1
|
Jayaraman A, Walachowski S, Bosmann M. The complement system: A key player in the host response to infections. Eur J Immunol 2024; 54:e2350814. [PMID: 39188171 DOI: 10.1002/eji.202350814] [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: 03/22/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024]
Abstract
Infections are one of the most significant healthcare and economic burdens across the world as underscored by the recent coronavirus pandemic. Moreover, with the increasing incidence of antimicrobial resistance, there is an urgent need to better understand host-pathogen interactions to design effective treatment strategies. The complement system is a key arsenal of the host defense response to pathogens and bridges both innate and adaptive immunity. However, in the contest between pathogens and host defense mechanisms, the host is not always victorious. Pathogens have evolved several approaches, including co-opting the host complement regulators to evade complement-mediated killing. Furthermore, deficiencies in the complement proteins, both genetic and therapeutic, can lead to an inefficient complement-mediated pathogen eradication, rendering the host more susceptible to certain infections. On the other hand, overwhelming infection can provoke fulminant complement activation with uncontrolled inflammation and potentially fatal tissue and organ damage. This review presents an overview of critical aspects of the complement-pathogen interactions during infection and discusses perspectives on designing therapies to mitigate complement dysfunction and limit tissue injury.
Collapse
Affiliation(s)
- Archana Jayaraman
- Department of Medicine, Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Sarah Walachowski
- Department of Medicine, Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Markus Bosmann
- Department of Medicine, Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
2
|
Haridevamuthu B, Raj D, Arshad A, Arockiaraj J. Comprehensive review of Argulus infestations in aquaculture: Biological impacts and advanced management strategies. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109851. [PMID: 39173980 DOI: 10.1016/j.fsi.2024.109851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
The aquaculture industry is hindered by various factors. One of the most noticeable factors is infection by parasites and pathogens. Argulus stands out as a prominent and economically significant ectoparasite in freshwater aquaculture. Argulus infestation causes severe immunomodulatory effects on its hosts by promoting argulosis, causing inflammation, extensive tissue damage, and death. Indian aquaculture sector faced a loss of 62.5 million USD due to Argulus infection. However, current control methods, such as pesticides, cause serious environmental damage. Herbal treatment methods are ineffective and have limitations. Hence, a more efficient and cost-effective control method is needed. In recent years, vaccine development has emerged as a promising avenue of research. Understanding the effect of the host-parasite relationship in the host immune system is essential to develop strategies for prevention, control, and management of argulosis. These interactions provide insights into the co-evolutionary dynamics between hosts and parasites. This review provides an overview of the current knowledge on the host-searching behaviour of Argulus, host-parasite interaction and control strategies. This review also highlights the need for further research and the development of sustainable control measures for Argulus infection.
Collapse
Affiliation(s)
- B Haridevamuthu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu District, Tamil Nadu, India
| | - David Raj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu District, Tamil Nadu, India
| | - Aziz Arshad
- Department of Aquaculture, Faculty of Agriculture, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
3
|
Wang X, Guo J, Yu Q, Zhao L, Gao X, Wang L, Wen M, Yan J, An M, Liu Y. Decellularized Matrices for the Treatment of Tissue Defects: from Matrix Origin to Immunological Mechanisms. Biomol Ther (Seoul) 2024; 32:509-522. [PMID: 39091238 PMCID: PMC11392660 DOI: 10.4062/biomolther.2024.050] [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: 03/23/2024] [Revised: 05/07/2024] [Accepted: 05/31/2024] [Indexed: 08/04/2024] Open
Abstract
Decellularized matrix transplantation has emerged as a promising therapeutic approach for repairing tissue defects, with numerous studies assessing its safety and efficacy in both animal models and clinical settings. The host immune response elicited by decellularized matrix grafts of natural biological origin plays a crucial role in determining the success of tissue repair, influenced by matrix heterogeneity and the inflammatory microenvironment of the wound. However, the specific immunologic mechanisms underlying the interaction between decellularized matrix grafts and the host immune system remain elusive. This article reviews the sources of decellularized matrices, available decellularization techniques, and residual immunogenic components. It focuses on the host immune response following decellularized matrix transplantation, with emphasis on the key mechanisms of Toll-like receptor, T-cell receptor, and TGF-β/SMAD signaling in the stages of post-transplantation immunorecognition, immunomodulation, and tissue repair, respectively. Furthermore, it highlights the innovative roles of TLR10 and miR-29a-3p in improving transplantation outcomes. An in-depth understanding of the molecular mechanisms underlying the host immune response after decellularized matrix transplantation provides new directions for the repair of tissue defects.
Collapse
Affiliation(s)
- Xinyue Wang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China
| | - Jiqiang Guo
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing Yu
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China
| | - Luyao Zhao
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China
| | - Xiang Gao
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China
| | - Li Wang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China
| | - Meiling Wen
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China
| | - Junrong Yan
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Meiwen An
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China
| | - Yang Liu
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
4
|
Hamad BS, Shnawa BH, Alrawi RA, Ahmed MH. Comparative analysis of host immune responses to Hydatid cyst in human and ovine hepatic cystic Echinococcosis. Vet Immunol Immunopathol 2024; 273:110775. [PMID: 38776648 DOI: 10.1016/j.vetimm.2024.110775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Hydatid disease is caused by the larval stages of the canine tapeworm Echinococcus granulosus. It is one of the most critical helminthic diseases, representing worldwide public health and socio-economic concern. AIM This study aimed to investigate the expression of apoptosis and immune response within hepatic tissues of humans and sheep infected with the Hydatid cyst. METHODS Paraffin-embedded tissue was prepared from each tissue sample and used for histopathological examination by Haematoxylin- Eosin. Also, toluidine blue staining was used for mast cell detection, while an immunohistochemical study was performed to assess CD3 T lymphocytes, CD4 helper T lymphocytes, CD8 cytotoxic T lymphocytes, CD20 memory B lymphocytes, CD68 macrophage, and caspase-3 antibodies. RESULTS The histological examination revealed significant changes, including the infiltration of inflammatory cells, predominantly lymphocytes with scattered giant cells, necrotic hepatic tissue, and fibrosis. Toluidine blue stain revealed a higher number of mast cells (5 cells/field) in humans compared to sheep (3.6 cells/field). The immunohistochemical analysis confirmed that the CD3 were the most predominant inflammatory cell in the hepatic tissue of humans (intensive 70%), and sheep (moderate 38.47%). Caspase-3 was observed in all samples in different grades and mostly in human liver tissue. CONCLUSION This data could aid in recognizing immunological markers for differentiating disease progression, as well as enhance the understanding of local immune responses to cystic Echinococcosis (CE). The findings could provide preliminary data for future studies on immune responses associated with Hydatid cysts.
Collapse
Affiliation(s)
- Bnar S Hamad
- Biology Department, Faculty of Science, Soran University, Soran, Kurdistan Region 30802, Iraq
| | - Bushra H Shnawa
- Biology Department, Faculty of Science, Soran University, Soran, Kurdistan Region 30802, Iraq
| | - Rafal A Alrawi
- Clinical Analysis Department, College of Pharmacy, Hawler Medical University, Kurdistan Region, Iraq
| | - Mukhtar H Ahmed
- SISAF Drug Delivery Nanotechnology, Ulster University, Belfast BT37 0QB, UK.
| |
Collapse
|
5
|
Jia Z, Yu W, Li J, Zhang M, Zhan B, Yan L, Ming Z, Cheng Y, Tian X, Shao S, Huang J, Zhu X. Crystal structure of Trichinella spiralis calreticulin and the structural basis of its complement evasion mechanism involving C1q. Front Immunol 2024; 15:1404752. [PMID: 38690267 PMCID: PMC11059001 DOI: 10.3389/fimmu.2024.1404752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
Helminths produce calreticulin (CRT) to immunomodulate the host immune system as a survival strategy. However, the structure of helminth-derived CRT and the structural basis of the immune evasion process remains unclarified. Previous study found that the tissue-dwelling helminth Trichinella spiralis produces calreticulin (TsCRT), which binds C1q to inhibit activation of the complement classical pathway. Here, we used x-ray crystallography to resolve the structure of truncated TsCRT (TsCRTΔ), the first structure of helminth-derived CRT. TsCRTΔ was observed to share the same binding region on C1q with IgG based on the structure and molecular docking, which explains the inhibitory effect of TsCRT on C1q-IgG-initiated classical complement activation. Based on the key residues in TsCRTΔ involved in the binding activity to C1q, a 24 amino acid peptide called PTsCRT was constructed that displayed strong C1q-binding activity and inhibited C1q-IgG-initiated classical complement activation. This study is the first to elucidate the structural basis of the role of TsCRT in immune evasion, providing an approach to develop helminth-derived bifunctional peptides as vaccine target to prevent parasite infections or as a therapeutic agent to treat complement-related autoimmune diseases.
Collapse
Affiliation(s)
- Zhihui Jia
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wen Yu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingmo Li
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Mingming Zhang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bin Zhan
- Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Liming Yan
- Ministry of Education Key Laboratory of Protein Science, School of Medicine, Tsinghua University, Beijing, China
| | - Zhenhua Ming
- College of Life Science and Technology, Guangxi University, Nanning, China
| | - Yuli Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaolin Tian
- Ministry of Education Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Shuai Shao
- Beijing institute of Clinical Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Menon SS, Ramirez-Toloza G, Wycoff KL, Ehinger S, Shaughnessy J, Ram S, Ferreira VP. Mechanisms by which Factor H protects Trypanosoma cruzi from the alternative pathway of complement. Front Immunol 2024; 15:1152000. [PMID: 38361922 PMCID: PMC10867245 DOI: 10.3389/fimmu.2024.1152000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Chagas disease, a chronic disabling disease caused by the protozoan Trypanosoma cruzi, has no standardized treatment or preventative vaccine. The infective trypomastigote form of T. cruzi is highly resistant to killing by the complement immune system. Factor H (FH), a negative regulator of the alternative pathway (AP) of complement on cell surfaces and in blood, contains 20 short consensus repeat domains. The four N-terminal domains of FH inactivate the AP, while the other domains interact with C3b/d and glycan markers on cell surfaces. Various pathogens bind FH to inactivate the AP. T. cruzi uses its trans-sialidase enzyme to transfer host sialic acids to its own surface, which could be one of the approaches it uses to bind FH. Previous studies have shown that FH binds to complement-opsonized T. cruzi and parasite desialylation increases complement-mediated lysis of trypomastigotes. However, the molecular basis of FH binding to T. cruzi remain unknown. Only trypomastigotes, but not epimastigotes (non-infective, complement susceptible) bound FH directly, independent of C3 deposition, in a dose-dependent manner. Domain mapping experiments using 3-5 FH domain fragments showed that domains 5-8 competitively inhibited FH binding to the trypomastigotes by ~35% but did not decrease survival in complement. FH-Fc or mutant FH-Fc fusion proteins (3-11 contiguous FH domains fused to the IgG Fc) also did not kill trypomastigotes. FH-related protein-5, whose domains bear significant sequence identity to all known polyanion-binding FH domains (6-7, 10-14, 19-20), fully inhibited FH binding to trypomastigotes and reduced trypomastigote survival to < 24% in the presence of serum. In conclusion, we have elucidated the role of FH in complement resistance of trypomastigotes.
