1
|
Sultana M, Islam MA, Khairnar R, Kumar S. A guide to pathophysiology, signaling pathways, and preclinical models of liver fibrosis. Mol Cell Endocrinol 2025; 598:112448. [PMID: 39755140 DOI: 10.1016/j.mce.2024.112448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
Liver fibrosis is potentially a reversible form of liver disease that evolved from the early stage of liver scarring as a consequence of chronic liver injuries. Recurrent injuries in the liver without any appropriate medication cause the injuries to get intense and deeper, which gradually leads to the progression of irreversible cirrhosis or carcinoma. Unfortunately, there are no approved treatment strategies for reversing hepatic fibrosis, making it one of the significant risk factors for developing advanced liver disorders and liver disease-associated mortality. Consequently, the interpretation of the fundamental mechanisms, etiology, and pathogenesis is crucial for identifying the potential therapeutic target as well as evaluating novel anti-fibrotic therapy. However, despite innumerable research, the functional mechanism and disease characteristics are still obscure. To accelerate the understanding of underlying disease pathophysiology, molecular pathways and disease progression mechanism, it is crucial to mimic human liver disease through the formation of precise disease models. Although various in vitro and in vivo liver fibrotic models have emerged and developed already, a perfect clinical model replicating human liver diseases is yet to be established, which is one of the major challenges in discovering proper therapeutics. This review paper will shed light on pathophysiology, signaling pathways, preclinical models of liver fibrosis, and their limitations.
Collapse
Affiliation(s)
- Mehonaz Sultana
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Md Asrarul Islam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Rhema Khairnar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| |
Collapse
|
2
|
Alfred MO, Ochola L, Okeyo K, Bae E, Ogongo P, Odongo D, Njaanake K, Robinson JP. Application of microphysiological systems to unravel the mechanisms of schistosomiasis egg extravasation. Front Cell Infect Microbiol 2025; 15:1521265. [PMID: 40041145 PMCID: PMC11876127 DOI: 10.3389/fcimb.2025.1521265] [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: 11/01/2024] [Accepted: 01/23/2025] [Indexed: 03/06/2025] Open
Abstract
Despite decades of control efforts, the prevalence of schistosomiasis remains high in many endemic regions, posing significant challenges to global health. One of the key factors contributing to the persistence of the disease is the complex life cycle of the Schistosoma parasite, the causative agent, which involves multiple stages of development and intricate interactions with its mammalian hosts and snails. Among the various stages of the parasite lifecycle, the deposition of eggs and their migration through host tissues is significant, as they initiate the onset of the disease pathology by inducing inflammatory reactions and tissue damage. However, our understanding of the mechanisms underlying Schistosoma egg extravasation remains limited, hindering efforts to develop effective interventions. Microphysiological systems, particularly organ-on-a-chip systems, offer a promising approach to study this phenomenon in a controlled experimental setting because they allow the replication of physiological microenvironments in vitro. This review provides an overview of schistosomiasis, introduces the concept of organ-on-a-chip technology, and discusses its potential applications in the field of schistosomiasis research.
Collapse
Affiliation(s)
- Martin Omondi Alfred
- Department of Medical Microbiology and Immunology, University of Nairobi, Hospital Road, Kenyatta National Hospital, Nairobi, Kenya
- Department of Tropical and Infectious Diseases, Kenya Institute of Primate Research, Nairobi, Kenya
| | - Lucy Ochola
- Department of Tropical and Infectious Diseases, Kenya Institute of Primate Research, Nairobi, Kenya
| | - Kennedy Okeyo
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| | - Euiwon Bae
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| | - Paul Ogongo
- Department of Tropical and Infectious Diseases, Kenya Institute of Primate Research, Nairobi, Kenya
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - David Odongo
- Department of Medical Microbiology and Immunology, University of Nairobi, Hospital Road, Kenyatta National Hospital, Nairobi, Kenya
| | - Kariuki Njaanake
- Department of Medical Microbiology and Immunology, University of Nairobi, Hospital Road, Kenyatta National Hospital, Nairobi, Kenya
| | - J. Paul Robinson
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
3
|
Saleh HA, Ragab TIM, Sayed SSM. Influence of Chlorella vulgaris and Pediastrum boryanum extracts carried on nanocellulose on the immune response of Biomphalaria alexandrina snails against Schistosoma mansoni infection. Int J Biol Macromol 2025; 289:138584. [PMID: 39689788 DOI: 10.1016/j.ijbiomac.2024.138584] [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: 06/25/2024] [Revised: 11/20/2024] [Accepted: 12/07/2024] [Indexed: 12/19/2024]
Abstract
Continuous efforts are made to explore alternative methods for reducing Schistosomiasis. So, this study evaluated the effectiveness of Chlorella vulgaris and Pediastrum boryanum extracts carried on their nanocrystalline cellulose (NCC) as immunostimulants for Biomphalaria alexandrina snails against Schistosoma mansoni infection. The results showed that the lowest cercarial shedding/snail was 340 and 330 with 400 mg/L of C. vulgaris extract and NCC + C. vulgaris extract, respectively. Meanwhile, it was decreased with 200 mg/L of P. boryanum extract and 400 mg/L of NCC + P. boryanum extract, with high survival rates for all treatments. In addition, snails treated with 400 mg/L of C. vulgaris extract and 200 and 400 mg/L of NCC + C. vulgaris extract showed a significant decrease in the contents of hydrogen peroxide (0.639, 0.426, and 0.564 mM/L), respectively, compared to the control group (0.695 mM/L). Furthermore, C. vulgaris extracts induced numerous hemocytes around immature cercariae and sporocysts. P. boryanum extracts showed degenerated sporocysts surrounded by plenty of hemocytes. Nanocellulose carriers improved the delivery of microalgal components within B. alexandrina snails, causing the cercariae to lose their ideal shape. Gas chromatography-mass spectrometry (GC-MS) analysis revealed that C. vulgaris and P. boryanum extracts contained oleic acid, which improved the immunological response of snails, and glutamic acid and flavonoids acted as immune modulators and antioxidants. Ultimately, the toxicity assay indicated that NCC + C. vulgaris extract and NCC + P. boryanum extract were the safest for Artemia salina as a non-target aquatic organism.
