1
|
Koike A, Brindley PJ. CRISPR/Cas genome editing, functional genomics, and diagnostics for parasitic helminths. Int J Parasitol 2025:S0020-7519(25)00092-X. [PMID: 40348052 DOI: 10.1016/j.ijpara.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Functional genomics using CRISPR (Clustered Regulatory Interspaced Short Palindromic Repeats)/Cas (CRISPR-associated endonuclease)-based approaches has revolutionized biomedical sciences. Gene editing is also widespread in parasitology generally and its use is increasing in studies on helminths including flatworm and roundworm parasites. Here, we survey the progress, specifically with experimental CRISPR-facilitated functional genomics to investigate helminth biology and pathogenesis, and also with the burgeoning use of CRISPR-based methods to assist in diagnosis of helminth infections. We also provide an historical timeline of the introduction and uses of CRISPR in helminth species to date.
Collapse
Affiliation(s)
- Akito Koike
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, D.C. 20037, USA
| | - Paul J Brindley
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, D.C. 20037, USA.
| |
Collapse
|
2
|
Apari P, Földvári G. How Do Trematodes Induce Cancer? A Possible Evolutionary Adaptation of an Oncogenic Agent Transmitted by Flukes. Evol Appl 2025; 18:e70070. [PMID: 39845579 PMCID: PMC11751881 DOI: 10.1111/eva.70070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/24/2025] Open
Abstract
There is strong epidemiological evidence that development of various cancer types is linked to infection with flukes (Platyhelminthes: Trematoda) in Africa, Asia and the Middle East. The exact nature of the mechanism by which cancer is induced by these parasites is unknown. Here, we provide a new hypothesis suggesting that flukes are not the primary cause of cancer but act as vectors of cancer-inducing microbial pathogens. These pathogens adaptively induce tumours to attract and help flukes to feed on blood from the tumour. Pathogen take-up by fluke vectors also takes place in the tumour; therefore, tumour formation in this case is the result of a mutualistic and adaptive relationship between the microbe and the helminth parasite. The suggested mechanism for cancer induction provided here may help us gain deeper understanding about cancer in general and its relationship with microbes and parasites. By further elaborating the unique nexus between flukes, carcinogenic microbes and cancer, in the future it will also help us to broaden our oncological perspective to reduce human death and suffering from this serious disease group.
Collapse
Affiliation(s)
| | - Gábor Földvári
- Institute of EvolutionHUN‐REN Centre for Ecological ResearchBudapestHungary
- Centre for Eco‐EpidemiologyNational Laboratory for Health SecurityBudapestHungary
| |
Collapse
|
3
|
Wagner M, Nishikawa H, Koyasu S. Reinventing type 2 immunity in cancer. Nature 2025; 637:296-303. [PMID: 39780006 DOI: 10.1038/s41586-024-08194-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 10/10/2024] [Indexed: 01/11/2025]
Abstract
Our understanding of type 2 immunity has undergone a substantial transformation in recent years, revealing previously unknown functions. Beyond its canonical role in defence against parasitic helminth infections, type 2 immunity safeguards the host through additional mechanisms, including the suppression of excessive type 1 immune responses, regulation of tissue repair and maintenance of adipose tissue homeostasis. However, unlike type 1 immune responses, type 2 immunity is perceived as a potential promoter of tumorigenesis. Emerging evidence challenges this perspective, painting a more nuanced picture in which type 2 immunity might protect against or even actively suppress tumour growth and progression. In this Review, we explore discoveries that highlight the potential of type 2 immunity in reshaping the landscape of cancer immunotherapies.
Collapse
Affiliation(s)
- Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network-PORT Polish Center for Technology Development, Wrocław, Poland.
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Cancer Immunology, Research Institute/EPOC, National Cancer Center, Tokyo, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- National Institutes for Quantum Science and Technology (QST), Chiba, Japan.
| |
Collapse
|
4
|
Lishai EA, Zaparina OG, Kapushchak YK, Sripa B, Hong SJ, Cheng G, Pakharukova MY. Comparative liver transcriptome analysis in hamsters infected with food-borne trematodes Opisthorchis felineus, Opisthorchis viverrini, or Clonorchis sinensis. PLoS Negl Trop Dis 2024; 18:e0012685. [PMID: 39652576 PMCID: PMC11627427 DOI: 10.1371/journal.pntd.0012685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Epidemiologically important food-borne trematodes Opisthorchis viverrini and Clonorchis sinensis are recognized as biological carcinogens of Group 1A, while Opisthorchis felineus is in Group 3 as noncarcinogenic to humans. Mechanisms of the biological carcinogenesis are still elusive. Some studies highlight chronic inflammation as a key factor and common pathway for cancer initiation and progression. Nonetheless, the chronic inflammation alone does not explain why these three species differ in carcinogenicity. We focused this study on genome-wide landscapes of liver gene expression and activation of cellular pathways in Mesocricetus auratus golden hamsters infected with C. sinensis (South Korea), O. viverrini (Thailand), or O. felineus (Russia) at 1 and 3 months after infection initiation. METHODOLOGY/PRINCIPAL FINDINGS Liver transcriptomes of golden hamsters (HiSeq Illumina, 2X150 bp) were sequenced at 1 and 3 months postinfection. Data processing was carried out using the following bioinformatic and experimental approaches: analysis of differential gene expression, estimates of proportions of affected liver cell types, liver histopathology, and examination of weighted gene coexpression networks. All infections caused enrichment with inflammatory response signaling pathways, fibrogenesis and cell proliferation, and IL2-STAT5, TNF-NF-κB, TGF-β, Hippo, MAPK, and PI3K-Akt signaling pathways. Nevertheless, species-specific responses to each infection were noted too. We also identified species-specific responses of liver cell types, differentially expressed gene clusters, and cellular pathways associated with structural liver damage (such as periductal fibrosis, epithelial neoplasia, and inflammation). CONCLUSIONS/SIGNIFICANCE This is the first comparative analysis of gene expression landscapes in the liver of experimental animals infected with O. viverrini, O. felineus, or C. sinensis. The trematodes have species-specific effects on the hepatobiliary system by triggering signaling pathways, thereby leading to differences in the severity of hepatobiliary structural lesions and contributing to the pathogenicity of closely related foodborne trematodes.
Collapse
Affiliation(s)
- Ekaterina A. Lishai
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Oxana G. Zaparina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), Novosibirsk, Russia
| | - Yaroslav K. Kapushchak
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), Novosibirsk, Russia
| | - Banchob Sripa
- Chung-Ang University College of Medicine, Seoul, Korea
| | - Sun-Jong Hong
- WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Guofeng Cheng
- Shanghai Tenth People’s Hospital, Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, Shanghai, China
| | - Maria Y. Pakharukova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
5
|
Smout MJ, Laha T, Chaiyadet S, Brindley PJ, Loukas A. Mechanistic insights into liver-fluke-induced bile-duct cancer. Trends Parasitol 2024; 40:1183-1196. [PMID: 39521672 DOI: 10.1016/j.pt.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Liver fluke infection is a major risk for cholangiocarcinoma (CCA). It has been established that the Asian liver flukes, Clonorchis sinensis and Opisthorchis viverrini secrete growth factors, digestive enzymes, and extracellular vesicles (EVs) which contribute to abnormal cell development in the bile ducts where the worms reside. These secretions - combined with aberrant inflammation and repeated cycles of chronic wounding at the site of parasite attachment and grazing on the epithelium - promote biliary hyperplasia and fibrosis and ultimately malignant transformation. Application of post-genomic and gene-editing tools to the study of liver fluke immunobiology and pathogenesis has accelerated the discovery of essential virulence factors to which targeted therapies and diagnostics can be directed.
