1
|
Omar M, Abdelal HO. Nitric oxide in parasitic infections: a friend or foe? J Parasit Dis 2022; 46:1147-1163. [PMID: 36457767 PMCID: PMC9606182 DOI: 10.1007/s12639-022-01518-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022] Open
Abstract
The complex interaction between the host and the parasite remains a puzzling question. Control of parasitic infections requires an efficient immune response that must be balanced against destructive pathological consequences. Nitric oxide is a nitrogenous free radical which has many molecular targets and serves diverse functions. Apart from being a signaling messenger, nitric oxide is critical for controlling numerous infections. There is still controversy surrounding the exact role of nitric oxide in the immune response against different parasitic species. It proved protective against intracellular protozoa, as well as extracellular helminths. At the same time, it plays a pivotal role in stimulating detrimental pathological changes in the infected hosts. Several reports have discussed the anti-parasitic and immunoregulatory functions of nitric oxide, which could directly influence the control of the infection. Nevertheless, there is scarce literature addressing the harmful cytotoxic impacts of this mediator. Thus, this review provides insights into the most updated concepts and controversies regarding the dual nature and opposing sides of nitric oxide during the course of different parasitic infections.
Collapse
Affiliation(s)
- Marwa Omar
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Gameyet Almohafza St. 1, Menya Al-Kamh, City of Zagazig, 44511 Sharkia Governorate Egypt
| | - Heba O. Abdelal
- LIS: Cross-National Data Center, Maison des Sciences Humaines - 5e étage, 11- porte des Sciences, L-4366 Esch-Belval, Luxembourg
| |
Collapse
|
2
|
Rahman MM, McFadden G, Ruthel G, Herbert DR, Freedman BD, Greenberg RM, Bais S. Oncolytic Myxoma virus infects and damages the tegument of the human parasitic flatworm Schistosoma mansoni. Exp Parasitol 2022; 239:108263. [PMID: 35598646 PMCID: PMC11003549 DOI: 10.1016/j.exppara.2022.108263] [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: 12/03/2021] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 11/23/2022]
Abstract
Schistosomiasis is a devastating disease caused by parasitic flatworms of the genus Schistosoma. Praziquantel (PZQ), the current treatment of choice, is ineffective against immature worms and cannot prevent reinfection. The continued reliance on a single drug for treatment increases the risk of the development of PZQ-resistant parasites. Reports of PZQ insusceptibility lends urgency to the need for new therapeutics. Here, we report that Myxoma virus (MYXV), an oncolytic pox virus which is non-pathogenic in all mammals except leporids, infects and replicates in S. mansoni schistosomula, juveniles, and adult male and female worms. MYXV infection results in the shredding of the tegument and reduced egg production in vitro, identifying MYXV as the first viral pathogen of schistosomes. MYXV is currently in preclinical studies to manage multiple human cancers, supporting its use in human therapeutics. Our findings raise the exciting possibility that MYXV virus represents a novel and safe class of potential anthelmintic therapeutics.
Collapse
Affiliation(s)
- Masmudur M Rahman
- Center for Immunotherapy, Vaccines and Virotherapy, Bio design Institute, Arizona State University, Tempe, AZ, USA
| | - Grant McFadden
- Center for Immunotherapy, Vaccines and Virotherapy, Bio design Institute, Arizona State University, Tempe, AZ, USA
| | - Gordon Ruthel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert M Greenberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Swarna Bais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Ogongo P, Nyakundi RK, Chege GK, Ochola L. The Road to Elimination: Current State of Schistosomiasis Research and Progress Towards the End Game. Front Immunol 2022; 13:846108. [PMID: 35592327 PMCID: PMC9112563 DOI: 10.3389/fimmu.2022.846108] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The new WHO Roadmap for Neglected Tropical Diseases targets the global elimination of schistosomiasis as a public health problem. To date, control strategies have focused on effective diagnostics, mass drug administration, complementary and integrative public health interventions. Non-mammalian intermediate hosts and other vertebrates promote transmission of schistosomiasis and have been utilized as experimental model systems. Experimental animal models that recapitulate schistosomiasis immunology, disease progression, and pathology observed in humans are important in testing and validation of control interventions. We discuss the pivotal value of these models in contributing to elimination of schistosomiasis. Treatment of schistosomiasis relies heavily on mass drug administration of praziquantel whose efficacy is comprised due to re-infections and experimental systems have revealed the inability to kill juvenile schistosomes. In terms of diagnosis, nonhuman primate models have demonstrated the low sensitivity of the gold standard Kato Katz smear technique. Antibody assays are valuable tools for evaluating efficacy of candidate vaccines, and sera from graded infection experiments are useful for evaluating diagnostic sensitivity of different targets. Lastly, the presence of Schistosomes can compromise the efficacy of vaccines to other infectious diseases and its elimination will benefit control programs of the other diseases. As the focus moves towards schistosomiasis elimination, it will be critical to integrate treatment, diagnostics, novel research tools such as sequencing, improved understanding of disease pathogenesis and utilization of experimental models to assist with evaluating performance of new approaches.
Collapse
Affiliation(s)
- Paul Ogongo
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
| | - Ruth K. Nyakundi
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
| | - Gerald K. Chege
- Primate Unit & Delft Animal Centre, South African Medical Research Council, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Lucy Ochola
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
- Department of Environmental Health, School of Behavioural and Lifestyle Sciences, Faculty of Health Sciences, Nelson Mandela University, Gqeberha, South Africa
| |
Collapse
|
4
|
Chen H, Li G, Zhang J, Zheng T, Chen Q, Zhang Y, Yang F, Wang C, Nie H, Zheng B, Gong Q. Sodium butyrate ameliorates Schistosoma japonicum-induced liver fibrosis by inhibiting HMGB1 expression. Exp Parasitol 2021; 231:108171. [PMID: 34736899 DOI: 10.1016/j.exppara.2021.108171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/16/2021] [Accepted: 10/31/2021] [Indexed: 11/25/2022]
Abstract
Schistosomiasis is a prevalent zoonotic parasitic disease caused by schistosomes. Its main threat to human health is hepatic granuloma and fibrosis due to worm eggs. Praziquantel remains the first choice for the treatment of schistosomiasis but has limited benefit in treating liver fibrosis. Therefore, the need to develop effective drugs for treating schistosomiasis-induced hepatic fibrosis is urgent. High-mobility group box 1 protein (HMGB1) is a potential immune mediator that is highly associated with the development of some fibrotic diseases and may be involved in the liver pathology of schistosomiasis. We speculated that HMGB1 inhibitors could have an anti-fibrotic effect. Sodium butyrate (SB), a potent inhibitor of HMGB1, has shown anti-inflammatory activity in some animal disease models. In this study, we evaluated the effects of SB on a murine schistosomiasis model. Mice were percutaneously infected with 20 ± 2 cercariae of Schistosoma japonicum. SB (500 mg/kg/day) was administered every 3 days for the entire experiment period. The activity of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), liver histopathology, HMGB1 expression, and the levels of interferon gamma (IFN-γ), transforming growth factor-β1 (TGF-β1), and interleukin-6 (IL-6) in serum were analyzed. SB reduced hepatic granuloma and fibrosis of schistosomiasis, reflected by the decreased levels of ALT and AST in serum and the reduced expression of pro-inflammatory and fibrogenic cytokines (IFN-γ, TGF-β1, and IL-6). The protective effect could be attributable to the inhibition of the expression of HMGB1 and release by SB.
Collapse
Affiliation(s)
- Hui Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Gang Li
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Department of Gastroenterology, Jingmen Second People's Hospital, Jingmen, Hubei Province, 448000, PR China
| | - Jianqiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Ting Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Qianglin Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Yanxiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Fei Yang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Chao Wang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Hao Nie
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Bing Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China.
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China.
