1
|
Weng M, Zhang R, Zhang Z, Wu J, Zheng W, Lu Q, Long S, Liu R, Wang Z, Cui J. A Novel Trichinella spiralis Galectin Strengthens the Macrophage ADCC Killing of Larvae via Driving M1 Polarization. Int J Mol Sci 2024; 25:10920. [PMID: 39456703 PMCID: PMC11506943 DOI: 10.3390/ijms252010920] [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: 09/08/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Galectin recognizes β-galactosides through its carbohydrate recognition domains (CRDs). This study aimed to determine the biological features of a novel Trichinella spiralis galectin (galactoside-binding lectin family protein, TsGLFP) and its role in driving macrophage M1 polarization and enhancing ADCC killing of larvae. TsGLFP belongs to the galectin family and has two CRDs. The complete TsGLFP cDNA sequence was cloned and then expressed in Escherichia coli BL21. The results of qPCR, Western blot, and indirect immunofluorescence tests (IIFTs) revealed that TsGLFP was expressed in various stages of T. spiralis worms and principally localized at the cuticle and around the female embryos of the nematode. rTsGLFP had the function of agglutinating mouse erythrocytes, and this agglutination activity could be inhibited by lactose. After the mouse macrophage RAW264.7 was incubated with rTsGLFP, the expression level of the M1 genes (iNOS, IL-6, and TNF-α) and NO production were obviously increased. After incubating macrophages with rTsGLFP, there was a noticeable rise in the expression levels of p-IκB-α and p-NF-κB p65. Additionally, rTsGLFP enhanced the macrophage's ability to kill newborn larvae by ADCC cytotoxicity. When the macrophages were pretreated with the specific p-NF-κB p65 inhibitor PDTC, and then stimulated with rTsGLFP, the expression levels of iNOS, NO, and p-NF-κB p65 and the macrophages' ADCC cytotoxicity were distinctly decreased. These findings indicated that rTsGLFP enhanced the macrophage ADCC killing of larvae by driving M1 polarization through activating the NF-κB pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhongquan Wang
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (M.W.); (R.Z.); (Z.Z.); (J.W.); (W.Z.); (Q.L.); (S.L.); (R.L.)
| | - Jing Cui
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (M.W.); (R.Z.); (Z.Z.); (J.W.); (W.Z.); (Q.L.); (S.L.); (R.L.)
| |
Collapse
|
2
|
Abstract
Complement factor D (FD) is a serine protease that plays an essential role in the activation of the alternative pathway (AP) by cleaving complement factor B (FB) and generating the C3 convertases C3(H2 O)Bb and C3bBb. FD is produced mainly from adipose tissue and circulates in an activated form. On the contrary, the other serine proteases of the complement system are mainly synthesized in the liver. The activation mechanism of FD has long been unknown. Recently, a serendipitous discovery in the mechanism of FD activation has been provided by a generation of Masp1 gene knockout mice lacking both the serine protease MASP-1 and its alternative splicing variant MASP-3, designated MASP-1/3-deficient mice. Sera from the MASP-1/3-deficient mice had little-to-no lectin pathway (LP) and AP activity with circulating zymogen or proenzyme FD (pro-FD). Sera from patients with 3MC syndrome carrying mutations in the MASP1 gene also had circulating pro-FD, suggesting that MASP-1 and/or MASP-3 are involved in activation of FD. Here, we summarize the current knowledge of the mechanism of FD activation that was finally elucidated using the sera of mice monospecifically deficient for MASP-1 or MASP-3. Sera of the MASP-1-deficient mice lacked LP activity, but those of the MASP-3-deficient mice lacked AP activity with pro-FD. This review illustrates the pivotal role of MASP-3 in the physiological activation of the AP via activation of FD.
Collapse
Affiliation(s)
- Hideharu Sekine
- Department of Immunology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takeshi Machida
- Department of Immunology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Teizo Fujita
- Fukushima Prefectural General Hygiene Institute, Fukushima, Japan
| |
Collapse
|
3
|
Infection and Immunity. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
4
|
Hao HN, Song YY, Ma KN, Wang BN, Long SR, Liu RD, Zhang X, Wang ZQ, Cui J. A novel C-type lectin from Trichinella spiralis mediates larval invasion of host intestinal epithelial cells. Vet Res 2022; 53:85. [PMID: 36258242 PMCID: PMC9580147 DOI: 10.1186/s13567-022-01104-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/05/2022] [Indexed: 11/10/2022] Open
Abstract
The aim of this study was to investigate the characteristics of a novel type C lectin from Trichinella spiralis (TsCTL) and its role in larval invasion of intestinal epithelial cells (IECs). TsCTL has a carbohydrate recognition domain (CRD) of C-type lectin. The full-length TsCTL cDNA sequence was cloned and expressed in Escherichia coli BL21. The results of qPCR, Western blotting and immunofluorescence assays (IFAs) showed that TsCTL was a surface and secretory protein that was highly expressed at the T. spiralis intestinal infective larva (IIL) stages and primarily located at the cuticle, stichosome and embryos of the parasite. rTsCTL could specifically bind with IECs, and the binding site was localized in the IEC nucleus and cytoplasm. The IFA results showed that natural TsCTL was secreted and bound to the enteral epithelium at the intestinal stage of T. spiralis infection. The rTsCTL had a haemagglutinating effect on murine erythrocytes, while mannose was able to inhibit the rTsCTL agglutinating effect for mouse erythrocytes. rTsCTL accelerated larval intrusion into the IECs, whereas anti-rTsCTL antibodies and mannose significantly impeded larval intrusion in a dose-dependent manner. The results indicated that TsCTL specifically binds to IECs and promotes larval invasion of intestinal epithelium, and it might be a potential target of vaccines against T. spiralis enteral stages.
