1
|
Carrera Silva EA, Puyssegur J, Errasti AE. Coevolutionary interplay: Helminths-trained immunity and its impact on the rise of inflammatory diseases. eLife 2025; 14:e105393. [PMID: 40231720 PMCID: PMC12002795 DOI: 10.7554/elife.105393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
The gut biome, a complex ecosystem of micro- and macro-organisms, plays a crucial role in human health. A disruption in this evolutive balance, particularly during early life, can lead to immune dysregulation and inflammatory disorders. 'Biome repletion' has emerged as a potential therapeutic approach, introducing live microbes or helminth-derived products to restore immune balance. While helminth therapy has shown some promise, significant challenges remain in optimizing clinical trials. Factors such as patient genetics, disease status, helminth species, and the optimal timing and dosage of their products or metabolites must be carefully considered to train the immune system effectively. We aim to discuss how helminths and their products induce trained immunity as prospective to treat inflammatory and autoimmune diseases. The molecular repertoire of helminth excretory/secretory products (ESPs), which includes proteins, peptides, lipids, and RNA-carrying extracellular vesicles (EVs), underscores their potential to modulate innate immune cells and hematopoietic stem cell precursors. Mimicking natural delivery mechanisms like synthetic exosomes could revolutionize EV-based therapies and optimizing production and delivery of ESP will be crucial for their translation into clinical applications. By deciphering and harnessing helminth-derived products' diverse modes of action, we can unleash their full therapeutic potential and pave the way for innovative treatments.
Collapse
Affiliation(s)
- Eugenio Antonio Carrera Silva
- EACS and JP Institute of Experimental Medicine, National Scientific and Technical Research Council, National Academy of Medicine (IMEX-CONICET-ANM)Buenos AiresArgentina
| | - Juliana Puyssegur
- EACS and JP Institute of Experimental Medicine, National Scientific and Technical Research Council, National Academy of Medicine (IMEX-CONICET-ANM)Buenos AiresArgentina
| | - Andrea Emilse Errasti
- AEE Institute of Pharmacology, School of Medicine, University of Buenos AiresBuenos AiresArgentina
- National Scientific and Technical Research Council (CONICET)Buenos AiresArgentina
| |
Collapse
|
2
|
Mohammad Rahimi H, Mahdavi F, Eslami N, Nemati S, Mirjalali H. The Effects of Extracellular Vesicles Derived from Hydatid Cyst Fluid on the Expression of microRNAs Involved in Liver Fibrosis. Acta Parasitol 2025; 70:89. [PMID: 40220059 DOI: 10.1007/s11686-025-01024-z] [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: 02/01/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025]
Abstract
INTRODUCTION Hydatidosis is a zoonotic neglected disease caused by the larval stage of Echinococcus granulosus. Evidence suggests a communication between hydatid cyst (HC) and hosts via extracellular vesicles (EVs). However, a little is known about the communication between EVs derived from HC fluid (HCF) and host cells. The current study aimed to investigate the effect of HCF derived EVs on expression of fibrotic and anti-fibrotic miRNAs in THP-1 cell line. METHODS In the current study, EVs were isolated using ultracentrifugation from wild-infected sheep HCF and characterized by western blot, electron microscope, and size distribution analysis. The effects of EVs on the expression levels of microRNAs (mir-16, mir-29a, and mir-155) involved in liver fibrosis were investigated using quantitative real-time PCR (qRT-PCR), 3 and 24 h after incubation. RESULTS Western blot analyses confirmed the expression of CD63 marker, while Calnexin and CD81 were absent in EVs samples. The SEM and morphology revealed round shape vesicles. The DLS analysis showed average size distribution 130.6 nm diameter. The expression levels of mir-16 and mir-29a were significantly upregulated after 3 h for 8.66 and 3.420, respectively, while they were significantly downregulated after 24 h for 3.853 and 1.859, respectively. CONCLUSION The main mechanism of the communication between EVs derived from HCF and their host remains unclear. Our results suggest that HC may modulate the expression of miRNAs, involved in liver fibrosis via EVs.
Collapse
Affiliation(s)
- Hanieh Mohammad Rahimi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mahdavi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasim Eslami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Nemati
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Zhang Y, Yue Y, Cheng Y, Jiao H, Yan M. Antigen B from Echinococcus granulosus regulates macrophage phagocytosis by controlling TLR4 endocytosis in immune thrombocytopenia. Chem Biol Interact 2025; 406:111350. [PMID: 39674446 DOI: 10.1016/j.cbi.2024.111350] [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: 10/01/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/16/2024]
Abstract
Immune thrombocytopenia (ITP) is characterized by a reduction in platelet counts, stemming from an autoimmune-mediated process where platelets are excessively cleared by macrophages. This enhanced phagocytosis is a cardinal pathogenic mechanism in ITP. Antigen B (AgB), a principal component of the Echinococcus granulosus cyst fluid, plays a pivotal role in safeguarding the parasite from host immune defenses by modulating macrophage activation. In this study, we explored the potential of AgB to regulate macrophage activation in the context of ITP. Our observations indicated a diminished presence of M1 macrophages and a reduced phagocytic capacity in patients infected with E. granulosus sensu stricto. We isolated AgB from E. granulosus cyst fluid (EgCF) and discovered that it could suppress the polarization of M1 macrophages and weaken their phagocytic activity via Fcγ receptors, consequently alleviating thrombocytopenia in an ITP mouse model. At the molecular level, AgB was found to suppress the activation of nuclear factor kappa B (NF-κB) and interferon regulatory factor 3 (IRF3) by impeding their nuclear translocation, leading to a reduction in the generation of inflammatory cytokines. Furthermore, AgB was shown to inhibit Toll-like receptor 4 (TLR4) endocytosis and the recycling of CD14. In aggregate, our findings uncover a novel immunomodulatory mechanism of AgB, which suppresses macrophage phagocytosis by regulating TLR4 endocytosis and the subsequent activation of NF-κB and IRF3 signaling pathways. These insights shed new light on the molecular intricacies of E. granulosus-induced immune evasion and suggest that AgB may serve as a promising therapeutic agent for ITP.
Collapse
Affiliation(s)
- Yunfei Zhang
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Yingbin Yue
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Yongfeng Cheng
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Hongjie Jiao
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Mei Yan
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| |
Collapse
|
4
|
Xiao J, Wu JW, Xin YZ, Song D, Gao XP, Yin M, Zhao W, Liu FL, Wang H, Wang J, Zhao JQ. A pilot study of microRNAs expression profile in plasma of patients with hydatid disease: potential immunomodulation of hydatid disease. Parasitol Res 2024; 123:336. [PMID: 39347812 DOI: 10.1007/s00436-024-08350-7] [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: 05/19/2024] [Accepted: 09/08/2024] [Indexed: 10/01/2024]
Abstract
Echinococcosis is a zoonotic disease, which seriously endangers human health. The immune game between parasite and host is not fully understood. Exosomes are thought to be one of the ways of information communication between parasite and host. In this study, we attempted to explore the communication between Echinococcus granulosus and its host through the medium of exosomes. We collected plasma from E. granulosus patients (CE-EXO) and healthy donors (HD-EXO) and extracted exosomes from the plasma. The expression profile of miRNA in plasma was determined by second generation sequencing. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to annotate the function of target genes of differential miRNAs. Meanwhile, we co-cultured plasma exosomes from healthy donors and plasma exosomes from E. granulosus patients with Jurkat T cells with or without phytohaemagglutinin (PHA) stimulation. The expression of CD69 on Jurkat T cells was detected by flow cytometry. The results showed that the miRNA of exosomes between healthy donors and E. granulosus patients was significantly different. GO and KEGG were used to annotate the function of target genes of differential miRNAs. The results indicate that many important pathways are involved in inflammation, metabolism, and immune response after parasite infection, such as p53 signaling pathway, PI3K-Akt signaling pathway, and glycolysis/gluconeogenesis. Flow cytometry showed that CE-EXO reduced the expression of CD69 + on Jurkat T cells. Our present results suggest that these differentially expressed miRNAs may be important regulators of parasite-host interactions. Meanwhile, functional prediction of its target genes provides valuable information for understanding the mechanism of host-parasite interactions. These results provide clues for future studies on E. granulosus escape from host immune attack, which could help control E. granulosus infection.
Collapse
Affiliation(s)
- Jing Xiao
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Shiyan Integrated Traditional Chinese and Western Medicine Hospital, Shiyan, China
| | - Jian-Wen Wu
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Scientific Technology Center of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Yun-Zhuo Xin
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Scientific Technology Center of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Dong Song
- Department of Otolaryngology Head and Neck Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
- Scientific Technology Center of Ningxia Medical University, Yinchuan, China
| | - Xiao-Ping Gao
- Department of Otolaryngology Head and Neck Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Mei Yin
- Department of Respiratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wei Zhao
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Scientific Technology Center of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Fu-Lin Liu
- Shiyan Integrated Traditional Chinese and Western Medicine Hospital, Shiyan, China
| | - Hao Wang
- Shiyan Integrated Traditional Chinese and Western Medicine Hospital, Shiyan, China
| | - Jie Wang
- Scientific Technology Center of Ningxia Medical University, Yinchuan, China.
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China.
| | - Jia-Qing Zhao
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China.
- Scientific Technology Center of Ningxia Medical University, Yinchuan, China.
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China.
| |
Collapse
|
5
|
Qadeer A, Wajid A, Rafey HA, Nawaz S, Khan S, Rahman SU, Alzahrani KJ, Khan MZ, Alsabi MNS, Ullah H, Safi SZ, Xia Z, Zahoor M. Exploring extracellular vesicles in zoonotic helminth biology: implications for diagnosis, therapeutic and delivery. Front Cell Infect Microbiol 2024; 14:1424838. [PMID: 39165921 PMCID: PMC11333462 DOI: 10.3389/fcimb.2024.1424838] [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: 04/28/2024] [Accepted: 07/03/2024] [Indexed: 08/22/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as key intercellular communication and pathogenesis mediators. Parasitic organisms' helminths, cause widespread infections with significant health impacts worldwide. Recent research has shed light on the role of EVs in the lifecycle, immune evasion, and disease progression of these parasitic organisms. These tiny membrane-bound organelles including microvesicles and exosomes, facilitate the transfer of proteins, lipids, mRNAs, and microRNAs between cells. EVs have been isolated from various bodily fluids, offering a potential diagnostic and therapeutic avenue for combating infectious agents. According to recent research, EVs from helminths hold great promise in the diagnosis of parasitic infections due to their specificity, early detection capabilities, accessibility, and the potential for staging and monitoring infections, promote intercellular communication, and are a viable therapeutic tool for the treatment of infectious agents. Exploring host-parasite interactions has identified promising new targets for diagnostic, therapy, and vaccine development against helminths. This literature review delves into EVS's origin, nature, biogenesis, and composition in these parasitic organisms. It also highlights the proteins and miRNAs involved in EV release, providing a comprehensive summary of the latest findings on the significance of EVs in the biology of helminths, promising targets for therapeutic and diagnostic biomarkers.