| |
Collapse
|
5
|
Tang CL, Li YH, Dai WQ, Zhu YW, Wu ZX, Li Y, Zuo T. GENE EXPRESSION LEVEL, IMMUNOLOCALIZATION, AND FUNCTION OF FATTY ACID-BINDING PROTEIN FROM SCHISTOSOMA JAPONICUM. J Parasitol 2021; 107:529-536. [PMID: 34198340 DOI: 10.1645/19-42] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The Schistosoma japonicum fatty acid-binding protein (FABP) is used in the cell membrane to absorb and transport fatty acids, which cannot be resynthesized by the organism and combined with hydrophobic ligands. Among the 5 stages of the worm life cycle examined, FABP messenger ribonucleic acid (mRNA) expression was highest in male adult worms, followed by the liver-stage schistosome, and was the lowest in the lung-stage schistosome. The fabp gene-coding region was cloned and expressed to obtain recombinant S. japonicum FABP (rSjFABP) with a molecular weight of approximately 18 kDa. Mice were then immunized against rSjFABP to prepare anti-FABP serum. Using immunohistochemical techniques, FABP protein was found to localize to the eggshell, parenchyma, and digestive tract. Double-stranded RNA-mediated knockdown of FABP mRNA by RNA interference decreased the number of transcripts by >70%. Moreover, the egg production rate decreased, whereas the abnormal egg ratio was significantly increased in the FABP-silenced group compared with the negative control group (P < 0.05). These results demonstrate that FABP localizes in adults and in various stages. FABP contributes to the egg-laying capacity of adults, which may be related to the reproductive function of S. japonicum.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Yan-Hong Li
- Department of Endocrinology, Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Wen-Qin Dai
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Ya-Wen Zhu
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Zhao-Xia Wu
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Yan Li
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Tao Zuo
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan 430030, China
| |
Collapse
|
6
|
Tang CL, Zhang RH, Li R, Li XR, Pan Q, Li L, Xiao JL. EFFECT OF ADENYLATE KINASE 1 ON THE GROWTH AND DEVELOPMENT OF SCHISTOSOMA JAPONICUM SCHISTOSOMULUM. J Parasitol 2021; 107:472-480. [PMID: 34153095 DOI: 10.1645/19-113] [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: 11/10/2022] Open
Abstract
We investigated the effect of Schistosoma japonicum adenylate kinase 1 (Sjak1) on the growth and development of schistosomula. Quantitative real-time PCR showed that Sjak1 mRNA was expressed in 3-, 10-, 14-, 18-, and 21-day-old schistosomula, and its levels increased gradually with the development of S. japonicum. Using immunohistochemical techniques, ak1 protein was found to be mainly distributed in the tegument and some parenchymal tissues of the schistosomula. Double-stranded RNA-mediated knockdowns of ak1 decreased ak1 mRNA transcripts by more than 90%, and western blot results showed that expression of ak1 protein was decreased by 66%. Scanning electron microscopy following the RNA-mediated ak1 knockdown showed that the sensory papillae did not develop. Transmission electron microscopy showed a lower mean thickness of the tegument in the Sjak1 interference group than in the negative control group. Terminal deoxynucleotidyl transferase dUTP nick-end labeling suggested higher apoptosis in the interference group than the negative control group. These results showed that ak1 may be involved in the growth and development of S. japonicum schistosomula and especially in the development of the integument. Consequently, ak1 may be a potential target in developing prevention methods for schistosomiasis in the future.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Rong-Hui Zhang
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Ru Li
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Xiu-Rong Li
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Qun Pan
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Li Li
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Jin-Lei Xiao
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| |
Collapse
|
7
|
Houlder EL, Costain AH, Cook PC, MacDonald AS. Schistosomes in the Lung: Immunobiology and Opportunity. Front Immunol 2021; 12:635513. [PMID: 33953712 PMCID: PMC8089482 DOI: 10.3389/fimmu.2021.635513] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/01/2021] [Indexed: 01/21/2023] Open
Abstract
Schistosome infection is a major cause of global morbidity, particularly in sub-Saharan Africa. However, there is no effective vaccine for this major neglected tropical disease, and re-infection routinely occurs after chemotherapeutic treatment. Following invasion through the skin, larval schistosomula enter the circulatory system and migrate through the lung before maturing to adulthood in the mesenteric or urogenital vasculature. Eggs released from adult worms can become trapped in various tissues, with resultant inflammatory responses leading to hepato-splenic, intestinal, or urogenital disease – processes that have been extensively studied in recent years. In contrast, although lung pathology can occur in both the acute and chronic phases of schistosomiasis, the mechanisms underlying pulmonary disease are particularly poorly understood. In chronic infection, egg-mediated fibrosis and vascular destruction can lead to the formation of portosystemic shunts through which eggs can embolise to the lungs, where they can trigger granulomatous disease. Acute schistosomiasis, or Katayama syndrome, which is primarily evident in non-endemic individuals, occurs during pulmonary larval migration, maturation, and initial egg-production, often involving fever and a cough with an accompanying immune cell infiltrate into the lung. Importantly, lung migrating larvae are not just a cause of inflammation and pathology but are a key target for future vaccine design. However, vaccine efforts are hindered by a limited understanding of what constitutes a protective immune response to larvae. In this review, we explore the current understanding of pulmonary immune responses and inflammatory pathology in schistosomiasis, highlighting important unanswered questions and areas for future research.
Collapse
Affiliation(s)
- Emma L Houlder
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Alice H Costain
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Peter C Cook
- MRC Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
8
|
Hambrook JR, Hanington PC. Immune Evasion Strategies of Schistosomes. Front Immunol 2021; 11:624178. [PMID: 33613562 PMCID: PMC7889519 DOI: 10.3389/fimmu.2020.624178] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/22/2020] [Indexed: 11/13/2022] Open
Abstract
Human schistosomes combat the unique immune systems of two vastly different hosts during their indirect life cycles. In gastropod molluscs, they face a potent innate immune response composed of variable immune recognition molecules and highly phagocytic hemocytes. In humans, a wide variety of innate and adaptive immune processes exist in proximity to these parasites throughout their lifespan. To survive and thrive as the second most common parasitic disease in humans, schistosomes have evolved many techniques to avoid and combat these targeted host responses. Among these techniques are molecular mimicry of host antigens, the utilization of an immune resistant outer tegument, the secretion of several potent proteases, and targeted release of specific immunomodulatory factors affecting immune cell functions. This review seeks to describe these key immune evasion mechanisms, among others, which schistosomes use to survive in both of their hosts. After diving into foundational observational studies of the processes mediating the establishment of schistosome infections, more recent transcriptomic and proteomic studies revealing crucial components of the host/parasite molecular interface are discussed. In order to combat this debilitating and lethal disease, a comprehensive understanding of schistosome immune evasion strategies is necessary for the development of novel therapeutics and treatment plans, necessitating the discussion of the numerous ways in which these parasitic flatworms overcome the immune responses of both hosts.
Collapse
Affiliation(s)
- Jacob R Hambrook
- School of Public Health, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
9
|
King M, Carson J, Stewart MT, Gobert GN. Revisiting the Schistosoma japonicum life cycle transcriptome for new insights into lung schistosomula development. Exp Parasitol 2021; 223:108080. [PMID: 33548219 DOI: 10.1016/j.exppara.2021.108080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 01/12/2021] [Accepted: 01/31/2021] [Indexed: 12/12/2022]
Abstract
Schistosome parasites are complex trematode blood flukes responsible for the disease schistosomiasis; a global health concern prevalent in many tropical and sub-tropical countries. While established transcriptomic databases are accessed ad hoc to facilitate studies characterising specific genes or gene families, a more comprehensive systematic updating of gene annotation and survey of the literature to aid in annotation and context is rarely addressed. We have reanalysed an online transcriptomic dataset originally published in 2009, where seven life cycle stages of Schistosoma japonicum were examined. Using the online pathway analysis tool Reactome, we have revisited key data from the original study. A key focus of this study was to improve the interpretation of the gene expression profile of the developmental lung-stage schistosomula, since it is one of the principle targets for worm elimination. Highly enriched transcripts, associated with lung schistosomula, were related to a number of important biological pathways including host immune evasion, energy metabolism and parasitic development. Revisiting large transcriptomic databases should be considered in the context of substantial new literature. This approach could aid in the improved understanding of the molecular basis of parasite biology. This may lead to the identification of new targets for diagnosis and therapies for schistosomes, and other helminths.
Collapse
Affiliation(s)
- Meághan King
- School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, UK
| | - Jack Carson
- School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, UK
| | - Michael T Stewart
- School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, UK
| | - Geoffrey N Gobert
- School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5DL, UK.
| |
Collapse
|
10
|
Lei Z, Tang R, Qi Q, Gu P, Wang J, Xu L, Wei C, Pu Y, Qi X, Chen Y, Yu B, Yu Y, Chen X, Zhu J, Li Y, Zhou S, Su C. Hepatocyte CD1d protects against liver immunopathology in mice with schistosomiasis japonica. Immunology 2020; 162:328-338. [PMID: 33283278 DOI: 10.1111/imm.13288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/17/2020] [Accepted: 11/21/2020] [Indexed: 12/16/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease with over 250 million people infected worldwide. The main clinically important species Schistosoma mansoni (S. mansoni) and Schistosoma japonicum (S. japonicum) cause inflammatory responses against tissue-trapped eggs, resulting in formation of granulomas mainly in host liver. Persistent granulomatous response results in severe fibrosis in the liver, leading to irreversible impairment of the liver and even death of the host. CD1d, a highly conserved MHC class I-like molecule, is expressed by both haematopoietic and non-haematopoietic cells. CD1d on antigen-presenting cells (APCs) of haematopoietic origin presents pathogen-derived lipid antigens to natural killer T (NKT) cells, which enables them to rapidly produce large amounts of various cytokines and facilitate CD4+ T helper (Th) cell differentiation upon invading pathogens. Noteworthy, hepatocytes of non-haematopoietic origin have recently been shown to be involved in maintaining liver NKT cell homeostasis through a CD1d-dependent manner. However, whether hepatocyte CD1d-dependent regulation of NKT cell homeostasis also modulates CD4+ Th cell responses and liver immunopathology in murine schistosomiasis remains to be addressed. Here, we show in mice that CD1d expression on hepatocytes was decreased dramatically upon S. japonicum infection, accompanied by increased NKT cells, as well as upregulated Th1 and Th2 responses. Overexpression of CD1d in hepatocytes significantly decreased local NKT numbers and cytokines (IFN-γ, IL-4, IL-13), concomitantly with downregulation of both Th1 and Th2 responses and alleviation in pathological damage in livers of S. japonicum-infected mice. These findings highlight the potential of hepatocyte CD1d-targeted therapies for liver immunopathology control in schistosomiasis.
