1
|
Liang L, Dang B, Ouyang X, Zhao X, Huang Y, Lin Y, Cheng X, Xie G, Lin J, Mi P, Ye Z, Guleng B, Cheng SC. Dietary succinate supplementation alleviates DSS-induced colitis via the IL-4Rα/Hif-1α Axis. Int Immunopharmacol 2025; 152:114408. [PMID: 40086056 DOI: 10.1016/j.intimp.2025.114408] [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: 12/05/2024] [Revised: 02/22/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Abstract
Inflammatory bowel disease (IBD) remains a pressing global health challenge, necessitating novel therapeutic strategies. Succinate, a metabolite known for its role in type 2 immunity and tuft cell activation in the small intestine, presents its potential in IBD management. However, its impact on colonic inflammation has not been explored. Here, we demonstrate that succinate administration induces a type 2 immune response, significantly alleviating dextran sulfate sodium (DSS)-induced colonic inflammation. Succinate enhances antibacterial capacity, reduces intestinal permeability, and reshapes the colonic cytokine milieu. Mechanistically, succinate promotes myeloid cell expansion in peripheral blood, mesenteric lymph nodes, and the colonic lamina propria. The protective effects of succinate were abolished in Ccr2-/- mice, confirming the role of monocyte recruitment, but persisted in Rag1-/- mice, indicating independence from adaptive immunity. Adoptive transfer of monocytes from succinate-treated donors mitigated intestinal inflammation in recipient mice. Transcriptomic analysis revealed heightened expression of Il1b and Il6, and higher lactate production in monocytes upon lipopolysaccharide (LPS) stimulation, highlighting a reprogrammed pro-inflammatory trained immunity phenotype. Finally, we identify the IL-4Rα/Hif-1α axis is critical for succinate-mediated protection. These findings reveal the ability of succinate to reprogram monocytes into protective intestinal macrophages via induction of type 2 response, restoring homeostasis through enhanced barrier function and immune modulation. Our study positions thus uncover succinate as a promising therapeutic candidate for IBD.
Collapse
Affiliation(s)
- Laiying Liang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China; Department of Laboratory Medicine, West China Xiamen Hospital of Sichuan University, Xiamen 361000, China
| | - Buyun Dang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China; State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xiaomei Ouyang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Xianling Zhao
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Yongdong Huang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Ying Lin
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Xiaoshen Cheng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Guijing Xie
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Junhui Lin
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Peng Mi
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Zhenyu Ye
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Bayasi Guleng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China.
| | - Shih-Chin Cheng
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Cancer Research Center & Institute of Microbial Ecology, School of Medicine, Xiamen University, Xiamen 361004, China; State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
2
|
El Skhawy N, Eissa MM, Allam M, Eleryan EM. Immunomodulatory role of Trichinella spiralis-derived antigen on imiquimod-induced psoriasis in mice model. Parasitol Res 2024; 123:397. [PMID: 39592463 DOI: 10.1007/s00436-024-08415-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024]
Abstract
The immunomodulatory activity of parasites has been extensively investigated in multiple immune-related diseases. However, dermatological diseases have been off the list for a long time despite their vast incidence and the deleterious consequences of some of them. This study explored the immunomodulatory role of autoclaved Trichinella spiralis (T. spiralis) larvae antigen (ATSLA) as a psoriasis immunotherapeutic candidate in a mice model. Psoriasis was induced in Swiss albino mice using commercial imiquimod cream (IMQ). Mice were randomly divided into the IMQ untreated control group and the IMQ treated group that was treated with ATSLA twice, on day 0 and day 3. Additional mice served as normal controls. Assessment of skin thickness, erythema, and scales was recorded. Total skin scores were calculated. Skin MDA levels, splenic indices, serum and skin IL-23, and tumor necrosis factor alpha (TNF-α) were measured. Skin sections were stained with H&E and immune stained for CD68-positive cells using immunohistochemistry. Treatment with ATSLA significantly reduced skin thickness, erythema, scales, and total skin scores in the IMQ-treated group compared to the untreated control. This was accompanied by a reduction in the splenic index, skin MDA levels, IL-23, and TNF-α in both the skin and serum of the treated group. Pathologically, skin sections of the treated group showed less epidermal thickness, acanthosis, hyperkeratosis, and CD68 cell count. The study concluded the immunotherapeutic activity of ATSLA in experimental psoriatic skin lesions. This will enrich the psoriasis immunotherapeutic list with novel candidates of parasitic origin.
Collapse
Affiliation(s)
- Nahla El Skhawy
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Maha M Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Maram Allam
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eman M Eleryan
- Department of Dermatology and Venereology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
3
|
Long SR, Shang WX, Zhang HR, Jiang M, Wang JJ, Liu RD, Wang ZQ, Cui J, Sun H. Trichinella-derived protein ameliorates colitis by altering the gut microbiome and improving intestinal barrier function. Int Immunopharmacol 2024; 127:111320. [PMID: 38064817 DOI: 10.1016/j.intimp.2023.111320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/29/2023] [Accepted: 11/27/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) encompasses Crohn's Disease and Ulcerative Colitis. Reports have highlighted the potential use of helminths or their byproducts as a possible treatment for IBD; however, the mechanisms underlying their ability to modulate inflammation remain incompletely understood. In the present study, we analyze the possible mechanism of a serine protease inhibitor from adult T. spiralis excretion-secretion products (rTsSPI) on the improvement of colitis. METHODS The immune protective effect of rTsSPI was studied by using DSS or Salmonella-induced colitis in female C56BL/6 mice. The effect of rTsSPI on the immune and inflammatory responses, gut microbiota, permeability of colon epithelium and junction proteins was analyzed. RESULTS Treating mice with rTsSPI induced type 2 immunity and significantly attenuated clinical symptoms, macroscopical and histological features of DSS or bacteria-induced colonic inflammation. This was accompanied by decreasing neutrophil recruitment in the colonic lamina propria, and reducing TNF-α mRNA levels in the colon; in contrast, the recruitment of M2 macrophages, the expression level of IL-10 and adhesion molecules increased in the colon tissue. Moreover, treatment with rTsSPI led to an improvement in gut microbiota diversity, as well as an increase in the abundance of the bacterial genera Bifidobacterium and Ruminclostridium 5. CONCLUSIONS Collective findings suggest that pretreatment with rTsSPI can ameliorate colitis in mice by inducing a Th2-type response with M2 macrophages. Data also indicate that immunotherapy with rTsSPI represents an additional strategy to ameliorate inflammatory processes in IBD by enhancing probiotic colonization and maintaining intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Shao Rong Long
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Wen Xuan Shang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Hui Ran Zhang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Miao Jiang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jing Jing Wang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jing Cui
