1
|
Hamidi F, Taghipour N. miRNA, New Perspective to World of Intestinal Protozoa and Toxoplasma. Acta Parasitol 2024; 69:1690-1703. [PMID: 39158784 DOI: 10.1007/s11686-024-00888-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND miRNAs are known as non-coding RNAs that can regulate gene expression. They are reported in many microorganisms and their host cells. Parasite infection can change or shift host miRNAs expression, which can aim at both parasite eradication and infection. PURPOSE This study dealt with examination of miRNA expressed in intestinal protozoan, coccidia , as well as profile changes in host cell miRNA after parasitic infection and their role in protozoan clearance/ survival. METHODS The authors searched ISI Web of Sciences, Pubmed, Scholar, Scopus, another databases and articles published up to 2024 were included. The keywords of miRNA, intestinal protozoa, toxoplasma and some words associated with topics were used in this search. RESULTS Transfection of miRNA mimics or inhibitors can control parasitic diseases, and be introduced as a new therapeutic option in parasitology. CONCLUSION This review can be used to provide up-to date knowledge for future research on these issues.
Collapse
Affiliation(s)
- Faezeh Hamidi
- Department of Laboratory Sciences and Microbiology, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Niloofar Taghipour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
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
|
3
|
Zhu Y, Li M, Li Z, Song J, Zhao W. Study on the mechanism of miRNAs on liver injury in the condition of Protoscocephalus alveolarus transhepatic portal vein infection. Immun Inflamm Dis 2024; 12:e1236. [PMID: 38652009 PMCID: PMC11037255 DOI: 10.1002/iid3.1236] [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: 10/17/2023] [Revised: 02/29/2024] [Accepted: 03/17/2024] [Indexed: 04/25/2024] Open
Abstract
OBJECTIVE To explore the role of miRNA in liver damage caused by Echinococcus multilocularis infection. METHODS Six female C57BL mice were randomly divided into two groups, the control group and the infection group. Mice in the control group were injected with 100 μL PBS through the hepatic portal vein, and mice in the infection group were infected with E. multilocularis via the hepatic portal vein to establish a mouse model of infection. Small RNA sequencing was performed for detecting the expression of miRNAs in the liver of mice infected with 2000 E. multilocularis after 3 months of infection, screen out miRNAs related to liver damage, and verify by RT-PCR. RESULTS Seventy-one differentially expressed miRNAs were found in the liver in comparison with control, and a total of 36 mouse miRNAs with |FC| >0.585 were screened out, respectively. In addition, Targetscan (V5.0) and miRanda (v3.3a) software were used to predict differential miRNAs target genes and functional enrichment of target genes. Functional annotation showed that "cytokine-cytokine interaction," "positive regulation of cytokine production," "inflammatory response," and "leukocyte activation" were enriched in the liver of E. multilocularis-infected mice. Moreover, the pathways "human cytomegalovirus infection," "cysteine and methionine metabolism," "Notch signaling pathway," and "ferroptosis" were involved in liver disease. Furthermore, four miRNAs (mmu-miR-30e-3p, mmu-miR-203-3p, mmu-miR-125b-5p, and mmu-miR-30c-2-3p) related to liver injury were screened and verified. CONCLUSION This study revealed that the expression profiling of miRNAs in the livers was changed after E. multilocularis infection, and improved our understanding of the transcriptomic landscape of hepatic echinococcosis in mice.
Collapse
Affiliation(s)
- Yazhou Zhu
- Department of Pathogen Biology, School of Basic MedicineNingxia Medical UniversityYinchuanChina
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesYinchuanChina
| | - Ming Li
- General Hospital of Ningxia Medical UniversityYinchuanChina
| | - Zihua Li
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesYinchuanChina
- Department of Cell Biology and GeneticsNingxia Medical UniversityYinchuanChina
| | - Jiahui Song
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesYinchuanChina
| | - Wei Zhao
- Department of Pathogen Biology, School of Basic MedicineNingxia Medical UniversityYinchuanChina
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesYinchuanChina
| |
Collapse
|
4
|
Wang SS, Wang X, He JJ, Zheng WB, Zhu XQ, Elsheikha HM, Zhou CX. Expression profiles of host miRNAs and circRNAs and ceRNA network during Toxoplasma gondii lytic cycle. Parasitol Res 2024; 123:145. [PMID: 38418741 PMCID: PMC10902104 DOI: 10.1007/s00436-024-08152-x] [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: 11/07/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
Toxoplasma gondii is an opportunistic protozoan parasite that is highly prevalent in the human population and can lead to adverse health consequences in immunocompromised patients and pregnant women. Noncoding RNAs, such as microRNAs (miRNAs) and circular RNAs (circRNAs), play important regulatory roles in the pathogenesis of many infections. However, the differentially expressed (DE) miRNAs and circRNAs implicated in the host cell response during the lytic cycle of T. gondii are unknown. In this study, we profiled the expression of miRNAs and circRNAs in human foreskin fibroblasts (HFFs) at different time points after T. gondii infection using RNA sequencing (RNA-seq). We identified a total of 7, 7, 27, 45, 70, 148, 203, and 217 DEmiRNAs and 276, 355, 782, 1863, 1738, 6336, 1229, and 1680 DEcircRNAs at 1.5, 3, 6, 9, 12, 24, 36, and 48 h post infection (hpi), respectively. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses revealed that the DE transcripts were enriched in immune response, apoptosis, signal transduction, and metabolism-related pathways. These findings provide new insight into the involvement of miRNAs and circRNAs in the host response to T. gondii infection.
Collapse
Affiliation(s)
- Sha-Sha Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, Gansu Province, China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, Gansu Province, China
| | - Jun-Jun He
- Key Laboratory of Veterinary Public Health of Higher Education of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650500, Yunnan Province, China
| | - Wen-Bin Zheng
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, China
| | - Xing-Quan Zhu
- Key Laboratory of Veterinary Public Health of Higher Education of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650500, Yunnan Province, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK.
| | - Chun-Xue Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250100, Shandong Province, China.
