, Li X, Dai J, Zhu Y, Zhang X. PAMAM-Lys, a novel vaccine delivery vector, enhances the protective effects of the SjC23 DNA vaccine against Schistosoma japonicum infection. PLoS One 2014; 9:e86578. [PMID: 24497955 PMCID: PMC3907433 DOI: 10.1371/journal.pone.0086578] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 12/12/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Schistosomiasis japonica remains a major public-health concern in China. Praziquantel-based chemotherapy effectively reduces both infections and intensity; however, it can not prevent re-infection. Furthermore, there is an increasing concern about praziquantel resistance following long-term repeated use of the drug in endemic areas. Therefore, development of a schistosomiasis vaccine, as a strategy to prevent and control schistosomiasis japonica, has been given high priority. The present study was conducted to develop PAMAM dendrimers as a novel vaccine delivery vector for a schistosomiasis japonica DNA vaccine and evaluate its ability to enhance protective effects against Schistosoma japonicum infection. METHODOLOGY/PRINCIPAL FINDINGS Lysine was used to modify 4.0G PAMAM, and the modified product PAMAM-Lys was synthesized. PAMAM-Lys showed both high transfection and low cytotocity for gene delivery in vitro. DNA vaccines combined with PAMAM-Lys produced higher level of protection compare with naked DNA vaccines against S. japonicum infection in a mouse model. Futhermore,antibodies from mice immunized with PAMAM-Lys combined DNA vaccines were significantly higher than those of mice immunized with the naked DNA vaccines. The PAMAM-Lys vector elicited a predominantly IgG2a antibody response and a tremendously increase in the production of IL-2 and IFN-γ. CONCLUSION/SIGNIFICANCE Lysine-modified PAMAM-Lys is an excellent vector. PAMAM-Lys may enhance the immunoreactivity of DNA vaccine and increase the protective effect of the SjC23 DNA vaccine against S. japonicum infection.
Collapse
Affiliation(s)
- Xiaoting Wang
- Jiangsu Stem Cell Key Laboratory, Institute of Medical Biotechnology, Medical College of Soochow University; Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Yang Dai
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Song Zhao
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Jianxia Tang
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Hongjun Li
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Yuntian Xing
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Guoli Qu
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Xinsong Li
- Biomaterials and Drug Delivery Laboratories, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Jianrong Dai
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Yinchang Zhu
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on Molecular Biology of Parasites, Wuxi, Jiangsu, China
- * E-mail: (YCZ); (XGZ)
| | - Xueguang Zhang
- Jiangsu Stem Cell Key Laboratory, Institute of Medical Biotechnology, Medical College of Soochow University; Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- * E-mail: (YCZ); (XGZ)
| |
Collapse
|
28
|
McWilliam HEG, Piedrafita D, Li Y, Zheng M, He Y, Yu X, McManus DP, Meeusen ENT. Local immune responses of the Chinese water buffalo, Bubalus bubalis, against Schistosoma japonicum larvae: crucial insights for vaccine design. PLoS Negl Trop Dis 2013; 7:e2460. [PMID: 24086786 PMCID: PMC3784499 DOI: 10.1371/journal.pntd.0002460] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 08/17/2013] [Indexed: 01/03/2023] Open
Abstract
Asian schistosomiasis is a zoonotic parasitic disease infecting up to a million people and threatening tens of millions more. Control of this disease is hindered by the animal reservoirs of the parasite, in particular the water buffalo (Bubalus bubalis), which is responsible for significant levels of human transmission. A transmission-blocking vaccine administered to buffaloes is a realistic option which would aid in the control of schistosomiasis. This will however require a better understanding of the immunobiology of schistosomiasis in naturally exposed buffaloes, particularly the immune response to migrating schistosome larvae, which are the likely targets of an anti-schistosome vaccine. To address this need we investigated the immune response at the major sites of larval migration, the skin and the lungs, in previously exposed and re-challenged water buffaloes. In the skin, a strong allergic-type inflammatory response occurred, characterised by leukocyte and eosinophil infiltration including the formation of granulocytic abscesses. Additionally at the local skin site, interleukin-5 transcript levels were elevated, while interleukin-10 levels decreased. In the skin-draining lymph node (LN) a predominant type-2 profile was seen in stimulated cells, while in contrast a type-1 profile was detected in the lung draining LN, and these responses occurred consecutively, reflecting the timing of parasite migration. The intense type-2 immune response at the site of cercarial penetration is significantly different to that seen in naive and permissive animal models such as mice, and suggests a possible mechanism for immunity. Preliminary data also suggest a reduced and delayed immune response occurred in buffaloes given high cercarial challenge doses compared with moderate infections, particularly in the skin. This study offers a deeper understanding into the immunobiology of schistosomiasis in a natural host, which may aid in the future design of more effective vaccines. Schistosomiasis is caused by a parasitic blood fluke, and in parts of Asia it infects both humans and livestock such as water buffaloes. This makes controlling the disease more difficult, because both humans and livestock must be treated regularly. A vaccine given to buffaloes is likely to reduce human infection rates and improve buffalo health by providing long-lasting protection from re-infection; at present no vaccines are available. Older buffaloes are known to have some immunity to schistosomiasis which is acquired over time; however how this occurs is not understood. In this study we investigated the immune response of buffalo against the schistosome larvae, which are vulnerable to immune attack, and hence are the ideal stage to target for vaccination. We found that the buffalo produces a profound allergic type-2 response as larvae penetrate the skin, with significant cellular infiltrates and abscesses. When the larvae move next to the lungs, a uniquely type-1 response was induced. This skin response is much greater than more susceptible animals such as mice, and may be a mechanism for larval killing in the buffalo. This study offers insight into the immunobiology of an important host for schistosomiasis and may help in designing better vaccines.
