1
|
Rinaldi G, Loukas A, Sotillo J. Trematode Genomics and Proteomics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1454:507-539. [PMID: 39008274 DOI: 10.1007/978-3-031-60121-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Trematode infections stand out as one of the frequently overlooked tropical diseases, despite their wide global prevalence and remarkable capacity to parasitize diverse host species and tissues. Furthermore, these parasites hold significant socio-economic, medical, veterinary and agricultural implications. Over the past decades, substantial strides have been taken to bridge the information gap concerning various "omic" tools, such as proteomics and genomics, in this field. In this edition of the book, we highlight recent progress in genomics and proteomics concerning trematodes with a particular focus on the advances made in the past 5 years. Additionally, we present insights into cutting-edge technologies employed in studying trematode biology and shed light on the available resources for exploring the molecular facets of this particular group of parasitic helminths.
Collapse
Affiliation(s)
- Gabriel Rinaldi
- Department of Life Sciences, Aberystwyth University, Aberystwyth, UK
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Javier Sotillo
- Laboratorio de Referencia e Investigación en Parasitología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Spain.
| |
Collapse
|
2
|
Ruangsuwast A, Smout MJ, Brindley PJ, Loukas A, Laha T, Chaiyadet S. Tetraspanins from the liver fluke Opisthorchis viverrini stimulate cholangiocyte migration and inflammatory cytokine production. Folia Parasitol (Praha) 2023; 70:2023.017. [PMID: 37752807 DOI: 10.14411/fp.2023.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/26/2023] [Indexed: 09/28/2023]
Abstract
The liver fluke Opisthorchis viverrini (Poirier, 1886) (Digenea) secretes extracellular vesicles (EVs) bearing CD63-like tetraspanins on their surface. Fluke EVs are actively internalised by host cholangiocytes in the bile ducts, where they drive pathology and promote neoplasia through induction of cellular proliferation and secretion of inflammatory cytokines. We investigated the effects of tetraspanins of the CD63 superfamily by co-culturing recombinant forms of the large extracellular loop (LEL) of O. viverrini tetraspanin-2 (rLEL-Ov-TSP-2) and tetraspanin-3 (rLEL-Ov-TSP-3) with non-cancerous human bile duct (H69) and cholangiocarcinoma (CCA, M213) cell lines. The results showed that cell lines co-cultured with excretory/secretory products from adult O. viverrini (Ov-ES) underwent significantly increased cell proliferation at 48 hours but not 24 hours compared to untreated control cells (P < 0.05), whereas rLEL-Ov-TSP-3 co-culture resulted in significantly increased cell proliferation at both 24 hours (P < 0.05) and 48 hours (P < 0.01) time points. In like fashion, H69 cholangiocytes co-cultured with both Ov-ES and rLEL-Ov-TSP-3 underwent significantly elevated Il-6 and Il-8 gene expression for at least one of the time points assessed. Finally, both rLEL-Ov-TSP-2 and rLEL-Ov-TSP-3 significantly enhanced migration of both M213 and H69 cell lines. These findings indicated that O. viverrini CD63 family tetraspanins can promote a cancerous microenvironment by enhancing innate immune responses and migration of biliary epithelial cells.
Collapse
Affiliation(s)
- Apisit Ruangsuwast
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Michael J Smout
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, D.C., USA
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sujittra Chaiyadet
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand *Address for correspondence: Sujittra Chaiyadet, Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand. E-mail
| |
Collapse
|
3
|
Ruangsuwast A, Smout MJ, Brindley PJ, Loukas A, Laha T, Chaiyadet S. Tetraspanins from Opisthorchis viverrini stimulate cholangiocyte migration and inflammatory cytokine production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544604. [PMID: 37398394 PMCID: PMC10312640 DOI: 10.1101/2023.06.12.544604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The liver fluke Opsithorchis viverrini secretes extracellular vesicles (EVs) bearing CD63-like tetraspanins on their surface. Fluke EVs are actively internalized by host cholangiocytes in the bile ducts, where they drive pathology and promote neoplasia through induction of cellular proliferation and secretion of inflammatory cytokines. We investigated the effects of tetraspanins of the CD63 superfamily by co-culturing recombinant forms of the large extracellular loop (LEL) of O. viverrini tetraspanin-2 (rLEL- Ov -TSP-2) and tetraspanin-3 (rLEL- Ov -TSP-3) with non-cancerous human bile duct (H69) and cholangiocarcinoma (CCA, M213) cell lines. The results showed that cell lines co-cultured with excretory/secretory products from adult O. viverrini ( Ov- ES) underwent significantly increased cell proliferation at 48 hours but not 24 hours compared to untreated control cells ( P <0.05), whereas rLEL- Ov -TSP-3 co-culture resulted in significantly increased cell proliferation at both 24 hr ( P <0.05) and 48 hr ( P <0.01) time points. In like fashion, H69 cholangiocytes co-cultured with both Ov -ES and rLEL- Ov -TSP-3 underwent significantly elevated Il-6 and Il-8 gene expression for at least one of the time points assessed. Finally, both rLEL- Ov -TSP-and rLEL- Ov -TSP-3 significantly enhanced migration of both M213 and H69 cell lines. These findings indicated that O. viverrini CD63 family tetraspanins can promote a cancerous microenvironment by enhancing innate immune responses and migration of biliary epithelial cells.
Collapse
Affiliation(s)
- Apisit Ruangsuwast
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Michael J. Smout
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, 4878, Australia
| | - Paul J. Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, D.C. 20037, USA
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, 4878, Australia
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sujittra Chaiyadet
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002 Thailand
| |
Collapse
|
4
|
Chaiyadet S, Sotillo J, Smout M, Cooper M, Doolan DL, Waardenberg A, Eichenberger RM, Field M, Brindley PJ, Laha T, Loukas A. Small extracellular vesicles but not microvesicles from Opisthorchis viverrini promote cell proliferation in human cholangiocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.540805. [PMID: 37292777 PMCID: PMC10245807 DOI: 10.1101/2023.05.22.540805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Chronic infection with O. viverrini has been linked to the development of cholangiocarcinoma (CCA), which is a major public health burden in the Lower Mekong River Basin countries, including Thailand, Lao PDR, Vietnam and Cambodia. Despite its importance, the exact mechanisms by which O. viverrini promotes CCA are largely unknown. In this study, we characterized different extracellular vesicle populations released by O. viverrini (OvEVs) using proteomic and transcriptomic analyses and investigated their potential role in host-parasite interactions. While 120k OvEVs promoted cell proliferation in H69 cells at different concentrations, 15k OvEVs did not produce any effect compared to controls. The proteomic analysis of both populations showed differences in their composition that could contribute to this differential effect. Furthermore, the miRNAs present in 120k EVs were analysed and their potential interactions with human host genes was explored by computational target prediction. Different pathways involved in inflammation, immune response and apoptosis were identified as potentially targeted by the miRNAs present in this population of EVs. This is the first study showing specific roles for different EV populations in the pathogenesis of a parasitic helminth, and more importantly, an important advance towards deciphering the mechanisms used in establishment of opisthorchiasis and liver fluke infection-associated malignancy.
Collapse
Affiliation(s)
- Sujittra Chaiyadet
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Javier Sotillo
- Parasitology Reference and Research Laboratory, Centro Nacional de Microbiologia, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Michael Smout
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Martha Cooper
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Denise L Doolan
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Ashley Waardenberg
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
- Current affiliation: i-Synapse, Cairns, QLD, Australia
| | - Ramon M Eichenberger
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Matt Field
- Centre for Tropical Bioinformatics and Molecular Biology, College of Public Health, Medical and Veterinary Science, James Cook University, Cairns, Australia
- Immunogenomics Lab, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Thailand
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| |
Collapse
|
5
|
Hembasat T, Chaiyadet S, Ittiprasert W, Smout MJ, Young ND, Loukas A, Brindley PJ, Laha T. Peptide derived from progranulin of the carcinogenic liver fluke, Opisthorchis viverrini stimulates cell hyperproliferation and proinflammatory cytokine production. RESEARCH SQUARE 2023:rs.3.rs-2586058. [PMID: 36993607 PMCID: PMC10055533 DOI: 10.21203/rs.3.rs-2586058/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Purpose Progranulin (PGRN) is a secreted glycoprotein growth factor with roles in wound healing, inflammation, angiogenesis and malignancy. An orthologue of the gene encoding human PGRN was identified in the carcinogenic liver fluke Opisthorchis viverrini. Methods Sequence structure, general characteristics and possible function of O. viverrini PGRN was analyzed using bioinformatics. Expression profiles were investigated with quantitative RT-PCR, western blot and immunolocalization. A specific peptide of Ov-PGRN was used to investigate a role for this molecule in pathogenesis. Results The structure of the gene coding for O. viverrini PGRN was 36,463 bp in length, and comprised of 13 exons, 12 introns, and a promoter sequence. The Ov-pgrn mRNA is 2,768 bp in length and encodes an 846 amino acids with a predicted molecular mass of 91.61 kDa. Ov-PGRN exhibited one half and seven complete granulin domains. Phylogenetic analysis revealed that Ov-PGRN formed its closest relationship with PGRN of liver flukes in the Opisthorchiidae. Transcripts of Ov-pgrn were detected in several developmental stages, with highest expression in the metacercaria, indicating that Ov-PGRN may participate as a growth factor in the early development of O. viverrini. Western blot analysis revealed the presence of detected Ov-PGRN in both soluble somatic or excretory/secretory products, and immunolocalization indicated high levels of expression in the tegument and parenchyma of the adult fluke. Co-culture of a human cholangiocyte cell line and a peptide fragment of Ov-PGRN stimulated proliferation of cholangiocytes and upregulation of expression of the cytokines IL6 and IL8. Conclusion Ov-PGRN is expressed throughout the life cycle of liver fluke, and likely plays a key role in development and growth.
Collapse
|
6
|
Wound healing approach based on excretory-secretory product and lysate of liver flukes. Sci Rep 2022; 12:21639. [PMID: 36517588 PMCID: PMC9751068 DOI: 10.1038/s41598-022-26275-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Exogenous bioactive peptides are considered promising for the wound healing therapy in humans. In this regard, parasitic trematodes proteins may potentially become a new perspective agents. Foodborne trematode Opisthorchis felineus is widespread in Europe and has the ability to stimulate proliferation of bile duct epithelium. In this study, we investigated skin wound healing potential of O. felineus proteins in mouse model. C57Bl/6 mice were inflicted with superficial wounds with 8 mm diameter. Experimental groups included several non-specific controls and specific treatment groups (excretory-secretory product and lysate). After 10 days of the experiment, the percentage of wound healing in the specific treatment groups significantly exceeded the control values. We also found that wound treatment with excretory-secretory product and worm lysate resulted in: (i) inflammation reducing, (ii) vascular response modulating, (iii) type 1 collagen deposition promoting dermal ECM remodeling. An additional proteomic analysis of excretory-secretory product and worm lysate samples was revealed 111 common proteins. The obtained data indicate a high wound-healing potential of liver fluke proteins and open prospects for further research as new therapeutic approaches.
Collapse
|
7
|
Pakharukova MY, Mordvinov VA. Similarities and differences among the Opisthorchiidae liver flukes: insights from Opisthorchis felineus. Parasitology 2022; 149:1306-1318. [PMID: 35570685 PMCID: PMC11010525 DOI: 10.1017/s0031182022000397] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/12/2022] [Accepted: 03/19/2022] [Indexed: 11/08/2022]
Abstract
The foodborne liver trematode Opisthorchis felineus (Rivolta, 1884) is a member of the triad of phylogenetically related epidemiologically important Opisthorchiidae trematodes, which also includes O. viverrini (Poirier, 1886) and Clonorchis sinensis (Loos, 1907). Despite similarity in the life cycle, Opisthorchiidae liver flukes also have marked differences. Two species (O. viverrini and C. sinensis) are recognized as Group 1A biological carcinogens, whereas O. felineus belongs to Group 3A. In this review, we focus on these questions: Are there actual differences in carcinogenicity among these 3 liver fluke species? Is there an explanation for these differences? We provide a recent update of our knowledge on the liver fluke O. felineus and highlight its differences from O. viverrini and C. sinensis. In particular, we concentrate on differences in the climate of endemic areas, characteristics of the life cycle, the range of intermediate hosts, genomic and transcriptomic features of the pathogens, and clinical symptoms and morbidity of the infections in humans. The discussion of these questions can stimulate new developments in comparative studies on the pathogenicity of liver flukes and should help to identify species-specific features of opisthorchiasis and clonorchiasis pathogenesis.
Collapse
Affiliation(s)
- Maria Y. Pakharukova
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), 10 Akad. Lavrentieva Ave., Novosibirsk 630090, Russia
- Department of Natural Sciences, Novosibirsk State University, 2 Pirogova Str., Novosibirsk 630090, Russia
| | - Viatcheslav A. Mordvinov
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), 10 Akad. Lavrentieva Ave., Novosibirsk 630090, Russia
| |
Collapse
|
8
|
Chaiyadet S, Tangkawattana S, Smout MJ, Ittiprasert W, Mann VH, Deenonpoe R, Arunsan P, Loukas A, Brindley PJ, Laha T. Knockout of liver fluke granulin, Ov-grn-1, impedes malignant transformation during chronic infection with Opisthorchis viverrini. PLoS Pathog 2022; 18:e1010839. [PMID: 36137145 PMCID: PMC9531791 DOI: 10.1371/journal.ppat.1010839] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/04/2022] [Accepted: 08/29/2022] [Indexed: 01/21/2023] Open
Abstract
Infection with the food-borne liver fluke Opisthorchis viverrini is the principal risk factor for cholangiocarcinoma (CCA) in the Mekong Basin countries of Thailand, Lao PDR, Vietnam, Myanmar and Cambodia. Using a novel model of CCA, involving infection with gene-edited liver flukes in the hamster during concurrent exposure to dietary nitrosamine, we explored the role of the fluke granulin-like growth factor Ov-GRN-1 in malignancy. We derived RNA-guided gene knockout flukes (ΔOv-grn-1) using CRISPR/Cas9/gRNA materials delivered by electroporation. Genome sequencing confirmed programmed Cas9-catalyzed mutations of the targeted genes, which was accompanied by rapid depletion of transcripts and the proteins they encode. Gene-edited parasites colonized the biliary tract of hamsters and developed into adult flukes. However, less hepatobiliary tract disease manifested during chronic infection with ΔOv-grn-1 worms in comparison to hamsters infected with control gene-edited and mock-edited parasites. Specifically, immuno- and colorimetric-histochemical analysis of livers revealed markedly less periductal fibrosis surrounding the flukes and less fibrosis globally within the hepatobiliary tract during infection with ΔOv-grn-1 genotype worms, minimal biliary epithelial cell proliferation, and significantly fewer mutations of TP53 in biliary epithelial cells. Moreover, fewer hamsters developed high-grade CCA compared to controls. The clinically relevant, pathophysiological phenotype of the hepatobiliary tract confirmed a role for this secreted growth factor in malignancy and morbidity during opisthorchiasis.