| |
Collapse
|
5
|
Su D, Zhu S, Hou Z, Hao F, Xu K, Xu F, Zhu Y, Liu D, Xu J, Tao J. Toxoplasma gondii infection regulates apoptosis of host cells via miR-185/ARAF axis. Parasit Vectors 2023; 16:371. [PMID: 37858158 PMCID: PMC10585723 DOI: 10.1186/s13071-023-05991-y] [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: 07/12/2023] [Accepted: 09/29/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Toxoplasmosis is a zoonosis with a worldwide presence that is caused by the intracellular parasite Toxoplasma gondii. Active regulation of apoptosis is an important immune mechanism by which host cells resist the growth of T. gondii or avoid excessive pathological damage induced by this parasite. Previous studies found that upregulated expression of microRNA-185 (miR-185) during T. gondii infection has a potential role in regulating the expression of the ARAF gene, which is reported to be associated with cell proliferation and apoptosis. METHODS The expression levels of miR-185 and the ARAF gene were evaluated by qPCR and Western blot, respectively, in mice tissues, porcine kidney epithelial cells (PK-15) and porcine alveolar macrophages (3D4/21) following infection with the T. gondii ToxoDB#9 and RH strains. The dual luciferase reporter assay was then used to verify the relationship between miR-185 and ARAF targets in PK-15 cells. PK-15 and 3D4/21 cell lines with stable knockout of the ARAF gene were established by CRISPR, and then the apoptosis rates of the cells following T. gondii infection were detected using cell flow cytometry assays. Simultaneously, the activities of cleaved caspase-3, as a key apoptosis executive protein, were detected by Western blot to evaluate the apoptosis levels of cells. RESULTS Infection with both the T. gondii ToxoDB#9 and RH strains induced an increased expression of miR-185 and a decreased expression of ARAF in mice tissues, PK-15 and 3D4/21 cells. MiR-185 mimic transfections showed a significantly negative correlation in expression levels between miR-185 and the ARAF gene. The dual luciferase reporter assay confirmed that ARAF was a target of miR-185. Functional investigation revealed that T. gondii infection induced the apoptosis of PK-15 and 3D4/21 cells, which could be inhibited by ARAF knockout or overexpression of miR-185. The expression levels of cleaved caspase-3 protein were significantly lower in cells with ARAF knockout than in normal cells, which were consistent with the results of the cell flow cytometry assays. CONCLUSIONS Toxoplasma gondii infection could lead to the upregulation of miR-185 and the downregulation of ARAF, which was not related to the strain of T. gondii and the host cells. Toxoplasma gondii infection could regulate the apoptosis of host cells via the miR-185/ARAF axis, which represents an additional strategy used by T. gondii to counteract host-cell apoptosis in order to maintain survival and reproduce in the host cells.
Collapse
Affiliation(s)
- Dingzeyang Su
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009 Jiangsu People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009 People’s Republic of China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Shifan Zhu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009 Jiangsu People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009 People’s Republic of China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009 Jiangsu People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009 People’s Republic of China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Fuxing Hao
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, 225300 People’s Republic of China
| | - Kangzhi Xu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009 Jiangsu People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009 People’s Republic of China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Fan Xu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009 Jiangsu People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009 People’s Republic of China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Yuyang Zhu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009 Jiangsu People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009 People’s Republic of China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009 Jiangsu People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009 People’s Republic of China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009 Jiangsu People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009 People’s Republic of China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009 Jiangsu People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009 People’s Republic of China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009 People’s Republic of China
| |
Collapse
|
6
|
Doghish AS, Ali MA, Elrebehy MA, Mohamed HH, Mansour R, Ghanem A, Hassan A, Elballal MS, Elazazy O, Elesawy AE, Abdel Mageed SS, Nassar YA, Mohammed OA, Abulsoud AI. The interplay between toxoplasmosis and host miRNAs: Mechanisms and consequences. Pathol Res Pract 2023; 250:154790. [PMID: 37683390 DOI: 10.1016/j.prp.2023.154790] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023]
Abstract
Toxoplasmosis is one of the highly prevalent zoonotic diseases worldwide caused by the parasite Toxoplasma gondii (T. gondii). The infection with T. gondii could pass unidentified in immunocompetent individuals; however, latent cysts remain dormant in their digestive tract, but they could be shed and excreted with feces infesting the environment. However, active toxoplasmosis can create serious consequences, particularly in newborns and infected persons with compromised immunity. These complications include ocular toxoplasmosis, in which most cases cannot be treated. Additionally, it caused many stillbirths and miscarriages. Circulating miRNAs are important regulatory molecules ensuring that the normal physiological role of various organs is harmonious. Upon infection with T. gondii, the tightly regulated miRNA profile is disrupted to favor the parasite's survival and further participate in the disease pathogenesis. Interestingly, this dysregulated profile could be useful in acute and chronic disease discrimination and in providing insights into the pathomechanisms of the disease. Thus, this review sheds light on the various roles of miRNAs in signaling pathways regulation involved in the pathogenesis of T. gondii and provides insights into the application of miRNAs clinically for its diagnosis and prognosis.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt.
| | - Mohamed A Ali
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| | - Hend H Mohamed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Reda Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt; Biology Department, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Aml Ghanem
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed Hassan
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Yara A Nassar
- Biology Department, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Osama A Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| |
Collapse
|
7
|
Hamidi F, Mohammadi-Yeganeh S, Haji Molla Hoseini M, Tabaei SJS, Taghipour N, Koochaki A, Hosseini V, Haghighi A. TGF-β Targeted by miR-27a Modulates Anti-Parasite Responses of Immune System. IRANIAN JOURNAL OF PARASITOLOGY 2023; 18:390-399. [PMID: 37886255 PMCID: PMC10597889 DOI: 10.18502/ijpa.v18i3.13762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/19/2023] [Indexed: 10/28/2023]
Abstract
Background Immune cells and their secreted cytokines are known as the first barrier against pathogens. Leishmania major as an intracellular protozoan produces anti-inflammatory cytokines that lead to proliferation and survival of the parasite in the macrophages. miRNAs are small non-coding RNA molecules that regulate mRNAs expression. We aimed to investigate the relationship between the expression of TGF-β and a bioinformatically candidate miRNA, in leishmaniasis as a model of TGF-β overexpression. Methods The miRNAs that target TGF-β -3'UTR were predicted and scored by bioinformatic tools. After cloning of TGF-β-3'UTR in psi-CHECK ™- 2 vector, targeting validation was confirmed using Luciferase assay. After miRNA mimic transfection, the expression of miR-27a, TGF-β, as well as Nitric Oxide concentration was evaluated. Results miR-27a received the highest score for targeting TGF-β in bioinformatic predictions. Luciferase assay confirmed that miR-27a is targeting TGF-β-3'UTR, since miR-27a transfection decreased the luciferase activity. After miRNA transfection, TGF-β expression and Nitric Oxide concentration were declined in L. major infected macrophages. Conclusion Bioinformatic prediction, luciferase assay, and miRNA transfection results showed that miR-27a targets TGF-β. Since miRNA and cytokine-base therapies are developing in infectious diseases, finding and validating miRNAs targeting regulatory cytokines can be a novel strategy for controlling and treating leishmaniasis.
Collapse
Affiliation(s)
- Faezeh Hamidi
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Laboratory Sciences and Microbiology, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Samira Mohammadi-Yeganeh
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Haji Molla Hoseini
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyyed Javad Seyyed Tabaei
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloofar Taghipour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ameneh Koochaki
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahedeh Hosseini
- Department of Molecular Medicine and Genetics, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ali Haghighi
- Department of Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Brandão YDO, Molento MB. A Systematic Review of Apicomplexa Looking into Epigenetic Pathways and the Opportunity for Novel Therapies. Pathogens 2023; 12:pathogens12020299. [PMID: 36839571 PMCID: PMC9963874 DOI: 10.3390/pathogens12020299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Interest in host epigenetic changes during apicomplexan infections increased in the last decade, mainly due to the emergence of new therapies directed to these alterations. This review aims to carry out a bibliometric analysis of the publications related to host epigenetic changes during apicomplexan infections and to summarize the main studied pathways in this context, pointing out those that represent putative drug targets. We used four databases for the article search. After screening, 116 studies were included. The bibliometric analysis revealed that the USA and China had the highest number of relevant publications. The evaluation of the selected studies revealed that Toxoplasma gondii was considered in most of the studies, non-coding RNA was the most frequently reported epigenetic event, and host defense was the most explored pathway. These findings were reinforced by an analysis of the co-occurrence of keywords. Even though we present putative targets for repurposing epidrugs and ncRNA-based drugs in apicomplexan infections, we understand that more detailed knowledge of the hosts' epigenetic pathways is still needed before establishing a definitive drug target.