Collapse
Affiliation(s)
- Zhigang Lei
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Tang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianqian Qi
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pan Gu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junling Wang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Xu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Wei
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanan Pu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Qi
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Chen
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Beibei Yu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanxiong Yu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojun Chen
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jifeng Zhu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yalin Li
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sha Zhou
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Su
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
11
|
Weatherhead JE, Gazzinelli-Guimaraes P, Knight JM, Fujiwara R, Hotez PJ, Bottazzi ME, Corry DB. Host Immunity and Inflammation to Pulmonary Helminth Infections. Front Immunol 2020; 11:594520. [PMID: 33193446 PMCID: PMC7606285 DOI: 10.3389/fimmu.2020.594520] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/30/2020] [Indexed: 01/04/2023] Open
Abstract
Helminths, including nematodes, cestodes and trematodes, are complex parasitic organisms that infect at least one billion people globally living in extreme poverty. Helminthic infections are associated with severe morbidity particularly in young children who often harbor the highest burden of disease. While each helminth species completes a distinct life cycle within the host, several helminths incite significant lung disease. This impact on the lungs occurs either directly from larval migration and host immune activation or indirectly from a systemic inflammatory immune response. The impact of helminths on the pulmonary immune response involves a sophisticated orchestration and activation of the host innate and adaptive immune cells. The consequences of activating pulmonary host immune responses are variable with several helminthic infections leading to severe, pulmonary compromise while others providing immune tolerance and protection against the development of pulmonary diseases. Further delineation of the convoluted interface between helminth infection and the pulmonary host immune responses is critical to the development of novel therapeutics that are critically needed to prevent the significant global morbidity caused by these parasites.
Collapse
Affiliation(s)
- Jill E. Weatherhead
- Department of Medicine, Infectious Diseases, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, Pediatric Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | | | - John M. Knight
- Department of Medicine, Pathology and Immunology, and the Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
| | - Ricardo Fujiwara
- Departamento de Parasitologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Peter J. Hotez
- Department of Pediatrics, Pediatric Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Center for Vaccine Development, Houston, TX, United States
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
- Department of Biology, Baylor University, Waco, TX, United States
- Hagler Institute for Advanced Study at Texas A&M University, College State, TX, United States
| | - Maria Elena Bottazzi
- Department of Pediatrics, Pediatric Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Center for Vaccine Development, Houston, TX, United States
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - David B. Corry
- Department of Medicine, Pathology and Immunology, and the Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Immunology, Allergy, Rheumatology, Baylor College of Medicine, Houston, TX, United States
- Michael E. DeBakey VA Center for Translational Research in Inflammatory Diseases, Houston, TX, United States
| |
Collapse
|
12
|
Role of adenylate kinase 1 in the integument development of Schistosoma japonicum schistosomula. Acta Trop 2020; 207:105467. [PMID: 32277925 DOI: 10.1016/j.actatropica.2020.105467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 03/14/2020] [Accepted: 03/29/2020] [Indexed: 11/21/2022]
Abstract
Schistosomula antigens play an important role in the growth and development of Schistosoma japonicum. We investigated the role of S. japonicum adenylate kinase 1 (SjAK1) in the growth and development of schistosomula. Quantitative real-time PCR showed that SjAK1 mRNA was expressed in all schistosomula stages, but increased gradually with the development of S. japonicum schistosomula. Using immunohistochemical techniques, the AK1 protein was found to be mainly distributed in the tegument and in some parenchymal tissues of the schistosomula. Double-stranded RNA-mediated knockdown of AK1 reduced AK1 mRNA transcripts by more than 90%; western blot analysis demonstrated that AK1 protein expression decreased by 66%. Scanning electron microscopy following RNA-mediated AK1 knockdown demonstrated that the sensory papillae degenerated significantly. Transmission electron microscopy demonstrated that the mean thickness of the tegument in the SjAK1 interference group was lower than that in the negative control group. Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) suggested that, compared with the negative control group, apoptosis increased in the interference group. These results show that AK1 may be involved in the growth and development of S. japonicum schistosomula, and thus may be a target when developing treatments for schistosomiasis.
Collapse
|
13
|
Melkus MW, Le L, Siddiqui AJ, Molehin AJ, Zhang W, Lazarus S, Siddiqui AA. Elucidation of Cellular Responses in Non-human Primates With Chronic Schistosomiasis Followed by Praziquantel Treatment. Front Cell Infect Microbiol 2020; 10:57. [PMID: 32154190 PMCID: PMC7050631 DOI: 10.3389/fcimb.2020.00057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
For decades, mass drug treatment with praziquantel (PZQ) has been utilized to treat schistosomiasis, yet reinfection and the risk of drug resistance are among the various factors precluding successful elimination of schistosomiasis. Tractable models that replicate "real world" field conditions are crucial to effectively evaluate putative schistosomiasis vaccines. Herein, we describe the cellular immune responses and cytokine expression profiles under field conditions that include prior infection with schistosomes followed by treatment with PZQ. Baboons were exposed to Schistosoma mansoni cercariae through trickle infection over 5 weeks, allowed for chronic disease to develop, and then treated with PZQ. Peripheral blood mononuclear cells (PBMCs) were monitored for cellular immune response(s) at each disease stage and PZQ therapy. After initial infection and during chronic disease, there was an increase in non-classical monocytes, NK and NKT cells while the CD4:CD8 T cell ratio inverted from a 2:1 to 1:2.5. The cytokine expressions of PBMCs after trickle infections were polarized more toward a Th2 response with a gradual increase in Th1 cytokine expression at chronic disease stage. Following PZQ treatment, with the exception of an increase in B cells, immune cell populations reverted back toward naïve levels; however, expression of almost all Th1, Th2, and Th17 cytokines was significantly increased. This preliminary study is the first to follow the cellular immune response and cytokine expression profiles in a non-human primate model simulating field conditions of schistosomiasis and PZQ therapy, providing a promising reference in predicting the immune response to future vaccines for schistosomiasis.
Collapse
Affiliation(s)
- Michael W Melkus
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Loc Le
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Arif J Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Biology, University of Hail, Hail, Saudi Arabia
| | - Adebayo J Molehin
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Samra Lazarus
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Afzal A Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
14
|
Zheng B, Zhang J, Chen H, Nie H, Miller H, Gong Q, Liu C. T Lymphocyte-Mediated Liver Immunopathology of Schistosomiasis. Front Immunol 2020; 11:61. [PMID: 32132991 PMCID: PMC7040032 DOI: 10.3389/fimmu.2020.00061] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/10/2020] [Indexed: 12/16/2022] Open
Abstract
The parasitic worms, Schistosoma mansoni and Schistosoma japonicum, reside in the mesenteric veins, where they release eggs that induce a dramatic granulomatous response in the liver and intestines. Subsequently, infection may further develop into significant fibrosis and portal hypertension. Over the past several years, uncovering the mechanism of immunopathology in schistosomiasis has become a major research objective. It is known that T lymphocytes, especially CD4+ T cells, are essential for immune responses against Schistosoma species. However, obtaining a clear understanding of how T lymphocytes regulate the pathological process is proving to be a daunting challenge. To date, CD4+ T cell subsets have been classified into several distinct T helper (Th) phenotypes including Th1, Th2, Th17, T follicular helper cells (Tfh), Th9, and regulatory T cells (Tregs). In the case of schistosomiasis, the granulomatous inflammation and the chronic liver pathology are critically regulated by the Th1/Th2 responses. Animal studies suggest that there is a moderate Th1 response to parasite antigens during the acute stage, but then, egg-derived antigens induce a sustained and dominant Th2 response that mediates granuloma formation and liver fibrosis. In addition, the newly discovered Th17 cells also play a critical role in the hepatic immunopathology of schistosomiasis. Within the liver, Tregs are recruited to hepatic granulomas and exert an immunosuppressive role to limit the granulomatous inflammation and fibrosis. Moreover, recent studies have shown that Tfh and Th9 cells might also promote liver granulomas and fibrogenesis in the murine schistosomiasis. Thus, during infection, T-cell subsets undergo complicated cross-talk with antigen presenting cells that then defines their various roles in the local microenvironment for regulating the pathological progression of schistosomiasis. This current review summarizes a vast body of literature to elucidate the contribution of T lymphocytes and their associated cytokines in the immunopathology of schistosomiasis.
