1
|
Banerjee S, Mukhopadhyay S, Das A, Banerjee S, Bose S, Banerjee S, Casarcia N, Bishayee A. Preparation, characterisation, anticancer potential and safety evaluation of a soy lecithin phytosome delivery system loaded with constituents from Barleria lupulina. J Microencapsul 2025:1-21. [PMID: 39992133 DOI: 10.1080/02652048.2025.2467046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/10/2025] [Indexed: 02/25/2025]
Abstract
In this study, antineoplastic effects of a novel soy lecithin-based phytosome drug delivery system containing Barleria lupulina Lindl. extract (BLSP) was evaluated. BLSP was prepared using the thin-film hydration method and analysed using energy-dispersive X-ray spectroscopy, scanning electron microscopy, X-ray diffraction, and Zetasizer technique. Phytosomes showed a mean-diameter of 135 ± 0.29 nm, zeta potential of -56 ± 1.16 mV, and entrapment efficiency of 57.24 ± 0.12%. The drug release profiles exhibited a two-phase pattern with a protracted and sustained release after the first release. BLSP had a cytotoxic potential against MCF-7 breast and HeLa cervical cancers and demonstrated a concentration-dependent reduction of reactive oxygen species and mitochondrial membrane potential. BLSP caused upregulation of B-cell lymphoma-2-associated-X protein, caspase-8, caspase-9, and cluster of differentiation-95, and downregulation of B-cell lymphoma-2. The in vivo toxicity study showed the safety of BLSP. Overall, BLSP has demonstrated potential as a promising formulation for delivering B. lupulina phytoconstituents to treat breast and cervical cancer.
Collapse
Affiliation(s)
- Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol, India
| | - Shibangi Mukhopadhyay
- Department of Pharmaceutics, Gupta College of Technological Sciences, Asansol, India
| | - Avik Das
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, India
| | - Subhasis Banerjee
- Department of Pharmaceutical Technology, Eminent College of Pharmaceutical Technology, Kolkata, India
| | - Sankhadip Bose
- School of Pharmacy, The Neotia University, Sarisa, India
| | - Santanu Banerjee
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, India
| | - Nicolette Casarcia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, USA
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, USA
| |
Collapse
|
2
|
Namulondo J, Mulindwa J, Nyangiri OA, Egesa M, Noyes H, Matovu E. Gene expression changes in mammalian hosts during schistosomiasis: a review. AAS Open Res 2021. [DOI: 10.12688/aasopenres.13312.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Schistosomiasis affects over 250 million people worldwide with an estimated mortality of more than 200,000 deaths per year in sub-Saharan Africa. Efforts to control schistosomiasis in the affected areas have mainly relied on mass administration of praziquantel, which kills adult but not immature worms of all Schistosoma species. Mammalian hosts respond differently to Schistosoma infection with some being more susceptible than others, which is associated with risk factors such as sociodemographic, epidemiological, immunological and/or genetic. Host genetic factors play a major role in influencing molecular processes in response to schistosomiasis as shown in gene expression studies. These studies highlight gene profiles expressed at different time points of infection using model animals. Immune function related genes; cytokines (Th1 and Th17) are upregulated earlier in infection and Th2 upregulated later indicating a mixed Th1/Th2 response. However, Th1 response has been shown to be sustained in S. japonicum infection. Immune mediators such as matrix metalloproteinases (Mmps) and tissue inhibitors of matrix metalloproteinases (Timps) are expressed later in the infection and these are linked to wound healing and fibrosis. Downregulation of metabolic associated genes is recorded in later stages of infection. Most mammalian host gene expression studies have been done using rodent models, with fewer in larger hosts such as bovines and humans. The majority of these studies have focused on S. japonicum infections and less on S. haematobium and S. mansoni infections (the two species that cause most global infections). The few human schistosomiasis gene expression studies so far have focused on S. japonicum and S. haematobium infections and none on S. mansoni, as far as we are aware. This highlights a paucity of gene expression data in humans, specifically with S. mansoni infection. This data is important to understand the disease pathology, identify biomarkers, diagnostics and possible drug targets.
Collapse
|
3
|
Carson JP, Gobert GN. Modulation of the Host Immune Response by Schistosome Egg-Secreted Proteins Is a Critical Avenue of Host-Parasite Communication. Pathogens 2021; 10:863. [PMID: 34358013 PMCID: PMC8308880 DOI: 10.3390/pathogens10070863] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/21/2022] Open
Abstract
During a schistosome infection, the interactions that occur between the mammalian host and the parasite change rapidly once egg laying begins. Both juvenile and adult schistosomes adapt to indefinitely avoid the host immune system. In contrast, the survival of eggs relies on quickly traversing from the host. Following the commencement of egg laying, the host immune response undergoes a shift from a type 1 helper (Th1) inflammatory response to a type 2 helper (Th2) granulomatous response. This change is driven by immunomodulatory proteins within the egg excretory/secretory products (ESPs), which interact with host cells and alter their behaviour to promote egg translocation. However, in parallel, these ESPs also provoke the development of chronic schistosomiasis pathology. Recent studies using high-throughput proteomics have begun to characterise the components of schistosome egg ESPs, particularly those of Schistosoma mansoni, S. japonicum and S. haematobium. Future application of this knowledge may lead to the identification of proteins with novel immunomodulatory activity or pathological importance. However, efforts in this area are limited by a lack of in situ or in vivo functional characterisation of these proteins. This review will highlight the current knowledge of the content and demonstrated functions of schistosome egg ESPs.
Collapse
Affiliation(s)
| | - Geoffrey N. Gobert
- School of Biological Sciences, Queen’s University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, UK;
| |
Collapse
|
4
|
You H, Mayer JU, Johnston RL, Sivakumaran H, Ranasinghe S, Rivera V, Kondrashova O, Koufariotis LT, Du X, Driguez P, French JD, Waddell N, Duke MG, Ittiprasert W, Mann VH, Brindley PJ, Jones MK, McManus DP. CRISPR/Cas9-mediated genome editing of Schistosoma mansoni acetylcholinesterase. FASEB J 2021; 35:e21205. [PMID: 33337558 DOI: 10.1096/fj.202001745rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/16/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022]
Abstract
CRISPR/Cas9-mediated genome editing shows cogent potential for the genetic modification of helminth parasites. We report successful gene knock-in (KI) into the genome of the egg of Schistosoma mansoni by combining CRISPR/Cas9 with single-stranded oligodeoxynucleotides (ssODNs). We edited the acetylcholinesterase (AChE) gene of S. mansoni targeting two guide RNAs (gRNAs), X5 and X7, located on exon 5 and exon 7 of Smp_154600, respectively. Eggs recovered from livers of experimentally infected mice were transfected by electroporation with a CRISPR/Cas9-vector encoding gRNA X5 or X7 combining with/ without a ssODN donor. Next generation sequencing analysis of reads of amplicon libraries spanning targeted regions revealed that the major modifications induced by CRISPR/Cas9 in the eggs were generated by homology directed repair (HDR). Furthermore, soluble egg antigen from AChE-edited eggs exhibited markedly reduced AChE activity, indicative that programed Cas9 cleavage mutated the AChE gene. Following injection of AChE-edited schistosome eggs into the tail veins of mice, an significantly enhanced Th2 response involving IL-4, -5, -10, and-13 was detected in lung cells and splenocytes in mice injected with X5-KI eggs in comparison to control mice injected with unmutated eggs. A Th2-predominant response, with increased levels of IL-4, -13, and GATA3, also was induced by X5 KI eggs in small intestine-draining mesenteric lymph node cells when the gene-edited eggs were introduced into the subserosa of the ileum of the mice. These findings confirmed the potential and the utility of CRISPR/Cas9-mediated genome editing for functional genomics in schistosomes.
