1
|
Jain S. Is Schistosoma mansoni playing a part in liver carcinogenesis? J Helminthol 2024; 98:e61. [PMID: 39469749 DOI: 10.1017/s0022149x24000506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
The relationship between Schistosoma mansoni (Sm) and hepatocellular carcinoma (HCC) has been evaluated by many studies that point towards a co-relation between schistosomal infection and HCC. While many such studies demonstrated that Sm infection in the presence of another carcinogenic factors leads to HCC, none of these studies could conclusively prove the cancer-inducing ability of Sm in humans, independent of other carcinogenic factors. The aim of this work is to present the current understanding on the association of Sm with HCC. Many epidemiological, pathological, and clinical studies have shown the role of multiple events like chronic inflammation and fibrosis as well as hepato-toxic agents like soluble egg antigens (SEAs), which help in creating a micro-environment which is suitable for HCC development. The role of Sm infection and deposited eggs in causing persistent inflammation, advanced fibrosis, and the role of SEAs, especially IPSE/alpha-1, is emphasised. This work concludes that Sm infection has the potential to induce cancer independently but the same has not been reported in humans to date. Extensive research is required to establish a causal relationship between Sm infection and HCC induction, or a complete lack thereof. However, Sm infection definitely acts along with other carcinogenic factors to induce HCC at a much faster pace and also leads to an aggressive form of liver cancer, which the other carcinogenic factor could not have achieved alone.
Collapse
Affiliation(s)
- S Jain
- Institute for Globally Distributed Open Research and Education (IGDORE), India
| |
Collapse
|
2
|
Peterková K, Konečný L, Macháček T, Jedličková L, Winkelmann F, Sombetzki M, Dvořák J. Winners vs. losers: Schistosoma mansoni intestinal and liver eggs exhibit striking differences in gene expression and immunogenicity. PLoS Pathog 2024; 20:e1012268. [PMID: 38814989 PMCID: PMC11166329 DOI: 10.1371/journal.ppat.1012268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/11/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
The eggs of the blood fluke Schistosoma mansoni are the main cause of the clinical manifestations of chronic schistosomiasis. After laying, the egg "winners" attach to the endothelium of the mesenteric vein and, after a period of development, induce the growth of a small granuloma, which facilitates their passage to the intestinal lumen. Egg "losers" carried by the bloodstream to non-specific tissues also undergo full development and induce large granuloma formation, but their life ends there. Although these trapped eggs represent a dead end in the parasite life cycle, the vast majority of studies attempting to describe the biology of the S. mansoni eggs have studied these liver-trapped "losers" instead of migrating intestinal "winners". This raises the fundamental question of how these eggs differ. With robust comparative transcriptomic analysis performed on S. mansoni eggs isolated 7 weeks post infection, we show that gene expression is critically dependent on tissue localization, both in the early and late stages of development. While mitochondrial genes and venom allergen-like proteins are significantly upregulated in mature intestinal eggs, well-described egg immunomodulators IPSE/alpha-1 and omega-1, together with micro-exon genes, are predominantly expressed in liver eggs. In addition, several proteases and protease inhibitors previously implicated in egg-host interactions display clear tissue-specific gene expression patterns. These major differences in gene expression could be then reflected in the observed different ability of liver and intestinal soluble egg antigens to elicit host immune responses and in the shorter viability of miracidia hatched from liver eggs. Our comparative analysis provides a new perspective on the biology of parasite's eggs in the context of their development and tissue localization. These findings could contribute to a broader and more accurate understanding of parasite eggs interactions with the host, which have historically been often restricted to liver eggs and sometimes inaccurately generalized.
