1
|
Zhang Y, Shen C, Zhu X, Leow CY, Ji M, Xu Z. Helminth-derived molecules: pathogenic and pharmacopeial roles. J Biomed Res 2024; 38:1-22. [PMID: 39314046 PMCID: PMC11629161 DOI: 10.7555/jbr.38.20240177] [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: 06/18/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024] Open
Abstract
Parasitic helminths, taxonomically comprising trematodes, cestodes, and nematodes, are multicellular invertebrates widely disseminated in nature and have afflicted people continuously for a long time. Helminths play potent roles in the host through generating a variety of novel molecules, including some excretory/secretory products and others that are involved in intracellular material exchange and information transfer as well as the initiation or stimulation of immune and metabolic activation. The helminth-derived molecules have developed powerful and diverse immunosuppressive effects to achieve immune evasion for parasite survival and establish chronic infections. However, they also improve autoimmune and allergic inflammatory responses and promote metabolic homeostasis by promoting metabolic reprogramming of various immune functions, and then inducing alternatively activated macrophages, T helper 2 cells, and regulatory T cells-mediated immune responses. Therefore, a deeper exploration of the immunopathogenic mechanism and immune regulatory mechanisms of helminth-derived molecules exerted in the host is crucial for understanding host-helminth interactions as well as the development of therapeutic drugs for infectious or non-infectious diseases. In this review, we focus on the properties of helminth-derived molecules to give an overview of the most recent scientific knowledge about their pathogenic and pharmacopeial roles in immune-metabolic homeostasis.
Collapse
Affiliation(s)
- Yu Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chunxiang Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xinyi Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia
| | - Minjun Ji
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zhipeng Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
2
|
Bao P, Wang X, Zhang X, Yu Y, Ma Y, Zhang H, Wang Y, Liu X, Gong P, Zhang N, Lee SO, Li X, Li J. Clonorchis sinensis aggravated liver fibrosis by activating PARP-1 signaling to induce parthanatos via DNA damage. Vet Parasitol 2024; 330:110217. [PMID: 38861911 DOI: 10.1016/j.vetpar.2024.110217] [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: 04/11/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024]
Abstract
Clonorchis sinensis is an important food-borne zoonotic parasite that is highly associated with liver fibrosis and cholangiocarcinoma. Further understanding of the pathogenesis of C. sinensis, especially liver fibrosis, could help us develop novel strategies for controlling clonorchiasis. Poly (ADP-ribose) polymerase-1 (PARP-1) can induce cellular parthanatos which is reported to be involved in liver fibrosis. Currently, whether C. sinensis could activate PARP-1 signaling to induce parthanatos or whether parthanatos play a role in C. sinensis-induced liver fibrosis is not clear. In the present study, the expression of PARP-1 and parthanatos indicators were detected in C. sinensis-infected mouse liver and in human intrahepatic biliary epithelial cells (HiBEpiCs) incubated with excretory/secretory products (ESPs) of C. sinensis. To explore the role of PARP-1 in C. sinensis infection, PARP-1 inhibitor NMS-P118 was used to block PARP-1 expression in vivo and vitro. The mortality rate, body weight, worm load, liver and bile duct lesions as well as PARP-1 and parthanatos indicators in C57BL/6 mice infected with C. sinensis, or in HiBEpiCs incubated with C. sinensis ESPs and NMS-P118 were analyzed and compared to the group without NMS-P118. The results showed that C. sinensis infection induced the activation of PARP-1 signaling as well as the translocation of AIF and MIF into the nucleus in mouse liver. ESPs of C. sinensis could induce PARP-1 up-regulation, ATP depletion and DNA damage in HiBEpiCs, indicating that C. sinensis could induce parthanatos. Inhibiting PARP-1 with NMS-P118 significantly reduced liver fibrosis and the number of larvae, increased the survival rate and body weight gain of the mice infected with C. sinensis. In addition, NMS-P118 decreased the expression of PARP-1 and alleviated ATP depletion as well as DNA damage in HiBEpiCs incubated with ESPs of C. sinensis. Our data indicated that C. sinensis and its ESPs could activate PARP-1 signaling to induce cellular parthanatos. NMS-P118 treatment alleviated liver fibrosis and promoted survival of the mice by inhibiting PARP-1, which suggested that PARP-1 could be used as a potential therapeutic target against clonorchiasis.