Collapse
Affiliation(s)
- Hui Nan Hao
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Yan Yan Song
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Kai Ning Ma
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Bo Ning Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Xi Zhang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| |
Collapse
|
5
|
Freeley SJ, Tham EL, Robson MG. The lectin pathway does not contribute to glomerular injury in the nephrotoxic nephritis model. Nephrology (Carlton) 2021; 27:208-214. [PMID: 34676615 DOI: 10.1111/nep.13988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/17/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022]
Abstract
AIMS Rapidly progressive crescentic glomerulonephritis occurs in number systemic and primary glomerular diseases, including anti-glomerular basement membrane disease, anti-neutrophil cytoplasmic antibody vasculitis and lupus nephritis. Our understanding of pathogenic mechanisms comes from animal models of disease such as the nephrotoxic nephritis model. The lectin pathway of complement activation has been shown to play a key role in several models of inflammation including renal ischaemia reperfusion. However, the lectin pathway is not required for crescentic glomerulonephritis in the anti-myeloperoxidase model of anti-neutrophil cytoplasmic antibody vasculitis. The aim of the current study was to explore the role of the lectin pathway in the nephrotoxic nephritis model, which is another model of crescentic glomerulonephritis. METHODS Nephrotoxic nephritis was induced in wild type and mannan-binding lectin-associated serine protease-2 deficient mice. Diseases were assessed by quantifying glomerular crescents and macrophages, in addition to albuminuria and serum creatinine. RESULTS There was no difference between wild type and MASP-2 deficient mice in any of the histological or biochemical parameters of disease assessed. In addition, there was no difference in the humoral immune response to sheep IgG. CONCLUSION These data show that the lectin pathway of complement activation is not required for the development of crescentic glomerulonephritis in the nephrotoxic nephritis model, reinforcing previous findings in the anti-myeloperoxidase model.
Collapse
Affiliation(s)
- Simon J Freeley
- Life Sciences and Medicine, King's College London, London, UK
| | - El Li Tham
- Life Sciences and Medicine, King's College London, London, UK
| | | |
Collapse
|
6
|
Wu X, Yao D, Bao L, Liu D, Xu X, An Y, Zhang X, Cao B. Ficolin A derived from local macrophages and neutrophils protects against lipopolysaccharide-induced acute lung injury by activating complement. Immunol Cell Biol 2020; 98:595-606. [PMID: 32339310 DOI: 10.1111/imcb.12344] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022]
Abstract
Ficolins are important and widely distributed pattern recognition molecules that can induce lectin complement pathway activation and initiate the innate immune response. Although ficolins can bind lipopolysaccharide (LPS) in vitro, the sources, dynamic changes and roles of local ficolins in LPS-induced pulmonary inflammation and injury remain poorly understood. In this study, we established a ficolin knockout mouse model by clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) technology, and used flow cytometry and hematoxylin and eosin staining to study the expressions and roles of local ficolins in LPS-induced pulmonary inflammation and injury. Our results show that besides ficolin B (FcnB), ficolin A (FcnA) is also expressed in leukocytes from the bone marrow, peripheral blood, lung and spleen. Further analyses showed that macrophages and neutrophils are the main sources of FcnA and FcnB, and T and B cells also express a small amount of FcnB. The intranasal administration of LPS induced local pulmonary inflammation with the increased recruitment of macrophages and neutrophils. LPS stimulation induced increased expression of FcnA and FcnB in neutrophils at the acute stage and in macrophages at the late stage. The severity of the lung injury and local inflammation of Fcna-/- mice was increased by the induction of extracellular complement activation. The recovery of LPS-induced local lung inflammation and injury was delayed in Fcnb-/- mice. Hence, these findings suggested that the local macrophage- and neutrophil-derived FcnA protects against LPS-induced acute lung injury by mediating extracellular complement activation.
Collapse
Affiliation(s)
- Xu Wu
- National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100029, China
| | - Duoduo Yao
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Linlin Bao
- NHC Key Laboratory of Human Disease Comparative Medicine , Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Di Liu
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoxue Xu
- Department of Core Facility Center, Capital Medical University, Beijing, 100069, China
| | - Yunqing An
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xulong Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Bin Cao
- National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100029, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100006, China
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, 100084, China
| |
Collapse
|
7
|
Deslyper G, Holland CV, Colgan TJ, Carolan JC. The liver proteome in a mouse model for Ascaris suum resistance and susceptibility: evidence for an altered innate immune response. Parasit Vectors 2019; 12:402. [PMID: 31412915 PMCID: PMC6693097 DOI: 10.1186/s13071-019-3655-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/03/2019] [Indexed: 01/08/2023] Open
Abstract
Background Ascariasis is a neglected tropical disease that affects 800 million people worldwide. Whereas most people only experience light worm burden, some people experience heavy worm burdens even after several rounds of chemotherapy, a phenomenon known as predisposition. Such heavy infections are associated with more severe symptoms and increased chronic morbidity. Methods In order to investigate potential mechanisms that may explain the observed predisposition, we infected mice with the porcine ascarid Ascaris suum using an established mouse model with two different mouse strains, where the C57BL/6J strain is more susceptible to infection and therefore a model for heavy infection and the CBA/Ca strain is more resistant and thus a model for light infection. At day 7 post-infection we investigated the liver proteome, using shotgun mass spectrometry, of both infected and control mice of each strain. Results We identified intrinsic differences, between the two mouse strains, in both oxidative phosphorylation proteins and proteins involved in retinol metabolism. Additionally, we found differences between the two mouse strains in activation of the complement system, where the CBA/Ca strain has higher protein abundances for lectin pathway proteins and the C57BL/6J strain has higher protein abundances for complement inhibiting proteins. The CBA/Ca strain had a higher abundance of proteins involved in the activation of the complement cascade via the lectin pathway. In contrast, the C57BL/6J strain demonstrated a higher abundance of proteins involved in arresting the complement pathway. Conclusions We observed clear differences between the two mouse strains both intrinsically and under infection. Electronic supplementary material The online version of this article (10.1186/s13071-019-3655-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gwendoline Deslyper
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, Dublin 2, Ireland.