Collapse
Affiliation(s)
- Hong You
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | - Rebecca L Johnston
- Genetics & Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Haran Sivakumaran
- Genetics & Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Shiwanthi Ranasinghe
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Vanessa Rivera
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Olga Kondrashova
- Genetics & Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Lambros T Koufariotis
- Genetics & Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Xiaofeng Du
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Patrick Driguez
- King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
| | - Juliet D French
- Genetics & Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nicola Waddell
- Genetics & Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Mary G Duke
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Wannaporn Ittiprasert
- Department of Microbiology, Immunology & Tropical Medicine, & Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA
| | - Victoria H Mann
- Department of Microbiology, Immunology & Tropical Medicine, & Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA
| | - Paul J Brindley
- Department of Microbiology, Immunology & Tropical Medicine, & Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA
| | - Malcolm K Jones
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,School of Veterinary Science, The University of Queensland, Gatton, QLD, Australia
| | - Donald P McManus
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
5
|
Fasciola hepatica hijacks host macrophage miRNA machinery to modulate early innate immune responses. Sci Rep 2021; 11:6712. [PMID: 33762636 PMCID: PMC7990952 DOI: 10.1038/s41598-021-86125-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 03/08/2021] [Indexed: 01/31/2023] Open
Abstract
Fasciola hepatica, a global worm parasite of humans and their livestock, regulates host innate immune responses within hours of infection. Host macrophages, essential to the first-line defence mechanisms, are quickly restricted in their ability to initiate a classic protective pro-inflammatory immune response. We found that macrophages from infected animals are enriched with parasite-derived micro(mi)RNAs. The most abundant of these miRNAs, fhe-miR-125b, is released by the parasite via exosomes and is homologous to a mammalian miRNA, hsa-miR-125b, that is known to regulate the activation of pro-inflammatory M1 macrophages. We show that the parasite fhe-miR-125b loads onto the mammalian Argonaut protein (Ago-2) within macrophages during infection and, therefore, propose that it mimics host miR-125b to negatively regulate the production of inflammatory cytokines. The hijacking of the miRNA machinery controlling innate cell function could be a fundamental mechanism by which worm parasites disarm the early immune responses of their host to ensure successful infection.
Collapse
|
6
|
Live imaging of collagen deposition during experimental hepatic schistosomiasis and recovery: a view on a dynamic process. J Transl Med 2019; 99:231-243. [PMID: 30401957 DOI: 10.1038/s41374-018-0154-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 01/01/2023] Open
Abstract
Hepatic fibrosis is the central cause of chronic clinical pathology resulting from infection by the blood flukes Schistosoma japonicum or S. mansoni. Much has been elucidated regarding the molecular, cellular and immunological responses that correspond to the formation of the granulomatous response to trapped schistosome eggs. A central feature of this Th2 response is the deposition of collagen around the periphery of the granuloma. To date, traditional histology and transcriptional methods have been used to quantify the deposition of collagen and to monitor the formation of the hepatic granuloma during experimental animal models of schistosomiasis. We have investigated the dynamic nature of granuloma formation through the use of a transgenic mouse model (B6.Collagen 1(A) luciferase mice (B6.Coll 1A-luc+)). With this model and whole-animal bioluminescence imaging, we followed the deposition of collagen during an active schistosome infection with Chinese and Philippines geographical strains of S. japonicum and after clearance of the adult parasites by the drug praziquantel. Individual mice were re-imaged over the time course to provide robust real-time quantitation of the development of chronic fibrotic disease. This model provides an improved method to follow the course of hepatic schistosomiasis-induced hepatic pathology and effectively supports the current dogma of the formation of hepatic fibrosis, originally elucidated from static traditional histology. This study demonstrates the first use of the B6.Coll 1A-luc+ mouse to monitor the dynamics of disease development and the treatment of pathogen-induced infection with the underlying pathology of fibrosis.
Collapse
|
7
|
Chan JD, Day TA, Marchant JS. Coalescing beneficial host and deleterious antiparasitic actions as an antischistosomal strategy. eLife 2018; 7:35755. [PMID: 30059006 PMCID: PMC6095690 DOI: 10.7554/elife.35755] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/24/2018] [Indexed: 12/28/2022] Open
Abstract
Conventional approaches for antiparasitic drug discovery center upon discovering selective agents that adversely impact parasites with minimal host side effects. Here, we show that agents with a broad polypharmacology, often considered ‘dirtier’ drugs, can have unique efficacy if they combine deleterious effects on the parasite with beneficial actions in the host. This principle is evidenced through a screen for drugs to treat schistosomiasis, a parasitic flatworm disease that impacts over 230 million people. A target-based screen of a Schistosoma serotoninergic G protein coupled receptor yielded the potent agonist, ergotamine, which disrupted worm movement. In vivo, ergotamine decreased mortality, parasite load and intestinal egg counts but also uniquely reduced organ pathology through engagement of host GPCRs that repressed hepatic stellate cell activation, inflammatory damage and fibrosis. The unique ability of ergotamine to engage both host and parasite GPCRs evidences a future strategy for anthelmintic drug design that coalesces deleterious antiparasitic activity with beneficial host effects. More than 200 million people worldwide are infected with parasitic worms that cause the disease schistosomiasis. Most cases occur in sub-Saharan Africa. Long-term infections can damage organs, and children who are affected may suffer delayed growth and learning difficulties. Despite its significant health and economic impact, schistosomiasis is still considered a ‘neglected’ tropical disease. This means there has not been adequate investment into developing new treatments or cures. A drug called praziquantel is currently the only treatment for schistosomiasis. However, the drug has unpleasant side effects, cannot cure all infected individuals, and there is a concern that worms may develop resistance to its effects. This means there is an urgent need to develop new therapies. One possible approach would be to develop drugs that interfere with the worm’s ability to move. Chan et al. screened thousands of existing chemicals for interactions with a protein that is known to control how the worms move. A drug called ergotamine, which is currently used to treat migraines, strongly interacted with the protein. Treating infected mice with ergotamine eliminated the parasites and reduced the organ damage caused by the infection. Praziquantel also reduced the number of parasites in the mice but it did not prevent organ damage. The results presented by Chan et al. show that a single drug can interact with targets in both the worm and the animals it infects. Searching for drugs that have this dual effect may help to develop more effective treatments for schistosomiasis and other diseases caused by parasites. Ergotamine itself is unlikely to be used to treat people for schistosomiasis because of the side effects produced when using it repeatedly. However, these findings will help researchers identify and develop safer drugs with similar benefits.