Collapse
Affiliation(s)
- Bing Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Jianqiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Hui Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Hao Nie
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Heather Miller
- Department of Intracellular Pathogens, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
15
|
Abstract
Immunofluorescence allows the detection, visualization, and localization of proteins by using the ability of antibodies to firmly bind to specific antigens. Proteins must be accessible to thorough interaction with the specific antibodies. Different immune evasion mechanisms of parasites are directed to hamper or prevent access of antibodies to critical proteins or virulence factors. The blood fluke Schistosoma mansoni would not survive a day in the host blood capillaries if antibodies were able to readily bind to proteins located at the surface and mediate its attrition and demise by the complement system and/or the FcγR- or FcαR-bearing leukocytes. The worm surface is the area of parasite-host interaction and the route to critical nutrients, but is selectively permeable, allowing access of nutrient molecules but not host antibodies. Gentle procedures, which, however, are not commonly in use in vivo, are required to increase the permeability of the parasite outer membrane shield to just allow access of specific antibodies and identify and localize the proteins at the apical surface. Robust methods involving acetone, methanol, and Triton X-100 treatment lead to disintegration of the dual lipid bilayer cover with exposure of the proteins located in the tegument beneath. Internal proteins may not be accessed except following cryostat or paraffin sectioning. Accordingly, vaccine-induced specific antibodies to the apical surface or tegument proteins are unable to harm intact parasites. Specific antibodies to surface membrane proteins may only add to the action of administered or endo schistosomicides via acceleration of killing and interference with repair of severely and lightly impacted parasites, respectively. Therefore, careful immunofluorescent localization of S. mansoni proteins is important for devising the different control strategies against infection.
Collapse
Affiliation(s)
- Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt.
| | - Hatem Tallima
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt.,Department of Chemistry, School of Science and Engineering, American University in Cairo, New Cairo, Egypt
| |
Collapse
|
16
|
Tedla BA, Sotillo J, Pickering D, Eichenberger RM, Ryan S, Becker L, Loukas A, Pearson MS. Novel cholinesterase paralogs of Schistosoma mansoni have perceived roles in cholinergic signalling and drug detoxification and are essential for parasite survival. PLoS Pathog 2019; 15:e1008213. [PMID: 31809524 PMCID: PMC6919630 DOI: 10.1371/journal.ppat.1008213] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 12/18/2019] [Accepted: 11/13/2019] [Indexed: 01/27/2023] Open
Abstract
Cholinesterase (ChE) function in schistosomes is essential for orchestration of parasite neurotransmission but has been poorly defined with respect to the molecules responsible. Interrogation of the S. mansoni genome has revealed the presence of three ChE domain-containing genes (Smche)s, which we have shown to encode two functional acetylcholinesterases (AChE)s (Smache1 –smp_154600 and Smache2 –smp_136690) and a butyrylcholinesterase (BChE) (Smbche1 –smp_125350). Antibodies to recombinant forms of each SmChE localized the proteins to the tegument of adults and schistosomula and developmental expression profiling differed among the three molecules, suggestive of functions extending beyond traditional cholinergic signaling. For the first time in schistosomes, we identified ChE enzymatic activity in fluke excretory/secretory (ES) products and, using proteomic approaches, attributed this activity to the presence of SmAChE1 and SmBChE1. Parasite survival in vitro and in vivo was significantly impaired by silencing of each smche, either individually or in combination, attesting to the essential roles of these molecules. Lastly, in the first characterization study of a BChE from helminths, evidence is provided that SmBChE1 may act as a bio-scavenger of AChE inhibitors as the addition of recombinant SmBChE1 to parasite cultures mitigated the effect of the anti-schistosome AChE inhibitor 2,2- dichlorovinyl dimethyl phosphate—dichlorvos (DDVP), whereas smbche1-silenced parasites displayed increased sensitivity to DDVP. Cholinesterases—aceytlcholinesterases (AChE)s and butyrylcholinesterases (BChE)s—are multi-functional enzymes that play a pivotal role in the nervous system of parasites by regulating neurotransmission through acetylcholine hydrolysis. Herein, we provide a detailed characterization of schistosome cholinesterases using molecular, enzymatic and gene-silencing approaches and show evidence for these molecules having roles in addition to their neuronal function. Further, we demonstrate the importance of these proteins to parasite development and survival through gene knockdown experiments in laboratory animals, providing evidence for the use of these proteins in the development of novel intervention strategies against schistosomiasis.
Collapse
Affiliation(s)
- Bemnet A. Tedla
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Javier Sotillo
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Darren Pickering
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Ramon M. Eichenberger
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
- Institute of Parasitology, University of Zurich, Zurich, Switzerland
| | - Stephanie Ryan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Luke Becker
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Mark S. Pearson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
- * E-mail:
| |
Collapse
|
17
|
Butrous G. Schistosome infection and its effect on pulmonary circulation. Glob Cardiol Sci Pract 2019; 2019:5. [PMID: 31024947 PMCID: PMC6472693 DOI: 10.21542/gcsp.2019.5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
Schistosomiasis is the most common parasitic disease associated with pulmonary hypertension. It induces remodelling via complex inflammatory processes, which eventually produce the clinical manifestation of pulmonary hypertension. The pulmonary hypertension shows clinical signs and symptoms that are not distinguishable from other forms of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Ghazwan Butrous
- Professor of Cardiopulmonary Sciences, Medway School of Pharmacy, University of Kent, UK and University of Greenwich, Central Ave, Gillingham, Chatham ME4 4BF, Kent, UK
| |
Collapse
|
18
|
Mäder P, Rennar GA, Ventura AMP, Grevelding CG, Schlitzer M. Chemotherapy for Fighting Schistosomiasis: Past, Present and Future. ChemMedChem 2018; 13:2374-2389. [DOI: 10.1002/cmdc.201800572] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Indexed: 01/22/2023]
Affiliation(s)
- Patrick Mäder
- Department of Pharmaceutical Chemistry; Philipps-Universität Marburg; Marbacher Weg 6 35032 Marburg Germany
| | - Georg A. Rennar
- Department of Pharmaceutical Chemistry; Philipps-Universität Marburg; Marbacher Weg 6 35032 Marburg Germany
| | - Alejandra M. Peter Ventura
- Department of Pharmaceutical Chemistry; Philipps-Universität Marburg; Marbacher Weg 6 35032 Marburg Germany
| | - Christoph G. Grevelding
- Institute of Parasitology, BFS; Justus-Liebig-Universität Gießen; Schubertstraße 81 35392 Gießen Germany
| | - Martin Schlitzer
- Department of Pharmaceutical Chemistry; Philipps-Universität Marburg; Marbacher Weg 6 35032 Marburg Germany
| |
Collapse
|
19
|
Souza COS, Espíndola MS, Fontanari C, Prado MKB, Frantz FG, Rodrigues V, Gardinassi LG, Faccioli LH. CD18 Regulates Monocyte Hematopoiesis and Promotes Resistance to Experimental Schistosomiasis. Front Immunol 2018; 9:1970. [PMID: 30233576 PMCID: PMC6127275 DOI: 10.3389/fimmu.2018.01970] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022] Open
Abstract
Infection with Schistosoma mansoni causes a chronic parasitic disease that progress to severe liver and gastrointestinal damage, and eventually death. During its development into mammalian hosts, immature schistosomula transit through the lung vasculature before they reach the liver to mature into adult worms. A low grade inflammatory reaction is induced during this process. However, molecules that are required for efficient leukocyte accumulation in the lungs of S. mansoni-infected subjects are unknown. In addition, specific leukocyte subsets that mediate pulmonary response during S. mansoni migration through the lung remain to be elucidated. β2 integrins are fundamental regulators of leukocyte trans-endothelial migration and function. Therefore, we investigated their role during experimental schistosomiasis. Mice that express low levels of CD18 (the common β2 integrin subunit) and wild type C57BL/6 mice were subcutaneously infected with S. mansoni cercariae. Cellular profiles of lungs and livers were evaluated in different time points after infection by flow cytometry. Low levels of CD18 affected the accumulation of patrolling Ly6Clow, intermediate Ly6Cinter monocytes, monocyte-derived macrophages and monocyte-derived dendritic cells in the lungs 7 days after infection. This correlated with increased TNF-α levels. Strikingly, low CD18 expression resulted in monocytopenia both in the peripheral blood and bone marrow during acute infection. After 48 days, S. mansoni worm burdens were higher in the hepatic portal system of CD18low mice, which also displayed reduced hepatic accumulation of patrolling Ly6Clow and intermediate Ly6Cinter, but not inflammatory Ly6Chigh monocytes. Higher parasite burden resulted in increased granulomatous lesions in the liver, increased egg deposition and enhanced mortality. Overall, our data point for a fundamental role of CD18 for monocyte hematopoiesis during infection, which promotes an efficient host response against experimental schistosomiasis.