Collapse
Affiliation(s)
- Abdul Qadeer
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Abdul Wajid
- Faculty of Pharmacy, Gomal University, Dera Ismail Khan, Pakistan
| | - Hafiz Abdul Rafey
- Shifa College of Pharmaceutical Sciences, Faculty of Pharmaceutical and Allied Health Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Saqib Nawaz
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Sawar Khan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Sajid Ur Rahman
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Khalid J. Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Muhammad Zahoor Khan
- College of Agricultural Science and Engineering, Liaocheng University, Liaocheng, Shandong, China
| | - Mohammad Nafi Solaiman Alsabi
- Department of Basic Veterinary Medical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Hanif Ullah
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu, China
| | - Sher Zaman Safi
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom, Selangor, Malaysia
| | - Zanxian Xia
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Muhammad Zahoor
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
6
|
Pinheiro AAS, Torrecilhas AC, Souza BSDF, Cruz FF, Guedes HLDM, Ramos TD, Lopes‐Pacheco M, Caruso‐Neves C, Rocco PRM. Potential of extracellular vesicles in the pathogenesis, diagnosis and therapy for parasitic diseases. J Extracell Vesicles 2024; 13:e12496. [PMID: 39113589 PMCID: PMC11306921 DOI: 10.1002/jev2.12496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 07/11/2024] [Indexed: 08/11/2024] Open
Abstract
Parasitic diseases have a significant impact on human and animal health, representing a major hazard to the public and causing economic and health damage worldwide. Extracellular vesicles (EVs) have long been recognized as diagnostic and therapeutic tools but are now also known to be implicated in the natural history of parasitic diseases and host immune response modulation. Studies have shown that EVs play a role in parasitic disease development by interacting with parasites and communicating with other types of cells. This review highlights the most recent research on EVs and their role in several aspects of parasite-host interactions in five key parasitic diseases: Chagas disease, malaria, toxoplasmosis, leishmaniasis and helminthiases. We also discuss the potential use of EVs as diagnostic tools or treatment options for these infectious diseases.
Collapse
Affiliation(s)
- Ana Acacia Sá Pinheiro
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health‐NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ)Rio de JaneiroBrazil
| | - Ana Claudia Torrecilhas
- Departamento de Ciências FarmacêuticasDiadema Campus, Instituto de Ciências Ambientais, Químicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)DiademaSão PauloBrazil
| | - Bruno Solano de Freitas Souza
- Center for Biotechnology and Cell TherapySão Rafael HospitalSalvadorBrazil
- D'Or Institute for Research and Education (IDOR)SalvadorBrazil
| | - Fernanda Ferreira Cruz
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health‐NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ)Rio de JaneiroBrazil
| | - Herbert Leonel de Matos Guedes
- Instituto de Microbiologia Paulo de Goés (IMPG)Universidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Fundação Oswaldo Cruz (FIOCRUZ)Instituto Oswaldo Cruz (IOC)Rio de JaneiroBrazil
| | - Tadeu Diniz Ramos
- Instituto de Microbiologia Paulo de Goés (IMPG)Universidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Fundação Oswaldo Cruz (FIOCRUZ)Instituto Oswaldo Cruz (IOC)Rio de JaneiroBrazil
| | - Miqueias Lopes‐Pacheco
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Deparment of PediatricsCenter for Cystic Fibrosis and Airway Disease ResearchEmory University School of MedicineAtlantaGeorgiaUSA
| | - Celso Caruso‐Neves
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health‐NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ)Rio de JaneiroBrazil
- National Institute of Science and Technology for Regenerative MedicineINCT‐REGENERARio de JaneiroBrazil
| | - Patricia R. M. Rocco
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health‐NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ)Rio de JaneiroBrazil
- National Institute of Science and Technology for Regenerative MedicineINCT‐REGENERARio de JaneiroBrazil
| |
Collapse
|
7
|
Zhang X, Yu C, Song L. Progress on the Regulation of the Host Immune Response by Parasite-Derived Exosomes. Pathogens 2024; 13:623. [PMID: 39204224 PMCID: PMC11357678 DOI: 10.3390/pathogens13080623] [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: 07/01/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Exosomes are membrane-bound structures released by cells into the external environment that carry a significant amount of important cargo, such as proteins, DNA, RNA, and lipids. They play a crucial role in intercellular communication. Parasites have complex life cycles and can release exosomes at different stages. Exosomes released by parasitic pathogens or infected cells contain parasitic nucleic acids, antigenic molecules, virulence factors, drug-resistant proteins, proteases, lipids, etc. These components can regulate host gene expression across species or modulate signaling pathways, thereby dampening or activating host immune responses, causing pathological damage, and participating in disease progression. This review focuses on the means by which parasitic exosomes modulate host immune responses, elaborates on the pathogenic mechanisms of parasites, clarifies the interactions between parasites and hosts, and provides a theoretical basis and research directions for the prevention and treatment of parasitic diseases.
Collapse
Affiliation(s)
| | - Chuanxin Yu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China;
| | - Lijun Song
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China;
| |
Collapse
|
8
|
Arbildi P, Muniz-Lagos AC, Fernández E, Giorgi R, Wiater K, Mourglia-Ettlin G, Fernández V. Immunization with a Mu-class glutathione transferase from Echinococcus granulosus induces efficient antibody responses and confers long-term protection against secondary cystic echinococcosis. Microbes Infect 2024; 26:105364. [PMID: 38777107 DOI: 10.1016/j.micinf.2024.105364] [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: 11/09/2023] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Cystic echinococcosis, a zoonosis caused by cestodes belonging to the Echinococcus granulosus sensu lato (s.l.) genetic complex, affects humans and diverse livestock species. Although a veterinary vaccine exhibiting high levels of antibody-mediated protection has successfully reached the market, the large genetic diversity among parasite isolates and their particular host preferences, makes still necessary the search for novel vaccine candidates. Glutathione transferases (GSTs) constitute attractive targets for immunoprophylaxis due to their outstanding relevance in helminth detoxification processes, against both exogenous and endogenous stressors. Among the six GSTs known to be expressed in E. granulosus s.l., EgGST1 (Mu-class), EgGST2 (Sigma-class), and EgGST3 (a still non-classifiable isoenzyme), show the highest proteomic expression. Therefore, their recombinant forms -rEgGST1, rEgGST2 and rEgGST3- were herein analyzed regarding their potential to induce long-term antiparasite protection in mice. Only immunization with rEgGST1 induced long-lasting protection; and accordingly, rEgGST1-specific antibodies enhanced the parasite killing through both the classical activation of the host complement system and the antibody-dependent cellular cytotoxicity by macrophages. These results support further testing of rEgGST1 as a vaccine candidate in diverse hosts due to the broad expression of EgGST1 in different parasite stages and tissues.
Collapse
Affiliation(s)
- Paula Arbildi
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay; Unidad Asociada de Inmunología, Instituto de Química Biológica (IQB), Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay; Departamento de Inmunología, Instituto de Higiene "Prof. Arnoldo Berta", Universidad de la República, Montevideo, Uruguay
| | - Ana Clara Muniz-Lagos
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Eugenia Fernández
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Rosina Giorgi
- Unidad Asociada de Inmunología, Instituto de Química Biológica (IQB), Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Kai Wiater
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Gustavo Mourglia-Ettlin
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay; Unidad Asociada de Inmunología, Instituto de Química Biológica (IQB), Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay; Departamento de Inmunología, Instituto de Higiene "Prof. Arnoldo Berta", Universidad de la República, Montevideo, Uruguay.
| | - Verónica Fernández
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay; Unidad Asociada de Inmunología, Instituto de Química Biológica (IQB), Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay; Departamento de Inmunología, Instituto de Higiene "Prof. Arnoldo Berta", Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
9
|
Espejo C, Ezenwa VO. Extracellular vesicles: an emerging tool for wild immunology. DISCOVERY IMMUNOLOGY 2024; 3:kyae011. [PMID: 39005930 PMCID: PMC11244269 DOI: 10.1093/discim/kyae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/12/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024]
Abstract
The immune system is crucial for defending organisms against pathogens and maintaining health. Traditionally, research in immunology has relied on laboratory animals to understand how the immune system works. However, there is increasing recognition that wild animals, due to their greater genetic diversity, lifespan, and environmental exposures, have much to contribute to basic and translational immunology. Unfortunately, logistical challenges associated with collecting and storing samples from wildlife, and the lack of commercially available species-specific reagents have hindered the advancement of immunological research on wild species. Extracellular vesicles (EVs) are cell-derived nanoparticles present in all body fluids and tissues of organisms spanning from bacteria to mammals. Human and lab animal studies indicate that EVs are involved in a range of immunological processes, and recent work shows that EVs may play similar roles in diverse wildlife species. Thus, EVs can expand the toolbox available for wild immunology research, helping to overcome some of the challenges associated with this work. In this paper, we explore the potential application of EVs to wild immunology. First, we review current understanding of EV biology across diverse organisms. Next, we discuss key insights into the immune system gained from research on EVs in human and laboratory animal models and highlight emerging evidence from wild species. Finally, we identify research themes in wild immunology that can immediately benefit from the study of EVs and describe practical considerations for using EVs in wildlife research.
Collapse
Affiliation(s)
- Camila Espejo
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Vanessa O Ezenwa
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| |
Collapse
|
10
|
Du X, Hua R, He X, Hou W, Li S, Yang A, Yang G. Echinococcus granulosus ubiquitin-conjugating enzymes (E2D2 and E2N) promote the formation of liver fibrosis in TGFβ1-induced LX-2 cells. Parasit Vectors 2024; 17:190. [PMID: 38643149 PMCID: PMC11031992 DOI: 10.1186/s13071-024-06222-8] [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: 10/16/2023] [Accepted: 02/29/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Cystic echinococcosis (CE) is a widespread zoonosis caused by the infection with Echinococcus granulosus sensu lato (E. granulosus s.l.). CE cysts mainly develop in the liver of intermediate hosts, characterized by the fibrotic tissue that separates host organ from parasite. However, precise mechanism underlying the formation of fibrotic tissue in CE remains unclear. METHODS To investigate the potential impact of ubiquitin-conjugating enzymes on liver fibrosis formation in CE, two members of ubiquitin-conjugating (UBC) enzyme of Echinococcus granulosus (EgE2D2 and EgE2N) were recombinantly expressed in Escherichia coli and analyzed for bioinformatics, immunogenicity, localization, and enzyme activity. In addition, the secretory pathway and their effects on the formation of liver fibrosis were also explored. RESULTS Both rEgE2D2 and rEgE2N possess intact UBC domains and active sites, exhibiting classical ubiquitin binding activity and strong immunoreactivity. Additionally, EgE2D2 and EgE2N were widely distributed in protoscoleces and germinal layer, with differences observed in their distribution in 25-day strobilated worms. Further, these two enzymes were secreted to the hydatid fluid and CE-infected sheep liver tissues via a non-classical secretory pathway. Notably, TGFβ1-induced LX-2 cells exposed to rEgE2D2 and rEgE2N resulted in increasing expression of fibrosis-related genes, enhancing cell proliferation, and facilitating cell migration. CONCLUSIONS Our findings suggest that EgE2D2 and EgE2N could secrete into the liver and may interact with hepatic stellate cells, thereby promoting the formation of liver fibrosis.
