1
|
Choudhuri S, Garg NJ. Hepatocyte Nuclear Factor 4 Alpha: A Key Regulator of Liver Disease Pathology and Haemostatic Disorders. Liver Int 2025; 45:e16245. [PMID: 40387433 DOI: 10.1111/liv.16245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE Hepatocyte nuclear factor 4 alpha (HNF4α) is a master regulator of hepatocyte differentiation in fetal and adult liver and exerts its transcriptional role in determining physiological functions of the liver. The objective of this review is to address the current knowledge of molecular mechanisms involved in HNF4α regulation in multiple aspects of liver disease pathogenesis. METHODS Based on available literature, this review summarises the current state of knowledge onthe mechanism of HNF4α dysregulation, and the role of HNF4α activity inregulating early to advanced stages of various liver diseases. RESULTS Patients with deranged HNF4α expression are at higher risk for the development of liver diseases such as viral hepatitis, alcoholic/nonalcoholic fatty liver disease, hepatocellular carcinoma, and haematological disorders such as coagulopathy and bleeding disorders. DISCUSSION HNF4α interactions with nuclear receptors and target genes promote liver disease pathology by regulating various metabolic pathways. The strong correlation between deranged HNF4α expression and the severity of liver diseases suggests that targeting HNF4α expression can offer potential therapeutic strategy in the prevention of liver disease pathology and haemostatic disorders.
Collapse
Affiliation(s)
- Subhadip Choudhuri
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas, USA
- Institute for Human Infections and Immunity (IHII), University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas, USA
- Institute for Human Infections and Immunity (IHII), University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| |
Collapse
|
2
|
Abulizi A, Tuergan T, Shalayiadang P, Zhang C, Zhang R, Jiang T, Guo Q, Wang H, Li L, Lin R, Shao Y, Aji T. Hepatic alveolar echinococcosis infection induces a decrease in NK cell function through high expression of NKG2A in patients. Front Immunol 2025; 16:1563248. [PMID: 40421027 PMCID: PMC12104054 DOI: 10.3389/fimmu.2025.1563248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Echinococcus multilocularis larval tapeworm infection in humans is considered a serious public health issue. The immune interaction between parasites and their hosts is extremely important. NK cells are known innate immune cells that play important roles during infection and tumour progression. However, the possible role of NK cells in hepatic alveolar echinococcosis is not completely clear. In this study, we investigated the functional decrease in NK cells in hepatic alveolar echinococcosis (AE) patients. Methods Using human liver tissue samples from 10 patients with hepatic AE, flow cytometry was used to detect the expression of NKG2A molecules on the surface of NK cells, and the correlations between NKG2A+ expression and lesion size, alkaline phosphatase (ALP) levels in close lesion tissues (CLTs) and distal lesion tissues (DLTs) in the liver, and the secretion of functional molecules by NKG2A+ NK cells were analysed. Results The expression of NKG2A on CD56dim and CD56bright NK cells in DLTs and CLTs revealed that the percentage of NKG2A+CD56dim NK cells in CLTs was significantly greater than that in DLTs. There was a negative correlation between the expression of NKG2A on NK cells in the CLT and alkaline phosphatase. Additionally, we analysed IFN-γ, TNF-α, granzyme B, and perforin production in NK cells. There was a significant reduction in IFN-γ production in CLTs compared with DLTs. There is a negative correlation between IFN-γ production levels and NKG2A expression in NK cells from the CLT. The capacity of NKG2A+ NK cells from CLT regions to produce IFN-γ and granzyme B was also significantly decreased. In contrast, the perforin level produced by NKG2A+ NK cells was much greater than that produced by NKG2A- NK cells. We also analysed the correlation between the ratio of the NKG2A expression area in CLT and DLT tissues and the PET-CT value and found a positive correlation between NKG2A expression and the PET-CT value. Conclusion The increased expression of NKG2A in NK cells induced a reduction in IFN-γ production, and the increased expression of NKG2A may improve lesion activity and fibrosis, which may be helpful for treating hepatic alveolar echinococcosis via immunity.