Collapse
Affiliation(s)
- Sujittra Chaiyadet
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Tropical Medicine Graduate Program, Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sirikachorn Tangkawattana
- Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand, and WHO Collaborating Center for Research and Control of Opisthorchiasis, Tropical Disease Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - Michael J. Smout
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Wannaporn Ittiprasert
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, District of Columbia, United States of America
| | - Victoria H. Mann
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, District of Columbia, United States of America
| | - Raksawan Deenonpoe
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Patpicha Arunsan
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, District of Columbia, United States of America
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Paul J. Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, District of Columbia, United States of America
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
9
|
Pakharukova MY, Zaparina O, Baginskaya NV, Mordvinov VA. Global changes in gene expression related to Opisthorchis felineus liver fluke infection reveal temporal heterogeneity of a mammalian host response. Food Waterborne Parasitol 2022; 27:e00159. [PMID: 35542180 PMCID: PMC9079687 DOI: 10.1016/j.fawpar.2022.e00159] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/03/2022] Open
Abstract
The food-borne trematode Opisthorchis felineus colonizes bile ducts of the liver of fish-eating mammals including humans. Among chronically infected individuals, this opisthorchiasis involves hepatobiliary problems, including chronic inflammation, periductal fibrosis, biliary intraepithelial neoplasia, and even cholangiocarcinoma. Despite numerous studies at the pathomorphological level, the systemic response and cellular pathogenesis of these disorders are not well studied. To conduct in-depth research and to gain insights into the mechanism by which O. felineus infection causes precancerous liver lesions, we (i) applied a next-generation-sequencing–based technology (high-throughput mRNA sequencing) to identify differentially expressed genes in the liver of golden hamsters infected with O. felineus at 1 and 3 months postinfection and (ii) verified the most pronounced changes in gene expression by western blotting and immunohistochemistry. A total of 2151 genes were found to be differentially expressed between uninfected and infected hamsters (“infection” factor), whereas 371 genes were differentially expressed when we analyzed “time × infection” interaction. Cluster analysis revealed that sets of activated genes of cellular pathways were different between acute (1 month postinfection) and chronic (3 months postinfection) opisthorchiasis. This enriched KEGG pathways were “Cell adhesion molecules”, “Hippo signaling”, “ECM-receptor interaction”, “Cell cycle”, “TGF-beta”, and “P53 signaling”. Moreover, epithelial–mesenchymal transition was the most enriched (q-value = 2.2E-07) MSigDB hallmark in the set of differentially expressed genes of all O. felineus–infected animals. Transcriptomic data were supported by the results of western blotting and immunohistochemistry revealing the upregulation of vimentin, N-cadherin, and α-smooth muscle actin postinfection. Our data expand knowledge about global changes in gene expression in the O. felineus–infected host liver and contribute to understanding the biliary neoplasia associated with the liver fluke infection. A total of 2151 genes were found to be differentially expressed during the infection. Epithelial-Mesenchymal Transition was the most enriched process in the infected animals. Different pathways were activated between acute and chronic opisthorchiasis.
Collapse
|
10
|
Ceci L, Zhou T, Lenci I, Meadows V, Kennedy L, Li P, Ekser B, Milana M, Zhang W, Wu C, Sato K, Chakraborty S, Glaser SS, Francis H, Alpini G, Baiocchi L. Molecular Mechanisms Linking Risk Factors to Cholangiocarcinoma Development. Cancers (Basel) 2022; 14:1442. [PMID: 35326593 PMCID: PMC8945938 DOI: 10.3390/cancers14061442] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/01/2023] Open
Abstract
The poor prognosis of cholangiocarcinoma in humans is related to several factors, such as (i) the heterogeneity of the disease, (ii) the late onset of symptoms and (iii) the limited comprehension of the carcinogenic pathways determining neoplastic changes, which all limit the pursuit of appropriate treatment. Several risk factors have been recognized, including different infective, immune-mediated, and dysmorphogenic disorders of the biliary tree. In this review, we report the details of possible mechanisms that lead a specific premalignant pathological condition to become cholangiocarcinoma. For instance, during liver fluke infection, factors secreted from the worms may play a major role in pathogenesis. In primary sclerosing cholangitis, deregulation of histamine and bile-acid signaling may determine important changes in cellular pathways. The study of these molecular events may also shed some light on the pathogenesis of sporadic (unrelated to risk factors) forms of cholangiocarcinoma, which represent the majority (nearly 75%) of cases.
Collapse
Affiliation(s)
- Ludovica Ceci
- Hepatology and Gastroenterology Division, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.C.); (T.Z.); (V.M.); (L.K.); (K.S.); (H.F.)
| | - Tianhao Zhou
- Hepatology and Gastroenterology Division, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.C.); (T.Z.); (V.M.); (L.K.); (K.S.); (H.F.)
| | - Ilaria Lenci
- Unit of Hepatology, Tor Vergata University, 00133 Rome, Italy; (I.L.); (M.M.)
| | - Vik Meadows
- Hepatology and Gastroenterology Division, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.C.); (T.Z.); (V.M.); (L.K.); (K.S.); (H.F.)
| | - Lindsey Kennedy
- Hepatology and Gastroenterology Division, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.C.); (T.Z.); (V.M.); (L.K.); (K.S.); (H.F.)
- Department of Research, Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Ping Li
- Department of Surgery, Division of Transplant Surgery, Indiana University, Indianapolis, IN 46202, USA; (P.L.); (B.E.); (W.Z.)
| | - Burcin Ekser
- Department of Surgery, Division of Transplant Surgery, Indiana University, Indianapolis, IN 46202, USA; (P.L.); (B.E.); (W.Z.)
| | - Martina Milana
- Unit of Hepatology, Tor Vergata University, 00133 Rome, Italy; (I.L.); (M.M.)
| | - Wenjun Zhang
- Department of Surgery, Division of Transplant Surgery, Indiana University, Indianapolis, IN 46202, USA; (P.L.); (B.E.); (W.Z.)
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA;
| | - Keisaku Sato
- Hepatology and Gastroenterology Division, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.C.); (T.Z.); (V.M.); (L.K.); (K.S.); (H.F.)
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA; (S.C.); (S.S.G.)
| | - Shannon S. Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA; (S.C.); (S.S.G.)
| | - Heather Francis
- Hepatology and Gastroenterology Division, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.C.); (T.Z.); (V.M.); (L.K.); (K.S.); (H.F.)
- Department of Research, Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Gianfranco Alpini
- Hepatology and Gastroenterology Division, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.C.); (T.Z.); (V.M.); (L.K.); (K.S.); (H.F.)
- Department of Research, Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Leonardo Baiocchi
- Unit of Hepatology, Tor Vergata University, 00133 Rome, Italy; (I.L.); (M.M.)
| |
Collapse
|
11
|
Drurey C, Lindholm HT, Coakley G, Poveda MC, Löser S, Doolan R, Gerbe F, Jay P, Harris N, Oudhoff MJ, Maizels RM. Intestinal epithelial tuft cell induction is negated by a murine helminth and its secreted products. J Exp Med 2022; 219:e20211140. [PMID: 34779829 PMCID: PMC8597987 DOI: 10.1084/jem.20211140] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/20/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023] Open
Abstract
Helminth parasites are adept manipulators of the immune system, using multiple strategies to evade the host type 2 response. In the intestinal niche, the epithelium is crucial for initiating type 2 immunity via tuft cells, which together with goblet cells expand dramatically in response to the type 2 cytokines IL-4 and IL-13. However, it is not known whether helminths modulate these epithelial cell populations. In vitro, using small intestinal organoids, we found that excretory/secretory products (HpES) from Heligmosomoides polygyrus blocked the effects of IL-4/13, inhibiting tuft and goblet cell gene expression and expansion, and inducing spheroid growth characteristic of fetal epithelium and homeostatic repair. Similar outcomes were seen in organoids exposed to parasite larvae. In vivo, H. polygyrus infection inhibited tuft cell responses to heterologous Nippostrongylus brasiliensis infection or succinate, and HpES also reduced succinate-stimulated tuft cell expansion. Our results demonstrate that helminth parasites reshape their intestinal environment in a novel strategy for undermining the host protective response.
Collapse
Affiliation(s)
- Claire Drurey
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Håvard T. Lindholm
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Gillian Coakley
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Marta Campillo Poveda
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Stephan Löser
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Rory Doolan
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - François Gerbe
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre national de la recherche scientifique UMR-5203, Institut National de la Santé et de la Recherche Médicale U1191, Montpellier, France
| | - Philippe Jay
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre national de la recherche scientifique UMR-5203, Institut National de la Santé et de la Recherche Médicale U1191, Montpellier, France
| | - Nicola Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Menno J. Oudhoff
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| |
Collapse
|
12
|
Wang C, He Q, Yin Y, Wu Y, Li X. Clonorchis sinensis Granulin Promotes Malignant Transformation of Hepatocyte Through EGFR-Mediated RAS/MAPK/ERK and PI3K/Akt Signaling Pathways. Front Cell Infect Microbiol 2021; 11:734750. [PMID: 34858869 PMCID: PMC8631275 DOI: 10.3389/fcimb.2021.734750] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
The biological functions of growth factor, such as granulins, have been explored in parasites, and we elucidated that Clonorchis sinensis granulin (CsGRN) promoted the metastasis of hepatocellular carcinoma in our previous study. However, it is still unclear for the malignant transformation role of CsGRN in normal human hepatocytes. In this study, by transfecting pEGFP-C1-CsGRN eukaryotic expression plasmid, a cell line with stable overexpression of CsGRN in normal hepatocyte (LO2-GRN cells) was constructed. The effects on cell proliferation were detected by carrying out cell counting kit-8 (CCK8) assay and colony formation assay. Additionally, we conducted flow cytometry analysis to determine whether the proliferation of CsGRN was due to cell cycle arrest. Subsequently, the migration ability and the invasion ability of LO2-GRN cells were evaluated through wound-healing assay and transwell assay. Meanwhile, the levels of the markers of RAS/MAPK/ERK and PI3K/Akt signaling pathways activation in LO2-GRN cells were assessed by quantitative RT-PCR and Western blot. Our results indicated that CsGRN promoted the proliferation of LO2 cells by regulating the expression of cell-cycle-related genes. Moreover, the overexpression of CsGRN regulates malignant metastasis of liver cells by inducing the upregulation of epithelial-mesenchymal transition (EMT) marker proteins. Furthermore, both mRNA and protein expression levels of p-EGFR, RAS, p-ERK, p-AKT, p-PI3K, and p-braf have been enhanced by CsGRN. These results showed that CsGRN promoted the malignant transformation of hepatocytes by regulating epidermal growth factor receptor (EGFR)-mediated RAS/MAPK/ERK and PI3K/Akt signaling pathways, which suggested that CsGRN could serve as a novel oncoprotein during Clonorchis sinensis-associated malignant transformation of hepatocytes.
Collapse
Affiliation(s)
- Caiqin Wang
- Department of Medical Oncology, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China.,China Atomic Energy Authority (CAEA) Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, China
| | - Qing He
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China.,China Atomic Energy Authority (CAEA) Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, China
| | - Yingxuan Yin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China.,China Atomic Energy Authority (CAEA) Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, China
| | - Yinjuan Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China.,China Atomic Energy Authority (CAEA) Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, China
| | - Xuerong Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China.,China Atomic Energy Authority (CAEA) Center of Excellence on Nuclear Technology Applications for Insect Control, Beijing, China
| |
Collapse
|
13
|
Arai T, Lopes F. Potential of human helminth therapy for resolution of inflammatory bowel disease: The future ahead. Exp Parasitol 2021; 232:108189. [PMID: 34848244 DOI: 10.1016/j.exppara.2021.108189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/06/2021] [Accepted: 11/24/2021] [Indexed: 12/23/2022]
Abstract
Inflammatory bowel disease (IBD) is associated with a dysregulated mucosal immune response in the gastrointestinal tract. The number of patients with IBD has increased worldwide, especially in highly industrialized western societies. The population of patients with IBD in North America is forecasted to reach about four million by 2030; meanwhile, there is no definitive therapy for IBD. Current anti-inflammatory, immunosuppressive, or biological treatment may induce and maintain remission, but not all patients respond to these treatments. Recent studies explored parasitic helminths as a novel modality of therapy due to their potent immunoregulatory properties in humans. Research using IBD animal models infected with a helminth or administered helminth-derived products such as excretory-secretory products has been promising, and helminth-microbiota interactions exert their anti-inflammatory effects by modulating the host immunity. Recent studies also indicate that evidence that helminth-derived metabolites may play a role in anticolitic effects. Thus, the helminth shows a potential benefit for treatment against IBD. Here we review the current feasibility of "helminth therapy" from the laboratory for application in IBD management.