Collapse
Affiliation(s)
- Xiaodi Du
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ruiqi Hua
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xue He
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Wei Hou
- Sichuan Center for Animal Disease Control and Prevention, Chengdu, 610041, China
| | - Shengqiong Li
- Sichuan Center for Animal Disease Control and Prevention, Chengdu, 610041, China
| | - Aiguo Yang
- Sichuan Center for Animal Disease Control and Prevention, Chengdu, 610041, China.
| | - Guangyou Yang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
11
|
Ancarola ME, Maldonado LL, García LCA, Franchini GR, Mourglia-Ettlin G, Kamenetzky L, Cucher MA. A Comparative Analysis of the Protein Cargo of Extracellular Vesicles from Helminth Parasites. Life (Basel) 2023; 13:2286. [PMID: 38137887 PMCID: PMC10744797 DOI: 10.3390/life13122286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/15/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Helminth parasites cause debilitating-sometimes fatal-diseases in humans and animals. Despite their impact on global health, mechanisms underlying host-parasite interactions are still poorly understood. One such mechanism involves the exchange of extracellular vesicles (EVs), which are membrane-enclosed subcellular nanoparticles. To date, EV secretion has been studied in helminth parasites, including EV protein content. However, information is highly heterogeneous, since it was generated in multiple species, using varied protocols for EV isolation and data analysis. Here, we compared the protein cargo of helminth EVs to identify common markers for each taxon. For this, we integrated published proteomic data and performed a comparative analysis through an orthology approach. Overall, only three proteins were common in the EVs of the seven analyzed species. Additionally, varied repertoires of proteins with moonlighting activity, vaccine antigens, canonical and non-canonical proteins related to EV biogenesis, taxon-specific proteins of unknown function and RNA-binding proteins were observed in platyhelminth and nematode EVs. Despite the lack of consensus on EV isolation protocols and protein annotation, several proteins were shown to be consistently detected in EV preparations from organisms at different taxa levels, providing a starting point for a selective biochemical characterization.
Collapse
Affiliation(s)
- María Eugenia Ancarola
- Department of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires C1121, Argentina; (M.E.A.); (L.L.M.)
- Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires C1121, Argentina
| | - Lucas L. Maldonado
- Department of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires C1121, Argentina; (M.E.A.); (L.L.M.)
- Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires C1121, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073, Argentina
| | - Lucía C. A. García
- Department of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires C1121, Argentina; (M.E.A.); (L.L.M.)
- Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires C1121, Argentina
| | - Gisela R. Franchini
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Facultad de Ciencias Médicas, Universidad Nacional de La Plata (UNLP)-Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), La Plata B1900, Argentina;
- Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), La Plata B1900, Argentina
| | - Gustavo Mourglia-Ettlin
- Área Inmunología, Departamento de Biociencias, Facultad de Química, Universidad de la República, Montevideo 11800, Uruguay;
| | - Laura Kamenetzky
- Instituto de Biociencias, Biotecnología y Biología Traslacional, Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428, Argentina;
| | - Marcela A. Cucher
- Department of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires C1121, Argentina; (M.E.A.); (L.L.M.)
- Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires C1121, Argentina
| |
Collapse
|
12
|
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
|
13
|
Sadr S, Borji H. Echinococcus granulosus as a Promising Therapeutic Agent against Triplenegative Breast Cancer. CURRENT CANCER THERAPY REVIEWS 2023; 19:292-297. [DOI: 10.2174/1573394719666230427094247] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/06/2022] [Accepted: 01/19/2023] [Indexed: 08/19/2024]
Abstract
Abstract:Breast cancer is a major cause of cancer deaths in women, with approximately 1.2 million new cases per year. Current treatment options for breast cancer include surgery, radiation, hormone therapy, and chemotherapy. However, the non-selective cytotoxicity of chemotherapeutic agents often leads to severe side effects, while drug resistance can worsen patient outcomes. Therefore, the development of more effective and less toxic anticancer drugs is a critical need. This study aimed to review the literature on Echinococcus granulosus antigens with anticancer potential against triple-negative breast cancer. Recent studies have suggested that certain parasite antigens may have potential anticancer effects. Specifically, research has shown that echinococcosis, a disease caused by the parasitic cestode Echinococcus granulosus, may have a protective effect against cancer. These findings offer new insights into the potential use of E. granulosus antigens in the development of novel cancer therapies and tumor cell vaccines. The findings of recent studies suggested that E. granulosus antigens may have the potential to be used in effective and less toxic cancer treatments. However, further research is needed to fully understand the mechanisms behind the anticancer effects of these antigens and develop new cancer therapies and vaccines
Collapse
Affiliation(s)
- Soheil Sadr
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
14
|
Fernandez‐Becerra C, Xander P, Alfandari D, Dong G, Aparici‐Herraiz I, Rosenhek‐Goldian I, Shokouhy M, Gualdron‐Lopez M, Lozano N, Cortes‐Serra N, Karam PA, Meneghetti P, Madeira RP, Porat Z, Soares RP, Costa AO, Rafati S, da Silva A, Santarém N, Fernandez‐Prada C, Ramirez MI, Bernal D, Marcilla A, Pereira‐Chioccola VL, Alves LR, Portillo HD, Regev‐Rudzki N, de Almeida IC, Schenkman S, Olivier M, Torrecilhas AC. Guidelines for the purification and characterization of extracellular vesicles of parasites. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e117. [PMID: 38939734 PMCID: PMC11080789 DOI: 10.1002/jex2.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/21/2023] [Accepted: 09/14/2023] [Indexed: 06/29/2024]
Abstract
Parasites are responsible for the most neglected tropical diseases, affecting over a billion people worldwide (WHO, 2015) and accounting for billions of cases a year and responsible for several millions of deaths. Research on extracellular vesicles (EVs) has increased in recent years and demonstrated that EVs shed by pathogenic parasites interact with host cells playing an important role in the parasite's survival, such as facilitation of infection, immunomodulation, parasite adaptation to the host environment and the transfer of drug resistance factors. Thus, EVs released by parasites mediate parasite-parasite and parasite-host intercellular communication. In addition, they are being explored as biomarkers of asymptomatic infections and disease prognosis after drug treatment. However, most current protocols used for the isolation, size determination, quantification and characterization of molecular cargo of EVs lack greater rigor, standardization, and adequate quality controls to certify the enrichment or purity of the ensuing bioproducts. We are now initiating major guidelines based on the evolution of collective knowledge in recent years. The main points covered in this position paper are methods for the isolation and molecular characterization of EVs obtained from parasite-infected cell cultures, experimental animals, and patients. The guideline also includes a discussion of suggested protocols and functional assays in host cells.
Collapse
Affiliation(s)
- Carmen Fernandez‐Becerra
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
- IGTP Institut d'Investigació Germans Trias i PujolBadalona (Barcelona)Spain
- CIBERINFECISCIII‐CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos IIIMadridSpain
| | - Patrícia Xander
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| | - Daniel Alfandari
- Department of Biomolecular SciencesWeizmann Institute of Science (WIS)RehovotIsrael
| | - George Dong
- The Research Institute of the McGill University Health CentreMcGill UniversityMontréalQuébecCanada
| | - Iris Aparici‐Herraiz
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
| | | | - Mehrdad Shokouhy
- Department of Immunotherapy and Leishmania Vaccine ResearchPasteur Institute of IranTehranIran
| | - Melisa Gualdron‐Lopez
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
| | - Nicholy Lozano
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| | - Nuria Cortes‐Serra
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
| | - Paula Abou Karam
- Department of Biomolecular SciencesWeizmann Institute of Science (WIS)RehovotIsrael
| | - Paula Meneghetti
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| | - Rafael Pedro Madeira
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| | - Ziv Porat
- Flow Cytometry UnitLife Sciences Core Facilities, WISRehovotIsrael
| | | | - Adriana Oliveira Costa
- Departamento de Análises Clínicas e ToxicológicasFaculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG)Belo HorizonteMinas GeraisBrasil
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine ResearchPasteur Institute of IranTehranIran
| | - Anabela‐Cordeiro da Silva
- Host‐Parasite Interactions GroupInstitute of Research and Innovation in HealthUniversity of PortoPortoPortugal
- Department of Biological SciencesFaculty of PharmacyUniversity of PortoPortoPortugal
| | - Nuno Santarém
- Host‐Parasite Interactions GroupInstitute of Research and Innovation in HealthUniversity of PortoPortoPortugal
- Department of Biological SciencesFaculty of PharmacyUniversity of PortoPortoPortugal
| | | | - Marcel I. Ramirez
- EVAHPI ‐ Extracellular Vesicles and Host‐Parasite Interactions Research Group Laboratório de Biologia Molecular e Sistemática de TripanossomatideosInstituto Carlos Chagas‐FiocruzCuritibaParanáBrasil
| | - Dolores Bernal
- Departament de Bioquímica i Biologia Molecular, Facultat de Ciències BiològiquesUniversitat de ValènciaBurjassotValenciaSpain
| | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i ParasitologiaUniversitat de ValènciaBurjassotValenciaSpain
| | - Vera Lucia Pereira‐Chioccola
- Laboratório de Biologia Molecular de Parasitas e Fungos, Centro de Parasitologia e MicologiaInstituto Adolfo Lutz (IAL)São PauloBrasil
| | - Lysangela Ronalte Alves
- Laboratório de Regulação da Expressão GênicaInstituto Carlos ChagasFiocruz ParanáCuritibaBrazil
- Research Center in Infectious DiseasesDivision of Infectious Disease and Immunity CHU de Quebec Research CenterDepartment of MicrobiologyInfectious Disease and ImmunologyFaculty of MedicineUniversity LavalQuebec CityQuebecCanada
| | - Hernando Del Portillo
- ISGlobal, Barcelona Institute for Global HealthHospital Clínic‐Universitatde BarcelonaBarcelonaSpain
- IGTP Institut d'Investigació Germans Trias i PujolBadalona (Barcelona)Spain
- ICREA Institució Catalana de Recerca i Estudis Avanc¸ats (ICREA)BarcelonaSpain
| | - Neta Regev‐Rudzki
- Department of Biomolecular SciencesWeizmann Institute of Science (WIS)RehovotIsrael
| | - Igor Correia de Almeida
- Department of Biological SciencesBorder Biomedical Research CenterThe University of Texas at El PasoEl PasoTexasUSA
| | - Sergio Schenkman
- Departamento de MicrobiologiaImunologia e Parasitologia, UNIFESPSão PauloBrazil
| | - Martin Olivier
- The Research Institute of the McGill University Health CentreMcGill UniversityMontréalQuébecCanada
| | - Ana Claudia Torrecilhas
- Departamento de Ciências FarmacêuticasLaboratório de Imunologia Celular e Bioquímica de Fungos e ProtozoáriosDepartamento de Ciências FarmacêuticasInstituto de Ciências AmbientaisQuímicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)São PauloBrazil
| |
Collapse
|
15
|
Khosravi M, Mohammad Rahimi H, Nazari A, Baghaei K, Asadzadeh Aghdaei H, Shahrokh S, Sharifdini M, Torrecilhas AC, Mehryab F, Mirjalali H, Shekari F, Zali MR. Characterisation of extracellular vesicles isolated from hydatid cyst fluid and evaluation of immunomodulatory effects on human monocytes. J Cell Mol Med 2023; 27:2614-2625. [PMID: 37530547 PMCID: PMC10468670 DOI: 10.1111/jcmm.17894] [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: 07/27/2022] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023] Open
Abstract
Hydatidosis is a disease caused by the larval stage of Echinococcus granulosus, which involves several organs of intermediate hosts. Evidence suggests a communication between hydatid cyst (HC) and hosts via extracellular vesicles. However, a little is known about the communication between EVs derived from HC fluid (HCF) and host cells. In the current study, EVs were isolated using differential centrifugation from sheep HCF and characterized by western blot, electron microscope and size distribution analysis. The uptake of EVs by human monocyte cell line (THP-1) was evaluated. The effects of EVs on the expression levels of pro- and anti-inflammatory cytokines were investigated using quantitative real-time PCR (RT-PCR), 3 and 24 h after incubation. Moreover, the cytokine level of IL-10 was evaluated in supernatant of THP-1 cell line at 3 and 24 h. EVs were successfully isolated and showed spherical shape with size distribution at 130.6 nm. After 3 h, the expression levels of pro-inflammatory cytokine genes (IL1Β, IL15 and IL8) were upregulated, while after 24 h, the expression levels of pro-inflammatory cytokines were decreased and IL13 gene expression showed upregulation. A statistically significant increase was seen in the levels of IL-10 after 24 h. The main mechanism of the communication between EVs derived from HCF and their host remains unclear; however, time-dependent anti-inflammatory effects in our study suggest that HC may modulate the immune responses via EVs.