Collapse
Affiliation(s)
- Abuduaini Abulizi
- Hepatobiliary & Hydatid Disease Department, Digestive & Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Talaiti Tuergan
- Hepatobiliary & Hydatid Disease Department, Digestive & Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Paizula Shalayiadang
- Hepatobiliary & Hydatid Disease Department, Digestive & Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chuanshan Zhang
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- State Key Laboratory on Pathogenesis Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, China
| | - Ruiqing Zhang
- Hepatobiliary & Hydatid Disease Department, Digestive & Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tiemin Jiang
- Hepatobiliary & Hydatid Disease Department, Digestive & Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qiang Guo
- Hepatobiliary & Hydatid Disease Department, Digestive & Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hui Wang
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- State Key Laboratory on Pathogenesis Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, China
| | - Liang Li
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- State Key Laboratory on Pathogenesis Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, China
| | - Renyong Lin
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- State Key Laboratory on Pathogenesis Prevention & Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, China
| | - Yingmei Shao
- Hepatobiliary & Hydatid Disease Department, Digestive & Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tuerganaili Aji
- Hepatobiliary & Hydatid Disease Department, Digestive & Vascular Surgery Center, First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- World Health Organization (WHO) Collaborating Center on Prevention and Management of Echinococcosis, Clinical Medicine Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
3
|
Zhou C, Zhang P, Ming Y. Characteristics of Intestinal Flora in Patients With Schistosoma japonicum Infection Undergoing Splenectomy. Parasite Immunol 2025; 47:e70008. [PMID: 40317954 PMCID: PMC12046944 DOI: 10.1111/pim.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 05/07/2025]
Abstract
Schistosomiasis japonica is a parasitic disease that seriously endangers human health. Patients with advanced Schistosoma japonicum infection often suffer from cirrhosis and portal hypertension. Splenectomy has been widely used in the treatment of these patients. Previous studies have confirmed that S. japonicum infection is closely related to the gut microbiota, but the impact of splenectomy on the gut microbiota of patients with advanced S. japonicum infection remains unclear. This study used 16sRNA sequencing technology to compare the differences in intestinal flora between patients with advanced S. japonicum infection who underwent splenectomy and non-surgical patients. We focused on the changes in the species composition, diversity and functions of the intestinal flora. Our study shows that dysbiosis of the gut microbiome occurred in patients with advanced S. japonicum infection, including changes in abundance and diversity and the disorder of biological function. The intestinal flora structure, diversity and function of patients who underwent splenectomy were significantly changed compared with those who did not undergo surgery.
Collapse
Affiliation(s)
- Chen Zhou
- Transplantation Center, Engineering and Technology Research Center for Transplantation Medicine of National Health ComissionThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Pengpeng Zhang
- Transplantation Center, Engineering and Technology Research Center for Transplantation Medicine of National Health ComissionThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Yingzi Ming
- Transplantation Center, Engineering and Technology Research Center for Transplantation Medicine of National Health ComissionThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| |
Collapse
|
4
|
Bansal A, Albano JE, Jayakumar D. Neuroschistosomiasis presenting as recurrent seizures: A case report. Surg Neurol Int 2025; 16:51. [PMID: 40041065 PMCID: PMC11878708 DOI: 10.25259/sni_1115_2024] [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: 12/25/2024] [Accepted: 01/28/2025] [Indexed: 03/06/2025] Open
Abstract
Background Cerebral pseudotumoral schistosomiasis is an uncommon and underreported condition, posing significant diagnostic challenges due to its ability to mimic other neurological conditions, especially in patients presenting with persistent seizures and imaging findings indicative of an infectious etiology. Case Description We report the case of a 16-year-old male who presented with persistent headaches and recurrent seizures despite adherence to antiseizure medications. Neuroimaging findings suggested an infectious process but were inconclusive in differentiating between a tuberculoma and cerebral schistosomiasis. Given the differing therapeutic approaches required for these conditions, a definitive diagnosis was pursued through a brain tissue biopsy, which confirmed cerebral schistosomiasis. This diagnosis guided appropriate treatment, leading to clinical improvement. Conclusion This case highlights the critical role of biopsy in establishing a definitive diagnosis when imaging results are inconclusive and suggests the importance of exploring the use of adjunct diagnostic methods like magnetic resonance spectroscopy, hence decreasing or potentially eliminating the need for an open biopsy.
Collapse
Affiliation(s)
- Anushree Bansal
- Department of Pediatrics, Davao Medical School Foundation, Medical Center, Davao City, Philippines
| | | | - Dheeraj Jayakumar
- Department of Pediatrics, Davao Medical School Foundation, Medical Center, Davao City, Philippines
| |
Collapse
|
5
|
Jain S. Can Schistosoma japonicum infection cause liver cancer? J Helminthol 2025; 99:e11. [PMID: 39924660 DOI: 10.1017/s0022149x24000762] [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] [Indexed: 02/11/2025]
Abstract
A co-relation between Schistosoma japonicum (Sj) and liver cancer (LC) in humans has been reported in the literature; however, this association is circumstantial. Due to the inconclusive nature of this association, the International Agency for Research on Cancer has placed Sj in Group 2B for LC, signifying it to be a 'possible carcinogen'. Many epidemiological, pathological and clinical studies have identified multiple factors, linked with Sj infection, which can lead to liver carcinogenesis. These factors include chronic inflammation in response to deposited eggs (which leads to fibrosis, cirrhosis and chromosomal instability at cellular level), hepatotoxic effects of egg-antigens, co-infection with hepatitis viruses, and up-regulation of glycolysis linked genes among others which predisposes hepatic tissue towards malignant transformation. The objective of this work is to present the current understanding on the association of Sj infection with LC. Mechanisms and factors linked with Sj infection that can lead to LC are emphasized, along with measures to diagnose and treat it. A comparison of liver carcinogenesis is also provided for cases linked with and independent of Sj infection. It appears that Sj, alone or with another carcinogen, is an important factor in liver carcinogenesis, but further studies are warranted to conclusively label 'infection with Sj alone' as a liver carcinogen.