Collapse
Affiliation(s)
- Toshio Arai
- Institution of Parasitology, McGill University, Quebec, Canada; Department of Gastroenterology, Hashimoto Municipal Hospital, Wakayama, Japan
| | - Fernando Lopes
- Institution of Parasitology, McGill University, Quebec, Canada.
| |
Collapse
|
14
|
Brindley PJ, Bachini M, Ilyas SI, Khan SA, Loukas A, Sirica AE, Teh BT, Wongkham S, Gores GJ. Cholangiocarcinoma. Nat Rev Dis Primers 2021; 7:65. [PMID: 34504109 PMCID: PMC9246479 DOI: 10.1038/s41572-021-00300-2] [Citation(s) in RCA: 328] [Impact Index Per Article: 109.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/03/2021] [Indexed: 02/08/2023]
Abstract
Cholangiocarcinoma (CCA) is a highly lethal adenocarcinoma of the hepatobiliary system, which can be classified as intrahepatic, perihilar and distal. Each anatomic subtype has distinct genetic aberrations, clinical presentations and therapeutic approaches. In endemic regions, liver fluke infection is associated with CCA, owing to the oncogenic effect of the associated chronic biliary tract inflammation. In other regions, CCA can be associated with chronic biliary tract inflammation owing to choledocholithiasis, cholelithiasis, or primary sclerosing cholangitis, but most CCAs have no identifiable cause. Administration of the anthelmintic drug praziquantel decreases the risk of CCA from liver flukes, but reinfection is common and future vaccination strategies may be more effective. Some patients with CCA are eligible for potentially curative surgical options, such as resection or liver transplantation. Genetic studies have provided new insights into the pathogenesis of CCA, and two aberrations that drive the pathogenesis of non-fluke-associated intrahepatic CCA, fibroblast growth factor receptor 2 fusions and isocitrate dehydrogenase gain-of-function mutations, can be therapeutically targeted. CCA is a highly desmoplastic cancer and targeting the tumour immune microenvironment might be a promising therapeutic approach. CCA remains a highly lethal disease and further scientific and clinical insights are needed to improve patient outcomes.
Collapse
Affiliation(s)
- Paul J. Brindley
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| | | | - Sumera I. Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Shahid A. Khan
- Liver Unit, Division of Digestive Diseases, Imperial College London, London, UK
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Alphonse E. Sirica
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre, Singapore, Singapore
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA,
| |
Collapse
|
15
|
Machicado C, Soto MP, La Chira LF, Torres J, Mendoza C, Marcos LA. In silico prediction of secretory proteins of Opisthorchis viverrini, Clonorchis sinensis and Fasciola hepatica that target the host cell nucleus. Heliyon 2021; 7:e07204. [PMID: 34337171 PMCID: PMC8318992 DOI: 10.1016/j.heliyon.2021.e07204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/21/2021] [Accepted: 05/31/2021] [Indexed: 12/01/2022] Open
Abstract
Liver flukes Fasciola hepatica, Opisthorchis viverrini and Clonorchis sinensis are causing agents of liver and hepatobiliary diseases. A remarkable difference between such worms is the fact that O. viverrini and C. sinensis are carcinogenic organisms whereas F. hepatica is not carcinogenic. The release of secretory factors by carcinogenic flukes seems to contribute to cancer development however if some of these target the host cell nuclei is unknown. We investigated the existence of O. viverrini and C. sinensis secretory proteins that target the nucleus of host cells and compared these with the corresponding proteins predicted in F. hepatica. Here we applied an algorithm composed by in silico approaches that screened and analyzed the potential genes predicted from genomes of liver flukes. We found 31 and 22 secretory proteins that target the nucleus of host cells in O. viverrini and C. sinensis, respectively, and that have no homologs in F. hepatica. These polypeptides have enriched the transcription initiation process and nucleic acid binding in O. viverrini and C. sinensis, respectively. In addition, other 11 secretory proteins of O. viverrini and C. sinensis, that target the nucleus of host cells, had F. hepatica homologs, have enriched RNA processing function. In conclusion, O. viverrini and C. sinensis have 31 and 22 genes, respectively, that may be involved in their carcinogenic action through a direct targeting on the host cell nuclei.
Collapse
Affiliation(s)
- Claudia Machicado
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Honorio Delgado 430, Lima 31, Peru.,Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, Spain
| | - Maria Pia Soto
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Honorio Delgado 430, Lima 31, Peru.,Laboratorio de Investigación en Biología Molecular y Farmacología Experimental, Universidad Católica de Santa María, Urb. San José, San Jose s/n, Arequipa, Peru
| | - Luis Felipe La Chira
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Honorio Delgado 430, Lima 31, Peru
| | - Joel Torres
- Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Carlos Germán Amezaga 375, Cercado de Lima, Peru
| | - Carlos Mendoza
- Facultad de Ciencias Biológicas, Universidad Nacional de Trujillo, Av. Juan Pablo II, Trujillo, 13011, Peru
| | - Luis A Marcos
- Department of Medicine (Division of Infectious Diseases), Department of Microbiology and Immunology, State University of New York at Stony Brook, NY, Stony Brook, USA
| |
Collapse
|
16
|
Kim JW, Yi J, Park J, Jeong JH, Kim J, Won J, Chung S, Kim TS, Pak JH. Transcriptomic profiling of three-dimensional cholangiocyte spheroids long term exposed to repetitive Clonorchis sinensis excretory-secretory products. Parasit Vectors 2021; 14:213. [PMID: 33879231 PMCID: PMC8056535 DOI: 10.1186/s13071-021-04717-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/01/2021] [Indexed: 12/16/2022] Open
Abstract
Background Biliary tract infection with the carcinogenic human liver fluke, Clonorchis sinensis, provokes chronic inflammation, epithelial hyperplasia, periductal fibrosis, and even cholangiocarcinoma. Complications are proportional to the intensity and duration of the infection. In addition to mechanical irritation of the biliary epithelia from worms, their excretory-secretory products (ESPs) cause chemical irritation, which leads to inflammation, proliferation, and free radical generation. Methods A three-dimensional in vitro cholangiocyte spheroid culture model was established, followed by ESP treatment. This allowed us to examine the intrinsic pathological mechanisms of clonorchiasis via the imitation of prolonged and repetitive in vivo infection. Results Microarray and RNA-Seq analysis revealed that ESP-treated cholangiocyte H69 spheroids displayed global changes in gene expression compared to untreated spheroids. In ESP-treated H69 spheroids, 185 and 63 probes were found to be significantly upregulated and downregulated, respectively, corresponding to 209 genes (p < 0.01, fold change > 2). RNA-Seq was performed for the validation of the microarray results, and the gene expression patterns in both transcriptome platforms were well matched for 209 significant genes. Gene ontology analysis demonstrated that differentially expressed genes were mainly classified into immune system processes, the extracellular region, and the extracellular matrix. Among the upregulated genes, four genes (XAF1, TRIM22, CXCL10, and BST2) were selected for confirmation using quantitative RT-PCR, resulting in 100% similar expression patterns in microarray and RNA-Seq. Conclusions These findings broaden our understanding of the pathological pathways of liver fluke-associated hepatobiliary disorders and suggest a novel therapeutic strategy for this infectious cancer. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-04717-2.
Collapse
Affiliation(s)
- Jung-Woong Kim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Junyeong Yi
- Department of Convergence Medicine, University of Ulsan College of Medicine and Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jinhong Park
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ji Hoon Jeong
- Department of Convergence Medicine, University of Ulsan College of Medicine and Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jinho Kim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jihee Won
- School of Mechanical Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Tong-Soo Kim
- Department of Tropical Medicine and Parasitology, Inha University School of Medicine, Incheon, 22212, Republic of Korea
| | - Jhang Ho Pak
- Department of Convergence Medicine, University of Ulsan College of Medicine and Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea.
| |
Collapse
|
17
|
da Costa JMC, Gouveia MJ, Rinaldi G, Brindley PJ, Santos J, Santos LL. Control Strategies for Carcinogenic-Associated Helminthiases: An Integrated Overview. Front Cell Infect Microbiol 2021; 11:626672. [PMID: 33842386 PMCID: PMC8025785 DOI: 10.3389/fcimb.2021.626672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/26/2021] [Indexed: 12/20/2022] Open
Abstract
Helminthiases are extremely prevalent in the developing world. In addition, the chronic infection with some parasitic worms are classified as carcinogenic. Therefore, it is utmost importance to understand the parasite-host interactions, the mechanisms underlay carcinogenesis and how they could be counteracted. This knowledge may ultimately guide novel control strategies that include chemotherapy-based approaches targeting these pathogens and associated pathologies caused by their infections. Little is known on how some helminthiases are associated with cancer; however, it has been hypothesized that chemical carcinogenesis may be involved in the process. Here, we summarize the current knowledge on chemical carcinogenesis associated with helminthiases, along with available therapeutic options and potential therapeutic alternatives including chemotherapy and/or immunotherapy. Ideally, the treatment of the carcinogenic helminthiases should target both the parasite and associated pathologies. The success of any chemotherapeutic regimen often depends on the host immune response during the infection and nutritional status among other factors. The close association between chemotherapy and cell-mediated immunity suggests that a dual therapeutic approach would be advantageous. In addition, there is a pressing need for complementary drugs that antagonize the carcinogenesis process associated with the helminth infections.
Collapse
Affiliation(s)
- José Manuel Correia da Costa
- Centre for the Study in Animal Science (CECA/ICETA), University of Porto, Porto, Portugal
- Centre for Parasite Immunology and Biology, Department of Infectious Diseases, National Institute for Health Dr Ricardo Jorge, Porto, Portugal
| | - Maria João Gouveia
- Centre for the Study in Animal Science (CECA/ICETA), University of Porto, Porto, Portugal
- Centre for Parasite Immunology and Biology, Department of Infectious Diseases, National Institute for Health Dr Ricardo Jorge, Porto, Portugal
- REQUIMTE, Department of Chemical Sciences, Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | | | - Paul J. Brindley
- Department of Microbiology, Immunology & Tropical Medicine, and Research Centre for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, United States
| | - Júlio Santos
- Deparment of Urology, Clínica da Sagrada Esperança, Luanda, Angola
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center of Instituto Português de Oncologia, Porto, Portugal
| |
Collapse
|
18
|
Mordvinov VA, Ershov NI, Zaparina OG, Pakharukova MY. Genomics and proteomics of the liver fluke Opisthorchis felineus. Vavilovskii Zhurnal Genet Selektsii 2021; 24:383-390. [PMID: 33659821 PMCID: PMC7716572 DOI: 10.18699/vj20.44-o] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
The causative agent of opisthorchiasis, the liver fluke Opisthorchis felineus (Rivolta, 1884) is one of the helminths of humans and animals in Russia. Together with closely related species of trematodes O. viverrini (Poirier, 1886) and Clonorchis sinensis (Loos, 1907), O. felineus is a part of a triad of epidemiologically important trematodes in the family Opisthorchiidae. Adult O. felineus worms infest the hepatobiliary system of warm-blooded animals and might provoke the development of severe pathologies, including malignancy of bile duct epithelium. The high medical importance of O. felineus attracts the attention of researchers. This review briefly summarizes the data about O. felineus genomics and proteomics. The review provides a comparative analysis of the number of genes and sizes of nuclear genomes of a number of flatworms, the distribution of intron lengths, as well as results of synteny between the O. felineus, O. viverrini and C. sinensis genomes. Special attention is paid to a particular form of RNA processing known as trans-splicing, widely presented in the opisthorchiid genomes. We also provide the results of a comparative analysis of the xenobiotic metabolizing system between parasitic and free-living flatworms. Moreover, data on parasitic granulins, which are potential promoters of cholangiocyte neoplasia, are also presented. Data on the O. felineus genomics and proteomics provide first insights into the structural and functional organization of the genome of this parasitic flatworm with a complex life cycle as well as provide a significant contribution to our understanding of “host-parasite” interaction and evolution of this group of parasitic flatworms.
Collapse
Affiliation(s)
- V A Mordvinov
- Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - N I Ershov
- Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - O G Zaparina
- Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - M Y Pakharukova
- Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
19
|
Porras-Silesky C, Mejías-Alpízar MJ, Mora J, Baneth G, Rojas A. Spirocerca lupi Proteomics and Its Role in Cancer Development: An Overview of Spirocercosis-Induced Sarcomas and Revision of Helminth-Induced Carcinomas. Pathogens 2021; 10:pathogens10020124. [PMID: 33530324 PMCID: PMC7911836 DOI: 10.3390/pathogens10020124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/19/2022] Open
Abstract
Spirocerca lupi is a parasitic nematode of canids that induces a myriad of clinical manifestations in its host and, in 25% of infections, leads to the formation of sarcomas. The description of the protein composition of the excretory and secretory products (Sl-ESP) of S. lupi has shed light on its possible interactions with the host environment, including migration within the host and mechanisms of immunomodulation. Despite this, the process by which S. lupi induces cancer in the dog remains poorly understood, and some hypotheses have arisen regarding these possible mechanisms. In this review, we discuss the role of specific ESP from the carcinogenic helminths Clonorchis sinensis, Opisthorchis viverrini and Schistosoma haematobium in inducing chronic inflammation and cancer in their host’s tissues. The parasitic worms Taenia solium, Echinococcus granulosus, Heterakis gallinarum, Trichuris muris and Strongyloides stercoralis, which have less-characterized mechanisms of cancer induction, are also analyzed. Based on the pathological findings in spirocercosis and the mechanisms by which other parasitic helminths induce cancer, we propose that the sustained inflammatory response in the dog´s tissues produced in response to the release of Sl-ESP homologous to those of other carcinogenic worms may lead to the malignant process in infected dogs.
Collapse
Affiliation(s)
- Catalina Porras-Silesky
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
| | - María José Mejías-Alpízar
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
| | - Javier Mora
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
| | - Gad Baneth
- Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot 7610001, Israel;
| | - Alicia Rojas
- Laboratory of Helminthology, Centro de Investigación en Enfermedades Tropicales, University of Costa Rica, 11501-2060 San José, Costa Rica; (C.P.-S.); (M.J.M.-A.); (J.M.)
