1
|
Mahdy A, Mostafa OMS, Aboueldahab MM, Nigm AH. Antiparasitic activity of Cerastes cerastes venom on Schistosoma mansoni infected mice. Exp Parasitol 2024; 268:108866. [PMID: 39617195 DOI: 10.1016/j.exppara.2024.108866] [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: 08/18/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
This study investigates whether Cerastes cerastes venom (CCV) administrated at different doses (3 and 6μg/mouse) and times (a week pre-infection, the first week post-infection, and the fifth week post-infection) possesses antischistosomal activity on Schistosoma mansoni infected mice. The results showed that treatment with half lethal dose (6 μg/mouse) of CCV, at various time schedules, led to a significant decrease in the total worm burden. However, quarter lethal dose (3μg/mouse) of CCV showed a significant decrease in the total worm burden only when administered a week pre-infection. The total number of deposited eggs by females of S. mansoni was significantly decreased in the liver and the intestine of mice treated with 3μg/mouse or 6μg/mouse CCV, associated with significant alterations in the oogram pattern with significant elevation in dead eggs levels and significant decrease in the number of mature eggs. Histological examinations illustrated a significant decrease in the number and diameter of hepatic granulomas in high dose (6μg/mouse) CCV-treated groups, while it was significant only a week pre-infection in low dose (3μg/mouse) CCV-treated groups. CCV also caused several tegumental changes in treated female and male worms, including loss of the normal surface architecture, tubercular destruction, loss of tubercles' spines, oedema, erosion, membrane blebbing, and swelling. S. mansoni-infected mice groups treated with CCV (6μg/mouse) a week before infection and at fifth week post-infection had, in all individuals up to a dilution of 1:1600, higher levels of antibodies against adult worm antigen. The current investigation found that C. cerastes venom has potential antischistosomal action in a time and dose-dependent manner (more enhanced antischistosomal effects at a dose of 6 μg and in the group treated in a week before infection), in addition to its potential immunomodulatory effect against schistosomiasis infection. More studies will be required to identify the venom's active ingredients that affect the host's immunology. This information could be used in the future to develop novel antischistosomal therapies.
Collapse
Affiliation(s)
- Asmaa Mahdy
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| | - Osama M S Mostafa
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| | - Marwa M Aboueldahab
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| | - Ahmed H Nigm
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| |
Collapse
|
2
|
Lima JC, Brito RMDM, Pereira LC, Pereira NDS, Nascimento MSL, de Melo AL, Guedes PMM. Innate immune receptors are differentially expressed in mice during experimental Schistosoma mansoni early infection. Mem Inst Oswaldo Cruz 2024; 119:e240013. [PMID: 38896633 PMCID: PMC11182339 DOI: 10.1590/0074-02760240013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/09/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND The impact of Schistosoma mansoni infection over the immune response and the mechanisms involved in pathogenesis are not yet completely understood. OBJECTIVES This study aimed to evaluate the expression of innate immune receptors in three distinct mouse lineages (BALB/c, C57BL/6 and Swiss) during experimental S. mansoni infection with LE strain. METHODS The parasite burden, intestinal tissue oogram and presence of hepatic granulomas were evaluated at 7- and 12-weeks post infection (wpi). The mRNA expression for innate Toll-like receptors, Nod-like receptors, their adaptor molecules, and cytokines were determined at 2, 7 and 12 wpi in the hepatic tissue by real-time quantitative polymerase chain reaction (qPCR). FINDINGS Swiss mice showed 100% of survival, had lower parasite burden and intestinal eggs, while infected BALB/c and C57BL/6 presented 80% and 90% of survival, respectively, higher parasite burden and intestinal eggs. The three mouse lineages displayed distinct patterns in the expression of innate immune receptors, their adaptor molecules and cytokines, at 2 and 7 wpi. MAIN CONCLUSIONS Our results suggest that the pathogenesis of S. mansoni infection is related to a dynamic early activation of innate immunity receptors and cytokines important for the control of developing worms.
Collapse
Affiliation(s)
- Janete Cunha Lima
- Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Biologia Parasitária, Natal, RN, Brasil
| | | | - Luanderson Cardoso Pereira
- Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Biologia Parasitária, Natal, RN, Brasil
| | - Nathalie de Sena Pereira
- Universidade Federal do Rio Grande do Norte, Departamento de Microbiologia e Parasitologia, Natal, RN, Brasil
| | | | - Alan Lane de Melo
- Universidade Federal de Minas Gerais, Departamento de Parasitologia, Belo Horizonte, MG, Brasil
| | - Paulo Marcos Matta Guedes
- Universidade Federal do Rio Grande do Norte, Programa de Pós-Graduação em Biologia Parasitária, Natal, RN, Brasil
| |
Collapse
|
3
|
Houlder EL, Costain AH, Nambuya I, Brown SL, Koopman JPR, Langenberg MCC, Janse JJ, Hoogerwerf MA, Ridley AJL, Forde-Thomas JE, Colombo SAP, Winkel BMF, Galdon AA, Hoffmann KF, Cook PC, Roestenberg M, Mpairwe H, MacDonald AS. Pulmonary inflammation promoted by type-2 dendritic cells is a feature of human and murine schistosomiasis. Nat Commun 2023; 14:1863. [PMID: 37012228 PMCID: PMC10070318 DOI: 10.1038/s41467-023-37502-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 03/17/2023] [Indexed: 04/05/2023] Open
Abstract
Schistosomiasis is a parasitic disease affecting over 200 million people in multiple organs, including the lungs. Despite this, there is little understanding of pulmonary immune responses during schistosomiasis. Here, we show type-2 dominated lung immune responses in both patent (egg producing) and pre-patent (larval lung migration) murine Schistosoma mansoni (S. mansoni) infection. Human pre-patent S. mansoni infection pulmonary (sputum) samples revealed a mixed type-1/type-2 inflammatory cytokine profile, whilst a case-control study showed no significant pulmonary cytokine changes in endemic patent infection. However, schistosomiasis induced expansion of pulmonary type-2 conventional dendritic cells (cDC2s) in human and murine hosts, at both infection stages. Further, cDC2s were required for type-2 pulmonary inflammation in murine pre-patent or patent infection. These data elevate our fundamental understanding of pulmonary immune responses during schistosomiasis, which may be important for future vaccine design, as well as for understanding links between schistosomiasis and other lung diseases.