Collapse
Affiliation(s)
- Kristýna Peterková
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
- Department of Zoology and Fisheries, Center of Infectious Animal Diseases, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, Prague, Czechia
| | - Lukáš Konečný
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
- Department of Ecology, Center of Infectious Animal Diseases, Faculty of Environmental Sciences, Czech University of Life Sciences, Prague, Czechia
| | - Tomáš Macháček
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Lucie Jedličková
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
- Department of Zoology and Fisheries, Center of Infectious Animal Diseases, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, Prague, Czechia
| | - Franziska Winkelmann
- Universitätsmedizin Rostock, Zentrum für Innere Medizin, Abteilung für Tropenmedizin, Infektionskrankheiten und Sektion Nephrologie, Rostock, Germany
| | - Martina Sombetzki
- Universitätsmedizin Rostock, Zentrum für Innere Medizin, Abteilung für Tropenmedizin, Infektionskrankheiten und Sektion Nephrologie, Rostock, Germany
| | - Jan Dvořák
- Department of Ecology, Center of Infectious Animal Diseases, Faculty of Environmental Sciences, Czech University of Life Sciences, Prague, Czechia
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
3
|
Abstract
Schistosomes are long lived, intravascular parasitic platyhelminths that infect >200 million people globally. The molecular mechanisms used by these blood flukes to dampen host immune responses are described in this review. Adult worms express a collection of host-interactive tegumental ectoenzymes that can cleave host signaling molecules such as the "alarmin" ATP (cleaved by SmATPDase1), the platelet activator ADP (SmATPDase1, SmNPP5), and can convert AMP into the anti-inflammatory mediator adenosine (SmAP). SmAP can additionally cleave the lipid immunomodulator sphingosine-1-phosphate and the proinflammatory anionic polymer, polyP. In addition, the worms release a barrage of proteins (e.g., SmCB1, SjHSP70, cyclophilin A) that can impinge on immune cell function. Parasite eggs also release their own immunoregulatory proteins (e.g., IPSE/α1, omega1, SmCKBP) as do invasive cercariae (e.g., Sm16, Sj16). Some schistosome glycans (e.g., LNFPIII, LNnT) and lipids (e.g., Lyso-PS, LPC), produced by several life stages, likewise affect immune cell responses. The parasites not only produce eicosanoids (e.g., PGE2, PGD2-that can be anti-inflammatory) but can also induce host cells to release these metabolites. Finally, the worms release extracellular vesicles (EVs) containing microRNAs, and these too have been shown to skew host cell metabolism. Thus, schistosomes employ an array of biomolecules-protein, lipid, glycan, nucleic acid, and more, to bend host biochemistry to their liking. Many of the listed molecules have been individually shown capable of inducing aspects of the polarized Th2 response seen following infection (with the generation of regulatory T cells (Tregs), regulatory B cells (Bregs) and anti-inflammatory, alternatively activated (M2) macrophages). Precisely how host cells integrate the impact of these myriad parasite products following natural infection is not known. Several of the schistosome immunomodulators described here are in development as novel therapeutics against autoimmune, inflammatory, and other, nonparasitic, diseases.
Collapse
Affiliation(s)
- Sreemoyee Acharya
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Akram A. Da’dara
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Patrick J. Skelly
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
4
|
von Bülow V, Lichtenberger J, Grevelding CG, Falcone FH, Roeb E, Roderfeld M. Does Schistosoma Mansoni Facilitate Carcinogenesis? Cells 2021; 10:1982. [PMID: 34440754 PMCID: PMC8393187 DOI: 10.3390/cells10081982] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/21/2021] [Accepted: 08/01/2021] [Indexed: 12/24/2022] Open
Abstract
Schistosomiasis is one of the most prominent parasite-induced infectious diseases, causing tremendous medical and socioeconomic problems. Current studies have reported on the spread of endemic regions and the fear of development of resistance against praziquantel, the only effective drug available. Among the Schistosoma species, only S. haematobium is classified as a Group 1 carcinogen (definitely cancerogenic to humans), causing squamous cell carcinoma of the bladder, whereas infection with S. mansoni is included in Group 3 of carcinogenic hazards to humans by the International Agency for Research on Cancer (IARC), indicating insufficient evidence to determine its carcinogenicity. Nevertheless, although S. mansoni has not been discussed as an organic carcinogen, the multiplicity of case reports, together with recent data from animal models and cell culture experiments, suggests that this parasite can predispose patients to or promote hepatic and colorectal cancer. In this review, we discuss the current data, with a focus on new developments regarding the association of S. mansoni infection with human cancer and the recently discovered biomolecular mechanisms by which S. mansoni may predispose patients to cancer development and carcinogenesis.