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Hualei Sun
- Department of Nutrition, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
4
|
Wu L, Yin W, Wen J, Wang S, Li H, Wang X, Zhang W, Duan S, Zhu Q, Gao E, Wu S, Zhan B, Zhou R, Yang X. Excretory/secretory products from Trichinella spiralis adult worms ameliorate myocardial infarction by inducing M2 macrophage polarization in a mouse model. Parasit Vectors 2023; 16:362. [PMID: 37845695 PMCID: PMC10577921 DOI: 10.1186/s13071-023-05930-x] [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: 05/26/2023] [Accepted: 08/14/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Ischemia-induced inflammatory response is the main pathological mechanism of myocardial infarction (MI)-caused heart tissue injury. It has been known that helminths and worm-derived proteins are capable of modulating host immune response to suppress excessive inflammation as a survival strategy. Excretory/secretory products from Trichinella spiralis adult worms (Ts-AES) have been shown to ameliorate inflammation-related diseases. In this study, Ts-AES were used to treat mice with MI to determine its therapeutic effect on reducing MI-induced heart inflammation and the immunological mechanism involved in the treatment. METHODS The MI model was established by the ligation of the left anterior descending coronary artery, followed by the treatment of Ts-AES by intraperitoneal injection. The therapeutic effect of Ts-AES on MI was evaluated by measuring the heart/body weight ratio, cardiac systolic and diastolic functions, histopathological change in affected heart tissue and observing the 28-day survival rate. The effect of Ts-AES on mouse macrophage polarization was determined by stimulating mouse bone marrow macrophages in vitro with Ts-AES, and the macrophage phenotype was determined by flow cytometry. The protective effect of Ts-AES-regulated macrophage polarization on hypoxic cardiomyocytes was determined by in vitro co-culturing Ts-AES-induced mouse bone marrow macrophages with hypoxic cardiomyocytes and cardiomyocyte apoptosis determined by flow cytometry. RESULTS We observed that treatment with Ts-AES significantly improved cardiac function and ventricular remodeling, reduced pathological damage and mortality in mice with MI, associated with decreased pro-inflammatory cytokine levels, increased regulatory cytokine expression and promoted macrophage polarization from M1 to M2 type in MI mice. Ts-AES-induced M2 macrophage polarization also reduced apoptosis of hypoxic cardiomyocytes in vitro. CONCLUSIONS Our results demonstrate that Ts-AES ameliorates MI in mice by promoting the polarization of macrophages toward the M2 type. Ts-AES is a potential pharmaceutical agent for the treatment of MI and other inflammation-related diseases.
Collapse
Affiliation(s)
- Lingqin Wu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Wenhui Yin
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Jutai Wen
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Shuying Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Weixiao Zhang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Shuyao Duan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Qiuyu Zhu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Erhe Gao
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Shili Wu
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rui Zhou
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
5
|
Ma ZR, Li ZL, Zhang N, Lu B, Li XW, Huang YH, Nouhoum D, Liu XS, Xiao KC, Cai LT, Xu SR, Yang XXO, Huang SQ, Wu X. Inhibition of GSDMD-mediated pyroptosis triggered by Trichinella spiralis intervention contributes to the alleviation of DSS-induced ulcerative colitis in mice. Parasit Vectors 2023; 16:280. [PMID: 37580819 PMCID: PMC10424392 DOI: 10.1186/s13071-023-05857-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/30/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is increasing worldwide. Although there is currently no completely curative treatment, helminthic therapy shows certain therapeutic potential for UC. Many studies have found that Trichinella spiralis (T.s) has a protective effect on UC, but the specific mechanism is still unclear. METHODS Balb/c mice drank dextran sulfate sodium (DSS) to induce acute colitis and then were treated with T.s. In vitro experiments, the LPS combination with ATP was used to induce the pyroptosis model, followed by intervention with crude protein from T.s (T.s cp). Additionally, the pyroptosis agonist of NSC or the pyroptosis inhibitor vx-765 was added to intervene to explore the role of pyroptosis in DSS-induced acute colitis. The degree of pyroptosis was evaluated by western blot, qPCR and IHC, etc., in vivo and in vitro. RESULTS T.s intervention significantly inhibited NLRP3 inflammasome activation and GSDMD-mediated pyroptosis by downregulating the expression of pyroptosis-related signatures in vitro (cellular inflammatory model) and in vivo (DSS-induced UC mice model). Furthermore, blockade of GSDMD-mediated pyroptosis by the caspase-1 inhibitor vx-765 has a similar therapeutic effect on DSS-induced UC mice with T.s intervention, thus indicating that T.s intervention alleviated DSS-induced UC in mice by inhibiting GSDMD-mediated pyroptosis. CONCLUSION This study showed that T.s could alleviate the pathological severity UC via GSDMD-mediated pyroptosis, and it provides new insight into the mechanistic study and application of helminths in treating colitis.
Collapse
Affiliation(s)
- Zhen-Rong Ma
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Zhuo-Lin Li
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Ni Zhang
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Bin Lu
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xuan-Wu Li
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Ye-Hong Huang
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Dibo Nouhoum
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xian-Shu Liu
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Ke-Chun Xiao
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Li-Ting Cai
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Shao-Rui Xu
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xue-Xian O Yang
- Department of Molecular & Genetic and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Shuai-Qin Huang
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China.
| | - Xiang Wu
- Department of Medical Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
6
|
Xie Y, Shao F, Duan X, Ding J, Ning Y, Sun X, Xia L, Pan J, Chen J, He S, Shen D, Qi C. Whole β-glucan particle attenuates AOM/DSS-induced colorectal tumorigenesis in mice via inhibition of intestinal inflammation. Front Pharmacol 2023; 14:1017475. [PMID: 36713833 PMCID: PMC9877317 DOI: 10.3389/fphar.2023.1017475] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Yeast β-glucan is a polysaccharide purified from the Saccharomyces cerevisiae cell wall, and its multiple biological activities are essential for immune regulation. However, the effect of β-glucan on the intestinal immune response during colitis-associated colorectal cancer (CAC) is unclear. Here, we explore the possible role of β-glucan in the development of CAC. Wild type (WT) mice with CAC induced by azoxmethane (AOM) and dextran sodium sulfate (DSS) had fewer tumors than untreated mice after oral β-glucan because of increased antitumor dendritic cells (DCs) in the tumor microenvironment, resulting in more CD8+ T cells and the production of related cytokines. β-glucan also increased resistance to DSS-induced chronic colitis by reshaping the inflammatory microenvironment. These data suggest that β-glucan improves experimental intestinal inflammation and delays the development of CAC. Therefore, β-glucan is feasible for treating chronic colitis and CAC in clinical practice.