- Correspondence: ; Tel.: +506-2511-8644
| |
Collapse
|
20
|
Avgustinovich D, Kovner A, Kashina E, Shatskaya N, Vishnivetskaya G, Bondar N, Lvova M. The pathogenic potential of the combined action of chronic Opisthorchis felineus infection and repeated social defeat stress in C57BL/6 mice. Int J Parasitol 2020; 51:353-363. [PMID: 33378706 DOI: 10.1016/j.ijpara.2020.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022]
Abstract
Parasitic food-borne diseases and chronic social stress are frequent attributes of day-to-day human life. Therefore, our aim was to model the combined action of chronic Opisthorchis felineus infection and repeated social defeat stress in C57BL/6 mice. Histological examination of the liver revealed inflammation sites, pronounced periductal fibrosis, and cholangiofibrosis together with proliferation of bile ducts and hepatocyte dystrophy in the infected mice, especially in the stress-exposed ones. Simultaneously with liver pathology, we detected significant structural changes in the cerebral cortex. Immunohistochemical analysis of the hippocampus indicated the highest increase in numerical density of Iba 1-, IL-6-, iNOS-, and Arg1-positive cells in mice simultaneously subjected to the two adverse factors. The number of GFAP-positive cells rose during repeated social defeat stress, most strongly in the mice subjected to both infection and stress. Real-time PCR analysis showed that the expression of genes Aif1 and Il6 differed among the analysed brain regions (hippocampus, hypothalamus, and frontal cortex) and depended on the adverse factors applied. In addition, among the brain regions, there was no consistent increase or decrease in these parameters when the two adverse treatments were combined: (i) in the hippocampus, there was upregulation of Aif1 and no change in Il6 expression; (ii) in the hypothalamus, expression levels of Aif1 and Il6 were not different from controls; and (iii) in the frontal cortex, Aif1 expression did not change while Il6 expression increased. It can be concluded that a combination of two long-lasting adverse factors, O. felineus infection and repeated social defeat stress, worsens not only the hepatic but also brain state, as evidenced behaviorally by disturbances of the startle response in mice.
Collapse
Affiliation(s)
- Damira Avgustinovich
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia; Institute of Solid State Chemistry and Mechanochemistry, SB RAS, Novosibirsk, Russia.
| | - Anna Kovner
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Elena Kashina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia; AO Vector-Best, Novosibirsk, Russia
| | - Natalia Shatskaya
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Galina Vishnivetskaya
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Natalia Bondar
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia; Novosibirsk State University, Novosibirsk, Russia
| | - Maria Lvova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| |
Collapse
|
21
|
Vanhamme L, Souopgui J, Ghogomu S, Ngale Njume F. The Functional Parasitic Worm Secretome: Mapping the Place of Onchocerca volvulus Excretory Secretory Products. Pathogens 2020; 9:pathogens9110975. [PMID: 33238479 PMCID: PMC7709020 DOI: 10.3390/pathogens9110975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 01/15/2023] Open
Abstract
Nematodes constitute a very successful phylum, especially in terms of parasitism. Inside their mammalian hosts, parasitic nematodes mainly dwell in the digestive tract (geohelminths) or in the vascular system (filariae). One of their main characteristics is their long sojourn inside the body where they are accessible to the immune system. Several strategies are used by parasites in order to counteract the immune attacks. One of them is the expression of molecules interfering with the function of the immune system. Excretory-secretory products (ESPs) pertain to this category. This is, however, not their only biological function, as they seem also involved in other mechanisms such as pathogenicity or parasitic cycle (molting, for example). We will mainly focus on filariae ESPs with an emphasis on data available regarding Onchocerca volvulus, but we will also refer to a few relevant/illustrative examples related to other worm categories when necessary (geohelminth nematodes, trematodes or cestodes). We first present Onchocerca volvulus, mainly focusing on the aspects of this organism that seem relevant when it comes to ESPs: life cycle, manifestations of the sickness, immunosuppression, diagnosis and treatment. We then elaborate on the function and use of ESPs in these aspects.
Collapse
Affiliation(s)
- Luc Vanhamme
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium; (J.S.); (F.N.N.)
- Correspondence:
| | - Jacob Souopgui
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium; (J.S.); (F.N.N.)
| | - Stephen Ghogomu
- Molecular and Cell Biology Laboratory, Biotechnology Unit, University of Buea, Buea P.O Box 63, Cameroon;
| | - Ferdinand Ngale Njume
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium; (J.S.); (F.N.N.)
- Molecular and Cell Biology Laboratory, Biotechnology Unit, University of Buea, Buea P.O Box 63, Cameroon;
| |
Collapse
|
22
|
Thanaphongdecha P, Karinshak SE, Ittiprasert W, Mann VH, Chamgramol Y, Pairojkul C, Fox JG, Suttiprapa S, Sripa B, Brindley PJ. Infection with Helicobacter pylori Induces Epithelial to Mesenchymal Transition in Human Cholangiocytes. Pathogens 2020; 9:E971. [PMID: 33233485 PMCID: PMC7700263 DOI: 10.3390/pathogens9110971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 02/08/2023] Open
Abstract
Recent reports suggest that the East Asian liver fluke infection, caused by Opisthorchis viverrini, which is implicated in opisthorchiasis-associated cholangiocarcinoma, serves as a reservoir of Helicobacter pylori. The opisthorchiasis-affected cholangiocytes that line the intrahepatic biliary tract are considered to be the cell of origin of this malignancy. Here, we investigated interactions in vitro among human cholangiocytes, Helicobacter pylori strain NCTC 11637, and the congeneric bacillus, Helicobacter bilis. Exposure to increasing numbers of H. pylori at 0, 1, 10, 100 bacilli per cholangiocyte of the H69 cell line induced phenotypic changes including the profusion of thread-like filopodia and a loss of cell-cell contact, in a dose-dependent fashion. In parallel, following exposure to H. pylori, changes were evident in levels of mRNA expression of epithelial to mesenchymal transition (EMT)-encoding factors including snail, slug, vimentin, matrix metalloprotease, zinc finger E-box-binding homeobox, and the cancer stem cell marker CD44. Analysis to quantify cellular proliferation, migration, and invasion in real-time by both H69 cholangiocytes and CC-LP-1 line of cholangiocarcinoma cells using the xCELLigence approach and Matrigel matrix revealed that exposure to 10 H. pylori bacilli per cell stimulated migration and invasion by the cholangiocytes. In addition, 10 bacilli of H. pylori stimulated contact-independent colony establishment in soft agar. These findings support the hypothesis that infection by H. pylori contributes to the malignant transformation of the biliary epithelium.
Collapse
Affiliation(s)
- Prissadee Thanaphongdecha
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
- Tropical Disease Research Laboratory, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Shannon E. Karinshak
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
| | - Wannaporn Ittiprasert
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
| | - Victoria H. Mann
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
| | - Yaovalux Chamgramol
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (C.P.)
| | - Chawalit Pairojkul
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (C.P.)
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Sutas Suttiprapa
- Tropical Disease Research Laboratory, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Banchob Sripa
- Tropical Disease Research Laboratory, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (C.P.)
| | - Paul J. Brindley
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
| |
Collapse
|
23
|
Folding of Truncated Granulin Peptides. Biomolecules 2020; 10:biom10081152. [PMID: 32781704 PMCID: PMC7463432 DOI: 10.3390/biom10081152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/01/2020] [Accepted: 08/04/2020] [Indexed: 12/01/2022] Open
Abstract
Granulins are a family of unique protein growth factors which are found in a range of species and have several bioactivities that include cell proliferation and wound healing. They typically contain six disulfide bonds, but the sequences, structures and bioactivities vary significantly. We have previously shown that an N-terminally truncated version of a granulin from the human liver fluke, Opisthorchis viverrini, can fold independently into a “mini-granulin” structure and has potent wound healing properties in vivo. The incorporation of a non-native third disulfide bond, with respect to the full-length granulin module, was critical for the formation of regular secondary structure in the liver fluke derived peptide. By contrast, this third disulfide bond is not required for a carp granulin-1 truncated peptide to fold independently. This distinction led us to explore granulins from the zebrafish model organism. Here we show that the mini-granulin fold occurs in a naturally occurring paragranulin (half-domain) from zebrafish, and is also present in a truncated form of a full-length zebrafish granulin, suggesting this structure might be a common property in either naturally occurring or engineered N-terminally truncated granulins and the carp granulin-1 folding is an anomaly. The in vitro folding yield is significantly higher in the naturally occurring paragranulin, but only the truncated zebrafish granulin peptide promoted the proliferation of fibroblasts consistent with a growth factor function, and therefore the function of the paragranulin remains unknown. These findings provide insight into the folding and evolution of granulin domains and might be useful in the elucidation of the structural features important for bioactivity to aid the design of more potent and stable analogues for the development of novel wound healing agents.
Collapse
|
24
|
Ryan SM, Eichenberger RM, Ruscher R, Giacomin PR, Loukas A. Harnessing helminth-driven immunoregulation in the search for novel therapeutic modalities. PLoS Pathog 2020; 16:e1008508. [PMID: 32407385 PMCID: PMC7224462 DOI: 10.1371/journal.ppat.1008508] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Parasitic helminths have coevolved with humans over millennia, intricately refining and developing an array of mechanisms to suppress or skew the host’s immune system, thereby promoting their long-term survival. Some helminths, such as hookworms, cause little to no overt pathology when present in modest numbers and may even confer benefits to their human host. To exploit this evolutionary phenomenon, clinical trials of human helminth infection have been established and assessed for safety and efficacy for a range of immune dysfunction diseases and have yielded mixed outcomes. Studies of live helminth therapy in mice and larger animals have convincingly shown that helminths and their excretory/secretory products possess anti-inflammatory drug-like properties and represent an untapped pharmacopeia. These anti-inflammatory moieties include extracellular vesicles, proteins, glycans, post-translational modifications, and various metabolites. Although the concept of helminth-inspired therapies holds promise, it also presents a challenge to the drug development community, which is generally unfamiliar with foreign biologics that do not behave like antibodies. Identification and characterization of helminth molecules and vesicles and the molecular pathways they target in the host present a unique opportunity to develop tailored drugs inspired by nature that are efficacious, safe, and have minimal immunogenicity. Even so, much work remains to mine and assess this out-of-the-box therapeutic modality. Industry-based organizations need to consider long-haul investments aimed at unraveling and exploiting unique and differentiated mechanisms of action as opposed to toe-dipping entries with an eye on rapid and profitable turnarounds.
Collapse
Affiliation(s)
- Stephanie M. Ryan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Ramon M. Eichenberger
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Roland Ruscher
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Paul R. Giacomin
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
- * E-mail:
| |
Collapse
|
25
|
Liver fluke granulin promotes extracellular vesicle-mediated crosstalk and cellular microenvironment conducive to cholangiocarcinoma. Neoplasia 2020; 22:203-216. [PMID: 32244128 PMCID: PMC7118280 DOI: 10.1016/j.neo.2020.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/25/2020] [Accepted: 02/29/2020] [Indexed: 12/13/2022] Open
Abstract
Crosstalk between malignant and neighboring cells contributes to tumor growth. In East Asia, infection with the liver fluke is a major risk factor for cholangiocarcinoma (CCA). The liver fluke Opisthorchis viverrini secretes a growth factor termed liver fluke granulin, a homologue of the human progranulin, which contributes significantly to biliary tract fibrosis and morbidity. Here, extracellular vesicle (EV)-mediated transfer of mRNAs from human cholangiocytes to naïve recipient cells was investigated following exposure to liver fluke granulin. To minimize the influence of endogenous progranulin, its cognate gene was inactivated using CRISPR/Cas9-based gene knock-out. Several progranulin-depleted cell lines, termed ΔhuPGRN-H69, were established. These lines exhibited >80% reductions in levels of specific transcript and progranulin, both in gene-edited cells and within EVs released by these cells. Profiles of extracellular vesicle RNAs (evRNA) from ΔhuPGRN-H69 for CCA-associated characteristics revealed a paucity of transcripts for estrogen- and Wnt-signaling pathways, peptidase inhibitors and tyrosine phosphatase related to cellular processes including oncogenic transformation. Several CCA-specific evRNAs including MAPK/AKT pathway members were induced by exposure to liver fluke granulin. By comparison, estrogen, Wnt/PI3K and TGF signaling and other CCA pathway mRNAs were upregulated in wild type H69 cells exposed to liver fluke granulin. Of these, CCA-associated evRNAs modified the CCA microenvironment in naïve cells co-cultured with EVs from ΔhuPGRN-H69 cells exposed to liver fluke granulin, and induced translation of MAPK phosphorylation related-protein in naïve recipient cells in comparison with control recipient cells. Exosome-mediated crosstalk in response to liver fluke granulin promoted a CCA-specific program through MAPK pathway which, in turn, established a CCA-conducive disposition.
Collapse
|
26
|
Kim TK, Tirloni L, Pinto AFM, Diedrich JK, Moresco JJ, Yates JR, da Silva Vaz I, Mulenga A. Time-resolved proteomic profile of Amblyomma americanum tick saliva during feeding. PLoS Negl Trop Dis 2020; 14:e0007758. [PMID: 32049966 PMCID: PMC7041860 DOI: 10.1371/journal.pntd.0007758] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 02/25/2020] [Accepted: 01/03/2020] [Indexed: 12/26/2022] Open
Abstract
Amblyomma americanum ticks transmit more than a third of human tick-borne disease (TBD) agents in the United States. Tick saliva proteins are critical to success of ticks as vectors of TBD agents, and thus might serve as targets in tick antigen-based vaccines to prevent TBD infections. We describe a systems biology approach to identify, by LC-MS/MS, saliva proteins (tick = 1182, rabbit = 335) that A. americanum ticks likely inject into the host every 24 h during the first 8 days of feeding, and towards the end of feeding. Searching against entries in GenBank grouped tick and rabbit proteins into 27 and 25 functional categories. Aside from housekeeping-like proteins, majority of tick saliva proteins belong to the tick-specific (no homology to non-tick organisms: 32%), protease inhibitors (13%), proteases (8%), glycine-rich proteins (6%) and lipocalins (4%) categories. Global secretion dynamics analysis suggests that majority (74%) of proteins in this study are associated with regulating initial tick feeding functions and transmission of pathogens as they are secreted within 24–48 h of tick attachment. Comparative analysis of the A. americanum tick saliva proteome to five other tick saliva proteomes identified 284 conserved tick saliva proteins: we speculate that these regulate critical tick feeding functions and might serve as tick vaccine antigens. We discuss our findings in the context of understanding A. americanum tick feeding physiology as a means through which we can find effective targets for a vaccine against tick feeding. The lone star tick, Amblyomma americanum, is a medically important species in US that transmits 5 of the 16 reported tick-borne disease agents. Most recently, bites of this tick were associated with red meat allergies in humans. Vaccination of animals against tick feeding has been shown to be a sustainable and an effective alternative to current acaricide based tick control method which has several limitations. The pre-requisite to tick vaccine development is to understand the molecular basis of tick feeding physiology. Toward this goal, this study has identified proteins that A. americanum ticks inject into the host at different phases of its feeding cycle. This data set has identified proteins that A. americanum inject into the host within 24–48 h of feeding before it starts to transmit pathogens. Of high importance, we identified 284 proteins that are present in saliva of other tick species, which we suspect regulate important role(s) in tick feeding success and might represent rich source target antigens for a tick vaccine. Overall, this study provides a foundation to understand the molecular mechanisms regulating tick feeding physiology.