Collapse
Affiliation(s)
- E L Houlder
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, Netherlands
| | - A H Costain
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, Netherlands
| | - I Nambuya
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - S L Brown
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - J P R Koopman
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, Netherlands
| | - M C C Langenberg
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, Netherlands
| | - J J Janse
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, Netherlands
| | - M A Hoogerwerf
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, Netherlands
| | - A J L Ridley
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - J E Forde-Thomas
- Department of Life Sciences, Aberystwyth University, Aberystwyth, SY23 3DA, UK
| | - S A P Colombo
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - B M F Winkel
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, Netherlands
| | - A A Galdon
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - K F Hoffmann
- Department of Life Sciences, Aberystwyth University, Aberystwyth, SY23 3DA, UK
| | - P C Cook
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - M Roestenberg
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, Netherlands
| | - H Mpairwe
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - A S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
| |
Collapse
|
4
|
Schistosoma mansoni egg-derived extracellular vesicles: A promising vaccine candidate against murine schistosomiasis. PLoS Negl Trop Dis 2021; 15:e0009866. [PMID: 34644290 PMCID: PMC8544836 DOI: 10.1371/journal.pntd.0009866] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/25/2021] [Accepted: 09/30/2021] [Indexed: 02/05/2023] Open
Abstract
Extracellular vesicles (EVs) are protein-loaded nano-scaled particles that are extracellularly released by eukaryotes and prokaryotes. Parasite's EVs manipulate the immune system, making them probable next-generation vaccines. Schistosomal EVs carry different proteins of promising immunizing potentials. For evaluating the immune-protective role of Schistosoma mansoni (S. mansoni) egg-derived EVs against murine schistosomiasis, EVs were isolated from cultured S. mansoni eggs by progressive sequential cooling ultra-centrifugation technique. Isolated EVs were structurally identified using transmission electron microscope and their protein was quantified by Lowry's technique. Experimental mice were subcutaneously immunized with three doses of 20 μg EVs (with or without alum adjuvant); every two weeks, then were challenged with S. mansoni cercariae two weeks after the last immunizing dose. Six weeks post infection, mice were sacrificed for vaccine candidate assessment. EVs protective efficacy was evaluated through parasitological, histopathological, and immunological parameters. Results showed significant reduction of tegumentally deranged adult worms, hepatic and intestinal egg counts reduction by 46.58%, 93.14% and 93.17% respectively, accompanied by remarkable amelioration of sizes, numbers and histopathology of hepatic granulomata mediated by high interferon gamma (IFN γ) and antibody level. Using sera from vaccinated mice, the molecular weight of EVs' protein components targeted by the antibody produced was recognized by western immunoblot. Results revealed two bands of ~ 14 KDa and ~ 21 KDa, proving that EVs are able to stimulate specific antibodies response. In conclusion, the present study highlighted the role of S. mansoni-egg derived EVs as a potential vaccine candidate against murine schistosomiasis mansoni.
Collapse
|
5
|
Springer LE, Patton JB, Zhan T, Rabson AB, Lin HC, Manser T, Lok JB, Hess JA, Abraham D. Strongyloides stercoralis and HTLV-1 coinfection in CD34+ cord blood stem cell humanized mice: Alteration of cytokine responses and enhancement of larval growth. PLoS Negl Trop Dis 2021; 15:e0009559. [PMID: 34314415 PMCID: PMC8315519 DOI: 10.1371/journal.pntd.0009559] [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: 11/30/2020] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Viral and parasitic coinfections are known to lead to both enhanced disease progression and altered disease states. HTLV-1 and Strongyloides stercoralis are co-endemic throughout much of their worldwide ranges resulting in a significant incidence of coinfection. Independently, HTLV-1 induces a Th1 response and S. stercoralis infection induces a Th2 response. However, coinfection with the two pathogens has been associated with the development of S. stercoralis hyperinfection and an alteration of the Th1/Th2 balance. In this study, a model of HTLV-1 and S. stercoralis coinfection in CD34+ umbilical cord blood hematopoietic stem cell engrafted humanized mice was established. An increased level of mortality was observed in the HTLV-1 and coinfected animals when compared to the S. stercoralis infected group. The mortality was not correlated with proviral loads or total viral RNA. Analysis of cytokine profiles showed a distinct shift towards Th1 responses in HTLV-1 infected animals, a shift towards Th2 cytokines in S. stercoralis infected animals and elevated TNF-α responses in coinfected animals. HTLV-1 infected and coinfection groups showed a significant, yet non-clonal expansion of the CD4+CD25+ T-cell population. Numbers of worms in the coinfection group did not differ from those of the S. stercoralis infected group and no autoinfective larvae were found. However, infective larvae recovered from the coinfection group showed an enhancement in growth, as was seen in mice with S. stercoralis hyperinfection caused by treatment with steroids. Humanized mice coinfected with S. stercoralis and HTLV-1 demonstrate features associated with human infection with these pathogens and provide a unique opportunity to study the interaction between these two infections in vivo in the context of human immune cells.
Collapse
Affiliation(s)
- Lauren E Springer
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - John B Patton
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Tingting Zhan
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Arnold B Rabson
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Hsin-Ching Lin
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, New Jersey, United States of America
| | - Tim Manser
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - James B Lok
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jessica A Hess
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - David Abraham
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
6
|
Mukendi JPK, Nakamura R, Uematsu S, Hamano S. Interleukin (IL)-33 is dispensable for Schistosoma mansoni worm maturation and the maintenance of egg-induced pathology in intestines of infected mice. Parasit Vectors 2021; 14:70. [PMID: 33482904 PMCID: PMC7821721 DOI: 10.1186/s13071-020-04561-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Schistosomes are trematode worms that dwell in their definitive host's blood vessels, where females lay eggs that need to be discharged into the environment with host excreta to maintain their life-cycle. Both worms and eggs require type 2 immunity for their maturation and excretion, respectively. However, the immune molecules that orchestrate such immunity remain unclear. Interleukin (IL)-33 is one of the epithelium-derived cytokines that induce type 2 immunity in tissues. The aim of this study was to determine the role of IL-33 in the maturation, reproduction and excretion of Schistosoma mansoni eggs, and in the maintenance of egg-induced pathology in the intestines of mice. METHODS The morphology of S. mansoni worms and the number of eggs in intestinal tissues were studied at different time points post-infection in S. mansoni-infected IL-33-deficient (IL-33-/-) and wild-type (WT) mice. IL-5 and IL-13 production in the spleens and mesenteric lymph nodes were measured. Tissue histology was performed on the terminal ilea of both infected and non-infected mice. RESULTS Worms from IL-33-/- and WT mice did not differ morphologically at 4 and 6 weeks post-infection (wpi). The number of eggs in intestinal tissues of IL-33-/- and WT mice differed only slightly. At 6 wpi, IL-33-/- mice presented impaired type 2 immunity in the intestines, characterized by a decreased production of IL-5 and IL-13 in mesenteric lymph nodes and fewer inflammatory infiltrates with fewer eosinophils in the ilea. There was no difference between IL-33-/- and WT mice in the levels of IL-25 and thymic stromal lymphopoietin (TSLP) in intestinal tissues. CONCLUSIONS Despite its ability to initiate type 2 immunity in tissues, IL-33 alone seems dispensable for S. mansoni maturation and its absence may not affect much the accumulation of eggs in intestinal tissues. The transient impairment of type 2 immunity observed in the intestines, but not spleens, highlights the importance of IL-33 over IL-25 and TSLP in initiating, but not maintaining, locally-induced type 2 immunity in intestinal tissues during schistosome infection. Further studies are needed to decipher the role of each of these molecules in schistosomiasis and clarify the possible interactions that might exist between them.
