1
|
Wu F, Chen Y, Chen X, Tong D, Zhou J, Du Z, Yao C, Yang Y, Du A, Ma G. Nematode serine protease inhibitor SPI-I8 negatively regulates host NF-κB signalling by hijacking MKRN1-mediated polyubiquitination of RACK1. Commun Biol 2025; 8:356. [PMID: 40032982 PMCID: PMC11876351 DOI: 10.1038/s42003-025-07803-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
Parasitic roundworms are remarkable for their ability to manipulate host immune systems and ameliorate inflammatory diseases. Although much is known about the nature of nematode effectors in immune modulation, little is known about the action mode of these molecules. Here, we report that a serine protease inhibitor SPI-I8 in the extracellular vesicles of blood-feeding nematodes like Ancylostoma ceylanicum, Haemonchus contortus and Nippostrongylus brasiliensis, effectively halts excessive inflammatory responses in vitro and in vivo. We demonstrate that H. contortus SPI-I8 promotes the role of a negative regulator of RACK1 and enhances the effects of RACK1 on tumor necrosis factor (TNF)-α-IκB kinases (IKKs)-nuclear factor kappa beta (NF-κB) axis in mammalian cells, by hijacking E3 ubiquitin protein ligase MKRN1-mediated polyubiquitination of RACK1. Administration of recombinant N. brasiliensis SPI-I8 effectively protects mice from dextran sulfate sodium (DSS)-induced colitis and lipopolysaccharide (LPS)-induced sepsis. Considering the structural and functional conservation of SPI-I8s among Strongylida nematodes and the conservation of interactive mediators (i.e., MKRN1 and RACK1) among mammals, our findings provide insights into the host-parasite interface where parasitic roundworms secret molecules to suppress host inflammatory responses. Harnessing these findings should underpin the exploitation of nematode's immunomodulators to relief excessive inflammation associated diseases in animals and humans.
Collapse
Affiliation(s)
- Fei Wu
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- College of Veterinary Medicine, Anhui Agricultural University, Hefei, China
| | - Yanqiong Chen
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xueqiu Chen
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Danni Tong
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingru Zhou
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- MOE Frontier Science Center for Brain and Brain-machine integration, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhendong Du
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chaoqun Yao
- Ross University School of Veterinary Medicine and One Health Center for Zoonoses and Tropical Veterinary Medicine, Ross University School of Veterinary Medicine, Basseterre, St. Kitts, Trinidad and Tobago
| | - Yi Yang
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Aifang Du
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Guangxu Ma
- Institute of Preventive Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- ZJU-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech Park, Xinchang, China.
| |
Collapse
|
2
|
Pillay R, Naidoo P, Mkhize-Kwitshana ZL. Exploring microRNA-Mediated Immune Responses to Soil-Transmitted Helminth and Herpes Simplex Virus Type 2 Co-Infections. Diseases 2025; 13:6. [PMID: 39851470 PMCID: PMC11765296 DOI: 10.3390/diseases13010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/24/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Over the last two decades, the field of microRNA (miRNA) research has grown significantly. MiRNAs are a class of short, single-stranded, non-coding RNAs that regulate gene expression post-transcriptionally. Thereby, miRNAs regulate various essential biological processes including immunity. Dysregulated miRNAs are associated with various infectious and non-infectious diseases. Recently co-infection with soil-transmitted helminths (STHs) and herpes simplex virus type 2 (HSV-2) has become a focus of study. Both pathogens can profoundly influence host immunity, particularly in under-resourced and co-endemic regions. It is well known that STHs induce immunomodulatory responses that have bystander effects on unrelated conditions. Typically, STHs induce T-helper 2 (Th2) and immunomodulatory responses, which may dampen the proinflammatory T-helper 1 (Th1) immune responses triggered by HSV-2. However, the extent to which STH co-infection influences the host immune response to HSV-2 is not well understood. Moreover, little is known about how miRNAs shape the immune response to STH/HSV-2 co-infection. In this article, we explore the potential influence that STH co-infection may have on host immunity to HSV-2. Because STH and HSV-2 infections are widespread and disproportionately affect vulnerable and impoverished countries, it is important to consider how STHs may impact HSV-2 immunity. Specifically, we explore how miRNAs contribute to both helminth and HSV-2 infections and discuss how miRNAs may mediate STH/HSV-2 co-infections. Insight into miRNA-mediated immune responses may further improve our understanding of the potential impact of STH/HSV-2 co-infections.
Collapse
Affiliation(s)
- Roxanne Pillay
- Department of Biomedical Sciences, Faculty of Natural Sciences, Mangosuthu University of Technology, Umlazi, Durban 4031, South Africa
- Department of Medical Microbiology, College of Health Sciences, School of Laboratory Medicine & Medical Sciences, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| | - Pragalathan Naidoo
- Department of Medical Microbiology, College of Health Sciences, School of Laboratory Medicine & Medical Sciences, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| | - Zilungile L. Mkhize-Kwitshana
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
- Biomedical Sciences Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Florida Campus, Johannesburg 1710, South Africa
| |
Collapse
|
3
|
Pillay R, Naidoo P, Duma Z, Bhengu KN, Mpaka-Mbatha MN, Nembe-Mafa N, Mkhize-Kwitshana ZL. Potential Interactions Between Soil-Transmitted Helminths and Herpes Simplex Virus Type II: Implications for Sexual and Reproductive Health in Sub-Saharan African. BIOLOGY 2024; 13:1050. [PMID: 39765717 PMCID: PMC11673149 DOI: 10.3390/biology13121050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/24/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025]
Abstract
Sub-Saharan Africa (SSA) bears a disproportionate and overlapping burden of soil-transmitted helminths (STHs) and sexually transmitted viral infections. An estimated 232 million pre-school and school-aged children in SSA are vulnerable to STH infections. Together with this, SSA has a high prevalence of herpes simplex virus type II (HSV-2), the primary cause of genital herpes. Studies have examined the immunological interactions between STHs and human immunodeficiency virus and human papillomavirus during co-infections. However, epidemiological and immunological studies on STH-HSV-2 co-infections are lacking, therefore their impact on sexual and reproductive health is not fully understood. STH-driven Th2 immune responses are known to downregulate Th1/Th17 immune responses. Therefore, during STH-HSV-2 co-infections, STH-driven immune responses may alter host immunity to HSV-2 and HSV-2 pathology. Herein, we provide an overview of the burden of STH and HSV-2 infections in SSA, and host immune responses to STH and HSV-2 infections. Further, we emphasize the relevance and urgent need for (i) focused research into the interactions between these important pathogens, and (ii) integrated approaches to improve the clinical detection and management of STH-HSV-2 co-infections in SSA.
Collapse
Affiliation(s)
- Roxanne Pillay
- Department of Biomedical Sciences, Faculty of Natural Sciences, Mangosuthu University of Technology, Umlazi, Durban 4031, South Africa
- Department of Medical Microbiology, College of Health Sciences, School of Laboratory Medicine & Medical Sciences, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| | - Pragalathan Naidoo
- Department of Medical Microbiology, College of Health Sciences, School of Laboratory Medicine & Medical Sciences, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| | - Zamathombeni Duma
- Department of Medical Microbiology, College of Health Sciences, School of Laboratory Medicine & Medical Sciences, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| | - Khethiwe N. Bhengu
- Department of Biomedical Sciences, Faculty of Natural Sciences, Mangosuthu University of Technology, Umlazi, Durban 4031, South Africa
- Department of Medical Microbiology, College of Health Sciences, School of Laboratory Medicine & Medical Sciences, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| | - Miranda N. Mpaka-Mbatha
- Department of Medical Microbiology, College of Health Sciences, School of Laboratory Medicine & Medical Sciences, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| | - Nomzamo Nembe-Mafa
- Department of Medical Microbiology, College of Health Sciences, School of Laboratory Medicine & Medical Sciences, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| | - Zilungile L. Mkhize-Kwitshana
- Department of Medical Microbiology, College of Health Sciences, School of Laboratory Medicine & Medical Sciences, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development, South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
- Department of Biomedical Sciences, University of Johannesburg, Doorfontein Campus, Johannesburg 2028, South Africa
- Biomedical Sciences Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Florida Campus, Johannesburg 1710, South Africa
| |
Collapse
|
4
|
Mugo RM, Rausch S, Musimbi ZD, Strube C, Raulf MK, Landt O, Gichuki PM, Ebner F, Mwacharo J, Odiere MR, Ndungu FM, Njomo DW, Hartmann S. Evaluation of copromicroscopy, multiplex-qPCR and antibody serology for monitoring of human ascariasis in endemic settings. PLoS Negl Trop Dis 2024; 18:e0012279. [PMID: 38889190 PMCID: PMC11216587 DOI: 10.1371/journal.pntd.0012279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 07/01/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The standard diagnosis of Ascaris lumbricoides and other soil-transmitted helminth (STH) infections relies on the detection of worm eggs by copromicroscopy. However, this method is dependent on worm patency and shows only limited accuracy in low-intensity infection settings. We aimed to decipher the diagnostic accuracy of different antibodies using various Ascaris antigens in reference to copromicroscopy and quantitative PCR (qPCR), four months after national STH preventative chemotherapy among school children in western Kenya. METHODOLOGY STH infection status of 390 school children was evaluated via copromicroscopy (Kato-Katz and mini-FLOTAC) and qPCR. In parallel, Ascaris-specific antibody profiles against larval and adult worm lysates, and adult worm excretory-secretory (ES) products were determined by enzyme-linked immunosorbent assay. Antibody cross-reactivity was evaluated using the closely related zoonotic roundworm species Toxocara cati and Toxocara canis. The diagnostic accuracy of each antibody was evaluated using receiver operating curve analysis and the correspondent area under the curve (AUC). PRINCIPAL FINDINGS Ascaris was the predominant helminth infection with an overall prevalence of 14.9% (58/390). The sensitivity of mini-FLOTAC and Kato-Katz for Ascaris diagnosis reached only 53.5% and 63.8%, respectively compared to qPCR. Although being more sensitive, qPCR values correlated with microscopic egg counts (R = -0.71, P<0.001), in contrast to antibody levels. Strikingly, IgG antibodies recognizing the ES products of adult Ascaris worms reliably diagnosed active Ascaris infection as determined by qPCR and microscopy, with IgG1 displaying the highest accuracy (AUC = 0.83, 95% CI: 0.75-0.91). CONCLUSION IgG1 antibody responses against adult Ascaris-ES products hold a promising potential for complementing the standard fecal and molecular techniques employed for monitoring Ascaris infections. This is of particular importance in the context of deworming programs as the antibody diagnostic accuracy was independent of egg counts.
Collapse
Affiliation(s)
- Robert M. Mugo
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Rausch
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Zaneta D. Musimbi
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Christina Strube
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Marie-Kristin Raulf
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Paul M. Gichuki
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research, Nairobi, Kenya
| | - Friederike Ebner
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
- Department of Molecular Life Sciences, School of Life Sciences, Technical University of Munich, Munich, Germany
| | | | - Maurice R. Odiere
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Francis M. Ndungu
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Doris W. Njomo
- Eastern and Southern Africa Centre of International Parasite Control, Kenya Medical Research, Nairobi, Kenya
| | - Susanne Hartmann
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
5
|
Getzlaff JM, Fulghum GH. Enterobius vermicularis (Pinworm) Appendicitis: The Real Vermiform Appendix. Mil Med 2024; 189:e894-e896. [PMID: 37674268 DOI: 10.1093/milmed/usad357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/05/2023] [Accepted: 08/30/2023] [Indexed: 09/08/2023] Open
Abstract
Enterobius vermicularis is the most common helminthic infection in the United States, infecting an estimated 40 million persons. A rare complication of E. vermicularis infestation is appendicitis. The diagnosis must be considered during pathologic examination to ensure appropriate treatment with anti-helminthic therapy.
Collapse
Affiliation(s)
- Jared M Getzlaff
- General Surgery Residency Program, Keesler AFB Medical Center, Biloxi, MS 39534, USA
| | - George H Fulghum
- General Surgery Residency Program, Keesler AFB Medical Center, Biloxi, MS 39534, USA
| |
Collapse
|
6
|
Ranasinghe S, Aspinall S, Beynon A, Ash A, Lymbery A. Traditional medicinal plants in the treatment of gastrointestinal parasites in humans: A systematic review and meta-analysis of clinical and experimental evidence. Phytother Res 2023; 37:3675-3687. [PMID: 37230485 DOI: 10.1002/ptr.7895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/22/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023]
Abstract
Gastrointestinal (GI) parasites cause significant morbidity and mortality worldwide. The use of conventional antiparasitic drugs is often inhibited due to limited availability, side effects or parasite resistance. Medicinal plants can be used as alternatives or adjuncts to current antiparasitic therapies. This systematic review and meta-analysis aimed to critically synthesise the literature on the efficacy of different plants and plant compounds against common human GI parasites and their toxicity profiles. Searches were conducted from inception to September 2021. Of 5393 screened articles, 162 were included in the qualitative synthesis (159 experimental studies and three randomised control trials [RCTs]), and three articles were included in meta-analyses. A total of 507 plant species belonging to 126 families were tested against different parasites, and most of these (78.4%) evaluated antiparasitic efficacy in vitro. A total of 91 plant species and 34 compounds were reported as having significant in vitro efficacy against parasites. Only a few plants (n = 57) were evaluated for their toxicity before testing their antiparasitic effects. The meta-analyses revealed strong evidence of the effectiveness of Lepidium virginicum L. against Entamoeba histolytica with a pooled mean IC50 of 198.63 μg/mL (95% CI 155.54-241.72). We present summary tables and various recommendations to direct future research.