Collapse
Affiliation(s)
- Tae Kwon Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Lucas Tirloni
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Antônio F. M. Pinto
- Foundation Peptide Biology Lab, Salk Institute for Biological Studies, La Jolla, Californai, United States of America
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jolene K. Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
- Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - James J. Moresco
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
- Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - John R. Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Albert Mulenga
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
- * E-mail:
| |
Collapse
|
27
|
Salao K, Spofford EM, Price C, Mairiang E, Suttiprapa S, Wright HL, Sripa B, Edwards SW. Enhanced neutrophil functions during Opisthorchis viverrini infections and correlation with advanced periductal fibrosis. Int J Parasitol 2020; 50:145-152. [PMID: 32006550 DOI: 10.1016/j.ijpara.2019.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/21/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022]
Abstract
Millions of people are infected with the liver fluke, Opisthorchis viverrini (OV), but only ~25% of those infected develop liver disease and even fewer develop cholangiocarcinoma. The reasons for these differential outcomes following infection are unknown but it has been proposed that differential immune responses to the parasite may play a role. We therefore measured granulocyte (neutrophil) function in OV-infected individuals, with and without advanced periductal fibrosis, to determine if these cells have a "pro-inflammatory" phenotype that may contribute to liver disease post-infection. A case-controlled study (n = 54 in each cohort) from endemic OV-infected areas of northeastern Thailand measured neutrophil functions in whole blood from non-infected (healthy controls) and OV-infected individuals with and without APF. We measured reactive oxygen species production, phagocytosis, receptor expression and apoptosis. Secreted products from OV cultures (obtained after in vitro culture of parasites) stimulated reactive oxygen species production in non-infected healthy controls, but levels were two-fold greater after OV infection (P < 0.0001); neutrophil reactive oxygen species production in individuals with APF was double that observed in those without APF (P < 0.0001). OV-infected neutrophils had elevated CD11b expression and greater phagocytic capacity, which was even three-fold higher in those with advanced periductal fibrosis (P < 0.0001). This "activated" phenotype of circulating neutrophils was further confirmed by the observation that isolated neutrophils had delayed apoptosis ex vivo. We believe this is the first study to show that circulating blood neutrophil function is enhanced following OV infection and is more activated in those with advanced periductal fibrosis. We propose that this activated phenotype could contribute to the pathology of liver disease. These data support the hypothesis of an activated innate inflammatory phenotype following OV infection and provide the first evidence for involvement of neutrophils in disease pathology.
Collapse
Affiliation(s)
- Kanin Salao
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Edward M Spofford
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Charlotte Price
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Eimorn Mairiang
- Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sutas Suttiprapa
- Tropical Disease Research Laboratory, Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Helen L Wright
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L69 7ZB, UK
| | - Banchob Sripa
- Tropical Disease Research Laboratory, Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Steven W Edwards
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK.
| |
Collapse
|
28
|
Pak JH, Lee JY, Jeon BY, Dai F, Yoo WG, Hong SJ. Cytokine Production in Cholangiocarcinoma Cells in Response to Clonorchis sinensis Excretory-Secretory Products and Their Putative Protein Components. THE KOREAN JOURNAL OF PARASITOLOGY 2019; 57:379-387. [PMID: 31533404 PMCID: PMC6753296 DOI: 10.3347/kjp.2019.57.4.379] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/01/2019] [Indexed: 01/23/2023]
Abstract
Clonorchis sinensis is a carcinogenic human liver fluke that promotes hepatic inflammatory environments via direct contact or through their excretory-secretory products (ESPs), subsequently leading to cholangitis, periductal fibrosis, liver cirrhosis, and even cholangiocarcinoma (CCA). This study was conducted to examine the host inflammatory responses to C. sinensis ESPs and their putative protein components selected from C. sinensis expressed sequenced tag (EST) pool databases, including TGF-β receptor interacting protein 1(CsTRIP1), legumain (CsLeg), and growth factor binding protein 2 (CsGrb2). Treatment of CCA cells (HuCCT1) with the ESPs or bacterial recombinant C. sinensis proteins differentially promoted the secretion of proinflammatory cytokines (IL-1β, IL-6, and TNF-α) as well as anti-inflammatory cytokines (IL-10, TGF-β1, and TGF-β2) in a time-dependent manner. In particular, recombinant C. sinensis protein treatment resulted in increase (at maximum) of ~7-fold in TGF-β1, ~30-fold in TGF-β2, and ~3-fold in TNF-α compared with the increase produced by ESPs, indicating that CsTrip1, CsLeg, and CsGrb2 function as strong inducers for secretion of these cytokines in host cells. These results suggest that C. sinensis ESPs contribute to the immunopathological response in host cells, leading to clonorchiasis-associated hepatobiliary abnormalities of greater severity.
Collapse
Affiliation(s)
- Jhang Ho Pak
- Department of Convergence Medicine, University of Ulsan College of Medicine and Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea
| | - Ji-Yun Lee
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul 06987, Korea
| | - Bo Young Jeon
- Department of Convergence Medicine, University of Ulsan College of Medicine and Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea
| | - Fuhong Dai
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul 06987, Korea.,Department of Parasitology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, 199 Ren-ai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, P.R. China
| | - Won Gi Yoo
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul 06987, Korea
| | - Sung-Jong Hong
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul 06987, Korea
| |
Collapse
|
29
|
Chaiyadet S, Sotillo J, Krueajampa W, Thongsen S, Brindley PJ, Sripa B, Loukas A, Laha T. Vaccination of hamsters with Opisthorchis viverrini extracellular vesicles and vesicle-derived recombinant tetraspanins induces antibodies that block vesicle uptake by cholangiocytes and reduce parasite burden after challenge infection. PLoS Negl Trop Dis 2019; 13:e0007450. [PMID: 31136572 PMCID: PMC6555531 DOI: 10.1371/journal.pntd.0007450] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 06/07/2019] [Accepted: 05/09/2019] [Indexed: 12/11/2022] Open
Abstract
Background The liver fluke Opisthorchis viverrini infects several million people in Southeast Asia. Adult flukes live in the bile ducts of humans, where they cause hepatobiliary pathology, including cholangiocarcinoma. Here, we investigated the potential of extracellular vesicles (EVs) secreted by the fluke and defined recombinant proteins derived from EVs to generate protective immunity in a hamster vaccination-challenge model. Methodology/Principal findings EVs isolated from the excretory-secretory products of O. viverrini and two recombinant EV surface proteins encoding the large extracellular loops (LEL) of Ov-TSP-2 (rOv-TSP-2) and Ov-TSP-3 (rOv-TSP-3) were adjuvanted and used to vaccinate hamsters intraperitoneally followed by challenge infection with O. viverrini metacercariae. The number of adult flukes recovered from hamsters immunized with EVs, rOv-TSP-2, rOv-TSP-3 and rOv-TSP-2+rOv-TSP-3 were significantly reduced compared to control animals vaccinated with adjuvant alone. The number of eggs per gram feces was also significantly reduced in hamsters vaccinated with rOv-TSP-2 compared to controls, but no significant differences were found in the other groups. The average length of worms recovered from hamsters vaccinated with EVs, rOv-TSP-2 and rOv-TSP-3 was significantly shorter than that of worms recovered from the control group. Anti-EV IgG levels in serum and bile were significantly higher in hamsters vaccinated with EVs compared to control hamsters both pre- and post-challenge. In addition, levels of anti-rOv-TSP antibodies in the serum and bile were significantly higher than control hamsters both pre- and post-challenge. Finally, antibodies against rOv-TSP-2 and rOv-TSP-3 blocked uptake of EVs by human primary cholangiocyte in vitro, providing a plausible mechanism by which these vaccines exert partial efficacy and reduce the intensity of O. viverrini infection. Conclusion/Significance Liver fluke EVs and recombinant tetraspanins derived from the EV surface when administered to hamsters induce antibody responses that block EV uptake by target bile duct cells and exert partial efficacy and against O. viverrini challenge. Cholangiocarcinoma (CCA) is a significant public health problem in countries throughout Southeast Asia. In these areas CCA has a strong association with chronic infection with the food-borne liver fluke Opisthorchis viverrini. Current control of the infection relies on chemotherapy and health education, however these approaches are not sustainable in isolation. Hence, there is an urgent need for a vaccine against this neglected tropical disease. A vaccine against O. viverrini would confer anti-cancer protection in similar fashion to the acclaimed vaccine for human papillomavirus and cervical cancer. Toward this goal, secreted extracellular vesicles (EVs) of O. viverrini and recombinant proteins from the surface of EVs were generated and tested as vaccines in a hamster challenge model. Vaccination of hamsters with EVs and recombinant proteins induced production of antibodies in serum and bile, and those antibodies blocked uptake of EVs by primary bile duct cells in vitro. Challenge of vaccinated hamsters with infective stage flukes markedly reduced adult fluke recovery compared to the adjuvant control group. This is the first report of successful vaccination of hamsters with O. viverrini EVs and recombinant vesicle surface proteins, and provides proof-of-concept for development of subunit vaccines for this carcinogenic infection.
Collapse
Affiliation(s)
- Sujittra Chaiyadet
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Javier Sotillo
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Watchara Krueajampa
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sophita Thongsen
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Paul J. Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, United States of America
| | - Banchob Sripa
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
- * E-mail: (AL); (TL)
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- * E-mail: (AL); (TL)
| |
Collapse
|
30
|
Ershov NI, Mordvinov VA, Prokhortchouk EB, Pakharukova MY, Gunbin KV, Ustyantsev K, Genaev MA, Blinov AG, Mazur A, Boulygina E, Tsygankova S, Khrameeva E, Chekanov N, Fan G, Xiao A, Zhang H, Xu X, Yang H, Solovyev V, Lee SMY, Liu X, Afonnikov DA, Skryabin KG. New insights from Opisthorchis felineus genome: update on genomics of the epidemiologically important liver flukes. BMC Genomics 2019; 20:399. [PMID: 31117933 PMCID: PMC6530080 DOI: 10.1186/s12864-019-5752-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/29/2019] [Indexed: 01/25/2023] Open
Abstract
Background The three epidemiologically important Opisthorchiidae liver flukes Opisthorchis felineus, O. viverrini, and Clonorchis sinensis, are believed to harbour similar potencies to provoke hepatobiliary diseases in their definitive hosts, although their populations have substantially different ecogeographical aspects including habitat, preferred hosts, population structure. Lack of O. felineus genomic data is an obstacle to the development of comparative molecular biological approaches necessary to obtain new knowledge about the biology of Opisthorchiidae trematodes, to identify essential pathways linked to parasite-host interaction, to predict genes that contribute to liver fluke pathogenesis and for the effective prevention and control of the disease. Results Here we present the first draft genome assembly of O. felineus and its gene repertoire accompanied by a comparative analysis with that of O. viverrini and Clonorchis sinensis. We observed both noticeably high heterozygosity of the sequenced individual and substantial genetic diversity in a pooled sample. This indicates that potency of O. felineus population for rapid adaptive response to control and preventive measures of opisthorchiasis is higher than in O. viverrini and C. sinensis. We also have found that all three species are characterized by more intensive involvement of trans-splicing in RNA processing compared to other trematodes. Conclusion All revealed peculiarities of structural organization of genomes are of extreme importance for a proper description of genes and their products in these parasitic species. This should be taken into account both in academic and applied research of epidemiologically important liver flukes. Further comparative genomics studies of liver flukes and non-carcinogenic flatworms allow for generation of well-grounded hypotheses on the mechanisms underlying development of cholangiocarcinoma associated with opisthorchiasis and clonorchiasis as well as species-specific mechanisms of these diseases. Electronic supplementary material The online version of this article (10.1186/s12864-019-5752-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nikita I Ershov
- Institute of Cytology and Genetics SB RAS, 10 Lavrentiev Ave, Novosibirsk, 630090, Russia.
| | | | - Egor B Prokhortchouk
- Russian Federal Research Center for Biotechnology, 33/2 Leninsky prospect, Moscow, 119071, Russia. .,ZAO Genoanalytica, 1 Leninskie Gory street, Moscow, 119234, Russia.