Collapse
Affiliation(s)
- Hassnaa A Saleh
- Environmental Research Department, Theodor Bilharz Research Institute, Imbaba, Giza 12411, Egypt
| | - Tamer I M Ragab
- Chemistry of Natural and Microbial Products Department, National Research Centre, Dokki, Giza 12622, Egypt.
| | - Sara S M Sayed
- Environmental Research Department, Theodor Bilharz Research Institute, Imbaba, Giza 12411, Egypt
| |
Collapse
|
4
|
Zhu Y, Chen X, Zheng H, Ma Q, Chen K, Li H. Anti-Inflammatory Effects of Helminth-Derived Products: Potential Applications and Challenges in Diabetes Mellitus Management. J Inflamm Res 2024; 17:11789-11812. [PMID: 39749005 PMCID: PMC11694023 DOI: 10.2147/jir.s493374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025] Open
Abstract
The global rise in diabetes mellitus (DM), particularly type 2 diabetes (T2D), has become a major public health challenge. According to the "hygiene hypothesis", helminth infections may offer therapeutic benefits for DM. These infections are known to modulate immune responses, reduce inflammation, and improve insulin sensitivity. However, they also carry risks, such as malnutrition, anemia, and intestinal obstruction. Importantly, helminth excretory/secretory products, which include small molecules and proteins, have shown therapeutic potential in treating various inflammatory diseases with minimal side effects. This review explores the anti-inflammatory properties of helminth derivatives and their potential to alleviate chronic inflammation in both type 1 diabetes and T2D, highlighting their promise as future drug candidates. Additionally, it discusses the possible applications of these derivatives in DM management and the challenges involved in translating these findings into clinical practice.
Collapse
Affiliation(s)
- Yunhuan Zhu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xintong Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hezheng Zheng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Qiman Ma
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hongyu Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
- Ocean College, Beibu Gulf University, Qinzhou, Guangxi, People’s Republic of China
| |
Collapse
|
5
|
Chaponda MM, Lam HYP. Schistosoma antigens: A future clinical magic bullet for autoimmune diseases? Parasite 2024; 31:68. [PMID: 39481080 PMCID: PMC11527426 DOI: 10.1051/parasite/2024067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
Autoimmune diseases are characterized by dysregulated immunity against self-antigens. Current treatment of autoimmune diseases largely relies on suppressing host immunity to prevent excessive inflammation. Other immunotherapy options, such as cytokine or cell-targeted therapies, have also been used. However, most patients do not benefit from these therapies as recurrence of the disease usually occurs. Therefore, more effort is needed to find alternative immune therapeutics. Schistosoma infection has been a significant public health problem in most developing countries. Schistosoma parasites produce eggs that continuously secrete soluble egg antigen (SEA), which is a known modulator of host immune responses by enhancing Th2 immunity and alleviating outcomes of Th1 and Th17 responses. Recently, SEA has shown promise in treating autoimmune disorders due to their substantial immune-regulatory effects. Despite this interest, how these antigens modulate human immunity demonstrates only limited pieces of evidence, and whether there is potential for Schistosoma antigens in other diseases in the future remains an unsolved question. This review discusses how SEA modulates human immune responses and its potential for development as a novel immunotherapeutic for autoimmune diseases. We also discuss the immune modulatory effects of other non-SEA schistosome antigens at different stages of the parasite's life cycle.
Collapse
Affiliation(s)
- Mphatso Mayuni Chaponda
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University Hualien Taiwan
| | - Ho Yin Pekkle Lam
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University Hualien Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University Hualien Taiwan
- Institute of Medical Science, Tzu Chi University Hualien Taiwan
| |
Collapse
|
6
|
Zhong H, Guan G, Jin Y. Roles of helminth extracellular vesicle-derived let-7 in host-parasite crosstalk. Front Immunol 2024; 15:1449495. [PMID: 39530086 PMCID: PMC11551607 DOI: 10.3389/fimmu.2024.1449495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/06/2024] [Indexed: 11/16/2024] Open
Abstract
Helminth infections are a major public health problem as they can cause long-term chronic infections in their hosts for which there is no effective vaccine. During the long-term interaction between helminths and their hosts, helminth-derived extracellular vesicles (EVs) can participate in host immunomodulatory processes by secreting bioactive molecules (BMAs). Growing data suggests that microRNAs (miRNAs) in helminth EVs have a significant impact on the host's immune system. The let-7 family is highly conserved among helminth EVs and highly homologous in the host, and its function in host-parasite crosstalk may reflect active selection for compatibility with the host miRNA machinery. In-depth studies targeting this aspect may better elucidate the mechanism of parasite-host interactions. Hence, this review summarizes the current studies on the cross-species involvement of helminth EV-derived let-7 in host immune regulation and discusses the barriers to related research and potential applications of helminth EVs.