Collapse
Affiliation(s)
- Camila O S Souza
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Milena S Espíndola
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Caroline Fontanari
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Morgana K B Prado
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Fabiani G Frantz
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Vanderlei Rodrigues
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz G Gardinassi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Lúcia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Chen L, Chen Q, Hou W, He L. High-throughput dynamic analysis of differentially expressed genes in splenic dendritic cells from mice infected with Schistosoma japonicum. Immunol Lett 2017; 184:15-22. [PMID: 28185924 DOI: 10.1016/j.imlet.2017.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 02/04/2017] [Accepted: 02/06/2017] [Indexed: 12/21/2022]
Abstract
Dendritic cells are the initiation and key point of immune response and play a role in immune regulation. So we explored the mechanisms involved in immune regulation of dendritic cells (DCs) against schistosomiasis using mice infected with Schistosoma japonicum. Splenic DCs from normal mice and mice with acute and chronic S. japonicum infection were sorted by flow cytometry. The numbers and functions of differentially expressed genes (DEGs) in DCs were determined by high-throughput analysis. All DEGs with transcription-level fold changes of ≥2 were selected and matched to corresponding genes in databases. Annotations and cluster analysis of DEGs were performed to compare differences between groups. Six important DEGs about immune regulation-CD86, TLR2, DC-SIGN, Capase3, PD-L2, and IL-7r were selected, and their transcription levels at different stages of schistosomisis were validated by qPCR. The Venn diagram of DEGs implied some genes are functional at all stages during S. japonicum infection, while others are only involved at certain stages. GO and KEGG pathway annotations indicated that these DEGs mainly belong to biological regulation, regulation of biological process, regulation of cellular process, antigen processing and presentation, cell adhesion molecules, cytokine-cytokine receptor interaction and Toll-like receptor signaling. Cluster analysis revealed immune regulation existed in splenic DCs. The results above indicated that the mechanisms underlying immune regulation to S. japonicum infection in mice are very complex. The present high-throughput dynamic analysis of DEGs in splenic DCs provides valuable insights into the molecular mechanisms underlying immune regulation in S. japonicum infection.
Collapse
Affiliation(s)
- Lin Chen
- Department of Parasitology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Qingzhou Chen
- State Key Laboratory of Virology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Wei Hou
- State Key Laboratory of Virology, Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Li He
- Department of Parasitology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
21
|
Morphew RM, Wilkinson TJ, Mackintosh N, Jahndel V, Paterson S, McVeigh P, Abbas Abidi SM, Saifullah K, Raman M, Ravikumar G, LaCourse J, Maule A, Brophy PM. Exploring and Expanding the Fatty-Acid-Binding Protein Superfamily in Fasciola Species. J Proteome Res 2016; 15:3308-21. [PMID: 27495901 DOI: 10.1021/acs.jproteome.6b00331] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The liver flukes Fasciola hepatica and F. gigantica infect livestock worldwide and threaten food security with climate change and problematic control measures spreading disease. Fascioliasis is also a foodborne disease with up to 17 million humans infected. In the absence of vaccines, treatment depends on triclabendazole (TCBZ), and overuse has led to widespread resistance, compromising future TCBZ control. Reductionist biology from many laboratories has predicted new therapeutic targets. To this end, the fatty-acid-binding protein (FABP) superfamily has proposed multifunctional roles, including functions intersecting vaccine and drug therapy, such as immune modulation and anthelmintic sequestration. Research is hindered by a lack of understanding of the full FABP superfamily complement. Although discovery studies predicted FABPs as promising vaccine candidates, it is unclear if uncharacterized FABPs are more relevant for vaccine formulations. We have coupled genome, transcriptome, and EST data mining with proteomics and phylogenetics to reveal a liver fluke FABP superfamily of seven clades: previously identified clades I-III and newly identified clades IV-VII. All new clade FABPs were analyzed using bioinformatics and cloned from both liver flukes. The extended FABP data set will provide new study tools to research the role of FABPs in parasite biology and as therapy targets.
Collapse
Affiliation(s)
- Russell M Morphew
- Aberystwyth University , Institute of Biological, Environmental and Rural Sciences, Aberystwyth SY23 3DA, United Kingdom
| | - Toby J Wilkinson
- Aberystwyth University , Institute of Biological, Environmental and Rural Sciences, Aberystwyth SY23 3DA, United Kingdom
| | - Neil Mackintosh
- Aberystwyth University , Institute of Biological, Environmental and Rural Sciences, Aberystwyth SY23 3DA, United Kingdom
| | - Veronika Jahndel
- University of Leipzig , Institute of Biochemistry, D-04103 Leipzig, Germany
| | - Steve Paterson
- University of Liverpool , School of Biological Sciences, Liverpool L69 7ZB, United Kingdom
| | - Paul McVeigh
- Queen's University Belfast , School of Biological Sciences, Belfast BT7 1NN, United Kingdom
| | | | - Khalid Saifullah
- Aligarh Muslim University , Aligarh, Uttar Pradesh 202002, India
| | - Muthusamy Raman
- Tamil Nadu Veterinary and Animal Sciences University , Chennai 600-051, India
| | | | - James LaCourse
- Liverpool School of Tropical Medicine , Liverpool L3 5QA, United Kingdom
| | - Aaron Maule
- Queen's University Belfast , School of Biological Sciences, Belfast BT7 1NN, United Kingdom
| | - Peter M Brophy
- Aberystwyth University , Institute of Biological, Environmental and Rural Sciences, Aberystwyth SY23 3DA, United Kingdom
| |
Collapse
|
22
|
Cercarial Dermatitis or Cercariosis: Defining the Condition. Trends Parasitol 2016; 32:352-353. [DOI: 10.1016/j.pt.2016.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 03/04/2016] [Indexed: 11/24/2022]
|
23
|
Cao X, Fu Z, Zhang M, Han Y, Han Q, Lu K, Li H, Zhu C, Hong Y, Lin J. Excretory/secretory proteome of 14-day schistosomula, Schistosoma japonicum. J Proteomics 2015; 130:221-30. [PMID: 26453986 DOI: 10.1016/j.jprot.2015.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/21/2015] [Accepted: 10/04/2015] [Indexed: 01/09/2023]
Abstract
Schistosomiasis remains a serious public health problem, with 200 million people infected and 779 million people at risk worldwide. The schistosomulum is the early stage of the complex lifecycle of Schistosoma japonicum in their vertebrate hosts, and is the main target of vaccine-induced protective immunity. Excretory/secretory (ES) proteins play a major role in host-parasite interactions and ES protein compositions of schistosomula of S. japonicum have not been characterized to date. In the present study, the proteome of ES proteins from 14 day schistosomula of S. japonicum was analyzed by liquid chromatography/tandem mass spectrometry and 713 unique proteins were finally identified. Gene ontology and pathway analysis revealed that identified proteins were mainly involved in carbohydrate metabolism, degradation, response to stimulus, oxidation-reduction, biological regulation and binding. Flow cytometry analysis demonstrated that thioredoxin peroxidase identified in this study had the effect on inhibiting MHCII and CD86 expression on LPS-activated macrophages. The present study provides insight into the growth and development of the schistosome in the final host and valuable information for screening vaccine candidates for schistosomiasis.
Collapse
Affiliation(s)
- Xiaodan Cao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Zhiqiang Fu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Min Zhang
- College of Animal Science and Technology, Henran University of Science and Technology, Luoyang, China
| | - Yanhui Han
- College of Animal Science, Henan Institute of Science and Technology, Xinxiang, China
| | - Qian Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Ke Lu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Hao Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Chuangang Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Yang Hong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China.
| | - Jiaojiao Lin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
24
|
Neves BJ, Braga RC, Bezerra JCB, Cravo PVL, Andrade CH. In silico repositioning-chemogenomics strategy identifies new drugs with potential activity against multiple life stages of Schistosoma mansoni. PLoS Negl Trop Dis 2015; 9:e3435. [PMID: 25569258 PMCID: PMC4287566 DOI: 10.1371/journal.pntd.0003435] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 11/23/2014] [Indexed: 12/12/2022] Open
Abstract
Morbidity and mortality caused by schistosomiasis are serious public health problems in developing countries. Because praziquantel is the only drug in therapeutic use, the risk of drug resistance is a concern. In the search for new schistosomicidal drugs, we performed a target-based chemogenomics screen of a dataset of 2,114 proteins to identify drugs that are approved for clinical use in humans that may be active against multiple life stages of Schistosoma mansoni. Each of these proteins was treated as a potential drug target, and its amino acid sequence was used to interrogate three databases: Therapeutic Target Database (TTD), DrugBank and STITCH. Predicted drug-target interactions were refined using a combination of approaches, including pairwise alignment, conservation state of functional regions and chemical space analysis. To validate our strategy, several drugs previously shown to be active against Schistosoma species were correctly predicted, such as clonazepam, auranofin, nifedipine, and artesunate. We were also able to identify 115 drugs that have not yet been experimentally tested against schistosomes and that require further assessment. Some examples are aprindine, gentamicin, clotrimazole, tetrabenazine, griseofulvin, and cinnarizine. In conclusion, we have developed a systematic and focused computer-aided approach to propose approved drugs that may warrant testing and/or serve as lead compounds for the design of new drugs against schistosomes. Schistosomiasis is a neglected tropical disease caused by schistosome parasites that affects millions of people worldwide. The current reliance on a single drug (Praziquantel) for treatment and control of the disease calls for the urgent discovery of novel schistosomicidal agents. One approach that can expedite drug discovery is to find new uses for existing approved drugs, a practice known as drug repositioning. Currently, modern drug repositioning strategies entail the search for compounds that act on a specific target, often a protein known or suspected to be required for survival of the parasite. Drug repositioning approaches for schistosomiasis are now greatly facilitated by the availability of comprehensive schistosome genome data in user-friendly databases. Here, we report a drug repositioning computational strategy that involves identification of novel schistosomicidal drug candidates using similarity between schistosome proteins and known drug targets. Researchers can now use the list of predicted drugs as a basis for deciding which potential schistosomicidal candidates can be tested experimentally.