Collapse
Affiliation(s)
- Smrithi S. Menon
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Galia Ramirez-Toloza
- Laboratory of Parasitology, Department of Animal Preventive Medicine, Faculty of Veterinary Medicine and Livestock Sciences, University of Chile, Santiago, Chile
| | | | - Sean Ehinger
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Jutamas Shaughnessy
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Sanjay Ram
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Viviana P. Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| |
Collapse
|
7
|
Lin R, Wang X, Ni C, Fu C, Yang C, Dong D, Wu X, Chen X, Wang L, Hou J. Echinococcus granulosus cyst fluid inhibits KDM6B-mediated demethylation of trimethylated histone H3 lysine 27 and interleukin-1β production in macrophages. Parasit Vectors 2023; 16:422. [PMID: 37974225 PMCID: PMC10652454 DOI: 10.1186/s13071-023-06041-3] [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: 04/17/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Echinococcus granulosus can manipulate its host's immune response to ensure its own survival. However, the effect of histone modifications on the regulation of the NOD-like receptor protein 3 (NLRP3) inflammasome and downstream interleukin-1β (IL-1β) production in response to the parasite is not fully understood. METHODS We evaluated IL-1β secretion through enzyme-linked immunosorbent assay and assessed reactive oxygen species levels using the dichlorodihydrofluorescein diacetate probe. Western blotting and quantitative real-time polymerase chain reaction were performed to examine the expression of NLRP3 and IL-1β in mouse peritoneal macrophages and Tohoku Hospital Pediatrics-1 cells, a human macrophage cell line. The presence of trimethylated histone H3 lysine 27 (H3K27me3) modification on NLRP3 and IL-1β promoters was studied by chromatin immunoprecipitation. RESULTS Treatment with E. granulosus cyst fluid (EgCF) considerably reduced IL-1β secretion in mouse and human macrophages, although reactive oxygen species production increased. EgCF also suppressed the expression of NLRP3 and IL-1β. Mechanistically, EgCF prompted the enrichment of repressive H3K27me3 modification on the promoters of both NLRP3 and IL-1β in macrophages. Notably, the presence of EgCF led to a significant reduction in the expression of the H3K27me3 demethylase KDM6B. CONCLUSIONS Our study revealed that EgCF inhibits KDM6B expression and H3K27me3 demethylation, resulting in the transcriptional inhibition of NLRP3 and IL-1β. These results provide new insights into the immune evasion mechanisms of E. granulosus.
Collapse
Affiliation(s)
- Ruolin Lin
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiaopeng Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Caiya Ni
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Chunxue Fu
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Chun Yang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | | | - Xiangwei Wu
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xueling Chen
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Lianghai Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| | - Jun Hou
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| |
Collapse
|
8
|
Mohammed MMD, Heikal EA, Ellessy FM, Aboushousha T, Ghareeb MA. Comprehensive chemical profiling of Bassia indica Wight. aerial parts extract using UPLC-ESI-MS/MS, and its antiparasitic activity in Trichinella spiralis infected mice: in silico supported in vivo study. BMC Complement Med Ther 2023; 23:161. [PMID: 37202749 DOI: 10.1186/s12906-023-03988-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 05/05/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Trichinellosis is a public health threat infected both animals and humans as a result of eating undercooked meat. It caused by Trichinella spiralis that has widespread drug resistance and even developed many sophisticated strategies for their survival, this increases the demand in searching for new anthelmintic drugs from natural source. METHODS Our objectives were to test the in vitro and in vivo anthelmintic activity of Bassia indica BuOH frac., and to characterize its chemical composition using UPLC-ESI-MS/MS. Besides an in silico molecular docking study with the prediction of the PreADMET properties. RESULTS In vitro investigation of B. indica BuOH frac., showed severe destruction of the adult worm and larvae, marked cuticle swelling, areas with vesicles, blebs and loss of annulations. This was assured via in vivo study, which revealed a significant reduction (P < 0.05) in the mean adult worm count with efficacy of 47.8% along with a significant decrease (P < 0.001) in the mean larval count per gram muscle with efficacy 80.7%. Histopathological examinations of the small intestine and muscular sections showed marked improvement. In addition, immunohistochemical findings demonstrated that B. indica BuOH frac. depressed the proinflammatory cytokines expressions of TNF-α, which was obviously upregulated by T. spiralis. Precise chemical investigation of the BuOH frac. using UPLC-ESI-MS/MS resulted in the identification of 13 oleanolic type triterpenoid saponins; oleanolic acid 3-O-6´-O-methyl-β-D-glucurono-pyranoside (1), chikusetsusaponin-IVa (2) and its methyl ester (3), chikusetsusaponin IV (4) and its methyl ester (5), momordin-Ic (6) and its methyl ester (7), betavulgaroside-I (8), -II (9) -IV (10), -X (11), licorice-saponin-C2 (12) and -J2 (13). In addition, 6 more phenolics were identified as syringaresinol (14), 3,4-di-O-caffeoylquinic acid (15), 3-O-caffeoyl-4-O-dihydrocaffeoylquinic acid (16), 3,4-di-O-caffeoylquinic acid butyl ester (17), 3,5-di-O-galloyl-4-O-digalloylquinic acid (18) and quercetin 3-O-(6´´-feruloyl)-sophoroside (19). The auspicious anthelmintic activity was further ascertained using in silico molecular docking approach that targeted certain protein receptors (β-tubulin monomer, tumor necrosis factor alpha (TNF-α), cysteine protease (Ts-CF1), calreticulin protein (Ts-CRT)), all the docked compounds (1-19) fit into the binding site of the active pocket with binding affinities noteworthy than albendazole. In addition, ADMET properties, drug score and drug likeness were predicted for all compounds.
Collapse
Affiliation(s)
- Magdy M D Mohammed
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki-12622, Cairo, Egypt.
| | - Elham A Heikal
- Department of Medical Parasitology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Fatma M Ellessy
- Department of Medical Parasitology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Tarek Aboushousha
- Department of Pathology, Theodor Bilharz Research Institute, Kornaish El-Nile, Warrak El-Hadar, Imbaba, (P.O. 30), Giza, 12411, Egypt
| | - Mosad A Ghareeb
- Medicinal Chemistry Department, Theodor Bilharz Research Institute, Kornaish El-Nile, Warrak El-Hadar, Imbaba, (P.O. 30), Giza, 12411, Egypt
| |
Collapse
|
9
|
Pereira-Filho AA, Queiroz DC, Saab NAA, D'Ávila Pessoa GC, Koerich LB, Pereira MH, Sant'Anna MRV, Araújo RN, Bartholomeu DC, Gontijo NF. Evasion of the complement system by Leishmania through the uptake of C4bBP, a complement regulatory protein, and probably by the action of GP63 on C4b molecules deposited on parasite surface. Acta Trop 2023; 242:106908. [PMID: 36963597 DOI: 10.1016/j.actatropica.2023.106908] [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: 01/19/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
The complement system is a primary component of the vertebrate innate immune system, and its activity is harmful to microorganisms and parasites. To evade complement attack, some pathogens, such as viruses, bacteria, and protozoa, can interact with complement regulatory proteins from their hosts. Our research group has described the ability of Leishmania species to bind Factor H from human serum and use it as a tool to evade the complement system. However, there is no description of the interaction of Leishmania with other complement regulatory proteins, such as the C4b-binding protein (C4bBP), a negative regulator of classical and lectins complement system pathways. The results presented in this manuscript suggest that Leishmania infantum, L. amazonensis, and L. braziliensis recruit C4bBP from human serum. The uptake of C4bBP by L. infantum was studied in detail to improve our understanding of this inhibitory mechanism. When exposed to this complement regulator, parasites with inactivated GP63 bind to C4bBP and inactivate C4b deposited on their surface after serum exposure. This inactivation occurs by the action of Factor I, a complement system protease. In addition to the C4bBP-Factor I inactivation mechanism, the surface parasite protease GP63 can also inactivate soluble C4b molecules and probably that C4b molecules deposited on the parasites surface. This manuscript shows that Leishmania has two independent strategies to inactivate C4b molecules, preventing the progress of classical and lectin pathways. The identification of theC4bBP receptor on the Leishmania membrane may provide a new vaccine target to fight leishmaniasis.