Collapse
Affiliation(s)
- Jean Pierre Kambala Mukendi
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Risa Nakamura
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan
- Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shinjiro Hamano
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| |
Collapse
|
7
|
Mbanefo EC, Fu CL, Ho CP, Le L, Ishida K, Hammam O, Hsieh MH. Interleukin-4 Signaling Plays a Major Role in Urogenital Schistosomiasis-Associated Bladder Pathogenesis. Infect Immun 2020; 88:e00669-19. [PMID: 31843965 PMCID: PMC7035943 DOI: 10.1128/iai.00669-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 11/26/2019] [Indexed: 11/20/2022] Open
Abstract
Interleukin-4 (IL-4) is crucial in many helminth infections, but its role in urogenital schistosomiasis, infection with Schistosoma haematobium worms, remains poorly understood due to a historical lack of animal models. The bladder pathology of urogenital schistosomiasis is caused by immune responses to eggs deposited in the bladder wall. A range of pathology occurs, including urothelial hyperplasia and cancer, but associated mechanisms and links to IL-4 are largely unknown. We modeled urogenital schistosomiasis by injecting the bladder walls of IL-4 receptor-alpha knockout (Il4ra-/- ) and wild-type mice with S. haematobium eggs. Readouts included bladder histology and ex vivo assessments of urothelial proliferation, cell cycle, and ploidy status. We also quantified the effects of exogenous IL-4 on urothelial cell proliferation in vitro, including cell cycle status and phosphorylation patterns of major downstream regulators in the IL-4 signaling pathway. There was a significant decrease in the intensity of granulomatous responses to bladder-wall-injected S. haematobium eggs in Il4ra-/- versus wild-type mice. S. haematobium egg injection triggered significant urothelial proliferation, including evidence of urothelial hyper-diploidy and cell cycle skewing in wild-type but not Il4ra-/- mice. Urothelial exposure to IL-4 in vitro led to cell cycle polarization and increased phosphorylation of AKT. Our results show that IL-4 signaling is required for key pathogenic features of urogenital schistosomiasis and that particular aspects of this signaling pathway may exert these effects directly on the urothelium. These findings point to potential mechanisms by which urogenital schistosomiasis promotes bladder carcinogenesis.
Collapse
Affiliation(s)
- Evaristus C Mbanefo
- Division of Urology, Children's National Medical Center, Washington, DC, USA
- Bladder Immunology Group, Biomedical Research Institute, Rockville, Maryland, USA
| | - Chi-Ling Fu
- Stanford University School of Medicine, Stanford, California, USA
| | - Christina P Ho
- Division of Urology, Children's National Medical Center, Washington, DC, USA
| | - Loc Le
- Bladder Immunology Group, Biomedical Research Institute, Rockville, Maryland, USA
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kenji Ishida
- Bladder Immunology Group, Biomedical Research Institute, Rockville, Maryland, USA
| | | | - Michael H Hsieh
- Division of Urology, Children's National Medical Center, Washington, DC, USA
- Bladder Immunology Group, Biomedical Research Institute, Rockville, Maryland, USA
- Department of Urology, The George Washington University, Washington, DC, USA
| |
Collapse
|
8
|
Doenhoff MJ, Modha J, Walker AJ. Failure of in vitro-cultured schistosomes to produce eggs: how does the parasite meet its needs for host-derived cytokines such as TGF-β? Int J Parasitol 2019; 49:747-757. [PMID: 31348959 DOI: 10.1016/j.ijpara.2019.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 12/20/2022]
Abstract
When adult schistosome worm pairs are transferred from experimental hosts to in vitro culture they cease producing viable eggs within a few days. Female worms in unisexual infections fail to mature, and when mature adult females are separated from male partners they regress sexually. Worms cultured from the larval stage are also permanently reproductively defective. The cytokine transforming growth factor beta derived from the mammalian host is considered important in stimulating schistosome female worm maturation and maintenance of fecundity. The means by which schistosomes acquire TGF-β have not been elucidated, but direct uptake in vivo seems unlikely as the concentration of free, biologically active cytokine in host blood is very low. Here we review the complexities of schistosome development and male-female interactions, and we speculate about two possibilities on how worms obtain the TGF-β they are assumed to need: (i) worms may have mechanisms to free active cytokine from the latency-inducing complex of proteins in which it is associated, and/or (ii) they may obtain the cytokine from alpha 2-macroglobulin, a blood-borne protease inhibitor to which TGF-β can bind. These ideas are experimentally testable.
