1
|
Jing Y, Bai Y, Liang C, Liu Y, Zhou J, Guo J, Cai X, Hu X, Fang Y, Ding X, Wu J, Hu D. Ingenol ameliorates silicosis via targeting the PTGS2/PI3K/AKT signaling axis: Implications for therapeutic intervention. Cell Signal 2025; 131:111780. [PMID: 40158708 DOI: 10.1016/j.cellsig.2025.111780] [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: 12/31/2024] [Revised: 03/11/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Silicosis, formerly known as silico, is an irreversible disease caused by prolonged inhalation of substantial amounts of free crystalline silica dust, characterized by pulmonary inflammation and extensive nodular fibrosis. The etiology of the disease remains unclear, which currently hinders the development of effective therapeutic drugs and interventions. Ingenol (Ing), a terpenoid active ingredient found in plants of the Euphorbiaceae family, including the entire herb of Euphorbia helioscopia, Euphorbia kansui, or Euphorbia lathyris, demonstrates significant anti-inflammatory and antiviral activities. In this study, we identified and confirmed that Ingenol can significantly ameliorate silicosis induced by silica dioxide by inhibiting the PTGS2/PI3K/AKT signaling pathway. In vivo, Ingenol improves pulmonary respiratory function and reduces inflammation and fibrosis in a murine model of CS-induced silicosis. In vitro, Ingenol inhibits the expression of cellular factors associated with inflammation and fibrosis, as well as macrophage apoptosis and fibroblast migration. Furthermore, it can modulate the expression of fibrosis-related proteins, thereby inhibiting CS-induced fibrotic responses. Mechanistically, a combination of bioinformatics, network pharmacology, and experimental validation indicates that Ingenol mitigates the progression of silicosis by modulating the PTGS2/PI3K/AKT signaling pathway. In summary, these findings suggest that Ingenol is a potential candidate for the treatment of silicosis.
Collapse
Affiliation(s)
- Yifan Jing
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Ying Bai
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Huainan Xinhua Medical Group Xinhua Hospital, China.
| | - Chao Liang
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Yafeng Liu
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jiawei Zhou
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jianqiang Guo
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Xiaolong Cai
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Xiaofei Hu
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Yujing Fang
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Xuansheng Ding
- Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jing Wu
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China; The First Affiliated Hospital of Anhui University of Science and Technology (Huainan First People's Hospital), School of Medicine, Huainan City, China.
| | - Dong Hu
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China; The First Affiliated Hospital of Anhui University of Science and Technology (Huainan First People's Hospital), School of Medicine, Huainan City, China; Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei City, China.
| |
Collapse
|
2
|
Zhu H, Zhang Q, Wang D, Zheng G, Wang S, Han Y, Xu Y, He H. Differential expression of Toll-like receptors and associated cytokines in the bursa of Eimeria tenella infected chickens. Res Vet Sci 2025; 188:105607. [PMID: 40086401 DOI: 10.1016/j.rvsc.2025.105607] [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: 08/18/2024] [Revised: 02/16/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Chicken coccidiosis, caused by Eimeria spp., is an economically important disease of commercial poultry. Innate immunity ensures an immediate response to invading parasites, and Toll-like receptors (TLRs) are major components of the innate immune system. However, few systematic studies have been reported on the roles of TLRs in chickens infected with Eimeria. In the present study, 14-day-old chickens were infected orally with 50,000 E. tenella oocysts and the bursa of Fabricius was dissected at different time points. Expression profiles of 10 chicken TLRs (chTLRs) and associated cytokines were determined by quantitative real-time PCR. The results showed that chTLR1a and chTLR2a peaked significantly at 3 h post-infection (p < 0.05), while other chTLRs displayed different expression profiles; chTLR1b, chTLR2b, chTLR5, and chTLR15 peaked at 48 h post-infection, while chTLR4, chTLR7, and chTLR21 peaked at 144 h post-infection. ChTLR3 expression was the highest among chTLRs, peaking at 96 h post-infection (p < 0.05). For cytokines, interleukin (IL)-6, IL-12, IL-17, and interferon-γ peaked at 12 h post-infection, while IL-4 peaked at 24 h post-infection. The results provide a valuable overview of the expression profiles of innate immune molecules during E. tenella infection in chicken bursa, and indicate that innate immune responses may mediate resistance to chicken coccidiosis.
Collapse
Affiliation(s)
- Huili Zhu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan 450000, PR China.