Collapse
Affiliation(s)
- John D Chan
- Department of Biomedical Sciences, Iowa State University, Ames, United States
| | - Timothy A Day
- Department of Biomedical Sciences, Iowa State University, Ames, United States
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
8
|
Sanchez MC, Krasnec KV, Parra AS, von Cabanlong C, Gobert GN, Umylny B, Cupit PM, Cunningham C. Effect of praziquantel on the differential expression of mouse hepatic genes and parasite ATP binding cassette transporter gene family members during Schistosoma mansoni infection. PLoS Negl Trop Dis 2017. [PMID: 28650976 PMCID: PMC5501684 DOI: 10.1371/journal.pntd.0005691] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Schistosomiasis is a chronic parasitic disease caused by sexually dimorphic blood flukes of the genus Schistosoma. Praziquantel (PZQ) is the only drug widely available to treat the disease but does not kill juvenile parasites. Here we report the use of next generation sequencing to study the transcriptional effect of PZQ on murine hepatic inflammatory, immune and fibrotic responses to Schistosoma mansoni worms and eggs. An initial T helper cell 1 (Th1) response is induced against schistosomes in mice treated with drug vehicle (Vh) around the time egg laying begins, followed by a T helper cell 2 (Th2) response and the induction of genes whose action leads to granuloma formation and fibrosis. When PZQ is administered at this time, there is a significant reduction in egg burden yet the hepatic Th1, Th2 and fibrotic responses are still observed in the absence of granuloma formation suggesting some degree of gene regulation may be induced by antigens released from the dying adult worms. Quantitative real-time PCR was used to examine the relative expression of 16 juvenile and adult S. mansoni genes during infection and their response to Vh and PZQ treatment in vivo. While the response of stress genes in adult parasites suggests the worms were alive immediately following exposure to PZQ, they were unable to induce transcription of any of the 9 genes encoding ATP-binding cassette (ABC) transporters tested. In contrast, juvenile schistosomes were able to significantly induce the activities of ABCB, C and G family members, underscoring the possibility that these efflux systems play a major role in drug resistance.
Collapse
Affiliation(s)
- Melissa C. Sanchez
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Katina V. Krasnec
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Amalia S. Parra
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Christian von Cabanlong
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Geoffrey N. Gobert
- School of Biological Sciences, Queen’s University, Belfast, United Kingdom
| | - Boris Umylny
- National Center for Genome Resources, Santa Fe, New Mexico, United States of America
| | - Pauline M. Cupit
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Charles Cunningham
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
9
|
The NIH-NIAID Schistosomiasis Resource Center at the Biomedical Research Institute: Molecular Redux. PLoS Negl Trop Dis 2016; 10:e0005022. [PMID: 27764112 PMCID: PMC5072641 DOI: 10.1371/journal.pntd.0005022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Schistosomiasis remains a health burden in many parts of the world. The complex life cycle of Schistosoma parasites and the economic and societal conditions present in endemic areas make the prospect of eradication unlikely in the foreseeable future. Continued and vigorous research efforts must therefore be directed at this disease, particularly since only a single World Health Organization (WHO)-approved drug is available for treatment. The National Institutes of Health (NIH)–National Institute of Allergy and Infectious Diseases (NIAID) Schistosomiasis Resource Center (SRC) at the Biomedical Research Institute provides investigators with the critical raw materials needed to carry out this important research. The SRC makes available, free of charge (including international shipping costs), not only infected host organisms but also a wide array of molecular reagents derived from all life stages of each of the three main human schistosome parasites. As the field of schistosomiasis research rapidly advances, it is likely to become increasingly reliant on omics, transgenics, epigenetics, and microbiome-related research approaches. The SRC has and will continue to monitor and contribute to advances in the field in order to support these research efforts with an expanding array of molecular reagents. In addition to providing investigators with source materials, the SRC has expanded its educational mission by offering a molecular techniques training course and has recently organized an international schistosomiasis-focused meeting. This review provides an overview of the materials and services that are available at the SRC for schistosomiasis researchers, with a focus on updates that have occurred since the original overview in 2008.
Collapse
|
10
|
Alves CC, Araujo N, Cassali GD, Fonseca CT. Parasitological, Pathological, and Immunological Parameters Associated with Schistosoma mansoni Infection and Reinfection in BALB/c AND C57BL/6 Mice. J Parasitol 2016; 102:336-41. [DOI: 10.1645/14-664] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Clarice Carvalho Alves
- Laboratório de Esquistossomose, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz. Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, Minas Gerais, Brazil
| | - Neusa Araujo
- Laboratório de Esquistossomose, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz. Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, Minas Gerais, Brazil
| | - Geovanni Dantas Cassali
- Laboratório de Esquistossomose, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz. Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, Minas Gerais, Brazil
| | - Cristina Toscano Fonseca
- Laboratório de Esquistossomose, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz. Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
11
|
Zhou G, Stevenson MM, Geary TG, Xia J. Comprehensive Transcriptome Meta-analysis to Characterize Host Immune Responses in Helminth Infections. PLoS Negl Trop Dis 2016; 10:e0004624. [PMID: 27058578 PMCID: PMC4826001 DOI: 10.1371/journal.pntd.0004624] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/21/2016] [Indexed: 12/19/2022] Open
Abstract
Helminth infections affect more than a third of the world’s population. Despite very broad phylogenetic differences among helminth parasite species, a systemic Th2 host immune response is typically associated with long-term helminth infections, also known as the “helminth effect”. Many investigations have been carried out to study host gene expression profiles during helminth infections. The objective of this study is to determine if there is a common transcriptomic signature characteristic of the helminth effect across multiple helminth species and tissue types. To this end, we performed a comprehensive meta-analysis of publicly available gene expression datasets. After data processing and adjusting for study-specific effects, we identified ~700 differentially expressed genes that are changed consistently during helminth infections. Functional enrichment analyses indicate that upregulated genes are predominantly involved in various immune functions, including immunomodulation, immune signaling, inflammation, pathogen recognition and antigen presentation. Down-regulated genes are mainly involved in metabolic process, with only a few of them are involved in immune regulation. This common immune gene signature confirms previous observations and indicates that the helminth effect is robust across different parasite species as well as host tissue types. To the best of our knowledge, this study is the first comprehensive meta-analysis of host transcriptome profiles during helminth infections. Many studies have been conducted to understand the immune modulatory effects in helminth infections. To determine whether there is a common transcriptomic signature characteristic of the helminth effect, we performed a comprehensive meta-analysis of publicly available gene expression datasets. The results revealed a distinct pattern of gene expression that is consistent across multiple helminth species and host tissue types, with upregulated genes dominated by those involved in immune regulation, Th2 immunity and inflammatory responses.