Collapse
Affiliation(s)
- Michael J Doenhoff
- School of Life Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Jay Modha
- Modha Biomedical Ltd, 9B St Cuthberts Avenue, Great Glen, Leicester LE8 9EJ, UK
| | - Anthony J Walker
- Molecular Parasitology Laboratory, School of Life Sciences Pharmacy and Chemistry, Kingston University, Kingston University, Penrhyn Road, Kingston upon Thames, Surrey KT1 2EE, UK
| |
Collapse
|
9
|
Soloviova K, Fox EC, Dalton JP, Caffrey CR, Davies SJ. A secreted schistosome cathepsin B1 cysteine protease and acute schistosome infection induce a transient T helper 17 response. PLoS Negl Trop Dis 2019; 13:e0007070. [PMID: 30653492 PMCID: PMC6353221 DOI: 10.1371/journal.pntd.0007070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 01/30/2019] [Accepted: 12/11/2018] [Indexed: 12/11/2022] Open
Abstract
The natural history of schistosome infection in the mammalian host is determined by CD4+ T helper responses mounted against different parasite life cycle stages. A T helper 2 (TH2) response to schistosome eggs is required for host survival and establishment of chronic infection. However, a TH2 cell-derived cytokine also contributes to an immune milieu that is conducive to schistosome growth and development. Thus, the same responses that allow for host survival have been co-opted by schistosomes to facilitate parasite development and transmission, underscoring the significance of CD4+ T cell responses to both worms and eggs in the natural history of schistosome infection. Here we show that a cathepsin B1 cysteine protease secreted by schistosome worms not only induces TH2 responses, but also TH1 and TH17 responses, by a mechanism that is dependent on the proteolytic activity of the enzyme. Further investigation revealed that, in addition to the expected TH1 and TH2 responses, acute schistosome infection also induces a transient TH17 response that is rapidly down-regulated at the onset of oviposition. TH17 responses are implicated in the development of severe egg-induced pathology. The regulation of worm-induced TH17 responses during acute infection could therefore influence the expression of high and low pathology states as infection progresses. Schistosomiasis, a neglected tropical disease caused by parasites of the genus Schistosoma, is prevalent throughout the developing world, with more than 230 million people infected. Left untreated, schistosome infection may cause relatively mild disease with some morbidity, or, in a minority of cases, result in severe pathology and death. These variable outcomes are recapitulated in animal models, where the natural history of schistosome infection is profoundly influenced by the responses of host CD4+ T helper cells. Type 2 CD4+ T cell (TH2) responses, which allow for host survival by limiting pathology, have ironically also been co-opted by schistosomes to promote parasite development. On the other hand, TH17 responses have been implicated in the development of severe pathology, in both experimentally infected animals and naturally infected humans. Here we show that a schistosome proteolytic enzyme (SmCB1), produced in the parasite gut and released into the bloodstream, induces both TH2 and TH17 responses by a mechanism that requires the enzyme’s inherent proteolytic activity. Further investigation revealed that acute schistosome infection also induces a transient TH17 response that is rapidly down-regulated once parasite egg-laying commences. Regulation of TH17 responses during early infection may help determine whether mild or severe pathology develops as the infection progresses.
Collapse
Affiliation(s)
- Kateryna Soloviova
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Ellen C. Fox
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - John P. Dalton
- School of Biological Sciences, Medical Biology Centre, Queen’s University Belfast, Northern Ireland, United Kingdom
| | - Conor R. Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Stephen J. Davies
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
10
|
Tallima H, Abou El Dahab M, El Ridi R. Role of T lymphocytes and papain enzymatic activity in the protection induced by the cysteine protease against Schistosoma mansoni in mice. J Adv Res 2019; 17:73-84. [PMID: 31193307 PMCID: PMC6526234 DOI: 10.1016/j.jare.2018.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/26/2018] [Accepted: 12/26/2018] [Indexed: 01/08/2023] Open
Abstract
Papain use deciphered the protection mechanism(s) of the schistosomiasis vaccine. Papain stimulation of innate immunity induced parasite egg attrition. Papain enzymatic and non-enzymatic sites activated T cells and innate immunity. IgG1 antibodies and liver uric acid and ARA levels correlated with protection. Identification of type 2 immunity-inducing cysteine peptidases motifs is required.
Papain, an experimental model protease, was used to decipher the protective mechanism(s) of the cysteine peptidase-based schistosomiasis vaccine. To examine the role of T lymphocytes, athymic nude (nu/nu) and immunocompetent haired (nu/+) mice were subcutaneously (sc) injected with 50 µg active papain two days before percutaneous exposure to 100 cercariae of Schistosoma mansoni. Highly significant (P < 0.005) reductions in worm burden required competent T lymphocytes, while significant increases (P < 0.05) of >80% in dead parasite ova in the small intestine were independent of T cell activity and likely relied on the innate immune axis. To investigate the role of enzymatic activity, immunocompetent mice were sc injected with 50 µg active or E-64-inactivated papain two days before exposure to cercariae. The reductions in worm burden were highly significant (P < 0.0001), reaching >65% and 40% in active and inactivated papain-treated mice, respectively. Similar highly significant (P < 0.0001) decreases of 85% in the viability of parasite ova in the small intestine occurred in both active and inactivated papain-treated mice. These findings indicated that immune responses elicited by one or more papain structural motifs are necessary and sufficient for induction of considerable parasite and egg attrition. Correlates of protection included IgG1-dominated antibody responses and increases in the levels of uric acid and arachidonic acid in the lung and liver upon parasite migration in these sites. Identification of the shared patterns or motifs in cysteine peptidases and evaluation of their immune protective potential will pave the way to the development of a safe, efficacious, storage-stable, and cost-effective schistosomiasis vaccine.
Collapse
Affiliation(s)
- Hatem Tallima
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt.,Department of Chemistry, School of Science and Engineering, American University in Cairo, New Cairo 11835, Egypt
| | - Marwa Abou El Dahab
- Zoology Department, Faculty of Science, Ein Shams University, Cairo 11566, Egypt
| | - Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
11
|
Costain AH, MacDonald AS, Smits HH. Schistosome Egg Migration: Mechanisms, Pathogenesis and Host Immune Responses. Front Immunol 2018; 9:3042. [PMID: 30619372 PMCID: PMC6306409 DOI: 10.3389/fimmu.2018.03042] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 12/10/2018] [Indexed: 12/22/2022] Open
Abstract
Many parasitic worms possess complex and intriguing life cycles, and schistosomes are no exception. To exit the human body and progress to their successive snail host, Schistosoma mansoni eggs must migrate from the mesenteric vessels, across the intestinal wall and into the feces. This process is complex and not always successful. A vast proportion of eggs fail to leave their definite host, instead becoming lodged within intestinal or hepatic tissue, where they can evoke potentially life-threatening pathology. Thus, to maximize the likelihood of successful egg passage whilst minimizing host pathology, intriguing egg exit strategies have evolved. Notably, schistosomes actively exert counter-inflammatory influences on the host immune system, discreetly compromise endothelial and epithelial barriers, and modulate granuloma formation around transiting eggs, which is instrumental to their migration. In this review, we discuss new developments in our understanding of schistosome egg migration, with an emphasis on S. mansoni and the intestine, and outline the host-parasite interactions that are thought to make this process possible. In addition, we explore the potential immune implications of egg penetration and discuss the long-term consequences for the host of unsuccessful egg transit, such as fibrosis, co-infection and cancer development.