Collapse
Affiliation(s)
- S Jain
- Independent Researcher, Institute for Globally Distributed Open Research and Education (IGDORE), Rewari, Haryana, India
| |
Collapse
|
6
|
Hu Y, Zhang Z, Adiham A, Li H, Gu J, Gong P. In Vivo and In Vitro Models of Hepatic Fibrosis for Pharmacodynamic Evaluation and Pathology Exploration. Int J Mol Sci 2025; 26:696. [PMID: 39859410 PMCID: PMC11766297 DOI: 10.3390/ijms26020696] [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: 10/22/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Hepatic fibrosis (HF) is an important pathological state in the progression of chronic liver disease to end-stage liver disease and is usually triggered by alcohol, nonalcoholic fatty liver, chronic hepatitis viruses, autoimmune hepatitis (AIH), or cholestatic liver disease. Research on novel therapies has become a hot topic due to the reversibility of HF. Research into the molecular mechanisms of the pathology of HF and potential drug screening relies on reliable and rational biological models, mainly including animals and cells. Hence, a number of modeling approaches have been attempted based on human dietary, pathological, and physiological factors in the development of HF. In this review, classical and novel methods of modeling HF in the last 10 years were collected from electronic databases, including Web of Science, PubMed, ScienceDirect, ResearchGate, Baidu Scholar, and CNKI. Animal models of HF are usually induced by chemical toxicants, special diets, pathogenic microorganisms, surgical operations, and gene editing. The advantages and limitations of hepatic stellate cells (HSCs), organoids, and 3D coculture-based HF modeling methods established in vitro were also proposed and summarized. This information provides a scientific basis for the discovery of the pathological mechanism and treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | - Jian Gu
- College of Pharmacy and Food, Southwest Minzu University, Chengdu 610093, China; (Y.H.); (Z.Z.); (A.A.); (H.L.)
| | - Puyang Gong
- College of Pharmacy and Food, Southwest Minzu University, Chengdu 610093, China; (Y.H.); (Z.Z.); (A.A.); (H.L.)
| |
Collapse
|
7
|
Jain S. Is Schistosoma mansoni playing a part in liver carcinogenesis? J Helminthol 2024; 98:e61. [PMID: 39469749 DOI: 10.1017/s0022149x24000506] [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] [Indexed: 10/30/2024]
Abstract
The relationship between Schistosoma mansoni (Sm) and hepatocellular carcinoma (HCC) has been evaluated by many studies that point towards a co-relation between schistosomal infection and HCC. While many such studies demonstrated that Sm infection in the presence of another carcinogenic factors leads to HCC, none of these studies could conclusively prove the cancer-inducing ability of Sm in humans, independent of other carcinogenic factors. The aim of this work is to present the current understanding on the association of Sm with HCC. Many epidemiological, pathological, and clinical studies have shown the role of multiple events like chronic inflammation and fibrosis as well as hepato-toxic agents like soluble egg antigens (SEAs), which help in creating a micro-environment which is suitable for HCC development. The role of Sm infection and deposited eggs in causing persistent inflammation, advanced fibrosis, and the role of SEAs, especially IPSE/alpha-1, is emphasised. This work concludes that Sm infection has the potential to induce cancer independently but the same has not been reported in humans to date. Extensive research is required to establish a causal relationship between Sm infection and HCC induction, or a complete lack thereof. However, Sm infection definitely acts along with other carcinogenic factors to induce HCC at a much faster pace and also leads to an aggressive form of liver cancer, which the other carcinogenic factor could not have achieved alone.