Collapse
Affiliation(s)
- Penglin Bao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xiaocen Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xu Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Yanhui Yu
- Second Affiliated Hospital, Jilin University, Changchun, China.
| | - Yeting Ma
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Haoyang Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Yuru Wang
- College of Public Health, Jilin Medical University, Jilin 132013, China.
| | - Xiaolei Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Pengtao Gong
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Nan Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Soon-Ok Lee
- Department of Medical Research Center for Bioreaction to Reactive Oxygen Species, Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Xin Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Jianhua Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
3
|
Xu L, Zhang Y, Ji N, Du Y, Jia T, Wei S, Wang W, Zhang S, Chen W. Tanshinone IIA regulates the TGF‑β1/Smad signaling pathway to ameliorate non‑alcoholic steatohepatitis‑related fibrosis. Exp Ther Med 2022; 24:486. [PMID: 35761808 PMCID: PMC9214595 DOI: 10.3892/etm.2022.11413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/04/2022] [Indexed: 11/19/2022] Open
Abstract
Tanshinone IIA (TIIA) is a major component extracted from the traditional herbal medicine Salvia miltiorrhiza and has been indicated to play a role in the treatment of organ fibrosis. However, the evidence supporting its antifibrotic effect is insufficient and the underlying mechanism is unclear. To investigate the therapeutic effect of TIIA on non-alcoholic steatohepatitis-related fibrosis (NASH-F), the present study used a methionine choline deficiency diet to induce NASH-F in rats, and explored the effect of TIIA on the transforming growth factor-β1 (TGF-β1)/Smad signaling pathway. Wistar rats were randomly divided into control, NASH-F and TIIA groups. After 8 weeks of treatment, the levels of serum markers associated with liver function and fibrosis were measured, liver fat vacuoles and inflammation were assessed by haematoxylin and eosin staining, and liver fibrosis was assessed by Masson's trichrome staining. TGF-β1, Smad2, Smad3, Smad7 and α-smooth muscle actin (α-SMA) mRNA expression, and TGF-β1, Smad2/3, phosphorylated (p)-Smad2/3, Smad7 and α-SMA protein levels were determined. The results revealed that TIIA could remarkably ameliorate liver fat vacuoles and inflammation in NASH-F rats, and could decrease the levels of serum aspartate aminotransferase, alanine aminotransferase, total bilirubin, total bile acid, hyaluronic acid, type Ⅳ collagen, laminin and type III collagen, while increasing the levels of total cholesterol and triglycerides; however, this was not statistically significance. TIIA markedly suppressed the increased TGF-β1, Smad2, Smad3 and α-SMA mRNA expression levels observed in the liver of NASH-F rats, while it increased the mRNA expression level of Smad7. Similarly, TIIA suppressed the increased TGF-β1, p-Smad2/3 and α-SMA protein levels observed in the liver of NASH-F rats, while it increased the protein expression level of Smad7 in vitro and in vivo. TIIA had no significant cytotoxic effect at 10, 20, 40 and 80 µmol/l on human LX-2 cell. In conclusion, the findings of the present study indicated that TIIA alleviated NASH-F by regulating the TGF-β1/Smad signaling pathway. TIIA may be a useful tool in the prevention and treatment of NASH-F.
Collapse
Affiliation(s)
- Lianjie Xu
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yurong Zhang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Nengbo Ji
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yan Du
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Tao Jia
- Department of Orthopedics, First Clinical Medical College of Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650021, P.R. China
| | - Shanshan Wei
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Wei Wang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Shan Zhang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Wenhui Chen
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
4
|
Yin F, Mao LC, Cai QQ, Jiang WH. Effect of Hepatocyte Growth Factor-Transfected Human Umbilical Cord Mesenchymal Stem Cells on Hepatic Stellate Cells by Regulating Transforming Growth Factor-β1/Smads Signaling Pathway. Stem Cells Dev 2021; 30:1070-1081. [PMID: 34514810 DOI: 10.1089/scd.2021.0136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Studies have shown that human umbilical cord mesenchymal stem cells (hUCMSCs) could ameliorate liver fibrosis (LF) through inhibiting the activation of hepatic stellate cells (HSCs). However, the specific mechanisms have not been studied clearly. The purpose of this study was to explore the possible mechanism of hepatocyte growth factor (HGF)-transfected hUCMSCs in inhibiting the proliferation and activation of HSCs-T6. The upper and lower double-cell coculture system was established among HGF-hUCMSCs, LV5-NC-hUCMSCs, hUCMSCs, and HSCs-T6 in experimental groups; HSCs-T6 were cultured alone as control group. After coculturing for 1, 2, and 3 days, results showed that HGF-transfected hUCMSCs could decrease cell viability of HSCs-T6 and promote apoptosis; inhibit their activation and reduce the expression of Collagen I, Collagen III, TGF-β1, Smad2 and Smad3, which may be related to inhibiting the activation of TGF-β1/Smads signaling pathway. These findings suggested that HGF-transfected hUCMSCs may be used as an alternative and novel therapeutic approach for the treatment of LF.