Collapse
Affiliation(s)
- Alice H. Costain
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Andrew S. MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Hermelijn H. Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
12
|
Hunter KS, Davies SJ. Host Adaptive Immune Status Regulates Expression of the Schistosome AMP-Activated Protein Kinase. Front Immunol 2018; 9:2699. [PMID: 30519243 PMCID: PMC6260181 DOI: 10.3389/fimmu.2018.02699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/01/2018] [Indexed: 12/25/2022] Open
Abstract
Schistosomes exhibit profound developmental adaptations in response to the immune status of their mammalian host, including significant attenuation of parasite growth, development and reproduction in response to deficits in host adaptive immunity. These observations led us to hypothesize that schistosomes regulate the utilization of energy resources in response to immunological conditions within the host. To test this hypothesis, we identified and characterized the Schistosoma mansoni AMP-activated protein kinase (AMPK), a heterotrimeric enzyme complex that is central to regulating energy metabolism at the cellular and organismal level in eukaryotes. We show that expression of the catalytic α subunit is developmentally regulated during the parasite life cycle, with peak expression occurring in adult worms. However, the protein is present and phosphorylated in all life cycle stages examined, suggesting a need for active regulation of energy resources throughout the life cycle. In contrast, transcription of the AMPK α gene is down-regulated in cercariae and schistosomula, suggesting that the protein in these life cycle stages is pre-synthesized in the sporocyst and that expression must be re-initiated once inside the mammalian host. We also show that schistosome AMPK α activity in adult worms is sensitive to changes in the parasite's environment, suggesting a mechanism by which schistosome metabolism may be responsive to host immune factors. Finally, we show that AMPK α expression is significantly down-regulated in parasites isolated from immunodeficient mice, suggesting that modulation of parasite energy metabolism may contribute to the attenuation of schistosome growth and reproduction in immunodeficient hosts. These findings provide insights into the molecular interactions between schistosomes and their vertebrate hosts and suggest that parasite energy metabolism may represent a novel target for anti-schistosome interventions.
Collapse
Affiliation(s)
- Kasandra S Hunter
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Stephen J Davies
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
13
|
Bais S, Greenberg RM. TRP channels as potential targets for antischistosomals. Int J Parasitol Drugs Drug Resist 2018; 8:511-517. [PMID: 30224169 PMCID: PMC6287577 DOI: 10.1016/j.ijpddr.2018.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 01/08/2023]
Abstract
Ion channels are membrane protein complexes that underlie electrical excitability in cells, allowing ions to diffuse through cell membranes in a regulated fashion. They are essential for normal functioning of the neuromusculature and other tissues. Ion channels are also validated targets for many current anthelmintics, yet the properties of only a small subset of ion channels in parasitic helminths have been explored in any detail. Transient receptor potential (TRP) channels comprise a widely diverse superfamily of ion channels with important roles in sensory signaling, regulation of ion homeostasis, organellar trafficking, and other functions. There are several subtypes of TRP channels, including TRPA1 and TRPV1 channels, both of which are involved in, among other functions, sensory, nociceptive, and inflammatory signaling in mammals. Several lines of evidence indicate that TRPA1-like channels in schistosomes exhibit pharmacological sensitivities that differ from their mammalian counterparts and that may signify unique physiological properties as well. Thus, in addition to responding to TRPA1 modulators, schistosome TRPA1-like channels also respond to compounds that in other organisms modulate TRPV1 channels. Notably, TRPV channel genes are not found in schistosome genomes. Here, we review the evidence leading to these conclusions and examine potential implications. We also discuss recent results showing that praziquantel, the current drug of choice against schistosomiasis, selectively targets host TRP channels in addition to its likely primary targets in the parasite. The results we discuss add weight to the notion that schistosome TRP channels are worthy of investigation as candidate therapeutic targets.
Collapse
Affiliation(s)
- Swarna Bais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia PA 19104, USA
| | - Robert M Greenberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia PA 19104, USA.
| |
Collapse
|
14
|
Host- and Helminth-Derived Endocannabinoids That Have Effects on Host Immunity Are Generated during Infection. Infect Immun 2018; 86:IAI.00441-18. [PMID: 30104215 DOI: 10.1128/iai.00441-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/07/2018] [Indexed: 02/07/2023] Open
Abstract
Helminths have coevolved with their hosts, resulting in the development of specialized host immune mechanisms and parasite-specific regulatory products. Identification of new pathways that regulate helminth infection could provide a better understanding of host-helminth interaction and may identify new therapeutic targets for helminth infection. Here we identify the endocannabinoid system as a new mechanism that influences host immunity to helminths. Endocannabinoids are lipid-derived signaling molecules that control important physiologic processes, such as feeding behavior and metabolism. Following murine infection with Nippostrongylus brasiliensis, an intestinal nematode with a life cycle similar to that of hookworms, we observed increased levels of endocannabinoids (2-arachidonoylglycerol [2-AG] or anandamide [AEA]) and the endocannabinoid-like molecule oleoylethanolamine (OEA) in infected lung and intestine. To investigate endocannabinoid function in helminth infection, we employed pharmacological inhibitors of cannabinoid subtype receptors 1 and 2 (CB1R and CB2R). Compared to findings for vehicle-treated mice, inhibition of CB1R but not CB2R resulted in increased N. brasiliensis worm burden and egg output, associated with significantly decreased expression of the T helper type 2 cytokine interleukin 5 (IL-5) in intestinal tissue and splenocyte cultures. Strikingly, bioinformatic analysis of genomic and transcriptome sequencing (RNA-seq) data sets identified putative genes encoding endocannabinoid biosynthetic and degradative enzymes in many parasitic nematodes. To test the novel hypothesis that helminth parasites produce their own endocannabinoids, we measured endocannabinoid levels in N. brasiliensis by mass spectrometry and quantitative PCR and found that N. brasiliensis parasites produced endocannabinoids, especially at the infectious larval stage. To our knowledge, this is the first report of helminth- and host-derived endocannabinoids that promote host immune responses and reduce parasite burden.