Collapse
Affiliation(s)
- Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Guiquan Guan
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
| | - Yamei Jin
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
7
|
Lu J, Zhang X, Dong P, Mei C, Yang Y, Yu C, Song L. Single-Cell Analysis Reveals the Cellular and Molecular Changes of Liver Injury and Fibrosis in Mice During the Progression of Schistosoma japonicum Infection. Curr Issues Mol Biol 2024; 46:11906-11926. [PMID: 39590301 PMCID: PMC11592686 DOI: 10.3390/cimb46110707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Schistosomiasis is a parasitic disease that poses a serious threat to human health. However, the pathogenic mechanism during the progression of Schistosoma japonicum infection remains unclear. In order to elucidate this mechanism, we used single-cell RNA sequencing (scRNA-seq) to investigate the transcriptome characteristics of the cellular (single-cell) landscape in the livers of mice infected with Schistosoma japonicum, which were divided into three groups: uninfected mice (0 week (w)), infected mice at 6 w post-infection (the acute phase), and infected mice at 10 w post-infection (the chronic phase). A total of 31,847 liver cells were included and clustered into 21 groups. The cells and T-cells had high heterogeneity in the liver during the progression of schistosome infection. The number and intensity of the intercellular interactions significantly increased at 6 w after infection but decreased at 10 w. The inflammatory signaling pathways chemoattractant cytokine ligand (CCL)5-chemokine C-C-motif receptor (CCR)5 between macrophages and T-cells were predominant at 6 w post-infection; the CCL6-CCR2 signaling pathway between macrophages was predominant at 10 w. The CD80 signaling pathway related to T-cell activation was increased at 6 w after infection, and increased expression of its receptor CD28 on the surfaces of CD4+ and CD8+ T-cells was confirmed by flow cytometry, suggesting an increase in their activation. In addition, scRNA-seq and quantitative reverse transcription polymerase chain reaction (qRT-PCR) confirmed that the intercellular communication between secretory phosphoprotein 1 (SPP1)-cluster of differentiation (CD44), insulin-like growth factor (IGF)-1-IGF1r and visfatin-insulin receptor (Insr) associated with bone metabolism and insulin metabolism was increased and enhanced in the liver at 6 w post-infection. Overall, we provide the comprehensive single-cell transcriptome landscape of the liver in mice during the progression of schistosome infection and delineate the key cellular and molecular events involved in schistosome infection-induced liver injury and fibrosis. The elevated CCL5-CCR5 and CCL6-CCR2 signaling pathways in the liver may be a drug target for liver injury and fibrosis caused by schistosome infection, respectively.
Collapse
Affiliation(s)
| | | | | | | | | | - Chuanxin Yu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China; (J.L.); (X.Z.); (P.D.); (C.M.); (Y.Y.)
| | - Lijun Song
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China; (J.L.); (X.Z.); (P.D.); (C.M.); (Y.Y.)
| |
Collapse
|
8
|
Afful P, Abotsi GK, Adu-Gyamfi CO, Benyem G, Katawa G, Kyei S, Arndts K, Ritter M, Asare KK. Schistosomiasis-Microbiota Interactions: A Systematic Review and Meta-Analysis. Pathogens 2024; 13:906. [PMID: 39452777 PMCID: PMC11510367 DOI: 10.3390/pathogens13100906] [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: 09/06/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
INTRODUCTION Schistosomiasis, a tropical disease affecting humans and animals, affected 251.4 million people in 2021. Schistosoma mansoni, S. haematobium, S. intercalatum, and S. japonicum are primary human schistosomes, causing tissue damage, granulomas, ulceration, hemorrhage, and opportunistic pathogen entry. The gut and urinary tract microbiota significantly impact a host's susceptibility to schistosomiasis, disrupting microbial balance; however, this relationship is not well understood. This systematic review and meta-analysis explores the intricate relationship between schistosomiasis and the host's microbiota, providing crucial insights into disease pathogenesis and management. METHODS This systematic review used PRISMA guidelines to identify peer-reviewed articles on schistosomiasis and its interactions with the host microbiome, using multiple databases and Google Scholar, providing a robust dataset for analysis. The study utilized Meta-Mar v3.5.1; descriptive tests, random-effects models, and subgroups were analyzed for the interaction between Schistosomiasis and the microbiome. Forest plots, Cochran's Q test, and Higgins' inconsistency statistic (I2) were used to assess heterogeneity. RESULTS The human Schistosoma species were observed to be associated with various bacterial species isolated from blood, stool, urine, sputum, skin, and vaginal or cervical samples. A meta-analysis of the interaction between schistosomiasis and the host microbiome, based on 31 studies, showed 29,784 observations and 5871 events. The pooled estimates indicated a significant association between schistosomiasis and changes in the microbiome of infected individuals. There was considerable heterogeneity with variance effect sizes (p < 0.0001). Subgroup analysis of Schistosoma species demonstrated that S. haematobium was the most significant contributor to the overall heterogeneity, accounting for 62.1% (p < 0.01). S. mansoni contributed 13.0% (p = 0.02), and the coinfection of S. haematobium and S. mansoni accounted for 16.8% of the heterogeneity (p < 0.01), contributing to the variability seen in the pooled analysis. Similarly, praziquantel treatment (RR = 1.68, 95% CI: 1.07-2.64) showed high heterogeneity (Chi2 = 71.42, df = 11, p < 0.01) and also indicated that Schistosoma infections in males (RR = 1.46, 95% CI: 0.00 to 551.30) and females (RR = 2.09, 95% CI: 0.24 to 18.31) have a higher risk of altering the host microbiome. CONCLUSIONS Schistosomiasis significantly disrupts the host microbiota across various bodily sites, leading to increased susceptibility to different bacterial taxa such as E. coli, Klebsiella, Proteus, Pseudomonas, Salmonella, Staphylococcus, Streptococcus, and Mycobacterium species (M. tuberculosis and M. leprae). This disruption enables these bacteria to produce toxic metabolites, which in turn cause inflammation and facilitate the progression of disease. The impact of schistosomiasis on the vaginal microbiome underscores the necessity for gender-specific approaches to treatment and prevention. Effective management of female genital schistosomiasis (FGS) requires addressing both the parasitic infection and the resulting microbiome imbalances. Additionally, praziquantel-treated individuals have different microbiome compositions compared to individuals with no praziquantel treatment. This suggests that combining praziquantel treatment with probiotics could potentially decrease the disease severity caused by an altered microbiome.
Collapse
Affiliation(s)
- Philip Afful
- Biomedical and Clinical Research Centre, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana; (P.A.); (G.K.A.); (C.O.A.-G.); (G.B.); (S.K.)
| | - Godwin Kwami Abotsi
- Biomedical and Clinical Research Centre, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana; (P.A.); (G.K.A.); (C.O.A.-G.); (G.B.); (S.K.)
| | - Czarina Owusua Adu-Gyamfi
- Biomedical and Clinical Research Centre, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana; (P.A.); (G.K.A.); (C.O.A.-G.); (G.B.); (S.K.)
| | - George Benyem
- Biomedical and Clinical Research Centre, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana; (P.A.); (G.K.A.); (C.O.A.-G.); (G.B.); (S.K.)
| | - Gnatoulma Katawa
- Unité de Recherche en Immunologie et Immunomodulation (UR2IM)/Laboratoire de Microbiologie et de Contrôle de Qualité des Denrées Alimentaires (LAMICODA), Ecole Supérieure des Techniques Biologiques et Alimentaires, Université de Lomé, Lomé, Togo;
| | - Samuel Kyei
- Biomedical and Clinical Research Centre, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana; (P.A.); (G.K.A.); (C.O.A.-G.); (G.B.); (S.K.)