Collapse
Affiliation(s)
- Sandamalie Ranasinghe
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| | - Sasha Aspinall
- School of Allied Health, College of Health and Education, Murdoch University, Perth, Western Australia, Australia
| | - Amber Beynon
- Department of Chiropractic, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie Park, New South Wales, Australia
| | - Amanda Ash
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| | - Alan Lymbery
- Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
7
|
Ranasinghe S, Armson A, Lymbery AJ, Zahedi A, Ash A. Medicinal plants as a source of antiparasitics: an overview of experimental studies. Pathog Glob Health 2023; 117:535-553. [PMID: 36805662 PMCID: PMC10392325 DOI: 10.1080/20477724.2023.2179454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
Despite advances in modern human and veterinary medicine, gastrointestinal (GI) parasitic infections remain a significant health issue worldwide, mainly in developing countries. Increasing evidence of the multi-drug resistance of these parasites and the side effects of currently available synthetic drugs have led to increased research on alternative medicines to treat parasitic infections. The exploration of potential botanical antiparasitics, which are inexpensive and abundant, may be a promising alternative in this context. This study summarizes the in vitro/in vivo antiparasitic efficacy of different medicinal plants and their components against GI parasites. Published literature from 1990-2020 was retrieved from Google Scholar, Web of Science, PubMed and Scopus. A total of 68 plant species belonging to 32 families have been evaluated as antiparasitic agents against GI parasites worldwide. The majority of studies (70%) were conducted in vitro. Most plants were from the Fabaceae family (53%, n = 18). Methanol (37%, n = 35) was the most used solvent. Leaf (22%, n = 16) was the most used plant part, followed by seed and rhizome (each 12%, n = 9). These studies suggest that herbal medicines hold a great scope for new drug discoveries against parasitic diseases and that the derivatives of these plants are useful structures for drug synthesis and bioactivity optimization.
Collapse
Affiliation(s)
- Sandamalie Ranasinghe
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| | - Anthony Armson
- Exercise Science and Chiropractic, College of Science, Health, Engineering and Education, Murdoch University, Perth, Western Australia, Australia
| | - Alan J. Lymbery
- Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| | - Alireza Zahedi
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| | - Amanda Ash
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
8
|
Lebu S, Kibone W, Muoghalu CC, Ochaya S, Salzberg A, Bongomin F, Manga M. Soil-transmitted helminths: A critical review of the impact of co-infections and implications for control and elimination. PLoS Negl Trop Dis 2023; 17:e0011496. [PMID: 37561673 PMCID: PMC10414660 DOI: 10.1371/journal.pntd.0011496] [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: 08/12/2023] Open
Abstract
Researchers have raised the possibility that soil-transmitted helminth (STH) infections might modify the host's immune response against other systemic infections. STH infections can alter the immune response towards type 2 immunity that could then affect the likelihood and severity of other illnesses. However, the importance of co-infections is not completely understood, and the impact and direction of their effects vary considerably by infection. This review synthesizes evidence regarding the relevance of STH co-infections, the potential mechanisms that explain their effects, and how they might affect control and elimination efforts. According to the literature reviewed, there are both positive and negative effects associated with STH infections on other diseases such as malaria, human immunodeficiency virus (HIV), tuberculosis, gestational anemia, pediatric anemia, neglected tropical diseases (NTDs) like lymphatic filariasis, onchocerciasis, schistosomiasis, and trachoma, as well as Coronavirus Disease 2019 (COVID-19) and human papillomavirus (HPV). Studies typically describe how STHs can affect the immune system and promote increased susceptibility, survival, and persistence of the infection in the host by causing a TH2-dominated immune response. The co-infection of STH with other diseases has important implications for the development of treatment and control strategies. Eliminating parasites from a human host can be more challenging because the TH2-dominated immune response induced by STH infection can suppress the TH1 immune response required to control other infections, resulting in an increased pathogen load and more severe disease. Preventive chemotherapy and treatment are currently the most common approaches used for the control of STH infections, but these approaches alone may not be adequate to achieve elimination goals. Based on the conclusions drawn from this review, integrated approaches that combine drug administration with water, sanitation and hygiene (WASH) interventions, hygiene education, community engagement, and vaccines are most likely to succeed in interrupting the transmission of STH co-infections. Gaining a better understanding of the behavior and relevance of STH co-infections in the context of elimination efforts is an important intermediate step toward reducing the associated burden of disease.
Collapse
Affiliation(s)
- Sarah Lebu
- The Water Institute at UNC, Department of Environmental Sciences and Engineering, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Winnie Kibone
- School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Chimdi C. Muoghalu
- The Water Institute at UNC, Department of Environmental Sciences and Engineering, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Stephen Ochaya
- Department of Microbiology and Immunology, Faculty of Medicine, Gulu University, Gulu, Uganda
- Department of Biology, Faculty of Science, Gulu University, Gulu, Uganda
- Department of Clinical Pathology, Uppsala Academic Hospital, Uppsala, Sweden
| | - Aaron Salzberg
- The Water Institute at UNC, Department of Environmental Sciences and Engineering, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Felix Bongomin
- Department of Microbiology and Immunology, Faculty of Medicine, Gulu University, Gulu, Uganda
| | - Musa Manga
- The Water Institute at UNC, Department of Environmental Sciences and Engineering, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
9
|
Zainab T, Khatoon S, Sahreen A, Saifullah MK. Assessment of antioxidant enzymes during the development of the digenetic trematode parasite Gastrothylax crumenifer, infecting the rumen of Indian water buffalo, Bubalus bubalis. J Parasit Dis 2023; 47:271-279. [PMID: 37193500 PMCID: PMC10182214 DOI: 10.1007/s12639-023-01564-z] [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: 11/13/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023] Open
Abstract
The presence of antioxidant enzymes in helminth parasites is well known. These enzymes help the parasites to survive in their hosts by detoxifying host-generated reactive oxygen species (ROS). The literature survey reveals that most of the studies related to antioxidant enzymes in helminth parasites are restricted to the adult stage while the larval stages are neglected. The present investigation is designed to evaluate the level of antioxidant enzymes in the adult and larval stages of rumen-infecting paramphistome parasites, Gastrothylax crumenifer. The larval stages include 0-day eggs, 4-day eggs, and eggs containing mature miracidia, cercariae, and metacercariae. Antioxidant enzyme assays were performed using standard assay protocols. Our findings revealed an increasing pattern in the level of Glutathione-S-Transferase (GST), Superoxide Dismutase (SOD), Glutathione Reductase (GR), and Glutathione Peroxidase (GPx) antioxidant enzymes during the development from 0-day eggs to the adult stage. Overall analysis shows that adult worms have higher antioxidant enzyme activity as compared to the larval stages, indicating that adult flukes are more adapted to oxidative stress. It can be concluded that the miracidia, cercarial, and metacercarial developmental stages of G. crumenifer possess a considerable level of antioxidant enzymes suitable to overcome the oxidative stress encountered during the development and help them in the completion of the life cycle and survival in the definitive host.
Collapse
Affiliation(s)
- Tahmina Zainab
- Parasite Biochemistry and Immunology Laboratory, Section of Parasitology, Department of Zoology, Aligarh Muslim University, Aligarh, UP 202002 India
| | - Sabiha Khatoon
- Department of Cell Biology and Biochemistry, Texas Tech, University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430 USA
| | - Anam Sahreen
- Parasite Biochemistry and Immunology Laboratory, Section of Parasitology, Department of Zoology, Aligarh Muslim University, Aligarh, UP 202002 India
| | - Mohammad Khalid Saifullah
- Parasite Biochemistry and Immunology Laboratory, Section of Parasitology, Department of Zoology, Aligarh Muslim University, Aligarh, UP 202002 India
| |
Collapse
|
10
|
Oyesola O, Downie AE, Howard N, Barre RS, Kiwanuka K, Zaldana K, Chen YH, Menezes A, Lee SC, Devlin J, Mondragón-Palomino O, Souza COS, Herrmann C, Koralov S, Cadwell K, Graham AL, Loke P. Genetic and Environmental interactions contribute to immune variation in rewilded mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533121. [PMID: 36993484 PMCID: PMC10055251 DOI: 10.1101/2023.03.17.533121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
The relative and synergistic contributions of genetics and environment to inter-individual immune response variation remain unclear, despite its implications for understanding both evolutionary biology and medicine. Here, we quantify interactive effects of genotype and environment on immune traits by investigating three inbred mouse strains rewilded in an outdoor enclosure and infected with the parasite, Trichuris muris. Whereas cytokine response heterogeneity was primarily driven by genotype, cellular composition heterogeneity was shaped by interactions between genotype and environment. Notably, genetic differences under laboratory conditions can be decreased following rewilding, and variation in T cell markers are more driven by genetics, whereas B cell markers are driven more by environment. Importantly, variation in worm burden is associated with measures of immune variation, as well as genetics and environment. These results indicate that nonheritable influences interact with genetic factors to shape immune variation, with synergistic impacts on the deployment and evolution of defense mechanisms.
Collapse
Affiliation(s)
- Oyebola Oyesola
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health; Bethesda, MD, USA
| | - Alexander E. Downie
- Department of Ecology and Evolutionary Biology, Princeton University; Princeton, NJ, USA
| | - Nina Howard
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health; Bethesda, MD, USA
| | - Ramya S. Barre
- Department of Ecology and Evolutionary Biology, Princeton University; Princeton, NJ, USA
| | - Kasalina Kiwanuka
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health; Bethesda, MD, USA
| | - Kimberly Zaldana
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health; Bethesda, MD, USA
| | - Ying-Han Chen
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University, Grossman School of Medicine; New York, NY, USA
| | - Arthur Menezes
- Department of Ecology and Evolutionary Biology, Princeton University; Princeton, NJ, USA
| | - Soo Ching Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health; Bethesda, MD, USA
| | - Joseph Devlin
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University, Grossman School of Medicine; New York, NY, USA
| | - Octavio Mondragón-Palomino
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health; Bethesda, MD, USA
| | - Camila Oliveira Silva Souza
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health; Bethesda, MD, USA
| | - Christin Herrmann
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University, Grossman School of Medicine; New York, NY, USA
| | - Sergei Koralov
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University, Grossman School of Medicine; New York, NY, USA
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University, Grossman School of Medicine; New York, NY, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Andrea L. Graham
- Department of Ecology and Evolutionary Biology, Princeton University; Princeton, NJ, USA
- Santa Fe Institute, Santa Fe, NM, USA
| | - P’ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health; Bethesda, MD, USA
| |
Collapse
|
11
|
Parker W, Patel E, Jirků-Pomajbíková K, Laman JD. COVID-19 morbidity in lower versus higher income populations underscores the need to restore lost biodiversity of eukaryotic symbionts. iScience 2023; 26:106167. [PMID: 36785786 PMCID: PMC9908430 DOI: 10.1016/j.isci.2023.106167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
The avoidance of infectious disease by widespread use of 'systems hygiene', defined by hygiene-enhancing technology such as sewage systems, water treatment facilities, and secure food storage containers, has led to a dramatic decrease in symbiotic helminths and protists in high-income human populations. Over a half-century of research has revealed that this 'biota alteration' leads to altered immune function and a propensity for chronic inflammatory diseases, including allergic, autoimmune and neuropsychiatric disorders. A recent Ethiopian study (EClinicalMedicine 39: 101054), validating predictions made by several laboratories, found that symbiotic helminths and protists were associated with a reduced risk of severe COVID-19 (adjusted odds ratio = 0.35; p<0.0001). Thus, it is now apparent that 'biome reconstitution', defined as the artificial re-introduction of benign, symbiotic helminths or protists into the ecosystem of the human body, is important not only for alleviation of chronic immune disease, but likely also for pandemic preparedness.
Collapse
Affiliation(s)
| | | | - Kateřina Jirků-Pomajbíková
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 370 05 České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, 370 05 České Budějovice, Czech Republic
| | - Jon D. Laman
- Department of Pathology and Medical Biology, University Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
12
|
Dagenais M, Tritten L. Hidden in plain sight: How helminths manage to thrive in host blood. FRONTIERS IN PARASITOLOGY 2023; 2:1128299. [PMID: 39816845 PMCID: PMC11732017 DOI: 10.3389/fpara.2023.1128299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/27/2023] [Indexed: 01/18/2025]
Abstract
Parasitic helminths have evolved a plethora of elegant stratagems to regulate and evade the host immune system, contributing to their considerable persistence and longevity in their vertebrate hosts. Various mechanisms to achieve this state have been described, ranging from interfering with or actively modulating host immune responses to hiding from immune recognition. Because they damage surrounding vessels and disturb blood flow, blood-borne and blood-feeding parasites in particular must deal with much more than immune effector cells. Management of the host complement system and coagulation cascade, as well as the development of processes of hiding and masking, represent hallmarks of life in blood. Here we review recent findings on putative evasion strategies employed by blood-borne parasitic helminths, focusing on the interaction with and utilisation of host serum components by nematodes and trematodes.
Collapse
Affiliation(s)
- Maude Dagenais
- Institute of Parasitology, McGill University, Ste-Anne-de-Bellevue, QC, Canada
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Lucienne Tritten
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Roose S, Leta GT, Vlaminck J, Getachew B, Mekete K, Peelaers I, Geldhof P, Levecke B. Comparison of coproprevalence and seroprevalence to guide decision-making in national soil-transmitted helminthiasis control programs: Ethiopia as a case study. PLoS Negl Trop Dis 2022; 16:e0010824. [PMID: 36197895 PMCID: PMC9534397 DOI: 10.1371/journal.pntd.0010824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/16/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND WHO recommends periodical assessment of the prevalence of any soil-transmitted helminth (STH) infections to adapt the frequency of mass drug administration targeting STHs. Today, detection of eggs in stool smears (Kato-Katz thick smear) remains the diagnostic standard. However, stool examination (coprology) has important operational drawbacks and impedes integrated surveys of multiple neglected tropical diseases. Therefore, the aim of the present study was to assess the potential of applying serology instead of coprology in STH control program decision-making. METHODOLOGY An antibody-ELISA based on extract of Ascaris lung stage larvae (AsLungL3-ELISA) was applied in ongoing monitoring activities of the Ethiopian national control program against schistosomiasis and soil-transmitted helminthiasis. Blood and stool samples were collected from over 6,700 students (median age: 11) from 63 schools in 33 woredas (districts) across the country. Stool samples of two consecutive days were analyzed applying duplicate Kato-Katz thick smear. PRINCIPAL FINDINGS On woreda level, qualitative (seroprevalence) and quantitative (mean optical density ratio) serology results were highly correlated, and hence seroprevalence was chosen as parameter. For 85% of the woredas, prevalence based on serology was higher than those based on coprology. The results suggested cross-reactivity of the AsLungL3-ELISA with Trichuris. When extrapolating the WHO coproprevalence thresholds, there was a moderate agreement (weighted κ = 0.43) in program decision-making. Using the same threshold values would predominantly lead to a higher frequency of drug administration. SIGNIFICANCE This is the first time that serology for soil-transmitted helminthiasis is applied on such large scale, thereby embedded in a control program context. The results underscore that serology holds promise as a tool to monitor STH control programs. Further research should focus on the optimization of the diagnostic assay and the refinement of serology-specific program decision-making thresholds.