Collapse
Affiliation(s)
- Yewen Xie
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Fang Shao
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Xuehan Duan
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Jun Ding
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Yongling Ning
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Xiao Sun
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Lei Xia
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Jie Pan
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Jie Chen
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Shuyan He
- Department of Oncology, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, Jiangsu, China
| | - Dong Shen
- Department of Oncology, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, Jiangsu, China,*Correspondence: Chunjian Qi, ; Dong Shen,
| | - Chunjian Qi
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,*Correspondence: Chunjian Qi, ; Dong Shen,
| |
Collapse
|
7
|
Khueangchiangkhwang S, Wu Z, Nagano I, Maekawa Y. Trichinella pseudospiralis-secreted 53 kDa protein ameliorates imiquimod-induced psoriasis by inhibiting the IL-23/IL-17 axis in mice. Biochem Biophys Rep 2022; 33:101415. [PMID: 36620087 PMCID: PMC9813687 DOI: 10.1016/j.bbrep.2022.101415] [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: 09/14/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Trichinella infection can experimentally ameliorate many autoimmune diseases. However, the immune mechanism of the amelioration and the identification of corresponding Trichinella-derived molecule(s) are still not fully elucidated. Fifty-three kDa excretory-secretory (ES) protein from Trichinella pseudospiralis (Tpp53) is a molecule like TsP53 reported as a protein exerting immune-inhibitory effect in T. spiralis. In this study, we investigated the immunomodulatory effect of Tpp53 using imiquimod (IMQ)-induced psoriasis-like dermatitis model, which is a mouse model of autoimmune disease with the pathogenic interleukin 17 (IL-17) producing CD4+ T cells (Th17) via IL-23/IL17 axis. Administrating the recombinant Tpp53 (rTpp53) mixed with IMQ cream on the skin of mice ameliorated psoriatic lesions, as revealed by the improvement of erythema, scaling, skin thickening, epidermis hyperplasia and parakeratosis, thickening of acanthosis cell layer, epidermal extension of dermis, less infiltration of inflammatory cells, and decreased expression of inflammatory marker. The increased expression of the factors related to the IL-23/IL-17 axis, including IL-17A, IL-6, Il17F and Il23a, in the skins of IMQ-treated mice was inhibited by rTpp53 treatment. Moreover, the expression of activated keratinocyte-produced cytokines, chemokines, and antimicrobial peptides in the skin was also down-regulated in rTpp53-treated IMQ-treated mice. Co-culture of splenocytes with rTpp53 inhibited IL-17A and treatment of macrophages with rTpp53 reduced IL-6 production. Overall, our study revealed that the Trichinella-secreted 53 kDa ES protein could ameliorate IMQ-induced psoriasis by inhibiting the IL-23/IL-17 axis, suggesting that Tpp53 might involve in regulating host Th17 for immune evasion and have an alternative potential for psoriasis therapy.
Collapse
Affiliation(s)
| | - Zhiliang Wu
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan,Cocorresponding author. 1-1 Yanagido, Gifu, 501-1194, Japan.
| | - Isao Nagano
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yoichi Maekawa
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan,Preemptive Food Research Center, Gifu University, Gifu, Japan,Corresponding author. 1-1 Yanagido, Gifu, 501-1194, Japan.
| |
Collapse
|
8
|
Peng J, Federman HG, Hernandez C, Siracusa MC. Communication is key: Innate immune cells regulate host protection to helminths. Front Immunol 2022; 13:995432. [PMID: 36225918 PMCID: PMC9548658 DOI: 10.3389/fimmu.2022.995432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Parasitic helminth infections remain a significant global health issue and are responsible for devastating morbidity and economic hardships. During infection, helminths migrate through different host organs, which results in substantial tissue damage and the release of diverse effector molecules by both hematopoietic and non-hematopoietic cells. Thus, host protective responses to helminths must initiate mechanisms that help to promote worm clearance while simultaneously mitigating tissue injury. The specialized immunity that promotes these responses is termed type 2 inflammation and is initiated by the recruitment and activation of hematopoietic stem/progenitor cells, mast cells, basophils, eosinophils, dendritic cells, neutrophils, macrophages, myeloid-derived suppressor cells, and group 2 innate lymphoid cells. Recent work has also revealed the importance of neuron-derived signals in regulating type 2 inflammation and antihelminth immunity. These studies suggest that multiple body systems coordinate to promote optimal outcomes post-infection. In this review, we will describe the innate immune events that direct the scope and intensity of antihelminth immunity. Further, we will highlight the recent progress made in our understanding of the neuro-immune interactions that regulate these pathways and discuss the conceptual advances they promote.
Collapse
Affiliation(s)
- Jianya Peng
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Hannah G. Federman
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Christina M. Hernandez
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
| | - Mark C. Siracusa
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, United States
- *Correspondence: Mark C. Siracusa,
| |
Collapse
|
9
|
Jin X, Liu Y, Vallee I, Karadjian G, Liu M, Liu X. Lentinan -triggered butyrate-producing bacteria drive the expulsion of the intestinal helminth Trichinella spiralis in mice. Front Immunol 2022; 13:926765. [PMID: 35967395 PMCID: PMC9371446 DOI: 10.3389/fimmu.2022.926765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022] Open
Abstract
Trichinellosis caused by Trichinella spiralis is a serious zoonosis with a worldwide distribution. Lentinan (LNT) is known to modulate the intestinal environment with noted health benefits, yet the effect of LNT against intestinal helminth is unknown. In our study, we first observed that LNT could trigger worm expulsion by promoting mucus layer functions through alteration of gut microbiota. LNT restored the abundance of Bacteroidetes and Proteobacteria altered by T. spiralis infection to the control group level. Interestingly, LNT triggered the production of butyrate. Then, we determined the deworming capacity of probiotics (butyrate-producing bacteria) in mice. Collectively, these findings indicated that LNT could modulate intestinal dysbiosis by T. spiralis, drive the expulsion of intestinal helminth and provided an easily implementable strategy to improve the host defence against T. spiralis infection.