- Department of Optometry and Vision Science, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Kathrin Arndts
- Institute for Medical Microbiology, Immunology, and Parasitology (IMMIP), University Hospital Bonn (UKB), 53127 Bonn, Germany;
- German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site Bonn, 53127 Bonn, Germany
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology, and Parasitology (IMMIP), University Hospital Bonn (UKB), 53127 Bonn, Germany;
- German-West African Centre for Global Health and Pandemic Prevention (G-WAC), Partner Site Bonn, 53127 Bonn, Germany
| | - Kwame Kumi Asare
- Biomedical and Clinical Research Centre, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana; (P.A.); (G.K.A.); (C.O.A.-G.); (G.B.); (S.K.)
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| |
Collapse
|
9
|
Gibbs LC, Oviedo JM, Ondigo BN, Fairfax KC. Maternal Helminth Infection Causes Dysfunctional B Cell Development in Male Offspring. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1157-1169. [PMID: 39185897 PMCID: PMC11537230 DOI: 10.4049/jimmunol.2400158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/31/2024] [Indexed: 08/27/2024]
Abstract
Infections during pregnancy are known to trigger alterations in offspring immunity, often leading to increased disease susceptibility. Maternal helminth infections correlate with lower Ab titers to certain childhood immunizations and putative decreased vaccine efficacy. The mechanisms that underlie how maternal infection blunts offspring humoral responses are unclear. Using our murine model of maternal schistosomiasis, we found that maternal helminth infection decreases the germinal center response of all offspring to tetanus immunization. However, only male offspring have defects in memory B cell and long-lived plasma cell generation. We found this sex-specific aberration begins during B cell development within the bone marrow via alteration of the IL-7 niche and persists throughout antigenic activation in the germinal center in the periphery. Critically, these defects in males are cell intrinsic, persisting following adoptive transfer to control male pups. Together, these data show that maternal infections can alter both the bone marrow microenvironment and the development of B lymphocytes in a sex-specific manner. This study correlates maternal infection induced defects in early life B cell development with ineffective Ab responses after vaccination.
Collapse
Affiliation(s)
- Lisa C. Gibbs
- Department of Pathology, University of Utah; Salt Lake City, UT, United States
| | - Juan M. Oviedo
- Department of Pathology, University of Utah; Salt Lake City, UT, United States
| | | | - Keke C. Fairfax
- Department of Pathology, University of Utah; Salt Lake City, UT, United States
| |
Collapse
|
10
|
Chatterji T, Khanna N, Alghamdi S, Bhagat T, Gupta N, Alkurbi MO, Sen M, Alghamdi SM, Bamagous GA, Sahoo DK, Patel A, Kumar P, Yadav VK. A Recent Advance in the Diagnosis, Treatment, and Vaccine Development for Human Schistosomiasis. Trop Med Infect Dis 2024; 9:243. [PMID: 39453270 PMCID: PMC11511416 DOI: 10.3390/tropicalmed9100243] [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: 08/28/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Schistosomiasis, which affects a large number of people worldwide, is among the most overlooked parasitic diseases. The disease is mainly prevalent in sub-Saharan Africa, southeast Asian countries, and South America due to the lack of adequate sanitation. The disease is mainly associated with poor hygiene, sanitation, and contaminated water, so it is also known as a disease of poverty. Three Schistosoma species (S. mansoni, S. japonicum, and S. haematobium) cause significant human infections. Co-infections with Schistosoma and other parasites are widely common. All these parasites may cause intestinal or urogenital schistosomiasis, where the disease may be categorized into the acute, sensitized, and chronic phases. The disease is more prevalent among school children, which may cause anemia and reduce development. Chronic infections frequently cause significant liver, intestinal, and bladder damage. Women exposed to contaminated water while performing normal duties like washing clothes might acquire urogenital schistosomiasis (UGS), which can cause tissue damage and raise the risk of blood-borne disease transmission, including human immunodeficiency virus (HIV) transmission. Praziquantel (PZQ) is the World Health Organization (WHO)-prescribed treatment for individuals who are known to be infected, but it does not prevent further re-infections with larval worms. Vaccine development and new molecular-based diagnosis techniques have promised to be a reliable approach to the diagnosis and prevention of schistosomiasis. The current review emphasizes the recent advancement in the diagnosis of schistosomiasis by molecular techniques and the treatment of schistosomiasis by combined and alternative regimes of drugs. Moreover, this review has also focused on the recent outbreak of schistosomiasis, the development of vaccines, and their clinical trials.
Collapse
Affiliation(s)
- Tanushri Chatterji
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), Adhyatmik Nagar, NH-09, Ghaziabad 201015, Uttar Pradesh, India;
| | - Namrata Khanna
- Department of Biochemistry, M A Rangoonwala College of Dental Sciences and Research Centre, 2390-B, K.B. Hidayatullah Road, Azam Campus, Camp, Pune 411001, Maharashtra, India;
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (S.A.); (M.O.A.)
| | - Tanya Bhagat
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), Adhyatmik Nagar, NH-09, Ghaziabad 201015, Uttar Pradesh, India;
| | - Nishant Gupta
- Engineering Department, River Engineering Pvt Ltd., Toy City, Ecotech–III, Greater Noida 201306, Uttar Pradesh, India;
| | - Mohammad Othman Alkurbi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (S.A.); (M.O.A.)