Collapse
Affiliation(s)
- Hamish E. G. McWilliam
- Biotechnology Research Laboratories, School of Biomedical Sciences, Monash University, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics, Monash University, Melbourne, Victoria, Australia
- * E-mail:
| | - David Piedrafita
- Biotechnology Research Laboratories, School of Biomedical Sciences, Monash University, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics, Monash University, Melbourne, Victoria, Australia
| | - Yuesheng Li
- Hunan Institute of Parasitic Diseases, Yueyang, Hunan, China
- Molecular Parasitology Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Mao Zheng
- Hunan Institute of Parasitic Diseases, Yueyang, Hunan, China
| | - Yongkang He
- Hunan Institute of Parasitic Diseases, Yueyang, Hunan, China
| | - Xinling Yu
- Hunan Institute of Parasitic Diseases, Yueyang, Hunan, China
| | - Donald P. McManus
- Molecular Parasitology Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Els N. T. Meeusen
- Biotechnology Research Laboratories, School of Biomedical Sciences, Monash University, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
A study of immunomodulatory genes responses to macrophages of Schistosoma japonicum infection during different stages by microarray analysis. Acta Trop 2013; 127:251-60. [PMID: 23732117 DOI: 10.1016/j.actatropica.2013.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 05/15/2013] [Accepted: 05/25/2013] [Indexed: 01/08/2023]
Abstract
Macrophages initiate, modulate, and also serve as final effector cells in immune responses during the course of schistosomal infections. In this study, we investigated the gene expression profile and functional changes of macrophages in immune responses against the Schistosoma japonicum by microarray analysis. Hierarchical clustering analysis demonstrated that a significant switch in gene transformation associated with a type-1 response and linked with a type-2 cytokine phenotype occurs between 4.5 and 8 weeks post-infection. Moreover, the gene profiles at 3 later time-points following egg challenge were similar in complexity and magnitude. The data also showed that there were mostly inhibition of gene expression related TLR, IFN, MHC and TNFrsf at the switch between 4.5 and 8 weeks post-infection, It is suggested that these immunomodulatory genes may be down-regulated in defense against S. japonicum eggs and granuloma pathology. The induction of alternatively activated macrophage (AAMϕ) was important for dampening the inflammation in hepatic granulomas and contributing to a decrease in cytotoxicity. The gene expressions involved in repair/remodeling during liver fibrosis were also observed after egg production. Understanding the immune mechanisms associated with parasitic resistance, pathology of parasite infection, and parasite growth will provide useful insight on host-schistosome interactions and for the control of schistosomiasis.
Collapse
|
30
|
Sun XJ, Li R, Sun X, Zhou Y, Wang Y, Liu XJ, Lu Q, Zhou CL, Wu ZD. Unique roles of Schistosoma japonicum protein Sj16 to induce IFN-γ and IL-10 producing CD4(+)CD25(+) regulatory T cells in vitro and in vivo. Parasite Immunol 2013; 34:430-9. [PMID: 22712636 DOI: 10.1111/j.1365-3024.2012.01377.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Various proteins are expressed during different stages of schistosome development that are essential for cercarial penetration of vertebrate skin and evasion of host immune response. CD4(+)CD25(+) regulatory T cells are important in modulating immune responses towards helminth infections. Schistosoma japonicum protein Sj16 present in the secretions of schistosomula has been shown to have anti-inflammatory effects; however, it is uncertain whether Sj16 can induce CD4(+)CD25(+) regulatory T cells to participate in the regulation of early infection. In this study, we demonstrate a relationship between recombinant Sj16 (rSj16) and the induction of CD4(+)CD25(+) Foxp3(+) regulatory T cells. An increase in CD4(+)CD25(+) T cells was observed both in splenic cells from mice injected with rSj16 and the cells pretreated with rSj16, respectively. The induced CD4(+)CD25(+) T cells suppressed CD4(+)CD25(-) T-cell proliferation; furthermore, IFN-γ and IL-10 released from rSj16-stimulated cells contribute to this suppression. Additionally, rSj16-treated bone marrow dendritic cells (BMDCs) demonstrate an immature phenotype and play a role in the conversion of CD4(+)CD25(-) T cells into suppressive CD4(+)CD25(+) regulatory T cells. Our study identified a new CD4(+)CD25(+) T-cell population that induced by rSj16 and suggests that an IFN-γ-biased microenvironment during early infection of schistosome may favour the establishment of infection.