Collapse
Affiliation(s)
- Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yi Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Isabelle Vallee
- UMR BIPAR, Anses, Ecole Nationale Vétérinaire d’Alfort, INRA, University Paris-Est, Animal Health Laboratory, Maisons-Alfort, France
| | - Gregory Karadjian
- UMR BIPAR, Anses, Ecole Nationale Vétérinaire d’Alfort, INRA, University Paris-Est, Animal Health Laboratory, Maisons-Alfort, France
| | - Mingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xiaolei Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
- *Correspondence: Xiaolei Liu,
| |
Collapse
|
10
|
β-Glucan-triggered Akkermansia muciniphila expansion facilitates the expulsion of intestinal helminth via TLR2 in mice. Carbohydr Polym 2022; 275:118719. [PMID: 34742442 DOI: 10.1016/j.carbpol.2021.118719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022]
Abstract
Trichinellosis caused by Trichinella spiralis is a serious zoonosis with a worldwide. β-Glucans (BG) are readily used across the world with noted health benefits, yet the effect and mechanism of BG on host defense against helminth infection remain poorly understood. We observed that BG could trigger worm expulsion via mucus layer independently of type 2 immunity, but was dependent on the gut microbiota in mice. BG restored the abundance of Bacteroidetes and Proteobacteria changed by T. spiralis infection to the control group level and markedly increased the relative abundance of Verrucomicrobia. Akkermansia (belonging to Verrucomicrobia) were significantly expanded in the BG + T. spiralis group. Notably, daily oral supplementation of pasteurized A. muciniphila has a stronger deworming effect than live bacteria and interacted with TLR2. These findings of this study is an easily implementable strategy to facilitate expulsion of gastrointestinal helminth.
Collapse
|
11
|
Arai T, Lopes F. Potential of human helminth therapy for resolution of inflammatory bowel disease: The future ahead. Exp Parasitol 2021; 232:108189. [PMID: 34848244 DOI: 10.1016/j.exppara.2021.108189] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/06/2021] [Accepted: 11/24/2021] [Indexed: 12/23/2022]
Abstract
Inflammatory bowel disease (IBD) is associated with a dysregulated mucosal immune response in the gastrointestinal tract. The number of patients with IBD has increased worldwide, especially in highly industrialized western societies. The population of patients with IBD in North America is forecasted to reach about four million by 2030; meanwhile, there is no definitive therapy for IBD. Current anti-inflammatory, immunosuppressive, or biological treatment may induce and maintain remission, but not all patients respond to these treatments. Recent studies explored parasitic helminths as a novel modality of therapy due to their potent immunoregulatory properties in humans. Research using IBD animal models infected with a helminth or administered helminth-derived products such as excretory-secretory products has been promising, and helminth-microbiota interactions exert their anti-inflammatory effects by modulating the host immunity. Recent studies also indicate that evidence that helminth-derived metabolites may play a role in anticolitic effects. Thus, the helminth shows a potential benefit for treatment against IBD. Here we review the current feasibility of "helminth therapy" from the laboratory for application in IBD management.
Collapse
Affiliation(s)
- Toshio Arai
- Institution of Parasitology, McGill University, Quebec, Canada; Department of Gastroenterology, Hashimoto Municipal Hospital, Wakayama, Japan
| | - Fernando Lopes
- Institution of Parasitology, McGill University, Quebec, Canada.
| |
Collapse
|
12
|
Liu Y, Liu X, Yang L, Qiu Y, Pang J, Hu X, Dong Z, Liu Z, Jin X. Adjuvanticity of β -Glucan for Vaccine Against Trichinella spiralis. Front Cell Dev Biol 2021; 9:701708. [PMID: 34322488 PMCID: PMC8313300 DOI: 10.3389/fcell.2021.701708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/15/2021] [Indexed: 11/30/2022] Open
Abstract
In the past 30 years, few researches focus on the efficacy of adjuvant against Trichinella spiralis infection. Identifying new, improved vaccine adjuvants for T. spiralis infection are required. β-glucan are effective and safe as adjuvant for infectious diseases. In this paper, we first observed the adjuvanticity of β-glucan as adjuvant for defensing helminth T. spiralis in vivo. We showed that IgG and IgE were elevated in the mice immunized with β-glucan combined with recombinant T. spiralis serine protease inhibitor (rTs-Serpin), which is one of the vaccine candidates. Furthermore, in vitro, the combination of β-glucan and rTs-Serpin enhanced the maturation of bone marrow dendritic cells (BMDCs) compared to rTs-Serpin alone. We showed that β-glucan + rTs-Serpin –treated BMDCs secreted higher production of IL-12 and IL-10. Moreover, β-glucan + rTs-Serpin –treated BMDCs not only promoted the population of CD4+ IFN-γ+ T cells, but also enhanced the population of CD4+ IL-4+ T cells. These findings suggested that β-glucan, as an adjuvant, have the capacity to protect against T. spiralis infection via activating both Th1 and Th2 immune response.
Collapse
Affiliation(s)
- Yi Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Li Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yangyuan Qiu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Jianda Pang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiaoxiang Hu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Zijian Dong
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Zengshan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xuemin Jin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| |
Collapse
|
13
|
Yim SK, Kim SW, Lee ST. Efficient Stool Collection Methods for Evaluating the Diarrhea Score in Mouse Diarrhea Models. In Vivo 2021; 35:2115-2125. [PMID: 34182487 DOI: 10.21873/invivo.12481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM The mouse diarrhea score is usually determined by evaluating stool consistency and shape. Thus, defecated stools should be collected without damage or contamination. The study aimed to develop improved mouse stool collection methods and diarrhea-scoring criteria. MATERIALS AND METHODS We developed improved stool collection methods (paper towel methods) and compared them with previously used ones (stool collection using regular cages containing bedding chips or filter paper and metabolic cages). RESULTS Compared to previously used methods, paper towel methods collected stools without bedding chips-induced contamination, mouse body/foot-induced damage, or sampling errors. When using paper towel methods, wet stools create water marks (diarrhea marks) on paper towels with strong water absorption capacity, by which diarrheal severity can be analyzed semi-quantitatively. To improve the objectivity in determining diarrhea scores, practical diarrhea-scoring criteria were also proposed. CONCLUSION These results would be helpful to researchers facing difficulties in evaluating the mouse diarrhea score.