Collapse
Affiliation(s)
- Mojdeh Khosravi
- Department of Pharmacy and Pharmaceutical Technology and ParasitologyUniversity of ValenciaValenciaSpain
| | - Hanieh Mohammad Rahimi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Abdoreza Nazari
- Department of Molecular Systems Biology at Cell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyTehranIran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Shabnam Shahrokh
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Meysam Sharifdini
- Department of Medical Parasitology and Mycology, School of MedicineGuilan University of Medical SciencesRashtIran
| | - Ana Claudia Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)DiademaBrazil
| | - Fatemeh Mehryab
- Department of Molecular Systems Biology at Cell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyTehranIran
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of PharmacyShahid Beheshti University of Medical SciencesTehranIran
| | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Faezeh Shekari
- Department of Molecular Systems Biology at Cell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyTehranIran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
16
|
Liu C, Cao J, Zhang H, Field MC, Yin J. Extracellular vesicles secreted by Echinococcus multilocularis: important players in angiogenesis promotion. Microbes Infect 2023; 25:105147. [PMID: 37142117 DOI: 10.1016/j.micinf.2023.105147] [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: 10/25/2022] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/06/2023]
Abstract
The involvement of Echinococcus multilocularis, and other parasitic helminths, in regulating host physiology is well recognized, but molecular mechanisms remain unclear. Extracellular vesicles (EVs) released by helminths play important roles in regulating parasite-host interactions by transferring materials to the host. Analysis of protein cargo of EVs from E. multilocularis protoscoleces in the present study revealed a unique composition exclusively associated with vesicle biogenesis. Common proteins in various Echinococcus species were identified, including the classical EVs markers tetraspanins, TSG101 and Alix. Further, unique tegumental antigens were identified which could be exploited as Echinococcus EV markers. Parasite- and host-derived proteins within these EVs are predicted to support important roles in parasite-parasite and parasite-host communication. In addition, the enriched host-derived protein payloads identified in parasite EVs in the present study suggested that they can be involved in focal adhesion and potentially promote angiogenesis. Further, increased angiogenesis was observed in livers of mice infected with E. multilocularis and the expression of several angiogenesis-regulated molecules, including VEGF, MMP9, MCP-1, SDF-1 and serpin E1 were increased. Significantly, EVs released by the E. multilocularis protoscolex promoted proliferation and tube formation by human umbilical vein endothelial cells (HUVECs) in vitro. Taken together, we present the first evidence that tapeworm-secreted EVs may promote angiogenesis in Echinococcus-infections, identifying central mechanisms of Echinococcus-host interactions.
Collapse
Affiliation(s)
- Congshan Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); NHC Key Laboratory of Parasite and Vector Biology; WHO Collaborating Center for Tropical Diseases; National Center for International Research on Tropical Diseases, Shanghai, 200025, China
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); NHC Key Laboratory of Parasite and Vector Biology; WHO Collaborating Center for Tropical Diseases; National Center for International Research on Tropical Diseases, Shanghai, 200025, China
| | - Haobing Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); NHC Key Laboratory of Parasite and Vector Biology; WHO Collaborating Center for Tropical Diseases; National Center for International Research on Tropical Diseases, Shanghai, 200025, China
| | - Mark C Field
- School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK; Institute of Parasitology, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Jianhai Yin
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); NHC Key Laboratory of Parasite and Vector Biology; WHO Collaborating Center for Tropical Diseases; National Center for International Research on Tropical Diseases, Shanghai, 200025, China.
| |
Collapse
|
17
|
Nicolao MC, Rodrigues CR, Coccimiglio MB, Ledo C, Docena GH, Cumino AC. Characterization of protein cargo of Echinococcus granulosus extracellular vesicles in drug response and its influence on immune response. Parasit Vectors 2023; 16:255. [PMID: 37516852 PMCID: PMC10387209 DOI: 10.1186/s13071-023-05854-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/28/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND The Echinococcus granulosus sensu lato species complex causes cystic echinococcosis, a zoonotic disease of medical importance. Parasite-derived small extracellular vesicles (sEVs) are involved in the interaction with hosts intervening in signal transduction related to parasite proliferation and disease pathogenesis. Although the characteristics of sEVs from E. granulosus protoscoleces and their interaction with host dendritic cells (DCs) have been described, the effect of sEVs recovered during parasite pharmacological treatment on the immune response remains unexplored. METHODS Here, we isolated and characterized sEVs from control and drug-treated protoscoleces by ultracentrifugation, transmission electron microscopy, dynamic light scattering, and proteomic analysis. In addition, we evaluated the cytokine response profile induced in murine bone marrow-derived dendritic cells (BMDCs) by qPCR. RESULTS The isolated sEVs, with conventional size between 50 and 200 nm, regardless of drug treatment, showed more than 500 cargo proteins and, importantly, 20 known antigens and 70 potential antigenic proteins, and several integral-transmembrane and soluble proteins mainly associated with signal transduction, immunomodulation, scaffolding factors, extracellular matrix-anchoring, and lipid transport. The identity and abundance of proteins in the sEV-cargo from metformin- and albendazole sulfoxide (ABZSO)-treated parasites were determined by proteomic analysis, detecting 107 and eight exclusive proteins, respectively, which include proteins related to the mechanisms of drug action. We also determined that the interaction of murine BMDCs with sEVs derived from control parasites and those treated with ABZSO and metformin increased the expression of pro-inflammatory cytokines such as IL-12 compared to control cells. Additionally, protoscolex-derived vesicles from metformin treatments induced the production of IL-6, TNF-α, and IL-10. However, the expression of IL-23 and TGF-β was downregulated. CONCLUSIONS We demonstrated that sEV-cargo derived from drug-treated E. granulosus protoscoleces have immunomodulatory functions, as they enhance DC activation towards a type 1 pro-inflammatory profile against the parasite, and therefore support the proposal of a new approach for the prevention and treatment of secondary echinococcosis.
Collapse
Affiliation(s)
- María Celeste Nicolao
- Laboratorio de Zoonosis Parasitarias, IIPROSAM, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, 7600, Mar del Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Christian Rodriguez Rodrigues
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, 7600, Mar del Plata, Argentina
| | - Magalí B Coccimiglio
- Laboratorio de Zoonosis Parasitarias, IIPROSAM, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, 7600, Mar del Plata, Argentina
| | - Camila Ledo
- Laboratorio de Zoonosis Parasitarias, IIPROSAM, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, 7600, Mar del Plata, Argentina
| | - Guillermo H Docena
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), La Plata, Argentina
| | - Andrea C Cumino
- Laboratorio de Zoonosis Parasitarias, IIPROSAM, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, 7600, Mar del Plata, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, 7600, Mar del Plata, Argentina.
| |
Collapse
|
18
|
Cucher MA, Mariconti M, Manciulli T, Vola A, Rosenzvit MC, Brehm K, Kamenetzky L, Brunetti E. Circulating Small RNA Profiling of Patients with Alveolar and Cystic Echinococcosis. BIOLOGY 2023; 12:biology12050715. [PMID: 37237528 DOI: 10.3390/biology12050715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/19/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
Alveolar (AE) and cystic (CE) echinococcosis are two parasitic diseases caused by the tapeworms Echinococcus multilocularis and E. granulosus sensu lato (s. l.), respectively. Currently, AE and CE are mainly diagnosed by means of imaging techniques, serology, and clinical and epidemiological data. However, no viability markers that indicate parasite state during infection are available. Extracellular small RNAs (sRNAs) are short non-coding RNAs that can be secreted by cells through association with extracellular vesicles, proteins, or lipoproteins. Circulating sRNAs can show altered expression in pathological states; hence, they are intensively studied as biomarkers for several diseases. Here, we profiled the sRNA transcriptomes of AE and CE patients to identify novel biomarkers to aid in medical decisions when current diagnostic procedures are inconclusive. For this, endogenous and parasitic sRNAs were analyzed by sRNA sequencing in serum from disease negative, positive, and treated patients and patients harboring a non-parasitic lesion. Consequently, 20 differentially expressed sRNAs associated with AE, CE, and/or non-parasitic lesion were identified. Our results represent an in-depth characterization of the effect E. multilocularis and E. granulosus s. l. exert on the extracellular sRNA landscape in human infections and provide a set of novel candidate biomarkers for both AE and CE detection.