Collapse
Affiliation(s)
- Guangyan Zhou
- Institute of Parasitology, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Centre for Host-Parasite Interactions, McGill University, Sainte Anne de Bellevue, Quebec, Canada
| | - Mary M. Stevenson
- Centre for Host-Parasite Interactions, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Departments of Medicine and Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Timothy G. Geary
- Institute of Parasitology, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Centre for Host-Parasite Interactions, McGill University, Sainte Anne de Bellevue, Quebec, Canada
| | - Jianguo Xia
- Institute of Parasitology, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Centre for Host-Parasite Interactions, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- Department of Animal Science, McGill University, Sainte Anne de Bellevue, Quebec, Canada
- * E-mail:
| |
Collapse
|
12
|
Chuah C, Jones MK, McManus DP, Nawaratna SK, Burke ML, Owen HC, Ramm GA, Gobert GN. Characterising granuloma regression and liver recovery in a murine model of schistosomiasis japonica. Int J Parasitol 2016; 46:239-52. [PMID: 26812024 DOI: 10.1016/j.ijpara.2015.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 02/07/2023]
Abstract
For hepatic schistosomiasis the egg-induced granulomatous response and the development of extensive fibrosis are the main pathologies. We used a Schistosoma japonicum-infected mouse model to characterise the multi-cellular pathways associated with the recovery from hepatic fibrosis following clearance of the infection with the anti-schistosomal drug, praziquantel. In the recovering liver splenomegaly, granuloma density and liver fibrosis were all reduced. Inflammatory cell infiltration into the liver was evident, and the numbers of neutrophils, eosinophils and macrophages were significantly decreased. Transcriptomic analysis revealed the up-regulation of fatty acid metabolism genes and the identification of Peroxisome proliferator activated receptor alpha as the upstream regulator of liver recovery. The aryl hydrocarbon receptor signalling pathway which regulates xenobiotic metabolism was also differentially up-regulated. These findings provide a better understanding of the mechanisms associated with the regression of hepatic schistosomiasis.
Collapse
Affiliation(s)
- Candy Chuah
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia; School of Veterinary Sciences, The University of Queensland, Gatton, Qld 4343, Australia; School of Medical Sciences, Universiti Sains Malaysia, 16150 Kelantan, Malaysia
| | - Malcolm K Jones
- School of Veterinary Sciences, The University of Queensland, Gatton, Qld 4343, Australia
| | - Donald P McManus
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia
| | | | - Melissa L Burke
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia
| | - Helen C Owen
- School of Veterinary Sciences, The University of Queensland, Gatton, Qld 4343, Australia
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia
| | - Geoffrey N Gobert
- QIMR Berghofer Medical Research Institute, Brisbane, Qld 4006, Australia.
| |
Collapse
|
13
|
Driguez P, McManus DP, Gobert GN. Clinical implications of recent findings in schistosome proteomics. Expert Rev Proteomics 2015; 13:19-33. [PMID: 26558506 DOI: 10.1586/14789450.2016.1116390] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Schistosomiasis is a neglected tropical disease of clinical significance that, despite years of research, still requires an effective vaccine and improved diagnostics for surveillance, control and potential elimination. Furthermore, the causes of host pathology during schistosomiasis are still not completely understood. The recent sequencing of the genomes of the three key schistosome species has enabled the discovery of many new possible vaccine and drug targets, as well as diagnostic biomarkers, using high-throughput and sensitive proteomics methods. This review focuses on the literature of the last 5 years that has reported on the use of proteomics to both better understand the biology of the schistosome parasites and the disease they cause in definitive mammalian hosts.
Collapse
Affiliation(s)
- Patrick Driguez
- a QIMR Berghofer Medical Research Institute, Infectious Disease Division , Brisbane , Queensland , Australia
| | - Donald P McManus
- a QIMR Berghofer Medical Research Institute, Infectious Disease Division , Brisbane , Queensland , Australia
| | - Geoffrey N Gobert
- a QIMR Berghofer Medical Research Institute, Infectious Disease Division , Brisbane , Queensland , Australia
| |
Collapse
|
14
|
Circulating miRNAs: Potential Novel Biomarkers for Hepatopathology Progression and Diagnosis of Schistosomiasis Japonica in Two Murine Models. PLoS Negl Trop Dis 2015; 9:e0003965. [PMID: 26230095 PMCID: PMC4521869 DOI: 10.1371/journal.pntd.0003965] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 07/08/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Schistosomiasis remains a major public health issue, with an estimated 230 million people infected worldwide. Novel tools for early diagnosis and surveillance of schistosomiasis are currently needed. Elevated levels of circulating microRNAs (miRNAs) are commonly associated with the initiation and progression of human disease pathology. Hence, serum miRNAs are emerging as promising biomarkers for the diagnosis of a variety of human diseases. This study investigated circulating host miRNAs commonly associated with liver diseases and schistosome parasite-derived miRNAs during the progression of hepatic schistosomiasis japonica in two murine models. METHODOLOGY/PRINCIPAL FINDINGS Two mouse strains (C57BL/6 and BALB/c) were infected with a low dosage of Schistosoma japonicum cercariae. The dynamic patterns of hepatopathology, the serum levels of liver injury-related enzymes and the serum circulating miRNAs (both host and parasite-derived) levels were then assessed in the progression of schistosomiasis japonica. For the first time, an inverse correlation between the severity of hepatocyte necrosis and the level of liver fibrosis was revealed during S. japonicum infection in BALB/c, but not in C57BL/6 mice. The inconsistent levels of the host circulating miRNAs, miR-122, miR-21 and miR-34a in serum were confirmed in the two murine models during infection, which limits their potential value as individual diagnostic biomarkers for schistosomiasis. However, their serum levels in combination may serve as a novel biomarker to mirror the hepatic immune responses induced in the mammalian host during schistosome infection and the degree of hepatopathology. Further, two circulating parasite-specific miRNAs, sja-miR-277 and sja-miR-3479-3p, were shown to have potential as diagnostic markers for schistosomiasis japonica. CONCLUSIONS/SIGNIFICANCE We provide the first evidence for the potential of utilizing circulating host miRNAs to indicate different immune responses and the severity of hepatopathology outcomes induced in two murine strains infected with S. japonicum. This study also establishes a basis for the early and cell-free diagnosis of schistosomiasis by targeting circulating schistosome parasite-derived miRNAs.