Collapse
|
15
|
Batugedara HM, Li J, Chen G, Lu D, Patel JJ, Jang JC, Radecki KC, Burr AC, Lo DD, Dillman AR, Nair MG. Hematopoietic cell-derived RELMα regulates hookworm immunity through effects on macrophages. J Leukoc Biol 2018; 104:855-869. [PMID: 29992625 DOI: 10.1002/jlb.4a0917-369rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 06/06/2018] [Accepted: 06/14/2018] [Indexed: 12/29/2022] Open
Abstract
Resistin-like molecule α (RELMα) is a highly secreted protein in type 2 (Th2) cytokine-induced inflammation including helminth infection and allergy. In infection with Nippostrongylus brasiliensis (Nb), RELMα dampens Th2 inflammatory responses. RELMα is expressed by immune cells, and by epithelial cells (EC); however, the functional impact of immune versus EC-derived RELMα is unknown. We generated bone marrow (BM) chimeras that were RELMα deficient (RELMα-/ - ) in BM or non BM cells and infected them with Nb. Non BM RELMα-/- chimeras had comparable inflammatory responses and parasite burdens to RELMα+/+ mice. In contrast, both RELMα-/- and BM RELMα-/- mice exhibited increased Nb-induced lung and intestinal inflammation, correlated with elevated Th2 cytokines and Nb killing. CD11c+ lung macrophages were the dominant BM-derived source of RELMα and can mediate Nb killing. Therefore, we employed a macrophage-worm co-culture system to investigate whether RELMα regulates macrophage-mediated Nb killing. Compared to RELMα+ /+ macrophages, RELMα-/- macrophages exhibited increased binding to Nb and functionally impaired Nb development. Supplementation with recombinant RELMα partially reversed this phenotype. Gene expression analysis revealed that RELMα decreased cell adhesion and Fc receptor signaling pathways, which are associated with macrophage-mediated helminth killing. Collectively, these studies demonstrate that BM-derived RELMα is necessary and sufficient to dampen Nb immune responses, and identify that one mechanism of action of RELMα is through inhibiting macrophage recruitment and interaction with Nb. Our findings suggest that RELMα acts as an immune brake that provides mutually beneficial effects for the host and parasite by limiting tissue damage and delaying parasite expulsion.
Collapse
Affiliation(s)
- Hashini M Batugedara
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Gang Chen
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Dihong Lu
- Department of Nematology, University of California Riverside, Riverside, California, USA
| | - Jay J Patel
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Jessica C Jang
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Kelly C Radecki
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Abigail C Burr
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - David D Lo
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Adler R Dillman
- Department of Nematology, University of California Riverside, Riverside, California, USA
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| |
Collapse
|
16
|
Bais S, Berry CT, Liu X, Ruthel G, Freedman BD, Greenberg RM. Atypical pharmacology of schistosome TRPA1-like ion channels. PLoS Negl Trop Dis 2018; 12:e0006495. [PMID: 29746471 PMCID: PMC5963811 DOI: 10.1371/journal.pntd.0006495] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/22/2018] [Accepted: 05/02/2018] [Indexed: 12/21/2022] Open
Abstract
Parasitic flatworms of the genus Schistosoma cause schistosomiasis, a neglected tropical disease estimated to affect over 200 million people worldwide. Praziquantel is the only antischistosomal currently available for treatment, and there is an urgent need for new therapeutics. Ion channels play key roles in physiology and are targets for many anthelmintics, yet only a few representatives have been characterized in any detail in schistosomes and other parasitic helminths. The transient receptor potential (TRP) channel superfamily comprises a diverse family of non-selective cation channels that play key roles in sensory transduction and a wide range of other functions. TRP channels fall into several subfamilies. Members of both the TRPA and TRPV subfamilies transduce nociceptive and inflammatory signals in mammals, and often also respond to chemical and thermal signals. We previously showed that although schistosomes contain no genes predicted to encode TRPV channels, TRPV1-selective activators such as capsaicin and resiniferatoxin elicit dramatic hyperactivity in adult worms and schistosomula. Surprisingly, this response requires expression of a S. mansoni TRPA1-like orthologue (SmTRPA). Here, we show that capsaicin induces a rise in intracellular Ca2+ in mammalian cells expressing either SmTRPA or a S. haematobium TRPA1 orthologue (ShTRPA). We also test SmTRPA and ShTRPA responses to various TRPV1 and TRPA1 modulators. Interestingly, in contrast to SmTRPA, ShTRPA is not activated by the TRPA1 activator AITC (allyl isothiocyanate), nor do S. haematobium adult worms respond to this compound, a potentially intriguing species difference. Notably, 4-hydroxynonenal (4-HNE), a host-derived, inflammatory product that directly activates mammalian TRPA1, also activates both SmTRPA and ShTRPA. Our results point to parasite TRPA1-like channels which exhibit atypical, mixed TRPA1/TRPV1-like pharmacology, and which may also function to transduce endogenous host signals. Schistosomes are parasitic flatworms that infect hundreds of millions of people worldwide. They cause schistosomiasis, a disease with major consequences for human health and economic development. There is only a single drug available for treatment and control of this highly prevalent disease, and there is an urgent need for development of new treatments. TRP ion channels play key roles in sensory (and other) functions. One type of TRP channel, TRPV1, is activated by capsaicin, the active ingredient in hot peppers. However, schistosomes do not have any TRPV-like channels. Nonetheless, we previously showed that capsaicin and similar compounds induce dramatic hyperactivity in schistosomes, and that this response is abolished by suppressing expression of SmTRPA, a schistosome TRPA1-like channel. Mammalian TRPA1 channels are not sensitive to capsaicin. Here, we show that the SmTRPA channel itself responds to capsaicin, resulting in an influx of Ca2+ into cells. ShTRPA, a TRPA1-like channel from another schistosome, S. haematobium, is also sensitive to capsaicin. Thus, the pharmacology of schistosome TRPA1 channels apparently differs from that of host mammalian channels, a characteristic that could indicate mixed TRPA/TRPV functionality and might be exploitable for development of new antischistosomal drugs. Furthermore, we show that schistosome TRPA1-like channels are activated by host-derived compounds, perhaps indicating a mechanism by which the parasite can respond to host signals.
Collapse
Affiliation(s)
- Swarna Bais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Corbett T. Berry
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Xiaohong Liu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gordon Ruthel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Bruce D. Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Robert M. Greenberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
17
|
High throughput data analyses of the immune characteristics of Microtus fortis infected with Schistosoma japonicum. Sci Rep 2017; 7:11311. [PMID: 28900150 PMCID: PMC5595801 DOI: 10.1038/s41598-017-11532-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/11/2017] [Indexed: 12/11/2022] Open
Abstract
Microtus fortis exhibits natural resistance against Schistosoma japonicum, and the parasite cannot grow and develop in M. fortis. Extensive research has been carried out, however, the associated mechanism remains unclear. In the present study, we analysed the combined data obtained from a cytokine chip assay, transcriptome, and metabolome. The cytokine profile from C57BL/6 and M. fortis mice was assessed before and after infection. Several cytokines increased during the second and third week post-infection. Some transcripts related to cytokine genes and associated proteins were also highly expressed (i.e., Hgf, C3, and Lbp). The liver metabolism of M. fortis following infection with S. japonicum was assessed. We identified 25 different metabolites between the uninfected and infected M. fortis, and 22 different metabolites between infected M. fortis and C57BL/6 mice. The metabolomic pathways of these differential metabolites were then analysed with MetPA, revealing that they were involved in histidine metabolism, valine, leucine, and isoleucine biosyntheses, and lysine degradation. Thus, the elevated expression of these metabolites and pathways may promote the phagocytic function of the neutrophils and natural killer cell activity following TLR activation. These results provide novel insight into the resistance mechanism of M. fortis against S. japonicum.