Collapse
Affiliation(s)
- Bruno J. Neves
- LabMol – Laboratory for Drug Design and Modeling, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Brazil
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Rodolpho C. Braga
- LabMol – Laboratory for Drug Design and Modeling, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Brazil
- Instituto de Química, Universidade Federal de Goiás, Goiaânia, Brazil
| | - José C. B. Bezerra
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Pedro V. L. Cravo
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
- Centro de Malária e Doenças Tropicais, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Carolina H. Andrade
- LabMol – Laboratory for Drug Design and Modeling, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, Brazil
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
- * E-mail:
| |
Collapse
|
25
|
Ye Q, Dong HF, Grevelding CG, Hu M. In vitro cultivation of Schistosoma japonicum-parasites and cells. Biotechnol Adv 2013; 31:1722-37. [DOI: 10.1016/j.biotechadv.2013.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/06/2013] [Accepted: 09/08/2013] [Indexed: 11/27/2022]
|
26
|
Abstract
Schistosomiasis is the second most important parasitic disease in the world in terms of public health impact. Globally, it is estimated that the disease affects over 200 million people and is responsible for 200,000 deaths each year. The three major schistosomes infecting humans are Schistosoma mansoni, S. japonicum, and S. haematobium. Much immunological research has focused on schistosomiasis because of the pathological effects of the disease, which include liver fibrosis and bladder dysfunction. This unit covers a wide range of aspects with respect to maintaining the life cycles of these parasites, including preparation of schistosome egg antigen, maintenance of intermediate snail hosts, infection of the definitive and intermediate hosts, and others. The unit primarily focuses on S. mansoni, but also includes coverage of S. japonicum and S. haematobium life cycles.
Collapse
Affiliation(s)
| | | | - Fred A Lewis
- Biomedical Research Institute, Rockville, Maryland
| | | | | |
Collapse
|
27
|
Thétiot-Laurent SAL, Boissier J, Robert A, Meunier B. Chemotherapie gegen Schistosomiasis. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201208390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
28
|
Thétiot-Laurent SAL, Boissier J, Robert A, Meunier B. Schistosomiasis chemotherapy. Angew Chem Int Ed Engl 2013; 52:7936-56. [PMID: 23813602 DOI: 10.1002/anie.201208390] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Indexed: 01/08/2023]
Abstract
After malaria, schistosomiasis (or bilharzia) is the second most prevalent disease in Africa, and is occurring in over 70 countries in tropical and subtropical regions. It is estimated that 600 million people are at risk of infection, 200 million people are infected, and at least 200,000 deaths per year are associated with the disease. All schistosome species are transmitted through contact with fresh water that is infested with free-swimming forms of the parasite, which is known as cercariae and produced by snails. When located in the blood vessels of the host, larval and adult schistosomes digest red cells to acquire amino acids for growth and development. Vaccine candidates have been unsuccessful up to now. Against such devastating parasitic disease, the antischistosomal arsenal is currently limited to a single drug, praziquantel, which has been used for more than 35 years. Because the question of the reduction of the activity of praziquantel was raised recently, it is thus urgent to create new and safe antischistosomal drugs that should be combined with praziquantel to develop efficient bitherapies.
Collapse
Affiliation(s)
- Sophie A-L Thétiot-Laurent
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, BP 44099, 31077 Toulouse cedex 4, France
| | | | | | | |
Collapse
|
29
|
Han H, Peng J, Gobert GN, Hong Y, Zhang M, Han Y, Fu Z, Shi Y, Xu J, Lin J, Tao J. Apoptosis phenomenon in the schistosomulum and adult worm life cycle stages of Schistosoma japonicum. Parasitol Int 2013; 62:100-8. [DOI: 10.1016/j.parint.2012.09.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Revised: 09/17/2012] [Accepted: 09/19/2012] [Indexed: 02/06/2023]
|
30
|
Schistosoma tegument proteins in vaccine and diagnosis development: an update. J Parasitol Res 2012; 2012:541268. [PMID: 23125917 PMCID: PMC3483795 DOI: 10.1155/2012/541268] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 09/24/2012] [Indexed: 01/16/2023] Open
Abstract
The development of a vaccine against schistosomiasis and also the availability of a more sensitive diagnosis test are important tools to help chemotherapy in controlling disease transmission. Bioinformatics tools, together with the access to parasite genome, published recently, should help generate new knowledge on parasite biology and search for new vaccines or therapeutic targets and antigens to be used in the disease diagnosis. Parasite surface proteins, especially those expressed in schistosomula tegument, represent interesting targets to be used in vaccine formulations and in the diagnosis of early infections, since the tegument represents the interface between host and parasite and its molecules are responsible for essential functions to parasite survival. In this paper we will present the advances in the development of vaccines and diagnosis tests achieved with the use of the information from schistosome genome focused on parasite tegument as a source for antigens.
Collapse
|
31
|
Zhang YL, Jia K, Zhao BP, Li Y, Yuan CX, Yang JM, Lin JJ, Feng XG. Identification of Th1 epitopes within molecules from the lung-stage schistosomulum of Schistosoma japonicum by combining prediction analysis of the transcriptome with experimental validation. Parasitol Int 2012; 61:586-93. [PMID: 22617496 DOI: 10.1016/j.parint.2012.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Revised: 05/04/2012] [Accepted: 05/14/2012] [Indexed: 01/01/2023]
Abstract
The lung-stage schistosomulum has been regarded as the main target of protective immunity induced by radiation-attenuated vaccines (RAV) in the mouse model of schistosomiasis, and immune mechanisms mediated by the CD4+ Th1 response play a major role in the RAV model. To identify Th1 epitopes rapidly within molecules from the lung schistosomulum of Schistosoma japonicum, in the present study we analyzed transcriptome data from normal and radiation-attenuated lung schistosomula of S. japonicum and Schistosoma mansoni. We selected six genes with high levels of expression of their transcripts as sample sequences from the lung schistosomula. From these six sequences, by using different algorithms, we predicted six promiscuous Th cell epitopes that are capable of binding to both murine and human MHC class II molecules. To validate our in silico prediction experimentally, first, the gene expressions of the six sequences in day 3 lung-stage schistosomula were assessed using reverse-transcription PCR (polymerase chain reaction) analysis. The result showed that all six sequences predicted can be expressed in normal day 3 schistosomula. Second, we measured the direct binding of the four peptides predicted above to APCs (Antigen Presenting Cells) from the BALB/c mouse strain using a fluorometric method, and found that the four peptides could bind to both I-Ad and I-Ed molecules of the mice. Finally, the proliferation and profiles of cytokine production by spleen lymphocytes from the BALB/c mice immunized with the six predicted peptides were detected in vitro using modified MTT (Methyl Thiazolyl Tetrazolium), and flow cytometry methods, respectively. The results showed that three of the six predicted peptides could induce a recall CD4+ Th1 response in vitro. These results demonstrate that potential Th1-type epitopes can be identified rapidly by a combination of in silico analysis of transcriptomes of lung-stage schistosomula with experimental validation.
Collapse
Affiliation(s)
- Yan Li Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, 518 Zi'yue Road Shanghai 200241, China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Coultas KA, Zhang SM. In vitro cercariae transformation: comparison of mechanical and nonmechanical methods and observation of morphological changes of detached cercariae tails. J Parasitol 2012; 98:1257-61. [PMID: 22519732 DOI: 10.1645/ge-3072.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Schistosomula, the larval stage of schistosomes in vertebrate hosts, are highly vulnerable and considered an ideal target for vaccine and drug development. Although the schistosomule stage is essential for biological studies, collecting sufficient numbers of schistosomula from their definitive hosts in vivo is difficult to accomplish. However, in vitro collection via cercariae transformation can effectively yield high numbers of schistosomula. We compared a current and widely used double-ended-needle mechanical transformation method to a culture medium based on a nonmechanical method. We found the rates of transformed cercariae, i.e., separated cercariae heads from tails, differed by only 2-7% at 0.5, 1, and 2 days in culture and that there was no significant difference in the number of transformed cercariae between the transformation methods at 3 and 4 days in culture. Notably, the mechanical and nonmechanical cercariae transformation methods both yielded significantly large and similar quantities of viable schistosomula. Given that the nonmechanical method is simpler and less damaging to the parasites, we recommend the use of it as an alternative way for in vitro cercariae transformation. In addition, we also observed morphological changes of the detached cercariae tails in culture medium. Interestingly, the tails are able to regenerate head-like organs/tissues and survive for at least 4 days. This intriguing change suggests unique biological features of the cells in the tails.
