1
|
Song M, Zhang C, Yang S, Lu J, Sun T, Li H, Tang L, Dai K, Liu C, Meng H, Wang J. Animal healer for refractory diseases: Myth or reality? Heliyon 2024; 10:e33056. [PMID: 39027544 PMCID: PMC11255451 DOI: 10.1016/j.heliyon.2024.e33056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/20/2024] Open
Abstract
A vast amount of knowledge has been acquired through human activities such as farming, hunting, and fishing. Throughout history, humans have utilized living creatures for disease treatment, relying on the natural world's healing powers. The special "healers" may be able to treat patients via the power of nature. However, there was no systematic introduction or summary of these treatments. Therefore, we conducted a literature review based on PubMed, Google Scholar, Web of Science, Scopus, CNKI and WanFang DATA. Here, we defined this unique method as "animal healer" and six common kinds of animal healers were reviewed. These are fish therapy, pet therapy, worm therapy, leech therapy, maggot therapy, and bee therapy. According to the different characteristics of healers, treatment methods mainly included bite, parasitism, contact and communication. With the advantages of green and effectiveness, animal healers have great therapy potential against a variety of refractory diseases. The main purpose of this review is to draw people's attention to animal healer, promote it to become a possible clinical treatment strategy, and make further exploration in species cultivation, mechanism research, animal welfare, standard setting, safety evaluation and other aspects. In the future, animal healers will play an increasingly important role in medicine and hopefully solve more medical problems and dilemmas.
Collapse
Affiliation(s)
- Mingzhi Song
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changru Zhang
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Simengge Yang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jialin Lu
- College of Humanities and Social Sciences, Dalian Medical University, Dalian, China
| | - Tianze Sun
- Department of Orthopaedics, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Heyue Li
- Department of Obstetrics and Gynecology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Tang
- Department of Orthopedic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kerong Dai
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Southwest Jiaotong University College of Medicine, Chengdu, China
| | - Chaozong Liu
- Institute of Orthopaedic & Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, London, United Kingdom
| | - He Meng
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Jinwu Wang
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Southwest Jiaotong University College of Medicine, Chengdu, China
| |
Collapse
|
2
|
Colomb F, Ogunkanbi A, Jamwal A, Dong B, Maizels RM, Finney CAM, Wasmuth JD, Higgins MK, McSorley HJ. IL-33-binding HpARI family homologues with divergent effects in suppressing or enhancing type 2 immune responses. Infect Immun 2024; 92:e0039523. [PMID: 38294241 PMCID: PMC10929406 DOI: 10.1128/iai.00395-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/27/2023] [Indexed: 02/01/2024] Open
Abstract
HpARI is an immunomodulatory protein secreted by the intestinal nematode Heligmosomoides polygyrus bakeri, which binds and blocks IL-33. Here, we find that the H. polygyrus bakeri genome contains three HpARI family members and that these have different effects on IL-33-dependent responses in vitro and in vivo, with HpARI1+2 suppressing and HpARI3 amplifying these responses. All HpARIs have sub-nanomolar affinity for mouse IL-33; however, HpARI3 does not block IL-33-ST2 interactions. Instead, HpARI3 stabilizes IL-33, increasing the half-life of the cytokine and amplifying responses to it in vivo. Together, these data show that H. polygyrus bakeri secretes a family of HpARI proteins with both overlapping and distinct functions, comprising a complex immunomodulatory arsenal of host-targeted proteins.