Collapse
Affiliation(s)
- Michael J Smout
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Thailand
| | - Sujittra Chaiyadet
- Tropical Medicine Graduate Program, Academic Affairs, 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, George Washington University, Washington, DC, USA
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia.
| |
Collapse
|
6
|
Yang Z, Chan KW, Abu Bakar MZ, Deng X. Unveiling Drimenol: A Phytochemical with Multifaceted Bioactivities. PLANTS (BASEL, SWITZERLAND) 2024; 13:2492. [PMID: 39273976 PMCID: PMC11397239 DOI: 10.3390/plants13172492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
Drimenol, a phytochemical with a distinct odor is found in edible aromatic plants, such as Polygonum minus (known as kesum in Malaysia) and Drimys winteri. Recently, drimenol has received increasing attention owing to its diverse biological activities. This review offers the first extensive overview of drimenol, covering its sources, bioactivities, and derivatives. Notably, drimenol possesses a wide spectrum of biological activities, including antifungal, antibacterial, anti-insect, antiparasitic, cytotoxic, anticancer, and antioxidant effects. Moreover, some mechanisms of its activities, such as its antifungal effects against human mycoses and anticancer activities, have been investigated. However, there are still several crucial issues in the research on drimenol, such as the lack of experimental understanding of its pharmacokinetics, bioavailability, and toxicity. By synthesizing current research findings, this review aims to present a holistic understanding of drimenol, paving the way for future studies and its potential utilization in diverse fields.
Collapse
Affiliation(s)
- Zhongming Yang
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Kim Wei Chan
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Md Zuki Abu Bakar
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Department of Veterinary Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Xi Deng
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| |
Collapse
|
7
|
Rajasekharan SK, Ravichandran V, Boya BR, Jayachandran A, Lee J. Repurposing methuosis-inducing anticancer drugs for anthelmintic therapy. PLoS Pathog 2024; 20:e1012475. [PMID: 39235992 PMCID: PMC11376546 DOI: 10.1371/journal.ppat.1012475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Abstract
Drug-resistant parasitic nematodes pose a grave threat to plants, animals, and humans. An innovative paradigm for treating parasitic nematodes is emphasized in this opinion. This approach relies on repurposing methuosis (a death characterized by accumulation of large vacuoles) inducing anticancer drugs as anthelmintics. We review drugs/chemicals that have shown to kill nematodes or cancerous cells by inducing multiple vacuoles that eventually coalesce and rupture. This perspective additionally offers a succinct summary on Structure-Activity Relationship (SAR) of methuosis-inducing small molecules. This strategy holds promise for the development of broad-spectrum anthelmintics, shedding light on shared molecular mechanisms between cancer and nematodes in response to these inducers, thereby potentially transforming both therapeutic domains.
Collapse
Affiliation(s)
- Satish Kumar Rajasekharan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, India
| | - Vinothkannan Ravichandran
- Centre for Drug Discovery and Development (CD3), Amity Institute of Biotechnology, Amity University Maharashtra, Bhatan, Panvel, Mumbai, Maharashtra, India
| | - Bharath Reddy Boya
- School of Chemical Engineering, Yeungnum University, Gyeongsan, Republic of Korea
| | - Anirudh Jayachandran
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, India
| | - Jintae Lee
- School of Chemical Engineering, Yeungnum University, Gyeongsan, Republic of Korea
| |
Collapse
|
8
|
Tangkawattana S, Suyapoh W, Thongrin T, Wendo WD, Salao K, Suttiprapa S, Saichua P, Tangkawattana P. Inflammatory responses to Opisthorchis viverrini infection in animal models: A comparison between susceptible and nonsusceptible hosts in different anatomical locations. Open Vet J 2024; 14:664-673. [PMID: 38549578 PMCID: PMC10970119 DOI: 10.5455/ovj.2024.v14.i2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/16/2024] [Indexed: 04/02/2024] Open
Abstract
Background Inflammation caused by Opisthorchis viverrini infection increases the risk of cholangitis, cholecystitis, and leads to bile duct cancer (cholangiocarcinoma or CCA). However, only certain infected individuals are susceptible to CCA, suggesting the involvement of host factors in cancer development. In addition, there are reports indicating differences in the locations of CCA. Aim This study aims to investigate cellular inflammatory responses in the common bile duct (CB), intrahepatic bile duct (IHB), and gallbladder (GB) in susceptible and non-susceptible hosts following O. viverrini infection. Methods Thirty Syrian golden hamsters (a susceptible host) and 30 BALB/c mice (a non-susceptible host) infected with O. viverrini were studied at six time points (five animals per group). Histopathological evaluations were conducted on samples from the IHB, CB, and GB. Inflammatory cell infiltration was quantitatively assessed and compared between groups and time points. Statistical analysis was performed using one-way ANOVA, with a significance level of p < 0.05. Results Inflammation was significantly more pronounced in the IHB compared to the other two biliary locations. In comparison between susceptible and non-susceptible hosts, the intensity of inflammation was higher in the OV+H group than in the OV+M group (p < 0.05). Conclusion This study highlights the association between host response to inflammation, tissue location, and host susceptibility, with the IHB showing particular susceptibility to inflammation and pathological changes. These findings contribute to our understanding of the increased risk of CCA in susceptible hosts.
Collapse
Affiliation(s)
- Sirikachorn Tangkawattana
- 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, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharapol Suyapoh
- Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - Theerayut Thongrin
- Graduate Program, Faculty of Veterinary Medicine, KhonKaen University, Khon Kaen, Thailand
- Faculty of Veterinary Medicine, Western University, Kanchanaburi, Thailand
| | - Woro Danur Wendo
- Graduate Program, Faculty of Veterinary Medicine, KhonKaen University, Khon Kaen, Thailand
- Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Kanin Salao
- Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sutas Suttiprapa
- WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Khon Kaen University, Khon Kaen, Thailand
- Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Prasert Saichua
- WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Khon Kaen University, Khon Kaen, Thailand
- Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | |
Collapse
|
9
|
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
|
10
|
Tangkawattana S, Suyapoh W, Taiki N, Tookampee P, Chitchak R, Thongrin T, Tangkawattana P. Unraveling the relationship among inflammatory responses, oxidative damage, and host susceptibility to Opisthorchis viverrini infection: A comparative analysis in animal models. Vet World 2023; 16:2303-2312. [PMID: 38152278 PMCID: PMC10750739 DOI: 10.14202/vetworld.2023.2303-2312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/16/2023] [Indexed: 12/29/2023] Open
Abstract
Background and Aim Opisthorchis viverrini infection-induced inflammation contributes to cholangiocarcinoma (CCA) development in humans and animals. Inflammation generates free radicals, such as reactive oxygen species and reactive nitrogen species (RNS), which damage the host's DNA. However, only 5% of O. viverrini-infected individuals develop malignancy, suggesting that variations in the inflammatory response of individuals to the parasite may influence susceptibility. Due to limitations in studying human susceptibility, we used an animal model to investigate the profiles of inflammatory reactions, oxidative burst, and irreversible DNA damage. This study aimed to explore the potential role of inflammation and RNS in causing DNA damage that may predispose susceptible hosts and non-susceptible animal models to cancer development in O. viverrini infection. Materials and Methods This experimental study was conducted on 30 Syrian golden hamsters (OV-H) and 30 BALB/c mice (OV-M) infected with O. viverrini, representing susceptible and non-susceptible models, respectively. Five animals per group were examined at six predetermined time points during the experiment. Biliary tract samples were systematically investigated using histopathological evaluation for inflammatory cell infiltration and immunohistochemical staining for RNS production and markers of DNA damage, including nitrotyrosine and 8-hydroxy-2'-deoxyguanosine. These features were quantified and compared among the experimental groups. Mann-Whitney U-test was used for statistical analysis, with p < 0.05 considered statistically significant. Results The comparison revealed that the OV-M group exhibited significantly earlier and higher rates of inflammatory cell infiltration during the acute phase, whereas the OV-H group exhibited chronic and more severe inflammation (p < 0.020). Intracellular RNS production and DNA damage were closely associated with the inflammatory response. Conclusion This study demonstrates differential responses in susceptible and non-susceptible models of O. viverrini infection regarding disease onset and duration, as well as intracellular RNS production and DNA damage caused by inflammation. Persistent inflammation generated oxidatively damaged DNA, which is a distinct pathological characteristic of susceptible hosts and may be critical for CCA development.