Collapse
Affiliation(s)
- Marcela A Cucher
- Department of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires C1121ABG, Argentina
- Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Mara Mariconti
- Unit of Infectious and Tropical Diseases, San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Tommaso Manciulli
- Unit of Infectious and Tropical Diseases, San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Ambra Vola
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Mara C Rosenzvit
- Department of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires C1121ABG, Argentina
- Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Klaus Brehm
- Institute for Hygiene and Microbiology, University of Würzburg, 97080 Würzburg, Germany
| | - Laura Kamenetzky
- Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Enrico Brunetti
- Immunology and Infectious Diseases, San Matteo Hospital Foundation, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
19
|
Kronenberg PA, Reinehr M, Eichenberger RM, Hasler S, Laurimäe T, Weber A, Deibel A, Müllhaupt B, Gottstein B, Müller N, Hemphill A, Deplazes P. Monoclonal antibody-based localization of major diagnostic antigens in metacestode tissue, excretory/secretory products, and extracellular vesicles of Echinococcus species. Front Cell Infect Microbiol 2023; 13:1162530. [PMID: 37009502 PMCID: PMC10061086 DOI: 10.3389/fcimb.2023.1162530] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Alveolar (AE) and cystic echinococcosis (CE) are severe parasitic zoonoses caused by the larval stages of Echinococcus multilocularis and E. granulosus sensu lato, respectively. A panel of 7 monoclonal antibodies (mAbs) was selected against major diagnostic epitopes of both species. The binding capacity of the mAbs to Echinococcus spp. excretory/secretory products (ESP) was analyzed by sandwich-ELISA, where mAb Em2G11 and mAb EmG3 detected in vitro extravesicular ESP of both E. multilocularis and E. granulosus s.s. These findings were subsequently confirmed by the detection of circulating ESP in a subset of serum samples from infected hosts including humans. Extracellular vesicles (EVs) were purified, and the binding to mAbs was analyzed by sandwich-ELISA. Transmission electron microscopy (TEM) was used to confirm the binding of mAb EmG3 to EVs from intravesicular fluid of Echinococcus spp. vesicles. The specificity of the mAbs in ELISA corresponded to the immunohistochemical staining (IHC-S) patterns performed on human AE and CE liver sections. Antigenic small particles designated as ''spems'' for E. multilocularis and ''spegs'' for E. granulosus s.l. were stained by the mAb EmG3IgM, mAb EmG3IgG1, mAb AgB, and mAb 2B2, while mAb Em2G11 reacted with spems and mAb Eg2 with spegs only. The laminated layer (LL) of both species was strongly visualized by using mAb EmG3IgM, mAb EmG3IgG1, mAb AgB, and mAb 2B2. The LL was specifically stained by mAb Em2G11 in E. multilocularis and by mAb Eg2 in E. granulosus s.l. In the germinal layer (GL), including the protoscoleces, a wide staining pattern with all structures of both species was observed with mAb EmG3IgG1, mAb EmG3IgM, mAb AgB, mAb 2B2, and mAb Em18. In the GL and protoscoleces, the mAb Eg2 displayed a strong E. granulosus s.l. specific binding, while mAb Em2G11 exhibited a weak granular E. multilocularis specific reaction. The most notable staining pattern in IHC-S was found with mAb Em18, which solely bound to the GL and protoscoleces of Echinococcus species and potentially to primary cells. To conclude, mAbs represent valuable tools for the visualization of major antigens in the most important Echinococcus species, as well as providing insights into parasite-host interactions and pathogenesis.
Collapse
Affiliation(s)
- Philipp A. Kronenberg
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Michael Reinehr
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ramon Marc Eichenberger
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
- Microbiology and Molecular Biology, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich University of Applied Sciences’ (ZHAW), Wädenswil, Switzerland
| | - Sina Hasler
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Teivi Laurimäe
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ansgar Deibel
- Department of Gastroenterology and Hepatology and Swiss HPB and Transplant Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Beat Müllhaupt
- Department of Gastroenterology and Hepatology and Swiss HPB and Transplant Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bruno Gottstein
- Institute for Infectious Diseases, Medical Faculty, University of Bern, Bern, Switzerland
| | - Norbert Müller
- Institute for Infectious Diseases, Medical Faculty, University of Bern, Bern, Switzerland
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Peter Deplazes
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
- Department of Gastroenterology and Hepatology and Swiss HPB and Transplant Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Mohammadi MA, Mansouri M, Derakhshani A, Rezaie M, Borhani M, Nasibi S, Mousavi SM, Afgar A, Macchiaroli N, Rosenzvit MC, Harandi MF. MicroRNA-Transcription factor regulatory networks in the early strobilar development of Echinococcus granulosus protoscoleces. BMC Genomics 2023; 24:114. [PMID: 36922762 PMCID: PMC10016175 DOI: 10.1186/s12864-023-09199-3] [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: 11/25/2022] [Accepted: 02/21/2023] [Indexed: 03/17/2023] Open
Abstract
BACKGROUND Echinococcus granulosus sensu lato has a complex developmental biology with a variety of factors relating to both intermediate and final hosts. To achieve maximum parasite adaptability, the development of the cestode is dependent on essential changes in transcript regulation. Transcription factors (TFs) and miRNAs are known as master regulators that affect the expression of downstream genes through a wide range of metabolic and signaling pathways. In this study, we aimed to develop a regulatory miRNA-Transcription factor (miRNA-TF) network across early developmental stages of E. granulosus protoscoleces by performing in silico analysis, and to experimentally validate TFs expression in protoscoleces obtained from in vitro culture, and from in vivo experiments. RESULTS We obtained list of 394 unique E. granulosus TFs and matched them with 818 differentially expressed genes which identified 41 predicted TFs with differential expression. These TFs were used to predict the potential targets of 31 differentially expressed miRNAs. As a result, eight miRNAs and eight TFs were found, and the predicted network was constructed using Cytoscape. At least four miRNAs (egr-miR-124a, egr-miR-124b-3p, egr-miR-745-3p, and egr-miR-87-3p) and their corresponding differentially expressed TFs (Zinc finger protein 45, Early growth response protein 3, Ecdysone induced protein 78c and ETS transcription factor elf 2) were highlighted in this investigation. The expression of predicted differentially expressed TFs obtained from in vitro and in vivo experiments, were experimentally validated by quantitative polymerase chain reaction. This confirmed findings of RNA-seq data. CONCLUSION miRNA-TF networks presented in this study control some of the most important metabolic and signaling pathways in the development and life cycle of E. granulosus, providing a potential approach for disrupting the early hours of dog infection and preventing the development of the helminth in the final host.
Collapse
Affiliation(s)
- Mohammad Ali Mohammadi
- Research Center for Hydatid Disease in Iran, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Mansouri
- Department of Agricultural Biotechnology, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Ali Derakhshani
- Research Center for Hydatid Disease in Iran, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Masoud Rezaie
- Student Research Committee, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Borhani
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Jilin, China
| | - Saeid Nasibi
- Research Center for Hydatid Disease in Iran, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed Mohammad Mousavi
- Research Center for Hydatid Disease in Iran, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Afgar
- Research Center for Hydatid Disease in Iran, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Natalia Macchiaroli
- Laboratorio Biología Molecular de Hidatidosis, Facultad de Medicina, Instituto de Microbiología Y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas Y Tecnológicas (CONICET), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Mara C. Rosenzvit
- Laboratorio Biología Molecular de Hidatidosis, Facultad de Medicina, Instituto de Microbiología Y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas Y Tecnológicas (CONICET), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Majid Fasihi Harandi
- Research Center for Hydatid Disease in Iran, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
21
|
Zhang X, Duan S, Li X, Ding J, Zuo L, Sun B, Zhang X, Jiang X, Gao Y, Hu X, Han S. Differences in the secretory exosomes of Clonorchis sinensis adults at different incubation times. Acta Trop 2022; 234:106604. [PMID: 35820469 DOI: 10.1016/j.actatropica.2022.106604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/16/2022] [Accepted: 07/08/2022] [Indexed: 11/26/2022]
Abstract
Exosomes are small membrane vesicles of endocytic origin and widely involved in a variety of physiological and pathological conditions. Exosome-like vesicles (ELVs) have been identified to mediate the parasite-host interactions and communication. Thus, increased knowledge of C. sinensis ELVs could provide insights into parasite-host interactions. In this experiment, ELVs was purified by ultracentrifugation from the culture medium of C. sinensis adults in vitro incubated for 24 h and 48 h, respectively. Transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) confirmed that the purified vesicles which ranged from 30 to 150 nm in size were present in the culture medium. Small RNA high-throughput sequencing analysis identified 51 miRNAs, including 37 known C. sinensis miRNAs, 3 novel C. sinensis miRNAs and 11 rat miRNAs. The sequencing data were validated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The biological function of targets of known C. sinensis miRNAs were proved to associated with signal transduction, infectious diseases and the immune system. Further, 15 miRNAs were classified as differentially expressed in the 24h-ELVs compared to the 48h-ELVs. We found that the numbers and expression levels of most miRNAs from 24h-ELVs were more and higer than 48h-ELVs'. Our work provides important data for understanding the molecular mechanisms underlying the pathogenesis of C. sinensis adults ELVs.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Parasitology, Harbin Medical University, Harbin, PR China
| | - Shanshan Duan
- Department of Parasitology, Harbin Medical University, Harbin, PR China.
| | - Xiang Li
- Department of Parasitology, Harbin Medical University, Harbin, PR China
| | - Jian Ding
- Department of Parasitology, Harbin Medical University, Harbin, PR China
| | - Lijiao Zuo
- Department of Parasitology, Harbin Medical University, Harbin, PR China.
| | - Beibei Sun
- Zhuhai Maternal and Child Health Hospital, Guangdong, PR China.
| | - Xueli Zhang
- Department of Parasitology, Harbin Medical University, Harbin, PR China
| | - Xu Jiang
- Department of Parasitology, Harbin Medical University, Harbin, PR China
| | - Yannan Gao
- Department of Graduate Studies, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, PR China.
| | - Xinyi Hu
- Department of Stomatology, Laixi People's Hospital, Shandong, PR China
| | - Su Han
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China; Department of Parasitology, Harbin Medical University, Harbin, PR China.
| |
Collapse
|
22
|
Expression profiling of exosomal miRNAs derived from different stages of infection in mice infected with Echinococcus granulosus protoscoleces using high-throughput sequencing. Parasitol Res 2022; 121:1993-2008. [PMID: 35511364 DOI: 10.1007/s00436-022-07536-1] [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: 12/13/2021] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
Echinococcosis is a worldwide zoonosis. The mechanism of the establishment, growth, and persistence of parasites in the host has not been fully understood. Exosomes are found to be a way of information exchange between parasites and hosts. They exist in various body fluids widely. There are few studies on host-derived exosomes and their miRNA expression profiles at different infection time points. In this study, BALB/c mice were intraperitoneally infected with protricercariae. Exosomes were extracted from plasma (0, 3, 9, and 20 weeks post infection), and the expression profiles of exosome miRNA in the peripheral blood of mice were determined using RNA-sequencing. Compared to the 0 week groups, 24, 35, and 22 differentially expressed miRNAs were detected in infected mouse at the three infection stages, respectively. The results showed that there were significant differences in the miRNAs of exosomes at different infection time points. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were used to annotate the different miRNAs. The results showed that the biological pathways of parasites changed significantly at different stages of infection, with many significant and abundant pathways involved in cell differentiation, inflammation, and immune response, such as MAPK signaling pathway, Th17 cell differentiation, Wnt signaling pathway, FoxO signaling pathway, Notch signaling pathway, etc. These results suggest that miRNA may be an important regulator of interactions between Echinococcus granulosus and host. The data provided here provide valuable information to increase understanding of the regulatory function of microRNAs in the host microenvironment and the mechanism of host-parasite interaction. This may help us to find targets for Echinococcus granulosus to escape host immune attack and control Echinococcus granulosus infection in the future.