Collapse
Affiliation(s)
- X J Sun
- Key Laboratory of Pathogen Biology of Jiangsu Province, Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Beschin A, De Baetselier P, Van Ginderachter JA. Contribution of myeloid cell subsets to liver fibrosis in parasite infection. J Pathol 2012; 229:186-97. [DOI: 10.1002/path.4112] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 08/24/2012] [Accepted: 09/13/2012] [Indexed: 12/23/2022]
Affiliation(s)
- Alain Beschin
- Myeloid Cell Immunology Laboratory; VIB Brussels Belgium
- Cellular and Molecular Immunology Unit; Vrije Universiteit Brussel; Brussels Belgium
| | - Patrick De Baetselier
- Myeloid Cell Immunology Laboratory; VIB Brussels Belgium
- Cellular and Molecular Immunology Unit; Vrije Universiteit Brussel; Brussels Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory; VIB Brussels Belgium
- Cellular and Molecular Immunology Unit; Vrije Universiteit Brussel; Brussels Belgium
| |
Collapse
|
32
|
Dvir E, Mellanby R, Kjelgaard-Hansen M, Schoeman J. Plasma IL-8 concentrations are increased in dogs with spirocercosis. Vet Parasitol 2012; 190:185-90. [DOI: 10.1016/j.vetpar.2012.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 06/08/2012] [Accepted: 06/10/2012] [Indexed: 02/04/2023]
|
33
|
Araujo JM, Melo TTD, Sena ICD, Alves CC, Araujo N, Durães FDV, Oliveira SC, Fonseca CT. Schistosoma mansoni schistosomula tegument (Smteg) immunization in absence of adjuvant induce IL-10 production by CD4+ cells and failed to protect mice against challenge infection. Acta Trop 2012; 124:140-6. [PMID: 22842304 DOI: 10.1016/j.actatropica.2012.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 07/18/2012] [Accepted: 07/19/2012] [Indexed: 10/28/2022]
Abstract
The Schistosoma mansoni tegument interaction with the immune system plays a key role in disease establishment or elimination. We have recently demonstrated that S. mansoni schistosomula tegument (Smteg) is able to activate innate immune response and to induce protective immunity in a vaccine formulation with Freunds adjuvant. In this work, we evaluated the ability of Smteg to elicit protection in the absence of adjuvant. Smteg mice immunization resulted in significant antibody production, increased percentage of CD4+IFN-g+ and CD4+IL-10+ cells in spleen and increased production of IFN-g and IL-10 by spleen cells, but failed to reduce parasite burden, female fecundity and morbidity. We also demonstrated that BMDC stimulation with Smteg resulted in significant IL-10 production. Our results demonstrate that Smteg has immune modulatory proprieties.
Collapse
|
34
|
Makepeace BL, Martin C, Turner JD, Specht S. Granulocytes in helminth infection -- who is calling the shots? Curr Med Chem 2012; 19:1567-86. [PMID: 22360486 PMCID: PMC3394172 DOI: 10.2174/092986712799828337] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 12/23/2011] [Accepted: 12/26/2011] [Indexed: 02/06/2023]
Abstract
Helminths are parasitic organisms that can be broadly described as “worms” due to their elongated body plan, but which otherwise differ in shape, development, migratory routes and the predilection site of the adults and larvae. They are divided into three major groups: trematodes (flukes), which are leaf-shaped, hermaphroditic (except for blood flukes) flatworms with oral and ventral suckers; cestodes (tapeworms), which are segmented, hermaphroditic flatworms that inhabit the intestinal lumen; and nematodes (roundworms), which are dioecious, cylindrical parasites that inhabit intestinal and peripheral tissue sites. Helminths exhibit a sublime co-evolution with the host´s immune system that has enabled them to successfully colonize almost all multicellular species present in every geographical environment, including over two billion humans. In the face of this challenge, the host immune system has evolved to strike a delicate balance between attempts to neutralize the infectious assault versus limitation of damage to host tissues. Among the most important cell types during helminthic invasion are granulocytes: eosinophils, neutrophils and basophils. Depending on the specific context, these leukocytes may have pivotal roles in host protection, immunopathology, or facilitation of helminth establishment. This review provides an overview of the function of granulocytes in helminthic infections.
Collapse
Affiliation(s)
- B L Makepeace
- Department of Infection Biology, Institute of Infection & Global Health, University of Liverpool, Liverpool L69 7ZJ, UK
| | | | | | | |
Collapse
|
35
|
The Mannose Receptor (CD206) is an important pattern recognition receptor (PRR) in the detection of the infective stage of the helminth Schistosoma mansoni and modulates IFNγ production. Int J Parasitol 2011; 41:1335-45. [DOI: 10.1016/j.ijpara.2011.08.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 08/26/2011] [Accepted: 08/29/2011] [Indexed: 01/06/2023]
|
36
|
CD4+CD25+ regulatory cells contribute to the regulation of colonic Th2 granulomatous pathology caused by schistosome infection. PLoS Negl Trop Dis 2011; 5:e1269. [PMID: 21858239 PMCID: PMC3153428 DOI: 10.1371/journal.pntd.0001269] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 06/22/2011] [Indexed: 12/30/2022] Open
Abstract
Eggs of the helminth Schistosoma mansoni accumulate in the colon following infection and generate Th2-biassed inflammatory granulomas which become down- modulated in size as the infection proceeds to chronicity. However, although CD4+CD25+FoxP3+ regulatory T cells (T(regs)) are known to suppress Th1-mediated colitis, it is not clear whether they control Th2-associated pathologies of the large intestine which characterise several helminth infections. Here we used a novel 3D-multiphoton confocal microscopy approach to visualise and quantify changes in the size and composition of colonic granulomas at the acute and chronic phases of S. mansoni infection. We observed decreased granuloma size, as well as reductions in the abundance of DsRed+ T cells and collagen deposition at 14 weeks (chronic) compared to 8 weeks (acute) post-infection. Th2 cytokine production (i.e. IL-4, IL-5) in the colonic tissue and draining mesenteric lymph node (mLN) decreased during the chronic phase of infection, whilst levels of TGF-β1 increased, co-incident with reduced mLN proliferative responses, granuloma size and fibrosis. The proportion of CD4+CD25+FoxP3+T(regs): CD4+ cells in the mLN increased during chronic disease, while within colonic granulomas there was an approximate 4-fold increase. The proportion of CD4+CD25+FoxP3+T(regs) in the mLN that were CD103+ and CCR5+ also increased indicating an enhanced potential to home to intestinal sites. CD4+CD25+ cells suppressed antigen-specific Th2 mLN cell proliferation in vitro, while their removal during chronic disease resulted in significantly larger granulomas, partial reversal of Th2 hypo-responsiveness and an increase in the number of eosinophils in colonic granulomas. Finally, transfer of schistosome infection-expanded CD4+CD25+T(regs) down-modulated the development of colonic granulomas, including collagen deposition. Therefore, CD4+CD25+FoxP3+T(regs) appear to control Th2 colonic granulomas during chronic infection, and are likely to play a role in containing pathology during intestinal schistosomiasis.