Collapse
Affiliation(s)
- Sirikachorn Tangkawattana
- Department of Veterinary Pathobiology, 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, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharapol Suyapoh
- Department of Veterinary Science, Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - Nathamon Taiki
- Doctor of Veterinary Medicine Program, Faculty of Veterinary Medicine, Khon Kaen University, Thailand
| | - Paramin Tookampee
- Doctor of Veterinary Medicine Program, Faculty of Veterinary Medicine, Khon Kaen University, Thailand
| | - Ravisara Chitchak
- Doctor of Veterinary Medicine Program, Faculty of Veterinary Medicine, Khon Kaen University, Thailand
| | - Theerayut Thongrin
- Master of Science Program in Veterinary Science, Faculty of Veterinary Medicine, Khon Kaen University, Thailand
| | - Prasarn Tangkawattana
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
11
|
Fonti N, Parisi F, Mancianti F, Freer G, Poli A. Cancerogenic parasites in veterinary medicine: a narrative literature review. Infect Agent Cancer 2023; 18:45. [PMID: 37496079 PMCID: PMC10373346 DOI: 10.1186/s13027-023-00522-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023] Open
Abstract
Parasite infection is one of the many environmental factors that can significantly contribute to carcinogenesis and is already known to be associated with a variety of malignancies in both human and veterinary medicine. However, the actual number of cancerogenic parasites and their relationship to tumor development is far from being fully understood, especially in veterinary medicine. Thus, the aim of this review is to investigate parasite-related cancers in domestic and wild animals and their burden in veterinary oncology. Spontaneous neoplasia with ascertained or putative parasite etiology in domestic and wild animals will be reviewed, and the multifarious mechanisms of protozoan and metazoan cancer induction will be discussed.
Collapse
Affiliation(s)
- Niccolò Fonti
- Dipartimento di Scienze veterinarie, Università di Pisa, Viale delle Piagge, 2, 56124, Pisa, Italy.
| | - Francesca Parisi
- Dipartimento di Scienze veterinarie, Università di Pisa, Viale delle Piagge, 2, 56124, Pisa, Italy
| | - Francesca Mancianti
- Dipartimento di Scienze veterinarie, Università di Pisa, Viale delle Piagge, 2, 56124, Pisa, Italy
| | - Giulia Freer
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Via Savi, 10, 56126, Pisa, Italy
| | - Alessandro Poli
- Dipartimento di Scienze veterinarie, Università di Pisa, Viale delle Piagge, 2, 56124, Pisa, Italy
| |
Collapse
|
12
|
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] [Grants] [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
|
13
|
Fedorova OS, Kovshirina AE, Kovshirina YV, Hattendorf J, Onishchenko SV, Katanakhova LL, Taslicki SS, Chizhikov AV, Tataurov IA, Vtorushin SV, Sripa B, Ogorodova LM, Odermatt P. Opisthorchis Felineus Infection is a Risk Factor for Cholangiocarcinoma in Western Siberia: A Hospital-based Case-control Study. Clin Infect Dis 2023; 76:e1392-e1398. [PMID: 35723279 DOI: 10.1093/cid/ciac497] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA), a fatal bile duct cancer, has a high incidence in Western Siberia, Russian Federation. In addition, Opisthorchis felineus, a bile duct-dwelling trematode liver fluke is highly endemic. Closely related species have been shown to be cancerogenic agents in Asia. We therefore examined the association between O felineus infection and CCA in Western Siberia. METHODS We conducted a hospital-based, individually matched case-control study between January 2017 and August 2020 in Tomsk Oblast and Khanty-Mansiysk Autonomous Okrug, Yugra, Russian Federation. Histologically confirmed CCA patients (cases) were compared with matched age, sex, and place of residence hospital controls. The examination of study participants included the diagnosis of current and past O felineus infection, abdominal ultrasonographical assessment, physical examination, and interview on exposures to potential risk factors. RESULTS We identified 40 patients with CCA and 160 controls. Exposures to O felineus infection was strongly associated with CCA (odds ratio [OR], 3.9; 95% confidence interval [CI], 1.4-10.8; P = .008). Also, cases reported more often that they were currently or in the past were infected by O felineus compared with controls (OR, 4.03; 95% CI, 1.7-9.5; P = .001). Furthermore, cases reported river fish consumption and fishing habits significantly more often than controls (OR, 5.5; 95% CI, 1.5-19.8; P = .009 and OR, 3.3; 95% CI, 1.4-7.7; P = .005). CONCLUSIONS The study results revealed a strong significantly increased risk for CCA development in O felineus-infected individuals. Elaboration of the guidelines on screening programs for early CCA diagnosis, prevention, and treatment is socially important in endemic regions.
Collapse
Affiliation(s)
- Olga S Fedorova
- Federal State Budget Educational Institution of Higher Education, Siberian State Medical University, Ministry of Healthcare of Russian Federation, Tomsk, Russian Federation
| | - Anna E Kovshirina
- Federal State Budget Educational Institution of Higher Education, Siberian State Medical University, Ministry of Healthcare of Russian Federation, Tomsk, Russian Federation
| | - Yulia V Kovshirina
- Federal State Budget Educational Institution of Higher Education, Siberian State Medical University, Ministry of Healthcare of Russian Federation, Tomsk, Russian Federation
| | - Jan Hattendorf
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland.,University of Basel, Basel, Switzerland
| | - Sergey V Onishchenko
- Surgut State University, Surgut, Russian Federation.,Surgut Regional Clinical Hospital 14, Surgut, Russian Federation
| | | | | | | | - Ilya A Tataurov
- Regional Clinical Hospital 40, Khanty-Mansiysk, Russian Federation
| | - Sergey V Vtorushin
- Federal State Budget Educational Institution of Higher Education, Siberian State Medical University, Ministry of Healthcare of Russian Federation, Tomsk, Russian Federation
| | | | - Ludmila M Ogorodova
- Federal State Budget Educational Institution of Higher Education, Siberian State Medical University, Ministry of Healthcare of Russian Federation, Tomsk, Russian Federation
| | - Peter Odermatt
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland.,University of Basel, Basel, Switzerland
| |
Collapse
|
14
|
microRNAs: Critical Players during Helminth Infections. Microorganisms 2022; 11:microorganisms11010061. [PMID: 36677353 PMCID: PMC9861972 DOI: 10.3390/microorganisms11010061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
microRNAs (miRNAs) are a group of small non-coding RNAs that regulate gene expression post-transcriptionally through their interaction with the 3' untranslated regions (3' UTR) of target mRNAs, affecting their stability and/or translation. Therefore, miRNAs regulate biological processes such as signal transduction, cell death, autophagy, metabolism, development, cellular proliferation, and differentiation. Dysregulated expression of microRNAs is associated with infectious diseases, where miRNAs modulate important aspects of the parasite-host interaction. Helminths are parasitic worms that cause various neglected tropical diseases affecting millions worldwide. These parasites have sophisticated mechanisms that give them a surprising immunomodulatory capacity favoring parasite persistence and establishment of infection. In this review, we analyze miRNAs in infections caused by helminths, emphasizing their role in immune regulation and its implication in diagnosis, prognosis, and the development of therapeutic strategies.
Collapse
|
15
|
Bellini I, Scribano D, Sarshar M, Ambrosi C, Pizzarelli A, Palamara AT, D’Amelio S, Cavallero S. Inflammatory Response in Caco-2 Cells Stimulated with Anisakis Messengers of Pathogenicity. Pathogens 2022; 11:1214. [PMID: 36297271 PMCID: PMC9611079 DOI: 10.3390/pathogens11101214] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 09/07/2024] Open
Abstract
Background: Anisakis spp. third-stage larvae (L3) are the causative agents of human zoonosis called anisakiasis. The accidental ingestion of L3 can cause acute and chronic inflammation at the gastric, intestinal, or ectopic levels. Despite its relevance in public health, studies on pathogenetic mechanisms and parasite-human interplay are scarce. The aim of this study was to investigate the human inflammatory response to different Anisakis vehicles of pathogenicity. Methods: Human colorectal adenocarcinoma (Caco-2) cells were exposed to Anisakis L3 (the initial contact with the host), extracellular vesicles (EVs, Anisakis-host communication), and crude extract (CE, the larval dying). The protein quantity and gene expression of two pro-inflammatory cytokines (IL-6 and IL-8) were investigated using an ELISA test (6 h and 24 h) and a qReal-Time PCR (1 h, 6 h, and 24 h), respectively. Results: The L3 and EVs induced a downregulation in both the Il-6 and Il-8 gene expression and protein quantity. On the contrary, the CE stimulated IL-6 gene expression and its protein release, not affecting IL-8. Conclusions: The Caco-2 cells seemed to not react to the exposure to the L3 and EVs, suggesting a parasite's immunomodulating action to remain alive in an inhospitable niche. Conversely, the dying larva (CE) could induce strong activation of the immune strategy of the host that, in vivo, would lead to parasite expulsion, eosinophilia, and/or granuloma formation.