| | - Celia V Holland
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Thomas J Colgan
- School of Biological, Earth and Environmental Sciences, University College Cork, Cork, Ireland
| | - James C Carolan
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|
8
|
Wang P, Zhuo XR, Tang L, Liu XS, Wang YF, Wang GX, Yu XQ, Wang JL. C-type lectin interacting with β-integrin enhances hemocytic encapsulation in the cotton bollworm, Helicoverpa armigera. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2017; 86:29-40. [PMID: 28572000 DOI: 10.1016/j.ibmb.2017.05.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/02/2017] [Accepted: 05/28/2017] [Indexed: 06/07/2023]
Abstract
The encapsulation reaction in invertebrates is analogous to granuloma formation in vertebrates, and this reaction is severely compromised when ecdysone signaling is blocked. However, the molecular mechanism underlying the encapsulation reaction and its regulation by ecdysone remains obscure. In our previous study, we found that the C-type lectin HaCTL3, from the cotton bollworm Helicoverpa armigera, is involved in anti-bacterial immune response, acting as a pattern recognition receptor (PRR). In the current study, we demonstrate that HaCTL3 is involved in defense against parasites and directly binds to the surface of nematodes. Our in vitro and in vivo studies indicate that HaCTL3 enhances hemocytic encapsulation and melanization, whereas H. armigera β-integrin (Haβ-integrin), located on the surface of hemocytes, participates in encapsulation. Additionally, co-immunoprecipitation experiments reveal HaCTL3 interacts with Haβ-integrin, and knockdown of Haβ-integrin leads to reduced encapsulation of HaCTL3-coated beads. These results indicate that Haβ-integrin serves as a hemocytic receptor of HaCTL3 during the encapsulation reaction. Furthermore, we demonstrate that 20-hydroxyecdysone (20E) treatment dramatically induces the expression of HaCTL3, and knockdown of the 20E receptor (EcR)/ultraspiracle (USP), abrogates this response. Overall, this study provides the first evidence of the presence of a hemocytic receptor (Haβ-integrin), that interacts with the PRR HaCTL3 to facilitate encapsulation reaction in insects and demonstrates the regulation of this process by the steroid hormone ecdysone.
Collapse
Affiliation(s)
- Pan Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xiao-Rong Zhuo
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Lin Tang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xu-Sheng Liu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Yu-Feng Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Guo-Xiu Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xiao-Qiang Yu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China; School of Biological Sciences, University of Missouri, Kansas City, MO 64110, USA
| | - Jia-Lin Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| |
Collapse
|
9
|
Yaseen S, Demopulos G, Dudler T, Yabuki M, Wood CL, Cummings WJ, Tjoelker LW, Fujita T, Sacks S, Garred P, Andrew P, Sim RB, Lachmann PJ, Wallis R, Lynch N, Schwaeble WJ. Lectin pathway effector enzyme mannan-binding lectin-associated serine protease-2 can activate native complement C3 in absence of C4 and/or C2. FASEB J 2017; 31:2210-2219. [PMID: 28188176 DOI: 10.1096/fj.201601306r] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/23/2017] [Indexed: 02/06/2023]
Abstract
All 3 activation pathways of complement-the classic pathway (CP), the alternative pathway, and the lectin pathway (LP)- converge into a common central event: the cleavage and activation of the abundant third complement component, C3, via formation of C3-activating enzymes (C3 convertases). The fourth complement component, C4, and the second component, C2, are indispensable constituents of the C3 convertase complex, C4bC2a, which is formed by both the CP and the LP. Whereas in the absence of C4, CP can no longer activate C3, LP retains a residual but physiologically critical capacity to convert native C3 into its activation fragments, C3a and C3b. This residual C4 and/or C2 bypass route is dependent on LP-specific mannan-binding lectin-associated serine protease-2. By using various serum sources with defined complement deficiencies, we demonstrate that, under physiologic conditions LP-specific C4 and/or C2 bypass activation of C3 is mediated by direct cleavage of native C3 by mannan-binding lectin-associated serine protease-2 bound to LP-activation complexes captured on ligand-coated surfaces.-Yaseen, S., Demopulos, G., Dudler, T., Yabuki, M., Wood, C. L., Cummings, W. J., Tjoelker, L. W., Fujita, T., Sacks, S., Garred, P., Andrew, P., Sim, R. B., Lachmann, P. J., Wallis, R., Lynch, N., Schwaeble, W. J. Lectin pathway effector enzyme mannan-binding lectin-associated serine protease-2 can activate native complement C3 in absence of C4 and/or C2.