Collapse
Affiliation(s)
- Verena von Bülow
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (V.v.B.); (J.L.); (E.R.)
| | - Jakob Lichtenberger
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (V.v.B.); (J.L.); (E.R.)
| | - Christoph G. Grevelding
- Institute of Parasitology, BFS, Justus Liebig University, 35392 Giessen, Germany; (C.G.G.); (F.H.F.)
| | - Franco H. Falcone
- Institute of Parasitology, BFS, Justus Liebig University, 35392 Giessen, Germany; (C.G.G.); (F.H.F.)
| | - Elke Roeb
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (V.v.B.); (J.L.); (E.R.)
| | - Martin Roderfeld
- Department of Gastroenterology, Justus Liebig University, 35392 Giessen, Germany; (V.v.B.); (J.L.); (E.R.)
| |
Collapse
|
5
|
Mbanefo EC, Agbo CT, Zhao Y, Lamanna OK, Thai KH, Karinshak SE, Khan MA, Fu CL, Odegaard JI, Saltikova IV, Smout MJ, Pennington LF, Nicolls MR, Jardetzky TS, Loukas A, Brindley PJ, Falcone FH, Hsieh MH. IPSE, an abundant egg-secreted protein of the carcinogenic helminth Schistosoma haematobium, promotes proliferation of bladder cancer cells and angiogenesis. Infect Agent Cancer 2020; 15:63. [PMID: 33101456 PMCID: PMC7578584 DOI: 10.1186/s13027-020-00331-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Schistosoma haematobium, the helminth causing urogenital schistosomiasis, is a known bladder carcinogen. Despite the causal link between S. haematobium and bladder cancer, the underlying mechanisms are poorly understood. S. haematobium oviposition in the bladder is associated with angiogenesis and urothelial hyperplasia. These changes may be pre-carcinogenic events in the bladder. We hypothesized that the Interleukin-4-inducing principle of Schistosoma mansoni eggs (IPSE), an S. haematobium egg-secreted "infiltrin" protein that enters host cell nuclei to alter cellular activity, is sufficient to induce angiogenesis and urothelial hyperplasia. Methods: Mouse bladders injected with S. haematobium eggs were analyzed via microscopy for angiogenesis and urothelial hyperplasia. Endothelial and urothelial cell lines were incubated with recombinant IPSE protein or an IPSE mutant protein that lacks the native nuclear localization sequence (NLS-) and proliferation measured using CFSE staining and real-time monitoring of cell growth. IPSE's effects on urothelial cell cycle status was assayed through propidium iodide staining. Endothelial and urothelial cell uptake of fluorophore-labeled IPSE was measured. Findings: Injection of S. haematobium eggs into the bladder triggers angiogenesis, enhances leakiness of bladder blood vessels, and drives urothelial hyperplasia. Wild type IPSE, but not NLS-, increases proliferation of endothelial and urothelial cells and skews urothelial cells towards S phase. Finally, IPSE is internalized by both endothelial and urothelial cells. Interpretation: IPSE drives endothelial and urothelial proliferation, which may depend on internalization of the molecule. The urothelial effects of IPSE depend upon its NLS. Thus, IPSE is a candidate pro-carcinogenic molecule of S. haematobium. SUMMARY Schistosoma haematobium acts as a bladder carcinogen through unclear mechanisms. The S. haematobium homolog of IPSE, a secreted schistosome egg immunomodulatory molecule, enhances angiogenesis and urothelial proliferation, hallmarks of pre-carcinogenesis, suggesting IPSE is a key pro-oncogenic molecule of S. haematobium.