Collapse
Affiliation(s)
- Ilaria Bellini
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Daniela Scribano
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Cecilia Ambrosi
- Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, IRCCS, 00166 Rome, Italy
| | - Antonella Pizzarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Anna Teresa Palamara
- Laboratory Affiliated to Institute Pasteur Italia—Cenci Bolognetti Foundation, Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
- Department of Infectious Diseases, National Institute of Health, 00185 Rome, Italy
| | - Stefano D’Amelio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Serena Cavallero
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
16
|
Liu X, Jiang Y, Ye J, Wang X. Helminth infection and helminth-derived products: A novel therapeutic option for non-alcoholic fatty liver disease. Front Immunol 2022; 13:999412. [PMID: 36263053 PMCID: PMC9573989 DOI: 10.3389/fimmu.2022.999412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely related to obesity, diabetes, and metabolic syndrome (MetS), and it has become the most common chronic liver disease. Helminths have co-evolved with humans, inducing multiple immunomodulatory mechanisms to modulate the host's immune system. By using their immunomodulatory ability, helminths and their products exhibit protection against various autoimmune and inflammatory diseases, including obesity, diabetes, and MetS, which are closely associated with NAFLD. Here, we review the pathogenesis of NAFLD from abnormal glycolipid metabolism, inflammation, and gut dysbiosis. Correspondingly, helminths and their products can treat or relieve these NAFLD-related diseases, including obesity, diabetes, and MetS, by promoting glycolipid metabolism homeostasis, regulating inflammation, and restoring the balance of gut microbiota. Considering that a large number of clinical trials have been carried out on helminths and their products for the treatment of inflammatory diseases with promising results, the treatment of NAFLD and obesity-related diseases by helminths is also a novel direction and strategy.
Collapse
Affiliation(s)
- Xi Liu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuyun Jiang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jixian Ye
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Nuclear Medicine and Institute of Digestive Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
17
|
Oliveira FMS, Cruz RE, Pinheiro GRG, Caliari MV. Comorbidities involving parasitic diseases: A look at the benefits and complications. Exp Biol Med (Maywood) 2022; 247:1819-1826. [PMID: 35876147 PMCID: PMC9679356 DOI: 10.1177/15353702221108387] [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] [Indexed: 12/29/2022] Open
Abstract
Parasitic infections acquired by the population cause substantial morbidity worldwide, with individuals from developing countries being most affected. Some parasites remain in the host for long periods, settling in different organs, manipulating the flow of nutrients and metabolites, and influencing the immune response, favoring their adaptation. The host attempts to counteract the metabolic and immunological alterations and the possible damage caused by infection. These metabolic and immunological changes experienced by the host can influence the progression of other existing morbidities or those that will be acquired in the future. Cancer and metabolic diseases are also frequent causes of morbidity in the world population. The large numbers of individuals affected by cancer and metabolic diseases and the high prevalence of morbidity caused by parasitic diseases favor the development of comorbidity involving these pathologies. This review provides an overview of major advances in research on cancer and metabolic diseases associated with parasitic infections. Information about hosts and parasites such as alterations of the immune response, metabolism and adaptation mechanisms of the parasites, and parasitic molecules with therapeutic potential is provided, as well as the beneficial results or complications related to the comorbidities discussed herein. We emphasize the need to conduct additional studies addressing comorbidities associated with parasitic infections to improve the understanding of the impact of this association on the progression of morbidities, as well as the possibility of the therapeutic use of and therapeutic approaches involving parasites.
Collapse
Affiliation(s)
- Fabrício Marcus Silva Oliveira
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Ruth Elizabeth Cruz
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Guilherme Rafael Gomide Pinheiro
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Marcelo Vidigal Caliari
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil,Marcelo Vidigal Caliari.
| |
Collapse
|
18
|
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: 3.0] [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
|
19
|
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: 44] [Impact Index Per Article: 14.7] [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
|
20
|
Rodpai R, Luvira V, Sadaow L, Sukeepaisarnjaroen W, Kitkhuandee A, Paonariang K, Sanpool O, Ittiprasert W, Mann VH, Intapan PM, Brindley PJ, Maleewong W. Rapid assessment of Opisthorchis viverrini IgG antibody in serum: a potential diagnostic biomarker to predict risk of cholangiocarcinoma in regions endemic for opisthorchiasis. Int J Infect Dis 2021; 116:80-84. [PMID: 34954313 PMCID: PMC9569029 DOI: 10.1016/j.ijid.2021.12.347] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/09/2021] [Accepted: 12/18/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Opisthorchiasis is caused by an infection with fish-borne liver flukes of the genus Opisthorchis. Opisthorchiasis frequently leads to chronic inflammation in the biliary tract and is classified as a group 1 biological carcinogen by the International Agency for Research on Cancer: a definitive risk for cholangiocarcinoma (CCA). Methods: We used the rapid immunochromatographic test (ICT) to detect anti-Opisthorchis viverrini IgG and IgG4 subclass antibodies in sera of patients with CCA. The ICT kits were developed based on soluble antigens excreted and secreted by O. viverrini adult worms. Results: ICT indicated sera was positive for IgG and IgG4 antibodies, respectively, in 22 (61.1%) and 15 (41.6%) participants of the 36 study participants diagnosed with CCA (P > 0.05). Our study also included groups with other cancers and with liver cirrhosis, where the IgG ICT and IgG4 ICT kits were 27.7% (13/47) and 25.5% (12/47) positive, respectively (P > 0.05). Neither total the IgG ICT nor the IgG4 ICT yielded positive results in a control group of 20 healthy participants. Moreover, the percentage positivity rate using the ICT for total IgG between the CCA group and the other cancers and liver cirrhosis group was significantly different (P < 0.05). By contrast, no significant difference between these groups was apparent in the ICT for IgG4 antibody. The CCA group was 6.53 times more likely to have positive anti–O. viverrini IgG antibody (odds ratio 6.53, P < 0.001) and 3.27 times more likely to have positive anti–O. viverrini IgG4 antibody (odds ratio 3.27, P = 0.010) than the non-CCA group. Conclusion: This information is of potential value for the development of a diagnostic biomarker to predict risk for O. viverrini infection-associated CCA.
Collapse
Affiliation(s)
- Rutchanee Rodpai
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Vor Luvira
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Lakkhana Sadaow
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | | | - Amnat Kitkhuandee
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Krisada Paonariang
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Oranuch Sanpool
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Wannaporn Ittiprasert
- Department of Microbiology, Immunology and Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine and Health Science, George Washington University, Washington, District of Columbia, USA
| | - Victoria H Mann
- Department of Microbiology, Immunology and Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine and Health Science, George Washington University, Washington, District of Columbia, USA
| | - Pewpan M Intapan
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine and Health Science, George Washington University, Washington, District of Columbia, USA
| | - Wanchai Maleewong
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Mekong Health Science Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
21
|
Drurey C, Maizels RM. Helminth extracellular vesicles: Interactions with the host immune system. Mol Immunol 2021; 137:124-133. [PMID: 34246032 PMCID: PMC8636279 DOI: 10.1016/j.molimm.2021.06.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/14/2021] [Accepted: 06/23/2021] [Indexed: 12/21/2022]
Abstract
As long-lived parasites, helminths depend upon immunomodulation of their hosts for survival. The release of excretory-secretory (ES) products, including proteins, lipids and RNAs is how successful host manipulation is achieved. It has recently been discovered that the ES products of helminths contain extracellular vesicles (EVs), with every species investigated found to secrete these lipid-bound structures. EVs are perfect for packaging and delivering immune modulators to target cell types. This review outlines the research carried out on helminth EVs and their constituents thus far, as well as their interaction with components of the mammalian immune system. We discuss how targeting EVs will aid treatment of helminth infection and consider how EVs and their immunomodulatory cargo could be used as therapeutics as we progress through this exciting era.