Collapse
Affiliation(s)
- Sadam Yaseen
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, United Kingdom.,Omeros Corporation, Seattle, Washington, USA.,Department of Biology, University of Mosul, Mosul, Iraq
| | | | | | | | | | | | | | - Teizo Fujita
- Fukushima Prefectural General Hygiene Institute, Fukushima Medical University, Fukushima City, Japan
| | - Steven Sacks
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Campus, London, United Kingdom
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Peter Andrew
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Robert B Sim
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Peter J Lachmann
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Russell Wallis
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Nicholas Lynch
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Wilhelm J Schwaeble
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, United Kingdom;
| |
Collapse
|
10
|
Nordmaj MA, Munthe-Fog L, Hein E, Skjoedt MO, Garred P. Genetically engineered fusion of MAP-1 and factor H domains 1-5 generates a potent dual upstream inhibitor of both the lectin and alternative complement pathways. FASEB J 2015; 29:4945-55. [PMID: 26260032 DOI: 10.1096/fj.15-277103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/03/2015] [Indexed: 01/01/2023]
Abstract
Inhibition of the complement cascade has emerged as an option for treatment of a range of diseases. Mannose-binding lectin/ficolin/collectin-associated protein (MAP-1) is a pattern recognition molecule (PRM)-associated inhibitor of the lectin pathway. The central regulator of the alternative pathway (AP) is complement factor H (FH). Our aim was to design a dual upstream inhibitor of both human lectin and APs by fusing MAP-1 with a part of FH. There were 2 different recombinant chimeric proteins comprising full-length human MAP-1 and the first 5 N-terminal domains of human FH designed. The FH domains were orientated either in the N- or C-terminal part of MAP-1. The complement inhibition potential in human serum was assessed. Both chimeric constructs displayed the characteristics of the native molecules and bound to the PRMs with an EC50 of ∼ 2 nM. However, when added to serum diluted 1:4 in a solid-phase functional assay, only the first 5 N-terminal domains of complement FH fused to the C-terminal part of full-length MAP-1 chimeric construct were able to combine inhibition of lectin and AP activation with an half maximal inhibitory concentration of ∼ 100 and 20 nM, respectively. No effect was seen on the classical pathway. Fusion of MAP-1 with FH domains represents a novel therapeutic approach for selective targeting upstream and central complement activation at sites of inflammation.
Collapse
Affiliation(s)
- Mie Anemone Nordmaj
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lea Munthe-Fog
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Estrid Hein
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel-Ole Skjoedt
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology-7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Robledo D, Ronza P, Harrison PW, Losada AP, Bermúdez R, Pardo BG, Redondo MJ, Sitjà-Bobadilla A, Quiroga MI, Martínez P. RNA-seq analysis reveals significant transcriptome changes in turbot (Scophthalmus maximus) suffering severe enteromyxosis. BMC Genomics 2014; 15:1149. [PMID: 25526753 PMCID: PMC4320470 DOI: 10.1186/1471-2164-15-1149] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 12/16/2014] [Indexed: 12/31/2022] Open
Abstract
Background Enteromyxosis caused by the intestinal myxozoan parasite Enteromyxum scophthalmi is a serious threat for turbot (Scophthalmus maximus, L.) aquaculture, causing severe catarrhal enteritis leading to a cachectic syndrome, with no therapeutic options available. There are still many aspects of host-parasite interaction and disease pathogenesis that are yet to be elucidated, and to date, no analysis of the transcriptomic changes induced by E. scophthalmi in turbot organs has been conducted. In this study, RNA-seq technology was applied to head kidney, spleen and pyloric caeca of severely infected turbot with the aim of furthering our understanding of the pathogenetic mechanisms and turbot immune response against enteromyxosis. Results A huge amount of information was generated with more than 23,000 identified genes in the three organs, amongst which 4,762 were differently expressed (DE) between infected and control fish. Associate gene functions were studied based on gene ontology terms and available literature, and the most interesting DE genes were classified into five categories: 1) immune and defence response; 2) apoptosis and cell proliferation; 3) iron metabolism and erythropoiesis; 4) cytoskeleton and extracellular matrix and 5) metabolism and digestive function. The analysis of down-regulated genes of the first category revealed evidences of a connexion failure between innate and adaptive immune response, especially represented by a high number of DE interferon-related genes in the three organs. Furthermore, we found an intense activation of local immune response at intestinal level that appeared exacerbated, whereas in kidney and spleen genes involved in adaptive immune response were mainly down-regulated. The apoptotic machinery was only clearly activated in pyloric caeca, while kidney and spleen showed a marked depression of genes related to erythropoiesis, probably related to disorders in iron homeostasis. The genetic signature of the causes and consequences of cachexia was also demonstrated by the down-regulation of the genes encoding structural proteins and those involved in the digestive metabolism. Conclusions This transcriptomic study has enabled us to gain a better understanding of the pathogenesis of enteromyxosis and identify a large number of DE target genes that bring us closer to the development of strategies designed to effectively combat this pathogen. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-1149) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - María Isabel Quiroga
- Departamento de Ciencias Clínicas Veterinarias, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo 27002, Spain.
| | | |
Collapse
|
12
|
Glycans expressed on Trichinella spiralis excretory-secretory antigens are important for anti-inflamatory immune response polarization. Comp Immunol Microbiol Infect Dis 2014; 37:355-67. [PMID: 25467036 DOI: 10.1016/j.cimid.2014.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/17/2014] [Accepted: 10/20/2014] [Indexed: 01/28/2023]
Abstract
Trichinella spiralis muscle larvae excretory-secretory antigens (ES L1) are most likely responsible for the induction of immune response during infection by this parasitic. The antigens bear carbohydrate structures that may contribute to immune system activation resulting in a Th2/anti-inflammatory immune response. We show that T. spiralis glycans affect the expression and the production of IL-4 and IL-10 in vivo. Alteration of carbohydrate structures on ES L1 altered dendritic cell (DC) maturation. Periodate treatment of ES L1 led to the reduction in both ERK and p38 phosphorylation which may be the cause of reduced IL-10 and IL-12p70 production. In vitro priming of naïve T cells with DCs stimulated with native and periodate-treated ES L1 emphasized the importance of intact glycans for IL-10 production. We conclude that T. spiralis glycans affect the anti-inflammatory environment and can interfere with the development of inflammatory diseases.