Collapse
Affiliation(s)
- Sara Roose
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
- * E-mail: (SR); (BL)
| | - Gemechu Tadesse Leta
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Johnny Vlaminck
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Birhanu Getachew
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Kalkidan Mekete
- Bacterial, Parasitic and Zoonotic Diseases Research Directorate, Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Iris Peelaers
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Peter Geldhof
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Bruno Levecke
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
- * E-mail: (SR); (BL)
| |
Collapse
|
14
|
Yeshi K, Ruscher R, Loukas A, Wangchuk P. Immunomodulatory and biological properties of helminth-derived small molecules: Potential applications in diagnostics and therapeutics. FRONTIERS IN PARASITOLOGY 2022; 1:984152. [PMID: 39816468 PMCID: PMC11731824 DOI: 10.3389/fpara.2022.984152] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2025]
Abstract
Parasitic helminths secrete and excrete a vast array of molecules known to help skew or suppress the host's immune response, thereby establishing a niche for sustained parasite maintenance. Indeed, the immunomodulatory potency of helminths is attributed mainly to excretory/secretory products (ESPs). The ESPs of helminths and the identified small molecules (SM) are reported to have diverse biological and pharmacological properties. The available literature reports only limited metabolites, and the identity of many metabolites remains unknown due to limitations in the identification protocols and helminth-specific compound libraries. Many metabolites are known to be involved in host-parasite interactions and pathogenicity. For example, fatty acids (e.g., stearic acid) detected in the infective stages of helminths are known to have a role in host interaction through facilitating successful penetration and migration inside the host. Moreover, excreted/secreted SM detected in helminth species are found to possess various biological properties, including anti-inflammatory activities, suggesting their potential in developing immunomodulatory drugs. For example, helminths-derived somatic tissue extracts and whole crude ESPs showed anti-inflammatory properties by inhibiting the secretion of proinflammatory cytokines from human peripheral blood mononuclear cells and suppressing the pathology in chemically-induced experimental mice model of colitis. Unlike bigger molecules like proteins, SM are ideal candidates for drug development since they are small structures, malleable, and lack immunogenicity. Future studies should strive toward identifying unknown SM and isolating the under-explored niche of helminth metabolites using the latest metabolomics technologies and associated software, which hold potential keys for finding new diagnostics and novel therapeutics.
Collapse
Affiliation(s)
- Karma Yeshi
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Cairns, QLD, Australia
| | | | | | | |
Collapse
|
15
|
Characterization of antigens of Enterobius vermicularis (pinworm) eggs. Sci Rep 2022; 12:14414. [PMID: 36002555 PMCID: PMC9402560 DOI: 10.1038/s41598-022-18303-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 08/09/2022] [Indexed: 11/08/2022] Open
Abstract
Enterobiasis (pinworm infection) caused by Enterobius vermicularis is a common parasitic infection prevalent worldwide especially in children. Infection is diagnosed by microscopic detection of E. vermicularis eggs on perianal swabs. This study aimed to characterize the antigens of E. vermicularis eggs as a preliminary step towards identifying diagnostic targets for detection in infected individuals. The study was conducted between October 2019 and February 2020, following approval from Ethics Review Committee of the Faculty of Medicine, University of Colombo (EC-19-034). E. vermicularis eggs were harvested from perianal swabs using acetone and purified with 1× PBS (pH 7.2). A portion of eggs was used for preparing antigen slides, while the rest were sonicated and vortexed with glass beads and inoculated subcutaneously (with weekly booster doses) into a Wistar rat for developing antibodies. Blood drawing from rat was done weekly for 5 weeks. Confirmation of the presence of antibodies was done by surface immunofluorescence against eggs on the antigen slides. Protein bands were determined using SDS-PAGE assay and immunogenic antigen bands were determined by reacting with antiserum after immunoblotting. The band sizes of the proteins were determined against corresponding bands of a protein ladder. Surface immunofluorescence was positive with serum obtained from day 14 post-inoculation from the Wistar rat as well as that obtained from a person with chronic enterobiasis. The most prominent and immunogenic protein bands identified from egg antigens were 21 kDa, 66 kDa, 83 kDa, 96 kDa, 112 kDa, 121 kDa, 140 kDa and 151 kDa. Methods used in this study were effective in obtaining E. vermicularis egg antigens which were immunogenic. Furthermore, surface antigens of intact eggs reacted with antibodies developed against crushed egg antigens. These findings may pave the way for the development of effective immunodiagnostics.
Collapse
|
16
|
Huang S, Qiu Y, Ma Z, Su Z, Hong W, Zuo H, Wu X, Yang Y. A secreted MIF homologue from Trichinella spiralis binds to and interacts with host monocytes. Acta Trop 2022; 234:106615. [PMID: 35901919 DOI: 10.1016/j.actatropica.2022.106615] [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: 04/28/2022] [Revised: 07/17/2022] [Accepted: 07/23/2022] [Indexed: 11/24/2022]
Abstract
Trichinella spiralis is a very successful parasite capable of surviving in many mammal hosts and residing in muscle tissues for long periods, indicating that it must have some effective strategies to escape from or guard against the host immune attack. The functions of MIF have been studied in other parasites and demonstrated to function as a virulence factor aiding in their survival by modulating the host immune response. However, the functions of Trichinella spiralis MIF (TsMIF) have not been addressed. Here, we successfully obtained the purified recombinant TsMIF and anti-TsMIF serum. Our results showed that TsMIF was expressed in all the Trichinella spiralis developmental stages, especially highly expressed in the muscle larvae (ML) and mainly located in stichocytes, midgut, cuticle, muscle cells of ML and around intrauterine embryos of female adults. We also observed TsMIF could be secreted from ML and bind to host monocytes. Next, our data demonstrated that TsMIF not only stimulated the phosphorylation of ERK1/2 and cell proliferation by binding to the host cell surface receptor CD74, but also interacted with a host intracellular protein, Jab1, which is a coactivator of AP-1 transcription. We concluded the secreted TsMIF plays an important role in the interaction between Trichinella spiralis and its host and could be a potential drug or vaccine target molecule against Trichinella spiralis infection.
Collapse
Affiliation(s)
- Shuaiqin Huang
- Department of Parasitology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yun Qiu
- Department of Biology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Zhenrong Ma
- Department of Parasitology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Zhiming Su
- Department of Biology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Wenbin Hong
- Department of Biology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Heng Zuo
- Department of Biology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Xiang Wu
- Department of Parasitology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yurong Yang
- Department of Biology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China.
| |
Collapse
|
17
|
Meza Cerda MI, Gray R, Thomson PC, Butcher L, Simpson K, Cameron A, Marcus AD, Higgins DP. Developing Immune Profiles of Endangered Australian Sea Lion ( Neophoca cinerea) Pups Within the Context of Endemic Hookworm ( Uncinaria sanguinis) Infection. Front Vet Sci 2022; 9:824584. [PMID: 35529837 PMCID: PMC9069138 DOI: 10.3389/fvets.2022.824584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
As a top predator, the endangered Australian sea lion (Neophoca cinerea) is a sentinel of ecosystem change, where population trends can reflect broader shifts in the marine environment. The population of this endemic pinniped was historically diminished by commercial sealing, and recovery has been slowed by fishery interactions, disease and, potentially, pollutants. Hookworm infects 100% of neonatal pups and has been identified as a contributor to population decline. Here, a multivariable approach using traditional serological and novel molecular tools such as qPCR and ddPCR was used to examine immune phenotypes of developing Australian sea lion pups infected with the endemic hookworm (Uncinaria sanguinis) from two South Australian colonies. Results show changing immunophenotypes throughout the patent period of infection represented by pro-inflammatory cytokines (IL-6), IgG and acute-phase proteins. Although cytokines may prove useful as markers of resistance, in this study, IL-6 is determined to be an early biomarker of inflammation in Australian sea lion pups, excluding the alternative hypothesis. Additionally, immunological differences between animals from high- and low-intensity hookworm seasons, as well as ivermectin-treated animals, indicate hookworm infection modulation of the host immune response, as evidenced by a lower IL-6 mRNA expression in the non-treated groups. This study of the Australian sea lion is an example of an ecoimmunological approach to disease investigation, which can be applied to evaluate the impact of environmental and anthropogenic factors on susceptibility to infectious diseases in free-ranging species
Collapse
Affiliation(s)
- María-Ignacia Meza Cerda
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Rachael Gray
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Peter C Thomson
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Loreena Butcher
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Kelly Simpson
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Abby Cameron
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Alan D Marcus
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Damien P Higgins
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
18
|
Donovan J, Tram TTB, Phu NH, Hiep NTT, Van VTT, Mui DTH, Ny NTH, Nghia HDT, Hanh NHH, Tan LV, Thuong NTT, Thwaites GE. Influence of Strongyloides stercoralis Coinfection on the Presentation, Pathogenesis, and Outcome of Tuberculous Meningitis. J Infect Dis 2022; 225:1653-1662. [PMID: 33104201 PMCID: PMC9071290 DOI: 10.1093/infdis/jiaa672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/21/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Helminth infections may modulate the inflammatory response to Mycobacterium tuberculosis and influence disease presentation and outcome. Strongyloides stercoralis is common among populations with high tuberculosis prevalence. Our aim was to determine whether S. stercoralis coinfection influenced clinical presentation, cerebrospinal fluid (CSF) inflammation, and outcome from tuberculous meningitis (TBM). METHODS From June 2017 to December 2019, 668 Vietnamese adults with TBM, enrolled in the ACT HIV or LAST ACT trials (NCT03092817 and NCT03100786), underwent pretreatment S. stercoralis testing by serology, stool microscopy, and/or stool polymerase chain reaction. Comparisons of pretreatment TBM severity, CSF inflammation (including cytokines), and 3-month clinical end points were performed in groups with or without active S. stercoralis infection. RESULTS Overall, 9.4% participants (63 of 668) tested positive for S. stercoralis. Active S. stercoralis infection was significantly associated with reduced pretreatment CSF neutrophil counts (median [interquartile range], 3/μL [0-25/μL] vs 14 /μL [1-83/μL]; P = .04), and with reduced CSF interferon ɣ, interleukin 2, and tumor necrosis factor α concentrations (11.4 vs 56.0 pg/mL [P = .01], 33.1 vs 54.5 pg/mL [P = .03], and 4.5 vs 11.9 pg/mL [P = .02], respectively), compared with uninfected participants. Neurological complications by 3 months were significantly reduced in participants with active S. stercoralis infection compared with uninfected participants (3.8% [1 of 26] vs 30.0% [33 of 110], respectively; P = .01). CONCLUSIONS S. stercoralis coinfection may modulate the intracerebral inflammatory response to M. tuberculosis and improve TBM clinical outcomes.
Collapse
Affiliation(s)
- Joseph Donovan
- Oxford University Clinical Research Unit, Centre for Tropical Medicine, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Trinh Thi Bich Tram
- Oxford University Clinical Research Unit, Centre for Tropical Medicine, Ho Chi Minh City, Vietnam
| | - Nguyen Hoan Phu
- Oxford University Clinical Research Unit, Centre for Tropical Medicine, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- School of Medicine, Vietnam National University of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Thu Hiep
- Oxford University Clinical Research Unit, Centre for Tropical Medicine, Ho Chi Minh City, Vietnam
| | - Vu Thi Thu Van
- Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | | | - Nguyen Thi Han Ny
- Oxford University Clinical Research Unit, Centre for Tropical Medicine, Ho Chi Minh City, Vietnam
| | - Ho Dang Trung Nghia
- Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | - Nguyen Ho Hong Hanh
- School of Medicine, Vietnam National University of Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Le Van Tan
- Oxford University Clinical Research Unit, Centre for Tropical Medicine, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nguyen Thuy Thuong Thuong
- Oxford University Clinical Research Unit, Centre for Tropical Medicine, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Guy E Thwaites
- Oxford University Clinical Research Unit, Centre for Tropical Medicine, Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Joardar N, Bhattacharya R, Halder S, Sen A, Biswas SR, Jana K, Babu SPS. Filarial thioredoxin reductase exerts anti-inflammatory effects upon lipopolysaccharide induced inflammation in macrophages. Int J Biol Macromol 2021; 193:1379-1390. [PMID: 34774593 DOI: 10.1016/j.ijbiomac.2021.10.200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/19/2021] [Accepted: 10/27/2021] [Indexed: 01/12/2023]
Abstract
Lymphatic filariasis and its associated health hazards have taken enormous tolls especially in the tropical and sub-tropical countries round the globe. Our present work contemplates the immunomodulatory role of filarial Thioredoxin reductase (TrxR) for the survival of the parasite inside the human host. For this, the protein TrxR was purified from the filarial parasite Setaria cervi and further substantiated through specific anti-TrxR antibody raised in mice. Both commercially available anti-TrxR antibody and laboratory raised antibody produced a single band with a molecular mass of ~80 kDa on western blot. The protein is optimally active at pH 7.0 and at temperature 37 °C. This protein contains both alpha helix and beta pleated sheet with selenocysteine at its active site. The Km was found to be 2.75 ± 0.49 mM. TrxR was found to downregulate lipopolysaccharide (LPS)-induced inflammation in macrophages due to inhibition of TLR4-NF-κB pathway. The result was further supported by the downregulation of inflammasome pathway and activation of alternatively activated macrophages upon TrxR treatment. Hence this study projects insights into the importance of filarial TrxR in host-parasite interface as well as it illustrates novel therapeutic strategy towards anti-filarial drug development.