Collapse
|
9
|
Chen JM, Zhao SS, Tao DL, Li JY, Yang X, Fan YY, Song JK, Liu Q, Zhao GH. Temporal transcriptomic changes in microRNAs involved in the host immune response and metabolism during Neospora caninum infection. Parasit Vectors 2023; 16:28. [PMID: 36694228 PMCID: PMC9872418 DOI: 10.1186/s13071-023-05665-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Neospora caninum infection is a major cause of abortion in cattle, which results in serious economic losses to the cattle industry. However, there are no effective drugs or vaccines for the control of N. caninum infections. There is increasing evidence that microRNAs (miRNAs) are involved in many physiological and pathological processes, and dysregulated expression of host miRNAs and the biological implications of this have been reported for infections by various protozoan parasites. However, to our knowledge, there is presently no published information on host miRNA expression during N. caninum infection. METHODS The expression profiles of miRNAs were investigated by RNA sequencing (RNA-seq) in caprine endometrial epithelial cells (EECs) infected with N. caninum at 24 h post infection (pi) and 48 hpi, and the functions of differentially expressed (DE) miRNAs were predicted by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. The transcriptome data were validated by using quantitative real-time polymerase chain reaction. One of the upregulated DEmiRNAs, namely chi-miR-146a, was selected to study the effect of DEmiRNAs on the propagation of N. caninum tachyzoites in caprine EECs. RESULTS RNA-seq showed 18 (17 up- and one downregulated) and 79 (54 up- and 25 downregulated) DEmiRNAs at 24 hpi and 48 hpi, respectively. Quantitative real-time polymerase chain reaction analysis of 13 randomly selected DEmiRNAs (10 up- and three downregulated miRNAs) confirmed the validity of the RNA-seq data. A total of 7835 messenger RNAs were predicted to be potential targets for 66 DEmiRNAs, and GO and KEGG enrichment analysis of these predicted targets revealed that DEmiRNAs altered by N. caninum infection may be involved in host immune responses (e.g. Fc gamma R-mediated phagocytosis, Toll-like receptor signaling pathway, tumor necrosis factor signaling pathway, transforming growth factor-β signaling pathway, mitogen-activated protein kinase signaling pathway) and metabolic pathways (e.g. lysine degradation, insulin signaling pathway, AMP-activated protein kinase signaling pathway, Rap1 signaling pathway, calcium signaling pathway). Upregulated chi-miR-146a was found to promote N. caninum propagation in caprine EECs. CONCLUSIONS This is, to our knowledge, the first report on the expression profiles of host miRNAs during infection with N. caninum, and shows that chi-miR-146a may promote N. caninum propagation in host cells. The novel findings of the present study should help to elucidate the interactions between host cells and N. caninum.
Collapse
Affiliation(s)
- Jin-Ming Chen
- grid.144022.10000 0004 1760 4150Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 China
| | - Shan-Shan Zhao
- grid.144022.10000 0004 1760 4150Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 China
| | - De-Liang Tao
- grid.144022.10000 0004 1760 4150Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 China
| | - Jing-Yu Li
- grid.144022.10000 0004 1760 4150Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 China
| | - Xin Yang
- grid.144022.10000 0004 1760 4150Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 China
| | - Ying-Ying Fan
- grid.144022.10000 0004 1760 4150Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 China
| | - Jun-Ke Song
- grid.144022.10000 0004 1760 4150Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 China
| | - Qun Liu
- grid.22935.3f0000 0004 0530 8290National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193 China
| | - Guang-Hui Zhao
- grid.144022.10000 0004 1760 4150Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 China
| |
Collapse
|
10
|
Amelkina O, da Silva AM, Silva AR, Comizzoli P. Feline microRNAome in ovary and testis: Exploration of in-silico miRNA-mRNA networks involved in gonadal function and cellular stress response. Front Genet 2022; 13:1009220. [PMID: 36226169 PMCID: PMC9548565 DOI: 10.3389/fgene.2022.1009220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
The aim of the study was to perform the first in-depth analysis of miRNAs in ovarian and testicular tissues of the domestic cat, a critical biomedical model. Specifically, potential miRNA involvement was explored in gonadal function, testis development, and cellular stress response to preservation protocols. We performed miRNA-sequencing on 20 ovarian and 20 testicular samples from 15 cats, including different ages and tissue treatments. Using fresh tissues (n = 15), we confirmed gonadal expression of 183 miRNA precursors and discovered additional 52 novel feline candidate precursors. We integrated the mRNA data from our previous study on the same age and treatment groups to create in-silico miRNA-mRNA networks and their functional enrichment, which allows comprehensive exploration into possible miRNA functions in cat gonads. Clusters of miRNAs united by shared differentially expressed mRNA targets are potentially involved in testicular development and spermatogenesis. MicroRNAs could play a significant role in ovarian tissue response to stress from microwave-assisted dehydration, with smaller roles in cellular response to vitrification in both ovary and testis. This new list of miRNAs with potential function in cat gonads is a major step towards understanding the gonadal biology, as well as optimizing fertility preservation protocols.
Collapse
Affiliation(s)
- Olga Amelkina
- Smithsonian’s National Zoo and Conservation Biology Institute, Washington, DC, United States
| | - Andreia M. da Silva
- Laboratory of Animal Germplasm Conservation, Federal Rural University of Semi-Arid—UFERSA, Mossoró, Brazil
| | - Alexandre R. Silva
- Laboratory of Animal Germplasm Conservation, Federal Rural University of Semi-Arid—UFERSA, Mossoró, Brazil
| | - Pierre Comizzoli
- Smithsonian’s National Zoo and Conservation Biology Institute, Washington, DC, United States
| |
Collapse
|
11
|
Medina L, Guerrero-Muñoz J, Castillo C, Liempi A, Fernández-Moya A, Araneda S, Ortega Y, Rivas C, Maya JD, Kemmerling U. Differential microRNAs expression during ex vivo infection of canine and ovine placental explants with Trypanosoma cruzi and Toxoplasma gondii. Acta Trop 2022; 235:106651. [PMID: 35964709 DOI: 10.1016/j.actatropica.2022.106651] [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: 05/27/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022]
Abstract
Trypanosoma cruzi and Toxoplasma gondii are two zoonotic parasites that constitute significant human and animal health threats, causing a significant economic burden worldwide. Both parasites can be transmitted congenitally, but transmission rates for T. gondii are high, contrary to what has been observed for T. cruzi. The probability of congenital transmission depends on complex interactions between the pathogen and the host, including the modulation of host cell gene expression by miRNAs. During ex vivo infection of canine and ovine placental explants, we evaluated the expression of 3 miRNAs (miR-30e-3p, miR-3074-5p, and miR-127-3p) previously associated with parasitic and placental diseases and modulated by both parasites. In addition, we identified the possible target genes of the miRNAs by using computational prediction tools and performed GO and KEGG enrichment analyses to identify the biological functions and associated pathologies. The three miRNAs are differentially expressed in the canine and ovine placenta in response to T. cruzi and T. gondii. We conclude that the observed differential expression and associated functions might explain, at least partially, the differences in transmission rates and susceptibility to parasite infection in different species.