Collapse
Affiliation(s)
- Sung Kyun Yim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jeonbuk National University Medical School and Hospital, Jeonju, Republic of Korea.,Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju, Republic of Korea
| | - Sang Wook Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jeonbuk National University Medical School and Hospital, Jeonju, Republic of Korea.,Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju, Republic of Korea
| | - Soo Teik Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jeonbuk National University Medical School and Hospital, Jeonju, Republic of Korea; .,Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
14
|
Jin X, Bai X, Zhao Y, Dong Z, Pang J, Liu M, Liu X. Nrf2 Participates in M2 Polarization by Trichinella spiralis to Alleviate TNBS-Induced Colitis in Mice. Front Immunol 2021; 12:698494. [PMID: 34249002 PMCID: PMC8261282 DOI: 10.3389/fimmu.2021.698494] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
Trichinella spiralis induced alternative activated macrophages (M2), leading to protect against Crohn's disease, known as Th1 -related inflammation, which enhances oxidative stress in the host. However, the relationship of oxidative stress and T. spiralis -mediated immune response is still unknown. In our study, we showed that nuclear factor erythroid 2-related factor-2 (Nrf2), a key transcription factor in antioxidant, participated in M2 polarization induced by T. spiralis muscle larval excretory/secretory (ES) products in vitro. ES -treated M2 were injected intravenously after TNBS challenge and we demonstrated that ES-M could alleviate the severity of the colitis in mice. Adoptive transfer of ES -treated M2 decreased the level of IFN-γ and increased the levels of IL-4 and IL-10 in vivo. However, the capacity of ES -treated Nrf2 KO macrophages to treat colitis was dramatically impaired. ES -treated Nrf2 KO macrophages was insufficient to result in the elevated levels of IL-4 and IL-10. These findings indicate that Nrf2 was required for M2 polarization induced by T. spiralis ES to alleviate colitis in mice.
Collapse
Affiliation(s)
- Xuemin Jin
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xue Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ying Zhao
- Department of Nephrology, First Hospital of Jilin University, Changchun, China
| | - Zijian Dong
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianda Pang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
15
|
Long SR, Liu RD, Kumar DV, Wang ZQ, Su CW. Immune Protection of a Helminth Protein in the DSS-Induced Colitis Model in Mice. Front Immunol 2021; 12:664998. [PMID: 33995396 PMCID: PMC8117093 DOI: 10.3389/fimmu.2021.664998] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/08/2021] [Indexed: 12/23/2022] Open
Abstract
Inflammatory bowel disease (IBD) increases the risk of colorectal cancer, and it has the potential to diminish the quality of life. Recent clinical and experimental evidence demonstrate protective aspects of parasitic helminth infection against IBD. Reports have highlighted the potential use of helminths and their byproducts as potential treatment for IBD. In the current study, we studied the effect of a newborn larvae-specific serine protease from Trichinella spiralis (TsSp) on the host immune and inflammatory responses. A 49-kDa recombinant TsSp (rTsSp) was expressed in Escherichia coli BL21 (DE3) and purified. The cytotoxicity of rTsSp was analyzed. The immune protective effect of rTsSp was studied by using dextran sodium sulfate (DSS)-induced mouse colitis model. The result illustrated that rTsSp has no toxic effects on cells. We further demonstrated that administration of the rTsSp without the additional adjuvant before the induction of DSS-induced colitis reduced the severity of intestinal inflammation and the disease index; it suppressed macrophage infiltration, reduced TNF-α secretion, and induced IL-10 expression. Our findings suggest therapeutic potential of rTsSp on colitis by altering the effect of macrophages. Data also suggest immunotherapy with rTsSp holds promise for use as an additional strategy to positively modulate inflammatory processes involved in IBD.
Collapse
Affiliation(s)
- Shao Rong Long
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China.,Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Ruo Dan Liu
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Deepak Vijaya Kumar
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Zhong Quan Wang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Chien-Wen Su
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
16
|
Li H, Qiu D, Yang H, Yuan Y, Wu L, Chu L, Zhan B, Wang X, Sun Y, Xu W, Yang X. Therapeutic Efficacy of Excretory-Secretory Products of Trichinella spiralis Adult Worms on Sepsis-Induced Acute Lung Injury in a Mouse Model. Front Cell Infect Microbiol 2021; 11:653843. [PMID: 33842398 PMCID: PMC8024484 DOI: 10.3389/fcimb.2021.653843] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/08/2021] [Indexed: 12/29/2022] Open
Abstract
Acute lung injury (ALI) is a common complication of systemic inflammation or sepsis with high morbidity and mortality. Although many studies have confirmed that helminth-derived proteins had strong immunomodulatory functions and could be used to treat inflammatory diseases, there is no report on the therapeutic effect of excretory-secretory products of Trichinella spiralis adult worms (Ts-AES) on sepsis-induced ALI. In this study, the therapeutic efficacy of Ts-AES on sepsis-induced ALI and the underlying immunological mechanism and the signaling pathway were investigated. The results indicated that after being treated with Ts-AES, the survival rate of mice with CLP-induced sepsis was significantly increased to 50% for 72 hours after CLP surgery compared to PBS control group with all mice died. The sepsis-induced ALI was largely mitigated characterized by reduced inflammation cell infiltration and pathological changes in lung tissue, with decreased lung injury scores and lung wet/dry weight ratio. The therapeutic efficacy of Ts-AES is associated with stimulated Tregs response with increased regulatory cytokines IL-10 and TGF-β and downregulated pro-inflammatory cytokines (TNF-α, IL-6, IL-1β). The expression of HMGB1, TLR2 and MyD88 in lung tissue was inhibited after treatment of Ts-AES. Our results demonstrated that Ts-AES play an important role in immunomodulation and confer a therapeutic effect on sepsis-induced ALI through inhibiting pro-inflammatory cytokines. The activation of Tregs and increased level of regulatory cytokines IL-10 and TGF-β are possibly involved in the immunomodulatory functions of Ts-AES through HMGB1/TLR2/MyD88 signal pathway. The findings suggest Ts-AES is a potential therapeutic agent for prevention and treatment of sepsis-induced ALI and other inflammatory diseases.