Collapse
Affiliation(s)
- S Jain
- Institute for Globally Distributed Open Research and Education (IGDORE), India
| |
Collapse
|
8
|
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
|
9
|
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: 2] [Impact Index Per Article: 2.0] [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
|
10
|
R LC, P.F. CM, M UE, V.J. BB. Hepatic schistosomiasis as a determining factor in the development of hepatic granulomas and liver fibrosis: a review of the current literature. Pathog Glob Health 2024; 118:529-537. [PMID: 39268619 PMCID: PMC11892069 DOI: 10.1080/20477724.2024.2400033] [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] [Indexed: 09/17/2024] Open
Abstract
Hepatic schistosomiasis is a neglected parasitosis that affects millions of people each year worldwide and leads to high healthcare costs and increased morbidity and mortality in infected humans. It is a disease that has been widely studied in terms of its pathophysiology; therefore, the signaling pathways that lead to liver damage, with the consequent development of liver fibrosis, are now better understood. Research has elucidated the role of soluble egg antigen in the development of hepatic granulomas and liver fibrosis, the signal transducer and activator of transcription 3 and its participation in liver damage, the role of heat shock protein 47 and its involvement in liver fibrosis, the anti-inflammatory effects caused by interleukin-37, and the role of natural killer and natural killer T cells in the development of the disease. Hepatic schistosomiasis can range from simple hepatomegaly to the development of portal hypertension combined with hepatic fibrosis. For diagnostic purposes, a microscopic examination of excreta remains the gold standard; however, abdominal ultrasound has recently taken on an important role in the assessment of liver lesions produced by the parasite. Praziquantel is considered the management drug of choice, and has been associated with a potential preventive antifibrotic effect.
Collapse
Affiliation(s)
- Lara-Cano R
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Benemérita Universidad Autónoma de Puebla, Mexico City, Mexico
| | | | - Uribe-Esquivel M
- Gastroenterology and Obesity Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | - Barbero-Becerra V.J.
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| |
Collapse
|
11
|
Chen YC, Lee YL, Lee CA, Lin TY, Hwu EET, Cheng PC. Development of a Lipid-encapsulated TGFβRI-siRNA Drug for Liver Fibrosis Induced by Schistosoma mansoni. PLoS Negl Trop Dis 2024; 18:e0012502. [PMID: 39264964 PMCID: PMC11421824 DOI: 10.1371/journal.pntd.0012502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/24/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Schistosoma mansoni infection leads to chronic schistosomiasis and severe hepatic fibrosis. We designed a liver-targeted lipid nanoparticle (LNP) carrying siRNA against type I TGF-β receptor (TGFβRI) mRNA to treat schistosomiasis-induced liver fibrosis in BALB/c mice. Knockdown of TGFβRI by LNP-siTGFβRI reduced LX-2 cell activation in vitro and alleviated liver fibrosis in S. mansoni-infected mice. αSMA and Col1a1 fibrotic markers in the liver tissues of infected mice were significantly suppressed in the treatment groups. In the serum of the LNP-siTGFβRI-treated groups, cytokines IFNγ, IL-1α, IL-6, IL-12, RANTES (CCL5), and TNFα increased, while GM-CSF, IL-2, IL-4, IL-10, IL-13, and KC (CXCL1) decreased compared to the control. Cell proportions were significantly altered in S. mansoni-infected mice, with increased CD56d NK cells and decreased CD19+ B cells and CD4+ T cells compared to naïve mice. Following LNP-siTGFβRI treatment, CD56d NK cells were downregulated, while B and memory Th cell populations were upregulated. The density of fibrotic regions significantly decreased with LNP-siTGFβRI treatment in a dose-dependent manner, and no systemic toxicity was observed in the major organs. This targeted siRNA delivery strategy effectively reduced granulomatous lesions in schistosomiasis-induced liver fibrosis without detectable side effects.
Collapse
Affiliation(s)
- Ying-Chou Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Drug Metabolism & Pharmacokinetics Department, Institute for Drug Evaluation Platform, Development Center for Biotechnology, Taipei, Taiwan
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, Taiwan
| | - Ching-An Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Yuan Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Edwin En-Te Hwu
- The Danish National Research Foundation and Villum Foundation’s Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics, Department of Health Technology, Technical University of Denmark
| | - Po-Ching Cheng
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Center for International Tropical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Alonzi T, Aiello A, Sali M, Delogu G, Villella VR, Raia V, Nicastri E, Piacentini M, Goletti D. Multiple antimicrobial and immune-modulating activities of cysteamine in infectious diseases. Biomed Pharmacother 2024; 178:117153. [PMID: 39024833 DOI: 10.1016/j.biopha.2024.117153] [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: 06/05/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024] Open
Abstract
Infectious diseases are a major threat to global health and cause millions of deaths every year, particularly in developing countries. The emergence of multidrug resistance challenges current antimicrobial treatments, inducing uncertainty in therapeutic protocols. New compounds are therefore necessary. A drug repurposing approach could play a critical role in developing new treatments used either alone or in combination with standard therapy regimens. Herein, we focused on cysteamine, an aminothiol endogenously synthesized by human cells during the degradation of coenzyme-A, which is a drug approved for the treatment of nephropathic cystinosis. Cysteamine influences many biological processes due to the presence of the highly reactive thiol group. This review provides an overview of cysteamine-mediated effects on different viruses, bacteria and parasites, with a particular focus on infections caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), Mycobacterium tuberculosis, non-tuberculous mycobacteria (NTM), and Pseudomonas aeruginosa. Evidences for a potential use of cysteamine as a direct antimicrobial agent and/or a host-directed therapy, either alone or in combination with other antimicrobial drugs, are described.