Collapse
Affiliation(s)
- Fei Yin
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, Jilin Province, China
| | - Li-Cui Mao
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, Jilin Province, China
| | - Qi-Qi Cai
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, Jilin Province, China
| | - Wen-Hua Jiang
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
5
|
Zhou QY, Yang HM, Liu JX, Xu N, Li J, Shen LP, Zhang YZ, Koda S, Zhang BB, Yu Q, Chen JX, Zheng KY, Yan C. MicroRNA-497 induced by Clonorchis sinensis enhances the TGF-β/Smad signaling pathway to promote hepatic fibrosis by targeting Smad7. Parasit Vectors 2021; 14:472. [PMID: 34521449 PMCID: PMC8442346 DOI: 10.1186/s13071-021-04972-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022] Open
Abstract
Background Various stimuli, including Clonorchis sinensis infection, can cause liver fibrosis. Liver fibrosis is characterized by the activation of hepatic stellate cells (HSCs) with massive production of extracellular matrix (ECM). Our previous study showed that the TGF-β1-induced Smad signaling pathway played a critical role in the activation of HSCs during liver fibrosis induced by worm infection; however, the mechanisms that modulate the TGF-β/Smad signaling pathway are still poorly understood. Accumulating evidence demonstrates that miRNAs act as an important regulator of activation of HSCs during liver fibrosis. Methods The target of miR-497 was determined by bioinformatics analysis combined with a dual-luciferase activity assay. LX-2 cells were transfected with miR-497 inhibitor and then stimulated with TGF-β1 or excretory/secretory products of C. sinensis (CsESPs), and activation of LX-2 was assessed using qPCR or western blot. In vivo, the mice treated with CCl4 were intravenously injected with a single dose of adeno-associated virus serotype 8 (AAV8) that overexpressed anti-miR-497 sequences or their scramble control for 6 weeks. Liver fibrosis and damage were assessed by hematoxylin and eosin (H&E) staining, Masson staining, and qPCR; the activation of the TGF-β/Smad signaling pathway was detected by qPCR or western blot. Results In the present study, the expression of miR-497 was increased in HSCs activated by TGF-β1 or ESPs of C. sinensis. We identified that Smad7 was the target of miR-497 using combined bioinformatics analysis with luciferase activity assays. Transfection of anti-miR-497 into HSCs upregulated the expression of Smad7, leading to a decrease in the level of p-Smad2/3 and subsequent suppression of the activation of HSCs induced by TGF-β1 or CsESPs. Furthermore, miR-497 inhibitor delivered by highly-hepatotropic (rAAV8) inhibited TGF-β/smads signaling pathway by targeting at Smad7 to ameliorate CCL4-induced liver fibrosis. Conclusions The present study demonstrates that miR-497 promotes liver fibrogenesis by targeting Smad7 to promote TGF-β/Smad signaling pathway transduction both in vivo and in vitro, which provides a promising therapeutic strategy using anti-miR-497 against liver fibrosis. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Qian-Yang Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China.,Department of Dermatology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, China
| | - Hui-Min Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China
| | - Ji-Xin Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China
| | - Na Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China
| | - Jing Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China
| | - Li-Ping Shen
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China
| | - Yu-Zhao Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China
| | - Stephane Koda
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China
| | - Bei-Bei Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China.,National Experimental Demonstration Center for Basic Medicine Education, Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Qian Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China.,National Experimental Demonstration Center for Basic Medicine Education, Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Jia-Xu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology, Ministry of Health, WHO Collaborating Center of Malaria, Schistosomiasis and Filariasis, Shanghai, 200025, People's Republic of China
| | - Kui-Yang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China. .,National Experimental Demonstration Center for Basic Medicine Education, Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China.