Collapse
Affiliation(s)
- Florent Colomb
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Adefunke Ogunkanbi
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
- Department of Biology, University of York, York, United Kingdom
| | - Abhishek Jamwal
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery and Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford, United Kingdom
| | - Beverly Dong
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Research Training Network, University of Calgary, Calgary, Alberta, Canada
| | - Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Constance A. M. Finney
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
- Host-Parasite Interactions Research Training Network, University of Calgary, Calgary, Alberta, Canada
| | - James D. Wasmuth
- Host-Parasite Interactions Research Training Network, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Matthew K. Higgins
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery and Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford, United Kingdom
| | - Henry J. McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
3
|
van Dinther M, Cunningham KT, Singh SP, White MPJ, Campion T, Ciancia C, van Veelen PA, de Ru AH, González-Prieto R, Mukundan A, Byeon CH, Staggers SR, Hinck CS, Hinck AP, Dijke PT, Maizels RM. CD44 acts as a coreceptor for cell-specific enhancement of signaling and regulatory T cell induction by TGM1, a parasite TGF-β mimic. Proc Natl Acad Sci U S A 2023; 120:e2302370120. [PMID: 37590410 PMCID: PMC10450677 DOI: 10.1073/pnas.2302370120] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/25/2023] [Indexed: 08/19/2023] Open
Abstract
Long-lived parasites evade host immunity through highly evolved molecular strategies. The murine intestinal helminth, Heligmosomoides polygyrus, down-modulates the host immune system through release of an immunosuppressive TGF-β mimic, TGM1, which is a divergent member of the CCP (Sushi) protein family. TGM1 comprises 5 domains, of which domains 1-3 (D1/2/3) bind mammalian TGF-β receptors, acting on T cells to induce Foxp3+ regulatory T cells; however, the roles of domains 4 and 5 (D4/5) remain unknown. We noted that truncated TGM1, lacking D4/5, showed reduced potency. Combination of D1/2/3 and D4/5 as separate proteins did not alter potency, suggesting that a physical linkage is required and that these domains do not deliver an independent signal. Coprecipitation from cells treated with biotinylated D4/5, followed by mass spectrometry, identified the cell surface protein CD44 as a coreceptor for TGM1. Both full-length and D4/5 bound strongly to a range of primary cells and cell lines, to a greater degree than D1/2/3 alone, although some cell lines did not respond to TGM1. Ectopic expression of CD44 in nonresponding cells conferred responsiveness, while genetic depletion of CD44 abolished enhancement by D4/5 and ablated the ability of full-length TGM1 to bind to cell surfaces. Moreover, CD44-deficient T cells showed attenuated induction of Foxp3 by full-length TGM1, to levels similar to those induced by D1/2/3. Hence, a parasite protein known to bind two host cytokine receptor subunits has evolved a third receptor specificity, which serves to raise the avidity and cell type-specific potency of TGF-β signaling in mammalian cells.
Collapse
Affiliation(s)
- Maarten van Dinther
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Kyle T. Cunningham
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Shashi Prakash Singh
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Madeleine P. J. White
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Tiffany Campion
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Claire Ciancia
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Peter A. van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden2333 ZC, The Netherlands
| | - Arnoud H. de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden2333 ZC, The Netherlands
| | - Román González-Prieto
- Andalusian Center for Molecular Biology and Regenerative Medicine, Universidad de Sevilla - CSIC - Universidad Pablo de Olavide, 41092Sevilla, Spain
- Department of Cell Biology, Faculty of Biology, University of Sevilla, 41013Sevilla, Spain
| | - Ananya Mukundan
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15260
| | - Chang-Hyeock Byeon
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15260
| | - Sophia R. Staggers
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15260
| | - Cynthia S. Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15260
| | - Andrew P. Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15260
| | - Peter ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden2300 RC, The Netherlands
| | - Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, GlasgowG12 8TA, United Kingdom
| |
Collapse
|
4
|
Gao X, Mao C, Zheng T, Xu X, Luo X, Zhang S, Liu J, Wang X, Chen X, Dong L. Schistosoma japonicum-derived peptide SJMHE1 ameliorates allergic symptoms and responses in mice with allergic rhinitis. Front Cell Infect Microbiol 2023; 13:1143950. [PMID: 37346033 PMCID: PMC10279851 DOI: 10.3389/fcimb.2023.1143950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/22/2023] [Indexed: 06/23/2023] Open
Abstract