Collapse
Affiliation(s)
- Huihui Li
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Dapeng Qiu
- Department of Orthopedics, Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Huijuan Yang
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Yuan Yuan
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Lingqin Wu
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Liang Chu
- Department of Orthopedics, Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Xiaoli Wang
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Yan Sun
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Wei Xu
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Xiaodi Yang
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| |
Collapse
|
17
|
Xie H, Wu L, Chen X, Gao S, Li H, Yuan Y, Liang J, Wang X, Wang S, Xu C, Chu L, Zhan B, Zhou R, Yang X. Schistosoma japonicum Cystatin Alleviates Sepsis Through Activating Regulatory Macrophages. Front Cell Infect Microbiol 2021; 11:617461. [PMID: 33718268 PMCID: PMC7943722 DOI: 10.3389/fcimb.2021.617461] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Multi-organ failure caused by the inflammatory cytokine storm induced by severe infection is the major cause of death for sepsis. Sj-Cys is a cysteine protease inhibitor secreted by Schistosoma japonicum with strong immunomodulatory functions on host immune system. Our previous studies have shown that treatment with Sj-Cys recombinant protein (rSj-Cys) attenuated inflammation caused by sepsis. However, the immunological mechanism underlying the immunomodulation of Sj-Cys for regulating inflammatory diseases is not yet known. In this study, we investigated the effect of Sj-Cys on the macrophage M2 polarization and subsequent therapeutic effect on sepsis. The rSj-Cys was expressed in yeast Pichia pastoris. Incubation of mouse bone marrow-derived macrophages (BMDMs) with yeast-expressed rSj-Cys significantly activated the polarization of macrophages to M2 subtype characterized by the expression of F4/80+ CD206+ with the elated secretion of IL-10 and TGF-β. Adoptive transfer of rSj-Cys treated BMDMs to mice with sepsis induced by cecal ligation and puncture (CLP) significantly improved their survival rates and the systemic clinical manifestations of sepsis compared with mice receiving non-treated normal BMDMs. The therapeutic effect of Sj-Cys-induced M2 macrophages on sepsis was also reflected by the reduced pathological damages in organs of heart, lung, liver and kidney and reduced serological levels of tissue damage-related ALT, AST, BUN and Cr, associated with downregulated pro-inflammatory cytokines (IFN-gamma and IL-6) and upregulated regulatory anti-inflammatory cytokines (IL-10 and TGF-β). Our results demonstrated that Sj-Cys is a strong immunomodulatory protein with anti-inflammatory features through activating M2 macrophage polarization. The findings of this study suggested that Sj-Cys itself or Sj-Cys-induced M2 macrophages could be used as therapeutic agents in the treatment of sepsis or other inflammatory diseases.
Collapse
Affiliation(s)
- Hong Xie
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Lingqin Wu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Pediatric, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xingzhi Chen
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Shifang Gao
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Yuan Yuan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Jinbao Liang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Shuying Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Pediatric, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Changyan Xu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Liang Chu
- Department of General Surgery, Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Rui Zhou
- Department of Pediatric, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases of Bengbu Medical College, Bengbu, China
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| |
Collapse
|
18
|
Xu J, Yue WW, Xu YXY, Hao HN, Liu RD, Long SR, Wang ZQ, Cui J. Molecular characterization of a novel aspartyl protease-1 from Trichinella spiralis. Res Vet Sci 2020; 134:1-11. [PMID: 33276221 DOI: 10.1016/j.rvsc.2020.11.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/28/2020] [Accepted: 11/15/2020] [Indexed: 02/06/2023]
Abstract
The aim of this study was to characterize the biological properties of a novel aspartic protease-1 from Trichinella spiralis (TsASP1) and evaluate its potential in inducing immune response. TsASP1 gene was cloned and expressed in Escherichia coli BL21 (DE3). On Western blotting analysis with anti-rTsASP1 serum, native TsASP1 was detected in various T. spiralis phases other than newborn larvae (NBL). qPCR results showed that TsASP1 transcription was the highest in intestinal infective larvae (IIL) and the lowest in the NBL stage. Immunofluorescence test result shows that native TsASP1 was principally localized in stichosome, muscle cells of muscle larvae (ML) and IIL, and surrounded intrauterine embryos in female adult worms (AW). After silencing TsASP1 gene of the ML by siRNA, the worm development was significantly inhibited, showed by shorter AW and more wrinkles and longitudinal crack on epicuticle of AW on scanning electron microscopy; the AW and ML burdens were reduced by 41.82 and 56.36% respectively, compared with the control siRNA or PBS group (P < 0.001). Immunization of mice with rTsASP1 elicited an evident antibody response (serum IgG, IgG1/IgG2a and enteral sIgA), and systemic (spleen) and intestinal local mucosal (mesenteric lymph node) cellular immune response, demonstrated by a prominent elevation of IFN-γ and IL-4. The results suggested TsASP1 participated in T. spiralis development and survival in host, and immunization of mice with rTsASP1 induced systemic/intestinal local mucosal humoral and cellular immune response against Trichinella.
Collapse
Affiliation(s)
- Jia Xu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, PR China
| | - Wen Wen Yue
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, PR China
| | - Yang Xiu Yue Xu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, PR China
| | - Hui Nan Hao
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, PR China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, PR China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, PR China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, PR China.
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
19
|
Qu Z, Jin X, Wang Y, Yang Y, Yang Li, Bai X, Yang Y, Xu N, Wang X, Liu M. Effect of recombinant serine protease from newborn larval stage of Trichinella spiralis on 2,4,6-trinitrobenzene sulfonic acid-induced experimental colitis in mice. Acta Trop 2020; 211:105553. [PMID: 32562622 DOI: 10.1016/j.actatropica.2020.105553] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/25/2022]
Abstract
Inflammatory bowel disease (IBD) is a complex immune-mediated disease of gastrointestinal tract that is mainly driven by Th1/Th17 immune response. "Helminth therapy" has emerged, and helminth-derived immunoregulatory molecules are being used as safe and new therapeutic antigens for IBD. Recombinant serine protease (SP) from newborn Trichinella spiralis (T. spiralis) larvae (NBL) was expressed and purified. BALB/c mice were immunized with NBL-SP at 100 µg three times at an interval of 5 days. Experimental colitis was induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS) administration. The disease activity index (DAI) and macroscopic and microscopic scores of the colon were assessed to identify the effect of NBL-SP on experimental colitis. Cytokine production in the serum was analysed by meso scale discovery (MSD). Cytokine production in the colon was detected by ELISA. CD4+T cell differentiation was measured by flow cytometry. NBL-SP alleviated TNBS-induced colitis in mice. The DAI, macroscopic and microscopic scores and colon length all showed a positive intervention effect of NBL-SP on experimental colitis. NBL-SP can weaken the increase in IFN-γ, TNF-α and IL-17 production as well as CD4+ IFN-γ+T cell and CD4+IL-17+T cell populations induced by colitis. Furthermore, the levels of Th2-related cytokines (IL-4, IL-5) and regulatory cytokines (IL-10, TGF-β) were elevated meanwhile the ratio of regulatory T cells (Tregs) and CD4+ IL-4 + T cells were increased by NBL-SP. NBL-SP of T. spiralis had a potential protective effect against IBD. NBL-SP skewed the Th1 and Th17-mediated response towards the Th2 and Treg response.