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Key Laboratory of Infection and Immunity, Xuzhou Medical University, Jiangsu, 221004, Xuzhou, People's Republic of China. .,National Experimental Demonstration Center for Basic Medicine Education, Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China.
| |
Collapse
|
6
|
Wu YJ, He Q, Shang M, Yin YX, Li Y, Du X, Li XR. The NF-κB signalling pathway and TM7SF3 contribute to liver fibrosis caused by secreted phospholipase A2 of Clonorchis sinensis. Parasit Vectors 2021; 14:152. [PMID: 33691755 PMCID: PMC7945307 DOI: 10.1186/s13071-021-04663-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 02/26/2021] [Indexed: 11/23/2022] Open
Abstract
Background The NF-κB signalling pathway has been reported to be related to liver fibrosis, and we investigated whether the NF-κB signalling pathway is involved in liver fibrosis caused by secreted phospholipase A2 of Clonorchis sinensis (CssPLA2). Furthermore, expression of the receptor of CssPLA2 on the cell surface of hepatic stellate cells (HSCs) may greatly contribute to liver fibrosis. Methods CssPLA2 was administered to BALB/c mice by abdominal injection. The levels of markers of NF-κB signalling pathway activation in mouse liver tissue were measured by quantitative RT-PCR, ELISA and western blot. Additionally, HSCs were incubated with CssPLA2, and an NF-κB signalling inhibitor (BAY 11-7082) was applied to test whether the NF-κB signalling pathway plays a role in the effect of CssPLA2. Then, the interaction between CssPLA2 and its receptor transmembrane 7 superfamily member 3 (TM7SF3) was confirmed by co-immunoprecipitation (co-IP) and GST pull-down. To determine how TM7SF3 influences the ability of CssPLA2 to cause liver fibrosis, a TM7SF3 antibody was used to block TM7SF3. Results The levels of the NF-ΚB signalling pathway activation markers TNF-α, IL-1β and phospho-p65 were increased by CssPLA2 in the context of liver fibrosis. In addition, the interaction between TM7SF3 and CssPLA2 was confirmed by co-IP and GST pull-down. When TM7SF3 was blocked by an antibody targeting 1–295 amino acids of TM7SF3, activation of HSCs caused by CssPLA2 was alleviated. Conclusions The NF-ΚB signalling pathway is involved in the activation of HSCs by CssPLA2. TM7SF3, the receptor of CssPLA2, plays important roles in liver fibrosis caused by CssPLA2.![]()
Collapse
Affiliation(s)
- Yin-Juan Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Qing He
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Mei Shang
- Department of Clinical Laboratory, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Ying-Xuan Yin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Ye Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Xue Du
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Xue-Rong Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China. .,Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China. .,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, Guangdong, 510080, People's Republic of China.
| |
Collapse
|
7
|
Shi W, He JJ, Mei XF, Lu KJ, Zeng ZX, Zhang YY, Sheng ZA, Elsheikha HM, Huang WY, Zhu XQ. Dysregulation of hepatic microRNA expression in C57BL/6 mice affected by excretory-secretory products of Fasciola gigantica. PLoS Negl Trop Dis 2020; 14:e0008951. [PMID: 33332355 PMCID: PMC7775122 DOI: 10.1371/journal.pntd.0008951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 12/31/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
The excretory-secretory products released by the liver fluke Fasciola gigantica (FgESPs) play important roles in regulating the host immune response during the infection. Identification of hepatic miRNAs altered by FgESPs may improve our understanding of the pathogenesis of F. gigantica infection. In this study, we investigated the alterations in the hepatic microRNAs (miRNAs) in mice treated with FgESPs using high-throughput small RNA (sRNA) sequencing and bioinformatics analysis. The expression of seven miRNAs was confirmed by quantitative stem-loop reverse transcription quantitative PCR (qRT-PCR). A total of 1,313 miRNAs were identified in the liver of mice, and the differentially expressed (DE) miRNAs varied across the time lapsed post exposure to FgESPs. We identified 67, 154 and 53 dysregulated miRNAs at 1, 4 and 12 weeks post-exposure, respectively. 5 miRNAs (miR-126a-3p, miR-150-5p, miR-155-5p, miR-181a-5p and miR-362-3p) were commonly dysregulated at the three time points. We also found that most of the DE miRNAs were induced by FgESPs in the mouse liver after 4 weeks of exposure. These were subjected to Gene Ontology (GO) enrichment analysis, which showed that the predicted targets of the hepatic DE miRNAs of mice 4 weeks of FgESPs injection were enriched in GO terms, including cell membrane, ion binding, cellular communication, organelle and DNA damage. KEGG analysis indicated that the predicted targets of the most downregulated miRNAs were involved in 15 neural activity-related pathways, 6 digestion-related pathways, 20 immune response-related pathways and 17 cancer-related pathways. These data provide new insights into how FgESPs can dysregulate hepatic miRNAs, which play important roles in modulating several aspects of F. gigantica pathogenesis.