Collapse
|
23
|
Dos Santos GB, da Silva ED, Kitano ES, Battistella ME, Monteiro KM, de Lima JC, Ferreira HB, Serrano SMDT, Zaha A. Proteomic profiling of hydatid fluid from pulmonary cystic echinococcosis. Parasit Vectors 2022; 15:99. [PMID: 35313982 PMCID: PMC8935821 DOI: 10.1186/s13071-022-05232-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/03/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Most cystic echinococcosis cases in Southern Brazil are caused by Echinococcus granulosus and Echinococcus ortleppi. Proteomic studies of helminths have increased our knowledge about the molecular survival strategies that are used by parasites. Here, we surveyed the protein content of the hydatid fluid compartment in E. granulosus and E. ortleppi pulmonary bovine cysts to better describe and compare their molecular arsenal at the host-parasite interface. METHODS Hydatid fluid samples from three isolates of each species were analyzed using mass spectrometry-based proteomics (LC-MS/MS). In silico functional analyses of the identified proteins were performed to examine parasite survival strategies. RESULTS The identified hydatid fluid protein profiles showed a predominance of parasite proteins compared to host proteins that infiltrate the cysts. We identified 280 parasitic proteins from E. granulosus and 251 from E. ortleppi, including 52 parasitic proteins that were common to all hydatid fluid samples. The in silico functional analysis revealed important molecular functions and processes that are active in pulmonary cystic echinococcosis, such as adhesion, extracellular structures organization, development regulation, signaling transduction, and enzyme activity. CONCLUSIONS The protein profiles described here provide evidence of important mechanisms related to basic cellular processes and functions that act at the host-parasite interface in cystic echinococcosis. The molecular tools used by E. granulosus and E. ortleppi for survival within the host are potential targets for new therapeutic approaches to treat cystic echinococcosis and other larval cestodiases.
Collapse
Affiliation(s)
- Guilherme Brzoskowski Dos Santos
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Edileuza Danieli da Silva
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo Shigueo Kitano
- Laboratório de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Maria Eduarda Battistella
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Karina Mariante Monteiro
- Laboratório de Genômica Estrutural E Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jeferson Camargo de Lima
- Laboratório de Genômica Estrutural E Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Henrique Bunselmeyer Ferreira
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratório de Genômica Estrutural E Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Solange Maria de Toledo Serrano
- Laboratório de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Arnaldo Zaha
- Laboratório de Biologia Molecular de Cestódeos, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil. .,Laboratório de Genômica Estrutural E Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
24
|
Shi C, Zhou X, Yang W, Wu J, Bai M, Zhang Y, Zhao W, Yang H, Nagai A, Yin M, Gao X, Ding S, Zhao J. Proteomic Analysis of Plasma-Derived Extracellular Vesicles From Mice With Echinococcus granulosus at Different Infection Stages and Their Immunomodulatory Functions. Front Cell Infect Microbiol 2022; 12:805010. [PMID: 35360110 PMCID: PMC8960237 DOI: 10.3389/fcimb.2022.805010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/14/2022] [Indexed: 01/15/2023] Open
Abstract
The globally distributed cystic echinococcosis (CE) is caused by the larval stage of Echinococcus granulosus (E. granulosus), a cosmopolitan and zoonotic disease with potentially life-threatening complications in humans. The emerging roles for extracellular vesicles (EVs) in parasitic infection include transferring proteins and modifying host cell gene expression to modulate host immune responses. Few studies focused on the host-derived EVs and its protein profiles. We focused on the EVs from mouse infected with E. granulosus at different stages. ExoQuick kit was used for isolating EVs from mouse plasma and ExoEasy Maxi kit was used for isolating protoscolex culture supernatant (PCS) and hydatid cyst fluid (HCF). Firstly, EVs were characterized by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and immunoblot. Secondly, the proteins of plasma EVs were identified using liquid chromatography-tandem mass spectrometry (LC–MS/MS). The resulting LC–MS/MS data were processed using Maxquant search engine (v 1.5.2.8). Tandem mass spectra were researched against the mice and E. granulosus proteins database in the NCBI. The differentially expressed proteins are performed by proteomic label-free quantitative analysis and bioinformatics. Thirdly, in vitro experiment, the results of co-culture of plasma EVs and spleen mononuclear cells showed that 7W-EVs can increase the relative abundance of regulatory T (Treg) cells and IL-10. We further verified that EVs can be internalized by CD4+ and CD8+ T cells, B cells, and myeloid-derived suppressor cells (MDSC). These results implied host-derived EVs are multidirectional immune modulators. The findings can contribute to a better understanding of the role of host-derived EVs which are the optimal vehicle to transfer important cargo into host immune system. In addition, we have found several important proteins associated with E. granulosus and identified in infected mouse plasma at different stages. Furthermore, our study further highlighted the proteomics and immunological function of EVs from mouse infected with E. granulosus protoscoleces at different infection stages. We have laid a solid foundation for the role of EVs in cystic echinococcosis in the future research and supplemented a unique dataset for this E. granulosus.
Collapse
Affiliation(s)
- Chunli Shi
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Department of Molecular Biology, Shanghai Centre for Clinical Laboratory, Shanghai, China
| | - Xiaojing Zhou
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Wenjuan Yang
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Jianwen Wu
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Min Bai
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Ying Zhang
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Wei Zhao
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Research Center for Medical Science and Technology, Ningxia Medical University, Yinchuan, China
- Ningxia Institute of Medical Science, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Hui Yang
- Research Center for Medical Science and Technology, Ningxia Medical University, Yinchuan, China
- Ningxia Institute of Medical Science, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Atsushi Nagai
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Mei Yin
- Department of Respiratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiaoping Gao
- Department of Otolaryngology Head and Neck Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Shuqin Ding
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- *Correspondence: Jiaqing Zhao, ; Shuqin Ding,
| | - Jiaqing Zhao
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Research Center for Medical Science and Technology, Ningxia Medical University, Yinchuan, China
- Ningxia Institute of Medical Science, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
- *Correspondence: Jiaqing Zhao, ; Shuqin Ding,
| |
Collapse
|
25
|
Ghabdian S, Parande Shirvan S, Maleki M, Borji H. Exacerbation of allergic asthma by somatic antigen of Echinococcus granulosus in allergic airway inflammation in BALB/c mice. Parasit Vectors 2022; 15:16. [PMID: 34991711 PMCID: PMC8734303 DOI: 10.1186/s13071-021-05125-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/11/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND There is ample evidence demonstrating a reverse relationship between helminth infection and immune-mediated diseases. Accordingly, several studies have shown that Echinococcus granulosus infection and hydatid cyst compounds are able to suppress immune responses in allergic airway inflammation. Previous studies have documented the ability of hydatid cysts to suppress aberrant Th2 immune response in a mouse model of allergic asthma. However, there is a paucity of research on the effects of protoscoleces on allergic asthma. Thus, this study was designed to evaluate the effects of somatic antigens of protoscoleces in a murine model of allergic airway inflammation. METHODS Ovalbumin (OVA)/aluminum hydroxide (alum) was injected intraperitoneally to sensitize BALB/c mice over a period of 0 to 7 days, followed by challenge with 1% OVA. The treatment group received somatic antigens of protoscoleces emulsified with PBS on these days in each sensitization before being challenged with 1% OVA on days 14, 15, and 16. The effects of somatic antigens of protoscoleces on allergic airway inflammation were evaluated by examining histopathological changes, the recruitment of inflammatory cells in the bronchoalveolar lavage, cytokine production in the homogenized lung tissue (IL-4, IL-5, IL-10, IL-17, and IFN-γ), and total antioxidant capacity in serum. RESULTS Overall, administration of somatic antigens of protoscoleces exacerbated allergic airway inflammation via increased Th2 cytokine levels in the lung homogenate, recruitment of eosinophils into bronchoalveolar lavage fluid, and pathological changes. In addition, total antioxidant capacity and IFN-γ levels declined following the administration of somatic antigens. CONCLUSIONS The results revealed that the co-administration of somatic products of protoscoleces with OVA/alum contributed to the exacerbation of allergic airway inflammation in BALB/c mice. Currently, the main cause of allergic-type inflammation exacerbation is unknown, and further research is needed to understand the mechanism of these interactions.
Collapse
Affiliation(s)
- Sara Ghabdian
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 91775-1793, Mashhad, Iran
| | - Sima Parande Shirvan
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 91775-1793, Mashhad, Iran
| | - Mohsen Maleki
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 91775-1793, Mashhad, Iran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P.O. Box: 91775-1793, Mashhad, Iran
| |
Collapse
|
26
|
Affiliation(s)
- Andrew Hemphill
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Parasitology, University of Bern, Länggass-Strasse 122, 3012Bern, Switzerland
| | - Britta Lundström-Stadelmann
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Parasitology, University of Bern, Länggass-Strasse 122, 3012Bern, Switzerland
| |
Collapse
|
27
|
Drurey C, Maizels RM. Helminth extracellular vesicles: Interactions with the host immune system. Mol Immunol 2021; 137:124-133. [PMID: 34246032 PMCID: PMC8636279 DOI: 10.1016/j.molimm.2021.06.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/14/2021] [Accepted: 06/23/2021] [Indexed: 12/21/2022]
Abstract
As long-lived parasites, helminths depend upon immunomodulation of their hosts for survival. The release of excretory-secretory (ES) products, including proteins, lipids and RNAs is how successful host manipulation is achieved. It has recently been discovered that the ES products of helminths contain extracellular vesicles (EVs), with every species investigated found to secrete these lipid-bound structures. EVs are perfect for packaging and delivering immune modulators to target cell types. This review outlines the research carried out on helminth EVs and their constituents thus far, as well as their interaction with components of the mammalian immune system. We discuss how targeting EVs will aid treatment of helminth infection and consider how EVs and their immunomodulatory cargo could be used as therapeutics as we progress through this exciting era.