Collapse
Affiliation(s)
- Evaristus C. Mbanefo
- Division of Urology, Department of Surgery, Children’s National Hospital, West Wing, 4th Floor, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | | | | | - Olivia K. Lamanna
- Division of Urology, Department of Surgery, Children’s National Hospital, West Wing, 4th Floor, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Kim H. Thai
- Baylor Scott and White Health, Temple, TX USA
| | - Shannon E. Karinshak
- Department of Microbiology, Immunology, and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC USA
| | - Mohammad Afzal Khan
- King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | | | | | - Irina V. Saltikova
- Guardant Health, Redwood City, CA USA
- Siberian State Medical University, Tomsk, Russian Federation
| | | | | | - Mark R. Nicolls
- Division of Pulmonology, Allergy, and Critical Care Medicine, Stanford University, Stanford, CA USA
| | | | - Alex Loukas
- James Cook University, Townsville, Australia
| | - Paul J. Brindley
- Department of Microbiology, Immunology, and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC USA
| | - Franco H. Falcone
- Institute of Parasitology, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Michael H. Hsieh
- Division of Urology, Department of Surgery, Children’s National Hospital, West Wing, 4th Floor, 111 Michigan Avenue NW, Washington, DC 20010 USA
| |
Collapse
|
6
|
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: 89] [Impact Index Per Article: 17.8] [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
|
7
|
Schwartz C, Fallon PG. Schistosoma "Eggs-Iting" the Host: Granuloma Formation and Egg Excretion. Front Immunol 2018; 9:2492. [PMID: 30459767 PMCID: PMC6232930 DOI: 10.3389/fimmu.2018.02492] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/09/2018] [Indexed: 12/12/2022] Open
Abstract
Schistosomiasis is a major cause of morbidity in humans invoked by chronic infection with parasitic trematodes of the genus Schistosoma. Schistosomes have a complex life-cycle involving infections of an aquatic snail intermediate host and a definitive mammalian host. In humans, adult male and female worms lie within the vasculature. Here, they propagate and eggs are laid. These eggs must then be released from the host to continue the life cycle. Schistosoma mansoni and Schistosoma japonicum reside in the mesenteric circulation of the intestines with egg excreted in the feces. In contrast, S. haematobium are present in the venus plexus of the bladder, expelling eggs in the urine. In an impressive case of exploitation of the host immune system, this process of Schistosome “eggs-iting” the host is immune dependent. In this article, we review the formation of the egg granuloma and explore how S. mansoni eggs laid in vasculature must usurp immunity to induce regulated inflammation, to facilitate extravasation through the intestinal wall and to be expelled in the feces. We highlight the roles of immune cell populations, stromal factors, and egg secretions in the process of egg excretion to provide a comprehensive overview of the current state of knowledge regarding a vastly unexplored mechanism.
Collapse
Affiliation(s)
- Christian Schwartz
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Padraic G Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland.,Trinity Translational Medicine Institute, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Haeberlein S, Obieglo K, Ozir-Fazalalikhan A, Chayé MAM, Veninga H, van der Vlugt LEPM, Voskamp A, Boon L, den Haan JMM, Westerhof LB, Wilbers RHP, Schots A, Schramm G, Hokke CH, Smits HH. Schistosome egg antigens, including the glycoprotein IPSE/alpha-1, trigger the development of regulatory B cells. PLoS Pathog 2017; 13:e1006539. [PMID: 28753651 PMCID: PMC5550006 DOI: 10.1371/journal.ppat.1006539] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 08/09/2017] [Accepted: 07/18/2017] [Indexed: 12/15/2022] Open
Abstract
Infection with the helminth Schistosoma (S.) mansoni drives the development of interleukin (IL)-10-producing regulatory B (Breg) cells in mice and man, which have the capacity to reduce experimental allergic airway inflammation and are thus of high therapeutic interest. However, both the involved antigen and cellular mechanisms that drive Breg cell development remain to be elucidated. Therefore, we investigated whether S. mansoni soluble egg antigens (SEA) directly interact with B cells to enhance their regulatory potential, or act indirectly on B cells via SEA-modulated macrophage subsets. Intraperitoneal injections of S. mansoni eggs or SEA significantly upregulated IL-10 and CD86 expression by marginal zone B cells. Both B cells as well as macrophages of the splenic marginal zone efficiently bound SEA in vivo, but macrophages were dispensable for Breg cell induction as shown by macrophage depletion with clodronate liposomes. SEA was internalized into acidic cell