| | - Manodeep Sen
- Department of Microbiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Vibhuti Khand, Gomti Nagar, Lucknow 226010, Uttar Pradesh, India;
| | - Saeed Mardy Alghamdi
- Respiratory Care Program, Clinical Technology Department, Faculty of Applied Medical Science, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Ghazi A. Bamagous
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan 384265, Gujarat, India;
| | - Pankaj Kumar
- Department of Environmental Science, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, Gujarat, India;
| | - Virendra Kumar Yadav
- Marwadi University Research Center, Department of Microbiology, Faculty of Sciences, Marwadi University, Rajkot 360003, Gujarat, India
| |
Collapse
|
11
|
Zhong H, Dong B, Zhu D, Fu Z, Liu J, Guan G, Jin Y. Schistosoma japonicumsja-let-7 Inhibits the Growth of Hepatocellular Carcinoma Cells via Cross-Species Regulation of Col1α2. Genes (Basel) 2024; 15:1165. [PMID: 39336756 PMCID: PMC11431810 DOI: 10.3390/genes15091165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Liver fibrosis, a critical precursor to hepatocellular carcinoma (HCC), results from chronic liver injury and significantly contributes to HCC progression. Schistosomiasis, a neglected tropical disease, is known to cause liver fibrosis; however, this process can be modulated by schistosome-derived miRNAs. Previous studies from our laboratory have demonstrated that Schistosoma japonicum extracellular vesicles (EVs) deliver sja-let-7 to hepatic stellate cells, leading to the inhibition of Col1α2 expression and alleviation of liver fibrosis. Given the well-documented antifibrotic and antiproliferative properties of the let-7 miRNA family, this study aims to preliminarily investigate the effects of the sja-let-7/Col1α2 axis on BALB/c mice and HCC cell line SNU387, providing a basis for the potential application of parasite-derived molecules in HCC therapy. In the present study, schistosome-induced fibrosis datasets were analyzed to identify the role of Col1α2 in extracellular matrix organization. Pan-cancer analysis revealed that Col1α2 is upregulated in various cancers, including HCC, with significant associations with immune cell infiltration and clinical parameters, highlighting its diagnostic importance. Functional assays demonstrated that transfection with sja-let-7 mimics significantly reduced Col1α2 expression, inhibited HCC cell proliferation, migration, and colony formation. These findings suggest that sja-let-7, by targeting Col1α2, has the potential to serve as a therapeutic agent in HCC treatment. This study indicates the pivotal role of Col1α2 in liver fibrosis and HCC, and the promising therapeutic application of helminth-derived miRNAs.
Collapse
Affiliation(s)
- Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (Z.F.); (J.L.)
| | - Bowen Dong
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (Z.F.); (J.L.)
| | - Danlin Zhu
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (Z.F.); (J.L.)
| | - Zhiqiang Fu
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (Z.F.); (J.L.)
| | - Jinming Liu
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (Z.F.); (J.L.)
| | - Guiquan Guan
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou 730046, China;
| | - Yamei Jin
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (Z.F.); (J.L.)
| |
Collapse
|
12
|
Rogers M, Kamath S, McManus D, Jones M, Gordon C, Navarro S. Schistosoma excretory/secretory products: an untapped library of tolerogenic immunotherapeutics against food allergy. Clin Transl Immunology 2024; 13:e70001. [PMID: 39221178 PMCID: PMC11359118 DOI: 10.1002/cti2.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/18/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Food allergy (FA) is considered the 'second wave' of the allergy epidemic in developed countries after asthma and allergic rhinitis with a steadily growing burden of 40%. The absence of early childhood pathogen stimulation embodied by the hygiene hypothesis is one explanation, and in particular, the eradication of parasitic helminths could be at play. Infections with parasites Schistosoma spp. have been found to have a negative correlation with allergic diseases. Schistosomes induce regulatory responses to evade immune detection and ensure their long-term survival. This is achieved via excretory/secretory (E/S) products, consisting of proteins, lipids, metabolites, nucleic acids and extracellular vesicles, representing an untapped therapeutic avenue for the treatment of FA without the unpleasant side-effects and risks associated with live infection. Schistosome-derived immunotherapeutic development is in its infancy and novel discoveries are heavily technology dependent; thus, it is essential to better understand how newly identified molecules interact with host immune systems to ensure safety and successful translation. This review will outline the identified Schistosoma-derived E/S products at all life cycle stages and discuss known mechanisms of action and their ability to suppress FA.
Collapse
Affiliation(s)
- Madeleine Rogers
- Faculty of MedicineUniversity of QueenslandBrisbaneQLDAustralia
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Sandip Kamath
- Institute of Pathophysiology and Allergy ResearchMedical University of ViennaViennaAustria
- Australian Institute of Tropical Health and MedicineJames Cook UniversityTownsvilleQLDAustralia
| | - Donald McManus
- Faculty of MedicineUniversity of QueenslandBrisbaneQLDAustralia
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Malcolm Jones
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
- Faculty of Science, School of Veterinary ScienceUniversity of QueenslandGattonQLDAustralia
| | - Catherine Gordon
- Faculty of MedicineUniversity of QueenslandBrisbaneQLDAustralia
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Severine Navarro
- Faculty of MedicineUniversity of QueenslandBrisbaneQLDAustralia
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
- Centre for Childhood Nutrition Research, Faculty of HealthQueensland University of TechnologyBrisbaneQLDAustralia
| |
Collapse
|
13
|
Wu P, Li W, Xie Y, Guan W, Yang S, Li J, Zhao Y. An insight into the gut microbiota after Schistosoma japonicum eggs immunization in an experimental ulcerative colitis model. FASEB J 2024; 38:e23721. [PMID: 38822662 DOI: 10.1096/fj.202302576rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/06/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
Schistosome infection and schistosome-derived products have been implicated in the prevention and alleviation of inflammatory bowel disease by manipulating the host immune response, whereas the role of gut microbiota in this protective effect remains poorly understood. In this study, we found that the intraperitoneal immunization with Schistosoma japonicum eggs prior to dextran sulfate sodium (DSS) application significantly ameliorated the symptoms of DSS-induced acute colitis, which was characterized by higher body weight, lower disease activity index score and macroscopic inflammatory scores. We demonstrated that the immunomodulatory effects of S. japonicum eggs were accompanied by an influence on gut microbiota composition, abundance, and diversity, which increased the abundance of genus Turicibacter, family Erysipelotrichaceae, phylum Firmicutes, and decreased the abundance of genus Odoribacter, family Marinifilaceae, order Bacteroidales, class Bacteroidia, phylum Bacteroidota. In addition, Lactobacillus was identified as a biomarker that distinguishes healthy control mice from DSS-induced colitis mice. The present study revealed the importance of the gut microbiota in S. japonicum eggs exerting protective effects in an experimental ulcerative colitis (UC) model, providing an alternative strategy for the discovery of UC prevention and treatment drugs.