Collapse
|
20
|
Yang Y, Liu L, Liu X, Zhang Y, Shi H, Jia W, Zhu H, Jia H, Liu M, Bai X. Extracellular Vesicles Derived From Trichinella spiralis Muscle Larvae Ameliorate TNBS-Induced Colitis in Mice. Front Immunol 2020; 11:1174. [PMID: 32595641 PMCID: PMC7300183 DOI: 10.3389/fimmu.2020.01174] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/12/2020] [Indexed: 01/01/2023] Open
Abstract
Helminths are masters at modulating the host immune response through a wide variety of versatile mechanisms. These complex strategies facilitate parasite survival in the host and can also be exploited to prevent chronic immune disorders by minimizing excessive inflammation. Extracellular vesicles (EVs) are small membrane-bound structures secreted by helminths which mediate immune evasion during parasite infection. The goal of this study was to investigate the immunoregulatory properties of Trichinella spiralis EVs (Ts-EVs) in a murine model of colitis. We found that Ts-EVs significantly ameliorated 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice. Ts-EVs alleviated intestinal epithelium barrier damage, markedly reduced pro-inflammatory cytokine secretion and neutrophil infiltration, and upregulated immunoregulatory cytokine expression in colon tissue. Ts-EVs also modulated the adaptive immune response by influencing T-cell composition. The numbers of Th1 and Th17 cells in MLNs, as well as the expression levels of Th1/Th17-associated cytokines and transcription factors in colon were reduced. In contrast, Th2 and Treg cells were increased after Ts-EVs treatment. Furthermore, sequencing of EV-derived microRNAs (miRNAs) indicated that an array of miRNAs was involved in the regulation of the host immune response, including inflammation. These findings expand our knowledge of host-parasite interactions, and may help design novel and effective strategies to prevent parasite infections or to treat inflammatory diseases like IBD. Further studies are needed to identify the specific cargo molecules carried by Ts-EVs and to clarify their roles during T. spiralis infection.
Collapse
Affiliation(s)
- Yong Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - YuanYuan Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Haining Shi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States
| | - Wanzhong Jia
- 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, China
| | - HongFei Zhu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hong Jia
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xue Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
21
|
Chen KY, Cheng CJ, Cheng CC, Jhan KY, Chen YJ, Wang LC. The excretory/secretory products of fifth-stage larval Angiostrongylus cantonensis induces autophagy via the Sonic hedgehog pathway in mouse brain astrocytes. PLoS Negl Trop Dis 2020; 14:e0008290. [PMID: 32479527 PMCID: PMC7289448 DOI: 10.1371/journal.pntd.0008290] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 06/11/2020] [Accepted: 04/13/2020] [Indexed: 12/22/2022] Open
Abstract
Angiostrongyliasis is induced by the nematode Angiostrongylus cantonensis and leads to eosinophilic meningitis and meningoencephalitis in humans. Excretory-secretory products (ESPs) are important investigation targets for studying the relationship between hosts and nematodes. These products assist worms in penetrating the blood-brain barrier and avoiding the host immune response. Autophagy is a catabolic process that is responsible for digesting cytoplasmic organelles, proteins, and lipids and removing them through lysosomes. This process is essential to cell survival and homeostasis during nutritional deficiency, cell injury and stress. In this study, we investigated autophagy induction upon treatment with the ESPs of the fifth-stage larvae (L5) of A. cantonensis and observed the relationship between autophagy and the Shh pathway. First, the results showed that A. cantonensis infection induced blood-brain barrier dysfunction and pathological changes in the brain. Moreover, A. cantonensis L5 ESPs stimulated autophagosome formation and the expression of autophagy molecules, such as LC3B, Beclin, and p62. The data showed that upon ESPs treatment, rapamycin elevated cell viability through the activation of the autophagy mechanism in astrocytes. Finally, we found that ESPs induced the activation of the Sonic hedgehog (Shh) signaling pathway and that the expression of autophagy molecules was increased through the Shh signaling pathway. Collectively, these results suggest that A. cantonensis L5 ESPs stimulate autophagy through the Shh signaling pathway and that autophagy has a protective effect in astrocytes. In helminthes, Excretory-secretory products (ESPs) contains a wide range of molecules, including proteins, lipids, glycans, and nucleic acids, that assist in the penetration of host defensive barriers, reduction of oxidative stress, and avoid the host immune attack. It has been known as a key factor for parasite development, including feeding, invasion and molting. Therefore, ESPs is a valuable target for the investigation of the host-parasite relationships. However, only a few researches about the function of Angiostrongyliasis cantonensis ESPs have been verified to date. Angiostrongyliasis cantonensis, a blood-feeding nematode, and it is an important causative agent of eosinophilic meningitis and meningoencephalitis in human. Recent our studies have demonstrated that the A. cantonensis ESPs can induce oxidative stress, apoptosis, and immune response. In this study, we will use a mouse astrocytes as a model to investigate the signaling mechanisms of autophagy induction by ESPs treatment. First, the Microarray, Western blotting, and Transmission electron microscopy data demonstrated that A. cantonensis ESPs can induce autophagy generation in astrocytes. Next, ESPs-induced autophagy was activated via Sonic hedgehog (Shh) signaling, and it has a protective potential for astrocytes. These finding will provide new insights into the mechanisms and effects of the A. cantonensis ESPs.
Collapse
Affiliation(s)
- Kuang-Yao Chen
- Department of Parasitology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chien-Ju Cheng
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Chieh Cheng
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kai-Yuan Jhan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Ju Chen
- Department of Parasitology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Lian-Chen Wang
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- * E-mail:
| |
Collapse
|
22
|
Li J, Liu X, Ding J, Tang B, Bai X, Wang Y, Li S, Liu M, Wang X. Effect of Trichinella spp. or derived antigens on chemically induced inflammatory bowel disease (IBD) in mouse models: A systematic review and meta-analysis. Int Immunopharmacol 2020; 85:106646. [PMID: 32485355 DOI: 10.1016/j.intimp.2020.106646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/14/2020] [Accepted: 05/25/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Trichinella or derived antigens have been suggested to be potential therapeutic agents for inflammatory bowel disease (IBD). We aimed to conduct a systematic review and meta-analysis of the available literature to estimate the effect of Trichinella or derived antigens on chemically induced IBD. METHODS Studies were identified by searching the Cochrane Central Register of Controlled Trials, PubMed, Scopus, Web of Science, and Science Direct from inception to February 2020. We included articles written in English that investigated the effect of Trichinella infection and/or derived products in mouse models of IBD. Studies were pooled, and the combined standard mean difference (SMD) and 95% confidence interval (CI) were calculated using a random-effect or fixed-effect model. RESULTS Thirteen studies were eventually included in the meta-analysis. The results indicated significant differences in the disease activity index (DAI), myeloperoxidase (MPO) activity, macroscopic inflammation score, and microscopic inflammation score between the experimental group and the control group. The anti-inflammatory cytokines interleukin (IL)-4, transforming growth factor-beta (TGF-β), IL-10 and IL-13 were significantly increased in the experimental group compared with the control group, whereas the levels of the proinflammatory cytokines interferon (IFN)-γ, IL-6, TNF-α, and IL-17 were significantly decreased. The percentage of regulatory T (Treg) cells was also significantly increased, while the level of the M1 phenotypic macrophage marker iNOS was significantly decreased and the expression of the M2 phenotypic macrophage marker Arg-1 was significantly increased. CONCLUSION Trichinella infection or derived antigens is effective for the alleviation of IBD in mouse models.