compartments of B cells and induced a 3-fold increase of IL-10, which was dependent on endosomal acidification and was further enhanced by CD40 ligation. IPSE/alpha-1, one of the major antigens in SEA, was also capable of inducing IL-10 in naïve B cells, which was reproduced by tobacco plant-derived recombinant IPSE. Other major schistosomal antigens, omega-1 and kappa-5, had no effect. SEA depleted of IPSE/alpha-1 was still able to induce Breg cells indicating that SEA contains more Breg cell-inducing components. Importantly, SEA- and IPSE-induced Breg cells triggered regulatory T cell development in vitro. SEA and recombinant IPSE/alpha-1 also induced IL-10 production in human CD1d+ B cells. In conclusion, the mechanism of S. mansoni-induced Breg cell development involves a direct targeting of B cells by SEA components such as the secretory glycoprotein IPSE/alpha-1. Infection with helminth parasites is known to be inversely associated with hyper-inflammatory disorders. While Schistosoma (S.) mansoni has been described to exert its down-modulatory effects on inflammation by inducing a network of regulatory immune cells such as regulatory B (Breg), the mechanisms of Breg cell induction remain unclear. Here, we use in vivo and in vitro approaches to show that antigens from S. mansoni eggs, among which the major glycoprotein IPSE/alpha-1, directly interact with splenic marginal zone B cells of mice which triggers them to produce the anti-inflammatory cytokine IL-10 and their capacity to induce regulatory T (Treg) cells. We also found that IPSE/alpha-1 induces IL-10 in human CD1d+ B cells, and that both natural and recombinant IPSE/alpha-1 are equally effective in driving murine and human Breg cells. Our study thus provides insight into the mechanisms of Breg cell induction by schistosomes, and an important step towards the development of helminth-based treatment strategies against hyper-inflammatory diseases.
Collapse
Affiliation(s)
- Simone Haeberlein
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Katja Obieglo
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Mathilde A. M. Chayé
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Henrike Veninga
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | | | - Astrid Voskamp
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Joke M. M. den Haan
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Lotte B. Westerhof
- Plant Science Department, Wageningen University and Research Centre, Droevendaalsesteeg, Wageningen, Netherlands
| | - Ruud H. P. Wilbers
- Plant Science Department, Wageningen University and Research Centre, Droevendaalsesteeg, Wageningen, Netherlands
| | - Arjen Schots
- Plant Science Department, Wageningen University and Research Centre, Droevendaalsesteeg, Wageningen, Netherlands
| | - Gabriele Schramm
- Experimental Pneumology, Priority Research Area Asthma & Allergy, Research Center Borstel, Parkallee, Borstel, Germany
| | - Cornelis H. Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Hermelijn H. Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- * E-mail:
| |
Collapse
|
9
|
Hagen J, Scheerlinck JPY, Young ND, Gasser RB, Kalinna BH. Prospects for Vector-Based Gene Silencing to Explore Immunobiological Features of Schistosoma mansoni. ADVANCES IN PARASITOLOGY 2015; 88:85-122. [PMID: 25911366 DOI: 10.1016/bs.apar.2015.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Schistosomiasis is a prevalent, socioeconomically important disease of humans caused by parasites of the genus Schistosoma (schistosomes or blood flukes). Currently, more than 200 million people worldwide are infected with schistosomes. Despite major research efforts, there is only one drug routinely used for effective treatment, and no vaccine is available to combat schistosomiasis. The purpose of the present article is to (1) provide a background on the parasites and different forms of disease; (2) describe key immunomolecular aspects of disease induced in the host; and (3) critically appraise functional genomic methods employed to explore parasite biology, parasite-host interactions and disease at the molecular level. Importantly, the article also describes the features and advantages of lentiviral delivery of artificial microRNAs to silence genes. It also discusses the first successful application of such an approach in schistosomes, in order to explore the immunobiological role of selected target proteins known to be involved in egg-induced disease. The lentiviral transduction system provides exciting prospects for future, fundamental investigations of schistosomes, and is likely to have broad applicability to other eukaryotic pathogens and infectious diseases. The ability to achieve effective and stable gene perturbation in parasites has major biotechnological implications, and might facilitate the development of radically new methods for the treatment and control of parasitic diseases.