Helminth derived excretory/secretory molecules have shown efficacy in the treatment of allergic asthma in mice, but their roles in allergic rhinitis (AR) are little known. In this study, we aimed to determine the intervention effect of SJMHE1, a Schistosoma japonicum derived small molecular peptide, on ovalbumin (OVA)-induced AR mice and investigate its possible mechanism. AR was induced in BALB/c mice, following which the mice were treated with phosphate-buffered saline (PBS), OVA323-339 and SJMHE1 respectively. SJMHE1 treatment improved clinical symptoms (rubbing and sneezing), suppressed infiltrates of inflammatory cells and eosinophils in nasal mucosa, modulated the production of type-2 (IL-4 and IL-13) and anti-inflammatory (IL-10) cytokines in the nasal lavage fluids (NLF), spleen, and serum. To investigate the underlying mechanism, fluorescein isothiocyanate (FITC)-labeled SJMHE1 was subcutaneously injected into AR mice, and we found that the FITC-SJMHE1 could accumulate in spleen, but not in nasal mucosa. FITC-SJMHE1 mainly bound to CD19 positive cells (B cells), and the SJMHE1 treatment significantly increased the proportion of regulatory B cells (Bregs) and B10 cells, along with the enhancement of PR domain containing protein 1 (Prdm1) protein levels. SJMHE1 may alleviate AR by upregulating Bregs, and has great potential as a new avenue for the AR treatment.
Collapse
Affiliation(s)
- Xuerong Gao
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chaoming Mao
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tingting Zheng
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiaowei Xu
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xinkai Luo
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shan Zhang
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiameng Liu
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xuefeng Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiaojun Chen
- Department of Pathogen Biology & Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liyang Dong
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
5
|
Karo-Atar D, Gregorieff A, King IL. Dangerous liaisons: how helminths manipulate the intestinal epithelium. Trends Parasitol 2023; 39:414-422. [PMID: 37076358 DOI: 10.1016/j.pt.2023.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 04/21/2023]
Abstract
Intestinal helminths remain highly pervasive throughout the animal kingdom by modulating multiple aspects of the host immune response. The intestinal epithelium functions as a physical barrier as well as a sentinel innate immune tissue with the ability to sense and respond to infectious agents. Although helminths form intimate interactions with the epithelium, comprehensive knowledge about host-helminth interactions at this dynamic interface is lacking. In addition, little is known about the ability of helminths to directly shape the fate of this barrier tissue. Here, we review the diverse pathways by which helminths regulate the epithelium and highlight the emerging field of direct helminth regulation of intestinal stem cell (ISC) fate and function.
Collapse
Affiliation(s)
- Danielle Karo-Atar
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada; McGill Regenerative Medicine Network, Montreal, Quebec, Canada.
| | - Alex Gregorieff
- McGill Regenerative Medicine Network, Montreal, Quebec, Canada; Department of Pathology, McGill University and Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Irah L King
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada; McGill Regenerative Medicine Network, Montreal, Quebec, Canada; McGill Centre for Microbiome Research, Montreal, Quebec, Canada.
| |
Collapse
|
6
|
Yang Y, Ding X, Chen F, Wu X, Chen Y, Zhang Q, Cao J, Wang J, Dai Y. Inhibition Effects of Nippostrongylus brasiliensis and Its Derivatives against Atherosclerosis in ApoE-/- Mice through Anti-Inflammatory Response. Pathogens 2022; 11:pathogens11101208. [PMID: 36297265 PMCID: PMC9610917 DOI: 10.3390/pathogens11101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis (AS) is a dominant and growing cause of death and disability worldwide that involves inflammation from its inception to the emergence of complications. Studies have demonstrated that intervention with helminth infections or derived products could modulate the host immune response and effectively prevent or mitigate the onset and progression of inflammation-related diseases. Therefore, to understand the molecular mechanisms underlying the development of atherosclerosis, we intervened in ApoE-/- mice maintained on a high-fat diet with Nippostrongylus brasiliensis (N. brasiliensis) infection and immunized with its derived products. We found that N. brasiliensis infection and its derived proteins had suitable protective effects both in the initial and progressive stages of atherosclerosis, effectively reducing aortic arch plaque areas and liver lipid contents and downregulating serum LDL levels, which may be associated with the significant upregulation of serum anti-inflammatory cytokines (IL-10 and IL-4) and the down-regulation of proinflammatory cytokines (TNF-α and IFN-γ) in the serum. In conclusion, these data highlighted the effective regulatory role of N. brasiliensis and its derived proteins in the development and progression of atherosclerosis. This could provide a promising new avenue for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yougui Yang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Xin Ding
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Fuzhong Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaomin Wu
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
- Microbiological Laboratory, Anhui Provincial Center for Disease Control and Prevention, Hefei 230601, China
| | - Yuying Chen
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Qiang Zhang
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Jun Cao
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Junhong Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Correspondence: (J.W.); (Y.D.)