Collapse
Affiliation(s)
- Tonino Alonzi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Michela Sali
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, India; Dipartimento di Scienze di Laboratorio ed Ematologiche, Fondazione Policlinico Universitario A. Gemelli, IRCCS; Rome, Italy
| | - Giovanni Delogu
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, India; Mater Olbia Hospital, Olbia, Italy
| | - Valeria Rachela Villella
- CEINGE, Advanced Biotechnologies Franco Salvatore, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Valeria Raia
- Pediatric Unit, Department of Translational Medical Sciences, Regional Cystic Fibrosis Center, Federico II University Naples, Naples, Italy
| | - Emanuele Nicastri
- Clinical Division of Infectious Diseases, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy; Cell Biology and Electron Microscopy Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy.
| |
Collapse
|
13
|
Gao R, Mao J. Noncoding RNA-Mediated Epigenetic Regulation in Hepatic Stellate Cells of Liver Fibrosis. Noncoding RNA 2024; 10:44. [PMID: 39195573 DOI: 10.3390/ncrna10040044] [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: 05/22/2024] [Revised: 07/09/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
Liver fibrosis is a significant contributor to liver-related disease mortality on a global scale. Despite this, there remains a dearth of effective therapeutic interventions capable of reversing this condition. Consequently, it is imperative that we gain a comprehensive understanding of the underlying mechanisms driving liver fibrosis. In this regard, the activation of hepatic stellate cells (HSCs) is recognized as a pivotal factor in the development and progression of liver fibrosis. The role of noncoding RNAs (ncRNAs) in epigenetic regulation of HSCs transdifferentiation into myofibroblasts has been established, providing new insights into gene expression changes during HSCs activation. NcRNAs play a crucial role in mediating the epigenetics of HSCs, serving as novel regulators in the pathogenesis of liver fibrosis. As research on epigenetics expands, the connection between ncRNAs involved in HSCs activation and epigenetic mechanisms becomes more evident. These changes in gene regulation have attracted considerable attention from researchers in the field. Furthermore, epigenetics has contributed valuable insights to drug discovery and the identification of therapeutic targets for individuals suffering from liver fibrosis and cirrhosis. As such, this review offers a thorough discussion on the role of ncRNAs in the HSCs activation of liver fibrosis.
Collapse
Affiliation(s)
- Ruoyu Gao
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jingwei Mao
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
14
|
Zhang X, Yu C, Song L. Progress on the Regulation of the Host Immune Response by Parasite-Derived Exosomes. Pathogens 2024; 13:623. [PMID: 39204224 PMCID: PMC11357678 DOI: 10.3390/pathogens13080623] [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: 07/01/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Exosomes are membrane-bound structures released by cells into the external environment that carry a significant amount of important cargo, such as proteins, DNA, RNA, and lipids. They play a crucial role in intercellular communication. Parasites have complex life cycles and can release exosomes at different stages. Exosomes released by parasitic pathogens or infected cells contain parasitic nucleic acids, antigenic molecules, virulence factors, drug-resistant proteins, proteases, lipids, etc. These components can regulate host gene expression across species or modulate signaling pathways, thereby dampening or activating host immune responses, causing pathological damage, and participating in disease progression. This review focuses on the means by which parasitic exosomes modulate host immune responses, elaborates on the pathogenic mechanisms of parasites, clarifies the interactions between parasites and hosts, and provides a theoretical basis and research directions for the prevention and treatment of parasitic diseases.
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;
| | - 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;
| |
Collapse
|
15
|
Vatankhah M, Panahizadeh R, Safari A, Ziyabakhsh A, Mohammadi-Ghalehbin B, Soozangar N, Jeddi F. The role of Nrf2 signaling in parasitic diseases and its therapeutic potential. Heliyon 2024; 10:e32459. [PMID: 38988513 PMCID: PMC11233909 DOI: 10.1016/j.heliyon.2024.e32459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 07/12/2024] Open
Abstract
In response to invading parasites, one of the principal arms of innate immunity is oxidative stress, caused by reactive oxygen species (ROS). However, oxidative stresses play dual functions in the disease, whereby free radicals promote pathogen removal, but they can also trigger inflammation, resulting in tissue injuries. A growing body of evidence has strongly supported the notion that nuclear factor erythroid 2-related factor 2 (NRF) signaling is one of the main antioxidant pathways to combat this oxidative burst against parasites. Given the important role of NRF2 in oxidative stress, in this review, we investigate the activation mechanism of the NRF2 antioxidant pathway in different parasitic diseases, such as malaria, leishmaniasis, trypanosomiasis, toxoplasmosis, schistosomiasis, entamoebiasis, and trichinosis.