Collapse
Affiliation(s)
- Jana Hagen
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Jean-Pierre Y Scheerlinck
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Neil D Young
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Robin B Gasser
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Bernd H Kalinna
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
10
|
Mishra A, Krishnan B, Srivastava SS, Sharma Y. Microbial βγ-crystallins. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:42-51. [PMID: 24594023 DOI: 10.1016/j.pbiomolbio.2014.02.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 01/24/2023]
Abstract
βγ-Crystallins have emerged as a superfamily of structurally homologous proteins with representatives across the domains of life. A major portion of this superfamily is constituted by members from microorganisms. This superfamily has also been recognized as a novel group of Ca(2+)-binding proteins with huge diversity. The βγ domain shows variable properties in Ca(2+) binding, stability and association with other domains. The various members present a series of evolutionary adaptations culminating in great diversity in properties and functions. Most of the predicted βγ-crystallins are yet to be characterized experimentally. In this review, we outline the distinctive features of microbial βγ-crystallins and their position in the βγ-crystallin superfamily.
Collapse
Affiliation(s)
- Amita Mishra
- CSIR - Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
| | - Bal Krishnan
- CSIR - Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
| | | | - Yogendra Sharma
- CSIR - Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India.
| |
Collapse
|
11
|
Oliveira RR, Figueiredo JP, Cardoso LS, Jabar RL, Souza RP, Wells MT, Carvalho EM, Fitzgerald DW, Barnes KC, Araújo MI, Glesby MJ. Factors associated with resistance to Schistosoma mansoni infection in an endemic area of Bahia, Brazil. Am J Trop Med Hyg 2012; 86:296-305. [PMID: 22302866 DOI: 10.4269/ajtmh.2012.11-0204] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Detailed knowledge of factors associated with resistance to Schistosoma mansoni infection in endemic areas might facilitate more effective schistosomiasis control. We conducted a cross-sectional study of persons resistant to schistosomiasis and found no association between socioeconomic status and resistance to infection. Mononuclear cells of resistant subjects produced higher levels of interleukin-5 (IL-5), IL-13 and interferon-γ upon stimulation with soluble egg antigen (SEA) compared with infected persons. When stimulated with Sm21.6 or Sm22.6, levels of IL-10 were higher in cell culture of resistant persons. Levels of IgE against soluble adult worm antigen (SWAP) and against interleukin-4-inducing principle from S. mansoni eggs (IPSE) and levels of IgG4 against SWAP, SEA, and Sm22.6 were lower in the resistant group compared with the susceptible group. Our data suggest that socioeconomic status could not fully explain resistance to S. mansoni infection observed in the studied area. However, a mixture of Th1 and Th2 immune responses and low levels of specific IgG4 against parasite antigens could be mediating resistance to infection.