| | - Qian Zhang
- Yebio Bioengineering Co., Ltd of Qingdao, Qingdao, Shandong 266108, PR China
| | - Danni Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan 450000, PR China
| | - Guijie Zheng
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan 450000, PR China
| | - Song Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan 450000, PR China
| | - Yanhui Han
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan 450000, PR China
| | - Yanzhao Xu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan 450000, PR China
| | - Hongxuan He
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, PR China; National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| |
Collapse
|
3
|
Day AW, Perez-Lozada J, DiLeo A, Blandino K, Maguire J, Kumamoto CA. Candida albicans Colonization Modulates Murine Ethanol Consumption and Behavioral Responses Through Elevation of Serum Prostaglandin E 2 and Impact on the Striatal Dopamine System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.640044. [PMID: 40060518 PMCID: PMC11888247 DOI: 10.1101/2025.02.25.640044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Candida albicans is a commensal yeast that is a common component of the gastrointestinal (GI) microbiome of humans. C. albicans has been shown to bloom in the GI tract of individuals with alcohol use disorder (AUD) and can promote and increase the severity of alcoholic liver disease (ALD). However, the effects of C. albicans blooms on the host in the context of AUD or AUD-related phenotypes, such as ethanol preference, have been unstudied. In this work, we report a reduction in ethanol consumption and preference in mice colonized with C. albicans. C. albicans-colonized mice exhibited elevated levels of serum PGE2 and reduced ethanol preference was reversed by injection with antagonists of PGE2 receptors. Further, injection of mice with a PGE2 derivative decreased their ethanol preference. These results show that PGE2 acting on its receptors EP1 and EP2 drives reduced ethanol preference in C. albicans-colonized mice. We also showed altered transcription of dopamine receptors in the dorsal striatum of C. albicans-colonized mice and more rapid acquisition of ethanol conditioned taste aversion, suggesting alterations to reinforcement or aversion learning. Finally, C. albicans-colonized mice were more susceptible to ethanol-induced motor coordination impairment showing significant alterations to the behavioral effects of ethanol. This study identifies a member of the fungal microbiome that alters ethanol preference and demonstrates a role for PGE2 signaling in these phenotypes.
Collapse
Affiliation(s)
- Andrew W. Day
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
| | - Jeyra Perez-Lozada
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
- current address: San Juan Bautista School of Medicine, Caguas, Puerto Rico, 00727, USA
| | - Alyssa DiLeo
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University, Boston, Massachusetts, 02111, USA
| | - Katrina Blandino
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University, Boston, Massachusetts, 02111, USA
| | - Jamie Maguire
- Department of Neuroscience, Tufts University, Boston, Massachusetts, 02111, USA
| | - Carol A. Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
| |
Collapse
|
4
|
Chen S, Zhou C, Huang J, Qiao Y, Wang N, Huang Y, Li B, Xu W, He X, Wang K, Zhi Y, Lv G, Shen S. Bioinformatics based exploration of the anti-NAFLD mechanism of Wang's empirical formula via TLR4/NF-κB/COX2 pathway. Mol Med 2024; 30:278. [PMID: 39730994 DOI: 10.1186/s10020-024-01022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 12/01/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has developed as a leading public wellness challenge as a result of changes in dietary patterns. Unfortunately, there is still a lack of effective pharmacotherapy methods for NAFLD. Wang's empirical formula (WSF) has demonstrated considerable clinical efficacy in treating metabolic disorders for years. Nevertheless, the protective effect of WSF against NAFLD and its underlying mechanism remains poorly understood. METHODS The NAFLD model was established using a 17-week high-sucrose and high-fat (HSHF) diet with 32 ICR mice. In assessing the therapeutic efficacy of WSF on NAFLD, we detected changes in body weight, viscera weight, biomarkers of glycolipid metabolism in serum and liver, transaminase levels and histopathology of liver with H&E and Oil Red O staining after oral administration. The chemical components in WSF were extensively identified and gathered utilizing the HPLC-Q-TOF/MS system, database mining from HMDB, MassBank, and TCMSP databases, alongside literature searches from CNKI, Wanfang and VIP databases. The forecast of network pharmacology approach was then utilized to investigate the probable mechanisms by which WSF improves NAFLD based on the performance of prospective target identification and pathway enrichment analysis. Besides, molecular docking was also conducted for the verification of combination activities between active components of WSF and core proteins related to NAFLD. In final, validation experiments of obtained pathways were conducted through ELISA, immunohistochemistry (IHC), and western blot (WB) analysis. RESULTS Pharmacodynamic outcomes indicated that WSF intervention effectively mitigated obesity, fat accumulation in organs, lipid metabolism disorders, abnormal transaminase levels and liver pathology injury in NAFLD mice (P < 0.05, 0.01). A total of 72 existent ingredients of WSF were acquired by HPLC-Q-TOF/MS and database, and 254 common targets (11.6% in total targets) of NAFLD and WSF were identified. Network pharmacology revealed that WSF presses NAFLD via modulating TNF, IL6, AKT1, IL1B, PTGS2 (COX2), and other targets, and the probable pathways were primarily inflammatory signaling pathways, as confirmed by molecular docking. Molecular biology experiments further conformed that WSF could decrease levels of inflammatory factors like IL-1β, IL-6 and TNF-α (P < 0.01) and expression of TLR4, NF-κB and COX-2 (P < 0.05, 0.01) in the liver. CONCLUSION WSF treatment effectively protects against lipid metabolism disorders and liver inflammation injury in HSHF diet-induced NAFLD mice, and its molecular mechanism might be via suppressing the TLR4/NF-κB/COX-2 inflammatory pathway to reduce the release of inflammatory cytokines in the liver.