Collapse
Affiliation(s)
- Peng Wu
- Department of Human Parasitology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Wenhao Li
- Department of Human Parasitology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Yiting Xie
- Department of Human Parasitology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Wei Guan
- Department of Human Parasitology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Shuguo Yang
- Department of Human Parasitology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Jian Li
- Department of Human Parasitology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Yanqing Zhao
- Department of Human Parasitology, School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
14
|
Golenser J, Birman I, Gold D. Considering ivermectin for treatment of schistosomiasis. Parasitol Res 2024; 123:180. [PMID: 38592544 PMCID: PMC11003930 DOI: 10.1007/s00436-024-08178-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/29/2024] [Indexed: 04/10/2024]
Abstract
Because of recent reports of praziquantel resistance in schistosome infections, there have been suggestions to employ ivermectin as a possible alternative, especially as its chemical composition is different from that of praziquantel, so cross-resistance is not expected. In order to ascertain possible damage and elimination of worms, we used ivermectin by oral gavage in infected mice, at a high dose (30.1 mg/kg, bordering toxicity). We also tested the efficacy of the drug at various times postinfection (PI), to check on possible effect on young and mature stages of the parasites. Thus, we treated mice on days 21 and 22 or on days 41 and 42 and even on days 21, 22, 41, and 42 PI. None of the treatment regimens resulted in cure rates or signs of lessened pathology in the mice. We also compared the effect of ivermectin to that of artemisone, an artemisinin derivative which had served us in the past as an effective anti-schistosome drug, and there was a stark difference in the artemisone's efficacy compared to that of ivermectin; while ivermectin was not effective, artemisone eliminated most of the worms, prevented egg production and granulomatous inflammatory response. We assume that the reported lack of activity of ivermectin, in comparison with praziquantel and artemisinins, originates from the difference in their mode of action. In wake of our results, we suggest that ivermectin is not a suitable drug for treatment of schistosomiasis.
Collapse
Affiliation(s)
- Jacob Golenser
- Department of Microbiology and Molecular Genetics, Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University - Hadassah Medical Center, Jerusalem, Israel.
| | - Ida Birman
- Department of Microbiology and Molecular Genetics, Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University - Hadassah Medical Center, Jerusalem, Israel
| | - Daniel Gold
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
15
|
Zhong H, Dong B, Zhu D, Li H, Lu K, Fu Z, Liu J, Jin Y. Sja-Let-7 Attenuates Carbon Tetrachloride-Induced Liver Fibrosis in a Mouse Model via Col1α2. BIOLOGY 2023; 12:1465. [PMID: 38132291 PMCID: PMC10740823 DOI: 10.3390/biology12121465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023]
Abstract
Liver fibrosis (LF) is a chronic progressive disease with no definitive treatment. The aim of this study was to assess helminth-derived molecules as potential therapeutic targets to prevent or reverse LF. A mouse model of carbon tetrachloride (CCL4)-induced LF was established and sja-let-7 was overexpressed by treatment with a miRNA agomir once per week. After four weeks, serum biochemistry, hepatic hydroxyproline content measurements, liver histology, mRNA expression profiling of fibrotic markers, the dual-luciferase reporter assay, and fluorescence in situ hybridization (FISH) were performed. Administration of the sja-let-7 agomir markedly ameliorated hepatosplenomegaly and reduced the liver hydroxyproline content. Liver histological analysis showed significant reductions in collagen deposition in the sja-let-7 agomir-treated mice. Additionally, the mRNA levels of both pro-fibrotic markers and pro-inflammatory cytokines were diminished after treatment. Furthermore, the dual-luciferase reporter assay and FISH identified the α2 chain of collagen type 1 (Col1α2) as the direct target of sja-let-7. Accordingly, the progression of LF was attenuated by targeting Col1α2 and the TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (H.L.); (K.L.); (Z.F.); (J.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Bowen Dong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (H.L.); (K.L.); (Z.F.); (J.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Danlin Zhu
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (H.L.); (K.L.); (Z.F.); (J.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Hao Li
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (H.L.); (K.L.); (Z.F.); (J.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Ke Lu
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (H.L.); (K.L.); (Z.F.); (J.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Zhiqiang Fu
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (H.L.); (K.L.); (Z.F.); (J.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jinming Liu
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (H.L.); (K.L.); (Z.F.); (J.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yamei Jin
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.Z.); (B.D.); (D.Z.); (H.L.); (K.L.); (Z.F.); (J.L.)
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| |
Collapse
|
16
|
Ma ZR, Li ZL, Zhang N, Lu B, Li XW, Huang YH, Nouhoum D, Liu XS, Xiao KC, Cai LT, Xu SR, Yang XXO, Huang SQ, Wu X. Inhibition of GSDMD-mediated pyroptosis triggered by Trichinella spiralis intervention contributes to the alleviation of DSS-induced ulcerative colitis in mice. Parasit Vectors 2023; 16:280. [PMID: 37580819 PMCID: PMC10424392 DOI: 10.1186/s13071-023-05857-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/30/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is increasing worldwide. Although there is currently no completely curative treatment, helminthic therapy shows certain therapeutic potential for UC. Many studies have found that Trichinella spiralis (T.s) has a protective effect on UC, but the specific mechanism is still unclear. METHODS Balb/c mice drank dextran sulfate sodium (DSS) to induce acute colitis and then were treated with T.s. In vitro experiments, the LPS combination with ATP was used to induce the pyroptosis model, followed by intervention with crude protein from T.s (T.s cp). Additionally, the pyroptosis agonist of NSC or the pyroptosis inhibitor vx-765 was added to intervene to explore the role of pyroptosis in DSS-induced acute colitis. The degree of pyroptosis was evaluated by western blot, qPCR and IHC, etc., in vivo and in vitro. RESULTS T.s intervention significantly inhibited NLRP3 inflammasome activation and GSDMD-mediated pyroptosis by downregulating the expression of pyroptosis-related signatures in vitro (cellular inflammatory model) and in vivo (DSS-induced UC mice model). Furthermore, blockade of GSDMD-mediated pyroptosis by the caspase-1 inhibitor vx-765 has a similar therapeutic effect on DSS-induced UC mice with T.s intervention, thus indicating that T.s intervention alleviated DSS-induced UC in mice by inhibiting GSDMD-mediated pyroptosis. CONCLUSION This study showed that T.s could alleviate the pathological severity UC via GSDMD-mediated pyroptosis, and it provides new insight into the mechanistic study and application of helminths in treating colitis.