Collapse
|
13
|
Trichinella spiralis excretory-secretory products protect against polymicrobial sepsis by suppressing MyD88 via mannose receptor. BIOMED RESEARCH INTERNATIONAL 2014; 2014:898646. [PMID: 25054155 PMCID: PMC4098621 DOI: 10.1155/2014/898646] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/10/2014] [Accepted: 06/15/2014] [Indexed: 12/31/2022]
Abstract
Trichinella spiralis (T. spiralis) or its excretory-secretory products (TsES) protect hosts from autoimmune diseases, which depend on inducing host T helper (Th) 2 immune response and inhibiting inflammatory factors. Sepsis is a systemic inflammatory response syndrome (SIRS) evoked by infection. Little is known about the effects of helminths or their excretory-secretory products on sepsis. Here, we investigated the effects of TsES in a mice model of polymicrobial sepsis. TsES improved survival, reduced organ injury, and enhanced bacterial clearance in septic mice. To investigate the molecular mechanism, macrophages from septic patients or the control group were incubated with TsES. TsES reduced sepsis-inducing inflammatory cytokines mediated by Toll-like receptors (TLR) in vitro by suppressing TLR adaptor-transducer myeloid differentiation factor 88 (MyD88) and nuclear factor- (NF-)-κB. Furthermore, TsES upregulated mannose receptor (MR) expression during sepsis. MR blocking attenuated the effects of TsES on MyD88 and NF-κB expression. In vivo, MR RNAi reduced the survival rate of septic mice treated with TsES, suggesting that TsES-mediated protection against polymicrobial sepsis is dependent on MR. Thus, TsES administration might be a potential therapeutic strategy for treating sepsis.
Collapse
|
14
|
Asgari E, Farrar CA, Lynch N, Ali YM, Roscher S, Stover C, Zhou W, Schwaeble WJ, Sacks SH. Mannan-binding lectin-associated serine protease 2 is critical for the development of renal ischemia reperfusion injury and mediates tissue injury in the absence of complement C4. FASEB J 2014; 28:3996-4003. [PMID: 24868011 DOI: 10.1096/fj.13-246306] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 05/19/2014] [Indexed: 01/19/2023]
Abstract
Mannan-binding lectin-associated serine protease 2 (MASP-2) has been described as the essential enzyme for the lectin pathway (LP) of complement activation. Since there is strong published evidence indicating that complement activation via the LP critically contributes to ischemia reperfusion (IR) injury, we assessed the effect of MASP-2 deficiency in an isogenic mouse model of renal transplantation. The experimental transplantation model used included nephrectomy of the remaining native kidney at d 5 post-transplantation. While wild-type (WT) kidneys grafted into WT recipients (n=7) developed acute renal failure (control group), WT grafts transplanted into MASP-2-deficient recipients (n=7) showed significantly better kidney function, less C3 deposition, and less IR injury. In the absence of donor or recipient complement C4 (n=7), the WT to WT phenotype was preserved, indicating that the MASP-2-mediated damage was independent of C4 activation. This C4-bypass MASP-2 activity was confirmed in mice deficient for both MASP-2 and C4 (n=7), where the protection from postoperative acute renal failure was no greater than in mice with MASP-2 deficiency alone. Our study highlights the role of LP activation in renal IR injury and indicates that injury occurs through MASP-2-dependent activation events independent of C4.
Collapse
Affiliation(s)
- Elham Asgari
- Medical Research Council Centre for Transplantation, King's College London, Guy's Campus, London, UK; and
| | - Conrad A Farrar
- Medical Research Council Centre for Transplantation, King's College London, Guy's Campus, London, UK; and
| | - Nicholas Lynch
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, UK
| | - Youssif M Ali
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, UK
| | - Silke Roscher
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, UK
| | - Cordula Stover
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, UK
| | - Wuding Zhou
- Medical Research Council Centre for Transplantation, King's College London, Guy's Campus, London, UK; and
| | - Wilhelm J Schwaeble
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, UK
| | - Steven H Sacks
- Medical Research Council Centre for Transplantation, King's College London, Guy's Campus, London, UK; and
| |
Collapse
|
15
|
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: 64] [Impact Index Per Article: 4.9] [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
|
16
|
The effect of anti- Anisakis simplexantibody levels on C3 and C4 complement components in human sera. J Helminthol 2012; 86:197-201. [DOI: 10.1017/s0022149x11000241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AbstractPreviously, anin vitroeffect was observed on the complement system not only of the excretory-secretory products but also of somatic antigens from L3Anisakis simplexlarvae. In the present work the effect of anti-A. simplexspecific antibodies on C3 and C4 levels in human sera was investigated. Up to 309 samples of sera were tested to determine levels of C3 and C4 and anti-A. simplexantibodies, including immunoglobulins IgG, IgM, IgA and IgE. Significant differences were observed between levels of C3 and C4 and all immunoglobulins except for IgE. In the case of immunoglobulins, the probability that an anti-A. simplexpositive subject has a C3 deficiency was 3.8 times higher than a subject without specific antibodies. In conclusion, an association between elevated levels of anti-A. simplexantibodies and C3 and C4 deficiency was demonstrated.