| | - Mariya Y Pakharukova
- Institute of Cytology and Genetics SB RAS, 10 Lavrentiev Ave, Novosibirsk, 630090, Russia.,Novosibirsk State University, 2 Pirogova Str, Novosibirsk, 630090, Russia
| | - Konstantin V Gunbin
- Institute of Cytology and Genetics SB RAS, 10 Lavrentiev Ave, Novosibirsk, 630090, Russia
| | - Kirill Ustyantsev
- Institute of Cytology and Genetics SB RAS, 10 Lavrentiev Ave, Novosibirsk, 630090, Russia
| | - Mikhail A Genaev
- Institute of Cytology and Genetics SB RAS, 10 Lavrentiev Ave, Novosibirsk, 630090, Russia
| | - Alexander G Blinov
- Institute of Cytology and Genetics SB RAS, 10 Lavrentiev Ave, Novosibirsk, 630090, Russia
| | - Alexander Mazur
- Russian Federal Research Center for Biotechnology, 33/2 Leninsky prospect, Moscow, 119071, Russia
| | | | | | | | - Nikolay Chekanov
- Russian Federal Research Center for Biotechnology, 33/2 Leninsky prospect, Moscow, 119071, Russia
| | - Guangyi Fan
- BGI-Shenzhen, 11 Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China.,State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - An Xiao
- BGI-Shenzhen, 11 Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - He Zhang
- BGI-Shenzhen, 11 Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Xun Xu
- BGI-Shenzhen, 11 Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Huanming Yang
- BGI-Shenzhen, 11 Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Victor Solovyev
- Softberry Inc., 116 Radio Circle, Suite 400, Mount Kisco, NY, 10549, USA
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xin Liu
- BGI-Shenzhen, 11 Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Dmitry A Afonnikov
- Institute of Cytology and Genetics SB RAS, 10 Lavrentiev Ave, Novosibirsk, 630090, Russia.,Novosibirsk State University, 2 Pirogova Str, Novosibirsk, 630090, Russia
| | - Konstantin G Skryabin
- Russian Federal Research Center for Biotechnology, 33/2 Leninsky prospect, Moscow, 119071, Russia.,Federal Research Center Kurchatov Institute, Moscow, Russia
| |
Collapse
|
31
|
Wang C, Wang B, Wang B, Wang Q, Liu G, Fan C, Zhang L. A novel granulin homologue isolated from the jellyfish Cyanea capillata promotes proliferation and migration of human umbilical vein endothelial cells through the ERK1/2-signaling pathway. Int J Biol Macromol 2019; 135:212-225. [PMID: 31108149 DOI: 10.1016/j.ijbiomac.2019.05.101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 11/24/2022]
Abstract
Jellyfish grow rapidly and have a strong regenerative ability, indicating that they may express high levels of growth factors. Therefore, the aim of this research was to isolate the growth-promoting components from the jellyfish Cyanea capillata (C. capillata) and to further explore the underlying mechanisms. In this study, we first isolated and identified a novel polypeptide from C. capillata tentacles using size-exclusion chromatography followed by reverse-phase HPLC. This peptide, consisting of 58 amino acids (MW 5782.9 Da), belonged to the granulin (GRN) family of growth factors; thus, we named it Cyanea capillata granulin-1 (CcGRN-1). Second, using CCK-8 assay and flow cytometry, we verified that CcGRN-1 at the 0.5 μg/ml concentration could promote cell proliferation and increase the expression of cell-cycle proteins (CyclinB1 and CyclinD1). Third, signaling pathways studies showed that CcGRN-1 could activate the PI3K/Akt- and ERK1/2 MAPK-signaling pathways but not the JNK MAPK- or NF-κB-signaling pathways. Subsequently, we further confirmed that the CcGRN-1-induced cell proliferation and migration were associated only with the ERK1/2 MAPK-signaling pathway. Considering all of these factors, CcGRN-1, as the first jellyfish-derived GRN homologue, possesses growth-promoting properties and may be a candidate for novel therapeutics to promote human wound healing in unfavorable conditions.
Collapse
Affiliation(s)
- Chao Wang
- Department of Marine Biotechnology, Faculty of Naval Medicine, Navy Medical University, Xiangyin Road No.800, Shanghai 200433, China
| | - Beilei Wang
- Department of Marine Biotechnology, Faculty of Naval Medicine, Navy Medical University, Xiangyin Road No.800, Shanghai 200433, China
| | - Bo Wang
- Department of Marine Biotechnology, Faculty of Naval Medicine, Navy Medical University, Xiangyin Road No.800, Shanghai 200433, China
| | - Qianqian Wang
- Department of Marine Biotechnology, Faculty of Naval Medicine, Navy Medical University, Xiangyin Road No.800, Shanghai 200433, China
| | - Guoyan Liu
- Department of Marine Biotechnology, Faculty of Naval Medicine, Navy Medical University, Xiangyin Road No.800, Shanghai 200433, China
| | - Chongxu Fan
- Beijing Institute of Pharmaceutical Chemistry, Wennan Road No.59, Beijing 102205, China.
| | - Liming Zhang
- Department of Marine Biotechnology, Faculty of Naval Medicine, Navy Medical University, Xiangyin Road No.800, Shanghai 200433, China.
| |
Collapse
|
32
|
Pakharukova MY, Zaparina OG, Kapushchak YK, Baginskaya NV, Mordvinov VA. Opisthorchis felineus infection provokes time-dependent accumulation of oxidative hepatobiliary lesions in the injured hamster liver. PLoS One 2019; 14:e0216757. [PMID: 31086416 PMCID: PMC6516637 DOI: 10.1371/journal.pone.0216757] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/27/2019] [Indexed: 12/15/2022] Open
Abstract
Opisthorchiasis caused by food-borne trematode Opisthorchis felineus is a substantial public health problem, with 17 million persons infected worldwide. This chronic disease is associated with hepatobiliary inflammation, cholangiocyte dysplasia, cholangiofibrosis, intraepithelial neoplasia, and even cholangiocarcinoma among chronically infected individuals. To provide first insights into the mechanism by which O. felineus infection causes precancerous liver lesions, we investigated the level of oxidative stress (lipid peroxidation byproducts and 8-hydroxy-2′-deoxyguanosine) as well as the time course profiles of chronic inflammation and fibrogenesis markers in the dynamics of opisthorchiasis from 1 month to 1.5 years postinfection in an experimental model based on golden hamsters Mesocricetus auratus. For the first time, we showed that O. felineus infection provokes time-dependent accumulation of oxidative hepatobiliary lesions in the injured liver of hamsters. In particular, over the course of infection, lipid peroxidation byproducts 4-hydroxynonenal and malondialdehyde were upregulated; these changes in general correlate with the dynamics of hepatic histopathological changes. We detected macrophages with various immunophenotypes and elevated levels of CD68, COX2, and CD163 in the O. felineus–infected animals. Meanwhile, there was direct time-dependent elevation of TNF-α (R = 0.79; p < 0.001) and CD163 protein levels (R = 0.58; p = 0.022). We also provide quantitative data about epithelial hyperplasia marker CK7 and a marker of myofibroblast activation (α smooth muscle actin). Our present data provide first insights into the histopathological mechanism by which O. felineus infection causes liver injuries. These findings support the inclusion of O. felineus in Group 1 of biological carcinogens.
Collapse
Affiliation(s)
- Mariya Y. Pakharukova
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
- * E-mail:
| | - Oxana G. Zaparina
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Yaroslav K. Kapushchak
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State Medical University, Novosibirsk, Russian Federation, Novosibirsk, Russia
| | - Nina V. Baginskaya
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Viatcheslav A. Mordvinov
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
33
|
Shabelnikov SV, Bobkov DE, Sharlaimova NS, Petukhova OA. Injury affects coelomic fluid proteome of the common starfish, Asterias rubens. ACTA ACUST UNITED AC 2019; 222:jeb.198556. [PMID: 30877231 DOI: 10.1242/jeb.198556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/11/2019] [Indexed: 01/04/2023]
Abstract
Echinoderms, possessing outstanding regenerative capabilities, provide a unique model system for the study of response to injury. However, little is known about the proteomic composition of coelomic fluid, an important biofluid circulating throughout the animal's body and reflecting the overall biological status of the organism. In this study, we used LC-MALDI tandem mass spectrometry to characterize the proteome of the cell-free coelomic fluid of the starfish Asterias rubens and to follow the changes occurring in response to puncture wound and blood loss. In total, 91 proteins were identified, of which 61 were extracellular soluble and 16 were bound to the plasma membrane. The most represented functional terms were 'pattern recognition receptor activity' and 'peptidase inhibitor activity'. A series of candidate proteins involved in early response to injury was revealed. Ependymin, β-microseminoprotein, serum amyloid A and avidin-like proteins, which are known to be involved in intestinal regeneration in the sea cucumber, were also identified as injury-responsive proteins. Our results expand the list of proteins potentially involved in defense and regeneration in echinoderms and demonstrate dramatic effects of injury on the coelomic fluid proteome.
Collapse
Affiliation(s)
- Sergey V Shabelnikov
- Laboratory of Regulation of Gene Expression, Institute of Cytology, Russian Academy of Sciences, 194064 St Petersburg, Russia
| | - Danila E Bobkov
- Department of Cell Cultures, Institute of Cytology, Russian Academy of Sciences, 194064 St Petersburg, Russia
| | - Natalia S Sharlaimova
- Department of Cell Cultures, Institute of Cytology, Russian Academy of Sciences, 194064 St Petersburg, Russia
| | - Olga A Petukhova
- Department of Cell Cultures, Institute of Cytology, Russian Academy of Sciences, 194064 St Petersburg, Russia
| |
Collapse
|
34
|
Arunsan P, Ittiprasert W, Smout MJ, Cochran CJ, Mann VH, Chaiyadet S, Karinshak SE, Sripa B, Young ND, Sotillo J, Loukas A, Brindley PJ, Laha T. Programmed knockout mutation of liver fluke granulin attenuates virulence of infection-induced hepatobiliary morbidity. eLife 2019; 8:e41463. [PMID: 30644359 PMCID: PMC6355195 DOI: 10.7554/elife.41463] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/12/2018] [Indexed: 12/26/2022] Open
Abstract
Infection with the food-borne liver fluke Opisthorchis viverrini is the principal risk factor (IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, 2012) for cholangiocarcinoma (CCA) in the Lower Mekong River Basin countries including Thailand, Lao PDR, Vietnam and Cambodia. We exploited this link to explore the role of the secreted growth factor termed liver fluke granulin (Ov-GRN-1) in pre-malignant lesions by undertaking programmed CRISPR/Cas9 knockout of the Ov-GRN-1 gene from the liver fluke genome. Deep sequencing of amplicon libraries from genomic DNA of gene-edited parasites revealed Cas9-catalyzed mutations within Ov-GRN-1. Gene editing resulted in rapid depletion of Ov-GRN-1 transcripts and the encoded Ov-GRN-1 protein. Gene-edited parasites colonized the biliary tract of hamsters and developed into adult flukes, but the infection resulted in reduced pathology as evidenced by attenuated biliary hyperplasia and fibrosis. Not only does this report pioneer programmed gene-editing in parasitic flatworms, but also the striking, clinically-relevant pathophysiological phenotype confirms the role for Ov-GRN-1 in virulence morbidity during opisthorchiasis.
Collapse
Affiliation(s)
- Patpicha Arunsan
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC, United States
- Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington DC, United States
| | - Wannaporn Ittiprasert
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC, United States
- Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington DC, United States
| | - Michael J Smout
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Christina J Cochran
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC, United States
- Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington DC, United States
| | - Victoria H Mann
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC, United States
- Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington DC, United States
| | - Sujittra Chaiyadet
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Shannon E Karinshak
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC, United States
- Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington DC, United States
| | - Banchob Sripa
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Neil David Young
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Victoria, Australia
| | - Javier Sotillo
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC, United States
- Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington DC, United States
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
35
|
Sotillo J, Pearson MS, Loukas A. Trematode Genomics and Proteomics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1154:411-436. [PMID: 31297769 DOI: 10.1007/978-3-030-18616-6_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Trematode infections are among the most neglected tropical diseases despite their worldwide distribution and extraordinary ability to parasitise many different host species and host tissues. Furthermore, these parasites are of great socioeconomic, medical, veterinary and agricultural importance. During the last 10 years, there have been increasing efforts to overcome the lack of information on different "omic" resources such as proteomics and genomics. Herein, we focus on the recent advances in genomics and proteomics from trematodes of human importance, including liver, blood, intestinal and lung flukes. We also provide information on the latest technologies applied to study the biology of trematodes as well as on the resources available for the study of the molecular aspects of this group of helminths.
Collapse
Affiliation(s)
- Javier Sotillo
- Centre for Molecular Therapeutics, Australian Institute for Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.
| | - Mark S Pearson
- Centre for Molecular Therapeutics, Australian Institute for Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute for Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
36
|
Maizels RM, Smits HH, McSorley HJ. Modulation of Host Immunity by Helminths: The Expanding Repertoire of Parasite Effector Molecules. Immunity 2018; 49:801-818. [PMID: 30462997 PMCID: PMC6269126 DOI: 10.1016/j.immuni.2018.10.016] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/13/2018] [Accepted: 10/30/2018] [Indexed: 02/09/2023]
Abstract
Helminths are extraordinarily successful parasites due to their ability to modulate the host immune response. They have evolved a spectrum of immunomodulatory molecules that are now beginning to be defined, heralding a molecular revolution in parasite immunology. These discoveries have the potential both to transform our understanding of parasite adaptation to the host and to develop possible therapies for immune-mediated disease. In this review we will summarize the current state of the art in parasite immunomodulation and discuss perspectives on future areas for research and discovery.
Collapse
Affiliation(s)
- Rick M Maizels
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | | | - Henry J McSorley
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
37
|
Laha T, Brindley PJ, Suyapoh W, Suttiprapa S. RNA Interference as an Approach to Functional Genomics Genetic Manipulation of Opisthorchis viverrini. ADVANCES IN PARASITOLOGY 2018; 102:25-43. [PMID: 30442309 DOI: 10.1016/bs.apar.2018.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The availability of genome and transcriptome data of the liver fluke Opisthorchis viverrini provides the foundation for exploration of gene function and its effect on host-parasite interactions and pathogenesis of O. viverrini-associated bile duct cancer. Functional genomics approaches address the function of DNA at levels of the gene, RNA transcript and protein product using informative manipulations of the genome, epigenome, transcriptome, proteome, microbiome and metabolome. Advances in functional genomics for O. viverrini have thus far focused on RNA interference. The flukes have been transfected with double-stranded RNAs aiming to silence target gene expression. In general, this approach for functional genomics investigation of this pathogen has been found to be tractable and efficient: suppression of messenger RNA expression in O. viverrini results in reduction of protein activity and phenotypic changes. Future perspectives for functional genomics of this liver fluke and close phylogenetic relatives are also discussed.