Collapse
Affiliation(s)
- Ricardo R Oliveira
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Madalosso RC, Oliveira GC, Martins MT, Vieira AED, Barbosa J, Caliari MV, Castilho RO, Tagliati CA. Campomanesia lineatifolia Ruiz & Pav. as a gastroprotective agent. JOURNAL OF ETHNOPHARMACOLOGY 2012; 139:772-779. [PMID: 22178179 DOI: 10.1016/j.jep.2011.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 10/27/2011] [Accepted: 12/03/2011] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Campomanesia lineatifolia Ruiz and Pav. (Myrtaceae) is a native edible species found in the Amazon Rainforest, commonly known as gabiroba. In Brazil, Campomanesia species are frequently used in traditional medicine for gastrointestinal disorders. MATERIALS AND METHODS The present study performed phytochemical analyses and determined both the in vitro antioxidant activity of the ethanolic extract of Campomanesia lineatifolia leaves (EEC) as well as its ethyl acetate fraction (EAFC). In this analysis, quercetin was used as a positive control. Gastroprotective activity was also investigated at different oral doses in two experimental models in rats - gastric lesion induced by ethanol and gastric lesion induced by indomethacin. In this analysis, cimetidine and sucralfate were used as positive controls. The area of gastric lesion underwent macroscopic and histomorphometric evaluations, while the mucus content was estimated by applying the periodic acid-Schiff stain. Oral acute toxicity was also assessed. RESULTS Phytochemical studies revealed the presence of flavonoids and tannins. Catechin and quercitrin were isolated by bioguided chromatographic fractionation of EAFC. EEC and EAFC presented in vitro antioxidant activity. The oral administration of EEC and EAFC at doses 100-400 mg/kg (ethanol model) and at doses of 400-1200 mg/kg (indomethacin model) proved to be effective in preventing gastric ulcerations in rats. Pretreatment with EAFC (400mg/kg) significantly increased the gastric mucus content in the ethanol model. No animals died during the acute oral toxicology test. CONCLUSIONS Results confirm the Brazilian ethnopharmacological use of Campomanesia lineatifolia as a gastroprotective agent and indicate that the anti-ulcer effect is most likely mediated by scavenging free radicals due to the polyphenol content and, at least in part, by increasing the mucus secretion and the mucosal defense. In addition, EEC and EAFC were found to be safe when applied to a 2000 mg/kg single oral dose.
Collapse
Affiliation(s)
- R C Madalosso
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Thakar J, Pathak AK, Murphy L, Albert R, Cattadori IM. Network model of immune responses reveals key effectors to single and co-infection dynamics by a respiratory bacterium and a gastrointestinal helminth. PLoS Comput Biol 2012; 8:e1002345. [PMID: 22253585 PMCID: PMC3257297 DOI: 10.1371/journal.pcbi.1002345] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 11/25/2011] [Indexed: 12/22/2022] Open
Abstract
Co-infections alter the host immune response but how the systemic and local processes at the site of infection interact is still unclear. The majority of studies on co-infections concentrate on one of the infecting species, an immune function or group of cells and often focus on the initial phase of the infection. Here, we used a combination of experiments and mathematical modelling to investigate the network of immune responses against single and co-infections with the respiratory bacterium Bordetella bronchiseptica and the gastrointestinal helminth Trichostrongylus retortaeformis. Our goal was to identify representative mediators and functions that could capture the essence of the host immune response as a whole, and to assess how their relative contribution dynamically changed over time and between single and co-infected individuals. Network-based discrete dynamic models of single infections were built using current knowledge of bacterial and helminth immunology; the two single infection models were combined into a co-infection model that was then verified by our empirical findings. Simulations showed that a T helper cell mediated antibody and neutrophil response led to phagocytosis and clearance of B. bronchiseptica from the lungs. This was consistent in single and co-infection with no significant delay induced by the helminth. In contrast, T. retortaeformis intensity decreased faster when co-infected with the bacterium. Simulations suggested that the robust recruitment of neutrophils in the co-infection, added to the activation of IgG and eosinophil driven reduction of larvae, which also played an important role in single infection, contributed to this fast clearance. Perturbation analysis of the models, through the knockout of individual nodes (immune cells), identified the cells critical to parasite persistence and clearance both in single and co-infections. Our integrated approach captured the within-host immuno-dynamics of bacteria-helminth infection and identified key components that can be crucial for explaining individual variability between single and co-infections in natural populations. Infections with different infecting agents can alter the immune response against any one parasite and the relative abundance and persistence of the infections within the host. This is because the immune system is not compartmentalized but acts as a whole to allow the host to maintain control of the infections as well as repair damaged tissues and avoid immuno-pathology. There is no comprehensive understanding of the immune responses during co-infections and of how systemic and local mechanisms interact. Here we integrated experimental data with mathematical modelling to describe the network of immune responses of single and co-infection by a respiratory bacterium and a gastrointestinal helminth. We were able to identify key cells and functions responsible for clearing or reducing both parasites and showed that some mechanisms differed between type of infection as a result of different signal outputs and cells contributing to the immune processes. This study highlights the importance of understanding the immuno-dynamics of co-infection as a host response, how immune mechanisms differ from single infections and how they may alter parasite persistence, impact and abundance.