Collapse
|
15
|
Transcriptional profiling of chronic clinical hepatic schistosomiasis japonica indicates reduced metabolism and immune responses. Parasitology 2015. [PMID: 26216487 DOI: 10.1017/s0031182015000682] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Schistosomiasis is a significant cause of human morbidity and mortality. We performed a genome-wide transcriptional survey of liver biopsies obtained from Chinese patients with chronic schistosomiasis only, or chronic schistosomiasis with a current or past history of viral hepatitis B. Both disease groups were compared with patients with no prior history or indicators of any liver disease. Analysis showed in the main, downregulation in gene expression, particularly those involved in signal transduction via EIF2 signalling and mTOR signalling, as were genes associated with cellular remodelling. Focusing on immune associated pathways, genes were generally downregulated. However, a set of three genes associated with granulocytes, MMP7, CLDN7, CXCL6 were upregulated. Differential gene profiles unique to schistosomiasis included the gene Granulin which was decreased despite being generally considered a marker for liver disease, and IGBP2 which is associated with increased liver size, and was the most upregulated gene in schistosomiasis only patients, all of which presented with hepatomegaly. The unique features of gene expression, in conjunction with previous reports in the murine model of the cellular composition of granulomas, granuloma formation and recovery, provide an increased understanding of the molecular immunopathology and general physiological processes underlying hepatic schistosomiasis.
Collapse
|
16
|
Gobert GN, Nawaratna SK, Harvie M, Ramm GA, McManus DP. An ex vivo model for studying hepatic schistosomiasis and the effect of released protein from dying eggs. PLoS Negl Trop Dis 2015; 9:e0003760. [PMID: 25965781 PMCID: PMC4428699 DOI: 10.1371/journal.pntd.0003760] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/14/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND We report the use of an ex vivo precision cut liver slice (PCLS) mouse model for studying hepatic schistosomiasis. In this system, liver tissue is unfixed, unfrozen, and alive for maintenance in culture and subsequent molecular analysis. METHODS AND FINDINGS Using thick naive mouse liver tissue and sterile culture conditions, the addition of soluble egg antigen (SEA) derived from Schistosoma japonicum eggs, followed 4, 24 and 48 hrs time points. Tissue was collected for transcriptional analysis and supernatants collected to quantitate liver enzymes, cytokines and chemokines. No significant hepatotoxicity was demonstrated by supernatant liver enzymes due to the presence of SEA. A proinflammatory response was observed both at the transcriptional level and at the protein level by cytokine and chemokine bead assay. Key genes observed elevated transcription in response to the addition of SEA included: IL1-α and IL1-β, IL6, all associated with inflammation. The recruitment of antigen presenting cells was reflected in increases in transcription of CD40, CCL4 and CSF1. Indications of tissue remodeling were seen in elevated gene expression of various Matrix MetalloProteinases (MMP3, 9, 10, 13) and delayed increases in TIMP1. Collagen deposition was significantly reduced in the presence of SEA as shown in COL1A1 expression by qPCR after 24 hrs culture. Cytokine and chemokine analysis of the culture supernatants confirmed the elevation of proteins including IL6, CCL3, CCL4 and CXCL5. CONCLUSIONS This ex vivo model system for the synchronised delivery of parasite antigen to liver tissue provides an insight into the early phase of hepatic schistosomiasis, corresponding with the release of soluble proteins from dying schistosome eggs.
Collapse
Affiliation(s)
- Geoffrey N. Gobert
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- * E-mail:
| | | | - Marina Harvie
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Grant A. Ramm
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Donald P. McManus
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| |
Collapse
|
17
|
Flórez-Vargas O, Bramhall M, Noyes H, Cruickshank S, Stevens R, Brass A. The quality of methods reporting in parasitology experiments. PLoS One 2014; 9:e101131. [PMID: 25076044 PMCID: PMC4116335 DOI: 10.1371/journal.pone.0101131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 06/03/2014] [Indexed: 12/23/2022] Open
Abstract
There is a growing concern both inside and outside the scientific community over the lack of reproducibility of experiments. The depth and detail of reported methods are critical to the reproducibility of findings, but also for making it possible to compare and integrate data from different studies. In this study, we evaluated in detail the methods reporting in a comprehensive set of trypanosomiasis experiments that should enable valid reproduction, integration and comparison of research findings. We evaluated a subset of other parasitic (Leishmania, Toxoplasma, Plasmodium, Trichuris and Schistosoma) and non-parasitic (Mycobacterium) experimental infections in order to compare the quality of method reporting more generally. A systematic review using PubMed (2000-2012) of all publications describing gene expression in cells and animals infected with Trypanosoma spp was undertaken based on PRISMA guidelines; 23 papers were identified and included. We defined a checklist of essential parameters that should be reported and have scored the number of those parameters that are reported for each publication. Bibliometric parameters (impact factor, citations and h-index) were used to look for association between Journal and Author status and the quality of method reporting. Trichuriasis experiments achieved the highest scores and included the only paper to score 100% in all criteria. The mean of scores achieved by Trypanosoma articles through the checklist was 65.5% (range 32-90%). Bibliometric parameters were not correlated with the quality of method reporting (Spearman's rank correlation coefficient <-0.5; p>0.05). Our results indicate that the quality of methods reporting in experimental parasitology is a cause for concern and it has not improved over time, despite there being evidence that most of the assessed parameters do influence the results. We propose that our set of parameters be used as guidelines to improve the quality of the reporting of experimental infection models as a pre-requisite for integrating and comparing sets of data.
Collapse
Affiliation(s)
- Oscar Flórez-Vargas
- Bio-health Informatics Group, School of Computer Science, University of Manchester, Manchester, United Kingdom
| | - Michael Bramhall
- Bio-health Informatics Group, School of Computer Science, University of Manchester, Manchester, United Kingdom
| | - Harry Noyes
- School of Biological Science, University of Liverpool, Liverpool, United Kingdom
| | - Sheena Cruickshank
- Manchester Immunology Group, Faculty of Life Science, University of Manchester, Manchester, United Kingdom
| | - Robert Stevens
- Bio-health Informatics Group, School of Computer Science, University of Manchester, Manchester, United Kingdom
| | - Andy Brass
- Bio-health Informatics Group, School of Computer Science, University of Manchester, Manchester, United Kingdom
- Manchester Immunology Group, Faculty of Life Science, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
18
|
Gobert GN, You H, McManus DP. Gaining biological perspectives from schistosome genomes. Mol Biochem Parasitol 2014; 196:21-8. [PMID: 25076011 DOI: 10.1016/j.molbiopara.2014.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/14/2014] [Accepted: 07/17/2014] [Indexed: 02/06/2023]
Abstract
Characterization of the genomic basis underlying schistosome biology is an important strategy for the development of future treatments and interventions. Genomic sequence is now available for the three major clinically relevant schistosome species, Schistosoma mansoni, S. japonicum and S. haematobium, and this information represents an invaluable resource for the future control of human schistosomiasis. The identification of a biologically important, but distinct from the host, schistosome gene product is the ultimate goal for many research groups. While the initial elucidation of the genome of an organism is critical for most biological research, continued improvement or curation of the genome construction should be an ongoing priority. In this review we will discuss prominent recent findings utilizing a systems approach to schistosome biology, as well as the increased use of interference RNA (RNAi). Both of these research strategies are aiming to place parasite genes into a more meaningful biological perspective.