Collapse
Affiliation(s)
- Nikhilesh Joardar
- Parasitology Laboratory, Department of Zoology, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India
| | - Rajarshi Bhattacharya
- Molecular Food Microbiology Laboratory, Department of Botany, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India
| | - Satyajit Halder
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India
| | - Animesh Sen
- Applied Phycology Laboratory, Department of Botany, Siksha-Bhavana, Visva-Bharati, Santiniketan 731235, West Bengal, India
| | - Swadesh Ranjan Biswas
- Molecular Food Microbiology Laboratory, Department of Botany, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India.
| | - Santi Prasad Sinha Babu
- Parasitology Laboratory, Department of Zoology, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India.
| |
Collapse
|
20
|
Evidence of MHC class I and II influencing viral and helminth infection via the microbiome in a non-human primate. PLoS Pathog 2021; 17:e1009675. [PMID: 34748618 PMCID: PMC8601626 DOI: 10.1371/journal.ppat.1009675] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/18/2021] [Accepted: 10/05/2021] [Indexed: 01/04/2023] Open
Abstract
Until recently, the study of major histocompability complex (MHC) mediated immunity has focused on the direct link between MHC diversity and susceptibility to parasite infection. However, MHC genes can also influence host health indirectly through the sculpting of the bacterial community that in turn shape immune responses. We investigated the links between MHC class I and II gene diversity gut microbiome diversity and micro- (adenovirus, AdV) and macro- (helminth) parasite infection probabilities in a wild population of non-human primates, mouse lemurs of Madagascar. This setup encompasses a plethora of underlying interactions between parasites, microbes and adaptive immunity in natural populations. Both MHC classes explained shifts in microbiome composition and the effect was driven by a few select microbial taxa. Among them were three taxa (Odoribacter, Campylobacter and Prevotellaceae-UCG-001) which were in turn linked to AdV and helminth infection status, correlative evidence of the indirect effect of the MHC via the microbiome. Our study provides support for the coupled role of MHC diversity and microbial flora as contributing factors of parasite infection. The selective pressure of the major histocompatibility complex (MHC) on microbial communities, and the potential role of this interaction in driving parasite resistance has been largely neglected. Using a natural population of the primate Microcebus griseorufus, we provide correlative evidence of two outstanding findings: that MHCI and MHCII diversity shapes the composition of the gut microbiota; and that select taxa associated with MHC diversity predicted adenovirus and helminth infection status. Our study highlights the importance of incorporating the microbiome when investigating parasite-mediated MHC selection.
Collapse
|
21
|
Schneider C. Tuft cell integration of luminal states and interaction modules in tissues. Pflugers Arch 2021; 473:1713-1722. [PMID: 34635955 PMCID: PMC8528756 DOI: 10.1007/s00424-021-02630-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/07/2023]
Abstract
Chemosensory processes are integral to the physiology of most organisms. This function is typically performed by specialized cells that are able to detect input signals and to convert them to an output dedicated to a particular group of target cells. Tuft cells are cholinergic chemosensory epithelial cells capable of producing immunologically relevant effector molecules. They are scattered throughout endoderm-derived hollow organs and function as sensors of luminal stimuli, which has been best studied in mucosal barrier epithelia. Given their epithelial origin and broad distribution, and based on their interplay with immune pathways, tuft cells can be considered a prototypical example of how complex multicellular organisms engage innate immune mechanisms to modulate and optimize organ physiology. In this review, I provide a concise overview of tuft cells and discuss how these cells influence organ adaptation to dynamic luminal conditions.
Collapse
Affiliation(s)
- Christoph Schneider
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.
| |
Collapse
|
22
|
Horn S, Borrero-Wolff D, Ritter M, Arndts K, Wiszniewsky A, Debrah LB, Debrah AY, Osei-Mensah J, Chachage M, Hoerauf A, Kroidl I, Layland LE. Distinct Immune Profiles of Exhausted Effector and Memory CD8 + T Cells in Individuals With Filarial Lymphedema. Front Cell Infect Microbiol 2021; 11:680832. [PMID: 34485170 PMCID: PMC8415778 DOI: 10.3389/fcimb.2021.680832] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/19/2021] [Indexed: 11/29/2022] Open
Abstract
CD8+ T cells are crucial for the clearance of viral infections, and current research begins to highlight their importance in parasitic diseases too. In-depth research about characteristics of CD8+ T-cell subsets and exhaustion remains uncertain, especially during filariasis, a chronic helminth infection. Lymphatic filariasis, elicited by Wuchereria bancrofti, remains a serious health problem in endemic areas in Ghana, especially in those suffering from morbidity due to lymphedema (LE). In this observational study, the characteristics and profiles of CD8+ T cells were compared between asymptomatic Wuchereria bancrofti-infected individuals, uninfected endemic normals, and those with LE (grades 2–6). Focusing on exhausted memory (CD8+exmem: CD8+ T-betdimEomeshi) and effector (CD8+exeff: CD8+T-bethiEomesdim) CD8+ T-cell subsets, advanced flow cytometry revealed that LE individuals presented reduced frequencies of IFN-γ+CD8+exmem T cells expressing Tim-3 or LAG-3 which negatively correlated to the presence of LE. Moreover, the LE cohort further showed significantly higher frequencies of IL-10+CD8+exeff T cells expressing either Tim-3, LAG-3, CD39, KLRG-1, or PD-1, all associated markers of exhaustion, and that these frequencies positively correlated with the presence of LE. In summary, this study shows that distinct exhausted CD8+ T-cell subsets are prominent in individuals suffering from LE, suggesting that enhanced inflammation and constant immune activation might drive exhaustion of CD8+ T cells. Since T-cell exhaustion is known to be associated with insufficient control of persisting antigen, the data presented here reveals that these CD8+ T-cell exhaustion patterns in filarial LE should be taken into consideration for prevention and control management of LE.
Collapse
Affiliation(s)
- Sacha Horn
- Division of Infectious Diseases and Tropical Medicine, University Hospital Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Dennis Borrero-Wolff
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany.,German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site, Bonn, Bonn, Germany
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany.,German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site, Bonn, Bonn, Germany
| | - Kathrin Arndts
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany.,German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site, Bonn, Bonn, Germany
| | - Anna Wiszniewsky
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany.,German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site, Bonn, Bonn, Germany
| | - Linda Batsa Debrah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Filariasis Unit, Kumasi, Ghana.,Department of Clinical Microbiology, School of Medicine and Dentistry, Kwame Nkrumah University of Sciences and Technology, Kumasi, Ghana.,German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site, Kumasi, Kumasi, Ghana
| | - Alexander Y Debrah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Filariasis Unit, Kumasi, Ghana.,German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site, Kumasi, Kumasi, Ghana.,Faculty of Allied Health Sciences, Kwame Nkrumah University of Sciences and Technology, Kumasi, Ghana
| | - Jubin Osei-Mensah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Filariasis Unit, Kumasi, Ghana.,German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site, Kumasi, Kumasi, Ghana
| | - Mkunde Chachage
- Division of Infectious Diseases and Tropical Medicine, University Hospital Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany.,National Institute for Medical Research (NIMR)-Mbeya Medical Research Center (MMRC), Department of Immunology, Mbeya, Tanzania.,University of Dar es Salaam-Mbeya College of Health and Allied Sciences (UDSM-MCHAS), Department of Microbiology and Immunology, Mbeya, Tanzania
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany.,German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site, Bonn, Bonn, Germany.,German Centre for Infection Research (DZIF), Neglected Tropical Disease, partner site, Bonn-Cologne, Bonn, Germany
| | - Inge Kroidl
- Division of Infectious Diseases and Tropical Medicine, University Hospital Munich, Ludwig-Maximilians-Universität (LMU), Munich, Germany.,German Centre for Infection Research (DZIF), Neglected Tropical Disease, partner site, Munich, Munich, Germany
| | - Laura E Layland
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany.,German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site, Bonn, Bonn, Germany.,German Centre for Infection Research (DZIF), Neglected Tropical Disease, partner site, Bonn-Cologne, Bonn, Germany
| |
Collapse
|
23
|
Wang C, Yang SH, Niu N, Tao J, Du XC, Yang JH, Zhu MX, Wang YN, Zhao W. lncRNA028466 regulates Th1/Th2 cytokine expression and associates with Echinococcus granulosus antigen P29 immunity. Parasit Vectors 2021; 14:295. [PMID: 34082780 PMCID: PMC8173744 DOI: 10.1186/s13071-021-04795-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023] Open
Abstract
Background Cystic echinococcosis (CE) is a parasitic disease that is caused by Echinococcus granulosus (Eg). The recombinant Echinococcus granulosus antigen P29 (rEg.P29) was shown to confer effective immunity to sheep and mice during E. granulosus secondary infection in our previous study. In this study, we sought to investigate the ability of long noncoding RNA 028466 (lncRNA028466) as a regulator for the protective immunity mediated by rEg.P29 vaccination and to study the effects of lncRNA028466 on CD4+T cell differentiation in mice spleen. Methods Female BALB/c mice were divided into two groups and were vaccinated subcutaneously with rEg.P29 antigen and PBS as a control (12 mice each group). Following prime-boost vaccination, CD4+T, CD8+T, and B cells from the spleen were isolated by flow cytometry. Quantitative real-time PCR (qRT-PCR) was performed to measure the expression of lncRNA028466 in these three kinds of cells. Then, lncRNA028466 was overexpressed and knocked down in naive CD4+T cells, and Th1 and Th2 cytokine expression was detected. qRT-PCR, western blot, and ELISA were performed to evaluate the production of IFN-γ, IL-2, IL-4, and IL-10, and flow cytometry was performed to detect the differentiation of Th1 and Th2 subgroups. Results lncRNA028466 was significantly decreased after the second week of immunization with rEg.P29 antigen. The proportion of CD4+ T cells was increased after rEg.P29 immunization. Overexpression of lncRNA028466 facilitated the production of IL-4, IL-10 and suppressed the production of IFN-γ, IL-2. Furthermore, after transfection with siRNA028466, IL-2 production was facilitated and IL-10 production was suppressed in naive CD4+ T cells. Conclusions Immunization with rEg.P29 downregulated the expression of lncRNA028466, which was related to a higher Th1 immune response and a lower Th2 immune response. Our results suggest that lncRNA028466 may be involved in rEg.P29-mediated immune response by regulating cytokine expression of Th1 and Th2. Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-04795-2.
Collapse
Affiliation(s)
- Chan Wang
- Department of Medical genetics and Cell biology, School of Basic Medical Science of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
| | - Song-Hao Yang
- Department of Medical genetics and Cell biology, School of Basic Medical Science of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
| | - Nan Niu
- Department of Medical genetics and Cell biology, School of Basic Medical Science of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
| | - Jia Tao
- Department of Medical genetics and Cell biology, School of Basic Medical Science of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
| | - Xian-Cai Du
- Department of Medical genetics and Cell biology, School of Basic Medical Science of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
| | - Ji-Hui Yang
- Center of Scientific Technology of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
| | - Ming-Xing Zhu
- Department of Medical genetics and Cell biology, School of Basic Medical Science of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.,Center of Scientific Technology of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
| | - Ya-Na Wang
- Department of Medical genetics and Cell biology, School of Basic Medical Science of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China
| | - Wei Zhao
- Center of Scientific Technology of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China. .,Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, People's Republic of China.
| |
Collapse
|
24
|
Amoani B, Gyan B, Sakyi SA, Abu EK, Nuvor SV, Barnes P, Sarkodie-Addo T, Ahenkorah B, Sewor C, Dwomoh D, Theisen M, Cappello M, Wilson MD, Adu B. Effect of hookworm infection and anthelmintic treatment on naturally acquired antibody responses against the GMZ2 malaria vaccine candidate and constituent antigens. BMC Infect Dis 2021; 21:332. [PMID: 33832450 PMCID: PMC8028774 DOI: 10.1186/s12879-021-06027-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/30/2021] [Indexed: 11/30/2022] Open
Abstract
Background Malaria and helminths diseases are co-endemic in most parts of sub-Saharan Africa. Immune responses from each of these pathogens interact, and these interactions may have implications on vaccines. The GMZ2 malaria vaccine candidate is a fusion protein of Plasmodium falciparum merozoite surface protein 3 (MSP3) and glutamate rich protein (GLURP R0). GMZ2 has recently showed modest efficacy in a phase IIb multicenter trial. Here, we assessed the effect of hookworm (Necator americanus) infection and anthelmintic treatment on naturally acquired antibody responses against GMZ2 and constituent antigens. Methods This longitudinal cross-sectional study was conducted in the Kintampo North Municipality of Ghana. Blood and stool samples were taken from 158 individuals (4–88 years old) infected with either P. falciparum alone (n = 59) or both hookworm and P. falciparum (n = 63) and uninfected endemic controls (n = 36). Stool hookworm infection was detected by the Kato-Katz method and PCR. Malaria parasitaemia was detected by RDT, light microscopy and P. falciparum-specific 18S rRNA gene PCR. Serum samples were obtained prior to hookworm treatment with a single dose of albendazole (400 mg) and 3 weeks (21 days) after treatment. Levels of IgG1, IgG3 and IgM against GMZ2, MSP3 and GLURP R0 were measured by ELISA and compared among the groups, before and after treatment. Results Participants with P. falciparum and hookworm co-infection had significantly higher IgG3 levels to GMZ2 than those with only P. falciparum infection and negative control (p < 0.05) at baseline. Treatment with albendazole led to a significant reduction in IgG3 levels against both GMZ2 and GLURP R0. Similarly, IgM and IgG1 levels against MSP3 also decreased following deworming treatment. Conclusion Individuals with co-infection had higher antibody responses to GMZ2 antigen. Treatment of hookworm/malaria co-infection resulted in a reduction in antibody responses against GMZ2 and constituent antigens after albendazole treatment. Thus, hookworm infection and treatment could have a potential implication on malaria vaccine efficacy.