Collapse
Affiliation(s)
- Lisvaneth Medina
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Jesús Guerrero-Muñoz
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Christian Castillo
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Núcleo de Investigación Aplicada en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Chile
| | - Ana Liempi
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alejandro Fernández-Moya
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sebastian Araneda
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Patología y Medicina Oral, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Yessica Ortega
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Facultad de Farmacia y Bioanálisis, Universidad de Los Andes, Mérida, Venezuela
| | - Cristian Rivas
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Juan Diego Maya
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ulrike Kemmerling
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
12
|
Hou Z, Zhang H, Xu K, Zhu S, Wang L, Su D, Liu J, Su S, Liu D, Huang S, Xu J, Pan Z, Tao J. Cluster analysis of splenocyte microRNAs in the pig reveals key signal regulators of immunomodulation in the host during acute and chronic Toxoplasma gondii infection. Parasit Vectors 2022; 15:58. [PMID: 35177094 PMCID: PMC8851844 DOI: 10.1186/s13071-022-05164-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is an obligate intracellular protozoan parasite that can cause a geographically widespread zoonosis. Our previous splenocyte microRNA profile analyses of pig infected with T. gondii revealed that the coordination of a large number of miRNAs regulates the host immune response during infection. However, the functions of other miRNAs involved in the immune regulation during T. gondii infection are not yet known. METHODS Clustering analysis was performed by K-means, self-organizing map (SOM), and hierarchical clustering to obtain miRNA groups with the similar expression patterns. Then, the target genes of the miRNA group in each subcluster were further analyzed for functional enrichment by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome pathway to recognize the key signaling molecules and the regulatory signatures of the innate and adaptive immune responses of the host during T. gondii infection. RESULTS A total of 252 miRNAs were successfully divided into 22 subclusters by K-means clustering (designated as K1-K22), 29 subclusters by SOM clustering (designated as SOM1-SOM29), and six subclusters by hierarchical clustering (designated as H1-H6) based on their dynamic expression levels in the different infection stages. A total of 634, 660, and 477 GO terms, 15, 26, and 14 KEGG pathways, and 16, 15, and 7 Reactome pathways were significantly enriched by K-means, SOM, and hierarchical clustering, respectively. Of note, up to 22 miRNAs mainly showing downregulated expression at 50 days post-infection (dpi) were grouped into one subcluster (namely subcluster H3-K17-SOM1) through the three algorithms. Functional analysis revealed that a large group of immunomodulatory signaling molecules were controlled by the different miRNA groups to regulate multiple immune processes, for instance, IL-1-mediated cellular response and Th1/Th2 cell differentiation partly depending on Notch signaling transduction for subclusters K1 and K2, innate immune response involved in neutrophil degranulation and TLR4 cascade signaling for subcluster K15, B cell activation for subclusters SOM17, SOM1, and SOM25, leukocyte migration, and chemokine activity for subcluster SOM9, cytokine-cytokine receptor interaction for subcluster H2, and interleukin production, chemotaxis of immune cells, chemokine signaling pathway, and C-type lectin receptor signaling pathway for subcluster H3-K17-SOM1. CONCLUSIONS Cluster analysis of splenocyte microRNAs in the pig revealed key regulatory properties of subcluster miRNA molecules and important features in the immune regulation induced by acute and chronic T. gondii infection. These results contribute new insight into the identification of physiological immune responses and maintenance of tolerance in pig spleen tissues during T. gondii infection.
Collapse
Affiliation(s)
- Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Hui Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Kangzhi Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Shifan Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Lele Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Dingzeyang Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Jiantao Liu
- YEBIO Bioengineering Co., Ltd. of QINGDAO, Qingdao, 266109, People's Republic of China
| | - Shijie Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Siyang Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Zhiming Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China. .,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China. .,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
13
|
Chadha A, Chadee K. The NF-κB Pathway: Modulation by Entamoeba histolytica and Other Protozoan Parasites. Front Cell Infect Microbiol 2021; 11:748404. [PMID: 34595137 PMCID: PMC8476871 DOI: 10.3389/fcimb.2021.748404] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Protozoan parasites have led to worldwide devastation because of their ability to cause infectious diseases. They have evolved as successful pathogens in part because of their remarkable and sophisticated ways to evade innate host defenses. This holds true for both intracellular and extracellular parasites that deploy multiple strategies to circumvent innate host defenses for their survival. The different strategies protozoan parasites use include hijacking the host cellular signaling pathways and transcription factors. In particular, the nuclear factor-κB (NF-κB) pathway seems to be an attractive target for different pathogens owing to their central role in regulating prompt innate immune responses in host defense. NF-κB is a ubiquitous transcription factor that plays an indispensable role not only in regulating immediate immune responses against invading pathogens but is also a critical regulator of cell proliferation and survival. The major immunomodulatory components include parasite surface and secreted proteins/enzymes and stimulation of host cells intracellular pathways and inflammatory caspases that directly or indirectly interfere with the NF-κB pathway to thwart immune responses that are directed for containment and/or elimination of the pathogen. To showcase how protozoan parasites exploits the NF-κB signaling pathway, this review highlights recent advances from Entamoeba histolytica and other protozoan parasites in contact with host cells that induce outside-in and inside-out signaling to modulate NF-κB in disease pathogenesis and survival in the host.
Collapse
Affiliation(s)
- Attinder Chadha
- Departments of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Kris Chadee
- Departments of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
14
|
Scalon MC, Martins CS, Ferreira GG, Schlemmer F, Titze-de-Almeida R, Paludo GR. RT-rtPCR quantification of circulating microRNAs in plasma and serum samples from healthy domestic cats. J Vet Diagn Invest 2021; 33:1151-1155. [PMID: 34301168 DOI: 10.1177/10406387211034843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at a post-transcriptional level by silencing targeted messenger RNA (mRNA). Most studies concerning miRNA expression use solid tissue samples. However, circulating miRNAs from different body fluids have recently emerged as diagnostic and prognostic molecules, given that they hold informative value and have increased stability in cell-free form. Blood sampling of cats can be challenging given their small body size and because they often experience distress when handled. We quantified miR-20a, -192, -365, -15b-5p, and -16-5p from plasma and serum samples of 10 healthy domestic cats. Our RT-rtPCR procedure used 100 µL of either plasma or serum samples as sources of biomarker molecules. However, serum provided higher amounts of miRNA than plasma samples, with a p < 0.0001 for miR-20a and p < 0.0002 for miR-16-5p.