Collapse
Affiliation(s)
- Claire Drurey
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK.
| |
Collapse
|
28
|
Wititkornkul B, Hulme BJ, Tomes JJ, Allen NR, Davis CN, Davey SD, Cookson AR, Phillips HC, Hegarty MJ, Swain MT, Brophy PM, Wonfor RE, Morphew RM. Evidence of Immune Modulators in the Secretome of the Equine Tapeworm Anoplocephala perfoliata. Pathogens 2021; 10:pathogens10070912. [PMID: 34358062 PMCID: PMC8308605 DOI: 10.3390/pathogens10070912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/15/2022] Open
Abstract
Anoplocephala perfoliata is a neglected gastro-intestinal tapeworm, commonly infecting horses worldwide. Molecular investigation of A. perfoliata is hampered by a lack of tools to better understand the host-parasite interface. This interface is likely influenced by parasite derived immune modulators released in the secretome as free proteins or components of extracellular vesicles (EVs). Therefore, adult RNA was sequenced and de novo assembled to generate the first A. perfoliata transcriptome. In addition, excretory secretory products (ESP) from adult A. perfoliata were collected and EVs isolated using size exclusion chromatography, prior to proteomic analysis of the EVs, the EV surface and EV depleted ESP. Transcriptome analysis revealed 454 sequences homologous to known helminth immune modulators including two novel Sigma class GSTs, five α-HSP90s, and three α-enolases with isoforms of all three observed within the proteomic analysis of the secretome. Furthermore, secretome proteomics identified common helminth proteins across each sample with known EV markers, such as annexins and tetraspanins, observed in EV fractions. Importantly, 49 of the 454 putative immune modulators were identified across the secretome proteomics contained within and on the surface of EVs in addition to those identified in free ESP. This work provides the molecular tools for A. perfoliata to reveal key players in the host-parasite interaction within the horse host.
Collapse
Affiliation(s)
- Boontarikaan Wititkornkul
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
- Faculty of Veterinary Science, Rajamangala University of Technology Srivijaya, Nakhon Si Thammarat 80240, Thailand
| | - Benjamin J. Hulme
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - John J. Tomes
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - Nathan R. Allen
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - Chelsea N. Davis
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - Sarah D. Davey
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - Alan R. Cookson
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - Helen C. Phillips
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - Matthew J. Hegarty
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - Martin T. Swain
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - Peter M. Brophy
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
| | - Ruth E. Wonfor
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
- Correspondence: (R.E.W.); (R.M.M.)
| | - Russell M. Morphew
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3DA, UK; (B.W.); (B.J.H.); (J.J.T.); (N.R.A.); (C.N.D.); (S.D.D.); (A.R.C.); (H.C.P.); (M.J.H.); (M.T.S.); (P.M.B.)
- Correspondence: (R.E.W.); (R.M.M.)
| |
Collapse
|
29
|
Miles S, Magnone J, García-Luna J, Ancarola ME, Cucher M, Dematteis S, Frischknecht F, Cyrklaff M, Mourglia-Ettlin G. Ultrastructural characterization of the tegument in protoscoleces of Echinococcus ortleppi. Int J Parasitol 2021; 51:989-997. [PMID: 34216624 DOI: 10.1016/j.ijpara.2021.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 12/27/2022]
Abstract
Cystic echinococcosis is a globally distributed zoonosis caused by cestodes of the Echinococcus granulosus sensu lato (s.l.) complex, with Echinococcus ortleppi mainly involved in cattle infection. Protoscoleces show high developmental plasticity, being able to differentiate into either adult worms or metacestodes within definitive or intermediate hosts, respectively. Their outermost cellular layer is called the tegument, which is important in determining the infection outcome through its immunomodulating activities. Herein, we report an in-depth characterization of the tegument of E. ortleppi protoscoleces performed through a combination of scanning and transmission electron microscopy techniques. Using electron tomography, a three-dimensional reconstruction of the tegumental cellular territories was obtained, revealing a novel structure termed the 'tegumental vesicular body' (TVB). Vesicle-like structures, possibly involved in endocytic/exocytic routes, were found within the TVB as well as in the parasite glycocalyx, distal cytoplasm and close inner structures. Furthermore, parasite antigens (GST-1 and AgB) were unevenly localised within tegumental structures, with both being detected in vesicles found within the TBV. Finally, the presence of host (bovine) IgG was also assessed, suggesting a possible endocytic route in protoscoleces. Our data forms the basis for a better understanding of E. ortleppi and E. granulosus s.l. structural biology.
Collapse
Affiliation(s)
- Sebastián Miles
- Área Inmunología, DEPBIO/IQB, Facultad de Química/Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay; Integrative Parasitology, Center for Infectious Diseases, Heidelberg University, Heidelberg, Germany
| | - Javier Magnone
- Área Inmunología, DEPBIO/IQB, Facultad de Química/Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Joaquín García-Luna
- Área Inmunología, DEPBIO/IQB, Facultad de Química/Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - María Eugenia Ancarola
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina; Departamento de Microbiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marcela Cucher
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina; Departamento de Microbiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sylvia Dematteis
- Área Inmunología, DEPBIO/IQB, Facultad de Química/Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University, Heidelberg, Germany; German Center for Infection Research (DZIF), partner site Heidelberg, Heidelberg, Germany
| | - Marek Cyrklaff
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University, Heidelberg, Germany.
| | - Gustavo Mourglia-Ettlin
- Área Inmunología, DEPBIO/IQB, Facultad de Química/Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
30
|
Mu Y, McManus DP, Gordon CA, Cai P. Parasitic Helminth-Derived microRNAs and Extracellular Vesicle Cargos as Biomarkers for Helminthic Infections. Front Cell Infect Microbiol 2021; 11:708952. [PMID: 34249784 PMCID: PMC8267863 DOI: 10.3389/fcimb.2021.708952] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022] Open
Abstract
As an adaption to their complex lifecycles, helminth parasites garner a unique repertoire of genes at different developmental stages with subtle regulatory mechanisms. These parasitic worms release differential components such as microRNAs (miRNAs) and extracellular vesicles (EVs) as mediators which participate in the host-parasite interaction, immune regulation/evasion, and in governing processes associated with host infection. MiRNAs are small (~ 22-nucleotides) non-coding RNAs that regulate gene expression at the post-transcriptional level, and can exist in stable form in bodily fluids such as serum/plasma, urine, saliva and bile. In addition to reports focusing on the identification of miRNAs or in the probing of differentially expressed miRNA profiles in different development stages/sexes or in specific tissues, a number of studies have focused on the detection of helminth-derived miRNAs in the mammalian host circulatory system as diagnostic biomarkers. Extracellular vesicles (EVs), small membrane-surrounded structures secreted by a wide variety of cell types, contain rich cargos that are important in cell-cell communication. EVs have attracted wide attention due to their unique functional relevance in host-parasite interactions and for their potential value in translational applications such as biomarker discovery. In the current review, we discuss the status and potential of helminth parasite-derived circulating miRNAs and EV cargos as novel diagnostic tools.
Collapse
Affiliation(s)
- Yi Mu
- Molecular Parasitology Laboratory, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Donald P McManus
- Molecular Parasitology Laboratory, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Catherine A Gordon
- Molecular Parasitology Laboratory, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Pengfei Cai
- Molecular Parasitology Laboratory, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
31
|
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: 13] [Impact Index Per Article: 3.3] [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
|
32
|
Sánchez-López CM, Trelis M, Bernal D, Marcilla A. Overview of the interaction of helminth extracellular vesicles with the host and their potential functions and biological applications. Mol Immunol 2021; 134:228-235. [PMID: 33836351 DOI: 10.1016/j.molimm.2021.03.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Helminth Extracellular Vesicles (EVs) have emerged as important mediators in host-parasite communications, participating in the parasite survival and its pathogenic effects. In the last decade, a growing amount of information reporting the isolation and characterization of EVs from different helminth species has appeared, but unfortunately, few reports have focused on functional studies of helminth EVs in different cell lines, organoids or animal models. We here review these in vitro and in vivo studies, which clearly demonstrate that helminths secrete EVs, which affect their environment. Helminth EVs are actively internalized by different cell lines, modulating cellular functions important for host-parasite communication. We discuss how these lines of investigation should provide potential new biomarkers of infection, and since helminth EVs can modulate the host immune response, we also discuss how they can provide a new landscape for the development of new vaccine tools against helminthiases as well as immunotherapy.
Collapse
Affiliation(s)
- Christian M Sánchez-López
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Av. V.A. Estellés, s/n, 46100, Burjassot, Valencia, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe-Universitat de València, 46026 Valencia, Spain
| | - María Trelis
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Av. V.A. Estellés, s/n, 46100, Burjassot, Valencia, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe-Universitat de València, 46026 Valencia, Spain
| | - Dolores Bernal
- Departament de Bioquimica i Biologia Molecular, Facultat de Ciencies Biològiques, Universitat de València, C/ Dr. Moliner, 50, 46100 Burjassot, Valencia, Spain
| | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Av. V.A. Estellés, s/n, 46100, Burjassot, Valencia, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe-Universitat de València, 46026 Valencia, Spain.
| |
Collapse
|
33
|
Song H, He X, Du X, Hua R, Xu J, He R, Xie Y, Gu X, Peng X, Yang G. Molecular characterization and expression analysis of annexin B3 and B38 as secretory proteins in Echinococcus granulosus. Parasit Vectors 2021; 14:103. [PMID: 33557917 PMCID: PMC7869467 DOI: 10.1186/s13071-021-04596-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/13/2021] [Indexed: 11/18/2022] Open
Abstract
Background Cystic echinococcosis is a parasitic zoonotic disease, which poses a threat to public health and animal husbandry, and causes significant economic losses. Annexins are a family of phospholipid-binding proteins with calcium ion-binding activity, which have many functions. Methods Two annexin protein family genes [Echinococcus granulosus annexin B3 (EgAnxB3) and EgAnxB38] were cloned and molecularly characterized using bioinformatic analysis. The immunoreactivity of recombinant EgAnxB3 (rEgAnxB3) and rEgAnxB38 was investigated using western blotting. The distribution of EgAnxB3 and EgAnxB38 in protoscoleces (PSCs), the germinal layer, 18-day strobilated worms and 45-day adult worms was analyzed by immunofluorescence localization, and their secretory characteristics were analyzed preliminarily; in addition, quantitative real-time reverse transcription polymerase chain reaction was used to analyze their transcript levels in PSCs and 28-day strobilated worms stages. The phospholipid-binding activities of rEgAnxB3 and rEgAnxB38 were also analyzed. Results EgAnxB3 and EgAnxB38 are conserved and contain calcium-binding sites. Both rEgAnxB3 and rEgAnxB38 could be specifically recognized by the serum samples from E. granulosus-infected sheep, indicating that they had strong immunoreactivity. EgAnxB3 and EgAnxB38 were distributed in all stages of E. granulosus, and their transcript levels were high in the 28-day strobilated worms. They were found in liver tissues near the cysts. In addition, rEgAnxB3 has Ca2+-dependent phospholipid-binding properties. Conclusions EgAnxB3 and EgAnxB38 contain calcium-binding sites, and rEgAnxB3 has Ca2+-dependent phospholipid-binding properties. EgAnxB3 and EgAnxB38 were transcribed in PSCs and 28-day strobilated worms. They were expressed in all stages of E. granulosus, and distributed in the liver tissues near the hydatid cyst, indicating that they are secreted proteins that play a crucial role in the development of E. granulosus. ![]()
Collapse
Affiliation(s)
- Hongyu Song
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, 611130, China
| | - Xue He
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, 611130, China
| | - Xiaodi Du
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, 611130, China
| | - Ruiqi Hua
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, 611130, China
| | - Jing Xu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, 611130, China
| | - Ran He
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, 611130, China
| | - Yue Xie
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, 611130, China
| | - Xiaobin Gu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, 611130, China
| | - Xuerong Peng
- Department of Chemistry, College of Life and Basic Science, Sichuan Agricultural University, Wenjiang, 611130, China
| | - Guangyou Yang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, 611130, China.