Collapse
Affiliation(s)
- Juilee Thakar
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Ashutosh K. Pathak
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Lisa Murphy
- Division of Animal Production and Public Health, Veterinary School, University of Glasgow, Glasgow, United Kingdom
| | - Réka Albert
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Physics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Isabella M. Cattadori
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
14
|
Abdulla MH, Lim KC, McKerrow JH, Caffrey CR. Proteomic identification of IPSE/alpha-1 as a major hepatotoxin secreted by Schistosoma mansoni eggs. PLoS Negl Trop Dis 2011; 5:e1368. [PMID: 22039561 PMCID: PMC3201919 DOI: 10.1371/journal.pntd.0001368] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 09/09/2011] [Indexed: 01/12/2023] Open
Abstract
Background Eggs deposited in the liver of the mammalian host by the blood fluke parasite, Schistosoma mansoni, normally drive a T-helper-2 (Th2)-mediated granulomatous response in immune-competent mice. By contrast, in mice deprived of T-cells and incapable of producing granulomata, egg-secreted proteins (ESP) induce acute hepatic injury and death. Previous work has shown that one such ESP, the T2 ribonuclease known as omega-1, is hepatotoxic in vivo in that specific antisera to omega-1 prevent hepatocyte damage. Methodology/Principal Findings Using an in vitro culture system employing mouse primary hepatocytes and alanine transaminase (ALT) activity as a marker of heptocyte injury, we demonstrated that S. mansoni eggs, egg-secreted proteins (ESP), soluble-egg antigen (SEA), and omega-1 are directly hepatotoxic and in a dose-dependent manner. Depletion of omega-1 using a monoclonal antibody abolished the toxicity of pure omega-1 and diminished the toxicity in ESP and SEA by 47 and 33%, respectively. Anion exchange chromatography of ESP yielded one predominant hepatotoxic fraction. Proteomics of that fraction identified the presence of IPSE/alpha-1 (IL-4 inducing principle from S. mansoni eggs), a known activator of basophils and inducer of Th2-type responses. Pure recombinant IPSE/alpha-1 also displayed a dose-dependent hepatotoxicity in vitro. Monoclonal antibody depletion of IPSE/alpha-1 abolished the latter's toxicity and diminished the total toxicity of ESP and SEA by 32 and 35%, respectively. Combined depletion of omega-1 and IPSE/alpha-1 diminished hepatotoxicity of ESP and SEA by 60 and 58% respectively. Conclusions We identified IPSE/alpha-1 as a novel hepatotoxin and conclude that both IPSE/alpha-1 and omega-1 account for the majority of the hepatotoxicity secreted by S. mansoni eggs. The flatworm disease, schistosomiasis, is a major public health problem in sub-Saharan Africa, South America and East Asia. A hallmark of infection with Schistosoma mansoni is the immune response to parasite eggs trapped in the liver and other organs. This response involves an infiltration of cells that surround the parasite egg forming a “granuloma.” In mice deprived of T-cells, this granulomatous response is lacking, and toxic products released by eggs quickly cause liver damage and death. Thus the granulomata protect the host from toxic egg products. Only one hepatotoxic molecule, omega-1, has been described to date. We set out to identify other S. mansoni egg hepatotoxins using liver cells grown in culture. We first showed that live eggs, their secretions, and pure omega-1 are toxic. Using a physical separation technique to prepare fractions from whole egg secretions, we identified the presence of IPSE/alpha-1, a protein that is known to strongly influence the immune system. We showed that IPSE/alpha-1 is also hepatotoxic, and that toxicity of both omega-1 and IPSE/alpha-1 can be prevented by first mixing the proteins with specific neutralizing antibodies. Both proteins constitute the majority of hepatotoxicity released by eggs.
Collapse
Affiliation(s)
- Maha-Hamadien Abdulla
- The Colorectal Research Center, Department of Surgery, King Khalid University Hospital, King Saud University, Riyadh, Saudi Arabia.
| | | | | | | |
Collapse
|