Collapse
Affiliation(s)
- Claire Drurey
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK.
| |
Collapse
|
22
|
Perera DJ, Ndao M. Promising Technologies in the Field of Helminth Vaccines. Front Immunol 2021; 12:711650. [PMID: 34489961 PMCID: PMC8418310 DOI: 10.3389/fimmu.2021.711650] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/26/2021] [Indexed: 12/18/2022] Open
Abstract
Helminths contribute a larger global burden of disease than both malaria and tuberculosis. These eukaryotes have caused human infections since before our earliest recorded history (i.e.: earlier than 1200 B.C. for Schistosoma spp.). Despite the prevalence and importance of these infections, helminths are considered a neglected tropical disease for which there are no vaccines approved for human use. Similar to other parasites, helminths are complex organisms which employ a plethora of features such as: complex life cycles, chronic infections, and antigenic mimicry to name a few, making them difficult to target by conventional vaccine strategies. With novel vaccine strategies such as viral vectors and genetic elements, numerous constructs are being defined for a wide range of helminth parasites; however, it has yet to be discussed which of these approaches may be the most effective. With human trials being conducted, and a pipeline of potential anti-helminthic antigens, greater understanding of helminth vaccine-induced immunity is necessary for the development of potent vaccine platforms and their optimal design. This review outlines the conventional and the most promising approaches in clinical and preclinical helminth vaccinology.
Collapse
Affiliation(s)
- Dilhan J. Perera
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Program of Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Momar Ndao
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Program of Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- National Reference Centre for Parasitology, Research Institute of McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
23
|
Masamba P, Kappo AP. Immunological and Biochemical Interplay between Cytokines, Oxidative Stress and Schistosomiasis. Int J Mol Sci 2021; 22:ijms22137216. [PMID: 34281269 PMCID: PMC8268096 DOI: 10.3390/ijms22137216] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/20/2021] [Accepted: 06/20/2021] [Indexed: 12/17/2022] Open
Abstract
The host–parasite schistosome relationship relies heavily on the interplay between the strategies imposed by the schistosome worm and the defense mechanisms the host uses to counter the line of attack of the parasite. The ultimate goal of the schistosome parasite entails five important steps: evade elimination tactics, survive within the human host, develop into adult forms, propagate in large numbers, and transmit from one host to the next. The aim of the parasitized host on the other hand is either to cure or limit infection. Therefore, it is a battle between two conflicting aspirations. From the host’s standpoint, infection accompanies a plethora of immunological consequences; some are set in place to defend the host, while most end up promoting chronic disease, which ultimately crosses paths with oxidative stress and cancer. Understanding these networks provides attractive opportunities for anti-schistosome therapeutic development. Hence, this review discusses the mechanisms by which schistosomes modulate the human immune response with ultimate links to oxidative stress and genetic instability.
Collapse
|
24
|
Hepatobiliary morbidities detected by ultrasonography in Opisthorchis viverrini-infected patients before and after praziquantel treatment: a five-year follow up study. Acta Trop 2021; 217:105853. [PMID: 33548204 DOI: 10.1016/j.actatropica.2021.105853] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 01/15/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022]
Abstract
Infection of the liver fluke, Opisthorchis viverrini (OV) is an important public health problem in northeast Thailand and adjacent countries, where people have a habit of eating raw or undercooked fish. A community case-control study was carried out with 8,936 participants from 89 villages, in Khon Kaen province, Thailand. There were 3,359 OV-infected participants all of whom underwent ultrasonography of upper abdomen for the evaluation of hepatobiliary morbidity. The participants with advanced periductal fibrosis (APF) by ultrasound (n = 785) were invited to undergo annual follow-up ultrasonography for five years after praziquantel treatment. The sonographer was blinded with respect to status of OV infection at each visit. The study findings revealed variability in the study population profile of the hepatobiliary morbidities before and after praziquantel treatment over the follow up interval. At the end of the study, 32 (30.8%) out of 104 participants showed no relapse of APF whereas, by contrast, 39 (37.5%) participants showed relapse or persistent APF since the outset of the study (≥ two consecutive visits). The APF in most follow-up visits was significantly associated with male sex, with intrahepatic duct stones, with the width of the gallbladder "pre" minus "post" fatty meal, and with the ratio of left lobe of the liver to aorta. Five cases of suspected cholangiocarcinoma were observed over the five years of follow-up. This long-term ultrasound follow-up study demonstrates a significant incidence of persistent APF in over one-third of opisthorchiasis cases after praziquantel treatment, findings that support the prospect of ongoing cholangiocarcinogenesis long after successful elimination of liver fluke infection among the population.
Collapse
|
25
|
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: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
26
|
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: 0.8] [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
|
27
|
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: 0.4] [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
|
28
|
Carson JP, Robinson MW, Hsieh MH, Cody J, Le L, You H, McManus DP, Gobert GN. A comparative proteomics analysis of the egg secretions of three major schistosome species. Mol Biochem Parasitol 2020; 240:111322. [PMID: 32961206 PMCID: PMC8059868 DOI: 10.1016/j.molbiopara.2020.111322] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/20/2022]
Abstract
Morbidity associated with hepatic and urogenital schistosomiasis stems primarily from the host immune response directed against schistosome eggs. When eggs become entrapped in host tissues, the development of fibrotic plaques drives downstream pathology. These events occur due to the antigenic nature of egg excretory/secretory products (ESPs). Both Schistosoma mansoni and S. japonicum ESPs have been shown to interact with several cell populations in the host liver including hepatocytes, macrophages, and hepatic stellate cells, with both immunomodulatory and pathological consequences. Several protein components of the ESPs of S. mansoni and S. japonicum eggs have been characterised; however, studies into the collective contents of schistosome egg ESPs are lacking. Utilising shotgun mass spectrometry and an array of in silico analyses, we identified 266, 90 and 50 proteins within the S. mansoni, S. japonicum and S. haematobium egg secretomes respectively. We identified numerous proteins with already established immunomodulatory activities, vaccine candidates and vesicle markers. Relatively few common orthologues within the ESPs were identified by BLAST, indicating that the three egg secretomes differ in content significantly. Having a clearer understanding of these components may lead to the identification of new proteins with uncharacterised immunomodulatory potential or pathological relevance. This will enhance our understanding of host-parasite interactions, particularly those occurring during chronic schistosomiasis, and pave the way towards novel therapeutics and vaccines.