Collapse
|
17
|
Trichinella spiralis paramyosin binds to C8 and C9 and protects the tissue-dwelling nematode from being attacked by host complement. PLoS Negl Trop Dis 2011; 5:e1225. [PMID: 21750743 PMCID: PMC3130009 DOI: 10.1371/journal.pntd.0001225] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 05/20/2011] [Indexed: 01/28/2023] Open
Abstract
Background Paramyosin is a thick myofibrillar protein found exclusively in invertebrates. Evidence suggested that paramyosin from helminths serves not only as a structural protein but also as an immunomodulatory agent. We previously reported that recombinant Trichinella spiralis paramyosin (Ts-Pmy) elicited a partial protective immunity in mice. In this study, the ability of Ts-Pmy to bind host complement components and protect against host complement attack was investigated. Methods and Findings In this study, the transcriptional and protein expression levels of Ts-Pmy were determined in T. spiralis newborn larva (NBL), muscle larva (ML) and adult worm developmental stages by RT-PCR and western blot analysis. Expression of Ts-Pmy at the outer membrane was observed in NBL and adult worms using immunogold electron microscopy and immunofluorescence staining. Functional analysis revealed that recombinant Ts-Pmy(rTs-Pmy) strongly bound to complement components C8 and C9 and inhibited the polymerization of C9 during the formation of the membrane attack complex (MAC). rTs-Pmy also inhibited the lysis of rabbit erythrocytes (ER) elicited by an alternative pathway-activated complement from guinea pig serum. Inhibition of native Ts-Pmy on the surface of NBL with a specific antiserum reduced larvae viability when under the attack of complement in vitro. In vivo passive transfer of anti-Ts-Pmy antiserum and complement-treated larvae into mice also significantly reduced the number of larvae that developed to ML. Conclusion These studies suggest that the outer membrane form of T. spiralis paramyosin plays an important role in the evasion of the host complement attack. Trichinellosis is a serious food borne parasitic disease caused by the consumption of meat contaminated with the infective larvae of Trichinella spiralis. The ability of the tissue-dwelling parasite to evade the host complement attack is essential for its survival and for establishing infection in the host. This study describes the expression of paramyosin, a muscular protein in invertebrates, on the surface of Trichinella spiralis and its role in the defense against the host complement attack as a survival strategy. Using a specific antiserum, expression of Trichinella spiralis paramyosin was detected on the outer membrane of the adult worms and newborn larvae. Functional analysis revealed that recombinant Trichinella spiralis paramyosin protein strongly bound human complement components C8 and C9 and inhibited the formation of the complement membrane attack complex. Neutralization with a specific antiserum greatly impaired the protective effect of paramyosin on the viability and infectivity of Trichinella spiralis newborn larva when under attack by complement. These studies suggest that the outer membrane form of Trichinella spiralis paramyosin plays an important role in the evasion of the host complement attack and is therefore a good target for vaccine and pharmaceutical development.
Collapse
|
18
|
Rees-Roberts D, Mullen LM, Gounaris K, Selkirk ME. Inactivation of the complement anaphylatoxin C5a by secreted products of parasitic nematodes. Int J Parasitol 2009; 40:527-32. [PMID: 19874826 PMCID: PMC2852653 DOI: 10.1016/j.ijpara.2009.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 10/04/2009] [Accepted: 10/08/2009] [Indexed: 11/30/2022]
Abstract
Given the importance of the complement anaphylatoxins in cellular recruitment during infection, the ability of secreted products from larval stages of Brugia malayi and Trichinella spiralis to influence C5a-mediated chemotaxis of human peripheral blood granulocytes in vitro was examined. Secreted products from B. malayi microfilariae almost completely abolished chemotaxis. This inhibition was blocked by phenylmethylsulphonyl fluoride, indicating the presence of a serine protease, which was subsequently shown to cleave C5a. In contrast, secreted products from T. spiralis infective larvae showed modest inhibition of C5a-mediated granulocyte chemotaxis, and this was blocked by potato carboxypeptidase inhibitor, an inhibitor of several metallocarboxypeptidases. Adult and larval stages of both parasites were demonstrated to secrete carboxypeptidases which cleaved hippuryl-L-lysine and hippuryl-L-arginine, and the T. spiralis enzyme was partially characterised. The data are discussed with reference to inflammation in parasitic nematode infection.