Collapse
|
37
|
Lin D, Tian F, Wu H, Gao Y, Wu J, Zhang D, Ji M, McManus DP, Driguez P, Wu G. Multiple vaccinations with UV- attenuated cercariae in pig enhance protective immunity against Schistosoma japonicum infection as compared to single vaccination. Parasit Vectors 2011; 4:103. [PMID: 21663666 PMCID: PMC3146440 DOI: 10.1186/1756-3305-4-103] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 06/10/2011] [Indexed: 12/04/2022] Open
Abstract
Background Schistosomiasis japonica is a major public health problem in the endemic areas of China, the Philippines, and Indonesia. To date, a vaccine has not been developed against this disease but immunization with UV-attenuated cercariae can induce a high level of protective immunity in Landrace/Yorkshire/Duroc crossbred pigs. To compare the efficacy of a single vaccination and multiple vaccinations with UV-attenuated Schistosoma japonicum cercariae, two groups of pigs received either one or three exposures to 10,000 cercariae attenuated with 400 μw UV. Results Pigs with a single immunization had a 59.33% reduction in adult worm burden, a 89.87% reduction in hepatic eggs and a 86.27% reduction in fecal eggs at eight weeks post-challenge (P < 0.01). After three immunizations, protection increased to 77.62%, 88.8% and 99.78% reduction in adult worms, hepatic eggs and fecal eggs, respectively (P < 0.01). Humoral and cellular immunological parameters measured indicated that schistosome-specific IgG1 and IgG2 levels in the vaccinated groups were higher than in the infection-control group. Triple vaccinations resulted in higher levels of antibodies, especially IgG2, compared with a single vaccination and IFN-γ levels increased with repeated immunization with UV-irradiated cercariae. Conclusion The high levels of protection against S. japonicum infection can be achieved with a UV-attenuated vaccine in pigs, and that three vaccinations were possibly more effective than a single vaccination. Moreover, triple vaccinations evoked a more vigorous IFN-γ response and a stronger antibody-mediated response, especially an increase in the levels of IgG2 antibodies.
Collapse
Affiliation(s)
- Dandan Lin
- Department of Pathogen Biology & Immunology, Nanjing Medical University, Nanjing, Jiangsu, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhou H, Sun X, Lv Z, Shen Y, Peng H, Yang L, Zheng H, Fung MC, Cao J, Wu Z. The secretions products from invading cercariae of S. japonicum (0-3hRP) restrain mouse dendritic cells to mature. Parasitol Res 2011; 110:119-26. [PMID: 21626155 DOI: 10.1007/s00436-011-2458-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Accepted: 05/11/2011] [Indexed: 11/29/2022]
Abstract
Schistosomiasis is a water-borne infection caused mainly by human schistosomes including Schistosoma mansoni (S. mansoni) and Schistosoma japonicum (S. japonicum). In the infected host, immune events in the skin appear to have an important initiating role in determining the type, and guiding the magnitude, of the ensuing acquired immune response. The previous studies of S. mansoni showed that dendritic cells (DCs) prime Th2 response after activating with products released by schistosome larvae (material released in the first 3 h after transformation, 0-3hRP). Therefore, it is interesting to know whether 0-3hRP from S. japonicum also activate DC and induce Th2 immune response. Here, we report 0-3hRP of S. japonicum failed to stimulate the maturation of bone marrow-derived DCs (BM-DCs), while soluble antigen of the body of cercariae preparation (SCAP) induced BM-DCs to mature which can be weakened by 0-3hRP. Moreover, using an in vitro ovalbumin peptide-restricted priming assay, DCs treated with 0-3hRP failed to induce T cells to secrete more cytokines than the negative control group by CD4(+) T cells from DO11.10 transgenic mice, while DCs treated with SCAP upregulated secretions of IFN-γ and IL-17A, but downregulated IL-4. Importantly, BM-DCs treated with 0-3hRP plus SCAP induced BM-DCs selective mature which drive Th2-type polarized response. Our in vitro results agree with the findings of in vivo studies by inoculation of DO11.10 mice with different stimulus-activated DCs pulsed with ovalbumin peptide. Our data demonstrate that the secretions from invading cercariae of S. japonicum (0-3hRP) impaired DCs to mature, which is potentially allowing the parasite to negotiate the immune recognition and attack.