Collapse
Affiliation(s)
- Marcela C Scalon
- Veterinary Clinical Pathology Laboratory, College of Agronomy and Veterinary Medicine, University of Brasilia, Brasilia, Brazil
| | - Christine S Martins
- Veterinary Clinical Pathology Laboratory, College of Agronomy and Veterinary Medicine, University of Brasilia, Brasilia, Brazil
| | - Gabriel G Ferreira
- Technology for Gene Therapy Laboratory, College of Agronomy and Veterinary Medicine, University of Brasilia, Brasilia, Brazil
| | - Franciele Schlemmer
- Technology for Gene Therapy Laboratory, College of Agronomy and Veterinary Medicine, University of Brasilia, Brasilia, Brazil
| | - Ricardo Titze-de-Almeida
- Technology for Gene Therapy Laboratory, College of Agronomy and Veterinary Medicine, University of Brasilia, Brasilia, Brazil
| | - Giane R Paludo
- Veterinary Clinical Pathology Laboratory, College of Agronomy and Veterinary Medicine, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
15
|
Quiarim TM, Maia MM, da Cruz AB, Taniwaki NN, Namiyama GM, Pereira-Chioccola VL. Characterization of extracellular vesicles isolated from types I, II and III strains of Toxoplasma gondii. Acta Trop 2021; 219:105915. [PMID: 33861971 DOI: 10.1016/j.actatropica.2021.105915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/15/2021] [Accepted: 03/27/2021] [Indexed: 12/31/2022]
Abstract
This study investigated the participation extracellular vesicles (EVs) in Toxoplasma gondii-host interaction. EVs of three T. gondii strains (RH, ME-49 and VEG) were purified by chromatography and ELISA. Results of "nanoparticle tracking analysis" and scanning electron microscopy showed that RH strain released more EVs than other strains. Images of transmission electron microscopy showed that in beginning of incubation (culture medium), EVs were inside of tachyzoites preparing to be released. After 24 hours, they were largely produced inside tachyzoites and were released through plasma membrane. The parasite burden of mice infected with RH strain plus EVs was increased and with early death of 1-2 days compared of those that received only parasites. EV proteins of ME-49 and VEG strains were poorly reactive to sera of infected patients in imunoblot. However, those from RH strain were reactive against sera of patients with cerebral toxoplasmosis. EVs stimulated murine splenocytes caused similar production of IFN-γ and IL-10 levels. RH strain derived EVs stimulated more TNF-α than those stimulated by other strains. T. gondii and infected hosts can express the same miRNAs (miR-155-5p, miR-125b-5p, miR-423-3p). In conclusion, T. gondii derived EVs promote host-parasite interactions, modulate host immune responses, carry virulent factors and cause an imbalance in cellular immune response.
Collapse
Affiliation(s)
- Talita Motta Quiarim
- Laboratório de Biologia Molecular de Parasitas e Fungos, Instituto Adolfo Lutz, Sao Paulo, Brazil.
| | - Marta Marques Maia
- Laboratório de Biologia Molecular de Parasitas e Fungos, Instituto Adolfo Lutz, Sao Paulo, Brazil.
| | - Allecineia Bispo da Cruz
- Laboratório de Biologia Molecular de Parasitas e Fungos, Instituto Adolfo Lutz, Sao Paulo, Brazil.
| | | | | | | |
Collapse
|
16
|
de Faria Junior GM, Murata FHA, Lorenzi HA, Castro BBP, Assoni LCP, Ayo CM, Brandão CC, de Mattos LC. The Role of microRNAs in the Infection by T. gondii in Humans. Front Cell Infect Microbiol 2021; 11:670548. [PMID: 34055667 PMCID: PMC8160463 DOI: 10.3389/fcimb.2021.670548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs are molecules belonging to an evolutionarily conserved family of small non-coding RNAs, which act on post-transcriptional gene regulation, causing messenger RNA (mRNA) degradation or inhibiting mRNA translation into proteins. These molecules represent potential biomarkers for diagnosis, non-invasive prognosis, and monitoring the development of the disease. Moreover, they may provide additional information on the pathophysiology of parasitic infections and guide strategies for treatment. The Apicomplexan parasite Toxoplasma gondii modifies the levels of microRNAs and mRNAs in infected host cells by modulating the innate and adaptive immune responses, facilitating its survival within the host. Some studies have shown that microRNAs are promising molecular markers for developing diagnostic tools for human toxoplasmosis. MicroRNAs can be detected in human specimens collected using non-invasive procedures. changes in the circulating host microRNAs have been associated with T. gondii infection in mice and ocular toxoplasmosis in humans. Besides, microRNAs can be amplified from samples using sensitive and molecular-specific approaches such as real-time PCR. This review presents recent findings of the role that microRNAs play during T. gondii infection and discuss their potential use of these small nuclei acid molecules to different approaches such as laboratory diagnosis, modulation of cell and tissue infected as other potential applications in human toxoplasmosis.
Collapse
Affiliation(s)
- Geraldo Magela de Faria Junior
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Fernando Henrique Antunes Murata
- Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, United States Department of Agriculture, Agricultural Research Service, Beltsville, MD, United States
| | | | - Bruno Bello Pede Castro
- Department of Preventive Veterinary Medicine and Animal Health, Faculty of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| | - Letícia Carolina Paraboli Assoni
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Christiane Maria Ayo
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Cinara Cássia Brandão
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Luiz Carlos de Mattos
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| |
Collapse
|
17
|
Multi-omic analyses in Abyssinian cats with primary renal amyloid deposits. Sci Rep 2021; 11:8339. [PMID: 33863921 PMCID: PMC8052419 DOI: 10.1038/s41598-021-87168-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/19/2021] [Indexed: 02/02/2023] Open
Abstract
The amyloidoses constitute a group of diseases occurring in humans and animals that are characterized by abnormal deposits of aggregated proteins in organs, affecting their structure and function. In the Abyssinian cat breed, a familial form of renal amyloidosis has been described. In this study, multi-omics analyses were applied and integrated to explore some aspects of the unknown pathogenetic processes in cats. Whole-genome sequences of two affected Abyssinians and 195 controls of other breeds (part of the 99 Lives initiative) were screened to prioritize potential disease-associated variants. Proteome and miRNAome from formalin-fixed paraffin-embedded kidney specimens of fully necropsied Abyssinian cats, three affected and three non-amyloidosis-affected were characterized. While the trigger of the disorder remains unclear, overall, (i) 35,960 genomic variants were detected; (ii) 215 and 56 proteins were identified as exclusive or overexpressed in the affected and control kidneys, respectively; (iii) 60 miRNAs were differentially expressed, 20 of which are newly described. With omics data integration, the general conclusions are: (i) the familial amyloid renal form in Abyssinians is not a simple monogenic trait; (ii) amyloid deposition is not triggered by mutated amyloidogenic proteins but is a mix of proteins codified by wild-type genes; (iii) the form is biochemically classifiable as AA amyloidosis.