Collapse
Affiliation(s)
- Mohammadamin Vatankhah
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Reza Panahizadeh
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Safari
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Alireza Ziyabakhsh
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Narges Soozangar
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
16
|
Yu Z, Jiang T, Xu F, Zhang J, Hu Y, Cao J. Inhibiting Liver Autophagy and Promoting Hepatocyte Apoptosis by Schistosoma Japonicum Infection. Trop Med Infect Dis 2024; 9:42. [PMID: 38393131 PMCID: PMC10892706 DOI: 10.3390/tropicalmed9020042] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
We established a mouse model of Schistosoma japonicum infection in order to study the effects of the infection on hepatocyte autophagy and apoptosis. We also stimulated HepG2 cells with soluble egg antigens (SEA) in vitro. At two, four, and six weeks post-infection, quantitative real-time PCR and Western blot (WB) were used to detect liver expression levels of autophagy and apoptosis-related proteins. HepG2 cells were treated with different concentrations of SEA. The changes in the levels of autophagy-related proteins and HepG2 cell apoptosis were detected. The Lc3b, Beclin1, Atg7, and Atg12 mRNA levels were significantly lower at four and six weeks after infection than those in the uninfected group. At four and six weeks following infection, the levels of Beclin1, LC3BII/I, Atg7, and p62 proteins were considerably lower than those in the uninfected group. The protein levels of pro-apoptotic Bax and cleaved caspase 3 and fibrosis-related proteins α-SMA and collagen 3 in the liver post-infection were significantly higher than those in uninfected mice. HepG2 cells stimulated with SEA showed decreased levels of Beclin1, p62, and Atg7 proteins and significantly increased apoptosis rates. The findings demonstrated that following infection with S. japonicum, mice's liver fibrosis worsened, hepatic autophagy was suppressed, and hepatocyte apoptosis was encouraged.
Collapse
Affiliation(s)
- Zhihao Yu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, Shanghai 200025, China; (Z.Y.); (T.J.); (F.X.); (J.Z.)
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China
- World Health Organization Collaborating Center for Tropical Diseases, Shanghai 200025, China
- World Health Organization Centre for Tropical Diseases, Shanghai 200025, China
| | - Tingting Jiang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, Shanghai 200025, China; (Z.Y.); (T.J.); (F.X.); (J.Z.)
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China
- World Health Organization Collaborating Center for Tropical Diseases, Shanghai 200025, China
- World Health Organization Centre for Tropical Diseases, Shanghai 200025, China
| | - Fangfang Xu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, Shanghai 200025, China; (Z.Y.); (T.J.); (F.X.); (J.Z.)
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China
- World Health Organization Collaborating Center for Tropical Diseases, Shanghai 200025, China
- World Health Organization Centre for Tropical Diseases, Shanghai 200025, China
| | - Jing Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, Shanghai 200025, China; (Z.Y.); (T.J.); (F.X.); (J.Z.)
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China
- World Health Organization Collaborating Center for Tropical Diseases, Shanghai 200025, China
- World Health Organization Centre for Tropical Diseases, Shanghai 200025, China
| | - Yuan Hu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, Shanghai 200025, China; (Z.Y.); (T.J.); (F.X.); (J.Z.)
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China
- World Health Organization Collaborating Center for Tropical Diseases, Shanghai 200025, China
- World Health Organization Centre for Tropical Diseases, Shanghai 200025, China
| | - Jianping Cao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China, Shanghai 200025, China; (Z.Y.); (T.J.); (F.X.); (J.Z.)