Collapse
|
18
|
Abstract
Cystic and alveolar echinococcosis are severe chronic helminthic diseases caused by the cystic growth or the intrahepatic tumour-like growth of the metacestode of Echinococcus granulosus or Echinococcus multilocularis, respectively. Both parasites have evolved sophisticated strategies to escape host immune responses, mainly by manipulating and directing this immune response towards anergy and/or tolerance. Recent research studies have revealed a number of respective immunoregulatory mechanisms related to macrophages and dendritic cell as well as T cell activities (regulatory T cells, Tregs). A better understanding of this complex parasite-host relationship, and the elucidation of specific crucial events that lead to disease, represents targets towards the development of novel treatment strategies and options.
Collapse
|
19
|
Bais S, Greenberg RM. TRP channels in schistosomes. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2016; 6:335-342. [PMID: 27496302 PMCID: PMC5196486 DOI: 10.1016/j.ijpddr.2016.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/13/2016] [Accepted: 07/17/2016] [Indexed: 12/22/2022]
Abstract
Praziquantel (PZQ) is effectively the only drug currently available for treatment and control of schistosomiasis, a disease affecting hundreds of millions of people worldwide. Many anthelmintics, likely including PZQ, target ion channels, membrane protein complexes essential for normal functioning of the neuromusculature and other tissues. Despite this fact, only a few classes of parasitic helminth ion channels have been assessed for their pharmacological properties or for their roles in parasite physiology. One such overlooked group of ion channels is the transient receptor potential (TRP) channel superfamily. TRP channels share a common core structure, but are widely diverse in their activation mechanisms and ion selectivity. They are critical to transducing sensory signals, responding to a wide range of external stimuli. They are also involved in other functions, such as regulating intracellular calcium and organellar ion homeostasis and trafficking. Here, we review current literature on parasitic helminth TRP channels, focusing on those in schistosomes. We discuss the likely roles of these channels in sensory and locomotor activity, including the possible significance of a class of TRP channels (TRPV) that is absent in schistosomes. We also focus on evidence indicating that at least one schistosome TRP channel (SmTRPA) has atypical, TRPV1-like pharmacological sensitivities that could potentially be exploited for future therapeutic targeting. We provide an overview of transient receptor potential (TRP) channels in schistosomes and other parasitic helminths. TRP channels are important for sensory signaling, ion homeostasis, organellar trafficking, and a host of other functions. Very little work has been done on TRP channels in parasitic helminths. TRPV channels, found throughout the Metazoa, appear not to be present in parasitic platyhelminths. TRP channels in schistosomes appear to have atypical pharmacology, perhaps an entrée for therapeutic targeting.
Collapse
Affiliation(s)
- Swarna Bais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Robert M Greenberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Huang L, Beiting DP, Gebreselassie NG, Gagliardo LF, Ruyechan MC, Lee NA, Lee JJ, Appleton JA. Eosinophils and IL-4 Support Nematode Growth Coincident with an Innate Response to Tissue Injury. PLoS Pathog 2015; 11:e1005347. [PMID: 26720604 PMCID: PMC4697774 DOI: 10.1371/journal.ppat.1005347] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/28/2015] [Indexed: 01/30/2023] Open
Abstract
It has become increasingly clear that the functions of eosinophils extend beyond host defense and allergy to metabolism and tissue regeneration. These influences have strong potential to be relevant in worm infections in which eosinophils are prominent and parasites rely on the host for nutrients to support growth or reproduction. The aim of this study was to investigate the mechanism underlying the observation that eosinophils promote growth of Trichinella spiralis larvae in skeletal muscle. Our results indicate that IL-4 and eosinophils are necessary for normal larval growth and that eosinophils from IL-4 competent mice are sufficient to support growth. The eosinophil-mediated effect operates in the absence of adaptive immunity. Following invasion by newborn larvae, host gene expression in skeletal muscle was compatible with a regenerative response and a shift in the source of energy in infected tissue. The presence of eosinophils suppressed local inflammation while also influencing nutrient homeostasis in muscle. Redistribution of glucose transporter 4 (GLUT4) and phosphorylation of Akt were observed in nurse cells, consistent with enhancement of glucose uptake and glycogen storage by larvae that is known to occur. The data are consistent with a mechanism in which eosinophils promote larval growth by an IL-4 dependent mechanism that limits local interferon-driven responses that otherwise alter nutrient metabolism in infected muscle. Our findings document a novel interaction between parasite and host in which worms have evolved a strategy to co-opt an innate host cell response in a way that facilitates their growth. Eosinophilia is a central feature of Type 2 immunity induced by infection with parasitic worms. Although early work showed that eosinophils could adhere to and damage parasite larvae in vitro, a definitive role for eosinophils during worm infection remained elusive for many years. Recent studies uncovered novel roles of eosinophils in regulating metabolism and tissue remodeling, observations that suggest that eosinophils may function as regulatory cells and modulate such processes during helminth infections. We investigated the eosinophil-dependent mechanism that promotes growth of Trichinella spiralis larvae. We found that larval growth is independent of adaptive immunity and requires IL-4/STAT6 signaling in eosinophils. Gene transcription profiles in infected muscle suggested that eosinophils promote larval growth by suppressing local inflammation and enhancing nutrient uptake and metabolism. Our study provides new insights into the interactions between a parasitic worm and its host.