Collapse
Affiliation(s)
- Adalberto Alves Pereira-Filho
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| | - Daniel Costa Queiroz
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| | - Natália Alvim Araújo Saab
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| | - Grasielle Caldas D'Ávila Pessoa
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| | - Leonardo Barbosa Koerich
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| | - Marcos Horácio Pereira
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| | - Mauricio Roberto Viana Sant'Anna
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| | - Ricardo Nascimento Araújo
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| | - Daniella Castanheira Bartholomeu
- Laboratório de Genômica de Parasitos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| | - Nelder Figueiredo Gontijo
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa postal 486, 31270-901, Belo Horizonte, MG, Brazil.
| |
Collapse
|
10
|
Raza A, Schulz BL, Nouwens A, Naseem MN, Kamran M, Mantilla Valdivieso EF, Kerr ED, Constantinoiu C, Jonsson NN, James P, Tabor AE. Application of quantitative proteomics to discover biomarkers for tick resistance in cattle. Front Immunol 2023; 14:1091066. [PMID: 36793724 PMCID: PMC9924087 DOI: 10.3389/fimmu.2023.1091066] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/04/2023] [Indexed: 01/31/2023] Open
Abstract
Introduction Breeding for tick resistance is a sustainable alternative to control cattle ticks due to widespread resistance to acaricidal drugs and the lack of a protective vaccine. The most accurate method used to characterise the phenotype for tick resistance in field studies is the standard tick count, but this is labour-intensive and can be hazardous to the operator. Efficient genetic selection requires reliable phenotyping or biomarker(s) for accurately identifying tick-resistant cattle. Although breed-specific genes associated with tick resistance have been identified, the mechanisms behind tick resistance have not yet been fully characterised. Methods This study applied quantitative proteomics to examine the differential abundance of serum and skin proteins using samples from naïve tick-resistant and -susceptible Brangus cattle at two-time points following tick exposure. The proteins were digested into peptides, followed by identification and quantification using sequential window acquisition of all theoretical fragment ion mass spectrometry. Results Resistant naïve cattle had a suite of proteins associated with immune response, blood coagulation and wound healing that were significantly (adjusted P < 10- 5) more abundant compared with susceptible naïve cattle. These proteins included complement factors (C3, C4, C4a), alpha-1-acid glycoprotein (AGP), beta-2-glycoprotein-1, keratins (KRT1 & KRT3) and fibrinogens (alpha & beta). The mass spectrometry findings were validated by identifying differences in the relative abundance of selected serum proteins with ELISA. The proteins showing a significantly different abundance in resistant cattle following early and prolonged tick exposures (compared to resistant naïve) were associated with immune response, blood coagulation, homeostasis, and wound healing. In contrast, susceptible cattle developed some of these responses only after prolonged tick exposure. Discussion Resistant cattle were able to transmigrate immune-response related proteins towards the tick bite sites, which may prevent tick feeding. Significantly differentially abundant proteins identified in this research in resistant naïve cattle may provide a rapid and efficient protective response to tick infestation. Physical barrier (skin integrity and wound healing) mechanisms and systemic immune responses were key contributors to resistance. Immune response-related proteins such as C4, C4a, AGP and CGN1 (naïve samples), CD14, GC and AGP (post-infestation) should be further investigated as potential biomarkers for tick resistance.
Collapse
Affiliation(s)
- Ali Raza
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Benjamin L Schulz
- The University of Queensland, School of Chemistry and Molecular Biosciences, St. Lucia, QLD, Australia
| | - Amanda Nouwens
- The University of Queensland, School of Chemistry and Molecular Biosciences, St. Lucia, QLD, Australia
| | - Muhammad Noman Naseem
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Muhammad Kamran
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Emily F Mantilla Valdivieso
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Edward D Kerr
- The University of Queensland, School of Chemistry and Molecular Biosciences, St. Lucia, QLD, Australia
| | - Constantin Constantinoiu
- College of Public Health, Medical & Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Nicholas N Jonsson
- Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Peter James
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia
| | - Ala E Tabor
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, St Lucia, QLD, Australia.,The University of Queensland, School of Chemistry and Molecular Biosciences, St. Lucia, QLD, Australia
| |
Collapse
|
11
|
Khattab A, Rezola M, Barroso M, Kyrklund M, Pihlajamaa T, Freitag TL, van Gemert GJ, Bousema T, Permi P, Turunen O, Sauerwein R, Luty AJF, Meri S. Hijacking the human complement inhibitor C4b-binding protein by the sporozoite stage of the Plasmodium falciparum parasite. Front Immunol 2022; 13:1051161. [PMID: 36479121 PMCID: PMC9720182 DOI: 10.3389/fimmu.2022.1051161] [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: 09/22/2022] [Accepted: 11/02/2022] [Indexed: 11/22/2022] Open
Abstract
The complement system is considered the first line of defense against pathogens. Hijacking complement regulators from blood is a common evasion tactic of pathogens to inhibit complement activation on their surfaces. Here, we report hijacking of the complement C4b-binding protein (C4bp), the regulator of the classical and lectin pathways of complement activation, by the sporozoite (SPZ) stage of the Plasmodium falciparum parasite. This was shown by direct binding of radiolabeled purified C4bp to live SPZs as well as by binding of C4bp from human serum to SPZs in indirect immunofluorescence assays. Using a membrane-bound peptide array, peptides from the N-terminal domain (NTD) of P. falciparum circumsporozoite protein (CSP) were found to bind C4bp. Soluble biotinylated peptide covering the same region on the NTD and a recombinantly expressed NTD also bound C4bp in a dose-dependent manner. NTD-binding site on C4bp was mapped to the CCP1-2 of the C4bp α-chain, a common binding site for many pathogens. Native CSP was also co-immunoprecipitated with C4bp from human serum. Preventing C4bp binding to the SPZ surface negatively affected the SPZs gliding motility in the presence of functional complement and malaria hyperimmune IgG confirming the protective role of C4bp in controlling complement activation through the classical pathway on the SPZ surface. Incorporating the CSP-C4bp binding region into a CSP-based vaccine formulation could induce vaccine-mediated immunity that neutralizes this immune evasion region and increases the vaccine efficacy.
Collapse
Affiliation(s)
- Ayman Khattab
- Department of Bacteriology and Immunology, Haartman Institute, and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland,Department of Nucleic Acid Research, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg El-Arab, Alexandria, Egypt,*Correspondence: Ayman Khattab,
| | - Mikel Rezola
- Department of Bacteriology and Immunology, Haartman Institute, and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Marta Barroso
- Department of Bacteriology and Immunology, Haartman Institute, and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Mikael Kyrklund
- Department of Bacteriology and Immunology, Haartman Institute, and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland,Department of Bioproducts and Biosystems, Aalto University, Espoo, Finland
| | - Tero Pihlajamaa
- Department of Clinical Chemistry, HUSLAB, Helsinki University Hospital, HUS Diagnostic Center, Helsinki, Finland
| | - Tobias L. Freitag
- Department of Bacteriology and Immunology, Haartman Institute, and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | | | - Teun Bousema
- Department of Medical Microbiology, Radboudumc, Nijmegen, Netherlands
| | - Perttu Permi
- Department of Chemistry, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland,Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Ossi Turunen
- Department of Bioproducts and Biosystems, Aalto University, Espoo, Finland,School of Forest Sciences, University of Eastern Finland, Joensuu, Finland
| | | | | | - Seppo Meri
- Department of Bacteriology and Immunology, Haartman Institute, and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland,HUSLAB Diagnostic Center, Helsinki University Central Hospital, Helsinki, Finland,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
12
|
Sikorski PM, Grigg ME. A Flow Cytometry-Based Assay to Measure C3b Deposition on Protozoan Parasites Such as Toxoplasma gondii. Curr Protoc 2022; 2:e593. [PMID: 36373989 DOI: 10.1002/cpz1.593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Flow cytometry is a powerful tool that can be used to study protozoan parasite interactions with the complement system. We developed a flow cytometric assay to measure the deposition of complement activation product C3b and to assess resistance to complement-mediated lysis. This assay involves exposing cultured parasites to human serum (the source of human complement) and staining parasites with antibodies against complement proteins to detect and quantify complement components on the parasite surface by flow cytometry. The assay can be used to compare complement activation across a variety of different species of protozoan parasites. As a proof of concept, we describe protocols to study C3 deposition on the single-cell protist Toxoplasma gondii. This parasite actively regulates C3 deposition and proteolytic inactivation to eliminate the formation of lytic pores targeted to the parasite surface coat, which is the end-product of the complement cascade. The antibodies used in this assay recognize both active and inactive forms of C3 deposited on parasite surfaces. Hence, the assay facilitates the identification and characterization of parasite resistance factors that regulate complement deposition and catabolic inactivation. © Published 2022. This article is a U.S. Government work and is in the public domain in the USA. Basic Protocol 1: Culturing human foreskin fibroblasts and Toxoplasma gondii strains Basic Protocol 2: In vitro complement activation assay Support Protocol: Screening of normal human serum Basic Protocol 3: Flow cytometric analysis of C3b deposition.
Collapse
Affiliation(s)
- Patricia M Sikorski
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
- Department of Microbiology and Immunology, Georgetown University Medical Center, Georgetown University, Washington, DC
| | - Michael E Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
13
|
Inklaar MR, Barillas-Mury C, Jore MM. Deceiving and escaping complement - the evasive journey of the malaria parasite. Trends Parasitol 2022; 38:962-974. [PMID: 36089499 PMCID: PMC9588674 DOI: 10.1016/j.pt.2022.08.013] [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: 05/13/2022] [Revised: 08/03/2022] [Accepted: 08/19/2022] [Indexed: 01/13/2023]
Abstract
During its life cycle, Plasmodium, the malaria parasite, is exposed to the human and mosquito complement systems. Early experiments demonstrated that activation of complement can pose a serious threat to parasites, but recent studies revealed complement-evasion mechanisms important for parasite survival. Blood-stage parasites and gametes recruit regulators to neutralize human complement activation, while ookinetes inhibit mosquito complement by disrupting epithelial nitration in response to midgut invasion. Here we provide an in-depth overview of the evasion mechanisms currently known and speculate on the existence of others not yet identified. Finally, we discuss how these mechanisms could provide novel targets for urgently needed malaria vaccines and therapeutics.