Collapse
Affiliation(s)
- Hejun Zhou
- Key Laboratory for Tropical Diseases Control, the Ministry of Education, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Cook PC, Aynsley SA, Turner JD, Jenkins GR, Van Rooijen N, Leeto M, Brombacher F, Mountford AP. Multiple helminth infection of the skin causes lymphocyte hypo-responsiveness mediated by Th2 conditioning of dermal myeloid cells. PLoS Pathog 2011; 7:e1001323. [PMID: 21445234 PMCID: PMC3060168 DOI: 10.1371/journal.ppat.1001323] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 02/16/2011] [Indexed: 12/03/2022] Open
Abstract
Infection of the mammalian host by schistosome larvae occurs via the skin, although
nothing is known about the development of immune responses to multiple exposures of
schistosome larvae, and/or their excretory/secretory (E/S) products. Here, we show
that multiple (4x) exposures, prior to the onset of egg laying by adult worms,
modulate the skin immune response and induce CD4+ cell
hypo-responsiveness in the draining lymph node, and even modulate the formation of
hepatic egg-induced granulomas. Compared to mice exposed to a single infection (1x),
dermal cells from multiply infected mice (4x), were less able to support lymph node
cell proliferation. Analysis of dermal cells showed that the most abundant in 4x mice
were eosinophils (F4/80+MHC-II−), but they did not
impact the ability of antigen presenting cells (APC) to support lymphocyte
proliferation to parasite antigen in vitro. However, two other cell
populations from the dermal site of infection appear to have a critical role. The
first comprises arginase-1+, Ym-1+ alternatively
activated macrophage-like cells, and the second are functionally compromised
MHC-IIhi cells. Through the administration of exogenous IL-12 to
multiply infected mice, we show that these suppressive myeloid cell phenotypes form
as a consequence of events in the skin, most notably an enrichment of IL-4 and IL-13,
likely resulting from an influx of RELMα-expressing eosinophils. We further
illustrate that the development of these suppressive dermal cells is dependent upon
IL-4Rα signalling. The development of immune hypo-responsiveness to schistosome
larvae and their effect on the subsequent response to the immunopathogenic egg is
important in appreciating how immune responses to helminth infections are modulated
by repeated exposure to the infective early stages of development. Schistosomiasis is a major helminth disease that infects more than 200 million people
in the tropics. Free-swimming aquatic cercariae infect through the skin after contact
with contaminated water, and in endemic areas this can occur frequently. However,
nothing is known about how multiple exposures affects innate immunity in the skin,
and/or whether it impacts the acquired immune response. Consequently, we have
developed an infection model in the mouse to examine the immune response to multiple
infections prior to the production of eggs. We show that multiple exposures to
schistosome larvae cause lymphocyte hypo-responsiveness, partly mediated by
macrophages and dendritic cells from the skin which have a
‘down-regulated’ phenotype and are not able to act as efficient antigen
presenting cells (APCs). These regulated APCs are conditioned amongst high levels of
the cytokines IL-4 and IL-13 which follow an influx of abundant eosinophils. In the
absence of the regulatory APCs, and in the absence of the common receptor chain for
IL-4 and IL-13 (i.e. IL-4Rα), lymphocyte proliferation is
restored. These findings are important in understanding how dermal immune responses
are modulated so that we can devise new strategies for vaccine delivery, or the
treatment of chronic inflammatory conditions of the skin.
Collapse
Affiliation(s)
- Peter C. Cook
- Centre for Immunology and Infection, Department
of Biology, The University of York, York, United Kingdom
| | - Sarah A. Aynsley
- Centre for Immunology and Infection, Department
of Biology, The University of York, York, United Kingdom
| | - Joseph D. Turner
- Centre for Immunology and Infection, Department
of Biology, The University of York, York, United Kingdom
| | - Gavin R. Jenkins
- Centre for Immunology and Infection, Department
of Biology, The University of York, York, United Kingdom
| | - Nico Van Rooijen
- Department of Molecular Cell Biology, Vrjie
Universiteit, Amsterdam, The Netherlands
| | - Mosiuoa Leeto
- Division of Infectious Immunology, University of
Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Division of Infectious Immunology, University of
Cape Town, Cape Town, South Africa
| | - Adrian P. Mountford
- Centre for Immunology and Infection, Department
of Biology, The University of York, York, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
ZHANG M, GAO Y, DU X, ZHANG D, JI M, WU G. Toll-like receptor (TLR) 2 and TLR4 deficiencies exert differential in vivo effects against Schistosoma japonicum. Parasite Immunol 2011; 33:199-209. [DOI: 10.1111/j.1365-3024.2010.01265.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
41
|
Kumar R, Sharma RK, Bansal DD, Patel DD, Mishra S, Miteva L, Dobreva Z, Gadjeva V, Stanilova S. Induction of immunostimulatory cytokine genes expression in human PBMCs by a novel semiquinone glucoside derivative (SQGD) isolated from a Bacillus sp. INM-1. Cell Immunol 2011; 267:67-75. [DOI: 10.1016/j.cellimm.2010.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 11/12/2010] [Accepted: 11/15/2010] [Indexed: 01/13/2023]
|
42
|
Antigen presenting cells may be able to distinguish between normal and radiated Schistosoma japonicum cercaria: an in vitro observation. J Biomed Res 2010; 24:285-91. [PMID: 23554642 PMCID: PMC3596594 DOI: 10.1016/s1674-8301(10)60040-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Indexed: 11/23/2022] Open
Abstract
Objective To observe the discrepancies of responses induced by Schistosoma japonicum (S. japonicum) normal cercaria antigen (NCA) and ultraviolet (UV) -radiation-attenuated cercaria antigen (UVACA) in an in vitro system. Methods S. japonicum cercariae were collected and UVACA and NCA were prepared. Mouse macrophage model cells (RAW 264.7) were treated with medium, NCA (40 µg/mL) or UVACA (40 µg/mL) in the presence or absence of recombinant mouse interferon gamma (rmIFN-γ; 4 ng/mL) for 48 h. Cell surface staining and flow cytometry were used to assess the major histocompatibility complex (MHC)γ; 4 ng/mL) for 48 h. Cell surface staining and flow cytometry were used to assess the major histocompatibility complex (MHC) II expression, and data were expressed as mean fluorescence intensities (MFI). Interleukin (IL) -10, IL-6 and prostaglandin E2 (PGE2) in cell culture supernatant were evaluated by commercial enzyme-linked immunosorbent assays. Results NCA significantly suppressed IFN-γ-induced MHC II expression on RAW 264.7 cells. In the presence of IFN-γ, NCA significantly promoted IL-6, IL-10 and PGE2 secretion from RAW 264.7 cells. In the presence of IFN-γ, UVACA significantly promoted IL-10 but not IL-6 and PGE2 secretion from RAW 264.7 cells and showed no effect on IFN-γ-induced MHC II expression. Compared with UVACA, NCA significantly suppressed IFN-γ-induced MHC II expression and significantly promoted IL-6, PGE2 and IL-10 secretion from RAW 264.7 cells. Conclusion RAW 264.7 cells respond differently to NCA and UVACA. NCA can significantly suppress IFN-γ-induced MHC II expression and significantly promote IL-6, IL-10 and PGE2 secretion from RAW 264.7 cells compared with UVACA.