Collapse
Affiliation(s)
- Benjamin Amoani
- Department of Biomedical Science, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana.,Department of Molecular Medicine, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Ben Gyan
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Samuel Asamoah Sakyi
- Department of Molecular Medicine, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.
| | - Emmanuel Kwasi Abu
- Department of Optometry, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Samuel Victor Nuvor
- Department of Microbiology and Immunology, School of Medical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Precious Barnes
- Department of Physician Assistant, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Tracy Sarkodie-Addo
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Benjamin Ahenkorah
- Department of Molecular Medicine, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.,Department of Medical Laboratory Science, Bolgatanga Technical University, Bolgatanga, Upper East Region, Ghana
| | - Christian Sewor
- Department of Biomedical Science, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Duah Dwomoh
- Department of Biostatistics, School of Public Health, University of Ghana, Accra, Ghana
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology, and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Michael Cappello
- Partnerships for Global Health, Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Michael D Wilson
- Parasitology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| |
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
|
Ntonifor HN, Chewa JS, Oumar M, Mbouobda HD. Intestinal helminths as predictors of some malaria clinical outcomes and IL-1β levels in outpatients attending two public hospitals in Bamenda, North West Cameroon. PLoS Negl Trop Dis 2021; 15:e0009174. [PMID: 33651792 PMCID: PMC7924769 DOI: 10.1371/journal.pntd.0009174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/24/2021] [Indexed: 11/19/2022] Open
Abstract
This study aimed at determining the impact of intestinal helminths on malaria parasitaemia, anaemia and pyrexia considering the levels of IL-1β among outpatients in Bamenda. A cohort of 358 consented participants aged three (3) years and above, both males and females on malaria consultation were recruited in the study. At enrolment, patients’ axillary body temperatures were measured and recorded. Venous blood was collected for haemoglobin concentration and malaria parasitaemia determination. Blood plasma was used to measure human IL-1β levels using Human ELISA Kit. The Kato-Katz technique was used to process stool samples. Five species of intestinal helminths Ascaris lumbricoides (6.4%), Enterobius vermicularis (5.0%), Taenia species (4.2%), Trichuris trichiura (1.1%) and hookworms (0.8%) were identified. The overall prevalence of Plasmodium falciparum and intestinal helminths was 30.4% (109/358) and 17.6% (63/358) respectively. The prevalence of intestinal helminths in malaria patients was 17.4% (19/109). Higher Geometric mean parasite density (GMPD ±SD) (malaria parasitaemia) was significantly observed in patients co-infected with Enterobius vermicularis (5548 ± 2829/μL, p = 0.041) and with Taenia species (6799 ± 4584/μL, p = 0.020) than in Plasmodium falciparum infected patients alone (651 ± 6076/ μL). Higher parasitaemia of (1393 ± 3031/μL) and (3464 ± 2828/μL) were recorded in patients co-infected with Ascaris lumbricoides and with hookworms respectively but the differences were not significant (p > 0.05). Anaemia and pyrexia prevalence was 27.1% (97/358) and 33.5% (120/358) respectively. Malaria patients co-infected with Enterobius vermicularis and Ascaris lumbricoides had increased risk of anaemia (OR = 13.712, p = 0.002 and OR = 16.969, p = 0.014) respectively and pyrexia (OR = 18.07, p = 0.001 and OR = 22.560, p = 0.007) respectively than their counterparts. Increased levels of IL-1β were significantly observed in anaemic (148.884 ± 36.073 pg/mL, t = 7.411, p = 0.000) and pyretic (127.737 ± 50.322 pg/mL, t = 5.028, p = 0.000) patients than in non-anaemic (64.335 ± 38.995pg/mL) and apyretic patients (58.479 ± 36.194pg/mL). Malaria patients co-infected with each species of intestinal helminths recorded higher IL-1β levels (IL-1β > 121.68 ± 58.86 pg/mL) and the overall mean (139.63 ± 38.33pg/mL) was higher compared with levels in malaria (121.68 ± 58.86 pg/mL) and helminth (61.78 ± 31.69pg/mL) infected patients alone. Intestinal helminths exacerbated the clinical outcomes of malaria in the patients and increased levels of IL-1β were observed in co-infected patients with anaemia, pyrexia and higher parasitaemia. Malaria and intestinal helminthiasis are endemic parasitic diseases in Sub Sahara Africa including Cameroon that has been associated with poverty. Humans are co-infected with these diseases. Intestinal helminths have been reported to improve or exacerbate malaria severities in co-infected patients. The precise mechanism through which they exert this is not well elucidated but there are speculations about possible immunological implications. It is therefore crucial to understand the mechanism how these neglected tropical disease (helminthiasis) impact malaria severities to develop robust integrated public health intervention strategies and treatment protocols that can effectively manage these diseases in endemic zones. In this study, we focused on the impact of these helminths on malaria parasitaemia, anaemia and pyrexia. We examined each patient for malaria and helminth infections while also measuring their haemoglobin concentrations and body temperatures. We found out that patients infected with intestinal helminths had increased risk of malaria infection and exacerbated malaria parasitaemia, anaemia and pyrexia in co-infected patients. We also observed that increased levels of IL-1β were higher in these co-infected patients than in patients infected with malaria parasite or helminths alone. Our study is informative about the possible involvement of intestinal helminths with the immune responses of the host that consequently affects malaria severity.
Collapse
Affiliation(s)
- Helen Ngum Ntonifor
- Department of Biological Sciences, Faculty of Science, University of Bamenda, Bambili, North West Region, Cameroon
- * E-mail:
| | - Julius Suh Chewa
- Department of Biological Sciences, Faculty of Science, University of Bamenda, Bambili, North West Region, Cameroon
| | - Mahamat Oumar
- Department of Biological Sciences, Faculty of Science, University of Bamenda, Bambili, North West Region, Cameroon
| | - Hermann Desire Mbouobda
- Department of Biology, Higher Teachers Training College, University of Bamenda, Bambili, North West Region, Cameroon
| |
Collapse
|
27
|
Sauer S, Beinart D, Finn SMB, Kumar SL, Cheng Q, Hwang SE, Parker W, Devi GR. Hymenolepis diminuta-based helminth therapy in C3(1)-TAg mice does not alter breast tumor onset or progression. Evol Med Public Health 2021; 9:131-138. [PMID: 33738103 PMCID: PMC7953836 DOI: 10.1093/emph/eoab007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 02/07/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND OBJECTIVES An individual's risk of breast cancer is profoundly affected by evolutionary mismatch. Mismatches in Western society known to increase the risk of breast cancer include a sedentary lifestyle and reproductive factors. Biota alteration, characterized by a loss of biodiversity from the ecosystem of the human body as a result of Western society, is a mismatch known to increase the risk of a variety of inflammation-related diseases, including colitis-associated colon cancer. However, the effect of biota alteration on breast cancer has not been evaluated. METHODOLOGY In this study, we utilized the C3(1)-TAg mouse model of breast cancer to evaluate the role of biota alteration in the development of breast cancer. This model has been used to recapitulate the role of exercise and pregnancy in reducing the risk of breast cancer. C3(1)-TAg mice were treated with Hymenolepis diminuta, a benign helminth that has been shown to reverse the effects of biota alteration in animal models. RESULTS No effect of the helminth H. diminuta was observed. Neither the latency nor tumor growth was affected by the therapy, and no significant effects on tumor transcriptome were observed based on RNAseq analysis. CONCLUSIONS AND IMPLICATIONS These findings suggest that biota alteration, although known to affect a variety of Western-associated diseases, might not be a significant factor in the high rate of breast cancer observed in Western societies. LAY SUMMARY An almost complete loss of intestinal worms in high-income countries has led to increases in allergic disorders, autoimmune conditions, and perhaps colon cancer. However, in this study, results using laboratory mice suggest that loss of intestinal worms might not be associated with breast cancer.
Collapse
Affiliation(s)
- Scott Sauer
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Dylan Beinart
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Sade M B Finn
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Sereena L Kumar
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Qing Cheng
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Shelley E Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - William Parker
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Gayathri R Devi
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
28
|
Human serum activates the tegument of female schistosomes and supports recovery from Praziquantel. Parasitol Res 2020; 120:209-221. [PMID: 33263166 PMCID: PMC7846515 DOI: 10.1007/s00436-020-06968-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 11/04/2020] [Indexed: 02/08/2023]
Abstract
Schistosomiasis is one of the most devastating parasitic disease in the world. Schistosoma spp. survive for decades within the vasculature of their human hosts. They have evolved a vast array of mechanisms to avoid the immune reaction of the host. Due to their sexual dimorphism, with the female worm lying within the gynecophoric canal of the male worm, it is the male that is exposed to the immediate environment and the soluble parts of the host’s immune response. To understand how the worms are so successful in fending off the immune attacks of the host, comparative analyses of both worm sexes in human serum (with or without Praziquantel) were performed using scanning electron microscopy, transmission electron microscopy, and immunohistochemistry. Further, gene expression analyses of tegument-specific genes were performed. Following the incubation in human serum, males and females out of pairs show morphological changes such as an altered structure of the pits below the surface and an increased number of pits per area. In addition, female schistosomes presented a marked tuft-like repulsion of their opsonized surface. The observed resistance of females to Praziquantel seemed to depend on active proteins in the human serum. Moreover, different expression profiles of tegument-specific genes indicate different functions of female_single and male_single teguments in response to human serum. Our results indicate that female schistosomes developed different evasion strategies toward the host’s immune system in comparison to males that might lead to more robustness and has to be taken into account for the development of new anti-schistosomal drugs.
Collapse
|
29
|
Corrêa F, Hidalgo C, Stoore C, Jiménez M, Hernández M, Paredes R. Cattle co-infection of Echinococcus granulosus and Fasciola hepatica results in a different systemic cytokine profile than single parasite infection. PLoS One 2020; 15:e0238909. [PMID: 32915902 PMCID: PMC7485845 DOI: 10.1371/journal.pone.0238909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
E. granulosus is a cestode that causes Cystic Echinococcosis (CE), a zoonotic disease with worldwide presence. The immune response generated by the host against the metacestode induces a permissive Th2 response, as opposed to pro-inflammatory Th1 response. In this view, mixed Th2 and regulatory responses allow parasite survival. Overall, larval Echinococcus infections induce strong regulatory responses. Fasciola hepatica, another common helminth parasite, represents a major infection in cattle. Co-infection with different parasite species in the same host, polyparasitism, is a common occurrence involving E. granulosus and F. hepatica in cattle. ‘While it is known that infection with F. hepatica also triggers a polarized Th2/Treg immune response, little is reported regarding effects on the systemic immune response of this example of polyparasitism. F. hepatica also triggers immune responses polarized to the Th2/ Treg spectrum. Serum samples from 107 animals were analyzed, and were divided according to their infection status and Echinococcal cysts fertility. Cytokines were measured utilizing a Milliplex Magnetic Bead Panel to detect IFN-γ, IL-1, IL-2, IL-4, IL-6, IL-10, IL-12 and IL-18. Cattle infected only with F. hepatica had the highest concentration of every cytokine analyzed, with both 4.24 and 3.34-fold increases in IL-10 and IL-4, respectively, compared to control animals, followed by E. granulosus and F. hepatica co-infected animals with two-fold increase in IL-10 and IL-4, compared to control animals, suggesting that E. granulosus co-infection dampens the cattle Th2/Treg immune response against F. hepatica. When considering Echinococcal cyst fertility and systemic cytokine concentrations, fertile cysts had higher IFN-γ, IL-6 and IL-18 concentrations, while infertile cysts had higher IL-10 concentrations. These results show that E. granulosus co-infection lowers Th1 and Th2 cytokine serological concentration when compared to F. hepatica infection alone. E. granulosus infections show no difference in IFN-γ, IL-1, IL-2, IL-6 and IL-18 levels compared with control animals, highlighting the immune evasion mechanisms of this cestode.
Collapse
Affiliation(s)
- Felipe Corrêa
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Christian Hidalgo
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
- Instituto de Ciencias Agroalimentarias, Animales y Ambientales (ICA3), Universidad de O’Higgins, San Fernando, Chile
| | - Caroll Stoore
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Mauricio Jiménez
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Marcela Hernández
- Laboratorio de Biología Periodontal y Departamento de Patología y Medicina Oral, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Rodolfo Paredes
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
- * E-mail:
| |
Collapse
|
30
|
Gouveia MJ, Brindley PJ, Gärtner F, Vale N. Activity of Combinations of Antioxidants and Anthelmintic Drugs against the Adult Stage of Schistosoma mansoni. J Parasitol Res 2020; 2020:8843808. [PMID: 32832132 PMCID: PMC7429017 DOI: 10.1155/2020/8843808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/19/2020] [Accepted: 07/15/2020] [Indexed: 12/25/2022] Open
Abstract
Schistosomiasis remains a major neglected tropical disease. The treatment and control of schistosomiasis rely on a single drug, praziquantel (PZQ). Despite its efficacy, treatment with PZQ presents some major drawbacks including an inability of the chemotherapy to reverse disease-induced fibrosis and the prospect of the emergence of drug resistance. Here, we investigated a novel therapeutic approach with antioxidant biomolecules in combination with PZQ against the adult developmental stage of Schistosoma mansoni and oviposition in vitro, given that this therapeutic approach achieved synergistic/additive activity against larval schistosomes. The antioxidants curcumin and oxadiazole per se exhibited antischistosomal activity against adult worms leading to severe morphological alterations and death. Additionally, the antioxidant flavone combined with vandetanib or imatinib improved antischistosomal activity against adult forms. By contrast, however, these antioxidant-anthelmintic combinations were not as effective against adults in comparison to larval schistosomes. Nevertheless, the antioxidants alone or combined with drugs inhibited oviposition.