| |
Collapse
|
34
|
Ancarola ME, Lichtenstein G, Herbig J, Holroyd N, Mariconti M, Brunetti E, Berriman M, Albrecht K, Marcilla A, Rosenzvit MC, Kamenetzky L, Brehm K, Cucher M. Extracellular non-coding RNA signatures of the metacestode stage of Echinococcus multilocularis. PLoS Negl Trop Dis 2020; 14:e0008890. [PMID: 33253209 PMCID: PMC7728270 DOI: 10.1371/journal.pntd.0008890] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/10/2020] [Accepted: 10/14/2020] [Indexed: 12/18/2022] Open
Abstract
Extracellular RNAs (ex-RNAs) are secreted by cells through different means that may involve association with proteins, lipoproteins or extracellular vesicles (EV). In the context of parasitism, ex-RNAs represent new and exciting communication intermediaries with promising potential as novel biomarkers. In the last years, it was shown that helminth parasites secrete ex-RNAs, however, most work mainly focused on RNA secretion mediated by EV. Ex-RNA study is of special interest in those helminth infections that still lack biomarkers for early and/or follow-up diagnosis, such as echinococcosis, a neglected zoonotic disease caused by cestodes of the genus Echinococcus. In this work, we have characterised the ex-RNA profile secreted by in vitro grown metacestodes of Echinococcus multilocularis, the casuative agent of alveolar echinococcosis. We have used high throughput RNA-sequencing together with RT-qPCR to characterise the ex-RNA profile secreted towards the extra- and intra-parasite milieus in EV-enriched and EV-depleted fractions. We show that a polarized secretion of small RNAs takes place, with microRNAs mainly secreted to the extra-parasite milieu and rRNA- and tRNA-derived sequences mostly secreted to the intra-parasite milieu. In addition, we show by nanoparticle tracking analyses that viable metacestodes secrete EV mainly into the metacestode inner vesicular fluid (MVF); however, the number of nanoparticles in culture medium and MVF increases > 10-fold when metacestodes show signs of tegument impairment. Interestingly, we confirm the presence of host miRNAs in the intra-parasite milieu, implying their internalization and transport through the tegument towards the MVF. Finally, our assessment of the detection of Echinococcus miRNAs in patient samples by RT-qPCR yielded negative results suggesting the tested miRNAs may not be good biomarkers for this disease. A comprehensive study of the secretion mechanisms throughout the life cycle of these parasites will help to understand parasite interaction with the host and also, improve current diagnostic tools. Extracellular RNAs (ex-RNAs) are secreted by cells through association with proteins or extracellular vesicles (EV). In the context of parasitism, ex-RNAs represent novel communication intermediaries with promising potential as biomarkers. In order to better understand the role ex-RNAs may play in the context of the zoonotic disease echinococcosis, we have characterised the RNA profile secreted by the larval stage (metacestode) of Echinococcus multilocularis. By analysing the products secreted towards the extra- and intra-parasite milieus, we demonstrate that the metacestode displays a polarized secretion of different classes of small non-coding RNAs (sRNAs). In addition, we show that EV secretion occurs mainly towards the inner fluid of the metacestodes. Interestingly, we confirm the presence of host sRNAs in the intra-parasite milieu, implying their internalization and transport through the tegument. Finally, the detection of Echinococcus miRNAs in patient samples yielded negative results suggesting the tested miRNAs may not be good biomarkers for this disease. In summary, our results provide a detailed description of the ex-RNA landscape of the E. multilocularis metacestode together with information on the distribution of the detected RNA classes in different extracellular compartments. This information is of importance to better understand host-parasite interaction and also, to improve current diagnostic tools.
Collapse
Affiliation(s)
- María Eugenia Ancarola
- Departament of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Gabriel Lichtenstein
- Departament of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Johannes Herbig
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Würzburg, Germany
| | - Nancy Holroyd
- Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Mara Mariconti
- Unit of Infectious and Tropical Diseases, San Matteo Hospital Foundation, Pavia, Italy
| | - Enrico Brunetti
- Unit of Infectious and Tropical Diseases, San Matteo Hospital Foundation, Pavia, Italy.,Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Krystyna Albrecht
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Würzburg, Germany
| | - Antonio Marcilla
- Departament de Farmàcia i Tecnologia Farmacéutica i Parasitologia, Universitat de València, València, Spain.,Joint Unit on Endocrinology, Nutrition and Clinical Dietetics, Instituto de Investigación Sanitaria-La Fe Valencia, València, Spain
| | - Mara Cecilia Rosenzvit
- Departament of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Laura Kamenetzky
- Departament of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Klaus Brehm
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Marcela Cucher
- Departament of Microbiology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Research on Microbiology and Medical Parasitology (IMPaM, UBA-CONICET), University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
35
|
Proteomic analysis of plasma exosomes from Cystic Echinococcosis patients provides in vivo support for distinct immune response profiles in active vs inactive infection and suggests potential biomarkers. PLoS Negl Trop Dis 2020; 14:e0008586. [PMID: 33017416 PMCID: PMC7535053 DOI: 10.1371/journal.pntd.0008586] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 07/10/2020] [Indexed: 02/07/2023] Open
Abstract
The reference diagnostic method of human abdominal Cystic Echinococcosis (CE) is imaging, particularly ultrasound, supported by serology when imaging is inconclusive. However, current diagnostic tools are neither optimal nor widely available. The availability of a test detecting circulating biomarkers would considerably improve CE diagnosis and cyst staging (active vs inactive), as well as treatments and follow-up of patients. Exosomes are extracellular vesicles involved in intercellular communication, including immune system responses, and are a recognized source of biomarkers. With the aim of identifying potential biomarkers, plasma pools from patients infected by active or inactive CE, as well as from control subjects, were processed to isolate exosomes for proteomic label-free quantitative analysis. Results were statistically processed and subjected to bioinformatics analysis to define distinct features associated with parasite viability. First, a few parasite proteins were identified that were specifically associated with either active or inactive CE, which represent potential biomarkers to be validated in further studies. Second, numerous identified proteins of human origin were common to active and inactive CE, confirming an overlap of several immune response pathways. However, a subset of human proteins specific to either active or inactive CE, and central in the respective protein-protein interaction networks, were identified. These include the Src family kinases Src and Lyn, and the immune-suppressive cytokine TGF-β in active CE, and Cdc42 in inactive CE. The Src and Lyn Kinases were confirmed as potential markers of active CE in totally independent plasma pools. In addition, insights were obtained on immune response profiles: largely consistent with previous evidence, our observations hint to a Th1/Th2/regulatory immune environment in patients with active CE and a Th1/inflammatory environment with a component of the wound healing response in the presence of inactive CE. Of note, our results were obtained for the first time from the analysis of samples obtained in vivo from a well-characterized, large cohort of human subjects.
Collapse
|
36
|
Stryiński R, Łopieńska-Biernat E, Carrera M. Proteomic Insights into the Biology of the Most Important Foodborne Parasites in Europe. Foods 2020; 9:E1403. [PMID: 33022912 PMCID: PMC7601233 DOI: 10.3390/foods9101403] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
Foodborne parasitoses compared with bacterial and viral-caused diseases seem to be neglected, and their unrecognition is a serious issue. Parasitic diseases transmitted by food are currently becoming more common. Constantly changing eating habits, new culinary trends, and easier access to food make foodborne parasites' transmission effortless, and the increase in the diagnosis of foodborne parasitic diseases in noted worldwide. This work presents the applications of numerous proteomic methods into the studies on foodborne parasites and their possible use in targeted diagnostics. Potential directions for the future are also provided.
Collapse
Affiliation(s)
- Robert Stryiński
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Elżbieta Łopieńska-Biernat
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Mónica Carrera
- Department of Food Technology, Marine Research Institute (IIM), Spanish National Research Council (CSIC), 36-208 Vigo, Spain
| |
Collapse
|
37
|
Sotillo J, Robinson MW, Kimber MJ, Cucher M, Ancarola ME, Nejsum P, Marcilla A, Eichenberger RM, Tritten L. The protein and microRNA cargo of extracellular vesicles from parasitic helminths - current status and research priorities. Int J Parasitol 2020; 50:635-645. [PMID: 32652128 DOI: 10.1016/j.ijpara.2020.04.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/16/2022]
Abstract
Helminth parasites have a remarkable ability to persist within their mammalian hosts, which is largely due to their secretion of molecules with immunomodulatory properties. Although the soluble components of helminth secretions have been extensively studied, the discovery that helminths release extracellular vesicles (EVs) has added further complexity to the host-parasite interaction. Whilst several studies have begun to characterise the molecules carried by helminth EVs, work aimed at investigating their biological functions has been hindered by a lack of helminth-specific EV markers. To begin to address this, we summarised helminth EV literature to date. With a focus on the protein and microRNA (miRNA) cargo, we aimed to detect similarities and differences across those major groups of helminths for which data are available; namely nematodes, trematodes and cestodes. Pfam analysis revealed that although there is no universal EV marker for all helminth species, the EF-hand protein family was present in all EV datasets from cestodes and trematodes, and could serve as a platyhelminth EV biomarker. In contrast, M13 metallopeptidases and actin may have potential as markers for nematode EVs. As with proteins, many miRNA families appeared to be species-, stage-, or dataset-specific. Two miRNA families were common to nematode EVs (mir-10 and let-7); the miRNA cargo of EVs secreted by clade I species appeared somewhat different from species from other clades. Five miRNA families (mir-71, mir-10, mir-190, let-7 and mir-2) were shared by all trematode species examined. Our analysis has identified novel markers that may be used in studies aimed at characterising helminth EVs and interrogating their function at the host-parasite interface. In addition, we discuss the heterogeneity of methods used for helminth EV isolation and emphasise the need for a standardised approach in reporting on helminth EV data.
Collapse
Affiliation(s)
- Javier Sotillo
- Centro Nacional de Microbiologia, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain.
| | - Mark W Robinson
- School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, Northern Ireland, United Kingdom
| | - Michael J Kimber
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Marcela Cucher
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Paraguay 2155 Piso 13 (CP1121), Buenos Aires, Argentina
| | - María Eugenia Ancarola
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM, UBA-CONICET), Paraguay 2155 Piso 13 (CP1121), Buenos Aires, Argentina
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 45, 8200 Aarhus, Denmark
| | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Valencia, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, Universitat de Valencia, Valencia, Spain
| | - Ramon M Eichenberger
- Institute of Parasitology, University of Zurich, Winterthurerstrasse 266a, CH-8057 Zurich, Switzerland.
| | - Lucienne Tritten
- Institute of Parasitology, University of Zurich, Winterthurerstrasse 266a, CH-8057 Zurich, Switzerland.
| |
Collapse
|