Collapse
|
43
|
de Oliveira Fraga LA, Torrero MN, Tocheva AS, Mitre E, Davies SJ. Induction of type 2 responses by schistosome worms during prepatent infection. J Infect Dis 2010; 201:464-72. [PMID: 20043751 DOI: 10.1086/649841] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
During natural schistosome infection, the induction of T helper type 2 (Th2) responses has been ascribed to parasite eggs, because exposure of the host to this life-cycle stage elicits a polarized Th2 response to egg antigens. In the present study, we show that schistosome worms also elicit systemic, antigen-specific type 2 responses during prepatent infection, before egg deposition begins. CD4(+) T cells producing interleukin (IL)-4 were induced by both male and female worms during single-sex infections, demonstrating that this response is independent of exposure to eggs. The Th2 response was accompanied by production of immunoglobulin E and the sensitization of circulating basophils to produce additional IL-4 in response to schistosome antigens. Together, our data show that schistosome worms establish an immunologic milieu where CD4(+) T cells and basophils are both primed to produce IL-4 before eggs are laid, suggesting that worms play a role in establishment of the Th2 response that is critical for host survival and parasite transmission.
Collapse
Affiliation(s)
- Lucia Alves de Oliveira Fraga
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | | | | | | | | |
Collapse
|
44
|
Paveley RA, Aynsley SA, Cook PC, Turner JD, Mountford AP. Fluorescent imaging of antigen released by a skin-invading helminth reveals differential uptake and activation profiles by antigen presenting cells. PLoS Negl Trop Dis 2009; 3:e528. [PMID: 19829705 PMCID: PMC2759291 DOI: 10.1371/journal.pntd.0000528] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 09/10/2009] [Indexed: 11/28/2022] Open
Abstract
Infection of the mammalian host by the parasitic helminth Schistosoma mansoni is accompanied by the release of excretory/secretory molecules (ES) from cercariae which aid penetration of the skin. These ES molecules are potent stimulants of innate immune cells leading to activation of acquired immunity. At present however, it is not known which cells take up parasite antigen, nor its intracellular fate. Here, we develop a technique to label live infectious cercariae which permits the imaging of released antigens into macrophages (MΦ) and dendritic cells (DCs) both in vitro and in vivo. The amine reactive tracer CFDA-SE was used to efficiently label the acetabular gland contents of cercariae which are released upon skin penetration. These ES products, termed ‘0-3hRP’, were phagocytosed by MHC-II+ cells in a Ca+ and actin-dependent manner. Imaging of a labelled cercaria as it penetrates the host skin over 2 hours reveals the progressive release of ES material. Recovery of cells from the skin shows that CFDA-SE labelled ES was initially (3 hrs) taken up by Gr1+MHC-II− neutrophils, followed (24 hrs) by skin-derived F4/80+MHC-IIlo MΦ and CD11c+ MHC-IIhi DC. Subsequently (48 hrs), MΦ and DC positive for CFDA-SE were detected in the skin-draining lymph nodes reflecting the time taken for antigen-laden cells to reach sites of immune priming. Comparison of in vitro-derived MΦ and DC revealed that MΦ were slower to process 0-3hRP, released higher quantities of IL-10, and expressed a greater quantity of arginase-1 transcript. Combined, our observations on differential uptake of cercarial ES by MΦ and DC suggest the development of a dynamic but ultimately balanced response that can be potentially pushed towards immune priming (via DC) or immune regulation (via MΦ). Schistosomiasis is caused by the parasitic worm Schistosoma with over 200 million people infected across 76 countries. The parasitic larvae (called cercariae) infect mammalian hosts via the skin, but the exact mechanisms by which dermal cells interact with molecules released by invading larvae are unclear. A better understanding of the infection process and stimulation of the early immune response would thus enable a targeted approach towards the development of drugs and vaccines. Here, we have used the fluorescent tracer CFDA-SE to label infectious cercariae and, together with confocal microscopy, have for the first time tracked in real time the parasite infecting via the epidermis and depositing excretory/secretory material in its wake. Phagocytic macrophages and dendritic cells in the skin internalised excretory/secretory molecules released by the larvae, and both cell types were subsequently located in the draining lymph nodes where priming of the acquired immune response occurs. In vitro studies determined that macrophages were slower to process released parasite material than dendritic cells; they also secreted lower levels of pro-inflammatory cytokines but greater quantities of regulatory IL-10. The relative abundance of macrophages versus dendritic cells in the skin infection site and their differential rates of antigen processing may be crucial in determining the success of adaptive immune priming in response to infection.