Collapse
Affiliation(s)
- Maria João Gouveia
- Department of Molecular Pathology and Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Center for the Study in Animal Science, University of Porto (CECA/ICETA), Rua de D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
| | - Paul J. Brindley
- Department of Microbiology, Immunology & Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Fátima Gärtner
- Department of Molecular Pathology and Immunology, 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 da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
31
|
Kadesch P, Quack T, Gerbig S, Grevelding CG, Spengler B. Tissue- and sex-specific lipidomic analysis of Schistosoma mansoni using high-resolution atmospheric pressure scanning microprobe matrix-assisted laser desorption/ionization mass spectrometry imaging. PLoS Negl Trop Dis 2020; 14:e0008145. [PMID: 32401760 PMCID: PMC7250470 DOI: 10.1371/journal.pntd.0008145] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 05/26/2020] [Accepted: 02/16/2020] [Indexed: 12/19/2022] Open
Abstract
Schistosomes are human pathogens causing the neglected tropical disease schistosomiasis, which occurs worldwide in (sub-)tropical regions. This infectious disease is often associated with poverty, and more than 700 million people are at risk of infection. Exploitation of novel habitats and limited therapeutic options brought schistosomes into research focus. Schistosomes are the only trematodes that have evolved separate sexes. They are covered by their metabolically active tegument, a surface area representing the interface between male and female in their permanent mating contact but also between parasite and host. The tegument comprises, besides others, numerous specific lipid compounds. Limited information is available on the exact lipid composition and its spatial distribution. We used atmospheric-pressure scanning microprobe matrix-assisted laser desorption/ionization (AP-SMALDI) mass spectrometry imaging (MSI) to characterize the Schistosoma mansoni tegument surface in comparison to tissue sections of whole worms or couples. We found that phosphatidylcholines (PC) and specific phosphatidylethanolamines (PE) are significantly more abundant inside the worm body compared to the tegument. On the other hand, the latter was found to be enriched in sphingomyelins (SM), phosphatidylserines (PS), lysophosphatidylcholines (LPC), and specific PE species. We further investigated lipid classes concerning number of carbon atoms in fatty acyl chains as well as the degree of unsaturation and found pronounced differences between the tegument and whole-worm body. Furthermore, differences between male and female teguments were found. The lipid composition of S. mansoni tissues has been investigated in an untargeted, spatially resolved manner for the first time. WHO-defined Neglected Tropical Diseases, including schistosomiasis, are a burden for a significant part of the human world population. The fight against the diecious trematode Schistosoma mansoni can be supported by investigations of the specific molecular communication in male/female and in worm/host interactions. Improving the knowledge about S. mansoni is mandatory, since there is justified fear of the possibility of resistance development against the only available drug Praziquantel. We used mass spectrometry imaging as a powerful tool to provide topographic and tissue-specific information on the parasite. We investigated single male and female worms, as well as mating couples, regarding both, their inner tissue, and their intact surfaces, the tegument. We found highly specific lipid species and visualized their local distributions and abundances in high-resolution molecular images. Our findings may help to improve knowledge of the complex life cycles and of molecular communication mechanisms of schistosomes and may help to develop new drugs and strategies for treatment of the infectious disease.
Collapse
Affiliation(s)
- Patrik Kadesch
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Thomas Quack
- Institute of Parasitology, Justus Liebig University Giessen, Biomedical Research Center Seltersberg (BFS), Giessen, Germany
| | - Stefanie Gerbig
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Christoph G. Grevelding
- Institute of Parasitology, Justus Liebig University Giessen, Biomedical Research Center Seltersberg (BFS), Giessen, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany
- * E-mail:
| |
Collapse
|
32
|
Zheng WB, Zou Y, Zhu XQ, Liu GH. Toxocara "omics" and the promises it holds for medicine and veterinary medicine. ADVANCES IN PARASITOLOGY 2020; 109:89-108. [PMID: 32381233 DOI: 10.1016/bs.apar.2020.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Toxocariasis is one of the most neglected worldwide zoonoses that is caused by larval nematode parasites of the genus Toxocara, Toxocara canis, and to a lesser extent, Toxocara cati, whose migration mechanism is still largely unknown. Fortunately, some advanced tools have been employed, such as genomics, transcriptomics, and proteomics, to better understand the molecular biology and regulatory mechanisms of Toxocara. Using genomics and transcriptomics, we can identify a large number of genes that participate in the development of Toxocara and the interaction of parasites and their hosts and can predict the functions of unknown genes by comparing them with other relevant species. Using proteomics, we can identify somatic proteins and excretory and secretory (ES) proteins that perform specific biological functions in tissue degradation, pathogen invasion, immune evasion or modulation. These "omics" techniques also can contribute enormously to the development of new drugs, vaccines and diagnostic tools for toxocariasis. In a word, by utilizing "omics", we can better understand the Toxocara and toxocariasis. In this review, we summarized the representative achievements in Toxocara and the interaction between Toxocara spp. and their hosts based on expressed sequence tags (ESTs), microarray gene expression, next-generation sequencing (NGS) technologies and liquid chromatography-tandem mass spectrometry (LC-MS/MS), hoping to better understand the molecular biology of Toxocara, and contribute to new progress in the application areas of new drugs, vaccines and diagnostic tool for toxocariasis in the future.
Collapse
Affiliation(s)
- Wen-Bin Zheng
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China; State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yang Zou
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University College of Veterinary Medicine, Yangzhou, China.
| | - Guo-Hua Liu
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.
| |
Collapse
|
33
|
Abuzeid AMI, Zhou X, Huang Y, Li G. Twenty-five-year research progress in hookworm excretory/secretory products. Parasit Vectors 2020; 13:136. [PMID: 32171305 PMCID: PMC7071665 DOI: 10.1186/s13071-020-04010-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 03/06/2020] [Indexed: 11/16/2022] Open
Abstract
Hookworm infection is a major public health problem that threatens about 500 million people throughout tropical areas of the world. Adult hookworms survive for many years in the host intestine, where they suck blood, causing iron deficiency anemia and malnutrition. Numerous molecules, named excretory/secretory (ES) products, are secreted by hookworm adults and/or larvae to aid in parasite survival and pathobiology. Although the molecular cloning and characterization of hookworm ES products began 25 years ago, the biological role and molecular nature of many of them are still unclear. Hookworm ES products, with distinct structures and functions, have been linked to many essential events in the disease pathogenesis. These events include host invasion and tissue migration, parasite nourishment and reproduction, and immune modulation. Several of these products represent promising vaccine targets for controlling hookworm disease and therapeutic targets for many inflammatory diseases. This review aims to summarize our present knowledge about hookworm ES products, including their role in parasite biology, host-parasite interactions, and as vaccine and pharmaceutical targets and to identify research gaps and future research directions in this field.![]()
Collapse
Affiliation(s)
- Asmaa M I Abuzeid
- Guangdong Provincial Zoonosis Prevention and Control Key Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xue Zhou
- Guangdong Provincial Zoonosis Prevention and Control Key Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yue Huang
- Guangdong Provincial Zoonosis Prevention and Control Key Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Guoqing Li
- Guangdong Provincial Zoonosis Prevention and Control Key Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
34
|
Moura VBL, Milhomem AC, Lima SB, Matos-Silva H, Sugita DM, Vinaud MC, Lino-Júnior RDS. Demyelination in experimental intraventricular neurocysticercosis. ARQUIVOS DE NEURO-PSIQUIATRIA 2020; 78:103-111. [PMID: 32022135 DOI: 10.1590/0004-282x20190155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 09/19/2019] [Indexed: 11/21/2022]
Abstract
Neurocysticercosis (NCC) is classified as a neglected tropical disease, which affects mainly Latin America and Africa in spite of some reports in North America and Europe. NCC represents the cause of up to 30% of the reported cases of epilepsy in endemic countries. The NCC injuries present direct relation to the development stage, location, and number of parasites as well as to the host immune response. This study aimed the characterization of the inflammatory response and tissue injuries by means of the analyses of the periventricular and parenchymatous demyelination through the experimental intraventricular NCC infection. Therefore, BALB/c mice were submitted to experimental NCC inoculation with Taenia crassiceps cysticerci. Their brains were removed at 7, 30, 60, and 90 days after the inoculation (DAI), and analyzed after staining with hematoxylin and eosin (HE), Luxol Fast Blue, and Nissl. It was possible to observe ventriculomegaly, inflammatory infiltration composed by polymorphonuclear and mononuclear cells, and foamy macrophages. The presence of inflammatory cells was associated with neurodegeneration detected by the areas with demyelination observed initially in the periventricular area and lately in the parenchyma. In conclusion, the presence of cysticerci and the consequent inflammation were able to promote initial periventricular demyelination followed by parenchymatous demyelination as the infection progressed.
Collapse
Affiliation(s)
| | - Analia Cirqueira Milhomem
- Universidade Federal de Goiás, Programa de Pós-Graduação em Medicina Tropical e Saúde Pública, Instituto de Patologia Tropical e Saúde Pública, Goiânia GO, Brazil
| | - Sarah Buzaim Lima
- Universidade Federal de Goiás, Programa de Pós-Graduação em Medicina Tropical e Saúde Pública, Instituto de Patologia Tropical e Saúde Pública, Goiânia GO, Brazil
| | | | | | - Mariana Clare Vinaud
- Universidade Federal de Goiás, Instituto de Patologia Tropical e Saúde Pública, Goiânia GO, Brazil
| | - Ruy de Souza Lino-Júnior
- Universidade Federal de Goiás, Instituto de Patologia Tropical e Saúde Pública, Goiânia GO, Brazil
| |
Collapse
|
35
|
Cytotoxic T-Lymphocyte-Associated Antigen 4 (CTLA-4)- and Programmed Death 1 (PD-1)-Mediated Regulation of Monofunctional and Dual Functional CD4 + and CD8 + T-Cell Responses in a Chronic Helminth Infection. Infect Immun 2019; 87:IAI.00469-19. [PMID: 31570560 DOI: 10.1128/iai.00469-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/23/2019] [Indexed: 11/20/2022] Open
Abstract
Chronic helminth infections are known to be associated with the modulation of antigen-specific T-cell responses. Strongyloides stercoralis infection is characterized by the downmodulation of antigen-specific Th1 and Th17 responses and the upregulation of Th2 and Th9 responses. Immune homeostasis is partially maintained by negative regulators of T-cell activation, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD-1), which dampen effector responses during chronic infections. However, their roles in S. stercoralis infection are yet to be defined. Therefore, we sought to determine the role of CTLA-4 and PD-1 in regulating CD4+ and CD8+ T-cell responses and examined the frequencies of monofunctional and dual functional Th1/T cytotoxic type 1 (Tc1), Th17/Tc17, Th2/Tc2, and Th9/Tc9 cells in S. stercoralis infection in 15 infected individuals stimulated with parasite antigen following CTLA-4 or PD-1 blockade. Our data reveal that CTLA-4 or PD-1 blockade results in significantly enhanced frequencies of monofunctional and dual functional Th1/Tc1 and Th17/Tc17 cells and, in contrast, diminishes the frequencies of monofunctional and dual functional Th2/Tc2 and Th9/Tc9 cells with parasite antigen stimulation in whole-blood cultures. Thus, we demonstrate that CTLA-4 and PD-1 limit the induction of particular T-cell subsets in S. stercoralis infection, which suggests the importance of CTLA-4 and PD-1 in immune modulation in a chronic helminth infection.
Collapse
|
36
|
Chen TT, Peng S, Wang Y, Hu Y, Shen Y, Xu Y, Yin J, Liu C, Cao J. Improvement of Mitochondrial Activity and Fibrosis by Resveratrol Treatment in Mice with Schistosoma japonicum Infection. Biomolecules 2019; 9:biom9110658. [PMID: 31717714 PMCID: PMC6920829 DOI: 10.3390/biom9110658] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 01/21/2023] Open
Abstract
Schistosomiasis caused by Schistosoma japonicum is a major parasitic disease in the People's Republic of China. Liver fibrosis is the main pathological mechanism of schistosomiasis, and it is also the major lesion. The common drug used for its treatment, praziquantel (PZQ), does not have a marked effect on liver fibrosis. Resveratrol (RSV), which is an antioxidant, improves mitochondrial function and also attenuates liver fibrosis. The combination of PZQ and RSV has been found to have a synergistic antischistosomal effect on Schistosoma mansoni; additionally, the activity of PZQ is enhanced in the presence of RSV. Here, we examine the therapeutic effects of RSV on the S. japonicum infection in a mouse model, and we investigate RSV as a novel therapeutic agent for mitochondrial function and schistosomiasis-associated liver fibrosis (SSLF). Mitochondrial membrane potential was examined using flow cytometry analysis. The expression of the mitochondrial biogenesis genes PGC-α and fibrosis-associated genes collagen I, collagen III and α-SMA were examined using western blot analysis. Fibrosis-associated histological changes were examined using Masson trichrome staining. Additionally, the effects of RSV on S. japonicum adult worms were examined using scanning electron microscopy and transmission electron microscopy. RSV treatment improved mitochondrial function by increasing membrane potential and increasing PGC-α expression (mitochondrial biogenesis). Further, RSV attenuated liver injury, including liver scarring, by decreasing collagen deposition and the extent of fibrosis, based on the decrease in expression of the fibrosis-related genes. RSV also decreased the adult worm count and caused considerable physical damage to the worm. These results indicate that RSV upregulates mitochondrial biogenesis and inhibits fibrosis. RSV may have potential as a therapeutic target for the treatment of fibrosis in schistosomiasis.
Collapse
Affiliation(s)
- Tina Tuwen Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology, MOH, Shanghai 200025, China; (T.T.C.); (Y.W.); (Y.H.); (Y.S.); (Y.X.); (J.Y.); (C.L.)
- National Center for International Research on Tropical Diseases, Shanghai 200025, China
- WHO Collaborating Center for Tropical Diseases, Shanghai 200025, China
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Shihyi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Yanjuan Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology, MOH, Shanghai 200025, China; (T.T.C.); (Y.W.); (Y.H.); (Y.S.); (Y.X.); (J.Y.); (C.L.)
- National Center for International Research on Tropical Diseases, Shanghai 200025, China
- WHO Collaborating Center for Tropical Diseases, Shanghai 200025, China
| | - Yuan Hu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology, MOH, Shanghai 200025, China; (T.T.C.); (Y.W.); (Y.H.); (Y.S.); (Y.X.); (J.Y.); (C.L.)
- National Center for International Research on Tropical Diseases, Shanghai 200025, China
- WHO Collaborating Center for Tropical Diseases, Shanghai 200025, China
| | - Yujuan Shen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology, MOH, Shanghai 200025, China; (T.T.C.); (Y.W.); (Y.H.); (Y.S.); (Y.X.); (J.Y.); (C.L.)
- National Center for International Research on Tropical Diseases, Shanghai 200025, China
- WHO Collaborating Center for Tropical Diseases, Shanghai 200025, China
| | - Yuxin Xu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology, MOH, Shanghai 200025, China; (T.T.C.); (Y.W.); (Y.H.); (Y.S.); (Y.X.); (J.Y.); (C.L.)