| | - Yang Dai
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
- Correspondence: (J.W.); (Y.D.)
| |
Collapse
|
7
|
Immunization with EmCRT-Induced Protective Immunity against Echinococcus multilocularis Infection in BALB/c Mice. Trop Med Infect Dis 2022; 7:tropicalmed7100279. [PMID: 36288020 PMCID: PMC9610995 DOI: 10.3390/tropicalmed7100279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Alveolar echinococcosis (AE) is a severe parasitic zoonosis caused by the larval stage of Echinococcus multilocularis. The identification of the antigens eliciting acquired immunity during infection is important for vaccine development against Echinococcus infection. Here, we identified that E. multilocularis calreticulin (EmCRT), a ubiquitous protein with a Ca2+-binding ability, could be recognized by the sera of mice infected with E. multilocularis. The native EmCRT was expressed on the surface of E. multilocularis larvae as well as in the secreted products of metacestode vesicles and protoscoleces (PSCs). The coding DNA for EmCRT was cloned from the mRNA of the E. multilocularis metacestode vesicles and a recombinant EmCRT protein (rEmCRT) was expressed in E. coli. Mice immunized with soluble rEmCRT formulated with Freund’s adjuvant (FA) produced a 43.16% larval vesicle weight reduction against the challenge of E. multilocularis PSCs compared to those that received the PBS control associated with a high titer of IgG, IgG1 and IgG2a antibody responses as well as high levels of Th1 cytokines (IFN-γ and IL-2) and Th2 cytokines (IL-4, IL-5 and IL-10), produced by splenocytes. Our results suggest that EmCRT is an immunodominant protein secreted by E. multilocularis larvae and a vaccine candidate that induces partial protective immunity in vaccinated mice against Echinococcus infection.
Collapse
|
8
|
Wen Z, Zhang Y, Feng J, Aimulajiang K, Aleem MT, Lu M, Xu L, Song X, Li X, Yan R. Excretory/secretory proteins inhibit host immune responses by downregulating the TLR4/NF-κB/MAPKs signaling pathway: A possible mechanism of immune evasion in parasitic nematode Haemonchus contortus. Front Immunol 2022; 13:1013159. [PMID: 36238295 PMCID: PMC9551057 DOI: 10.3389/fimmu.2022.1013159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Haemonchus contortus is an important parasitic nematode of ruminants. Previous studies showed that H. contortus escape the immunity through complex mechanisms, including releasing excretory/secretory proteins (ESPs) to modulate the host immune response. However, the detailed mechanism through which H. contortus excretory/secretory proteins (HcESPs) promote immune evasion remains unknown. In the present study, we demonstrated that HcESPs inhibit the adaptive immune response of goats including downregulation of immune cell antigen presentation, upregulation of immune checkpoint molecules, activation of the STAT3/PD-L1 pathway, and activation of immunosuppressive regulatory T (Treg) cells. Furthermore, HcESPs reversed the LPS-induced upregulation of pro-inflammatory mediators in PBMCs by inhibiting the TLR4/NF-κB/MAPKs/NLRP3 signaling pathway. Our study provides a better understanding of the evasion mechanisms for H. contortus, which could be helpful in providing an alternative way to prevent the infection of this parasite.