Collapse
Affiliation(s)
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboudumc, The Netherlands.
| |
Collapse
|
14
|
Rashidi S, Mansouri R, Ali-Hassanzadeh M, Muro A, Nguewa P, Manzano-Román R. The Defensive Interactions of Prominent Infectious Protozoan Parasites: The Host's Complement System. Biomolecules 2022; 12:1564. [PMID: 36358913 PMCID: PMC9687244 DOI: 10.3390/biom12111564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/16/2022] [Accepted: 10/21/2022] [Indexed: 12/30/2023] Open
Abstract
The complement system exerts crucial functions both in innate immune responses and adaptive humoral immunity. This pivotal system plays a major role dealing with pathogen invasions including protozoan parasites. Different pathogens including parasites have developed sophisticated strategies to defend themselves against complement killing. Some of these strategies include the employment, mimicking or inhibition of host's complement regulatory proteins, leading to complement evasion. Therefore, parasites are proven to use the manipulation of the complement system to assist them during infection and persistence. Herein, we attempt to study the interaction´s mechanisms of some prominent infectious protozoan parasites including Plasmodium, Toxoplasma, Trypanosoma, and Leishmania dealing with the complement system. Moreover, several crucial proteins that are expressed, recruited or hijacked by parasites and are involved in the modulation of the host´s complement system are selected and their role for efficient complement killing or lysis evasion is discussed. In addition, parasite's complement regulatory proteins appear as plausible therapeutic and vaccine targets in protozoan parasitic infections. Accordingly, we also suggest some perspectives and insights useful in guiding future investigations.
Collapse
Affiliation(s)
- Sajad Rashidi
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein 38811, Iran
- Department of Medical Laboratory Sciences, Khomein University of Medical Sciences, Khomein 38811, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd 8915173143, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft 7861615765, Iran
| | - Antonio Muro
- Infectious and Tropical Diseases Group (e-INTRO), Institute of Biomedical Research of Salamanca-Research Center for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37008 Salamanca, Spain
| | - Paul Nguewa
- Department of Microbiology and Parasitology, ISTUN Institute of Tropical Health, IdiSNA (Navarra Institute for Health Research), University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain
| | - Raúl Manzano-Román
- Infectious and Tropical Diseases Group (e-INTRO), Institute of Biomedical Research of Salamanca-Research Center for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37008 Salamanca, Spain
| |
Collapse
|
15
|
Arora G, Lynn GE, Tang X, Rosen CE, Hoornstra D, Sajid A, Hovius JW, Palm NW, Ring AM, Fikrig E. CD55 Facilitates Immune Evasion by Borrelia crocidurae, an Agent of Relapsing Fever. mBio 2022; 13:e0116122. [PMID: 36036625 PMCID: PMC9600505 DOI: 10.1128/mbio.01161-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/19/2022] [Indexed: 11/20/2022] Open
Abstract
Relapsing fever, caused by diverse Borrelia spirochetes, is prevalent in many parts of the world and causes significant morbidity and mortality. To investigate the pathoetiology of relapsing fever, we performed a high-throughput screen of Borrelia-binding host factors using a library of human extracellular and secretory proteins and identified CD55 as a novel host binding partner of Borrelia crocidurae and Borrelia persica, two agents of relapsing fever in Africa and Eurasia. CD55 is present on the surface of erythrocytes, carries the Cromer blood group antigens, and protects cells from complement-mediated lysis. Using flow cytometry, we confirmed that both human and murine CD55 bound to B. crocidurae and B. persica. Given the expression of CD55 on erythrocytes, we investigated the role of CD55 in pathological B. crocidurae-induced erythrocyte aggregation (rosettes), which enables spirochete immune evasion. We showed that rosette formation was partially dependent on host cell CD55 expression. Pharmacologically, soluble recombinant CD55 inhibited erythrocyte rosette formation. Finally, CD55-deficient mice infected with B. crocidurae had a lower pathogen load and elevated proinflammatory cytokine and complement factor C5a levels. In summary, our results indicate that CD55 is a host factor that is manipulated by the causative agents of relapsing fever for immune evasion. IMPORTANCE Borrelia species are causative agents of Lyme disease and relapsing fever infections in humans. B. crocidurae causes one of the most prevalent relapsing fever infections in parts of West Africa. In the endemic regions, B. crocidurae is present in ~17% of the ticks and ~11% of the rodents that serve as reservoirs. In Senegal, ~7% of patients with acute febrile illness were found to be infected with B. crocidurae. There is little information on host-pathogen interactions and how B. crocidurae manipulates host immunity. In this study, we used a high-throughput screen to identify host proteins that interact with relapsing fever-causing Borrelia species. We identified CD55 as one of the host proteins that bind to B. crocidurae and B. persica, the two causes of relapsing fever in Africa and Eurasia. We show that the interaction of B. crocidurae with CD55, present on the surface of erythrocytes, is key to immune evasion and successful infection in vivo. Our study further shows the role of CD55 in complement regulation, regulation of inflammatory cytokine levels, and innate immunity during relapsing fever infection. Overall, this study sheds light on host-pathogen interactions during relapsing fever infection in vivo.
Collapse
Affiliation(s)
- Gunjan Arora
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Geoffrey E. Lynn
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xiaotian Tang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Connor E. Rosen
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dieuwertje Hoornstra
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam Infection and Immunity, Amsterdam, Netherlands
| | - Andaleeb Sajid
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joppe W. Hovius
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam Infection and Immunity, Amsterdam, Netherlands
| | - Noah W. Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Aaron M. Ring
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
16
|
Uthailak N, Adisakwattana P, Thiangtrongjit T, Limpanont Y, Chusongsang P, Chusongsang Y, Tanasarnprasert K, Reamtong O. Discovery of Schistosoma mekongi circulating proteins and antigens in infected mouse sera. PLoS One 2022; 17:e0275992. [PMID: 36227939 PMCID: PMC9562170 DOI: 10.1371/journal.pone.0275992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by an infection of the parasitic flatworms schistosomes. Schistosoma mekongi is a restricted Schistosoma species found near the Mekong River, mainly in southern Laos and northern Cambodia. Because there is no vaccine or effective early diagnosis available for S. mekongi, additional biomarkers are required. In this study, serum biomarkers associated with S. mekongi-infected mice were identified at 14-, 28-, 42-, and 56-days post-infection. Circulating proteins and antigens of S. mekongi in mouse sera were analyzed using mass spectrometry-based proteomics. Serine protease inhibitors and macrophage erythroblast attacher were down-regulated in mouse sera at all infection timepoints. In addition, 54 circulating proteins and 55 antigens of S. mekongi were identified. Notable circulating proteins included kyphoscoliosis peptidase and putative tuberin, and antigens were detected at all four infection timepoints, particularly in the early stages (12 days). The putative tuberin sequence of S. mekongi was highly similar to homologs found in other members of the genus Schistosoma and less similar to human and murine sequences. Our study provided the identity of promising diagnostic biomarkers that could be applicable in early schistosomiasis diagnosis and vaccine development.
Collapse
Affiliation(s)
- Naphatsamon Uthailak
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Poom Adisakwattana
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tipparat Thiangtrongjit
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yanin Limpanont
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phiraphol Chusongsang
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yupa Chusongsang
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kanthi Tanasarnprasert
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
17
|
Kang JM, Lê HG, Võ TC, Yoo WG, Sohn WM, Na BK. Mapping of the Complement C9 Binding Region on Clonorchis sinensis Paramyosin. THE KOREAN JOURNAL OF PARASITOLOGY 2022; 60:255-259. [PMID: 36041487 PMCID: PMC9441447 DOI: 10.3347/kjp.2022.60.4.255] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022]
Abstract
Heliminthic paramyosin is a multifunctional protein that not only acts as a structural protein in muscle layers but as an immune-modulatory molecule interacting with the host immune system. Previously, we found that paramyosin from Clonorchis sinensis (CsPmy) is bound to human complement C9 protein (C9). To analyze the C9 binding region on CsPmy, overlapping recombinant fragments of CsPmy were produced and their binding activity to human C9 was investigated. The fragmental expression of CsPmy and C9 binding assays revealed that the C9 binding region was located at the C-terminus of CsPmy. Further analysis of the C-terminus of CsPmy to narrow the C9 binding region on CsPmy indicated that the region flanking731Leu–780 Leu was a potent C9 binding region. The CsPmy fragments corresponding to the region effectively inhibited human C9 polymerization. These results provide a precise molecular basis for CsPmy as a potent immunomodulator to evade host immune defenses by inhibiting complement attack.
Collapse
Affiliation(s)
- Jung-Mi Kang
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
| | - Hương Giang Lê
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
| | - Tuấn Cường Võ
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
| | - Won Gi Yoo
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
| | - Woon-Mok Sohn
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
| | - Byoung-Kuk Na
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
- Corresponding author ()
| |
Collapse
|
18
|
de Castro Neto AL, da Silveira JF, Mortara RA. Role of Virulence Factors of Trypanosomatids in the Insect Vector and Putative Genetic Events Involved in Surface Protein Diversity. Front Cell Infect Microbiol 2022; 12:807172. [PMID: 35573777 PMCID: PMC9097677 DOI: 10.3389/fcimb.2022.807172] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Trypanosomatids are flagellate protozoans that can infect several invertebrate and vertebrate hosts, including insects and humans. The three most studied species are the human pathogens Trypanosoma brucei, Trypanosoma cruzi and Leishmania spp. which are the causative agents of Human African Trypanosomiasis (HAT), Chagas disease and different clinical forms of leishmaniasis, respectively. These parasites possess complex dixenous life cycles, with zoonotic and anthroponotic stages, and are transmitted by hematophagous insects. To colonize this myriad of hosts, they developed mechanisms, mediated by virulence factors, to infect, propagate and survive in different environments. In insects, surface proteins play roles in parasite attachment and survival in the insect gut, whilst in the mammalian host, the parasites have a whole group of proteins and mechanisms that aid them invading the host cells and evading its immune system components. Many studies have been done on the impact of these molecules in the vertebrate host, however it is also essential to notice the importance of these virulence factors in the insect vector during the parasite life cycle. When inside the insect, the parasites, like in humans, also need to survive defense mechanisms components that can inhibit parasite colonization or survival, e.g., midgut peritrophic membrane barrier, digestive enzymes, evasion of excretion alongside the digested blood meal, anatomic structures and physiological mechanisms of the anterior gut. This protection inside the insect is often implemented by the same group of virulence factors that perform roles of immune evasion in the mammalian host with just a few exceptions, in which a specific protein is expressed specifically for the insect vector form of the parasite. This review aims to discuss the roles of the virulence molecules in the insect vectors, showing the differences and similarities of modes of action of the same group of molecules in insect and humans, exclusive insect molecules and discuss possible genetic events that may have generated this protein diversity.