- National Center for International Research on Tropical Diseases, Shanghai 200025, China
- WHO Collaborating Center for Tropical Diseases, Shanghai 200025, China
| | - Jianhai Yin
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology, MOH, Shanghai 200025, China; (T.T.C.); (Y.W.); (Y.H.); (Y.S.); (Y.X.); (J.Y.); (C.L.)
- National Center for International Research on Tropical Diseases, Shanghai 200025, China
- WHO Collaborating Center for Tropical Diseases, Shanghai 200025, China
| | - Congshan Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology, MOH, Shanghai 200025, China; (T.T.C.); (Y.W.); (Y.H.); (Y.S.); (Y.X.); (J.Y.); (C.L.)
- National Center for International Research on Tropical Diseases, Shanghai 200025, China
- WHO Collaborating Center for Tropical Diseases, Shanghai 200025, China
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology, MOH, Shanghai 200025, China; (T.T.C.); (Y.W.); (Y.H.); (Y.S.); (Y.X.); (J.Y.); (C.L.)
- National Center for International Research on Tropical Diseases, Shanghai 200025, China
- WHO Collaborating Center for Tropical Diseases, Shanghai 200025, China
- Correspondence:
| |
Collapse
|
37
|
Amoani B, Adu B, Frempong MT, Sarkodie-Addo T, Victor Nuvor S, Abu EK, Harrison LM, Cappello M, Gyan B, Wilson MD. Cytokine profiles of Necator americanus and Plasmodium falciparum co-infected patients in rural Ghana. Cytokine X 2019; 1:100014. [PMID: 33604551 PMCID: PMC7885886 DOI: 10.1016/j.cytox.2019.100014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/21/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022] Open
Abstract
Co-infection of hookworm with P. falciparum modulates blood parasitemia levels. Cytokine levels were higher in the parasite infected individuals. Serum eotaxin level correlate negatively with hookworm intensity. Deworming drug treatment alters cytokine profiles in hookworm infected subjects.
Background Necator americanus (hookworm) and Plasmodium falciparum co-infections are common in endemic communities in rural Ghana. Human immune responses to P. falciparum and hookworm are complex, and the dynamics of cytokine levels and effector mediators are poorly understood. This study aimed to determine the effect of hookworm and P. falciparum co-infection on parasite intensities and cytokine profiles in individuals before and after deworming drug treatment. Methods In this cross-sectional study conducted in the Kintampo North Municipality of Ghana blood and stool samples were analyzed from 984 participants (aged 4–88 years). Stool samples were collected at baseline from all participants and examined for the presence of hookworm using the Kato-Katz method. Blood and stool samples were analysed again two weeks after albendazole treatment of hookworm infected individuals. Malaria parasitaemia was estimated by light microscopy and P. falciparum-specific 18S rRNA gene PCR method used for species identification. Serum levels of circulating cytokines interleukins -5, -10 (IL-5, IL-10), tumor necrosis factor [TNF]-α, and eotaxin [CCL11] were determined using ELISA based methods. Results Malaria parasitaemia was significantly reduced in hookworm and P. falciparum co-infected individuals (p = 0.0018) while hookworm intensity was similar between groups. IL-10 level was significantly higher in the co-infected individuals (39.9 ± 12.2 pg/ml) compared to the single infected or the uninfected group (10.7 ± 7.6 mg/ml). IL-5 level was higher in the hookworm only infected individual. TNF-α levels were higher in all infected groups compared to the uninfected controls. CCL11 levels were significantly higher in subjects infected with hookworm only or co-infected with hookworm and P. falciparum. There was a significantly negative correlation (rs = −0.39, p = 0.021) between hookworm eggs per gram of stool and CCL11 levels in the group mono-infected with hookworm which was not affected by treatment. Treatment with albendazole led to a significant reduction of TNF-α (p = 0.041), IL-5 (p = 0.01) and IL-10 (p = 0.001) levels. Conclusion This study shows that in the absence of other helminths, co-infection of hookworm with P. falciparum may modulate blood parasitemia levels and cytokine responses. Data also show that deworming drug treatment alters these cytokine profiles in hookworm infected subjects. Future studies to elucidate the potential mechanisms underlying these observations should include an assessment of parasite specific cellular responses.
Collapse
Affiliation(s)
- Benjamin Amoani
- Department of Biomedical Science, College of Health Sciences, University of Cape Coast, Cape Coast, Ghana.,Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana.,Molecular Medicine Department, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Ghana
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Margaret T Frempong
- Molecular Medicine Department, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Ghana
| | - Tracy Sarkodie-Addo
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Samuel Victor Nuvor
- Department of Microbiology, College of Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Emmanuel Kwasi Abu
- Department of Optometry, College of Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Lisa M Harrison
- Partnerships for Global Health, Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Michael Cappello
- Partnerships for Global Health, Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Ben Gyan
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Michael D Wilson
- Parasitology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| |
Collapse
|
38
|
Engineering universal cells that evade immune detection. Nat Rev Immunol 2019; 19:723-733. [DOI: 10.1038/s41577-019-0200-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2019] [Indexed: 12/15/2022]
|
39
|
Gouveia MJ, Brindley PJ, Azevedo C, Gärtner F, da Costa JMC, Vale N. The antioxidants resveratrol and N-acetylcysteine enhance anthelmintic activity of praziquantel and artesunate against Schistosoma mansoni. Parasit Vectors 2019; 12:309. [PMID: 31221193 PMCID: PMC6585032 DOI: 10.1186/s13071-019-3566-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 06/15/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Treatment of schistosomiasis has relied on the anthelmintic drug praziquantel (PZQ) for more than a generation. Despite its celebrated performance for treatment and control of schistosomiasis and other platyhelminth infections, praziquantel has some shortcomings and the inability of this drug to counteract disease sequelae prompts the need for novel therapeutic strategies. METHODS Using a host-parasite model involving Biomphalaria glabrata and Schistosoma mansoni we established mechanical transformation of S. mansoni cercariae into newly transformed schistosomula (NTS) and characterized optimal culture conditions. Thereafter, we investigated the antischistosomal activity and ability of the antioxidants N-acetylcysteine (NAC) and resveratrol (RESV) to augment the performance of praziquantel and/or artesunate (AS) against larval stages of the parasite. Drug effects were evaluated by using an automated microscopical system to study live and fixed parasites and by transmission electron microscopy (TEM). RESULTS Transformation rates of cercariae to schistosomula reached ~ 70% when the manipulation process was optimized. Several culture media were tested, with M199 supplemented with HEPES found to be suitable for S. mansoni NTS. Among the antioxidants studied, RESV alone or combined with anthelminthic drugs achieved better results rather N-acetylcysteine (NAC). TEM observations demonstrated that the combination of AS + RESV induced severe, extensive alterations to the tegument and subtegument of NTS when compared to the constituent compounds alone. Two anthelmintic-antioxidant combinations, praziquantel-resveratrol [combination index (CI) = 0.74] and artesunate-resveratrol (CI = 0.34) displayed moderate and strong synergy, respectively. CONCLUSIONS The use of viability markers including staining with propidium iodide increased the accuracy of drug screening assays against S. mansoni NTS. The synergies observed might be the consequence of increased action by RESV on targets of AS and PZQ and/or they may act through concomitantly on discrete targets to enhance overall antischistosomal action. Combinations of active agents, preferably with discrete modes of action including activity against developmental stages and/or the potential to ameliorate infection-associated pathology, might be pursued in order to identify novel therapeutic interventions.
Collapse
Affiliation(s)
- Maria João Gouveia
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- ICBAS-UP, Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-343 Porto, Portugal
| | - Paul J. Brindley
- Department of Microbiology, Immunology and Tropical Medicine and Research Center for Neglected Diseases of Poverty, School of Medicine and Health Sciences, George Washington University, 20037 Washington, DC USA
| | - Carlos Azevedo
- Laboratory of Cell Biology, Institute of Biomedical Sciences (ICBAS/UP), University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Fátima Gärtner
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- ICBAS-UP, Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-343 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - José M. C. da Costa
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- National Health Institute Dr. Ricardo Jorge (INSA), Rua Alexandre Herculano, 321, 4000-055 Porto, Portugal
| | - Nuno Vale
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- ICBAS-UP, Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-343 Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
40
|
Ahmed SA, El-Moselhy A, El-Moammaly A, El-Shewy K. Strongyloides stercoralis in Patients on Corticosteroids Therapy Using Enzyme-Linked Immunosorbent Assay and Gelatin Particles Indirect Agglutination Tests: A Diagnostic Approach. Acta Parasitol 2019; 64:394-405. [PMID: 31069645 DOI: 10.2478/s11686-019-00060-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/24/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Patients on corticosteroids therapy (POCT) are more likely to flare up concealed Strongyloides infection and develop Strongyloides hyperinfection syndrome and/or dissemination. Such critical complications can lead to high mortality rates. Rapid detection methods are, therefore, necessary to detect Strongyloides infection in POCT with the advantage of being applicable in a developing country. METHODS Two hundred POCT have been enrolled in this study to determine the rate of infection with Strongyloides. Three different groups of POCT (Strongyloides infected, non-infected, infected with other parasites) were used to evaluate the antibodies detection capability of two serological techniques (enzyme-linked immunosorbent assay (ELISA) and gelatin particles indirect agglutination (GPIAT) against the results of the gold standard agar plate culture (APC). RESULTS With APC, the infection rate of Strongyloides stercoralis in POCT was 9.5% (19/200). POCT with Strongyloides infection displayed related risk factors (job, rural settlements, and soil contact) for infection combined with the subtropical nature of Ismailia Governorate. With regard to serology, ELISA detection results were poor compared to APC with sensitivity and specificity of 42.1% and 82.6%, respectively, and positive and negative predictive values of 72% and 30%. GPIAT appeared to be closely related to APC with sensitivity and specificity of 89.4% and 81.8%, respectively, and positive and negative predictive values of 80.9% and 96.7%. Statistical moderate correlation was detected between GPIAT and ELISA. CONCLUSION The GPIAT technique is more convenient, easier, cheaper and faster to rule out the infection of Strongyloides in POCT. It might be the test of choice for routine immunodiagnosis of human strongyloidiasis.
Collapse
Affiliation(s)
- Shahira A Ahmed
- Department of Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | - Amany El-Moselhy
- Department of Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Amal El-Moammaly
- Department of Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Khalid El-Shewy
- Department of Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
41
|
Karo-Atar D, Bitton A, Benhar I, Munitz A. Therapeutic Targeting of the Interleukin-4/Interleukin-13 Signaling Pathway: In Allergy and Beyond. BioDrugs 2019; 32:201-220. [PMID: 29736903 DOI: 10.1007/s40259-018-0280-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammation triggered by interleukin-4 (IL-4)/IL-13 is mediated by IL-4 and IL-13 receptors that are present on multiple cell types, including epithelial cells, smooth muscle, fibroblasts endothelial cells and immune cells. IL-4 exerts its activities by interacting with two specific cell surface receptors: one designated the type 1 IL-4 receptor (IL-4R); the other designated the type 2 IL-4R, a receptor complex that is also the functional receptor for IL-13. "Traditionally," IL-4 and IL-13 have been studied in the context of T helper 2-associated immune responses (i.e., type 2 immunity). In these settings, IL-4, IL-13 and their cognate receptor chains display pivotal roles where IL-4 is considered an instigator of type 2 immune responses and IL-13 an effector molecule. Thus, therapeutic targeting of the IL-4/IL-13 pathway is under extensive research, mainly for the treatment of allergic diseases. Nonetheless, in addition to IL-4's and IL-13's roles in type 2 immune responses, recent data highlight key activities for IL-4 and IL-13 in additional settings including metabolism, bone resorption, and even cognitive learning. This review summarizes the established knowledge that has accumulated regarding the roles of IL-4, IL-13, and their receptors in allergic diseases, with an emphasis on asthma, atopic dermatitis and eosinophilic esophagitis. Further, we provide an overview of the pharmacological entities targeting these cytokines and/or their receptors, which have been developed and clinically examined over the years. Finally, we will briefly highlight emerging evidence of potential new roles for IL-4 and IL-13 in other pathologies.
Collapse
Affiliation(s)
- Danielle Karo-Atar
- Biotherapeutics Cluster, Augmanity Nano LTD, Rehovot, Israel. .,Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel.
| | - Almog Bitton
- School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel
| | - Itai Benhar
- School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel
| | - Ariel Munitz
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978, Tel-Aviv, Israel.
| |
Collapse
|
42
|
Piecyk A, Ritter M, Kalbe M. The right response at the right time: Exploring helminth immune modulation in sticklebacks by experimental coinfection. Mol Ecol 2019; 28:2668-2680. [PMID: 30993799 PMCID: PMC6852435 DOI: 10.1111/mec.15106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 02/06/2023]
Abstract
Parasites are one of the strongest selective agents in nature. They select for hosts that evolve counter‐adaptive strategies to cope with infection. Helminth parasites are special because they can modulate their hosts’ immune responses. This phenomenon is important in epidemiological contexts, where coinfections may be affected. How different types of hosts and helminths interact with each other is insufficiently investigated. We used the three‐spined stickleback (Gasterosteus aculeatus) – Schistocephalus solidus model to study mechanisms and temporal components of helminth immune modulation. Sticklebacks from two contrasting populations with either high resistance (HR) or low resistance (LR) against S. solidus, were individually exposed to S. solidus strains with characteristically high growth (HG) or low growth (LG) in G. aculeatus. We determined the susceptibility to another parasite, the eye fluke Diplostomum pseudospathaceum, and the expression of 23 key immune genes at three time points after S. solidus infection. D. pseudospathaceum infection rates and the gene expression responses depended on host and S. solidus type and changed over time. Whereas the effect of S. solidus type was not significant after three weeks, T regulatory responses and complement components were upregulated at later time points if hosts were infected with HG S. solidus. HR hosts showed a well orchestrated immune response, which was absent in LR hosts. Our results emphasize the role of regulatory T cells and the timing of specific immune responses during helminth infections. This study elucidates the importance to consider different coevolutionary trajectories and ecologies when studying host‐parasite interactions.