Collapse
|
18
|
Inhibition of anti-inflammatory cytokines, IL-10 and TGF-β, in Leishmania major infected macrophage by miRNAs: A new therapeutic modality against leishmaniasis. Microb Pathog 2021; 153:104777. [PMID: 33592260 DOI: 10.1016/j.micpath.2021.104777] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/18/2020] [Accepted: 01/28/2021] [Indexed: 01/03/2023]
Abstract
Leishmania major (L. major) applies several mechanisms to escape the immune system. Interleukin-10 (IL-10) and Transforming Growth Factor (TGF-β) downregulate nitric oxide synthase (iNOS) leading to the survival of Leishmania within macrophages. The miRNAs are known as the modulators of the immune system. The present study was conducted to assess the effect of synthetic miR-340 mimic on cytokines (IL-10 and TGF-β1) involved in L. major infected macrophages. The miRNAs targeting of IL-10 and TGF-β1 was predicted using bioinformatic tools. Relative expression of predicted miRNA, IL-10, and TGF-β1 was measured by RT-qPCR before and after synthetic miRNA mimic transfection. Concentration of IL-10 and TGF-β was measured in posttreatment condition using ELISA method. Also, infectivity was assessed by Giemsa staining. mmu-miR-340 received the highest score for targeting cytokines. The expression of miR-340 was downregulated in L. major infected macrophages. By contrast, expression of IL-10 and TGF-β1 was upregulated in infected macrophages. After miRNA transfection, TGF-β1 and IL-10 were both downregulated and interestingly, the combination of miR-340 and miR-27a had a stronger effect on the downregulation of target genes. This research revealed that transfection of infected macrophages with miR-340 alone or in combination with miR-27a mimic can reduce macrophage infectivity and might be introduced as a novel therapeutic agent for cutaneous leishmaniasis.
Collapse
|
19
|
Medina L, Castillo C, Liempi A, Guerrero-Muñoz J, Rojas-Pirela M, Maya JD, Prieto H, Kemmerling U. Trypanosoma cruzi and Toxoplasma gondii Induce a Differential MicroRNA Profile in Human Placental Explants. Front Immunol 2020; 11:595250. [PMID: 33240284 PMCID: PMC7677230 DOI: 10.3389/fimmu.2020.595250] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022] Open
Abstract
Trypanosoma cruzi and Toxoplasma gondii are two parasites than can be transmitted from mother to child through the placenta. However, congenital transmission rates are low for T. cruzi and high for T. gondii. Infection success or failure depends on complex parasite-host interactions in which parasites can alter host gene expression by modulating non-coding RNAs such as miRNAs. As of yet, there are no reports on altered miRNA expression in placental tissue in response to either parasite. Therefore, we infected human placental explants ex vivo by cultivation with either T. cruzi or T. gondii for 2 h. We then analyzed the miRNA expression profiles of both types of infected tissue by miRNA sequencing and quantitative PCR, sequence-based miRNA target prediction, pathway functional enrichment, and upstream regulator analysis of differentially expressed genes targeted by differentially expressed miRNAs. Both parasites induced specific miRNA profiles. GO analysis revealed that the in silico predicted targets of the differentially expressed miRNAs regulated different cellular processes involved in development and immunity, and most of the identified KEGG pathways were related to chronic diseases and infection. Considering that the differentially expressed miRNAs identified here modulated crucial host cellular targets that participate in determining the success of infection, these miRNAs might explain the differing congenital transmission rates between the two parasites. Molecules of the different pathways that are regulated by miRNAs and modulated during infection, as well as the miRNAs themselves, may be potential targets for the therapeutic control of either congenital Chagas disease or toxoplasmosis.
Collapse
Affiliation(s)
- Lisvaneth Medina
- Programa de Anatomía y Biología del Desarrollo, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Christian Castillo
- Programa de Anatomía y Biología del Desarrollo, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Ana Liempi
- Programa de Anatomía y Biología del Desarrollo, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Jesús Guerrero-Muñoz
- Programa de Anatomía y Biología del Desarrollo, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Maura Rojas-Pirela
- Instituto de Biología, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Juan Diego Maya
- Programa de Farmacología Molecular y Clínica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Humberto Prieto
- Instituto de Investigaciones Agropecuarias, Ministerio de Agricultura, Santiago, Chile
| | - Ulrike Kemmerling
- Programa de Anatomía y Biología del Desarrollo, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
20
|
Acetylome analysis of the feline small intestine following Toxoplasma gondii infection. Parasitol Res 2020; 119:3649-3657. [PMID: 32951143 PMCID: PMC7502155 DOI: 10.1007/s00436-020-06880-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/07/2020] [Indexed: 10/26/2022]
Abstract
Toxoplasma gondii is a protozoan parasite capable of infecting a large number of warm-blooded animals and causes serious health complications in immunocompromised patients. T. gondii infection of the feline small intestine is critical for the completion of the life cycle and transmission of T. gondii. Protein acetylation is an important posttranslational modification, which plays roles in the regulation of various cellular processes. Therefore, understanding of how T. gondii reprograms the protein acetylation status of feline definitive host can help to thwart the production and spread of T. gondii. Here, we used affinity enrichment and high-resolution liquid chromatography with tandem mass spectrometry to profile the alterations of the acetylome in cat small intestine 10 days after infection by T. gondii Prugniuad (Pru) strain. Our analysis showed that T. gondii induced significant changes in the acetylation of proteins in the cat intestine. We identified 2606 unique lysine acetylation sites in 1357 acetylated proteins. The levels of 334 acetylated peptides were downregulated, while the levels of 82 acetylated peptides were increased in the infected small intestine. The proteins with differentially acetylated peptides were particularly enriched in the bioenergetics-related processes, such as tricarboxylic acid cycle, oxidative phosphorylation, and oxidation-reduction. These results provide the first baseline of the global acetylome of feline small intestine following T. gondii infection and should facilitate further analysis of the role of acetylated protein in the pathogenesis of T. gondii infection in its definitive host.
Collapse
|
21
|
Abo-Al-Ela HG. Toxoplasmosis and Psychiatric and Neurological Disorders: A Step toward Understanding Parasite Pathogenesis. ACS Chem Neurosci 2020; 11:2393-2406. [PMID: 31268676 DOI: 10.1021/acschemneuro.9b00245] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Toxoplasmosis, a disease that disrupts fetal brain development and severely affects the host's brain, has been linked to many behavioral and neurological disorders. There is growing interest in how a single-celled neurotropic parasite, Toxoplasma gondii, can control or change the behavior of the host as well as how it dominates the host's neurons. Secrets beyond these could be answered by decoding the Toxoplasma gondii genome, unravelling the function of genomic sequences, and exploring epigenetics and mRNAs alterations, as well as the postulated mechanisms contributing to various neurological and psychiatric symptoms caused by this parasite. Substantial efforts have been made to elucidate the action of T. gondii on host immunity and the biology of its infection. However, the available studies on the molecular aspects of toxoplasmosis that affect central nervous system (CNS) circuits remain limited, and much research is still needed on this interesting topic. In my opinion, this parasite is a gift for studying the biology of the nervous system and related diseases. We should utilize the unique features of Toxoplasma, such as its abilities to modulate brain physiology, for neurological studies or as a possible tool or approach to cure neurological disease.