Collapse
Affiliation(s)
- Zhen-Rong Ma
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Zhuo-Lin Li
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Ni Zhang
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Bin Lu
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xuan-Wu Li
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Ye-Hong Huang
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Dibo Nouhoum
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xian-Shu Liu
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Ke-Chun Xiao
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Li-Ting Cai
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Shao-Rui Xu
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xue-Xian O Yang
- Department of Molecular & Genetic and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Shuai-Qin Huang
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China.
| | - Xiang Wu
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
17
|
Johnson D, Jiang W. Infectious diseases, autoantibodies, and autoimmunity. J Autoimmun 2023; 137:102962. [PMID: 36470769 PMCID: PMC10235211 DOI: 10.1016/j.jaut.2022.102962] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 11/27/2022] [Indexed: 12/04/2022]
Abstract
Infections are known to trigger flares of autoimmune diseases in humans and serve as an inciting cause of autoimmunity in animals. Evidence suggests a causative role of infections in triggering antigen-specific autoimmunity, previous thought mainly through antigen mimicry. However, an infection can induce bystander autoreactive T and B cell polyclonal activation, believed to result in non-pathogenic and pathogenic autoimmune responses. Lastly, epitope spreading in autoimmunity is a mechanism of epitope changes of autoreactive cells induced by infection, promoting the targeting of additional self-epitopes. This review highlights recent research findings, emphasizes infection-mediated autoimmune responses, and discusses the possible mechanisms involved.
Collapse
Affiliation(s)
- Douglas Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA; Divison of Infectious Disease, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
18
|
Zhong H, Jin Y. Single-sex schistosomiasis: a mini review. Front Immunol 2023; 14:1158805. [PMID: 37153566 PMCID: PMC10154636 DOI: 10.3389/fimmu.2023.1158805] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by dioecious blood flukes of the genus Schistosoma and second to malaria as a parasitic disease with significant socio-economic impacts. Mating is essential for maturation of male and female schistosomes and for females to lay of eggs, which are responsible for the pathogenesis and propagation of the life cycle beyond the mammalian host. Single-sex schistosomes, which do not produce viable eggs without mating, have been overlooked given the symptomatic paucity of the single-sex schistosomiasis and limited diagnostic toolkit. Besides, single-sex schistosomes are less sensitive to praziquantel. Therefore, these issues should be considered to achieve the elimination of this infection disease. The aim of this review is to summarize current progress in research of single-sex schistosomes and host-parasite interactions.
Collapse
Affiliation(s)
- Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yamei Jin
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- *Correspondence: Yamei Jin,
| |
Collapse
|
19
|
Hou X, Zhu F, Zheng W, Jacques ML, Huang J, Guan F, Lei J. Protective effect of Schistosoma japonicum eggs on TNBS-induced colitis is associated with regulating Treg/Th17 balance and reprogramming glycolipid metabolism in mice. Front Cell Infect Microbiol 2022; 12:1028899. [PMID: 36304936 PMCID: PMC9592807 DOI: 10.3389/fcimb.2022.1028899] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/28/2022] [Indexed: 11/28/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) have been classified as modern refractory diseases. However, safe, well-tolerated, and effective treatments for IBDs are still lacking. Therefore, there is an urgent need to develop novel therapeutic targets with fewer undesirable adverse reactions. A growing body of research has shown that infection with live helminths or exposure to defined helminth-derived components can downregulate pathogenic inflammation due to their immunoregulatory ability. Here we were to explore the protective role of Schistosoma japonicum eggs on murine experimental colitis caused by trinitrobenzene sulfonic acid (TNBS) and the underlying mechanism. Frequencies of splenic Treg and Th17 cells were detected by flow cytometry. Protein and mRNA expressions of Foxp3 and RORγt were investigated by Western Blot and quantitative real-time polymerase chain reaction (qPCR), respectively. Concentrations of transforming growth factor-beta1 (TGF-β1), interleukin-10 (IL-10) and IL-17A were assessed with ELISA. Expression levels of genes related to glycolipid metabolism were measured with qPCR. The results showed that pre-exposure to S. japonicum eggs contributed to the relief of colitis in the TNBS model, evidenced by improved body weight loss, reversing spleen enlargement and colon shortening, and decreased histology scores. Compared with the TNBS group, the TNBS+Egg group had increased Treg immune response, accompanied by decreased Th17 immune response, leading to the reconstruction of Treg/Th17 balance. In addition, a ratio of Treg/Th17 was correlated negatively with the histological scores in the experiment groups. Furthermore, the regulation of Treg/Th17 balance by S. japonicum eggs was associated with inhibiting the glycolysis pathway and lipogenesis, along with promoting fatty acid oxidation in the TNBS+Egg group. These data indicate that S. japonicum eggs have a protective effect against TNBS-induced colitis, which is related to restoring Treg/Th17 balance and regulating glucose and lipid metabolism.