Collapse
Affiliation(s)
- Kristen A Coultas
- Center for Evolutionary and Theoretical Immunology, Department of Biology, MSC03 2020, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | | |
Collapse
|
33
|
Wen X, He L, Chi Y, Zhou S, Hoellwarth J, Zhang C, Zhu J, Wu C, Dhesi S, Wang X, Liu F, Su C. Dynamics of Th17 cells and their role in Schistosoma japonicum infection in C57BL/6 mice. PLoS Negl Trop Dis 2011; 5:e1399. [PMID: 22102924 PMCID: PMC3216943 DOI: 10.1371/journal.pntd.0001399] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Accepted: 09/30/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The current knowledge of immunological responses to schistosomiasis, a major tropical helminthic disease, is insufficient, and a better understanding of these responses would support vaccine development or therapies to control granuloma-associated immunopathology. CD4(+) T cells play critical roles in both host immune responses against parasitic infection and immunopathology in schistosomiasis. The induction of T helper (Th)1, Th2 and T regulatory (Treg) cells and their roles in schistosome infections are well-illustrated. However, little in vivo data are available on the dynamics of Th17 cells, another important CD4(+) T cell subset, after Schistosoma japonicum infection or whether these cells and their defining IL-17 cytokine mediate host protective responses early in infection. METHODOLOGY Levels of Th17 and the other three CD4(+) T cell subpopulations and the cytokines related to induction or repression of Th17 cell generation in different stages of S. japonicum infection were observed. Contrary to reported in vitro studies, our results showed that the Th17 cells were induced along with the Th1, Th2, Treg cells and the IFN-γ and IL-4 cytokines in S. japonicum infected mice. The results also suggested that S. japonicum egg antigens but not adult worm antigens preferentially induced Th17 cell generation. Furthermore, decreasing IL-17 with a neutralizing anti-IL-17 monoclonal antibody (mAb) increased schistosome-specific antibody levels and partial protection against S. japonicum infection in mice. CONCLUSIONS Our study is the first to report the dynamics of Th17 cells during S. japonicum infection and indicate that Th17 cell differentiation results from the integrated impact of inducing and suppressive factors promoted by the parasite. Importantly, our findings suggest that lower IL-17 levels may result in favorable host protective responses. This study significantly contributes to the understanding of immunity to schistosomiasis and may aid in developing interventions to protect hosts from infection or restrain immunopathology.
Collapse
Affiliation(s)
- Xiaoyun Wen
- Department of Pathogen Biology and Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lei He
- Department of Pathogen Biology and Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Ying Chi
- Department of Pathogen Biology and Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Sha Zhou
- Department of Pathogen Biology and Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jason Hoellwarth
- Department of Educational Affairs, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Cui Zhang
- Department of Pathogen Biology and Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jifeng Zhu
- Department of Pathogen Biology and Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Calvin Wu
- Department of Educational Affairs, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Shawn Dhesi
- Department of Educational Affairs, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Xuefeng Wang
- Department of Pathogen Biology and Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Feng Liu
- Department of Pathogen Biology and Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Chuan Su
- Department of Pathogen Biology and Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, People's Republic of China
- * E-mail:
| |
Collapse
|
34
|
Liao Q, Yuan X, Xiao H, Liu C, Lv Z, Zhao Y, Wu Z. Identifying Schistosoma japonicum excretory/secretory proteins and their interactions with host immune system. PLoS One 2011; 6:e23786. [PMID: 21887319 PMCID: PMC3161075 DOI: 10.1371/journal.pone.0023786] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 07/25/2011] [Indexed: 12/22/2022] Open
Abstract
Schistosoma japonicum is a major infectious agent of schistosomiasis. It has been reported that large number of proteins excreted and secreted by S. japonicum during its life cycle are important for its infection and survival in definitive hosts. These proteins can be used as ideal candidates for vaccines or drug targets. In this work, we analyzed the protein sequences of S. japonicum and found that compared with other proteins in S. japonicum, excretory/secretory (ES) proteins are generally longer, more likely to be stable and enzyme, more likely to contain immune-related binding peptides and more likely to be involved in regulation and metabolism processes. Based on the sequence difference between ES and non-ES proteins, we trained a support vector machine (SVM) with much higher accuracy than existing approaches. Using this SVM, we identified 191 new ES proteins in S. japonicum, and further predicted 7 potential interactions between these ES proteins and human immune proteins. Our results are useful to understand the pathogenesis of schistosomiasis and can serve as a new resource for vaccine or drug targets discovery for anti-schistosome.
Collapse
Affiliation(s)
- Qi Liao
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
- Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China
- Bioinformatics Research Group, Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xiongying Yuan
- Bioinformatics Research Group, Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Hui Xiao
- Bioinformatics Research Group, Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Changning Liu
- Bioinformatics Research Group, Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Zhiyue Lv
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
- Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yi Zhao
- Bioinformatics Research Group, Key Laboratory of Intelligent Information Processing, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
- * E-mail: (YZ); (ZW)
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
- Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China
- * E-mail: (YZ); (ZW)
| |
Collapse
|
35
|
Torben W, Ahmad G, Zhang W, Siddiqui AA. Role of antibodies in Sm-p80-mediated protection against Schistosoma mansoni challenge infection in murine and nonhuman primate models. Vaccine 2011; 29:2262-71. [PMID: 21277404 DOI: 10.1016/j.vaccine.2011.01.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 12/23/2010] [Accepted: 01/13/2011] [Indexed: 01/23/2023]
Abstract
Schistosomiasis is an important public health concern in more than 76 developing countries. Advent of an anti-schistosome vaccine would undoubtedly add to the existing control measures and may eventually help in the elimination of this disease. In the present study we have attempted to dissect the role(s) of antibodies in Sm-p80 mediated protection by intravenously transferring pooled sera from mice immunized with Sm-p80-pcDNA3 or purified IgG from baboons immunized with Sm-p80-pcDNA3, into naïve C57BL/6 mice, respectively, prior to challenge with cercariae. The passive transfer of antibodies from protected mice (homologous transfers) as well as transfer of total IgG from baboons (heterologous transfers), into naïve mice showed statistically significant reductions in worm burden and in the number of eggs in the tissues. Immunizations of antibody knockout mice (μMt-/-; B10.129S2 (B6)-Igh-6(tm1Cgn)/J) with recombinant Sm-p80 in the presence of CpG-motif oligodeoxynucleotides as an adjuvant, resulted in substantial reduction of Sm-p80-mediated protection, compared to C57BL/6 (normal) control group of mice. Down regulation of cytokines that have important effects on B cell proliferation as well as the recovery of higher number of parasites in antibody knockout indicated a significant role(s) of antibodies in Sm-p80-mediated protection against Schistosoma mansoni in mice. In toto, these studies appear to suggest that antibodies play a significant role in Sm-p80 mediated protection.
Collapse
Affiliation(s)
- Workineh Torben
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | | | |
Collapse
|
36
|
Araújo AP, Frezza TF, Allegretti SM, Giorgio S. Hypoxia, hypoxia-inducible factor-1α and vascular endothelial growth factor in a murine model of Schistosoma mansoni infection. Exp Mol Pathol 2010; 89:327-33. [PMID: 20858486 DOI: 10.1016/j.yexmp.2010.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 09/09/2010] [Accepted: 09/09/2010] [Indexed: 11/28/2022]
Abstract
Schistosomiasis mansoni is a chronic parasitic disease where much of the symptomatology is attributed to granuloma formation, an immunopathological reaction against Schistosoma eggs. To more clearly understand the immunopathology of schistosomiasis, the tissue microenvironment generated by S. mansoni infected mice was investigated. Using the hypoxia marker pimonidazole, we provide immunohistochemical evidence that hypoxia occurred in inflammatory cells infiltrated around the eggs and cells surrounding granulomas in the liver, intestine, spleen and lungs of infected mice. Hypoxia-inducible factor-1α (HIF-1α) was mainly expressed in inflammatory cells surrounding the eggs and in hepatocytes surrounding cellular and fibrocellular granulomas in infected mouse liver. HIF-1α expression was also verified in granulomas in the other tissues tested (intestine, spleen and lungs). Vascular endothelial growth factor (VEGF) expression was observed in the extracellular space surrounding inflammatory cells in liver granuloma. The VEGF expression pattern verified in infected mouse liver was very similar to that observed in the other tissues tested. A strong positive correlation occurred between pimonidazole binding and HIF-1α and VEGF expression in the tissues tested, except for lung. This work is the first evidence that infection by a helminth parasite, S. mansoni, produces a hypoxic tissue microenvironment and induces HIF-1α and VEGF expression.
Collapse
Affiliation(s)
- Alexandra Paiva Araújo
- Department of Animal Biology, Biology Institute, Universidade Estadual de Campinas, Caixa Postal 6109, Cep 13083-970, Campinas, SP, Brazil
| | | | | | | |
Collapse
|
37
|
Activation-induced T helper cell death contributes to Th1/Th2 polarization following murine Schistosoma japonicum infection. J Biomed Biotechnol 2010; 2010:202397. [PMID: 20111738 PMCID: PMC2810471 DOI: 10.1155/2010/202397] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Accepted: 10/15/2009] [Indexed: 02/07/2023] Open
Abstract
In chronic infectious diseases, such as schistosomiasis, pathogen growth and immunopathology are affected by the induction of a proper balanced Th1/Th2 response to the pathogen and by antigen-triggered activation-induced T cell death. Here, by using S. japonicum infection or schistosome antigens-immunized mouse model, or antigens in vitro stimulation, we report that during the early stage of S. japonicum infection, nonegg antigens trigger Th2 cell apoptosis via the granzyme B signal pathway, contributing to Th1 polarization, which is thought to be associated with worm clearance and severe schistosomiasis. Meanwhile, after the adult worms lay their eggs, the egg antigens trigger Th1 cell apoptosis via the caspase pathway, contributing to Th2 polarization, which is associated with mild pathology and enhanced survival of both worms and their hosts. Thus, our study suggests that S. japonicum antigen-induced Th1 and Th2 cell apoptosis involves the Th1/Th2 shift and favorites both hosts and parasites.