Collapse
Affiliation(s)
- Haitham G. Abo-Al-Ela
- Animal Health Research Institute, Agriculture Research Center, Shibin Al-Kom, El-Minufiya 7001, Egypt
| |
Collapse
|
22
|
Pereira IDS, Maia MM, da Cruz AB, Telles JPM, Vidal JE, Gava R, Meira-Strejevitch CS, Pereira-Chioccola VL. Plasma extracellular microRNAs are related to AIDS/cerebral toxoplasmosis co-infection. Parasite Immunol 2020; 42:e12696. [PMID: 31945196 DOI: 10.1111/pim.12696] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/21/2022]
Abstract
This study investigated the potential of five miRNA candidates for cerebral toxoplasmosis/HIV co-infection (CT/HIV) biomarkers. miR-155-5p, miR-146a-5p, miR-21-5p, miR-125b-5p and miR-29c-3p were tested in 79 plasma divided into groups: 32 CT/HIV patients; 27 individuals with asymptomatic toxoplasmosis (AT); and 20 individuals seronegative for toxoplasmosis (NC). From each was collected peripheral blood/EDTA for laboratory diagnosis. Blood cells for DNA extractions (molecular diagnosis), plasma for RNA extractions (gene expression) and ELISA (serological diagnosis). miRNA expression was performed by qPCR, and values were expressed in Relative Quantification (RQ). Among the five miRNAs, miR-21-5p and miR-146a-5p were up-expressed in CT/HIV group when compared with AT and NC groups. RQ means for miR-21-5p and miR-146a-5p in CT/HIV group were 3.829 and 2.500, while in AT group, were 1.815 and 1.661, respectively. Differences between 3 groups were statistically significant (Kruskal-Wallis ANOVA test), as well as CT/HIV and AT groups (Mann-Whitney test). Plasma of CT/HIV and AT groups expressed similar levels of miR-29c-3p, miR-155-5p and miR-125b-5p. As NC group was different of CT/HIV and AT groups, differences between three groups were statistically significant (Kruskal-Wallis ANOVA test). No difference was shown between CT/HIV and AT groups (Mann-Whitney test). These results suggest the host miRNAs modulation by Toxoplasma gondii.
Collapse
Affiliation(s)
| | - Marta Marques Maia
- Laboratório de Biologia Molecular de Parasitas e Fungos, Instituto Adolfo Lutz, Sao Paulo, Brazil
| | - Allecineia Bispo da Cruz
- Laboratório de Biologia Molecular de Parasitas e Fungos, Instituto Adolfo Lutz, Sao Paulo, Brazil
| | | | - Jose Ernesto Vidal
- Instituto de Infectologia Emilio Ribas, São Paulo, Brazil.,Faculdade de Medicina, Hospital das Clínicas, da Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Investigação Médica (LIM) 49, Instituto de Medicina Tropical da Universidade de São Paulo, São Paulo, Brazil
| | - Ricardo Gava
- Laboratório de Biologia Molecular de Parasitas e Fungos, Instituto Adolfo Lutz, Sao Paulo, Brazil
| | | | | |
Collapse
|
23
|
Acuña SM, Floeter-Winter LM, Muxel SM. MicroRNAs: Biological Regulators in Pathogen-Host Interactions. Cells 2020; 9:E113. [PMID: 31906500 PMCID: PMC7016591 DOI: 10.3390/cells9010113] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
An inflammatory response is essential for combating invading pathogens. Several effector components, as well as immune cell populations, are involved in mounting an immune response, thereby destroying pathogenic organisms such as bacteria, fungi, viruses, and parasites. In the past decade, microRNAs (miRNAs), a group of noncoding small RNAs, have emerged as functionally significant regulatory molecules with the significant capability of fine-tuning biological processes. The important role of miRNAs in inflammation and immune responses is highlighted by studies in which the regulation of miRNAs in the host was shown to be related to infectious diseases and associated with the eradication or susceptibility of the infection. Here, we review the biological aspects of microRNAs, focusing on their roles as regulators of gene expression during pathogen-host interactions and their implications in the immune response against Leishmania, Trypanosoma, Toxoplasma, and Plasmodium infectious diseases.
Collapse
Affiliation(s)
| | | | - Sandra Marcia Muxel
- Department of Physiology, Universidade de São Paulo, 05508-090 São Paulo, Brazil; (S.M.A.); (L.M.F.-W.)
| |
Collapse
|
24
|
Menard KL, Haskins BE, Denkers EY. Impact of Toxoplasma gondii Infection on Host Non-coding RNA Responses. Front Cell Infect Microbiol 2019; 9:132. [PMID: 31157172 PMCID: PMC6530353 DOI: 10.3389/fcimb.2019.00132] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022] Open
Abstract
As an intracellular microbe, Toxoplasma gondii must establish a highly intimate relationship with its host to ensure success as a parasite. Many studies over the last decade-and-a-half have highlighted how the host reshapes its immunoproteome to survive infection, and conversely how the parasite regulates host responses to ensure persistence. The role of host non-protein-coding RNA during infection is a vast and largely unexplored area of emerging interest. The potential importance of this facet of the host-parasite interaction is underscored by current estimates that as much as 80% of the host genome is transcribed into non-translated RNA. Here, we review the current state of knowledge with respect to two major classes of non-coding RNA, microRNA (miRNA) and long non-coding RNA (lncRNA), in the host response to T. gondii infection. These two classes of regulatory RNA are known to have profound and widespread effects on cell function. However, their impact on infection and immunity is not well-understood, particularly for the response to T. gondii. Nevertheless, numerous miRNAs have been identified that are upregulated by Toxoplasma, and emerging evidence suggests a functional role during infection. While the field of lncRNA is in its infancy, it is already clear that Toxoplasma is also a strong trigger for this class of regulatory RNA. Non-coding RNA responses induced by T. gondii are likely to be major determinants of the host's ability to resist infection and the parasite's ability to establish long-term latency.
Collapse
Affiliation(s)
- Kayla L Menard
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| | - Breanne E Haskins
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| | - Eric Y Denkers
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
25
|
Hou Z, Liu D, Su S, Wang L, Zhao Z, Ma Y, Li Q, Jia C, Xu J, Zhou Y, Tao J. Comparison of splenocyte microRNA expression profiles of pigs during acute and chronic toxoplasmosis. BMC Genomics 2019; 20:97. [PMID: 30700253 PMCID: PMC6354428 DOI: 10.1186/s12864-019-5458-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 01/17/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is an obligate intracellular parasite that infects humans and other warm-blooded animals. Previous quantitative proteomic analyses of infected host cells revealed that the expression of many host proteins is modulated by T. gondii infection. However, at present limited data are available on the differentially expressed miRNAs (DEMs) associated with the pathology and host immune responses induced by acute and chronic infection with T. gondii in pigs in vivo. In this study, high-throughput sequencing was used to investigate expression profiles of spleen miRNAs at 10, 25 and 50 days post-infection (DPI) in pigs infected with Chinese I genotype strain T. gondii isolated from a dead pig. RESULTS When compared to the control group, 34, 6 and 86 DEMs were found in spleens of infected pigs at 10, 25 and 50 DPI, respectively. Gene Ontology (GO) enrichment analysis of the target genes of DEMs showed that no GO terms were enriched at 25 DPI, whereas 28 and 241 GO terms, of which two and 215 were sample-specific, were significantly enriched at 10 and 50 DPI, respectively. The top 20 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of the target genes of DEMs included signal transduction, immune system, metabolism and diseases. miRNA-gene network analysis revealed that the DEMs played important roles in the host immune response to T. gondii infection by modulating expression levels of cellular immunity-related cytokines and immune-related C-type lectins. CONCLUSION Our results not only showed that host miRNA expression is altered by T. gondii but also revealed differences in the regulation of key biological processes and pathways involved in host responses to acute versus chronic T. gondii infection. This will aid future research into miRNA-target interactions during T. gondii infection in pigs and the development of novel therapies against T. gondii.