Collapse
|
19
|
De Marco Verissimo C, Jewhurst HL, Dobó J, Gál P, Dalton JP, Cwiklinski K. Fasciola hepatica is refractory to complement killing by preventing attachment of mannose binding lectin (MBL) and inhibiting MBL-associated serine proteases (MASPs) with serpins. PLoS Pathog 2022; 18:e1010226. [PMID: 35007288 PMCID: PMC8782513 DOI: 10.1371/journal.ppat.1010226] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 01/21/2022] [Accepted: 12/22/2021] [Indexed: 11/18/2022] Open
Abstract
The complement system is a first-line innate host immune defence against invading pathogens. It is activated via three pathways, termed Classical, Lectin and Alternative, which are mediated by antibodies, carbohydrate arrays or microbial liposaccharides, respectively. The three complement pathways converge in the formation of C3-convertase followed by the assembly of a lethal pore-like structure, the membrane attack complex (MAC), on the pathogen surface. We found that the infectious stage of the helminth parasite Fasciola hepatica, the newly excysted juvenile (NEJ), is resistant to the damaging effects of complement. Despite being coated with mannosylated proteins, the main initiator of the Lectin pathway, the mannose binding lectin (MBL), does not bind to the surface of live NEJ. In addition, we found that recombinantly expressed serine protease inhibitors secreted by NEJ (rFhSrp1 and rFhSrp2) selectively prevent activation of the complement via the Lectin pathway. Our experiments demonstrate that rFhSrp1 and rFhSrp2 inhibit native and recombinant MBL-associated serine proteases (MASPs), impairing the primary step that mediates C3b and C4b deposition on the NEJ surface. Indeed, immunofluorescence studies show that MBL, C3b, C4b or MAC are not deposited on the surface of NEJ incubated in normal human serum. Taken together, our findings uncover new means by which a helminth parasite prevents the activation of the Lectin complement pathway to become refractory to killing via this host response, in spite of presenting an assortment of glycans on their surface.
Collapse
Affiliation(s)
- Carolina De Marco Verissimo
- Centre for One Health and Ryan Institute, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Heather L. Jewhurst
- Centre for One Health and Ryan Institute, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - John P. Dalton
- Centre for One Health and Ryan Institute, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Krystyna Cwiklinski
- Centre for One Health and Ryan Institute, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
20
|
Schistosoma mansoni Adult Worm Protective and Diagnostic Proteins in n-Butanol Extracts Revealed by Proteomic Analysis. Pathogens 2021; 11:pathogens11010022. [PMID: 35055970 PMCID: PMC8777762 DOI: 10.3390/pathogens11010022] [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: 11/08/2021] [Revised: 12/09/2021] [Accepted: 12/21/2021] [Indexed: 12/02/2022] Open
Abstract
The S. mansoni adult worm n-butanol extract (Sm-AWBE) has been previously shown to contain specific S. mansoni antigens that have been used for immunodiagnosis of schistosomiasis in solid phase alkaline phosphatase immunoassay (APIA) and western blot (WB) analyses. Sm-AWBE was also used in immunoprotection studies against a fatal live-cercariae challenge in experimental mouse vaccination (~43% protection). The Sm-AWBE fraction was prepared by mixing adult worm membranous suspensions with aqueous-saturated n-butanol, centrifuging and recovering n-butanol-resistant proteins in the aqueous phase. Here we report a preliminary identification of Sm-AWBE protein components as revealed from a qualitative proteomic study after processing Sm-AWBE by 1D-gel electrophoresis, in-gel and in-solution tryptic digestions, and mass spectrometry analyses. We identified 33 proteins in Sm-AWBE, all previously known S. mansoni proteins and antigens; among them, immunomodulatory proteins and proteins mostly involved in host–parasite interactions. About 81.8% of the identified Sm-AWBE proteins are antigenic. STRING analysis showed a set of Sm-AWBE proteins configuring a small network of interactive proteins and a group of proteins without interactions. Functional groups of proteins included muscle contraction, antioxidant, GPI-anchored phosphoesterases, regulatory 14-3-3, various enzymes and stress proteins. The results widen the possibilities to design novel antigen combinations for better diagnostic and immunoprotective strategies for schistosomiasis control.
Collapse
|
21
|
Medina-Rincón GJ, Gallo-Bernal S, Jiménez PA, Cruz-Saavedra L, Ramírez JD, Rodríguez MJ, Medina-Mur R, Díaz-Nassif G, Valderrama-Achury MD, Medina HM. Molecular and Clinical Aspects of Chronic Manifestations in Chagas Disease: A State-of-the-Art Review. Pathogens 2021; 10:pathogens10111493. [PMID: 34832648 PMCID: PMC8619182 DOI: 10.3390/pathogens10111493] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic manifestations of Chagas disease present as disabling and life-threatening conditions affecting mainly the cardiovascular and gastrointestinal systems. Although meaningful research has outlined the different molecular mechanisms underlying Trypanosoma cruzi’s infection and the host-parasite interactions that follow, prompt diagnosis and treatment remain a challenge, particularly in developing countries and also in those where the disease is considered non-endemic. This review intends to present an up-to-date review of the parasite’s life cycle, genetic diversity, virulence factors, and infective mechanisms, as well as the epidemiology, clinical presentation, diagnosis, and treatment options of the main chronic complications of Chagas disease.
Collapse
Affiliation(s)
- Germán J. Medina-Rincón
- School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia; (S.G.-B.); (M.D.V.-A.); (H.M.M.)
- Correspondence: ; Tel.: +57-310-817-2369
| | - Sebastián Gallo-Bernal
- School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia; (S.G.-B.); (M.D.V.-A.); (H.M.M.)
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Paula A. Jiménez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia; (P.A.J.); (L.C.-S.); (J.D.R.)
| | - Lissa Cruz-Saavedra
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia; (P.A.J.); (L.C.-S.); (J.D.R.)
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia; (P.A.J.); (L.C.-S.); (J.D.R.)
| | - María Juliana Rodríguez
- Division of Cardiology, Fundación Cardioinfantil-Instituto de Cardiología, Bogotá 110131, Colombia; (M.J.R.); (R.M.-M.)
| | - Ramón Medina-Mur
- Division of Cardiology, Fundación Cardioinfantil-Instituto de Cardiología, Bogotá 110131, Colombia; (M.J.R.); (R.M.-M.)
| | - Gustavo Díaz-Nassif
- Division of Gastroenterology and Liver Diseases, Fundación Cardioinfantil-Instituto de Cardiología, Bogotá 111221, Colombia;
| | | | - Héctor M. Medina
- School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia; (S.G.-B.); (M.D.V.-A.); (H.M.M.)
- Division of Cardiology, Fundación Cardioinfantil-Instituto de Cardiología, Bogotá 110131, Colombia; (M.J.R.); (R.M.-M.)
| |
Collapse
|
22
|
Nakada-Tsukui K, Nozaki T. Trogocytosis in Unicellular Eukaryotes. Cells 2021; 10:cells10112975. [PMID: 34831198 PMCID: PMC8616307 DOI: 10.3390/cells10112975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/25/2021] [Accepted: 10/17/2021] [Indexed: 12/12/2022] Open
Abstract
Trogocytosis is a mode of internalization of a part of a live cell by nibbling and is mechanistically distinct from phagocytosis, which implies internalization of a whole cell or a particle. Trogocytosis has been demonstrated in a broad range of cell types in multicellular organisms and is also known to be involved in a plethora of functions. In immune cells, trogocytosis is involved in the "cross-dressing" between antigen presenting cells and T cells, and is thus considered to mediate intercellular communication. On the other hand, trogocytosis has also been reported in a variety of unicellular organisms including the protistan (protozoan) parasite Entamoeba histolytica. E. histolytica ingests human T cell line by trogocytosis and acquires complement resistance and cross-dresses major histocompatibility complex (MHC) class I on the cell surface. Furthermore, trogocytosis and trogocytosis-like phenomena (nibbling of a live cell, not previously described as trogocytosis) have also been reported in other parasitic protists such as Trichomonas, Plasmodium, Toxoplasma, and free-living amoebae. Thus, trogocytosis is conserved in diverse eukaryotic supergroups as a means of intercellular communication. It is depicting the universality of trogocytosis among eukaryotes. In this review, we summarize our current understanding of trogocytosis in unicellular organisms, including the history of its discovery, taxonomical distribution, roles, and molecular mechanisms.
Collapse
Affiliation(s)
- Kumiko Nakada-Tsukui
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
- Correspondence: (K.N.-T.); (T.N.); Tel.: +81-3-5285-1111 (K.N.-T.); +81-3-5841-3526 (T.N.)
| | - Tomoyoshi Nozaki
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
- Correspondence: (K.N.-T.); (T.N.); Tel.: +81-3-5285-1111 (K.N.-T.); +81-3-5841-3526 (T.N.)
| |
Collapse
|
23
|
Chulanetra M, Chaicumpa W. Revisiting the Mechanisms of Immune Evasion Employed by Human Parasites. Front Cell Infect Microbiol 2021; 11:702125. [PMID: 34395313 PMCID: PMC8358743 DOI: 10.3389/fcimb.2021.702125] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
For the establishment of a successful infection, i.e., long-term parasitism and a complete life cycle, parasites use various diverse mechanisms and factors, which they may be inherently bestowed with, or may acquire from the natural vector biting the host at the infection prelude, or may take over from the infecting host, to outmaneuver, evade, overcome, and/or suppress the host immunity, both innately and adaptively. This narrative review summarizes the up-to-date strategies exploited by a number of representative human parasites (protozoa and helminths) to counteract the target host immune defense. The revisited information should be useful for designing diagnostics and therapeutics as well as vaccines against the respective parasitic infections.