Collapse
Affiliation(s)
- Zhaohai Wen
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yue Zhang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiajun Feng
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Kalibixiati Aimulajiang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Muhammad Tahir Aleem
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Mingmin Lu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Lixin Xu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaokai Song
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiangrui Li
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ruofeng Yan
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Ruofeng Yan,
| |
Collapse
|
9
|
Hodge SH, McSorley HJ. A Good Day for Helminths: how parasite‐derived GDH suppresses inflammatory responses. EMBO Rep 2022; 23:e55054. [PMID: 35357756 PMCID: PMC9066059 DOI: 10.15252/embr.202255054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 11/28/2022] Open
Abstract
Parasitic helminths are often associated with immunoregulation, which allows them to survive in their hosts in the face of type 2 immune responses. They achieve this feat through the secretion of multiple immunomodulatory factors. In this issue of EMBO Reports, Prodjinotho et al show that the parasitic cestode Taenia solium induces regulatory T‐cell responses in mice and humans through the release of the metabolic enzyme Glutamate dehydrogenase (GDH), which may be a conserved pathway of immunoregulation in many helminths (Prodjinotho et al, 2022).
Collapse
Affiliation(s)
- Suzanne H Hodge
- Division of Cell Signalling and Immunology University of Dundee Dundee UK
| | - Henry J McSorley
- Division of Cell Signalling and Immunology University of Dundee Dundee UK
| |
Collapse
|
10
|
Shields VE, Cooper J. Use of helminth therapy for management of ulcerative colitis and Crohn's disease: a systematic review. Parasitology 2022; 149:145-154. [PMID: 34579797 PMCID: PMC10090591 DOI: 10.1017/s0031182021001670] [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: 08/12/2021] [Revised: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 11/06/2022]
Abstract
The incidence rate of inflammatory bowel diseases is increasing in developed countries. As such there is an increasing demand for new therapies. The aim of this systematic review was to investigate whether there is evidence to support the use of helminth therapy for the management of Crohn's disease and ulcerative colitis. Four databases (PubMed, Embase, Medline and the Cochrane Central Register of Control Trials) were searched for primary evidence in the form of clinical studies. Nine studies were suitable for inclusion: five double-blind randomized control trials and four open-label studies. This review divided the results of the studies into two categories: (a) the efficacy of helminth therapy and (b) the safety of helminth therapy. Results regarding the efficacy were mixed and a conclusive answer could not be reached, as there was not enough evidence to rule out a placebo effect. More research is needed, particularly studies with control groups to address the possibility of a placebo effect. Despite this, all nine studies concluded helminth therapy was safe and tolerable, and therefore there is currently no evidence against further exploration of this treatment option.
Collapse
Affiliation(s)
| | - Jan Cooper
- Warwick Medical School, University of Warwick, CoventryCV4 7AL, UK
| |
Collapse
|
11
|
Zhang Z, Liu J, Mao C, Zhang S, Wang X, Dong L. SJMHE1 protects against excessive iodine-induced pyroptosis in human thyroid follicular epithelial cells through a toll-like receptor 2-dependent pathway. Int J Med Sci 2022; 19:631-639. [PMID: 35582426 PMCID: PMC9108411 DOI: 10.7150/ijms.66167] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
To elucidate the effect of Schistosoma japonicum peptide (SJMHE1) on pyroptosis in thyroid follicular epithelial cells (TFCs) induced by excessive iodine and the potential mechanism, the effects of SJMHE1 were investigated in NaI-treated Nthy-ori 3-1 cells; and the involvement of the ROS/MAPK/NF-κB signaling pathways in these effects was evaluated by employing CCK-8 assays, flow cytometry, ELISA, and Western blotting experiments. We found that SJMHE1 significantly reduced NLRP3, N-terminus of gasdermin D (GSDMD-N) and cleaved caspase-1 (C-caspase-1) expression, and decreased IL-1β secretion in TFCs. SJMHE1 also markedly reduced reactive oxygen species (ROS) production, and decreased the phosphorylation levels of MAPK and NF-κB pathway members. Moreover, blocking of the Toll-like receptor 2 significantly impaired SJMHE1-mediated protection from excessive iodine-induced pyroptosis in TFCs. Therefore, our results suggested a protective role of SJMHE1 in excessive iodine-induced pyroptosis in TFCs, which might be attributed to its suppression for ROS/MAPK/NF-κB signaling pathway by binding of SJMHE1 with TLR2.