Collapse
|
38
|
Oliveira KC, Carvalho MLP, Venancio TM, Miyasato PA, Kawano T, DeMarco R, Verjovski-Almeida S. Identification of the Schistosoma mansoni TNF-alpha receptor gene and the effect of human TNF-alpha on the parasite gene expression profile. PLoS Negl Trop Dis 2009; 3:e556. [PMID: 19956564 PMCID: PMC2779652 DOI: 10.1371/journal.pntd.0000556] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 10/20/2009] [Indexed: 11/23/2022] Open
Abstract
Background Schistosoma mansoni is the major causative agent of schistosomiasis. The parasite takes advantage of host signals to complete its development in the human body. Tumor necrosis factor-alpha (TNF-α) is a human cytokine involved in skin inflammatory responses, and although its effect on the adult parasite's metabolism and egg-laying process has been previously described, a comprehensive assessment of the TNF-α pathway and its downstream molecular effects is lacking. Methodology/Principal Findings In the present work we describe a possible TNF-α receptor (TNFR) homolog gene in S. mansoni (SmTNFR). SmTNFR encodes a complete receptor sequence composed of 599 amino acids, and contains four cysteine-rich domains as described for TNFR members. Real-time RT-PCR experiments revealed that SmTNFR highest expression level is in cercariae, 3.5 (±0.7) times higher than in adult worms. Downstream members of the known human TNF-α pathway were identified by an in silico analysis, revealing a possible TNF-α signaling pathway in the parasite. In order to simulate parasite's exposure to human cytokine during penetration of the skin, schistosomula were exposed to human TNF-α just 3 h after cercariae-to-schistosomula in vitro transformation, and large-scale gene expression measurements were performed with microarrays. A total of 548 genes with significantly altered expression were detected, when compared to control parasites. In addition, treatment of adult worms with TNF-α caused a significantly altered expression of 1857 genes. Interestingly, the set of genes altered in adults is different from that of schistosomula, with 58 genes in common, representing 3% of altered genes in adults and 11% in 3 h-old early schistosomula. Conclusions/Significance We describe the possible molecular elements and targets involved in human TNF-α effect on S. mansoni, highlighting the mechanism by which recently transformed schistosomula may sense and respond to this host mediator at the site of cercarial penetration into the skin. Schistosoma mansoni is the major causative agent of schistosomiasis in the Americas. This parasite takes advantage of host signaling molecules such as cytokines and hormones to complete its development inside the host. Tumor necrosis factor-alpha (TNF-α) is one of the most important host cytokines involved in the inflammatory response. When cercariae, the infective stage, penetrates the human skin the release of TNF-α is started. In this work the authors describe the complete sequence of a possible TNF-α receptor in S. mansoni and detect that the receptor is most highly expressed in cercariae among all life cycle stages. Aiming to mimic the situation at the site of skin penetration, cercariae were mechanically transformed in vitro into schistosomula and exposed to human TNF-α. Exposure of early-developing schistosomula to the human hormone caused a large-scale change in the expression of parasite genes. Exposure of adult worms to human TNF-α caused gene expression changes as well, and the set of parasite altered genes in the adult parasite was different from that of schistosomula. This work increases the number of known signaling pathways of the parasite, and opens new perspectives into understanding the molecular components of TNF-α response as well as into possibly interfering with parasite–host interaction.
Collapse
Affiliation(s)
- Katia C. Oliveira
- Laboratory of Gene Expression in Eukaryotes, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Mariana L. P. Carvalho
- Laboratory of Gene Expression in Eukaryotes, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Thiago M. Venancio
- Laboratory of Gene Expression in Eukaryotes, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | - Toshie Kawano
- Laboratory of Parasitology, Instituto Butantan, São Paulo, Brazil
| | - Ricardo DeMarco
- Departamento de Física e Informática, Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | - Sergio Verjovski-Almeida
- Laboratory of Gene Expression in Eukaryotes, Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
39
|
Jurberg AD, Gonçalves T, Costa TA, de Mattos ACA, Pascarelli BM, de Manso PPA, Ribeiro-Alves M, Pelajo-Machado M, Peralta JM, Coelho PMZ, Lenzi HL. The embryonic development of Schistosoma mansoni eggs: proposal for a new staging system. Dev Genes Evol 2009; 219:219-34. [PMID: 19415326 DOI: 10.1007/s00427-009-0285-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 04/03/2009] [Indexed: 11/25/2022]
Abstract
Schistosomiasis is a water-borne parasitic illness caused by neoophoran trematodes of the genus Schistosoma. Using classical histological techniques and whole-mount preparations, the present work describes the embryonic development of Schistosoma mansoni eggs in the murine host and compares it with eggs maintained under in vitro conditions. Two pre-embryonic stages occur inside the female worm: the prezygotic stage is characterized by the release of mature oocytes from the female ovary until its fertilization. The zygotic stage encompasses the migration of the zygote through the ootype, where the eggshell is formed, to the uterus. Fully formed eggs are laid still undeveloped, without having suffered any cleavage. In the outside environment, eight embryonic stages can be defined: stage 1 refers to early cleavages and the beginning of yolk fusion. Stage 2 represents late cleavage, with the formation of a stereoblastula and the onset of outer envelope differentiation. Stage 3 is defined by the elongation of the embryonic primordium and the onset of inner envelope formation. At stage 4, the first organ primordia arise. During stages 5 to 7, tissue and organ differentiation occurs (neural mass, epidermis, terebratorium, musculature, and miracidial glands). Stage 7 is characterized by the nuclear condensation of neurons of the central neural mass. Stage 8 refers to the fully formed larva, presenting muscular contraction, cilia, and flame-cell beating. This staging system was compared to a previous classification and could underlie further studies on egg histoproteomics (morphological localizome). The differentiation of embryonic structures and their probable roles in granulomatogenesis are discussed herein.
Collapse
Affiliation(s)
- Arnon D Jurberg
- Laboratório de Patologia, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz, Pavilhão Gomes de Faria. Av. Brasil, 4365-Manguinhos, Rio de Janeiro 21040-900, RJ, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang X, Gobert GN, Feng X, Fu Z, Jin Y, Peng J, Lin J. Analysis of early hepatic stage schistosomula gene expression by subtractive expressed sequence tags library. Mol Biochem Parasitol 2009; 166:62-9. [PMID: 19428674 DOI: 10.1016/j.molbiopara.2009.02.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Revised: 02/24/2009] [Accepted: 02/25/2009] [Indexed: 01/12/2023]
Abstract
Schistosome parasites require a complex lifecycle requiring two hosts and aquatic phases of development. The schistosomula is a key phase of parasite development within the mammalian host, however relatively little is understood about the molecular processes underlying this stage. In this study 5723 subtractive expressed sequence tags (ESTs) were randomly selected from a 7 day hepatic schistosomula enriched library constructed using suppression subtractive hybridization method. Sequence analysis of these ESTs identified 1762 unique genes (contigs). Among them, 989 contigs were annotated with known genes, 311 contigs were homologous to established genes, 101 contigs were similar to established genes, 72 contigs were weakly similar to established genes and 289 sequences did not match any published sequences. Genes identified related to metabolism, cellular development, immune evasion and host-parasite interactions were identified as enriched in the hepatic schistosomula stage. The future identification of poorly annotated but stage-specific genes may potentially represent new drugs or vaccine targets, applicable for the future controlling of schistosomiasis.
Collapse
Affiliation(s)
- Xinzhi Wang
- Shanghai Institute of Veterinary Research, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, PR China
| | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The trematode parasites in the family Schistosomatidae (phylum Platyhelminthes) infect a wide range of vertebrates. Three species of the genus Schistosoma are of major medical importance. This unit deals exclusively with the parasite Schistosoma mansoni, which is the species most frequently maintained in the laboratory. Among the far-ranging investigations in the immunology of schistosomiasis are studies in vaccine development, immunopathology of granulomatous inflammation and fibrosis, eosinophil function, and in vivo regulation of T(H)1 and T(H)2 responses. This unit describes maintenance and collection procedures for various stages of the schistosome that have immunologic interest, including infection of mice with cercariae, collection of cercariae, preparation of in vitro-derived schistosomules and in vivo-derived schistosomules, and collection of adult worms and eggs. Included also are techniques for preparing soluble egg antigen (SEA), one of the more commonly used schistosome antigenic preparations. A discussion is given of the basic steps that are important in maintaining the snail intermediate host, and infecting the snails with schistosome miracidia. The unit deals exclusively with the parasite Schistosoma mansoni, which is the species most frequently maintained in the laboratory. Since part of the life cycle of all schistosomes involves a snail host, a description of proper maintenance for the snails is provided. Often, problems in experiments can be traced back to improper snail and parasite maintenance, or lack of attention to detail during mammalian exposure to the infective stage (cercaria) of the parasite.
Collapse
Affiliation(s)
- F Lewis
- Biomedical Research Institute, Rockville, Maryland, USA
| |
Collapse
|