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China
- World Health Organization Collaborating Center for Tropical Diseases, Shanghai 200025, China
- World Health Organization Centre for Tropical Diseases, Shanghai 200025, China
- The School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
17
|
Sharaf-El-Deen S, Soliman S, Brakat R. Evaluation of the antiparasitic and antifibrotic effects of gallic acid on experimental hepatic schistosomiasis mansoni. J Helminthol 2024; 98:e3. [PMID: 38167243 DOI: 10.1017/s0022149x23000937] [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] [Indexed: 01/05/2024]
Abstract
Schistosomiasis afflicts approximately 120 million individuals globally. The hepatic pathology that occurs due to egg-induced granuloma and fibrosis is commonly attributed to this condition. However, there is currently no efficacious treatment available for either of these conditions.Our study aimed to investigate the potential antifibrotic and antiparasitic properties of different doses of gallic acid (GA) in experimental schistosomiasis mansoni. In addition, we investigated the outcomes of co-administering it with the standard anti-schistosomiasis treatment, praziquantel (PZQ).In experiment I, Schistosoma mansoni-infected mice were administered GA at doses of 10, 20, or 40 mg/kg. Their effectiveness was evaluated through parasitological (worm and egg loads, granuloma number and diameter), pathological (fibrosis percentage and H-score of hepatic stellate cells (HSCs)), and functional (liver enzymes) tests. In experiment II, we investigated the optimal dosage that yielded the best outcomes. This dosage was administered in conjunction with PZQ and was evaluated regarding the parasitological, pathological, functional, and immunological (fibrosis-regulating cytokines) activities.Our findings indicate that the administration of 40 mg/kg GA exhibited the highest level of effectiveness in experiment I. In experiment II, it exhibited lower antiparasitic efficacy in comparison to PZQ. However, it surpassed PZQ in other tests. It showed enhanced outcomes when combined with PZQ.In conclusion, our findings reveal that GA only slightly increased the antischistosomal activity of PZQ. However, it was linked to decreased fibrosis, particularly when administrated with PZQ. Our pilot study identifies GA as a natural antifibrotic agent, which could be administered with PZQ to mitigate the development of fibrosis.
Collapse
Affiliation(s)
- S Sharaf-El-Deen
- Parasitology Department, Faculty of Medicine, Menoufia University, Shebin-el-kom, Menoufia, Egypt
| | - S Soliman
- Public Health and Community Medicine Department, Faculty of Medicine, Menoufia University, Shebin-el-kom, Menoufia, Egypt
| | - R Brakat
- Parasitology Department, Faculty of Medicine, Menoufia University, Shebin-el-kom, Menoufia, Egypt
| |
Collapse
|
18
|
Xie S, Zhang Y, Li J, Zhou J, Li J, Zhang P, Liu Y, Luo Y, Ming Y. IgG persistence showed weak clinical aspects in chronic schistosomiasis patients. Sci Rep 2023; 13:13222. [PMID: 37580417 PMCID: PMC10425409 DOI: 10.1038/s41598-023-40082-z] [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: 02/13/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023] Open
Abstract
Schistosomiasis is a chronic parasitic disease, which affects the quality of daily life of patients and imposes a huge burden on society. Hepatic fibrosis in response to continuous insult of eggs to the liver is a significant cause of morbidity and mortality. However, the mechanisms of hepatic fibrosis in schistosomiasis are largely undefined. The purpose of our study is to detect the indicator to hepatic fibrosis in schistosomiasis. A total of 488 patients with chronic schistosomiasis japonica were enrolled in our study. The patients were divided into two groups according to liver ultrasound examination, which could indicate liver fibrosis of schistosomiasis with unique reticular changes. Logistic regression analysis showed that globulin, albumin/globulin, GGT levels and anti-Schistosoma IgG were independently associated with liver fibrosis in patients with schistosomiasis and IgG was the largest association of liver fibrosis (OR 2.039, 95% CI 1.293-3.213). We further compared IgG+ patients with IgG- patients. IgG+ patients (ALT 25 U/L, GGT 31 U/L) slightly higher than IgG- patients (ALT 22 U/L, GGT 26 U/L) in ALT and GGT. However, the fibrosis of liver in IgG+ patients (Grade II(19.7%), Grade III(7.3%)) were more severe than that in IgG- patients(Grade II(12.5%), Grade III(2.9%)) according to the grade of liver ultrasonography. Our results showed anti-Schistosoma IgG was independently associated with liver fibrosis in patients with chronic schistosomiasis japonica and patients with persistent anti-Schistosoma IgG might have more liver fibrosis than negative patients despite no obvious clinical signs or symptoms.
Collapse
Affiliation(s)
- Shudong Xie
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Comission, Changsha, Hunan, People's Republic of China
| | - Yu Zhang
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Comission, Changsha, Hunan, People's Republic of China
| | - Junhui Li
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Comission, Changsha, Hunan, People's Republic of China
| | - Jie Zhou
- Hunan Institute of Schistosomiasis Control, Yueyang, Hunan, People's Republic of China
| | - Jun Li
- Hunan Institute of Schistosomiasis Control, Yueyang, Hunan, People's Republic of China
| | - Pengpeng Zhang
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Comission, Changsha, Hunan, People's Republic of China
| | - Yang Liu
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Comission, Changsha, Hunan, People's Republic of China
| | - Yulin Luo
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Comission, Changsha, Hunan, People's Republic of China
| | - Yingzi Ming
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China.