Collapse
Affiliation(s)
- Geoffrey N Gobert
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | - Hong You
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Donald P McManus
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
19
|
Chuah C, Jones MK, Burke ML, McManus DP, Owen HC, Gobert GN. Defining a pro-inflammatory neutrophil phenotype in response to schistosome eggs. Cell Microbiol 2014; 16:1666-77. [PMID: 24898449 DOI: 10.1111/cmi.12316] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/22/2014] [Indexed: 12/21/2022]
Abstract
Neutrophils contribute to the pathological processes of a number of inflammatory disorders, including rheumatoid arthritis, sepsis and cystic fibrosis. Neutrophils also play prominent roles in schistosomiasis japonica liver fibrosis, being central mediators of inflammation following granuloma formation. In this study, we investigated the interaction between Schistosoma japonicum eggs and neutrophils, and the effect of eggs on the inflammatory phenotype of neutrophils. Our results showed significant upregulated expression of pro-inflammatory cytokines (IL-1α, IL-1β and IL-8) and chemokines (CCL3, CCL4 and CXCL2) in neutrophils after 4 h in vitro stimulation with S. japonicum eggs. Furthermore, mitochondrial DNA was released by stimulated neutrophils, and induced the production of matrix metalloproteinase 9 (MMP-9), a protease involved in inflammation and associated tissue destruction. We also found that intact live eggs and isolated soluble egg antigen (SEA) triggered the release of neutrophil extracellular traps (NETs), but, unlike those reported in bacterial or fungal infection, NETs did not kill schistosome eggs in vitro. Together these show that S. japonicum eggs can induce the inflammatory phenotype of neutrophils, and further our understanding of the host-parasite interplay that takes place within the in vivo microenvironment of schistosome-induced granuloma. These findings represent novel findings in a metazoan parasite, and confirm characteristics of NETs that have until now, only been observed in response to protozoan pathogens.
Collapse
Affiliation(s)
- Candy Chuah
- Parasite Cell Biology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Qld, 4006, Australia; School of Veterinary Sciences, The University of Queensland, Gatton, Qld, 4343, Australia; School of Medical Sciences, Universiti Sains Malaysia, 16150, Kelantan, Malaysia
| | | | | | | | | | | |
Collapse
|
20
|
The effect of recombinant sTGFβ1RII and sIL13Rα2 receptor proteins on schistosomiasis japonica, hepatic fibrosis and signal transduction in a mouse model of schistosome disease. Exp Parasitol 2014; 142:17-26. [PMID: 24746639 DOI: 10.1016/j.exppara.2014.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 03/11/2014] [Accepted: 04/06/2014] [Indexed: 11/20/2022]
Abstract
This study was designed to investigate the effect of recombinant sTGFβ1RII and sIL13Rα2 receptor proteins on schistosomiasis japonica, hepatic fibrosis and the expression of SMAD3 and STAT6. The proteins sTGFβ1RII and sIL13Rα2 were expressed in Escherichiacoli, purified using affinity chromatography and characterized by Western blotting. Female BALB/C mice (48) were randomly divided into eight groups and infected with Schistosoma japonicum. Five weeks after infection, test groups were injected with the recombinant proteins at different doses. Eight weeks after infection, lung and hepatic tissue samples were obtained and stained with hematoxylin and eosin (HE) and Masson's trichrome. Immunohistochemical staining was used to detect the expression of SMAD3 and STAT6. The recombinant proteins sTGFβ1RII and sIL13Rα2 were successfully expressed, purified, and characterized. The granuloma area, hepatic hydroxyproline (HYP) level and hepatic fibrosis of the protein therapeutic groups were significantly smaller than those of the positive control group (P<0.01). Treatment with sTGFβ1RII was more effective when the protein was administered for 4weeks rather than 2 (P<0.01). Hepatic fibrosis in the groups using a low dose of protein sTGFβ1 was lower that of the combination group (P<0.05). The expression level of STAT6 was significantly lower in groups treated with sIL13Rα2 than in groups not treated with the protein (P<0.01). The recombinant proteins TGFβ1RII and sIL13Rα2 were able to decrease granuloma area and hepatic fibrosis in schistosomiasis japonica, and also reduced the expression of the signal transduction proteins SMAD3 and STAT6. The proteins were more effective when used in combination than when applied singly.
Collapse
|
21
|
Chuah C, Jones MK, Burke ML, McManus DP, Gobert GN. Cellular and chemokine-mediated regulation in schistosome-induced hepatic pathology. Trends Parasitol 2014; 30:141-50. [PMID: 24433721 DOI: 10.1016/j.pt.2013.12.009] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 02/06/2023]
Abstract
In hepatic schistosomiasis, pathology arises when schistosome eggs become lodged in the host liver, evoking an interleukin 4 (IL-4)- and IL-13-mediated dominant CD4(+) Th2 immune response. This response leads to the development of granulomas and fibrosis, with eosinophils, neutrophils, macrophages, hepatic stellate cells, and lymphocytes all identified as major cellular contributors to these events. This review outlines the cellular and molecular mechanisms of hepatic schistosomiasis, with an emphasis on the major cellular components and their release of chemokines. The differences between Schistosoma mansoni- and Schistosoma japonicum-induced hepatic granuloma are also discussed. This comprehensive overview of the processes associated with hepatic schistosomiasis may provide new insights into improved treatment for both schistosomiasis and other granulofibrotic diseases.
Collapse
Affiliation(s)
- Candy Chuah
- Parasite Cell Biology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, 4006, Australia; School of Veterinary Sciences, The University of Queensland, Gatton, 4343, Australia; School of Medical Sciences, Universiti Sains Malaysia, 16150, Kelantan, Malaysia
| | - Malcolm K Jones
- Parasite Cell Biology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, 4006, Australia; School of Veterinary Sciences, The University of Queensland, Gatton, 4343, Australia
| | - Melissa L Burke
- Division of Mycobacterial Research, National Institute for Medical Research, London, NW7 1AA, UK
| | - Donald P McManus
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, 4006, Australia
| | - Geoffrey N Gobert
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, 4006, Australia.
| |
Collapse
|
22
|
A study of immunomodulatory genes responses to macrophages of Schistosoma japonicum infection during different stages by microarray analysis. Acta Trop 2013; 127:251-60. [PMID: 23732117 DOI: 10.1016/j.actatropica.2013.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 05/15/2013] [Accepted: 05/25/2013] [Indexed: 01/08/2023]
Abstract
Macrophages initiate, modulate, and also serve as final effector cells in immune responses during the course of schistosomal infections. In this study, we investigated the gene expression profile and functional changes of macrophages in immune responses against the Schistosoma japonicum by microarray analysis. Hierarchical clustering analysis demonstrated that a significant switch in gene transformation associated with a type-1 response and linked with a type-2 cytokine phenotype occurs between 4.5 and 8 weeks post-infection. Moreover, the gene profiles at 3 later time-points following egg challenge were similar in complexity and magnitude. The data also showed that there were mostly inhibition of gene expression related TLR, IFN, MHC and TNFrsf at the switch between 4.5 and 8 weeks post-infection, It is suggested that these immunomodulatory genes may be down-regulated in defense against S. japonicum eggs and granuloma pathology. The induction of alternatively activated macrophage (AAMϕ) was important for dampening the inflammation in hepatic granulomas and contributing to a decrease in cytotoxicity. The gene expressions involved in repair/remodeling during liver fibrosis were also observed after egg production. Understanding the immune mechanisms associated with parasitic resistance, pathology of parasite infection, and parasite growth will provide useful insight on host-schistosome interactions and for the control of schistosomiasis.