Collapse
Affiliation(s)
- Jian Li
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Jing Ding
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Bin Tang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Xue Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Yang Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Shicun Li
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China
| | - Xuelin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, OIE Collaborating Center on Foodborne Parasites in the Asian-Pacific Region, Changchun 130062, China.
| |
Collapse
|
23
|
Qin Y, Ren Y, Yi C, Chileshe N, Chen Y, Zheng L, Dai X, Dong Y, Cui Y. Effect of Wortmannilactone F on Trichinella spiralis Enteral in Mice. Vector Borne Zoonotic Dis 2020; 20:205-211. [DOI: 10.1089/vbz.2019.2482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Yuanhua Qin
- Department of Parasitology, Dalian Medical University, Dalian, China
| | - Yixin Ren
- Department of Parasitology, Dalian Medical University, Dalian, China
| | - Cen Yi
- Clinical Laboratory, Wuhan No.1 Hospital, Wuhan, China
| | - Nawila Chileshe
- 2017 International Education College, Dalian Medical University, Dalian, China
| | - Yufeng Chen
- Department of Microorganism Examination, Dalian Center for Disease Control and Prevention, Dalian, China
| | - Lili Zheng
- Department of Parasitology, Dalian Medical University, Dalian, China
| | - Xiaodong Dai
- Department of Parasitology, Dalian Medical University, Dalian, China
| | - Yuesheng Dong
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Yu Cui
- Department of Parasitology, Dalian Medical University, Dalian, China
| |
Collapse
|
24
|
Wang N, Bai X, Tang B, Yang Y, Wang X, Zhu H, Luo X, Yan H, Jia H, Liu M, Liu X. Primary characterization of the immune response in pigs infected with Trichinella spiralis. Vet Res 2020; 51:17. [PMID: 32085808 PMCID: PMC7035712 DOI: 10.1186/s13567-020-0741-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022] Open
Abstract
Trichinellosis, which is caused by Trichinella spiralis (T. spiralis), is a serious zoonosis. Pigs play an important role in the transmission of human trichinellosis. Characterizing the immune response to T. spiralis infection is key to elucidating host–parasite interactions. However, most studies on the immune response to T. spiralis infection have employed murine models. In this study, we investigated the immune response to T. spiralis infection in pigs. The results showed that the average numbers of larvae per gram (lpg) for the 100-muscle larvae (ML), 1000-ML, and 10 000-ML groups were 1.502, 35.947, and 398.811, respectively. The percentages of CD3+ T cells, B cells, CD4+ T cells, Treg cells, and Th17 cells were elevated in the infection groups compared to the control animals. In contrast, CD8+ T cell percentages were reduced after infection in the low-dose group. The number of neutrophils was increased at 3–17 days post-infection (dpi). Th1 cytokine IL-2 levels were significantly decreased at 7 dpi, and Th2 cytokine IL-4 levels were significantly elevated at 3 dpi. Treg cytokine IL-10 levels were significantly elevated between 7 dpi and 30 dpi. Th17 cytokine IL-17A levels were significantly increased beginning at 11 dpi. These results confirmed that pigs infected with T. spiralis predominantly induced Th2 and Treg immune responses, which suppress the Th1 immune responses. This study provides novel insights into the immune response of pigs infected with T. spiralis.
Collapse
Affiliation(s)
- Nan Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, 130000, China
| | - Xue Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, 130000, China
| | - Bin Tang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, 130000, China
| | - Yong Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, 130000, China
| | - Xuelin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, 130000, China
| | - Hongfei Zhu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Xuenong Luo
- 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, 730046, China
| | - Hongbin Yan
- 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, 730046, China
| | - Hong Jia
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, 130000, China.
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Changchun, 130000, China.
| |
Collapse
|
25
|
Lu Y, Xu HM, Han Y, Zhang YL. Analgesic effect of resveratrol on colitis-induced visceral pain via inhibition of TRAF6/NF-κB signaling pathway in the spinal cord. Brain Res 2019; 1724:146464. [DOI: 10.1016/j.brainres.2019.146464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 08/08/2019] [Accepted: 09/15/2019] [Indexed: 12/15/2022]
|
26
|
Osada Y, Morita K, Tahara S, Ishihara T, Wu Z, Nagano I, Maekawa Y, Nakae S, Sudo K, Kanazawa T. Th2 signals are not essential for the anti-arthritic effects of Trichinella spiralis in mice. Parasite Immunol 2019; 42:e12677. [PMID: 31605645 DOI: 10.1111/pim.12677] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/01/2019] [Accepted: 10/09/2019] [Indexed: 12/31/2022]
Abstract
AIMS Many parasitic helminths are known to alter host immune responses and consequently affect the progression of autoimmune and allergic diseases. The parasitic nematode Trichinella sp has been reported to suppress several experimental diseases in rodents, including experimental autoimmune encephalomyelitis, type 1 diabetes, colitis, airway inflammation and autoimmune arthritis. We tried to clarify requirement of Th2 cytokines in the anti-arthritic effects of Trichinella spiralis (Ts) against collagen-induced arthritis (CIA). METHODS AND RESULTS We infected Ts and then induced CIA in STAT6KO DBA/1 mice, comparing the disease progression with that in wild-type (WT) DBA/1 mice, Ts significantly mitigated arthritis in WT mice, in addition to the impairment of anti-type II collagen (IIC) IgG production in a subclass-independent manner. The genetic absence of STAT6 in the mice did not abrogate the anti-arthritic effects of Ts. Alteration of splenic cytokines was not related to the anti-arthritic effects of the parasite. Moreover, lack of IL-10 did not abrogate the anti-arthritic effects of Ts. CONCLUSION Our results suggest that the anti-arthritic effects of Ts do not require host Th2 signals.
Collapse
Affiliation(s)
- Yoshio Osada
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kentaro Morita
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Sayaka Tahara
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tsubasa Ishihara
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Zhiliang Wu
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Isao Nagano
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yoichi Maekawa
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan.,Domain of Integrated Life Systems, Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu, Japan
| | - Susumu Nakae
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Katsuko Sudo
- Animal Research Center, Tokyo Medical University, Tokyo, Japan
| | - Tamotsu Kanazawa
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|