Collapse
Affiliation(s)
- Monrat Chulanetra
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
24
|
Lin K, Zhou D, Li M, Meng J, He F, Yang X, Dong D, Wang X, Wu X, Chen X, Hou J. Echinococcus granulosus cyst fluid suppresses inflammatory responses by inhibiting TRAF6 signalling in macrophages. Parasitology 2021; 148:887-894. [PMID: 33775265 PMCID: PMC11010193 DOI: 10.1017/s0031182021000548] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 11/07/2022]
Abstract
Echinococcus granulosus sensu lato has complex defence mechanisms that protect it from the anti-parasitic immune response for long periods. Echinococcus granulosus cyst fluid (EgCF) is involved in the immune escape. Nevertheless, whether and how EgCF modulates the inflammatory response in macrophages remains poorly understood. Here, real-time polymerase chain reaction and enzyme-linked immunosorbent assay revealed that EgCF could markedly attenuate the lipopolysaccharide (LPS)-induced production of pro-inflammatory factors including tumour necrosis factor-α, interleukin (IL)-12 and IL-6 but increase the expression of IL-10 at mRNA and protein levels in mouse peritoneal macrophages and RAW 264.7 cells. Mechanically, western blotting and immunofluorescence assay showed that EgCF abolished the activation of nuclear factor (NF)-κB p65, p38 mitogen-activated protein kinase (MAPK) and ERK1/2 signalling pathways by LPS stimulation in mouse macrophages. EgCF's anti-inflammatory role was at least partly contributed by promoting proteasomal degradation of the critical adaptor TRAF6. Moreover, the EgCF-promoted anti-inflammatory response and TRAF6 proteasomal degradation were conserved in human THP-1 macrophages. These findings collectively reveal a novel mechanism by which EgCF suppresses inflammatory responses by inhibiting TRAF6 and the downstream activation of NF-κB and MAPK signalling in both human and mouse macrophages, providing new insights into the molecular mechanisms underlying the E. granulosus-induced immune evasion.
Collapse
Affiliation(s)
- Ke Lin
- Department of Immunology, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Di Zhou
- Medical Laboratory, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Min Li
- Nursing School of Shihezi University, Shihezi, Xinjiang, China
| | - Jin Meng
- Department of Immunology, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Feiming He
- Department of Immunology, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiaofeng Yang
- Department of Immunology, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Dan Dong
- Department of Immunology, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xian Wang
- Department of Immunology, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiangwei Wu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xueling Chen
- Department of Immunology, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Jun Hou
- Department of Immunology, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| |
Collapse
|
25
|
Ilgová J, Salát J, Kašný M. Molecular communication between the monogenea and fish immune system. FISH & SHELLFISH IMMUNOLOGY 2021; 112:179-190. [PMID: 32800986 DOI: 10.1016/j.fsi.2020.08.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/21/2020] [Accepted: 08/11/2020] [Indexed: 06/11/2023]
Abstract
Monogeneans parasitise mainly the outer structures of fish, such as the gills, fins, and skin, that is, tissues covered with a mucous layer. While attached by sclerotised structures to host's surface, monogeneans feed on its blood or epidermal cells and mucus. Besides being a rich source of nutrients, these tissues also contain humoral immune factors and immune cells, which are ready to launch defence mechanisms against the tegument or gastrointestinal tract of these invaders. The exploitation of hosts' resources by the Monogenea must, therefore, be accompanied by suppressive and immunomodulatory mechanisms which protect the parasites against attacks by host immune system. Elimination of hosts' cytotoxic molecules and evasion of host immune response is often mediated by proteins secreted by the parasites. The aim of this review is to summarise existing knowledge on fish immune responses against monogeneans. Results gleaned from experimental infections illustrate the various interactions between parasites and the innate and adaptive immune system of the fish. The involvement of monogenean molecules (mainly inhibitors of peptidases) in molecular communication with host immune system is discussed.
Collapse
Affiliation(s)
- Jana Ilgová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic.
| | - Jiří Salát
- Department of Virology, Veterinary Research Institute, Hudcova 296/70, Brno, 621 00, Czech Republic
| | - Martin Kašný
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, Brno, 611 37, Czech Republic
| |
Collapse
|
26
|
de Castro Neto AL, da Silveira JF, Mortara RA. Comparative Analysis of Virulence Mechanisms of Trypanosomatids Pathogenic to Humans. Front Cell Infect Microbiol 2021; 11:669079. [PMID: 33937106 PMCID: PMC8085324 DOI: 10.3389/fcimb.2021.669079] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/30/2021] [Indexed: 11/23/2022] Open
Abstract
Trypanosoma brucei, Leishmania spp., and T. cruzi are flagellate protozoans of the family Trypanosomatidae and the causative agents of human African trypanosomiasis, leishmaniasis, and Chagas disease, respectively. These diseases affect humans worldwide and exert a significant impact on public health. Over the course of evolution, the parasites associated with these pathologies have developed mechanisms to circumvent the immune response system throughout the infection cycle. In cases of human infection, this function is undertaken by a group of proteins and processes that allow the parasites to propagate and survive during host invasion. In T. brucei, antigenic variation is promoted by variant surface glycoproteins and other proteins involved in evasion from the humoral immune response, which helps the parasite sustain itself in the extracellular milieu during infection. Conversely, Leishmania spp. and T. cruzi possess a more complex infection cycle, with specific intracellular stages. In addition to mechanisms for evading humoral immunity, the pathogens have also developed mechanisms for facilitating their adhesion and incorporation into host cells. In this review, the different immune evasion strategies at cellular and molecular levels developed by these human-pathogenic trypanosomatids have been discussed, with a focus on the key molecules responsible for mediating the invasion and evasion mechanisms and the effects of these molecules on virulence.
Collapse
Affiliation(s)
- Artur Leonel de Castro Neto
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
27
|
Vorel J, Cwiklinski K, Roudnický P, Ilgová J, Jedličková L, Dalton JP, Mikeš L, Gelnar M, Kašný M. Eudiplozoon nipponicum (Monogenea, Diplozoidae) and its adaptation to haematophagy as revealed by transcriptome and secretome profiling. BMC Genomics 2021; 22:274. [PMID: 33858339 PMCID: PMC8050918 DOI: 10.1186/s12864-021-07589-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background Ectoparasites from the family Diplozoidae (Platyhelminthes, Monogenea) belong to obligate haematophagous helminths of cyprinid fish. Current knowledge of these worms is for the most part limited to their morphological, phylogenetic, and population features. Information concerning the biochemical and molecular nature of physiological processes involved in host–parasite interaction, such as evasion of the immune system and its regulation, digestion of macromolecules, suppression of blood coagulation and inflammation, and effect on host tissue and physiology, is lacking. In this study, we report for the first time a comprehensive transcriptomic/secretome description of expressed genes and proteins secreted by the adult stage of Eudiplozoon nipponicum (Goto, 1891) Khotenovsky, 1985, an obligate sanguivorous monogenean which parasitises the gills of the common carp (Cyprinus carpio). Results RNA-seq raw reads (324,941 Roche 454 and 149,697,864 Illumina) were generated, de novo assembled, and filtered into 37,062 protein-coding transcripts. For 19,644 (53.0%) of them, we determined their sequential homologues. In silico functional analysis of E. nipponicum RNA-seq data revealed numerous transcripts, pathways, and GO terms responsible for immunomodulation (inhibitors of proteolytic enzymes, CD59-like proteins, fatty acid binding proteins), feeding (proteolytic enzymes cathepsins B, D, L1, and L3), and development (fructose 1,6-bisphosphatase, ferritin, and annexin). LC-MS/MS spectrometry analysis identified 721 proteins secreted by E. nipponicum with predominantly immunomodulatory and anti-inflammatory functions (peptidyl-prolyl cis-trans isomerase, homolog to SmKK7, tetraspanin) and ability to digest host macromolecules (cathepsins B, D, L1). Conclusions In this study, we integrated two high-throughput sequencing techniques, mass spectrometry analysis, and comprehensive bioinformatics approach in order to arrive at the first comprehensive description of monogenean transcriptome and secretome. Exploration of E. nipponicum transcriptome-related nucleotide sequences and translated and secreted proteins offer a better understanding of molecular biology and biochemistry of these, often neglected, organisms. It enabled us to report the essential physiological pathways and protein molecules involved in their interactions with the fish hosts. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07589-z.
Collapse
Affiliation(s)
- Jiří Vorel
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic.
| | - Krystyna Cwiklinski
- Molecular Parasitology Laboratory, Centre for One Health, Ryan Institute, National University of Ireland Galway, Galway, Ireland
| | - Pavel Roudnický
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic.,Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Jana Ilgová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic
| | - Lucie Jedličková
- Department of Parasitology, Faculty of Science, Charles University, Viničná 7, 128 44, Prague, Czech Republic.,Department of Zoology and Fisheries, Centre of Infectious Animal Diseases, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00, Prague, Czech Republic
| | - John P Dalton
- Molecular Parasitology Laboratory, Centre for One Health, Ryan Institute, National University of Ireland Galway, Galway, Ireland
| | - Libor Mikeš
- Department of Parasitology, Faculty of Science, Charles University, Viničná 7, 128 44, Prague, Czech Republic
| | - Milan Gelnar
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic
| | - Martin Kašný
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37, Brno, Czech Republic
| |
Collapse
|
28
|
Moore SR, Menon SS, Cortes C, Ferreira VP. Hijacking Factor H for Complement Immune Evasion. Front Immunol 2021; 12:602277. [PMID: 33717083 PMCID: PMC7947212 DOI: 10.3389/fimmu.2021.602277] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
The complement system is an essential player in innate and adaptive immunity. It consists of three pathways (alternative, classical, and lectin) that initiate either spontaneously (alternative) or in response to danger (all pathways). Complement leads to numerous outcomes detrimental to invaders, including direct killing by formation of the pore-forming membrane attack complex, recruitment of immune cells to sites of invasion, facilitation of phagocytosis, and enhancement of cellular immune responses. Pathogens must overcome the complement system to survive in the host. A common strategy used by pathogens to evade complement is hijacking host complement regulators. Complement regulators prevent attack of host cells and include a collection of membrane-bound and fluid phase proteins. Factor H (FH), a fluid phase complement regulatory protein, controls the alternative pathway (AP) both in the fluid phase of the human body and on cell surfaces. In order to prevent complement activation and amplification on host cells and tissues, FH recognizes host cell-specific polyanionic markers in combination with complement C3 fragments. FH suppresses AP complement-mediated attack by accelerating decay of convertases and by helping to inactivate C3 fragments on host cells. Pathogens, most of which do not have polyanionic markers, are not recognized by FH. Numerous pathogens, including certain bacteria, viruses, protozoa, helminths, and fungi, can recruit FH to protect themselves against host-mediated complement attack, using either specific receptors and/or molecular mimicry to appear more like a host cell. This review will explore pathogen complement evasion mechanisms involving FH recruitment with an emphasis on: (a) characterizing the structural properties and expression patterns of pathogen FH binding proteins, as well as other strategies used by pathogens to capture FH; (b) classifying domains of FH important in pathogen interaction; and (c) discussing existing and potential treatment strategies that target FH interactions with pathogens. Overall, many pathogens use FH to avoid complement attack and appreciating the commonalities across these diverse microorganisms deepens the understanding of complement in microbiology.