Collapse
Affiliation(s)
- Ross A. Paveley
- Department of Biology, The University of York, York, United Kingdom
| | - Sarah A. Aynsley
- Department of Biology, The University of York, York, United Kingdom
| | - Peter C. Cook
- Department of Biology, The University of York, York, United Kingdom
| | - Joseph D. Turner
- Department of Biology, The University of York, York, United Kingdom
| | - Adrian P. Mountford
- Department of Biology, The University of York, York, United Kingdom
- * E-mail:
| |
Collapse
|
45
|
Durães FV, Carvalho NB, Melo TT, Oliveira SC, Fonseca CT. IL-12 and TNF-alpha production by dendritic cells stimulated with Schistosoma mansoni schistosomula tegument is TLR4- and MyD88-dependent. Immunol Lett 2009; 125:72-7. [PMID: 19539649 DOI: 10.1016/j.imlet.2009.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 06/01/2009] [Accepted: 06/08/2009] [Indexed: 12/31/2022]
Abstract
Schistosoma mansoni schistosomula are the most susceptible parasite life stage to host immune system attack. Complex host-parasite interactions take place on Schistosoma tegument, which is a unique double membrane structure involved in nutrition and immune evasion. Herein, we have demonstrated that schistosomula tegument (Smteg) activates Dendritic cells to produce IL-12p40, TNF-alpha and also to up-regulate the co-stimulatory molecules CD40 and CD86. Moreover, using DCs derived from MyD88-, TLR2-, TLR4- and TLR9-deficient mice we have shown that the ability of Smteg to activate DCs to produce IL-12 and TNF-alpha involves TLR4/Smteg interaction and MyD88 signaling pathway. Finally, our findings lead us to conclude that TLR4 is a key receptor involved in Smteg induction of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Fernanda V Durães
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, A. Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | |
Collapse
|
46
|
Hu S, Wu Z, Yang L, Fung MC. Molecular cloning and expression of a functional anti-inflammatory protein, Sj16, of Schistosoma japonicum. Int J Parasitol 2008; 39:191-200. [PMID: 18804475 DOI: 10.1016/j.ijpara.2008.06.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 06/02/2008] [Accepted: 06/30/2008] [Indexed: 10/21/2022]
Abstract
Schistosomes are the causative agent of schistosomiasis. In the infected host, significant inflammatory response to the parasite is not observed. Previous studies of Schistosoma mansoni showed that this subdued inflammatory response was due to a 16-kDa protein, Sm16, which is present in high levels in the secretions of schistosomula. Here we report the cloning and characterization of a gene (named Sj16) from Schistosoma japonicum. Sequence analysis showed that Sj16 shares 99% identity with Sm16 in its nucleotide sequence, and 100% identity in its protein sequence. While previous studies reportedly failed to obtain the soluble recombinant protein of Sm16, we expressed and purified recombinant Sj16 (rSj16) from Escherichia coli. Western blot and ELISA analyses showed that S. japonicum-infected rabbit sera could not recognize rSj16, indicating that native Sj16 may fail to induce circulating antibodies during S. japonicum infection. In vivo, rSj16 dramatically suppressed the recruitment of thioglycollate-mediated leukocytes to the peritoneal cavity of BALB/c mice, accompanied by marked up-regulation of IL-10 and IL-1RA transcripts, and down-regulation of IL-12p35, IL-1 beta and MIP-2 transcripts in peritoneal cells. Further analysis revealed that rSj16 also suppressed thioglycollate-induced peritoneal macrophage maturation. These results demonstrate that rSj16 has an anti-inflammatory function.
Collapse
Affiliation(s)
- Shaomin Hu
- Department of Biology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | | | | |
Collapse
|
47
|
Ferret-Bernard S, Curwen RS, Mountford AP. Proteomic profiling reveals that Th2-inducing dendritic cells stimulated with helminth antigens have a 'limited maturation' phenotype. Proteomics 2008; 8:980-93. [PMID: 18324723 DOI: 10.1002/pmic.200700538] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Dendritic cells (DCs) are important in the initiation of primary immune responses against pathogens. To aid understanding of how DCs guide T helper (Th)2-type responses, we employed 2-DE in association with MS/MS to identify proteins which characterise pro-Th2 DCs (matured with zero-to-three hours released proteins (0-3hRP), released by Schistosoma mansoni cercariae) versus pro-Th1 DCs (matured with lipopolysaccharide, LPS) and immature DCs. Software analysis of average 2-DE gels (three replicates per DC type) showed many similarities in the pattern of spots between the three groups of DCs but also marked changes. The major and significant changes in protein expression mainly affected cytoskeletal proteins. Other differences included chaperone proteins and enzymes involved in protein folding, S100 calcium-binding proteins, peroxiredoxin 1, superoxide dismutase 1, several annexins and arginase 1. Our study demonstrates that pro-Th2 DCs matured with 0-3hRP exhibit a proteome that is intermediate between that of immature DCs and pro-Th1 DCs. Finally, the differential regulation of protein spots identified by MALDI-MS/MS as having cytoskeletal and morphological functions was confirmed by contrast, confocal and scanning electron microscopy examination of DCs. Together, our results support the view that Th2 differentiation results from a 'limited maturation' of DCs.