Collapse
Affiliation(s)
- Xiao Hou
- Department of Clinical Laboratory, The General Hospital of Central Theater Command, The People's Liberation Army, Wuhan, China
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feifan Zhu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenwen Zheng
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Muziazia Lupemba Jacques
- Department of Parasitology, Kinshasa Institute of Medical, Kinshasa, Democratic Republic of the Congo
| | - Jin Huang
- Department of Clinical Laboratory, Wuhan Pu’ai Hospital, Wuhan, China
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jiahui Lei,
| |
Collapse
|
20
|
Liu X, Jiang Y, Ye J, Wang X. Helminth infection and helminth-derived products: A novel therapeutic option for non-alcoholic fatty liver disease. Front Immunol 2022; 13:999412. [PMID: 36263053 PMCID: PMC9573989 DOI: 10.3389/fimmu.2022.999412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely related to obesity, diabetes, and metabolic syndrome (MetS), and it has become the most common chronic liver disease. Helminths have co-evolved with humans, inducing multiple immunomodulatory mechanisms to modulate the host's immune system. By using their immunomodulatory ability, helminths and their products exhibit protection against various autoimmune and inflammatory diseases, including obesity, diabetes, and MetS, which are closely associated with NAFLD. Here, we review the pathogenesis of NAFLD from abnormal glycolipid metabolism, inflammation, and gut dysbiosis. Correspondingly, helminths and their products can treat or relieve these NAFLD-related diseases, including obesity, diabetes, and MetS, by promoting glycolipid metabolism homeostasis, regulating inflammation, and restoring the balance of gut microbiota. Considering that a large number of clinical trials have been carried out on helminths and their products for the treatment of inflammatory diseases with promising results, the treatment of NAFLD and obesity-related diseases by helminths is also a novel direction and strategy.
Collapse
Affiliation(s)
- Xi Liu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuyun Jiang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jixian Ye
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Nuclear Medicine and Institute of Digestive Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
21
|
Zhong H, Jin Y. Multifunctional Roles of MicroRNAs in Schistosomiasis. Front Microbiol 2022; 13:925386. [PMID: 35756064 PMCID: PMC9218868 DOI: 10.3389/fmicb.2022.925386] [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: 04/21/2022] [Accepted: 05/20/2022] [Indexed: 11/22/2022] Open
Abstract
Schistosomiasis is a parasitic disease that is caused by helminths of the genus Schistosoma. The dioecious schistosomes mate and lay eggs after undergoing a complex life cycle. Schistosome eggs are mostly responsible for the transmission of schistosomiasis and chronic fibrotic disease induced by egg antigens is the main cause of the high mortality rate. Currently, chemotherapy with praziquantel (PZQ) is the only effective treatment against schistosomiasis, although the potential of drug resistance remains a concern. Hence, there is an urgent demand for new and effective strategies to combat schistosomiasis, which is the second most prevalent parasitic disease after malaria. MicroRNAs (miRNAs) are small non-coding RNAs that play pivotal regulatory roles in many organisms, including the development and sexual maturation of schistosomes. Thus, miRNAs are potential targets for treatment of schistosomiasis. Moreover, miRNAs can serve as multifunctional “nano-tools” for cross-species delivery in order to regulate host-parasite interactions. In this review, the multifunctional roles of miRNAs in the growth and development of schistosomes are discussed. The various regulatory functions of host-derived and worm-derived miRNAs on the progression of schistosomiasis are also thoroughly addressed, especially the promotional and inhibitory effects on schistosome-induced liver fibrosis. Additionally, the potential of miRNAs as biomarkers for the diagnosis and treatment of schistosomiasis is considered.
Collapse
Affiliation(s)
- Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China.,Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yamei Jin
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China.,Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
22
|
Cai P, Mu Y, McManus DP. The Fight Against Severe COVID-19: Can Parasitic Worms Contribute? Front Immunol 2022; 13:849465. [PMID: 35222441 PMCID: PMC8874793 DOI: 10.3389/fimmu.2022.849465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 12/15/2022] Open
Affiliation(s)
- Pengfei Cai
- *Correspondence: Pengfei Cai, .; Donald P. McManus,
| | | | | |
Collapse
|
23
|
Gaballah EM, Morita K, Shimizu S, Elhenawy AA, Nabih N, Elsawey AM, Abdel-Mageed SA, Osada Y. Non-lethal rodent malarial infection prevents collagen-induced arthritis in mice via anti-arthritic immunomodulation. Parasite Immunol 2021; 44:e12901. [PMID: 34931316 DOI: 10.1111/pim.12901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/29/2022]
Abstract
AIMS Immunomodulatory effects of parasitic infections on the outcomes of allergic or autoimmune disorders have been addressed in many experimental studies. We examined the effects of Plasmodium yoelii 17X NL (Py) infection on collagen-induced arthritis (CIA). METHODS AND RESULTS Male DBA/1J mice were immunized with bovine type II collagen (IIC). Py inoculation was induced at three different time points (1, 4 weeks after or 4 weeks before the immunization). Only the inoculation at 4 weeks after IIC immunization significantly inhibited arthritis development. Non-malarial anaemia induced by phenylhydrazine hydrochloride (PHZ) did not affect arthritis development. In the infected mice, anti-IIC IgG levels were transiently reduced. In addition, splenic production of pro-arthritic cytokines (IL-17 and TNF-α) and IFN-γ decreased, whereas IL-10 production increased. Flow cytometric analysis clarified that the main IL-10 producers in Py-infected mice had the CD4+ CD25- Foxp3- phenotype, presumably Tr1 cells. CONCLUSION We demonstrated that experimental malarial infection alleviated autoimmune arthritis via immunomodulation, suggesting the importance of malaria in the hygiene hypothesis and the significance of searching for therapeutic immunomodulatory molecules from malarial parasites.
Collapse
Affiliation(s)
- Eman M Gaballah
- Department of Medical Parasitology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Department of Parasitology and Immunology, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan.,Department of Medical Parasitology, Faculty of Medicine, Damietta University, Damietta, Egypt
| | - Kentaro Morita
- Department of Parasitology and Immunology, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Shoichi Shimizu
- Department of Parasitology and Immunology, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Abeer A Elhenawy
- Department of Medical Parasitology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nairmen Nabih
- Department of Medical Parasitology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Aliaa M Elsawey
- Department of Medical Parasitology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Salama A Abdel-Mageed
- Department of Medical Parasitology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Yoshio Osada
- Department of Parasitology and Immunology, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|