Collapse
Affiliation(s)
- Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Shijie Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Lele Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Zhenxing Zhao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yifei Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Qiaoqiao Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Chuanli Jia
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yonghua Zhou
- Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China. .,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, People's Republic of China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
26
|
Wang M, Zhang FK, Elsheikha HM, Zhang NZ, He JJ, Luo JX, Zhu XQ. Transcriptomic insights into the early host-pathogen interaction of cat intestine with Toxoplasma gondii. Parasit Vectors 2018; 11:592. [PMID: 30428922 PMCID: PMC6236892 DOI: 10.1186/s13071-018-3179-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022] Open
Abstract
Background Although sexual reproduction of the parasite Toxoplasma gondii exclusively occurs in the cat intestine, knowledge about the alteration of gene expression in the intestine of cats infected with T. gondii is still limited. Here, we investigated the temporal transcriptional changes that occur in the cat intestine during T. gondii infection. Methods Cats were infected with 100 T. gondii cysts and their intestines were collected at 6, 12, 18, 24, 72 and 96 hours post-infection (hpi). RNA sequencing (RNA-Seq) Illumina technology was used to gain insight into the spectrum of genes that are differentially expressed due to infection. Quantitative RT-PCR (qRT-PCR) was also used to validate the level of expression of a set of differentially expressed genes (DEGs) obtained by sequencing. Results Our transcriptome analysis revealed 2363 DEGs that were clustered into six unique patterns of gene expression across all the time points after infection. Our analysis revealed 56, 184, 404, 508, 400 and 811 DEGs in infected intestines compared to uninfected controls at 6, 12, 18, 24, 72 and 96 hpi, respectively. RNA-Seq results were confirmed by qRT-PCR. DEGs were mainly enriched in catalytic activity and metabolic process based on gene ontology enrichment analysis. Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that transcriptional changes in the intestine of infected cats evolve over the course of infection, and the largest difference in the enriched pathways was observed at 96 hpi. The anti-T. gondii defense response of the feline host was mediated by Major Histocompatibility Complex class I, proteasomes, heat-shock proteins and fatty acid binding proteins. Conclusions This study revealed novel host factors, which may be critical for the successful establishment of an intracellular niche during T. gondii infection in the definitive feline host. Electronic supplementary material The online version of this article (10.1186/s13071-018-3179-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Meng Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - Fu-Kai Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK
| | - Nian-Zhang Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China.
| | - Jian-Xun Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China. .,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University College of Veterinary Medicine, Yangzhou, Jiangsu Province, 225009, People's Republic of China.
| |
Collapse
|
27
|
Hu RS, He JJ, Elsheikha HM, Zhang FK, Zou Y, Zhao GH, Cong W, Zhu XQ. Differential Brain MicroRNA Expression Profiles After Acute and Chronic Infection of Mice With Toxoplasma gondii Oocysts. Front Microbiol 2018; 9:2316. [PMID: 30333806 PMCID: PMC6176049 DOI: 10.3389/fmicb.2018.02316] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/11/2018] [Indexed: 12/16/2022] Open
Abstract
Brain microRNAs (miRNAs) change in abundance in response to Toxoplasma gondii infection. However, their precise role in the pathogenesis of cerebral infection with T. gondii oocyst remains unclear. We studied the abundance of miRNAs in the brain of mice on days 11 and 33 post-infection (dpi) in order to identify miRNA pattern specific to early (11 dpi) and late (33 dpi) T. gondii infection. Mice were challenged with T. gondii oocysts (Type II strain) and on 11 and 33 dpi, the expression of miRNAs in mouse brain was investigated using small RNA (sRNA) sequencing. miRNA expression was confirmed by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed to identify the biological processes, molecular functions, and cellular components, as well as pathways involved in infection. More than 1,500 miRNAs (1,352 known and 150 novel miRNAs) were detected in the infected and control mice. The expression of miRNAs varied across time after infection; 3, 38, and 108 differentially expressed miRNAs (P < 0.05) were detected during acute infection, chronic infection and chronic vs. acute infection, respectively. GO analysis showed that chronically infected mice had more predicted targets of dysregulated miRNAs than acutely infected mice. KEGG analysis indicated that most predicted targets were involved in immune- or disease-related pathways. Our data indicate that T. gondii infection alters the abundance of miRNAs in mouse brain particularly at the chronic stage, probably to fine-tune conditions required for the establishment of a latent brain infection.
Collapse
Affiliation(s)
- Rui-Si Hu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, The University of Nottingham, Loughborough, United Kingdom
| | - Fu-Kai Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yang Zou
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Guang-Hui Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Wei Cong
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Marine Science, Shandong University at Weihai, Weihai, China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
28
|
Chen XQ, Elsheikha HM, Hu RS, Hu GX, Guo SL, Zhou CX, Zhu XQ. Hepatic Metabolomics Investigation in Acute and Chronic Murine Toxoplasmosis. Front Cell Infect Microbiol 2018; 8:189. [PMID: 29922602 PMCID: PMC5996072 DOI: 10.3389/fcimb.2018.00189] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 05/17/2018] [Indexed: 11/29/2022] Open
Abstract
Toxoplasma gondii poses a great threat to human health, with no approved vaccine available for the treatment of T. gondii infection. T. gondii infections are not limited to the brain, and may also affect other organs especially the liver. Identification of host liver molecules or pathways involved in T. gondii replication process may lead to the discovery of novel anti-T. gondii targets. Here, we analyzed the metabolic profile of the liver of mice on 11 and 30 days postinfection (dpi) with type II T. gondii Pru strain. Global metabolomics using liquid chromatography-tandem mass spectrometry (LC-MS/MS) identified 389 significant metabolites from acutely infected mice; and 368 from chronically infected mice, when compared with control mice. Multivariate statistical analysis revealed distinct metabolic signatures from acutely infected, chronically infected and control mice. Infection influenced several metabolic processes, in particular those for lipids and amino acids. Metabolic pathways, such as steroid hormone biosynthesis, primary bile acid biosynthesis, bile secretion, and biosynthesis of unsaturated fatty acids were perturbed during the whole infection process, particularly during the acute stage of infection. The present results provide insight into hepatic metabolic changes that occur in BALB/c mice during acute and chronic T. gondii infection.
Collapse
Affiliation(s)
- Xiao-Qing Chen
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Rui-Si Hu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Gui-Xue Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Shu-Ling Guo
- Department of Parasitology, Shandong University School of Basic Medicine, Jinan, China
| | - Chun-Xue Zhou
- Department of Parasitology, Shandong University School of Basic Medicine, Jinan, China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|