Collapse
|
48
|
Scott JE, ElKhal A, Freyschmidt EJ, MacArthur DH, McDonald D, Howell MD, Leung DYM, Laouar A, Manjunath N, Bianchi T, Boes M, Oettgen HC, Geha RS. Impaired immune response to vaccinia virus inoculated at the site of cutaneous allergic inflammation. J Allergy Clin Immunol 2007; 120:1382-8. [PMID: 17889291 DOI: 10.1016/j.jaci.2007.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Revised: 07/30/2007] [Accepted: 08/01/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Patients with atopic dermatitis (AD) exposed to the vaccinia virus (VV) smallpox vaccine have an increased risk of developing eczema vaccinatum. OBJECTIVE To investigate the effects of local allergic skin inflammation on vaccinia immunity. METHODS BALB/c mice were epicutaneously sensitized with ovalbumin (OVA) to induce allergic skin inflammation or with saline control, then inoculated with an attenuated VV strain by skin scarification or intraperitoneally. After 8 days, serum IgG anti-VV and cytokine secretion by splenocytes were measured. RESULTS Mice inoculated with VV at sites of epicutaneous sensitization with OVA, but not control mice inoculated at saline exposed sites, developed satellite pox lesions and had impaired secretion of T(H)1 cytokines in response to VV, decreased VV specific serum IgG(2a), increased VV specific serum IgG(1), and impaired upregulation of IFN-alpha, but not the cathelicidin-related antimicrobial peptide, at the infection site. The VV immune response of OVA-sensitized mice inoculated with VV at distant skin sites or intraperitoneally was normal. CONCLUSION Local immune dysregulation at sites of allergic skin inflammation underlies the impaired T(H)1 immune response to VV introduced at these sites and the increased susceptibility to develop satellite pox lesions, a characteristic of eczema vaccinatum in patients with AD. CLINICAL IMPLICATIONS In a mouse model of AD, inoculation of VV at inflamed skin sites is associated with increased numbers of satellite pox lesions and an abnormal immune response to the virus. This may contribute to the susceptibility of patients with AD to virus dissemination after smallpox vaccination.
Collapse
Affiliation(s)
- Jordan E Scott
- Department of Pediatrics, Division of Allergy and Immunology, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hewitson JP, Hamblin PA, Mountford AP. In the absence of CD154, administration of interleukin-12 restores Th1 responses but not protective immunity to Schistosoma mansoni. Infect Immun 2007; 75:3539-47. [PMID: 17485453 PMCID: PMC1932915 DOI: 10.1128/iai.00252-07] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cytokine interplay during the development of protective immunity to the radiation-attenuated (RA) schistosome vaccine has been extensively characterized over recent years, yet the role of costimulatory molecules in the development of cell-mediated immunity is much less well understood. Here we demonstrate the importance of CD40/CD154 in vaccine-induced immunity, as CD154(-/-) mice exposed to RA schistosomes develop no protection to challenge infection. We showed that vaccinated CD154(-/-) mice have defective Th1-associated immune responses in the skin-draining lymph nodes and the lungs, with reduced or absent levels of interleukin-12p40 (IL-12p40), gamma interferon, and nitric oxide, but elevated levels of lung IL-4 and IL-5. The expression of major histocompatibility complex II (MHC-II) on antigen-presenting cells recovered from the lungs of vaccinated CD154(-/-) mice was also severely compromised. The administration of anti-CD40 monoclonal antibody (MAb) to CD154(-/-) mice did not reconstitute sustained Th1 responses in the lymph nodes or the lungs, nor did the MAb restore anti-parasite immunoglobulin G production or protective immunity. On the other hand, the administration of recombinant IL-12 (rIL-12) to CD154(-/-) mice shortly after vaccination caused elevated and sustained levels of Th1-associated cytokines, rescued MHC-II expression by lung CD11c(+) cells, and restored the appearance of inflammatory effector foci in the lungs. However, the treatment of CD154(-/-) mice with rIL-12 did not restore protection. We conclude that protective immunity to the RA schistosome vaccine is CD154 dependent but is independent of IL-12-orchestrated cellular immune mechanisms in the lungs.
Collapse
Affiliation(s)
- James P Hewitson
- Department of Biology (Area 5), University of York, York, United Kingdom
| | | | | |
Collapse
|
50
|
Kumkate S, Jenkins GR, Paveley RA, Hogg KG, Mountford AP. CD207+ Langerhans cells constitute a minor population of skin-derived antigen-presenting cells in the draining lymph node following exposure to Schistosoma mansoni. Int J Parasitol 2006; 37:209-20. [PMID: 17157855 PMCID: PMC1847335 DOI: 10.1016/j.ijpara.2006.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 08/30/2006] [Accepted: 10/20/2006] [Indexed: 11/25/2022]
Abstract
Infectious cercariae of Schistosoma mansoni gain entry to the mammalian host through the skin where they induce a transient inflammatory influx of mononuclear cells. Some of these cells have antigen-presenting cell function (MHCII+) and have been reported to migrate to the skin-draining lymph nodes (sdLN) where they have the potential to prime CD4+ cells of the acquired immune response. Here, in mice exposed to vaccinating radiation-attenuated schistosome larvae, which induce high levels of protective immunity to challenge infection, we describe the parasite-induced migration of Langerhans cells (LCs) from the epidermal site of immunisation to the sdLN using a specific monoclonal antibody that recognises langerin (CD207). CD207+ cells with dendritic morphology were abundant in the epidermis at all times and their migration into the dermis was detected soon after vaccination. All CD207+ LCs were MHCII+ but not all MHCII+ cells in the skin were CD207+. LCs migrated from the dermis in enhanced numbers after vaccination, as detected in dermal exudate populations recovered after in vitro culture of skin biopsies. Elevated numbers of CD207+ LCs were also detected in the sdLN from 24 h to 4 days after vaccination. However, compared with other dermal-derived antigen-presenting cells that were CD207−MHCII+ or CD207−CD11c+, the relative numbers of CD207+ cells in the dermal exudate population and in the sdLN were very small. Furthermore, the migration of CD207+ cells after exposure to ‘protective’ radiation-attenuated, compared with ‘non-protective’ normal cercariae, was similar in terms of numbers and kinetics. Together, these studies suggest that CD207+ LCs are only a minor component of the antigen-presenting cell population that migrates from the epidermis and they are unlikely to be important in the priming of protective CD4+ cells in the sdLN.
Collapse
|