Collapse
Affiliation(s)
- Zhu Zhang
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, P. R. China.,Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 212000, P. R. China
| | - Jiameng Liu
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, P. R. China
| | - Chaoming Mao
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, P. R. China
| | - Shan Zhang
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, P. R. China
| | - Xuefeng Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, P. R. China
| | - Liyang Dong
- Department of Nuclear Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, P. R. China
| |
Collapse
|
12
|
Chen Y, Zhang M, Ding X, Yang Y, Chen Y, Zhang Q, Fan Y, Dai Y, Wang J. Mining Anti-Inflammation Molecules From Nippostrongylus brasiliensis-Derived Products Through the Metabolomics Approach. Front Cell Infect Microbiol 2021; 11:781132. [PMID: 34858883 PMCID: PMC8632049 DOI: 10.3389/fcimb.2021.781132] [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: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 01/13/2023] Open
Abstract
Hookworm is one type of soil-transmitted helminth, which could exert an anti-inflammatory effect in human or animal host, which provides a beneficial possibility for the discovery of inflammatory-related disease interventions. The identification of hookworm-derived anti-inflammatory molecules is urgently needed for future translational research. The emergence of metabolomics has become a powerful approach to comprehensively characterize metabolic alterations in recent times. Herein, excretory and secretory products (ESPs) were collected from cultured adult worm, while small intestinal contents were obtained from Nippostrongylus brasiliensis (N. brasiliensis, Nb)-infected mice. Through ultra-high-performance liquid chromatography coupled with mass spectrometry (UHPLC-MS) platform, metabolomics analysis was used to explore the identification of anti-inflammatory molecules. Out of 45 differential metabolites that were discovered from ESPs, 10 of them showed potential anti-inflammatory properties, which could be subclassed into amino acids, furanocoumarins, linear diarylheptanoids, gamma butyrolactones, and alpha-keto acids. In terms of intestinal contents that were derived from N. brasiliensis-infected mice, 14 out of 301 differential metabolites were discovered to demonstrate anti-inflammatory effects, with possible subclassification into amino acids, benzylisoquinolines, quaternary ammonium salts, pyrimidines, pregnane steroids, purines, biphenyls, and glycerophosphocholines. Furthermore, nine of the differential metabolites appeared both in ESPs and infected intestinal contents, wherein four were proven to show anti-inflammation properties, namely, L-glutamine, glutamine (Gln), pyruvate, and alanine-Gln (Ala-Gln). In summary, we have provided a method for the identification and analysis of parasite-derived molecules with potential anti-inflammatory properties in the present study. This array of anti-inflammatory metabolites could provide clues for future evaluation and translational study of these anti-inflammatory molecules.