Collapse
Affiliation(s)
- Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Tropical Diseases of Poverty, School of Medicine & Health Sciences, The George Washington University, Washington DC, United States
| | - Watcharapol Suyapoh
- Biomedical Science Graduate Program, Graduate School, Khon Kaen University, Khon Kaen, Thailand
| | - Sutas Suttiprapa
- Tropical Disease Research Center, Tropical Medicine Graduate Program, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
38
|
Upontain S, Sereerak P, Laha T, Sripa B, Tangkawatana P, Brindley PJ, Tangkawatana S. Granulin Expression in Hamsters during Opisthorchis viverrini Infection-Induced Cholangiocarcinogenesis. Asian Pac J Cancer Prev 2018; 19:2437-2445. [PMID: 30255697 PMCID: PMC6249462 DOI: 10.22034/apjcp.2018.19.9.2437] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 07/30/2018] [Indexed: 02/07/2023] Open
Abstract
The secreted growth factor granulin (GRN) is upregulated during diverse epithelial cancers. GRN stimulates cell growth and development while inhibiting apoptosis. Orthologues of vertebrate granulins evolved in other animals including the liver fluke Opisthorchis viverrini. Curiously, liver fluke granulin, termed Ov-GRN-1 promotes cholangiocarcinogenesis during chronic opisthorchiasis but, by contrast, limited information is available concerning mammalian GRN during liver fluke infection-induced cholangiocarcinoma (CCA). Here we investigated the expression of mammalian granulin in the O. viverrini-associated a hamster model of opisthorchiasis and liver fluke infection-induced CCA. Male Syrian golden hamsters were assigned to one of four treatment groups, each group included 30 hamsters: 1) normal (control), 2) infected with O. viverrini (OV); 3) exposed to N-dimethylnitrosamine in drinking water (DMN); and 4) infected with O. viverrini and exposed to DMN (OVDMN). Immunohistochemistry using an anti-granulin specific probe for mammalian granulin was undertaken to monitor expression and location in hepatobiliary tissues of the hamsters. In parallel, cognate studies of transcription of mRNA and protein. Histopathological examination revealed development of proliferative lesions from the onset and eruption of CCA onwards, an outcome that was most prominent in the OVDMN hamsters. Proliferating cell nuclear antigen (PCNA) index rose continuously from initiation of infection and increased with lesion progression in OV, DMN and markedly in OVDMN hamsters. Expression of GRN in biliary was elevated in biliary epithelial cells in CCA lesions in hamsters in the DMN and OVDMN groups. Expression of GRN as assayed by western blot and RT-PCR reflected the same trend as seen with PCNA. Together the histopathogical and molecular assay based findings revealed marked expression of granulin during cholangiocarcinoma in these hamsters, and highlighted the prospect that granulin represents a potential prognostic marker for cholangiocarcinoma.
Collapse
Affiliation(s)
- Songkiad Upontain
- Graduate School, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
- WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease)/Tropical Disease Research Center (TDRC), Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| | | | | | | | | | | | | |
Collapse
|
39
|
Dastpeyman M, Bansal PS, Wilson D, Sotillo J, Brindley PJ, Loukas A, Smout MJ, Daly NL. Structural Variants of a Liver Fluke Derived Granulin Peptide Potently Stimulate Wound Healing. J Med Chem 2018; 61:8746-8753. [PMID: 30183294 DOI: 10.1021/acs.jmedchem.8b00898] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Granulins are a family of growth factors involved in cell proliferation. The liver-fluke granulin, Ov-GRN-1, isolated from a carcinogenic liver fluke Opisthorchis viverrini, can significantly accelerate wound repair in vivo and in vitro. However, it is difficult to express Ov-GRN-1 in recombinant form at high yield, impeding its utility as a drug lead. Previously we reported that a truncated analogue ( Ov-GRN12-35_3s) promotes healing of cutaneous wounds in mice. NMR analysis of this analogue indicates the presence of multiple conformations, most likely as a result of proline cis/ trans isomerization. To further investigate whether the proline residues are involved in adopting the multiple confirmations, we have synthesized analogues involving mutation of the proline residues. We have shown that the proline residues have a significant influence on the structure, activity, and folding of Ov-GRN12-35_3s. These results provide insight into improving the oxidative folding yield and bioactivity of Ov-GRN12-35_3s and might facilitate the development of a novel wound healing agent.
Collapse
Affiliation(s)
- Mohadeseh Dastpeyman
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , QLD 4870 , Australia
| | - Paramjit S Bansal
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , QLD 4870 , Australia
| | - David Wilson
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , QLD 4870 , Australia
| | - Javier Sotillo
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , QLD 4870 , Australia
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine and Health Sciences , George Washington University , Washington, D.C. 20052 , United States
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , QLD 4870 , Australia
| | - Michael J Smout
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , QLD 4870 , Australia
| | - Norelle L Daly
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , QLD 4870 , Australia
| |
Collapse
|
40
|
McSorley HJ, Chayé MAM, Smits HH. Worms: Pernicious parasites or allies against allergies? Parasite Immunol 2018; 41:e12574. [PMID: 30043455 PMCID: PMC6585781 DOI: 10.1111/pim.12574] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/13/2018] [Accepted: 07/18/2018] [Indexed: 12/14/2022]
Abstract
Type 2 immune responses are most commonly associated with allergy and helminth parasite infections. Since the discovery of Th1 and Th2 immune responses more than 30 years ago, models of both allergic disease and helminth infections have been useful in characterizing the development, effector mechanisms and pathological consequences of type 2 immune responses. The observation that some helminth infections negatively correlate with allergic and inflammatory disease led to a large field of research into parasite immunomodulation. However, it is worth noting that helminth parasites are not always benign infections, and that helminth immunomodulation can have stimulatory as well as suppressive effects on allergic responses. In this review, we will discuss how parasitic infections change host responses, the consequences for bystander immunity and how this interaction influences clinical symptoms of allergy.
Collapse
Affiliation(s)
- Henry J McSorley
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mathilde A M Chayé
- Department of Parasitology, Leiden Immunology of Parasitic Infections Group, Leiden University Medical Centre, ZA Leiden, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden Immunology of Parasitic Infections Group, Leiden University Medical Centre, ZA Leiden, The Netherlands
| |
Collapse
|
41
|
Wang P, Chitramuthu B, Bateman A, Bennett HPJ, Xu P, Ni F. Structure dissection of zebrafish progranulins identifies a well-folded granulin/epithelin module protein with pro-cell survival activities. Protein Sci 2018; 27:1476-1490. [PMID: 29732682 DOI: 10.1002/pro.3441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/02/2018] [Accepted: 05/02/2018] [Indexed: 01/07/2023]
Abstract
The ancient and pluripotent progranulins contain multiple repeats of a cysteine-rich sequence motif of ∼60 amino acids, called the granulin/epithelin module (GEM) with a prototypic structure of four β-hairpins zipped together by six inter-hairpin disulfide bonds. Prevalence of this disulfide-enforced structure is assessed here by an expression screening of 19 unique GEM sequences of the four progranulins in the zebrafish genome, progranulins 1, 2, A and B. While a majority of the expressed GEM peptides did not exhibit uniquely folded conformations, module AaE from progranulin A and AbB from progranulin B were found to fold into the protopypic 4-hairpin structure along with disulfide formation. Module AaE has the most-rigid three-dimensional structure with all four β-hairpins defined using high-resolution (H-15 N) NMR spectroscopy, including 492 inter-proton nuclear Overhauser effects, 23 3 J(HN,Hα ) coupling constants, 22 hydrogen bonds as well as 45 residual dipolar coupling constants. Three-dimensional structure of AaE and the partially folded AbB re-iterate the conformational stability of the N-terminal stack of two beta-hairpins and varying degrees of structural flexibility for the C-terminal half of the 4-hairpin global fold of the GEM repeat. A cell-based assay demonstrated a functional activity for the zebrafish granulin AaE in promoting the survival of neuronal cells, similarly to what has been found for the corresponding granulin E module in human progranulin. Finally, this work highlights the remaining challenges in structure-activity studies of proteins containing the GEM repeats, due to the apparent prevalence of structural disorder in GEM motifs despite potentially a high density of intramolecular disulfide bonds.
Collapse
Affiliation(s)
- Ping Wang
- Department of Biochemistry, McIntyre Medical Sciences Building, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, H3G 1Y6, Canada.,Protein NMR Laboratory, Advanced Analytics Section, Human Health Therapeutics Research Center, National Research Council Canada, 6100 Royalmount Avenue, Montreal, Quebec, H4P 2R2, Canada
| | - Babykumari Chitramuthu
- Division of Experimental Medicine, Research Institute of the McGill University Health Center, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada.,Center for Translational Biology, The Research Institute of McGill University Health Center, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada
| | - Andrew Bateman
- Division of Experimental Medicine, Research Institute of the McGill University Health Center, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada.,Center for Translational Biology, The Research Institute of McGill University Health Center, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada
| | - Hugh P J Bennett
- Division of Experimental Medicine, Research Institute of the McGill University Health Center, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada.,Center for Translational Biology, The Research Institute of McGill University Health Center, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada
| | - Ping Xu
- Protein NMR Laboratory, Advanced Analytics Section, Human Health Therapeutics Research Center, National Research Council Canada, 6100 Royalmount Avenue, Montreal, Quebec, H4P 2R2, Canada
| | - Feng Ni
- Department of Biochemistry, McIntyre Medical Sciences Building, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, H3G 1Y6, Canada.,Protein NMR Laboratory, Advanced Analytics Section, Human Health Therapeutics Research Center, National Research Council Canada, 6100 Royalmount Avenue, Montreal, Quebec, H4P 2R2, Canada.,Division of Experimental Medicine, Research Institute of the McGill University Health Center, 1001 Decarie Boulevard, Montreal, Quebec, H4A 3J1, Canada
| |
Collapse
|
42
|
Chaidee A, Onsurathum S, Intuyod K, Pannangpetch P, Pongchaiyakul C, Pinlaor P, Pairojkul C, Ittiprasert W, Cochran CJ, Mann VH, Brindley PJ, Pinlaor S. Co-occurrence of opisthorchiasis and diabetes exacerbates morbidity of the hepatobiliary tract disease. PLoS Negl Trop Dis 2018; 12:e0006611. [PMID: 29953446 PMCID: PMC6040770 DOI: 10.1371/journal.pntd.0006611] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/11/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022] Open
Abstract
Complications arising from infection with the carcinogenic liver fluke Opisthorchis viverrini cause substantial morbidity and mortality in Thailand and adjacent lower Mekong countries. In parallel, the incidence rate of diabetes mellitus (DM) is increasing in this same region, and indeed worldwide. Many residents in opisthorchiasis-endemic regions also exhibit DM, but the hepatobiliary disease arising during the co-occurrence of these two conditions remains to be characterized. Here, the histopathological profile during co-occurrence of opisthorchiasis and DM was investigated in a rodent model of human opisthorchiasis in which diabetes was induced with streptozotocin. The effects of excretory/secretory products from the liver fluke, O. viverrini (OVES) on hepatocyte and cholangiocyte responses during hyperglycemic conditions also were monitored. Both the liver fluke-infected hamsters (OV group) and hamsters with DM lost weight compared to control hamsters. Weight loss was even more marked in the hamsters with both opisthorchiasis and DM (OD group). Hypertrophy of hepatocytes, altered biliary canaliculi, and biliary hyperplasia were more prominent in the OD group, compared with OV and DM groups. Profound oxidative DNA damage, evidenced by 8-oxo-2'-deoxyguanosine, proliferating cell nuclear antigen, and periductal fibrosis characterized the OD compared to OV and DM hamsters. Upregulation of expression of cytokines in response to infection and impairment of the pathway for insulin receptor substrate (IRS)/phosphatidylinositol-3-kinases (PI3K)/protein kinase B (AKT) signaling attended these changes. In vitro, OVES and glucose provoked time- and dose-dependent effects on the proliferation of both hepatocytes and cholangiocytes. In overview, the co-occurrence of opisthorchiasis and diabetes exacerbated pathophysiological damage to the hepatobiliary tract. We speculate that opisthorchiasis and diabetes together aggravate hepatobiliary pathogenesis through an IRS/PI3K/AKT-independent pathway.
Collapse
Affiliation(s)
- Apisit Chaidee
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Sudarat Onsurathum
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Kitti Intuyod
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | | | | | - Porntip Pinlaor
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Chawalit Pairojkul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Wannaporn Ittiprasert
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, The George Washington University, Washington, D.C., United States of America
| | - Christina J. Cochran
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, The George Washington University, Washington, D.C., United States of America
| | - Victoria H. Mann
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, The George Washington University, Washington, D.C., United States of America
| | - Paul J. Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, The George Washington University, Washington, D.C., United States of America
| | - Somchai Pinlaor
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
43
|
Roeksomtawin S, Navasumrit P, Waraprasit S, Parnlob V, Sricharunrat T, Bhudhisawasdi V, Savaraj N, Ruchirawat M. Decreased argininosuccinate synthetase expression in Thai patients with cholangiocarcinoma and the effects of ADI-PEG20 treatment in CCA cell lines. Oncol Lett 2018; 16:1529-1538. [PMID: 30008833 PMCID: PMC6036342 DOI: 10.3892/ol.2018.8807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a severe cancer with poor prognosis. The aim of the present study was to explore the expression of argininosuccinate synthetase (ASS), as well as the possibility of using pegylated arginine deiminase (ADI-PEG20) for the treatment of CCA. ASS expression was determined in CCA specimens from 40 patients in Thailand. Immunohistochemical detection of ASS and determination of the proliferative index, Ki-67, were carried out in paraffin-embedded sections of these specimens, as well as in two CCA cell lines, HuCCA and RmCCA-1, derived from CCA samples from patients in Thailand. In total, ~45% of the CCA specimens had low ASS expression, and the level of expression was significantly negatively associated with cell differentiation (P<0.05) and Ki-67 expression (P<0.05). The level of ASS expression in tumor cells was significantly lower than that in non-tumor cells (1.3-fold, P<0.05). The HuCCA cell line had significantly lower levels (P<0.05) of ASS expression at the mRNA and protein levels relative to those of normal human immortalized fibroblast cells (BJ-1). By contrast, the RmCCA-1 cell line showed no significant difference. In addition, the effects of ADI-PEG20 on growth inhibition, apoptosis and cell cycle arrest were determined in HuCCA and RmCCA-1 cells. ADI-PEG20 treatment reduced cell viability and cell proliferation in the two CCA cell lines, though it had no effect in immortalized BJ-1 cells. Furthermore, ADI-PEG20 treatment significantly increased G0/G1 cell cycle arrest in HuCCA, though not in RmCCA-1 cells. ASS silencing in the RmCCA-1 cell line significantly enhanced its sensitivity to ADI-PEG20 treatment. Results from the in vitro study demonstrated that ADI-PEG20 has antitumor activity against CCA with low ASS expression.