Collapse
Affiliation(s)
- Sara R Moore
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Smrithi S Menon
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Claudio Cortes
- Department of Foundational Medical Sciences, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| |
Collapse
|
29
|
Sikorski PM, Commodaro AG, Grigg ME. A Protective and Pathogenic Role for Complement During Acute Toxoplasma gondii Infection. Front Cell Infect Microbiol 2021; 11:634610. [PMID: 33692968 PMCID: PMC7937796 DOI: 10.3389/fcimb.2021.634610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/06/2021] [Indexed: 11/13/2022] Open
Abstract
The infection competence of the protozoan pathogen Toxoplasma gondii is critically dependent on the parasite’s ability to inactivate the host complement system. Toxoplasma actively resists complement-mediated killing in non-immune serum by recruiting host-derived complement regulatory proteins C4BP and Factor H (FH) to the parasite surface to inactivate surface-bound C3 and limit formation of the C5b-9 membrane attack complex (MAC). While decreased complement activation on the parasite surface certainly protects Toxoplasma from immediate lysis, the biological effector functions of C3 split products C3b and C3a are maintained, which includes opsonization of the parasite for phagocytosis and potent immunomodulatory effects that promote pro-inflammatory responses and alters mucosal defenses during infection, respectively. In this review, we discuss how complement regulation by Toxoplasma controls parasite burden systemically but drives exacerbated immune responses locally in the gut of genetically susceptible C57BL/6J mice. In effect, Toxoplasma has evolved to strike a balance with the complement system, by inactivating complement to protect the parasite from immediate serum killing, it generates sufficient C3 catabolites that signal through their cognate receptors to stimulate protective immunity. This regulation ultimately controls tachyzoite proliferation and promotes host survival, parasite persistence, and transmissibility to new hosts.
Collapse
Affiliation(s)
- Patricia M Sikorski
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Department of Microbiology and Immunology, Georgetown University Medical Center, Georgetown University, Washington, DC, United States
| | - Alessandra G Commodaro
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Michael E Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
30
|
Tritten L, Gillis-Germitsch N, Kockmann T, Schnyder M. Quantitative proteomics analysis of Angiostrongylus vasorum-induced alterations in dog serum sheds light on the pathogenesis of canine angiostrongylosis. Sci Rep 2021; 11:283. [PMID: 33431914 PMCID: PMC7801463 DOI: 10.1038/s41598-020-79459-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 12/09/2020] [Indexed: 12/22/2022] Open
Abstract
Blood contains hundreds of proteins, reflecting ongoing cellular processes and immune reactions. Infections with the blood-dwelling cardiopulmonary nematode Angiostrongylus vasorum in dogs manifest with a broad spectrum of clinical signs including respiratory distress, bleeding diathesis and neurological signs, and are associated with a perturbed blood protein profile in dogs. However, current knowledge does not completely explain the observed pathologies induced by A. vasorum infections, including bleeding disorders. Using sera from experimentally infected dogs, dog serum proteome was analysed by quantitative mass spectrometry methods over several time points before and after inoculation. Following computational analysis, we identified 139 up- and downregulated proteins after infection (log2 ratio cut-off ≥ 1.0; q-value ≤ 0.05). Among upregulated proteins were chitinase 3-like 1 and pulmonary surfactant-associated protein B (log2 fold-changes ≥ 5). Pathway enrichment revealed the complement (especially the lectin pathway) and coagulation cascades as significantly affected upon analysis of downregulated proteins. Among them were mannan-binding lectin serine peptidases, ficolin, and coagulation factor XIII-B. These results bring new elements towards understanding the underlying pathomechanisms of bleeding diatheses observed in some A. vasorum-infected dogs.
Collapse
Affiliation(s)
- Lucienne Tritten
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| | - Nina Gillis-Germitsch
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Tobias Kockmann
- Functional Genomics Center Zurich, ETH/UZH Zurich, Zurich, Switzerland
| | - Manuela Schnyder
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
31
|
Chen Y, Shao S, Huang J, Gu Y, Cheng Y, Zhu X. Therapeutic Efficacy of a Trichinella Spiralis Paramyosin-Derived Peptide Modified With a Membrane-Targeting Signal in Mice With Antigen-Induced Arthritis. Front Microbiol 2020; 11:608380. [PMID: 33424810 PMCID: PMC7785802 DOI: 10.3389/fmicb.2020.608380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Helminth-derived molecules have the ability to modulate the host immune system. Our previous study identified a tetradecapeptide derived from Trichinella spiralis paramyosin (Ts-pmy) that could bind to human complement component C9 to inhibit its polymerization, making the peptide a candidate therapeutic agent for complement-related immune disorders. Here, the peptide underwent an N-terminal modification with a membrane-targeting signal (a unique myristoylated peptide) to improve its therapeutic efficacy. We found that the modified peptide had a binding affinity to human C9 that was similar to that of the original peptide, as confirmed by microscale thermophoresis assays. The binding of the modified peptide to human C9 resulted in the inhibition of C9-related complement activation, as reflected by the decreased Zn2+-induced C9 polymerization and the decreased C9-dependent lysis of rabbit erythrocytes. In addition, the original and modified peptides could both bind to recombinant mouse C9 and inhibit the C9-dependent lysis of rabbit erythrocytes in normal mouse serum (NMS), which meant that the peptides could cross the species barrier to inhibit complement activity in mice. Further in vitro and in vivo analyses confirmed that the peptide modification increased the retention time of the peptide. Furthermore, intraarticular injection of the modified peptide markedly ameliorated knee swelling and joint damage in mice with antigen-induced arthritis (AIA), as assessed histologically. These results suggested that the Ts-pmy-derived peptide modified with a membrane-targeting signal was a reasonable candidate therapeutic agent for membrane attack complex (MAC)-related diseases [such as rheumatoid arthritis (RA)] and the study presented a new modification method to improve the potential therapeutic effects of the peptide.
Collapse
Affiliation(s)
- Yi Chen
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuai Shao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuan Gu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuli Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Shao S, Hao C, Zhan B, Zhuang Q, Zhao L, Chen Y, Huang J, Zhu X. Trichinella spiralis Calreticulin S-Domain Binds to Human Complement C1q to Interfere With C1q-Mediated Immune Functions. Front Immunol 2020; 11:572326. [PMID: 33329535 PMCID: PMC7710684 DOI: 10.3389/fimmu.2020.572326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/20/2020] [Indexed: 01/21/2023] Open
Abstract
Helminths develop strategies to escape host immune responses that facilitate their survival in the hostile host immune environment. Trichinella spiralis, a tissue-dwelling nematode, has developed a sophisticated strategy to escape complement attack. Our previous study demonstrated that T. spiralis secretes calreticulin (TsCRT) to inhibit host classical complement activation through binding to C1q; however, the C1q binding site in TsCRT and the specific mechanism involved with complement-related immune evasion remains unknown. Using molecular docking modeling and fragment expression, we determined that TsCRT-S, a 153-aa domain of TsCRT, is responsible for C1q binding. Recombinant TsCRT-S protein expressed in Escherichia coli had the same capacity to bind and inhibit human C1q-induced complement and neutrophil activation, as full-length TsCRT. TsCRT-S inhibited neutrophil reactive oxygen species and elastase release by binding to C1q and reduced neutrophil killing of newborn T. spiralis larvae. Binding of TsCRT-S to C1q also inhibited formation of neutrophil extracellular traps (NETs), which are involved in autoimmune pathologies and have yet to be therapeutically targeted. These findings provide evidence that the TsCRT-S fragment, rather than the full-length TsCRT, is a potential target for vaccine or therapeutic development for trichinellosis, as well as for complement-related autoimmune disease therapies.
Collapse
Affiliation(s)
- Shuai Shao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chunyue Hao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bin Zhan
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Qinghui Zhuang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Limei Zhao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yi Chen
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Abdo AIK, Ngoh YY, Lew MH, Dass SA, Rahumatullah A, Noordin R, Tye GJ. Generation of human scFv-IgG1Fc antibodies for detection of lymphatic filarial recombinant antigens, BmR1 and BmSXP. Biotechnol Appl Biochem 2020; 69:70-76. [PMID: 33258152 DOI: 10.1002/bab.2082] [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: 06/15/2020] [Accepted: 11/22/2020] [Indexed: 11/08/2022]
Abstract
Lymphatic filariasis is a neglected parasitic disease that affects millions in tropical and subtropical countries and is caused by Wuchereria and Brugia species. Specific and sensitive detection methods are essential in mapping infected areas where rapid tests are needed to cover underdeveloped and remote regions, which facilitates eliminating the disease as a public health problem. A few commercialized rapid tests based on antigen or antibody detection are available, but the former only detects infection by Wuchereria species and cross-reacts with nonlymphatic filaria, whereas antibody detection might provide positive results of previous infection. Here, we report the production of three different recombinant immunoglobulin gamma (IgG)1 antibodies based on scFvs previously generated via human antibody phage display technology, that is, anti-BmR1 clone 4, anti-BmXSP clone 5B, and anti-BmXSP clone 2H2. The scFv sequences were cloned into a pCMV-IgG1 vector, then transfected into a HEK293F cell line. The generated antibodies were found to be able to bind to their respective targets even at relatively low concentration. Conjugation of Fc to scFv induces binder stability and provides multiple labeling sites for probes and signaling molecules that can be used in rapid tests.