Collapse
Affiliation(s)
- Wei Shi
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, People’s Republic of China
- School of Animal Science and Technology, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, People’s Republic of China
- * E-mail:
| | - Xue-Fang Mei
- School of Animal Science and Technology, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Ke-Jing Lu
- School of Animal Science and Technology, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Zi-Xuan Zeng
- School of Animal Science and Technology, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Yao-Yao Zhang
- School of Animal Science and Technology, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Zhao-An Sheng
- School of Animal Science and Technology, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Hany M. Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Wei-Yi Huang
- School of Animal Science and Technology, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, People’s Republic of China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province, People’s Republic of China
| |
Collapse
|
8
|
Brattig NW, Bergquist R, Qian MB, Zhou XN, Utzinger J. Helminthiases in the People's Republic of China: Status and prospects. Acta Trop 2020; 212:105670. [PMID: 32841589 DOI: 10.1016/j.actatropica.2020.105670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Helminth infections, many of them listed as neglected tropical diseases by the World Health Organization, remain a public health issue in many parts of the world. The People's Republic of China (P.R. China) stands out due to impressive progress in the control and local elimination of helminth infections. An important contextual factor is P.R. China's sustained social and economic development that allowed implementation of health-related poverty alleviation, improving water, sanitation and hygiene, enhancing information, education and communication, coupled with major engineering and infrastructure development and intersectoral collaboration. Nonetheless, food-borne trematodiases, soil-transmitted helminthiases, echinococcosis, cysticercosis/taeniasis and schistosomiasis still exert a considerable burden in P.R. China, even though the numbers of infected people have decreased substantially since the new millennium. This special issue of Acta Tropica provides a comprehensive update of the current knowledge of the main helminth infections in P.R. China, summarises progress in research and discusses future prospects for gaining and sustaining control towards the final goal of breaking transmission and hence, eliminating helminthiases. It consists of 34 articles with a wide coverage that can be grouped into six domains: (i) epidemiological assessment and disease burden estimates; (ii) diagnostics and antigen characterisation; (iii) drug and vaccine development; (iv) host-parasite interactions and snail genetics; (v) surveillance and public health response; and (vi) capacity building and international cooperation. The control and elimination of helminthiases not only furthers the health and wellbeing of the Chinese people, but also provides innovative approaches, tools and strategies, which can be adopted and applied in other countries and regions of the world where helminthiases still prevail.
Collapse
|
9
|
Bai X, Wang N, Zhou J, Cui M, Jing X, Liu N. DX5 + NKT cells' increase was correlated with liver damage in FVB mice not in BALB/c mice infected by Clonorchis sinensis. Parasite Immunol 2020; 43:e12796. [PMID: 32984976 DOI: 10.1111/pim.12796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/26/2022]
Abstract
AIMS DX5+ NKT cells' distribution and population change in BALB/c and FVB mice infected by C sinensis and their function in liver damage were investigated. METHODS AND RESULTS Mice were infected by Clonorchis sinensis metacercariae, and lymphocytes were isolated from the livers, spleens and peripheral blood. NK, DX5+ NKT, INF-γ+ DX5+ NKT cells and liver fibrosis were analysed. The DX5+ NKT cells displayed the largest amount in normal BALB/c mice liver followed by peripheral blood and spleen. Although the hepatic DX5+ NKT cells of BALB/c mice were more than that of FVB mice, they did not show significant percentage change after C sinensis infection. The hepatic DX5+ NKT cells of FVB mice increased remarkably after infection accompanied with heavier liver injury and fibrosis than the BALB/c mice. And hydroxyproline content was also positively correlated with DX5+ NKT cells only in FVB mice. However, the increase of IFN-γ producing DX5+ NKT cells was lower in FVB mice than in BALB/c mice which showed sharp increase with mild liver damage after infection. The frequencies of anti-fibrotic NK cells were similar in both of the two mouse strains. CONCLUSIONS C sinensis could induce different DX5+ NKT cells responses in different mouse strains which may play roles in liver injury and fibrosis in FVB mice.
Collapse
Affiliation(s)
- Xuelian Bai
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Nan Wang
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Jie Zhou
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Min Cui
- Department of Pediatrics, Binzhou City People's Hospital, Binzhou, China
| | - Xuening Jing
- Department of Immunology, Shandong College of Traditional Chinese Medicine, Yantai, China
| | - Naiguo Liu
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|