- Engineering and Technology Research Center for Transplantation Medicine of National Health Comission, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
19
|
Cao S, Wang D, Wu Y, Zhang J, Pu L, Luo X, Zhang X, Sun X, Zheng Y, Wang S, Guo X. mmu-miRNA-342-3p promotes hepatic stellate cell activation and hepatic fibrosis induced by Echinococcus multilocularis infection via targeting Zbtb7a. PLoS Negl Trop Dis 2023; 17:e0011520. [PMID: 37490505 PMCID: PMC10403128 DOI: 10.1371/journal.pntd.0011520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 07/08/2023] [Indexed: 07/27/2023] Open
Abstract
Liver fibrosis is one of the histopathological characters during Echinococcus multilocularis infection. The activation of hepatic stellate cells (HSCs) is a key event in the development of liver fibrosis. However, the molecular mechanism of HSC activation in the E. multilocularis infection-induced liver fibrosis remains largely unclear. Here, we reported that mmu-miR-342-3p was most dominantly expressed in HSCs and was upregulated in the HSCs in response to E. multilocularis infection. We further showed that mmu-miR-342-3p was able to bind to the 3' UTR of the Zbtb7a gene and regulated its expression. Moreover, mmu-miR-342-3p expression was negatively correlated with its target gene Zbtb7a in HSCs during E. multilocularis infection. Knockdown of mmu-miR-342-3p promoted the expression of Gfap in the activated HSCs in vitro. In the E. multilocularis-infected mice, knockdown of mmu-miR-342-3p suppressed the expression of α-Sma, Col1α1, and TGF-β but promoted the expression of Gfap. Therefore, mmu-miR-342-3p is a key regulator for activation of HSCs, and inhibiting mmu-miR-342-3p to promote Zbtb7a-mediated TGF-β signaling in activated HSCs could be a novel strategy to treat liver fibrosis induced by E. multilocularis.
Collapse
Affiliation(s)
- Shanling Cao
- 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 Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Dexian Wang
- 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 Sciences, Lanzhou, Gansu, China
| | - Yixuan Wu
- 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 Sciences, Lanzhou, Gansu, China
| | - Junmei Zhang
- 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 Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Lixia Pu
- 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 Sciences, Lanzhou, Gansu, China
| | - Xuenong Luo
- 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 Sciences, Lanzhou, Gansu, China
| | - Xueyong Zhang
- 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 Sciences, Lanzhou, Gansu, China
- Qinghai Academy of Animal Sciences and Veterinary Medicine, Qinghai University, Xining P. R. China
| | - Xiaolin Sun
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yadong Zheng
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, China
| | - Shuai Wang
- 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 Sciences, Lanzhou, Gansu, China
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaola Guo
- 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 Sciences, Lanzhou, Gansu, China
| |
Collapse
|
20
|
Differential Analysis of Key Proteins Related to Fibrosis and Inflammation in Soluble Egg Antigen of Schistosoma mansoni at Different Infection Times. Pathogens 2023; 12:pathogens12030441. [PMID: 36986363 PMCID: PMC10054402 DOI: 10.3390/pathogens12030441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Schistosomiasis is a major global health problem. Schistosomes secrete antigens into the host tissue that bind to chemokines or inhibit immune cell receptors, regulating the immune responses to allow schistosome development. However, the detailed mechanism of chronic schistosome infection-induced liver fibrosis, including the relationship between secreted soluble egg antigen (SEA) and hepatic stellate cell (HSC) activation, is still unknown. We used mass spectrometry to identify the SEA protein sequences from different infection weeks. In the 10th and 12th infection weeks, we focused on the SEA components and screened out the special protein components, particularly fibrosis- and inflammation-related protein sequences. Our results have identified heat shock proteins, phosphorylation-associated enzymes, or kinases, such as Sm16, GSTA3, GPCRs, EF1-α, MMP7, and other proteins linked to schistosome-induced liver fibrosis. After sorting, we found many special proteins related to fibrosis and inflammation, but studies proving their association with schistosomiasis infection are limited. Follow-up studies on MICOS, MATE1, 14-3-3 epsilon, and CDCP1 are needed. We treated the LX-2 cells with the SEA from the 8th, 10th, and 12th infection weeks to test HSC activation. In a trans-well cell model in which PBMCs and HSCs were co-cultured, the SEA could significantly induce TGF-β secretion, especially from the 12th week of infection. Our data also showed that TGF-β secreted by PBMC after the SEA treatment activates LX-2 and upregulates hepatic fibrotic markers α-SMA and collagen 1. Based on these results, the CUB domain-containing protein 1 (CDCP1) screened at the 12th infection week could be investigated further. This study clarifies the trend of immune mechanism variation in the different stages of schistosome infection. However, how egg-induced immune response transformation causes liver tissue fibrosis needs to be studied further.
Collapse
|