Collapse
Affiliation(s)
- Lu Huang
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nebiat G. Gebreselassie
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Lucille F. Gagliardo
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Maura C. Ruyechan
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Nancy A. Lee
- Department of Biochemistry and Molecular Biology, Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
| | - James J. Lee
- Department of Biochemistry and Molecular Biology, Division of Pulmonary Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, United States of America
| | - Judith A. Appleton
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
21
|
Yang J, Fu Z, Hong Y, Wu H, Jin Y, Zhu C, Li H, Lu K, Shi Y, Yuan C, Cheng G, Feng X, Liu J, Lin J. The Differential Expression of Immune Genes between Water Buffalo and Yellow Cattle Determines Species-Specific Susceptibility to Schistosoma japonicum Infection. PLoS One 2015; 10:e0130344. [PMID: 26125181 PMCID: PMC4488319 DOI: 10.1371/journal.pone.0130344] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 05/19/2015] [Indexed: 01/10/2023] Open
Abstract
Water buffalo are less susceptible to Schistosoma japonicum infection than yellow cattle. The factors that affect such differences in susceptibility remain unknown. A Bos taurus genome-wide gene chip was used to analyze gene expression profiles in the peripheral blood of water buffalo and yellow cattle pre- and post-infection with S. japonicum. This study showed that most of the identified differentially expressed genes (DEGs) between water buffalo and yellow cattle pre- and post-infection were involved in immune-related processes, and the expression level of immune genes was lower in water buffalo. The unique DEGs (390) in yellow cattle were mainly associated with inflammation pathways, while the unique DEGs (2,114) in water buffalo were mainly associated with immune-related factors. The 83 common DEGs may be the essential response genes during S. japonicum infection, the highest two gene ontology (GO) functions were associated with the regulation of fibrinolysis. The pathway enrichment analysis showed that the DEGs constituted similar immune-related pathways pre- and post-infection between the two hosts. This first analysis of the transcriptional profiles of natural hosts has enabled us to gain new insights into the mechanisms that govern their susceptibility or resistance to S. japonicum infections.
Collapse
Affiliation(s)
- Jianmei Yang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Zhiqiang Fu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Yang Hong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Haiwei Wu
- Department of Pathology and Laboratory Medicine, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Yamei Jin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Chuangang Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Hao Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Ke Lu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Yaojun Shi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Chunxiu Yuan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Guofeng Cheng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Xingang Feng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Jinming Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
| | - Jiaojiao Lin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People’s Republic of China
| |
Collapse
|
22
|
Abstract
Schistosomiasis is one of the most prevalent, insidious and serious of the tropical parasitic diseases. Although the effective anthelmintic drug, praziquantel, is widely available and cheap, it does not protect against re-infection, drug-resistant schistosome may evolve and mass drug administration programmes based around praziquantel are probably unsustainable long term. Whereas protective anti-schistosome vaccines are not yet available, the zoonotic nature of Schistosoma japonicum provides a novel approach for developing a transmission-blocking veterinary vaccine in domestic animals, especially bovines, which are major reservoir hosts, being responsible for up to 90% of environmental egg contamination in China and the Philippines. However, a greater knowledge of schistosome immunology is required to understand the processes associated with anti-schistosome protective immunity and to reinforce the rationale for vaccine development against schistosomiasis japonica. Importantly as well, improved diagnostic tests, with high specificity and sensitivity, which are simple, rapid and able to diagnose light S. japonicum infections, are required to determine the extent of transmission interruption and the complete elimination of schistosomiasis following control efforts. This article discusses aspects of the host immune response in schistosomiasis, the current status of vaccine development against S. japonicum and reviews approaches for diagnosing and detecting schistosome infections in mammalian hosts.
Collapse
|
23
|
von Moltke J, Locksley RM. I-L-C-2 it: type 2 immunity and group 2 innate lymphoid cells in homeostasis. Curr Opin Immunol 2014; 31:58-65. [PMID: 25458996 DOI: 10.1016/j.coi.2014.09.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/24/2014] [Accepted: 09/28/2014] [Indexed: 12/17/2022]
Abstract
Innate type 2 immune cells are activated in response to helminths, allergens, and certain types of proteases and particulates. Recently, innate type 2 immune pathways have also been implicated in protective host responses to homeostatic perturbations, such as metabolic dysfunction, atherosclerosis, and tissue injury. In this context, innate type 2 cytokines stimulate local tissues, recruit eosinophils, and alternatively activate macrophages to restore homeostasis. As the major source of innate interleukin (IL)-5 and IL-13, group 2 innate lymphoid cells are positioned to initiate and maintain homeostatic type 2 responses. The absence of exogenous stimuli in these processes implicates endogenous pathways in the activation of type 2 immunity and suggests an alternative evolutionary trajectory for type 2 immunity, apart from its role in response to helminths and allergens.
Collapse
Affiliation(s)
- Jakob von Moltke
- Departments of Microbiology and Immunology and Medicine, Howard Hughes Medical Institute, University of California, San Francisco, CA 94143, USA
| | - Richard M Locksley
- Departments of Microbiology and Immunology and Medicine, Howard Hughes Medical Institute, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
24
|
El Ridi R, Tallima H, Dalton JP, Donnelly S. Induction of protective immune responses against schistosomiasis using functionally active cysteine peptidases. Front Genet 2014; 5:119. [PMID: 24847355 PMCID: PMC4021144 DOI: 10.3389/fgene.2014.00119] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/17/2014] [Indexed: 01/08/2023] Open
Abstract
Each year schistosomiasis afflicts up to 600 million people in 74 tropical and sub-tropical countries, predominantly in the developing world. Yet we depend on a single drug, praziquantel, for its treatment and control. There is no vaccine available but one is urgently needed especially since praziquantel-resistant parasites are likely to emerge at some time in the future. The disease is caused by several worm species of the genus Schistosoma. These express several classes of papain-like cysteine peptidases, cathepsins B and L, in various tissues but particularly in their gastrodermis where they employ them as digestive enzymes. We have shown that sub-cutaneous injection of recombinant and functionally active Schistosoma mansoni cathepsin B1 (SmCB1), or a cathepsin L from a related parasite Fasciola hepatica (FhCL1), elicits highly significant protection (up to 73%) against an experimental challenge worm infection in murine models of schistosomiasis. The immune modulating properties of this subcutaneous injection can boost protection levels (up to 83%) when combined with other S. mansoni vaccine candidates, glyceraldehyde 3-phosphate dehydrogenase (SG3PDH) and peroxiredoxin (PRX-MAP). Here, we discuss these data in the context of the parasite's biology and development, and provide putative mechanism by which the native-like cysteine peptidase induce protective immune responses.
Collapse
Affiliation(s)
- Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University Cairo, Egypt
| | - Hatem Tallima
- Zoology Department, Faculty of Science, Cairo University Cairo, Egypt
| | - John P Dalton
- Medical Biology Centre, School of Biological Sciences, Queen's University Belfast Belfast, Northern Ireland
| | - Sheila Donnelly
- The i-three Institute, University of Technology at Sydney Ultimo, Sydney, NSW, Australia
| |
Collapse
|