Collapse
Affiliation(s)
- Jack P Carson
- School of Biological Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Mark W Robinson
- School of Biological Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Michael H Hsieh
- Division of Urology, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | | | | | - Hong You
- Molecular Parasitology Laboratory, Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Donald P McManus
- Molecular Parasitology Laboratory, Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Geoffrey N Gobert
- School of Biological Sciences, Queen's University Belfast, Belfast, United Kingdom.
| |
Collapse
|
29
|
Bernardo C, Santos J, Costa C, Tavares A, Amaro T, Marques I, Gouveia MJ, Félix V, Afreixo V, Brindley PJ, Costa JM, Amado F, Helguero L, Santos LL. Estrogen receptors in urogenital schistosomiasis and bladder cancer: Estrogen receptor alpha-mediated cell proliferation. Urol Oncol 2020; 38:738.e23-738.e35. [PMID: 32507545 DOI: 10.1016/j.urolonc.2020.04.022] [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: 11/16/2019] [Revised: 03/30/2020] [Accepted: 04/21/2020] [Indexed: 01/22/2023]
Abstract
Estrogen-like metabolites have been identified in S. haematobium, the helminth parasite that causes urogenital schistosomiasis (UGS) and in patients´ blood and urine during UGS. Estrogen receptor (ER) activation is enriched in the luminal molecular subtype bladder cancer (BlaCa). To date, the significance of ER to these diseases remains elusive. We evaluated ERα and ERβ expression in UGS-related BlaCa (n = 27), UGS-related non-malignant lesions (n = 35), and noninfected BlaCa (n = 80). We investigated the potential of ERα to recognize S. haematobium-derived metabolites by docking and molecular dynamics simulations and studied ERα modulation in vitro using 3 BlaCa cell lines, T24, 5637 and HT1376. ERα was expressed in tumor and stromal cells in approximately 20% noninfected cases and in 30% of UGS-related BlaCa, predominantly in the epithelial cells. Overall, ERα expression was associated with features of tumor aggressiveness such as high proliferation and p53 positive expression. ERα expression correlated with presence of schistosome eggs. ERβ was widely expressed in both cohorts but weaker in UGS-related cases. molecular dynamics simulations of the 4 most abundant S. haematobium-derived metabolites revealed that smaller metabolites have comparable affinity for the ERα active state than 17β-estradiol, while the larger metabolites present higher affinity. Our in vitro findings suggested that ERα activation promotes proliferation in ERα expressing BlaCa cells and that this can be reverted with anti-estrogenic therapy. In summary, we report differential ER expression between UGS-related BlaCa and noninfected BlaCa and provide evidence supporting a role of active ERα during UGS and UGS-induced carcinogenesis.
Collapse
Affiliation(s)
- Carina Bernardo
- Hormones and Cancer Lab, Department of Medical Sciences, Institute of Biomedicine, iBiMED, University of Aveiro, Aveiro, Portugal; Experimental Pathology and Therapeutics, Research Center, Portuguese Oncology Institute - Porto (IPO-Porto), Porto, Portugal
| | - Júlio Santos
- Urology Department, Hospital Américo Boavida, Luanda, Angola; Center for the Study of Animal Science, CECA/ICETA, University of Porto, Porto, Portugal
| | - Céu Costa
- Fernando Pessoa Energy, Environment and Health Research Unit/Biomedical Research Center (FP-ENAS/CEBIMED), Faculty of Health Sciences, Fernando Pessoa University, Porto, Portugal
| | - Ana Tavares
- Experimental Pathology and Therapeutics, Research Center, Portuguese Oncology Institute - Porto (IPO-Porto), Porto, Portugal
| | - Teresina Amaro
- Department of Pathology, Hospital Pedro Hispano, Matosinhos, Portugal
| | - Igor Marques
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Maria João Gouveia
- Center for the Study of Animal Science, CECA/ICETA, University of Porto, Porto, Portugal; Department of Infectious Diseases, R&D Unit, INSA-National Health Institute Dr. Ricardo Jorge, Porto, Portugal
| | - Vítor Félix
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Vera Afreixo
- Center for Research and Development in Mathematics and Applications (CIDMA), Department of Mathematics, Aveiro, Portugal
| | - Paul J Brindley
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC, USA
| | - José Manuel Costa
- Center for the Study of Animal Science, CECA/ICETA, University of Porto, Porto, Portugal; Department of Infectious Diseases, R&D Unit, INSA-National Health Institute Dr. Ricardo Jorge, Porto, Portugal
| | - Francisco Amado
- Mass Spectrometry Group, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Luisa Helguero
- Hormones and Cancer Lab, Department of Medical Sciences, Institute of Biomedicine, iBiMED, University of Aveiro, Aveiro, Portugal
| | - Lúcio L Santos
- Experimental Pathology and Therapeutics, Research Center, Portuguese Oncology Institute - Porto (IPO-Porto), Porto, Portugal; Department of Surgical Oncology, Portuguese Oncology Institute - Porto (IPO-Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
| |
Collapse
|
30
|
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: 3.2] [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
|
31
|
Roy S, Glaser S, Chakraborty S. Inflammation and Progression of Cholangiocarcinoma: Role of Angiogenic and Lymphangiogenic Mechanisms. Front Med (Lausanne) 2019; 6:293. [PMID: 31921870 PMCID: PMC6930194 DOI: 10.3389/fmed.2019.00293] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022] Open
Abstract
Cholangiocarcinoma (CCA), or cancer of the biliary epithelium is a relatively rare but aggressive form of biliary duct cancer which has a 5-year survival rate post metastasis of 2%. Although a number of risk factors are established for CCA growth and progression, a careful evaluation of the existing literature on CCA reveals that an inflammatory environment near the biliary tree is the most common causal link between the risk factors and the development of CCA. The fact that inflammation predisposes affected individuals to CCA is further bolstered by multiple observations where the presence and maintenance of an inflammatory microenvironment at the site of the primary tumor plays a significant role in the development and metastasis of CCA. In addition, mechanisms activating the tumor vasculature and enhancing angiogenesis and lymphangiogenesis significantly contribute to CCA aggressiveness and metastasis. This review aims to address the role of an inflammatory microenvironment-CCA crosstalk and will present the basic concepts, observations, and current perspectives from recent research studies in the field of tumor stroma of CCA.
Collapse
Affiliation(s)
- Sukanya Roy
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, TX, United States
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, TX, United States
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, TX, United States
| |
Collapse
|
32
|
Arora N, Kaur R, Anjum F, Tripathi S, Mishra A, Kumar R, Prasad A. Neglected Agent Eminent Disease: Linking Human Helminthic Infection, Inflammation, and Malignancy. Front Cell Infect Microbiol 2019; 9:402. [PMID: 31867284 PMCID: PMC6909818 DOI: 10.3389/fcimb.2019.00402] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Helminthic parasitic infection is grossly prevalent across the globe and is considered a significant factor in human cancer occurrence induced by biological agents. Although only three helminths (Schistosoma haematobium, Clonorchis sinensis, and Opisthorchis viverrini) so far have been directly associated with carcinogenesis; there are evidence suggesting the involvement of other species too. Broadly, human helminthiasis can cause chronic inflammation, genetic instability, and host immune modulation by affecting inter- and intracellular communications, disruption of proliferation-anti-proliferation pathways, and stimulation of malignant stem cell progeny. These changes ultimately lead to tumor development through the secretion of soluble factors that interact with host cells. However, the detailed mechanisms by which helminths introduce and promote malignant transformation of host cells are still not clear. Here, we reviewed the current understanding of immune-pathogenesis of helminth parasites, which have been associated with carcinogenesis, and how these infections initiate carcinogenesis in the host.
Collapse
Affiliation(s)
- Naina Arora
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Rimanpreet Kaur
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Farhan Anjum
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Shweta Tripathi
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Karwar, India
| | - Rajiv Kumar
- Institute for Himalayan Bioresource Technology (CSIR), Palampur, India
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| |
Collapse
|
33
|
Gouveia MJ, Nogueira V, Araújo B, Gärtner F, Vale N. Inhibition of the Formation In Vitro of Putatively Carcinogenic Metabolites Derived from S. haematobium and O. viverrini by Combination of Drugs with Antioxidants. Molecules 2019; 24:E3842. [PMID: 31731402 PMCID: PMC6864706 DOI: 10.3390/molecules24213842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 11/24/2022] Open
Abstract
Infections caused by Schistosoma haematobium and Opisthorchis viverrini are classified as carcinogenic. Although carcinogenesis might be a multifactorial process, it has been postulated that these helminth produce/excrete oxysterols and estrogen-like metabolites that might act as initiators of their infection-associated carcinogenesis. Current treatment and control of these infections rely on a single drug, praziquantel, that mainly targets the parasites and not the pathologies related to the infection including cancer. Thus, there is a need to search for novel therapeutic alternatives that might include combinations of drugs and drug repurposing. Based on these concepts, we propose a novel therapeutic strategy that combines drugs with molecule antioxidants. We evaluate the efficacy of a novel therapeutic strategy to prevent the formation of putative carcinogenic metabolites precursors and DNA adducts. Firstly, we used a methodology previously established to synthesize metabolites precursors and DNA adducts in the presence of CYP450. Then, we evaluated the inhibition of their formation induced by drugs and antioxidants alone or in combination. Drugs and resveratrol alone did not show a significant inhibitory effect while N-acetylcysteine inhibited the formation of most metabolite precursors and DNA adducts. Moreover, the combinations of classical drugs with antioxidants were more effective rather than compounds alone. This strategy might be a valuable tool to prevent the initiation of helminth infection-associated carcinogenesis.