Collapse
Affiliation(s)
- Somphon Roeksomtawin
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand.,Chulabhorn Graduate Institute, Bangkok 10210, Thailand
| | - Panida Navasumrit
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand.,Chulabhorn Graduate Institute, Bangkok 10210, Thailand.,Center of Excellence on Environmental Health and Toxicology, CHE, Ministry of Education, Bangkok 10300, Thailand
| | - Somchamai Waraprasit
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Varabhorn Parnlob
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | | | - Vajarabhongsa Bhudhisawasdi
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kean 40000, Thailand.,Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Niramol Savaraj
- Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33125, USA
| | - Mathuros Ruchirawat
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand.,Chulabhorn Graduate Institute, Bangkok 10210, Thailand.,Center of Excellence on Environmental Health and Toxicology, CHE, Ministry of Education, Bangkok 10300, Thailand
| |
Collapse
|
44
|
Dastpeyman M, Smout MJ, Wilson D, Loukas A, Daly NL. Folding of granulin domains. Pept Sci (Hoboken) 2018. [DOI: 10.1002/pep2.24062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Mohadeseh Dastpeyman
- Centre for Biodiscovery and Molecular Development of Therapeutics; Australian Institute of Tropical Health and Medicine, James Cook University; Cairns Queensland Australia
| | - Michael J. Smout
- Centre for Biodiscovery and Molecular Development of Therapeutics; Australian Institute of Tropical Health and Medicine, James Cook University; Cairns Queensland Australia
| | - David Wilson
- Centre for Biodiscovery and Molecular Development of Therapeutics; Australian Institute of Tropical Health and Medicine, James Cook University; Cairns Queensland Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics; Australian Institute of Tropical Health and Medicine, James Cook University; Cairns Queensland Australia
| | - Norelle L. Daly
- Centre for Biodiscovery and Molecular Development of Therapeutics; Australian Institute of Tropical Health and Medicine, James Cook University; Cairns Queensland Australia
| |
Collapse
|
45
|
Haugen B, Karinshak SE, Mann VH, Popratiloff A, Loukas A, Brindley PJ, Smout MJ. Granulin Secreted by the Food-Borne Liver Fluke Opisthorchis viverrini Promotes Angiogenesis in Human Endothelial Cells. Front Med (Lausanne) 2018; 5:30. [PMID: 29503819 PMCID: PMC5820972 DOI: 10.3389/fmed.2018.00030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/29/2018] [Indexed: 01/05/2023] Open
Abstract
The liver fluke Opisthorchis viverrini is a food-borne, zoonotic pathogen endemic to Thailand and adjacent countries in Southeast Asia. The adult developmental stage of the O. viverrini parasite excretes and secretes numerous proteins within the biliary tract including the gall bladder. Lesions caused by the feeding activities of the liver fluke represent wounds that undergo protracted cycles of healing and re-injury during chronic infection, which can last for decades. Components of the excretory/secretory (ES) complement released by the worms capably drive proliferation of bile duct epithelial cells and are implicated in establishing the oncogenic milieu that leads to bile duct cancer, cholangiocarcinoma. An ES protein, the secreted granulin-like growth factor termed Ov-GRN-1, accelerates wound resolution in mice and in vitro. To investigate angiogenesis (blood vessel development) that may contribute to wound healing promoted by liver fluke granulin and, by implication, to carcinogenesis during chronic opisthorchiasis, we employed an in vitro tubule formation assay (TFA) where human umbilical vein endothelial cells were grown on gelled basement matrix. Ten and 40 nM Ov-GRN-1 significantly stimulated angiogenesis as monitored by cellular proliferation and by TFA in real time. This demonstration of potent angiogenic property of Ov-GRN-1 bolsters earlier reports on the therapeutic potential for chronic non-healing wounds of diabetics, tobacco users, and the elderly and, in addition, showcases another of the hallmark of cancer characteristic of this carcinogenic liver fluke.
Collapse
Affiliation(s)
- Brandon Haugen
- Department of Microbiology, Immunology and Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, United States.,Department of Biology, University of the District of Columbia, Washington, DC, United States
| | - Shannon E Karinshak
- Department of Microbiology, Immunology and Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, United States
| | - Victoria H Mann
- Department of Microbiology, Immunology and Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, United States
| | - Anastas Popratiloff
- Nanofabrication and Imaging Center, Office of VP for Research, George Washington University, Washington, DC, United States
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, United States
| | - Michael J Smout
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
46
|
Suttiprapa S, Sotillo J, Smout M, Suyapoh W, Chaiyadet S, Tripathi T, Laha T, Loukas A. Opisthorchis viverrini Proteome and Host-Parasite Interactions. ADVANCES IN PARASITOLOGY 2018; 102:45-72. [PMID: 30442310 DOI: 10.1016/bs.apar.2018.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The omics technologies have improved our understanding of the molecular events that underpin host-parasite interactions and the pathogenesis of parasitic diseases. In the last decade, proteomics and genomics in particular have been used to characterize the surface and secreted products of the carcinogenic liver fluke Opisthorchis viverrini and revealed important roles for proteins at the host-parasite interface to ensure that the flukes can migrate, feed and reproduce in a hostile environment. This review summarizes the advances made in this area, primarily focusing on discoveries enabled by the publication of the fluke secreted proteomes over the last decade. Protein families that will be covered include proteases, antioxidants, oncogenic proteins and the secretion of exosome-like extracellular vesicles. Roles of these proteins in host-parasite interactions and pathogenesis of fluke-induced hepatobiliary diseases, including cholangiocarcinogenesis, are discussed. Future directions for the application of this knowledge to control infection and disease will also be discussed.
Collapse
|
47
|
Sripa B, Tangkawattana S, Brindley PJ. Update on Pathogenesis of Opisthorchiasis and Cholangiocarcinoma. ADVANCES IN PARASITOLOGY 2018; 102:97-113. [PMID: 30442312 DOI: 10.1016/bs.apar.2018.10.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Infection with the food-borne liver fluke Opisthorchis viverrini causes cholangiocarcinoma (CCA). Whereas the cause of CCA in the West remains obscure, the principal risk factor in Thailand is opisthorchiasis. Here, we review recent findings on the pathogenesis of opisthorchiasis and CCA focusing on helminth molecules/toxic metabolites, host-parasite interaction, endocytosis, immunopathology/inflammatory responses, free radical production, molecular genetic alterations, and multifactorial including coinfections driving to CCA development.
Collapse
|
48
|
Sundaraneedi MK, Tedla BA, Eichenberger RM, Becker L, Pickering D, Smout MJ, Rajan S, Wangchuk P, Keene FR, Loukas A, Collins JG, Pearson MS. Polypyridylruthenium(II) complexes exert anti-schistosome activity and inhibit parasite acetylcholinesterases. PLoS Negl Trop Dis 2017; 11:e0006134. [PMID: 29240773 PMCID: PMC5746282 DOI: 10.1371/journal.pntd.0006134] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/28/2017] [Accepted: 11/23/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Schistosomiasis affects over 200 million people and there are concerns whether the current chemotherapeutic control strategy (periodic mass drug administration with praziquantel (PZQ)-the only licenced anti-schistosome compound) is sustainable, necessitating the development of new drugs. METHODOLOGY/PRINCIPAL FINDINGS We investigated the anti-schistosome efficacy of polypyridylruthenium(II) complexes and showed they were active against all intra-mammalian stages of S. mansoni. Two compounds, Rubb12-tri and Rubb7-tnl, which were among the most potent in their ability to kill schistosomula and adult worms and inhibit egg hatching in vitro, were assessed for their efficacy in a mouse model of schistosomiasis using 5 consecutive daily i.v. doses of 2 mg/kg (Rubb12-tri) and 10 mg/kg (Rubb7-tnl). Mice treated with Rubb12-tri showed an average 42% reduction (P = 0.009), over two independent trials, in adult worm burden. Liver egg burdens were not significantly decreased in either drug-treated group but ova from both of these groups showed significant decreases in hatching ability (Rubb12-tri-68%, Rubb7-tnl-56%) and were significantly morphologically altered (Rubb12-tri-62% abnormal, Rubb7-tnl-35% abnormal). We hypothesize that the drugs exerted their activity, at least partially, through inhibition of both neuronal and tegumental acetylcholinesterases (AChEs), as worms treated in vitro showed significant decreases in activity of these enzymes. Further, treated parasites exhibited a significantly decreased ability to uptake glucose, significantly depleted glycogen stores and withered tubercules (a site of glycogen storage), implying drug-mediated interference in this nutrient acquisition pathway. CONCLUSIONS/SIGNIFICANCE Our data provide compelling evidence that ruthenium complexes are effective against all intra-mammalian stages of schistosomes, including schistosomula (refractory to PZQ) and eggs (agents of disease transmissibility). Further, the results of this study suggest that schistosome AChE is a target of ruthenium drugs, a finding that can inform modification of current compounds to identify analogues which are even more effective and selective against schistosomes.
Collapse
Affiliation(s)
- Madhu K. Sundaraneedi
- School of Physical, Environmental and Mathematical Sciences, UNSW Canberra, Canberra, Australian Capital Territory, Australia
| | - Bemnet A. Tedla
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Ramon M. Eichenberger
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Luke Becker
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Darren Pickering
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Michael J. Smout
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Siji Rajan
- School of Physical, Environmental and Mathematical Sciences, UNSW Canberra, Canberra, Australian Capital Territory, Australia
| | - Phurpa Wangchuk
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - F. Richard Keene
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
- School of Physical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - J. Grant Collins
- School of Physical, Environmental and Mathematical Sciences, UNSW Canberra, Canberra, Australian Capital Territory, Australia
| | - Mark S. Pearson
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| |
Collapse
|
49
|
Jin A, Dekan Z, Smout MJ, Wilson D, Dutertre S, Vetter I, Lewis RJ, Loukas A, Daly NL, Alewood PF. Conotoxin Φ‐MiXXVIIA from the Superfamily G2 Employs a Novel Cysteine Framework that Mimics Granulin and Displays Anti‐Apoptotic Activity. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201708927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ai‐Hua Jin
- Institute for Molecular Bioscience The University of Queensland St Lucia Queensland 4072 Australia
| | - Zoltan Dekan
- Institute for Molecular Bioscience The University of Queensland St Lucia Queensland 4072 Australia
| | - Michael J. Smout
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM James Cook University Smithfield, Cairns QLD 4878 Australia
| | - David Wilson
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM James Cook University Smithfield, Cairns QLD 4878 Australia
| | - Sébastien Dutertre
- Institute for Molecular Bioscience The University of Queensland St Lucia Queensland 4072 Australia
- Institut des Biomolécules Max Mousseron, UMR 5247 Université Montpellier, CNRS Place Eugène Bataillon 34095 Montpellier Cedex 5 France
| | - Irina Vetter
- Institute for Molecular Bioscience The University of Queensland St Lucia Queensland 4072 Australia
| | - Richard J. Lewis
- Institute for Molecular Bioscience The University of Queensland St Lucia Queensland 4072 Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM James Cook University Smithfield, Cairns QLD 4878 Australia
| | - Norelle L. Daly
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM James Cook University Smithfield, Cairns QLD 4878 Australia
| | - Paul F. Alewood
- Institute for Molecular Bioscience The University of Queensland St Lucia Queensland 4072 Australia
| |
Collapse
|
50
|
Jin AH, Dekan Z, Smout MJ, Wilson D, Dutertre S, Vetter I, Lewis RJ, Loukas A, Daly NL, Alewood PF. Conotoxin Φ-MiXXVIIA from the Superfamily G2 Employs a Novel Cysteine Framework that Mimics Granulin and Displays Anti-Apoptotic Activity. Angew Chem Int Ed Engl 2017; 56:14973-14976. [PMID: 28984021 DOI: 10.1002/anie.201708927] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Indexed: 12/12/2022]
Abstract
Conotoxins are a large family of disulfide-rich peptides that contain unique cysteine frameworks that target a broad range of ion channels and receptors. We recently discovered the 33-residue conotoxin Φ-MiXXVIIA from Conus miles with a novel cysteine framework comprising three consecutive cysteine residues and four disulfide bonds. Regioselective chemical synthesis helped decipher the disulfide bond connectivity and the structure of Φ-MiXXVIIA was determined by NMR spectroscopy. The 3D structure displays a unique topology containing two β-hairpins that resemble the N-terminal domain of granulin. Similar to granulin, Φ-MiXXVIIA promotes cell proliferation (EC50 17.85 μm) while inhibiting apoptosis (EC50 2.2 μm). Additional framework XXVII sequences were discovered with homologous signal peptides that define the new conotoxin superfamily G2. The novel structure and biological activity of Φ-MiXXVIIA expands the repertoire of disulfide-rich conotoxins that recognize mammalian receptors.
Collapse
Affiliation(s)
- Ai-Hua Jin
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Zoltan Dekan
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Michael J Smout
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM, James Cook University, Smithfield, Cairns, QLD, 4878, Australia
| | - David Wilson
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM, James Cook University, Smithfield, Cairns, QLD, 4878, Australia
| | - Sébastien Dutertre
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia.,Institut des Biomolécules Max Mousseron, UMR 5247, Université Montpellier, CNRS, Place Eugène Bataillon, 34095, Montpellier Cedex 5, France
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM, James Cook University, Smithfield, Cairns, QLD, 4878, Australia
| | - Norelle L Daly
- Centre for Biodiscovery and Molecular Development of Therapeutics, AITHM, James Cook University, Smithfield, Cairns, QLD, 4878, Australia
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia
| |
Collapse
|