Collapse
Affiliation(s)
- Yuying Chen
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Mingming Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Ding
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Yougui Yang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Yujia Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang Zhang
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Yinwen Fan
- Department of Cardiology, The Friendship Hospital of Ili Kazakh Autonomous Prefecture Ili & Jiangsu Joint Institute of Health, Ili, China
| | - Yang Dai
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Key Laboratory of Jiangsu Province on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Junhong Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Cardiology, The Friendship Hospital of Ili Kazakh Autonomous Prefecture Ili & Jiangsu Joint Institute of Health, Ili, China
| |
Collapse
|
13
|
Silveira GO, Coelho HS, Amaral MS, Verjovski-Almeida S. Long non-coding RNAs as possible therapeutic targets in protozoa, and in Schistosoma and other helminths. Parasitol Res 2021; 121:1091-1115. [PMID: 34859292 DOI: 10.1007/s00436-021-07384-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/14/2021] [Indexed: 12/26/2022]
Abstract
Long non-coding RNAs (lncRNAs) emerged in the past 20 years due to massive amounts of scientific data regarding transcriptomic analyses. They have been implicated in a plethora of cellular processes in higher eukaryotes. However, little is known about lncRNA possible involvement in parasitic diseases, with most studies only detecting their presence in parasites of human medical importance. Here, we review the progress on lncRNA studies and their functions in protozoans and helminths. In addition, we show an example of knockdown of one lncRNA in Schistosoma mansoni, SmLINC156349, which led to in vitro parasite adhesion, motility, and pairing impairment, with a 20% decrease in parasite viability and 33% reduction in female oviposition. Other observed phenotypes were a decrease in the proliferation rate of both male and female worms and their gonads, and reduced female lipid and vitelline droplets that are markers for well-developed vitellaria. Impairment of female worms' vitellaria in SmLINC156349-silenced worms led to egg development deficiency. All those results demonstrate the great potential of the tools and methods to characterize lncRNAs as potential new therapeutic targets. Further, we discuss the challenges and limitations of current methods for studying lncRNAs in parasites and possible solutions to overcome them, and we highlight the future directions of this exciting field.
Collapse
Affiliation(s)
- Gilbert O Silveira
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, SP, 05503-900, Brazil.,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Helena S Coelho
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | - Murilo S Amaral
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, SP, 05503-900, Brazil.
| | - Sergio Verjovski-Almeida
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, SP, 05503-900, Brazil. .,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil.
| |
Collapse
|
14
|
Chauhan A, Sharma A, Tripathi JK, Sun Y, Sukumran P, Singh BB, Mishra BB, Sharma J. Helminth derived factors inhibit neutrophil extracellular trap formation and inflammation in bacterial peritonitis. Sci Rep 2021; 11:12718. [PMID: 34135384 PMCID: PMC8209178 DOI: 10.1038/s41598-021-92001-9] [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: 03/16/2021] [Accepted: 06/03/2021] [Indexed: 01/18/2023] Open
Abstract
Despite their protective antimicrobial function, neutrophil extracellular traps (NETs) have been implicated in propagation of inflammatory responses in several disease conditions including sepsis. Highly diffusible exogenous ROS produced under such inflammatory conditions, can induce exuberant NETs, thus making inhibition of NETs desirable in inflammatory diseases. Here we report that helminth parasite excretory/secretory factors termed as parasitic ligands (PL) inhibit ROS-induced NETs by blocking the activation of nonselective calcium permeable channel Transient Receptor Potential Melastatin 2 (TRPM2). Therapeutic implication of PL mediated blockage of NET formation was tested in preclinical model of septic peritonitis, where PL treatment regulated neutrophil cell death modalities including NET formation and mitigated neutrophil mediated inflammatory response. This translated into improved survival and reduced systemic and local bacterial load in infected mice. Overall, our results posit PL as an important biological regulator of neutrophil functions with implications to a variety of inflammatory diseases including peritonitis.
Collapse
Affiliation(s)
- Arun Chauhan
- Department of Biomedical Sciences, The University of North Dakota School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND, 58202-9037, USA
| | - Atul Sharma
- Department of Biomedical Sciences, The University of North Dakota School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND, 58202-9037, USA
- Department of Critical Care, Division of Anesthesiology, Critical Care and Pain Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 110, Houston, TX, 77030-4009, USA
| | - Jitendra K Tripathi
- Department of Biomedical Sciences, The University of North Dakota School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND, 58202-9037, USA
- Department of Geriatrics, The University of North Dakota School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND, 58202-9037, USA
| | - Yuyang Sun
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Pramod Sukumran
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Brij B Singh
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Bibhuti B Mishra
- Department of Biomedical Sciences, The University of North Dakota School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND, 58202-9037, USA.
| | - Jyotika Sharma
- Department of Biomedical Sciences, The University of North Dakota School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND, 58202-9037, USA.
- Department of Critical Care, Division of Anesthesiology, Critical Care and Pain Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 110, Houston, TX, 77030-4009, USA.
| |
Collapse
|