Collapse
Affiliation(s)
- Agnes Piecyk
- Evolutionary Ecology, Max Planck Institute for Evolutionary Biology, Plön, Germany.,Marine Evolutionary Ecology, GEOMAR Helmholtz Centre for Ocean Research Kiel, Kiel, Germany
| | - Marc Ritter
- Evolutionary Ecology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Martin Kalbe
- Evolutionary Ecology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| |
Collapse
|
43
|
Dietrich CF, Chaubal N, Hoerauf A, Kling K, Piontek MS, Steffgen L, Mand S, Dong Y. Review of Dancing Parasites in Lymphatic Filariasis. Ultrasound Int Open 2019; 5:E65-E74. [PMID: 31312785 PMCID: PMC6629997 DOI: 10.1055/a-0918-3678] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/04/2019] [Accepted: 05/01/2019] [Indexed: 02/07/2023] Open
Abstract
Lymphatic filariasis is an infection transmitted by blood-sucking mosquitoes with filarial nematodes of the species Wuchereria bancrofti, Brugia malayi und B. timori . It is prevalent in tropical countries throughout the world, with more than 60 million people infected and more than 1 billion living in areas with the risk of transmission. Worm larvae with a length of less than 1 mm are transmitted by mosquitoes, develop in human lymphatic tissue to adult worms with a length of 7-10 cm, live in the human body for up to 10 years and produce millions of microfilariae, which can be transmitted further by mosquitoes. The adult worms can be easily observed by ultrasonography because of their size and fast movements (the so-called "filarial dance sign"), which can be differentiated from other movements (e. g., blood in venous vessels) by their characteristic movement profile in pulsed-wave Doppler mode. Therapeutic options include (combinations of) ivermectin, albendazole, diethylcarbamazine and doxycycline. The latter depletes endosymbiotic Wolbachia bacteria from the worms and thus sterilizes and later kills the adult worms (macrofilaricidal or adulticidal effect).
Collapse
Affiliation(s)
| | - Nitin Chaubal
- Thane Ultrasound Centre, Thane Ultrasound Centre, Thane, India
| | - Achim Hoerauf
- Institut für Med. Mikrobiologie, Immunologie und Parasitologie (IMMIP), Universität Bonn, Bonn, Germany
| | - Kerstin Kling
- Department of Infectious Disease Epidemiology, Robert Koch-Institute, Berlin, Germany
| | - Markus Schindler Piontek
- Caritas Krankenhaus Bad Mergentheim, Academic Teaching Hospital of the University of Würzburg, Medical Clinic 2, Bad Mergentheim, Germany
| | - Ludwig Steffgen
- Trainings-Zentrum Ultraschall-Diagnostik LS GmbH, Ultrasound, Mainleus, Germany
| | - Sabine Mand
- Institut für Med. Mikrobiologie, Immunologie und Parasitologie (IMMIP), Universität Bonn, Bonn, Germany
| | - Yi Dong
- Zhongshan Hospital, Ultrasound, Shanghai, China
| |
Collapse
|
44
|
Kadesch P, Quack T, Gerbig S, Grevelding CG, Spengler B. Lipid Topography in Schistosoma mansoni Cryosections, Revealed by Microembedding and High-Resolution Atmospheric-Pressure Matrix-Assisted Laser Desorption/Ionization (MALDI) Mass Spectrometry Imaging. Anal Chem 2019; 91:4520-4528. [DOI: 10.1021/acs.analchem.8b05440] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Patrik Kadesch
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Thomas Quack
- Institute of Parasitology, Justus Liebig University Giessen, BFS, Schubertstrasse 81, 35392 Giessen, Germany
| | - Stefanie Gerbig
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Christoph G. Grevelding
- Institute of Parasitology, Justus Liebig University Giessen, BFS, Schubertstrasse 81, 35392 Giessen, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| |
Collapse
|
45
|
Gouveia MJ, Brindley PJ, Rinaldi G, Gärtner F, Correia da Costa JM, Vale N. Combination Anthelmintic/Antioxidant Activity Against Schistosoma Mansoni. Biomolecules 2019; 9:E54. [PMID: 30764562 PMCID: PMC6406910 DOI: 10.3390/biom9020054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 01/11/2023] Open
Abstract
Schistosomiasis is a major neglected tropical disease. Treatment for schistosomiasis with praziquantel (PZQ), which is effective against the parasite, by itself is not capable to counteract infection-associated disease lesions including hepatic fibrosis. There is a pressing need for novel therapies. Due to their biological properties, antioxidant biomolecules might be useful in treating and reverting associated pathological sequelae. Here, we investigated a novel therapy approach based on a combination of anthelmintic drugs with antioxidant biomolecules. We used a host-parasite model involving Bioamphalaria glabrata and newly transformed schistosomula (NTS) of Schistosoma mansoni. For in vitro drug screening assays, was selected several antioxidants and evaluated not only antischistosomal activity but also ability to enhance activity of the anthelmintic drugs praziquantel (PZQ) and artesunate (AS). The morphological alterations induced by compounds alone/combined were assessed on daily basis using an inverted and automated microscope to quantify NTS viability by a fluorometric-based method. The findings indicated that not only do some antioxidants improve antischistosomal activity of the two anthelmintics, but they exhibit activity per se, leading to high mortality of NTS post-exposure. The combination index (CI) of PZQ + Mel (CI = 0.80), PZQ + Resv (CI = 0.74), AS + Resv (CI = 0.34), AS + NAC (CI = 0.89), VDT + Flav (CI = 1.03) and VDT + Resv (CI = 1.06) reveal that they display moderate to strong synergism. The combination of compounds with discrete mechanisms of action might provide a valuable adjunct to contribution for treatment of schistosomiasis-associated disease.
Collapse
Affiliation(s)
- Maria João Gouveia
- Center for the Study in Animal Science, University of Porto, (CECA/ICETA), Rua de D. Manuel II, Apt 55142, 4051-401 Porto, Portugal.
- Department of Molecular Pathology and Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
- Department of Drug Sciences, Laboratory of Pharmacology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - Paul J Brindley
- Department of Microbiology, Immunology & Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA.
| | - Gabriel Rinaldi
- Department of Microbiology, Immunology & Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA.
| | - Fátima Gärtner
- Department of Molecular Pathology and Immunology, 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.
- University of Porto, i3S, Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.
| | - José Manuel Correia da Costa
- Center for the Study in Animal Science, University of Porto, (CECA/ICETA), Rua de D. Manuel II, Apt 55142, 4051-401 Porto, Portugal.
- Department of Infectious Diseases, INSA-National Institute of Health Dr. Ricardo Jorge, Rua Alexandre Herculano 321, 4000-055 Porto, Portugal.
| | - Nuno Vale
- Department of Molecular Pathology and Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
- Department of Drug Sciences, Laboratory of Pharmacology, Faculty of Pharmacy, 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.
- University of Porto, i3S, Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.
| |
Collapse
|
46
|
Irvine MA, Kazura JW, Hollingsworth TD, Reimer LJ. Understanding heterogeneities in mosquito-bite exposure and infection distributions for the elimination of lymphatic filariasis. Proc Biol Sci 2019; 285:rspb.2017.2253. [PMID: 29386362 PMCID: PMC5805933 DOI: 10.1098/rspb.2017.2253] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/08/2018] [Indexed: 11/24/2022] Open
Abstract
It is well known that individuals in the same community can be exposed to a highly variable number of mosquito bites. This heterogeneity in bite exposure has consequences for the control of vector-borne diseases because a few people may be contributing significantly to transmission. However, very few studies measure sources of heterogeneity in a way which is relevant to decision-making. We investigate the relationship between two classic measures of heterogeneity, spatial and individual, within the context of lymphatic filariasis, a parasitic mosquito-borne disease. Using infection and mosquito-bite data for five villages in Papua New Guinea, we measure biting characteristics to model what impact bed-nets have had on control of the disease. We combine this analysis with geospatial modelling to understand the spatial relationship between disease indicators and nightly mosquito bites. We found a weak association between biting and infection heterogeneity within villages. The introduction of bed-nets increased biting heterogeneity, but the reduction in mean biting more than compensated for this, by reducing prevalence closer to elimination thresholds. Nightly biting was explained by a spatial heterogeneity model, while parasite load was better explained by an individual heterogeneity model. Spatial and individual heterogeneity are qualitatively different with profoundly different policy implications.
Collapse
Affiliation(s)
- Michael A Irvine
- School of Life Sciences, University of Warwick, Warwick, UK .,Institute of Applied Mathematics, University of British Columbia, Vancouver, Canada
| | - James W Kazura
- Center for Global Health and Disease, Case Western Reserve University, Cleveland, OH, USA
| | - T Deirdre Hollingsworth
- School of Life Sciences, University of Warwick, Warwick, UK.,Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Lisa J Reimer
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
47
|
Idris OA, Wintola OA, Afolayan AJ. Helminthiases; prevalence, transmission, host-parasite interactions, resistance to common synthetic drugs and treatment. Heliyon 2019; 5:e01161. [PMID: 30775568 PMCID: PMC6357211 DOI: 10.1016/j.heliyon.2019.e01161] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/17/2018] [Accepted: 01/21/2019] [Indexed: 01/08/2023] Open
Abstract
The morbidity caused by parasite worms on susceptible hosts is of great concern and studies were carried out to explain the mechanism of infection, prevalence, host-parasite interaction and resistance of the parasite to treatment. This review elucidates the prevalence of parasitic worm infections; which is on the increases with the increase in the world population, global warming, poor standard of living particularly in troubled regions and developing nations. The neglect of the disease coupled with the resistance of these parasites to the few available drugs becomes a huge challenge that influences global disease burden. Helminths infections pose a life threat and increase the disability-adjusted life year (DALYs) of the poor and vulnerable people. On the other hand, exploration of medicinal plants as an alternative source of treatment against drugs resistance helminths, attract insufficient attention. This review focused on providing a general overview of the prevalence of helminths, host-parasite interactions, the resistance of helminths and the medicinal plants used to treat helminthic infections.
Collapse
Affiliation(s)
- Oladayo Amed Idris
- Medicinal Plants and Economic Development (MPED) Research Centre, Department of Botany, University of Fort Hare, Alice, 5700, South Africa
| | | | | |
Collapse
|
48
|
Schroeder JC, Jones D, Maranich A. Peripheral Eosinophilia Found in Pediatric Enterobius vermicularis Infections. Clin Pediatr (Phila) 2019; 58:13-16. [PMID: 30280584 DOI: 10.1177/0009922818805193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Enterobius vermicularis, the common pinworm, is well known in North America as a parasitic infection, mainly affecting children. It is a very contagious organism, and it is responsible for a high number of infections in the United States each year. A rise in eosinophilia is linked to most parasitic infections. However, the correlation between eosinophilia and enterobiasis infections is not well documented in the literature. In this article, we present 3 cases involving patients seen for pediatric gastroenterology consultation with concern for inflammatory bowel disease. As part of their evaluation, each patient was found to have eosinophilia of unknown significance with an ultimate diagnosis of pinworm infections made by endoscopy. Their illness presentation did not include classic enterobiasis symptoms such as rectal pruritus or nighttime irritability. These cases support a link between eosinophilia and enterobiasis that may be instructive for pediatric providers seeing patients with eosinophilia for which there is no readily apparent underlying cause.
Collapse
Affiliation(s)
| | - David Jones
- 2 San Antonio Military Medical Center, San Antonio, TX, USA
| | | |
Collapse
|
49
|
Effect of Time and Mixing in Thermal Pretreatment on Faecal Indicator Bacteria Inactivation. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15061225. [PMID: 29891776 PMCID: PMC6024947 DOI: 10.3390/ijerph15061225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 05/28/2018] [Accepted: 06/08/2018] [Indexed: 11/28/2022]
Abstract
Faecal indicator bacteria of faecal coliform, Salmonella spp., and faecal Streptococcus are present at high levels in faecal sludge and affect human health. Mesophilic anaerobic digestion cannot reduce faecal indicator bacteria to meet the standards for biosolids; therefore, the thermal pretreatment of faecal sludge is essential. The main objectives of this research were to evaluate the effect of thermal (70 °C) pretreatment time (20, 40, 60, 80, 100, and 120 min) and mixing velocity gradient (no mixing, 133, 191, and 238 s−1) on faecal indicator bacteria inactivation and determine the kinetics of the inactivation. The results showed that mixing has a more positive effect on pretreatment; thermal pretreatment with mixing was able to completely inactivate faecal indicator bacteria within 80 min, and inactivation followed first-order kinetics. In addition, under optimal mixing at a velocity gradient 191 s−1, the thermal pretreatment with mixing had a positive effect on the sludge solubilisation. Soluble chemical oxygen demand (SCOD, 71,430 mg L−1) and soluble protein (7.96 g L−1) were higher than the values obtained with thermal pretreatment without mixing, which were a SCOD value of 63,600 mg L−1 and soluble protein of 6.78 g L−1.
Collapse
|
50
|
Gonzaga HT, Nunes DDS, Ribeiro VDS, Feliciano ND, Cunha-Junior JPD, Costa-Cruz JM. Metaperiodate deglycosylation of Strongyloides venezuelensis larvae: Immunochemical characterization and antigen production for human strongyloidiasis diagnosis. Acta Trop 2018; 182:27-33. [PMID: 29454735 DOI: 10.1016/j.actatropica.2018.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/24/2018] [Accepted: 02/10/2018] [Indexed: 10/18/2022]
Abstract
Strongyloidiasis is an important helminthiasis affecting million people worldwide. The aim of this study was to use sodium metaperiodate (MP) treatment to immunochemically characterize Strongyloides venezuelensis filariform larvae and use MP-treated heterologous antigen to detect IgG and subclasses in serum. Samples from individuals with definitive diagnosis of strongyloidiasis (n = 50), other parasitic diseases (n = 60) and negative endemic (n = 50) were tested. TG-ROC and two-way ANOVA were applied. MP-treatment resulted on differential localization of carbohydrates at larval structure and no carbohydrate content in saline extract (SE). Electrophoretic profiles were similar before and after treatment. ELISA sensitivity and specificity were: 90%; 88.2% for SE and 92.0%; 94.6% for MP, respectively. When using MP treated antigen we observed reduction in IgG1 and IgG3 detection in strongyloidiasis group and decrease of cross reactions in control groups. Our data demonstrate the role of carbohydrate residues in cross reactions and on the recognition of anti-Strongyloides IgG and its subclasses.
Collapse
|