Collapse
|
23
|
Krementsov DN, Wall EH, Martin RA, Subramanian M, Noubade R, Rio RD, Mawe GM, Bond JP, Poynter ME, Blankenhorn EP, Teuscher C. Histamine H(3) receptor integrates peripheral inflammatory signals in the neurogenic control of immune responses and autoimmune disease susceptibility. PLoS One 2013; 8:e62743. [PMID: 23894272 PMCID: PMC3718788 DOI: 10.1371/journal.pone.0062743] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/23/2013] [Indexed: 12/15/2022] Open
Abstract
Histamine H3 receptor (Hrh3/H3R) is primarily expressed by neurons in the central nervous system (CNS) where it functions as a presynaptic inhibitory autoreceptor and heteroreceptor. Previously, we identified an H3R-mediated central component in susceptibility to experimental allergic encephalomyelitis (EAE), the principal autoimmune model of multiple sclerosis (MS), related to neurogenic control of blood brain barrier permeability and peripheral T cell effector responses. Furthermore, we identified Hrh3 as a positional candidate for the EAE susceptibility locus Eae8. Here, we characterize Hrh3 polymorphisms between EAE-susceptible and resistant SJL and B10.S mice, respectively, and show that Hrh3 isoform expression in the CNS is differentially regulated by acute peripheral inflammatory stimuli in an allele-specific fashion. Next, we show that Hrh3 is not expressed in any subpopulations of the immune compartment, and that secondary lymphoid tissue is anatomically poised to be regulated by central H3R signaling. Accordingly, using transcriptome analysis, we show that, inflammatory stimuli elicit unique transcriptional profiles in the lymph nodes of H3RKO mice compared to WT mice, which is indicative of negative regulation of peripheral immune responses by central H3R signaling. These results further support a functional link between the neurogenic control of T cell responses and susceptibility to CNS autoimmune disease coincident with acute and/or chronic peripheral inflammation. Pharmacological targeting of H3R may therefore be useful in preventing the development and formation of new lesions in MS, thereby limiting disease progression.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Central Nervous System/immunology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Gene Expression Regulation
- Genetic Predisposition to Disease/genetics
- Hematopoiesis/genetics
- Hematopoiesis/immunology
- Humans
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/pathology
- Intracellular Space/metabolism
- Lymph Nodes/immunology
- Male
- Mice
- Molecular Sequence Data
- Polymorphism, Single Nucleotide
- Protein Isoforms/chemistry
- Protein Isoforms/genetics
- Protein Structure, Tertiary
- Receptors, Histamine H3/chemistry
- Receptors, Histamine H3/genetics
- Signal Transduction/genetics
- Signal Transduction/immunology
Collapse
Affiliation(s)
- Dimitry N. Krementsov
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
| | - Emma H. Wall
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
| | - Rebecca A. Martin
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
| | - Meenakumari Subramanian
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
| | - Rajkumar Noubade
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
| | - Roxana Del Rio
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
| | - Gary M. Mawe
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
| | - Jeffrey P. Bond
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
| | - Matthew E. Poynter
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
| | - Elizabeth P. Blankenhorn
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Cory Teuscher
- Department of Medicine, Immunobiology Program, University of Vermont, Burlington, Vermont, United States of America
- Department of Pathology, University of Vermont, Burlington, Vermont, United States of America
- * E-mail:
| |
Collapse
|
24
|
Cytokine and chemokine responses to helminth and protozoan parasites and to fungus and mite allergens in neonates, children, adults, and the elderly. IMMUNITY & AGEING 2013; 10:29. [PMID: 23855879 PMCID: PMC3720251 DOI: 10.1186/1742-4933-10-29] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 06/29/2013] [Indexed: 01/13/2023]
Abstract
Background In rural sub-Saharan Africa, endemic populations are often infected concurrently with several intestinal and intravascular helminth and protozoan parasites. A specific, balanced and, to an extent, protective immunity will develop over time in response to repeated parasite encounters, with immune responses initially being poorly adapted and non-protective. The cellular production of pro-inflammatory and regulatory cytokines and chemokines in response to helminth, protozoan antigens and ubiquitous allergens were studied in neonates, children, adults and the elderly. Results In children schistosomiasis prevailed (33%) while hookworm and Entamoeba histolytica/E. dispar was found in up to half of adults and the elderly. Mansonella perstans filariasis was only present in adults (24%) and the elderly (25%). Two or more parasite infections were diagnosed in 41% of children, while such polyparasitism was present in 34% and 38% of adults and the elderly. Cytokine and chemokine production was distinctively inducible by parasite antigens; pro-inflammatory Th2-type cytokine IL-19 was activated by Entamoeba and Ascaris antigens, being low in neonates and children while IL-19 production enhanced “stepwise” in adults and elderly. In contrast, highest production of MIP-1delta/CCL15 was present in neonates and children and inducible by Entamoeba-specific antigens only. Adults and the elderly had enhanced regulatory IL-27 cytokine responses, with Th2-type chemokines (MCP-4/CCL13, Eotaxin-2/CCL24) and cytokines (IL-33) being notably inducible by helminth- and Entamoeba-specific antigens and fungus-derived allergens. The lower cellular responsiveness in neonates and children highlighted the development of a parasite-specific cellular response profile in response to repeated episodes of exposure and re-infection. Conclusions Following repeated exposure to parasites, and as a consequence of host inability to prevent or eliminate intestinal helminth or protozoa infections, a repertoire of immune responses will evolve with lessened pro-inflammatory and pronounced regulatory cytokines and chemokines; this is required for partial parasite control as well as for preventing inadequate and excessive host tissue and organ damage.