Collapse
Affiliation(s)
- Dominic Rees-Roberts
- Division of Cell & Molecular Biology, Department of Life Sciences, Imperial College London, London SW72AZ, UK
| | | | | | | |
Collapse
|
19
|
Salman HH, Irache JM, Gamazo C. Immunoadjuvant capacity of flagellin and mannosamine-coated poly(anhydride) nanoparticles in oral vaccination. Vaccine 2009; 27:4784-90. [DOI: 10.1016/j.vaccine.2009.05.091] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 04/20/2009] [Accepted: 05/31/2009] [Indexed: 12/26/2022]
|
20
|
Abstract
SUMMARYInfection with parasitic helminths takes a heavy toll on the health and well-being of humans and their domestic livestock, concomitantly resulting in major economic losses. Analyses have consistently revealed bioactive molecules in extracts of helminths or in their excretory/secretory products that modulate the immune response of the host. It is our view that parasitic helminths are an untapped source of immunomodulatory substances that, in pure form, could become new drugs (or models for drug design) to treat disease. Here, we illustrate the range of immunomodulatory molecules in selected parasitic trematodes, cestodes and nematodes, their impact on the immune cells in the host and how the host may recognize these molecules. There are many examples of the partial characterization of helminth-derived immunomodulatory molecules, but these have not yet translated into new drugs, reflecting the difficulty of isolating and fully characterizing proteins, glycoproteins and lipid-based molecules from small amounts of parasite material. However, this should not deter the investigator, since analytical techniques are now being used to accrue considerable structural information on parasite-derived molecules, even when only minute quantities of tissue are available. With the introduction of methodologies to purify and structurally-characterize molecules from small amounts of tissue and the application of high throughput immunological assays, one would predict that an assessment of parasitic helminths will yield a variety of novel drug candidates in the coming years.
Collapse
|
21
|
Perrigoue JG, Marshall FA, Artis D. On the hunt for helminths: innate immune cells in the recognition and response to helminth parasites. Cell Microbiol 2008; 10:1757-64. [PMID: 18505479 DOI: 10.1111/j.1462-5822.2008.01174.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The generation of protective immunity to helminth parasites is critically dependent upon the development of a CD4(+) T helper type 2 cytokine response. However, the host-parasite interactions responsible for initiating this response are poorly understood. This review will discuss recent advances in our understanding of how helminth-derived products are recognized by innate immune cells. Specifically, interactions between helminth excretory/secretory products and host Toll-like receptors and lectins will be discussed as well as the putative functions of helminth proteases and chitin in activating and recruiting innate immune cells. In addition, the functional significance of pattern recognition by epithelial cells, granulocytes, dendritic cells and macrophages including expression of alarmins, thymic stromal lymphopoetin, interleukin (IL)-25, IL-33 and Notch ligands in the development of adaptive anti-parasite Th2 cytokine responses will be examined.
Collapse
|
22
|
Giacomin PR, Gordon DL, Botto M, Daha MR, Sanderson SD, Taylor SM, Dent LA. The role of complement in innate, adaptive and eosinophil-dependent immunity to the nematode Nippostrongylus brasiliensis. Mol Immunol 2008; 45:446-55. [PMID: 17675237 DOI: 10.1016/j.molimm.2007.05.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Revised: 05/25/2007] [Accepted: 05/30/2007] [Indexed: 01/21/2023]
Abstract
Complement may be important for immunity to infection with parasitic helminths, by promoting the recruitment of leukocytes to infected tissues and by modulating the function of cytotoxic effector leukocytes. However, the importance of complement in vivo during helminth infection is poorly understood. In this study, mice lacking classical (C1q-deficient), alternative (factor B-deficient) or all pathways of complement activation (C3-deficient) were used to assess the role of complement in immunity to the nematode Nippostrongylus brasiliensis. Double-mutant complement-deficient/IL-5 transgenic (Tg) mice were used to determine if complement is required for the strong eosinophil-dependent resistance to this parasite. Complement activation on larvae (C3 deposition), extracellular eosinophil peroxidase activity, larval aggregation and eosinophil recruitment to the skin 30 min post-injection (p.i.) of larvae were reduced in factor B-deficient mice. Inhibition of the C5a receptor with the antagonist PMX53 impaired eosinophil and neutrophil recruitment to the skin. C3 deposition on larvae was minimal by 150 min p.i. and at this time cell adherence, larval aggregation, eosinophil recruitment and degranulation were complement-independent. Factor B and C3 deficiency were associated with higher lung larval burdens in primary infections. Complement-deficient/IL-5 Tg mice were highly resistant to N. brasiliensis, suggesting that eosinophils can limit infection in a complement-independent manner. Potent secondary immunity was similarly complement-independent. In conclusion, although the alternative pathway is important for parasite recognition and leukocyte recruitment early in N. brasiliensis infections, the parasite soon becomes resistant to complement and other factors can compensate to promote eosinophil-dependent immunity.
Collapse
Affiliation(s)
- Paul R Giacomin
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia 5005, Australia
| | | | | | | | | | | | | |
Collapse
|
23
|
Zaccone P, Burton OT, Cooke A. Interplay of parasite-driven immune responses and autoimmunity. Trends Parasitol 2007; 24:35-42. [PMID: 18055264 DOI: 10.1016/j.pt.2007.10.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 09/29/2007] [Accepted: 10/01/2007] [Indexed: 12/24/2022]
Abstract
As more facts emerge regarding the ways in which parasite-derived molecules modulate the host immune response, it is possible to envisage how a lack of infection by agents that once infected humans commonly might contribute to the rise in autoimmune disease. Through effects on cells of both the innate and adaptive arms of the immune response, parasites can orchestrate a range of outcomes that are beneficial not only to parasites, in terms of facilitating their life cycles, but also to their host, in limiting pathology.
Collapse
Affiliation(s)
- Paola Zaccone
- Department of Pathology, Tennis Court Road, Cambridge, CB2 1QP, UK
| | | | | |
Collapse
|
24
|
Abstract
Nematode parasites cause significant infections in both humans and animals. They are complex, multicellular organisms that present unique challenges for the host, in particular with respect to the recognition of their unusual surface structures by the innate defence system. The innate immune system is now recognized to be a critical component in the development of an adaptive effector response as well as a driver of vaccine-induced immunity. This paper will give an overview of current research on the innate barriers and immune mechanisms, cells, and receptors involved in the innate host response to nematode parasites. It will also review the 'nematode-associated molecular patterns' that may be specifically recognized by the host, in addition to other signals, such as nervous stimulation and tissue damage, that may alert the innate system to parasite invasion.