Collapse
Affiliation(s)
- Maria João Gouveia
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (M.J.G.); (V.N.); (B.A.)
- Department of Molecular Pathology and Immnunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira Apartado 55142, 4051-401 Porto, Portugal
| | - Verónica Nogueira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (M.J.G.); (V.N.); (B.A.)
- Department of Molecular Pathology and Immnunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Bruno Araújo
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (M.J.G.); (V.N.); (B.A.)
- Department of Molecular Pathology and Immnunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Fátima Gärtner
- Department of Molecular Pathology and Immnunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, university of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Nuno Vale
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (M.J.G.); (V.N.); (B.A.)
- Department of Molecular Pathology and Immnunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, university of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| |
Collapse
|
34
|
Affiliation(s)
- Sidhant Jain
- Department of Zoology, University of Delhi, North Campus, New Delhi, India
| | - Madhumita Sengupta
- Department of Zoology, University of Delhi, North Campus, New Delhi, India
| | - Pooja Jain
- Department of Obstetrics and Gynaecology, Bhagwati Hospital, New Delhi, India
| |
Collapse
|
35
|
Differential responses of epithelial cells from urinary and biliary tract to eggs of Schistosoma haematobium and S. mansoni. Sci Rep 2019; 9:10731. [PMID: 31341177 PMCID: PMC6656753 DOI: 10.1038/s41598-019-46917-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/28/2019] [Indexed: 01/09/2023] Open
Abstract
Chronic urogenital schistosomiasis can lead to squamous cell carcinoma of the bladder. The International Agency for Research on Cancer classifies the infection with S. haematobium as a group 1 carcinogen, a definitive cause of cancer. By contrast, hepatointestinal schistosomiasis due to the chronic infection with S. mansoni or S. japonicum associated with liver periportal fibrosis, does not apparently lead to malignancy. The effects of culturing human epithelial cells, HCV29, established from normal urothelium, and H69, established from cholangiocytes, in the presence of S. haematobium or S. mansoni eggs were investigated. Cell growth of cells co-cultured with schistosome eggs was monitored in real time, and gene expression analysis of oncogenesis, epithelial to mesenchymal transition and apoptosis pathways was undertaken. Schistosome eggs promoted proliferation of the urothelial cells but inhibited growth of cholangiocytes. In addition, the tumor suppressor P53 pathway was significantly downregulated when exposed to schistosome eggs, and downregulation of estrogen receptor was predicted in urothelial cells exposed only to S. haematobium eggs. Overall, cell proliferative responses were influenced by both the tissue origin of the epithelial cells and the schistosome species.
Collapse
|
36
|
Sotillo J, Pearson MS, Becker L, Mekonnen GG, Amoah AS, van Dam G, Corstjens PLAM, Murray J, Mduluza T, Mutapi F, Loukas A. In-depth proteomic characterization of Schistosoma haematobium: Towards the development of new tools for elimination. PLoS Negl Trop Dis 2019; 13:e0007362. [PMID: 31091291 PMCID: PMC6538189 DOI: 10.1371/journal.pntd.0007362] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/28/2019] [Accepted: 04/05/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Schistosomiasis is a neglected disease affecting hundreds of millions worldwide. Of the three main species affecting humans, Schistosoma haematobium is the most common, and is the leading cause of urogenital schistosomiasis. S. haematobium infection can cause different urogenital clinical complications, particularly in the bladder, and furthermore, this parasite has been strongly linked with squamous cell carcinoma. A comprehensive analysis of the molecular composition of its different proteomes will contribute to developing new tools against this devastating disease. METHODS AND FINDINGS By combining a comprehensive protein fractionation approach consisting of OFFGEL electrophoresis with high-throughput mass spectrometry, we have performed the first in-depth characterisation of the different discrete proteomes of S. haematobium that are predicted to interact with human host tissues, including the secreted and tegumental proteomes of adult flukes and secreted and soluble egg proteomes. A total of 662, 239, 210 and 138 proteins were found in the adult tegument, adult secreted, soluble egg and secreted egg proteomes, respectively. In addition, we probed these distinct proteomes with urine to assess urinary antibody responses from naturally infected human subjects with different infection intensities, and identified adult fluke secreted and tegument extracts as being the best predictors of infection. CONCLUSION We provide a comprehensive dataset of proteins from the adult and egg stages of S. haematobium and highlight their utility as diagnostic markers of infection intensity. Protein composition was markedly different between the different extracts, highlighting the distinct subsets of proteins that different development stages present in their different niches. Furthermore, we have identified adult fluke ES and tegument extracts as best predictors of infection using urine antibodies of naturally infected people. This study provides the first steps towards the development of novel tools to control this important neglected tropical disease.
Collapse
Affiliation(s)
- Javier Sotillo
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
- Laboratorio de Referencia en Parasitología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Mark S. Pearson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Luke Becker
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Gebeyaw G. Mekonnen
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Abena S. Amoah
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Govert van Dam
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul L. A. M. Corstjens
- Department of Molecular Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Janice Murray
- Institute of Immunology & Infection Research, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, King's Buildings, Edinburgh, United Kingdom
| | - Takafira Mduluza
- Biochemistry Department, University of Zimbabwe, Mount Pleasant, Harare, Zimbabwe
- TIBA Partnership, NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Zimbabwe
| | - Francisca Mutapi
- Institute of Immunology & Infection Research, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, King's Buildings, Edinburgh, United Kingdom
- TIBA Partnership, NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Zimbabwe
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| |
Collapse
|
37
|
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: 8.2] [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
|
38
|
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: 0.5] [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
|
39
|
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
|
40
|
Liu M, Veryser C, Lu JG, Wenseleers T, De Borggraeve WM, Jiang ZH, Luyten W. Bioassay-guided isolation of active substances from Semen Torreyae identifies two new anthelmintic compounds with novel mechanism of action. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:421-428. [PMID: 29933012 DOI: 10.1016/j.jep.2018.06.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/13/2018] [Accepted: 06/18/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Semen Torreyae, the seeds of Torreya grandis Fortune ex Lindley (Cephalotaxaceae) is a well-known traditional Chinese medicinal plant recorded in the Chinese Pharmacopeia (2010 version). It is widely used for treating intestinal parasites in China, owing to its desirable efficacy and safety. However, the anthelmintic compounds in Semen Torreyae have not yet been identified. AIM OF THE STUDY This study aims to identify the compounds active against helminths from Semen Torreyae. In addition, we tested whether C. elegans strains resistant to currently-used anthelmintic drugs showed cross-resistance to these compounds. METHODS A bioassay-guided isolation of anthelmintic compounds from Semen Torreyae was performed using a Caenorhabditis elegans (C. elegans) testing model. The structures of active compounds were elucidated by a combination of GC-MS, high resolution MS, and NMR. The median-effect method was employed to generate a combination index (CI) to evaluate the synergistic effect of the anthelmintic compounds. A panel of C. elegans mutant strains resistant against the major anthelmintic drug classes was used to study the cross-resistance to currently-used anthelmintic drugs. A panel of transient receptor potential (TRP) channel mutant strains was also tested to explore the possible mechanisms of action of the anthelmintic compounds. RESULTS The bioassay-guided isolation led to two active compounds, i.e. galangal acetate (IC50: 58.5 ± 8.9 μM) and miogadial (IC50: 25.1 ± 5.4 μM). The combination of galangal acetate and miogadial resulted in a synergistic effect at IC50, IC70, and IC90 levels (CIs < 1). Galangal acetate and miogadial demonstrated similar activity against drug-resistant C. elegans strains compared to the wild-type strain. In addition, none of the TRP mutants was significantly resistant to galangal acetate or miogadial compared to wild type worms. CONCLUSIONS We identified the bioactive compounds from Semen Torreyae responsible for its anthelmintic activity: galangal acetate and miogadial. The two anthelmintic compounds demonstrated a synergistic effect against C. elegans. Galangal acetate and miogadial are unlikely to act on the targets of currently-used anthelmintics (ivermectin, levamisole, benomyl and aldicarb), and an action on TRP channels appears to be ruled out as well. In summary, galangal acetate and miogadial are promising anthelmintic hits worth further investigation.