Collapse
|
25
|
MicroRNA-gene expression network in murine liver during Schistosoma japonicum infection. PLoS One 2013; 8:e67037. [PMID: 23825609 PMCID: PMC3692539 DOI: 10.1371/journal.pone.0067037] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 05/13/2013] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Schistosomiasis japonica remains a significant public health problem in China and Southeast Asian countries. The most typical and serious outcome of the chronic oriental schistosomiasis is the progressive granuloma and fibrosis in the host liver, which has been a major medical challenge. However, the molecular mechanism underling the hepatic pathogenesis is still not clear. METHODOLOGY AND PRINCIPAL FINDINGS Using microarrays, we quantified the temporal gene expression profiles in the liver of Schistosoma japonicum-infected BALB/c mice at 15, 30, and 45 day post infection (dpi) with that from uninfected mice as controls. Gene expression alternation associated with liver damage was observed in the initial phase of infection (dpi 15), which became more magnificent with the onset of egg-laying. Up-regulated genes were dominantly associated with inflammatory infiltration, whereas down-regulated genes primarily led to the hepatic functional disorders. Simultaneously, microRNA profiles from the same samples were decoded by Solexa sequencing. More than 130 miRNAs were differentially expressed in murine liver during S. japonicum infection. MiRNAs significantly dysregulated in the mid-phase of infection (dpi 30), such as mmu-miR-146b and mmu-miR-155, may relate to the regulation of hepatic inflammatory responses, whereas miRNAs exhibiting a peak expression in the late phase of infection (dpi 45), such as mmu-miR-223, mmu-miR-146a/b, mmu-miR-155, mmu-miR-34c, mmu-miR-199, and mmu-miR-134, may represent a molecular signature of the development of schistosomal hepatopathy. Further, a dynamic miRNA-gene co-expression network in the progression of infection was constructed. CONCLUSIONS AND SIGNIFICANCE This study presents a global view of dynamic expression of both mRNA and miRNA transcripts in murine liver during S. japonicum infection, and highlights that miRNAs may play a variety of regulatory roles in balancing the immune responses during the development of hepatic pathology. The data provide robust information for further researches on the pathogenesis and molecular events of hepatopathy induced by schistosome eggs.
Collapse
|
26
|
Anthony BJ, James KR, Gobert GN, Ramm GA, McManus DP. Schistosomajaponicum Eggs Induce a Proinflammatory, Anti-Fibrogenic Phenotype in Hepatic Stellate Cells. PLoS One 2013; 8:e68479. [PMID: 23840855 PMCID: PMC3686876 DOI: 10.1371/journal.pone.0068479] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 06/04/2013] [Indexed: 01/18/2023] Open
Abstract
Hepatic fibrosis induced by egg deposition is the most serious pathology associated with chronic schistosomiasis, in which the hepatic stellate cell (HSC) plays a central role. While the effect of Schistosoma mansoni eggs on the fibrogenic phenotype of HSCs has been investigated, studies determining the effect of eggs of S. japonicum on HSCs are lacking. Disease caused by S. japonicum is much more severe than that resulting from S. mansoni infection so it is important to compare the pathologies caused by these two parasites, to determine whether this phenotype is due to the species interacting differently with the mammalian host. Accordingly, we investigated the effect of S. japonicum eggs on the human HSC cell line, LX-2, with and without TGF-β (Transforming Growth Factor beta) co-treatment, so as to determine the impact on genes associated with fibrogenesis, inflammation and matrix re-organisation. Activation status of HSCs was assessed by αSMA (Alpha Smooth Muscle Actin) immunofluorescence, accumulation of Oil Red O-stained lipid droplets and the relative expression of selected genes associated with activation. The fibrogenic phenotype of HSCs was inhibited by the presence of eggs both with or without TGF-β treatment, as evidenced by a lack of αSMA staining and reduced gene expression of αSMA and Col1A1 (Collagen 1A1). Unlike S. mansoni-treated cells, however, expression of the quiescent HSC marker PPAR-γ (Peroxisome Proliferator-Activated Receptor gamma) was not increased, nor was there accumulation of lipid droplets. In contrast, S. japonicum eggs induced the mRNA expression of MMP-9 (Matrix Metalloproteinase 9), CCL2 (Chemokine (C-C motif) Ligand 2) and IL-6 (Interleukin 6) in HSCs indicating that rather than inducing complete HSC quiescence, the eggs induced a proinflammatory phenotype. These results suggest HSCs in close proximity to S. japonicum eggs in the liver may play a role in the proinflammatory regulation of hepatic granuloma formation.
Collapse
Affiliation(s)
- Barrie J. Anthony
- Molecular Parasitology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
- * E-mail:
| | - Kylie R. James
- Molecular Parasitology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
| | - Geoffrey N. Gobert
- Molecular Parasitology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
| | - Grant A. Ramm
- The Hepatic Fibrosis Group, Queensland Institute of Medical Research, Brisbane, Australia
- Faculty of Health Sciences, The University of Queensland, Brisbane, Australia
| | - Donald P. McManus
- Molecular Parasitology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
| |
Collapse
|
27
|
Chuah C, Jones MK, Burke ML, Owen HC, Anthony BJ, McManus DP, Ramm GA, Gobert GN. Spatial and temporal transcriptomics ofSchistosoma japonicum-induced hepatic granuloma formation reveals novel roles for neutrophils. J Leukoc Biol 2013; 94:353-65. [DOI: 10.1189/jlb.1212653] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
28
|
Beschin A, De Baetselier P, Van Ginderachter JA. Contribution of myeloid cell subsets to liver fibrosis in parasite infection. J Pathol 2012; 229:186-97. [DOI: 10.1002/path.4112] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 08/24/2012] [Accepted: 09/13/2012] [Indexed: 12/23/2022]
Affiliation(s)
- Alain Beschin
- Myeloid Cell Immunology Laboratory; VIB Brussels Belgium
- Cellular and Molecular Immunology Unit; Vrije Universiteit Brussel; Brussels Belgium
| | - Patrick De Baetselier
- Myeloid Cell Immunology Laboratory; VIB Brussels Belgium
- Cellular and Molecular Immunology Unit; Vrije Universiteit Brussel; Brussels Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory; VIB Brussels Belgium
- Cellular and Molecular Immunology Unit; Vrije Universiteit Brussel; Brussels Belgium
| |
Collapse
|
29
|
Migrating Schistosoma japonicum schistosomula induce an innate immune response and wound healing in the murine lung. Mol Immunol 2011; 49:191-200. [PMID: 21917316 DOI: 10.1016/j.molimm.2011.08.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 08/10/2011] [Accepted: 08/18/2011] [Indexed: 01/13/2023]
Abstract
The migrating schistosomulum is an important stage of the schistosome lifecycle and represents a key target for elimination of infection by natural and vaccine-induced host immune responses. To gain a better understanding of how schistosomes initiate a primary host immune response we have characterised the host lung response to migrating Schistosoma japonicum schistosomula using a combination of histopathology, microarray analysis and real-time PCR. Our findings indicate that the early pulmonary response to these migrating larvae is characteristic of innate inflammation and wound healing. This response is associated with significant up-regulation of several genes with immunoregulatory function including Ch25h, Hmox1 and Retnla which may act to control the nature or magnitude of the inflammatory response to the migrating schistosomula, promoting both parasite and host survival. These findings contribute to our understanding of host-parasite interactions associated with schistosome and, especially, S. japonicum infection, and may aid the future design of novel vaccines that target the lung stage schistosomulum.
Collapse
|