Collapse
Affiliation(s)
- M J de Veer
- Animal Biotechnology Research Laboratories, School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | | | | |
Collapse
|
25
|
Gruden-Movsesijan A, Milosavljevic LS. The involvement of the macrophage mannose receptor in the innate immune response to infection with parasite Trichinella spiralis. Vet Immunol Immunopathol 2006; 109:57-67. [PMID: 16135385 DOI: 10.1016/j.vetimm.2005.07.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2004] [Revised: 06/16/2005] [Accepted: 07/19/2005] [Indexed: 11/18/2022]
Abstract
The macrophage mannose receptor (MR) is a pattern recognition receptor of the innate immune system that binds to microbial structures bearing mannose, fucose and N-acetylglucosamine on their surface. The MR can mediate endocytosis and phagocytosis, as well as activation of macrophages and antigen presentation. Since Trichinella spiralis antigens are rich in oligomannose residues, we investigated whether a mannose-recognizing receptor, such as the MR, participated in the host-parasite interaction. The results show that the MR (either on the surface of macrophages or in the purified form) recognizes and binds components of T. spiralis muscle larvae. The presence of parasites provoked activation of peritoneal macrophages, which was indicated by down-regulation of MR expression, and the stimulation of NO secretion. In vitro stimulation of macrophages with T. spiralis components resulted in increased NO and IL-6 production. However, while the MR was partially involved in stimulation of NO production, it did not mediate IL-6 secretion.
Collapse
Affiliation(s)
- A Gruden-Movsesijan
- Institute for the Application of Nuclear Energy-INEP, Banatska 31b, 11080 Belgrade, Serbia and Montenegro.
| | | |
Collapse
|
26
|
Ambrosio AR, De Messias-Reason IJT. Leishmania (Viannia) braziliensis: interaction of mannose-binding lectin with surface glycoconjugates and complement activation. An antibody-independent defence mechanism. Parasite Immunol 2005; 27:333-40. [PMID: 16149991 DOI: 10.1111/j.1365-3024.2005.00782.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The activation of complement on the surface of Leishmania promastigotes appears to be an important factor for parasite infectivity in the mammalian host, allowing their attachment and the invasion of macrophages via complement receptors. Mannose-binding lectin (MBL) is a well-known complement activator and an efficient opsonine. We have investigated here whether serum and purified MBL bind to and promote lysis of live promastigotes of L. braziliensis; and evaluated the deposition of MBL, C1q, C4 and C3 on the parasite surface after interaction with non-immune normal human serum (NHS). We observed that both serum MBL and the purified MBL-MASP complex bind to the surface of L. braziliensis and that this binding occurred via the carbohydrate recognition domains of MBL. The binding of MBL, however, did not affect the lytic effect of complement on the parasites. The deposition of C1q, C4, C3 and parasite lysis was observed after incubation with NHS. EDTA but not EGTA abolished C3 deposition on the parasite surface, indicating the involvement of the alternative pathway in this process. Our results indicate that MBL binds to L. braziliensis and that this is mediated by a specific carbohydrate on the surface of parasites and provides evidence for antibody-independent mechanisms that complement activation on the parasite surface.
Collapse
Affiliation(s)
- A R Ambrosio
- Immunopathology Laboratory and Department of Clinical Pathology, Federal University of Parana, Curitiba, Brazil
| | | |
Collapse
|
27
|
Giacomin PR, Wang H, Gordon DL, Botto M, Dent LA. Loss of complement activation and leukocyte adherence as Nippostrongylus brasiliensis develops within the murine host. Infect Immun 2005; 73:7442-9. [PMID: 16239545 PMCID: PMC1273855 DOI: 10.1128/iai.73.11.7442-7449.2005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Complement activation and C3 deposition on the surface of parasitic helminths may be important for recruitment of leukocytes and for damage to the target organism via cell-mediated mechanisms. Inhibition of complement activation would therefore be advantageous to parasites, minimizing damage and enhancing migration through tissues. The aim of this study was to determine ex vivo if complement activation by, and leukocyte adherence to, the nematode Nippostrongylus brasiliensis change as the parasite matures and migrates through the murine host. Pathways of activation of complement and the mechanism of adherence of leukocytes were also defined using sera from mice genetically deficient in either C1q, factor B, C1q and factor B, C3, or C4. Substantive deposition of C3 and adherence of eosinophil-rich leukocytes were seen with infective-stage (L3) but not with lung-stage (L4) larvae. Adult intestinal worms had low to intermediate levels of both C3 and leukocyte binding. For L3 and adult worms, complement deposition was principally dependent on the alternative pathway. For lung-stage larvae, the small amount of C3 detected was dependent to similar degrees on both the lectin and alternative pathways. The classical pathway was not involved for any of the life stages of the parasite. These results suggest that in primary infections, the infective stage of N. brasiliensis is vulnerable to complement-dependent attack by leukocytes. However, within the first 24 h of infection, N. brasiliensis acquires the ability to largely avoid complement-dependent immune responses.
Collapse
Affiliation(s)
- Paul R Giacomin
- School of Molecular and Biomedical Science, University of Adelaide, North Terrace, Adelaide, South Australia, Australia 5005
| | | | | | | | | |
Collapse
|