Collapse
Affiliation(s)
- Ahmad Ismail Khaled Abdo
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Ying Yuan Ngoh
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Min Han Lew
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Sylvia Annabel Dass
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Anizah Rahumatullah
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Rahmah Noordin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Penang, Malaysia
| |
Collapse
|
34
|
The Electrophoretic Pattern of Serum Proteins in Sheep with Naturally Acquired Gastrointestinal Nematode Infections. ACTA VET-BEOGRAD 2020. [DOI: 10.2478/acve-2020-0024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Abstract
The objective of this study was to describe the serum protein pattern in sheep with naturally acquired gastrointestinal parasitosis and to compare the distribution of protein fractions with the results obtained in animals with negative fecal examination results. Fecal and blood samples were taken from twenty-nine sheep positive for nematode eggs and twenty-four animals with negative results of fecal examination. Between the evaluated groups of sheep significant differences were noted in the relative mean values for all protein fractions and for albumin/globulin ratio (p<0.01 and p<0.001). The concentrations of total proteins showed no significant differences between both groups of sheep. The absolute mean values of albumin, α2-globulins and A/G ratio were significantly lower, the mean concentrations of α1-, β-, γ1- and γ2-globulins significantly higher in the nematode positive group of sheep (p<0.01 and p<0.001). In the nematode positive group the protein electrophoretic pattern showed a double α2-zone in three sheep and the γ-globulin zones were characterized by a diffuse higher broad and wide peaks. The presented results indicate that the gastrointestinal parasitic infections in sheep alter the distribution of serum proteins, and suggest their usefulness in animals with unchanged serum protein concentrations. The study brings new findings and extends the knowledge about the metabolic responses and consequences of gastrointestinal parasitic infections in sheep, particularly with regard to alterations in protein metabolism.
Collapse
|
35
|
Sharma S, Bhatnagar R, Gaur D. Complement Evasion Strategies of Human Pathogenic Bacteria. Indian J Microbiol 2020; 60:283-296. [PMID: 32655196 PMCID: PMC7329968 DOI: 10.1007/s12088-020-00872-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
Human pathogens need to overcome an elaborate network of host defense mechanisms in order to establish their infection, colonization, proliferation and eventual dissemination. The interaction of pathogens with different effector molecules of the immune system results in their neutralization and elimination from the host. The complement system is one such integral component of innate immunity that is critically involved in the early recognition and elimination of the pathogen. Hence, under this immune pressure, all virulent pathogens capable of inducing active infections have evolved immune evasive strategies that primarily target the complement system, which plays an essential and central role for host defense. Recent reports on several bacterial pathogens have elucidated the molecular mechanisms underlying complement evasion, inhibition of opsonic phagocytosis and cell lysis. This review aims to comprehensively summarize the recent findings on the various strategies adopted by pathogenic bacteria to escape complement-mediated clearance.
Collapse
Affiliation(s)
- Shikhar Sharma
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067 India
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067 India
| |
Collapse
|
36
|
Braden LM, Monaghan SJ, Fast MD. Salmon immunological defence and interplay with the modulatory capabilities of its ectoparasite Lepeophtheirus salmonis. Parasite Immunol 2020; 42:e12731. [PMID: 32403169 DOI: 10.1111/pim.12731] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 03/13/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022]
Abstract
The salmon louse Lepeophtheirus salmonis (Lsal) is an ectoparasitic copepod that exerts immunomodulatory and physiological effects on its host Atlantic salmon. Over 30 years of research on louse biology, control, host responses and the host-parasite relationship has provided a plethora of information on the intricacies of host resistance and parasite adaptation. Atlantic salmon exhibit temporal and spatial impairment of the immune system and wound healing ability during infection. This immunosuppression may render Atlantic salmon less tolerant to stress and other confounders associated with current management strategies. Contrasting susceptibility of salmonid hosts exists, and early pro-inflammatory Th1 type responses are associated with resistance. Rapid cellular responses to larvae appear to tip the balance of the host-parasite relationship in favour of the host, preventing severe immune-physiological impacts of the more invasive adults. Immunological, transcriptomic, genomic and proteomic evidence suggests pathological impacts occur in susceptible hosts through modulation of host immunity and physiology via pharmacologically active molecules. Co-evolutionary and farming selection pressures may have incurred preference of Atlantic salmon as a host for Lsal reflected in their interactome. Here, we review host-parasite interactions at the primary attachment/feeding site, and the complex life stage-dependent molecular mechanisms employed to subvert host physiology and immune responses.
Collapse
Affiliation(s)
- Laura M Braden
- AquaBounty Canada, Bay Fortune, PEI, Canada.,Department of Pathology and Microbiology, Atlantic Veterinary College-UPEI, Charlottetown, PEI, Canada
| | - Sean J Monaghan
- Institute of Aquaculture, University of Stirling, Stirling, UK
| | - Mark D Fast
- Department of Pathology and Microbiology, Atlantic Veterinary College-UPEI, Charlottetown, PEI, Canada
| |
Collapse
|
37
|
Sharma S, Bhatnagar R, Gaur D. Bacillus anthracis Poly-γ-D-Glutamate Capsule Inhibits Opsonic Phagocytosis by Impeding Complement Activation. Front Immunol 2020; 11:462. [PMID: 32296419 PMCID: PMC7138205 DOI: 10.3389/fimmu.2020.00462] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/28/2020] [Indexed: 01/18/2023] Open
Abstract
Bacillus anthracis poly-γ-D-glutamic acid (PGA) capsule is an essential virulent factor that helps the bacterial pathogen to escape host immunity. Like other encapsulated bacterial species, the B. anthracis capsule may also inhibit complement-mediated clearance and ensure bacterial survival in the host. Previous reports suggest that B. anthracis spore proteins inhibit complement activation. However, the mechanism through which the B. anthracis capsule imparts a survival advantage to the active bacteria has not been demonstrated till date. Thus, to evaluate the role of the PGA capsule in evading host immunity, we have undertaken the present head-to-head comparative study of the phagocytosis and complement activation of non-encapsulated and encapsulated B. anthracis strains. The encapsulated virulent strain exhibited resistance toward complement-dependent and complement-independent bacterial phagocytosis by human macrophages. The non-encapsulated Sterne strain was highly susceptible to phagocytosis by THP-1 macrophages, after incubation with normal human serum (NHS), heat-inactivated serum, and serum-free media, thus indicating that the capsule inhibited both complement-dependent and complement-independent opsonic phagocytosis. An increased binding of C3b and its subsequent activation to C3c and C3dg, which functionally act as potent opsonins, were observed with the non-encapsulated Sterne strain compared with the encapsulated strain. Other known mediators of complement fixation, IgG, C-reactive protein (CRP), and serum amyloid P component (SAP), also bound more prominently with the non-encapsulated Sterne strain. Studies with complement pathway-specific, component-deficient serum demonstrated that the classical pathway was primarily involved in mediating C3b binding on the non-encapsulated bacteria. Both strains equally bound the complement regulatory proteins C4BP and factor H. Importantly, we demonstrated that the negative charge of the PGA capsule was responsible for the differential binding of the complement proteins between the non-encapsulated and encapsulated strains. At lower pH closer to the isoelectric point of PGA, the neutralization of the negative charge was associated with an increased binding of C3b and IgG with the encapsulated B. anthracis strain. Overall, our data have demonstrated that the B. anthracis capsule inhibits complement fixation and opsonization resulting in reduced phagocytosis by macrophages, thus allowing the bacterial pathogen to evade host immunity.
Collapse
Affiliation(s)
- Shikhar Sharma
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
38
|
Sikorski PM, Commodaro AG, Grigg ME. Toxoplasma gondii Recruits Factor H and C4b-Binding Protein to Mediate Resistance to Serum Killing and Promote Parasite Persistence in vivo. Front Immunol 2020; 10:3105. [PMID: 32010145 PMCID: PMC6979546 DOI: 10.3389/fimmu.2019.03105] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/19/2019] [Indexed: 01/14/2023] Open
Abstract
Regulating complement is an important step in the establishment of infection by microbial pathogens. Toxoplasma gondii actively resists complement-mediated killing in non-immune human serum (NHS) by inactivating C3b, however the precise molecular basis is unknown. Here, a flow cytometry-based C3b binding assay demonstrated that Type II strains had significantly higher levels of surface-bound C3b than Type I strains. However, both strains efficiently inactivated C3b and were equally resistant to serum killing, suggesting that resistance is not strain-dependent. Toxoplasma activated both the lectin (LP) and alternative (AP) pathways, and the deposition of C3b was both strain and lectin-dependent. A flow cytometry-based lectin binding assay identified strain-specific differences in the level and heterogeneity of surface glycans detected. Specifically, increased lectin-binding by Type II strains correlated with higher levels of the LP recognition receptor mannose binding lectin (MBL). Western blot analyses demonstrated that Toxoplasma recruits both classical pathway (CP) and LP regulator C4b-binding proteins (C4BP) and AP regulator Factor H (FH) to the parasite surface to inactivate bound C3b-iC3b and C3dg and limit formation of the C5b-9 attack complex. Blocking FH and C4BP contributed to increased C5b-9 formation in vitro. However, parasite susceptibility in vitro was only impacted when FH was blocked, indicating that down regulation of the alternative pathway by FH may be more critical for parasite resistance. Infection of C3 deficient mice led to uncontrolled parasite growth, acute mortality, and reduced antibody production, indicating that both the presence of C3, and the ability of the parasite to inactivate C3, was protective. Taken together, our results establish that Toxoplasma regulation of the complement system renders mice resistant to acute infection by limiting parasite proliferation in vivo, but susceptible to chronic infection, with all mice developing transmissible cysts to maintain its life cycle.
Collapse
Affiliation(s)
- Patricia M Sikorski
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Department of Microbiology and Immunology, Georgetown University Medical Centre, Georgetown University, Washington, DC, United States
| | - Alessandra G Commodaro
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Michael E Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|