Collapse
Affiliation(s)
- Maoxuan Liu
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Naamsestraat 59, box 2465, 3000 Leuven, Belgium; Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, box 921, 3000 Leuven, Belgium.
| | - Cedrick Veryser
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven, Celestijnenlaan 200F, box 2404, 3000 Leuven, Belgium
| | - Jing-Guang Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Tom Wenseleers
- Department of Ecology, Evolution and Biodiversity Conservation, KU Leuven, Naamsestraat 59, box 2466, 3000 Leuven, Belgium
| | - Wim M De Borggraeve
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven, Celestijnenlaan 200F, box 2404, 3000 Leuven, Belgium
| | - Zhi-Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Walter Luyten
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Naamsestraat 59, box 2465, 3000 Leuven, Belgium
| |
Collapse
|
41
|
Liu M, Kipanga P, Mai AH, Dhondt I, Braeckman BP, De Borggraeve W, Luyten W. Bioassay-guided isolation of three anthelmintic compounds from Warburgia ugandensis Sprague subspecies ugandensis, and the mechanism of action of polygodial. Int J Parasitol 2018; 48:833-844. [PMID: 30031002 DOI: 10.1016/j.ijpara.2017.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 11/27/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022]
Abstract
Parasitic helminths continue to pose problems in human and veterinary medicine, as well as in agriculture. Resistance to current anthelmintics has prompted the search for new drugs. Anthelmintic metabolites from medicinal plants could be good anthelmintic drug candidates. However, the compounds active against nematodes have not been identified in most medicinal plants with anthelmintic activity. In this study, we aimed to identify the active compounds against helminths in Warburgia ugandensis Sprague subspecies ugandensis (Canellaceae) and study the underlying mechanism of action. A bioassay-guided isolation of anthelmintic compounds from the plant was performed using a Caenorhabditis elegans (C. elegans) test model with a WMicrotracker instrument to monitor motility. Three active compounds were purified and identified by nuclear magnetic resonance and high resolution MS: warburganal (IC50: 28.2 ± 8.6 μM), polygodial (IC50: 13.1 ± 5.3 μM) and alpha-linolenic acid (ALA, IC50: 70.1 ± 17.5 μM). A checkerboard assay for warburganal and ALA as well as polygodial and ALA showed a fractional inhibitory concentration index of 0.41 and 0.37, respectively, suggesting that polygodial and ALA, as well as warburganal and ALA, have a synergistic effect against nematodes. A preliminary structure-activity relationship study for polygodial showed that the α,β-unsaturated 1,4-dialdehyde structural motif is essential for the potent activity. None of a panel of C. elegans mutant strains, resistant against major anthelmintic drug classes, showed significant resistance to polygodial, implying that polygodial may block C. elegans motility through a mechanism which differs from that of currently marketed drugs. Further measurements showed that polygodial inhibits mitochondrial ATP synthesis of C. elegans in a dose-dependent manner (IC50: 1.8 ± 1.0 μM). Therefore, we believe that the underlying mechanism of action of polygodial is probably inhibition of mitochondrial ATP synthesis. In conclusion, polygodial could be a promising anthelmintic drug candidate worth considering for further development.
Collapse
Affiliation(s)
- Maoxuan Liu
- Faculty of Pharmaceutical Sciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Naamsestraat 59, Box 2465, 3000 Leuven, Belgium.
| | - Purity Kipanga
- Faculty of Pharmaceutical Sciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Anh Hung Mai
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven, Celestijnenlaan 200F, Box 2404, 3000 Leuven, Belgium
| | - Ineke Dhondt
- Department of Biology, Faculty of Sciences, Ghent University, K.L. Ledeganckstraat 35, 9000 Gent, Belgium
| | - Bart P Braeckman
- Department of Biology, Faculty of Sciences, Ghent University, K.L. Ledeganckstraat 35, 9000 Gent, Belgium
| | - Wim De Borggraeve
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven, Celestijnenlaan 200F, Box 2404, 3000 Leuven, Belgium
| | - Walter Luyten
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Naamsestraat 59, Box 2465, 3000 Leuven, Belgium
| |
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: 1.4] [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
|
Scholte LLS, Pascoal-Xavier MA, Nahum LA. Helminths and Cancers From the Evolutionary Perspective. Front Med (Lausanne) 2018; 5:90. [PMID: 29713629 PMCID: PMC5911458 DOI: 10.3389/fmed.2018.00090] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/22/2018] [Indexed: 01/20/2023] Open
Abstract
Helminths include free-living and parasitic Platyhelminthes and Nematoda which infect millions of people worldwide. Some Platyhelminthes species of blood flukes (Schistosoma haematobium, Schistosoma japonicum, and Schistosoma mansoni) and liver flukes (Clonorchis sinensis and Opisthorchis viverrini) are known to be involved in human cancers. Other helminths are likely to be carcinogenic. Our main goals are to summarize the current knowledge of human cancers caused by Platyhelminthes, point out some helminth and human biomarkers identified so far, and highlight the potential contributions of phylogenetics and molecular evolution to cancer research. Human cancers caused by helminth infection include cholangiocarcinoma, colorectal hepatocellular carcinoma, squamous cell carcinoma, and urinary bladder cancer. Chronic inflammation is proposed as a common pathway for cancer initiation and development. Furthermore, different bacteria present in gastric, colorectal, and urogenital microbiomes might be responsible for enlarging inflammatory and fibrotic responses in cancers. Studies have suggested that different biomarkers are involved in helminth infection and human cancer development; although, the detailed mechanisms remain under debate. Different helminth proteins have been studied by different approaches. However, their evolutionary relationships remain unsolved. Here, we illustrate the strengths of homology identification and function prediction of uncharacterized proteins from genome sequencing projects based on an evolutionary framework. Together, these approaches may help identifying new biomarkers for disease diagnostics and intervention measures. This work has potential applications in the field of phylomedicine (evolutionary medicine) and may contribute to parasite and cancer research.
Collapse
Affiliation(s)
- Larissa L. S. Scholte
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
- Vice-Presidência de Pesquisa e Coleções Biológicas, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Marcelo A. Pascoal-Xavier
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
- Departamento de Anatomia Patológica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laila A. Nahum
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
- Faculdade Promove de Tecnologia, Belo Horizonte, Brazil
| |
Collapse
|
44
|
|
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: 2.1] [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: 3.6] [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: 52] [Impact Index Per Article: 7.4] [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
|
Extracellular vesicles as a target for the development of anti-helminth vaccines. Emerg Top Life Sci 2017; 1:659-665. [PMID: 33525849 DOI: 10.1042/etls20170095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/12/2017] [Accepted: 11/14/2017] [Indexed: 01/29/2023]
Abstract
There is a rapidly growing body of evidence that production of extracellular vesicles (EVs) is a universal feature of cellular life. More recently, EVs have been identified in a broad range of both unicellular and multicellular parasites where they play roles in parasite-parasite intercommunication as well as parasite-host interactions. Parasitic helminth-derived EVs traverse host target cell membranes whereupon they offload their molecular cargo - proteins, lipids, and genetic information such as mRNAs and miRNAs - which are thought to hijack the target cell and modulate its gene expression to promote parasite survival. As such, EVs represent a novel mechanism of intercellular communication that could be targeted for vaccine-mediated interruption, given the abundance of surface antigens expressed on helminth EVs, and the ability of antibodies to block their uptake by target cells. In this Perspective article, we review recent developments in the field of helminth-derived EVs and highlight their roles in helminth vaccine discovery and development.
Collapse
|
49
|
|