Collapse
Affiliation(s)
- Suhong Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang, 313200, China
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, No. 548, Binwen Road, Biniang District, Hangzhou, 310053, Zhejiang, China
| | - Chuanjie Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang, 313200, China
| | - Jiahui Huang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang, 313200, China
| | - Yunlong Qiao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang, 313200, China
| | - Ning Wang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang, 313200, China
| | - Yuzhen Huang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang, 313200, China
| | - Bo Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang, 313200, China
| | - Wanfeng Xu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang, 313200, China
| | - Xinglishang He
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Gongshu District, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products. Huzhou, Zhejiang, 313200, China
| | - Kungen Wang
- Disease Prevention and Health Management Center, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang, China.
- Kun-Gen Wang National Famous Chinese Medicine Doctor Studio, Hangzhou, 310006, Zhejiang, China.
| | - Yihui Zhi
- Disease Prevention and Health Management Center, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang, China.
- Kun-Gen Wang National Famous Chinese Medicine Doctor Studio, Hangzhou, 310006, Zhejiang, China.
| | - Guiyuan Lv
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, No. 548, Binwen Road, Biniang District, Hangzhou, 310053, Zhejiang, China.
| | - Shuhua Shen
- Disease Prevention and Health Management Center, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang, China.
- Kun-Gen Wang National Famous Chinese Medicine Doctor Studio, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
5
|
Qiang E, Xu H. PGE 2 synthesis and signaling in the liver physiology and pathophysiology: An update. Prostaglandins Other Lipid Mediat 2024; 174:106875. [PMID: 39019102 DOI: 10.1016/j.prostaglandins.2024.106875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
The liver plays a central role in systemic metabolism and drug degradation. However, it is highly susceptible to damage due to various factors, including metabolic imbalances, excessive alcohol consumption, viral infections, and drug influences. These factors often result in conditions such as fatty liver, hepatitis, and acute or chronic liver injury. Failure to address these injuries could promptly lead to the development of liver cirrhosis and potentially hepatocellular carcinoma (HCC). Prostaglandin E2 (PGE2) is a metabolite of arachidonic acid that belongs to the class of polyunsaturated fatty acids (PUFA) and is synthesized via the cyclooxygenase (COX) pathway. By binding to its G protein coupled receptors (i.e., EP1, EP2, EP3 and EP4), PGE2 has a wide range of physiological and pathophysiology effects, including pain, inflammation, fever, cardiovascular homeostasis, etc. Recently, emerging studies showed that PGE2 plays an indispensable role in liver health and disease. This review focus on the research progress of the role of PGE2 synthase and its receptors in liver physiological and pathophysiological processes and discuss the possibility of developing liver protective drugs targeting the COXs/PGESs/PGE2/EPs axis.
Collapse
Affiliation(s)
- Erjiao Qiang
- Department of Pathology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Hu Xu
- Health Science Center, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
6
|
Khaksari M, Pourali M, Rezaei Talabon S, Gholizadeh Navashenaq J, Bashiri H, Amiresmaili S. Protective effects of 17-β-estradiol on liver injury: The role of TLR4 signaling pathway and inflammatory response. Cytokine 2024; 181:156686. [PMID: 38991382 DOI: 10.1016/j.cyto.2024.156686] [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: 04/20/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
Liver injury, a major global health issue, stems from various causes such as alcohol consumption, nonalcoholic steatohepatitis, obesity, diabetes, metabolic syndrome, hepatitis, and certain medications. The liver's unique susceptibility to ischemia and hypoxia, coupled with the critical role of the gut-liver axis in inflammation, underscores the need for effective therapeutic interventions. The study highlights E2's interaction with estrogen receptors (ERs) and its modulation of the Toll-like receptor 4 (TLR4) signaling pathway as key mechanisms in mitigating liver injury. Activation of TLR4 leads to the release of pro-inflammatory cytokines and chemokines, exacerbating liver inflammation and injury. E2 down-regulates TLR4 expression, reduces oxidative stress, and inhibits pro-inflammatory cytokines, thereby protecting the liver. Both classic (ERα and ERβ) and non-classic [G protein-coupled estrogen receptor (GPER)] receptors are influenced by E2. ERα is particularly crucial for liver regeneration, preventing liver failure by promoting hepatocyte proliferation. Furthermore, E2 exerts anti-inflammatory, antioxidant, and anti-apoptotic effects by inhibiting cytokines such as IL-6, IL-1β, TNF-α, and IL-17, and by reducing lipid peroxidation and free radical damage. The article calls for further clinical research to validate these findings and to develop estrogen-based treatments for liver injuries. Overall, the research emphasizes the significant potential of E2 as a therapeutic agent for liver injuries. It advocates for extensive clinical studies to validate E2 hepatoprotective properties and develop effective estrogen-based treatments.
Collapse
Affiliation(s)
- Mohammad Khaksari
- Neuroscince and Endocrinology and Metabolism Research Centers, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | | | | | | - Hamideh Bashiri
- Neuroscience Research Center, Institute of Neuropharmacology, Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Ira
| | | |
Collapse
|
7
|
Li Q, Wang J, Lv J, Liu D, Xiao S, Mo J, Lu Z, Qiu R, Li C, Tang L, He S, Tang Z, Cheng Q, Zhan T. Total flavonoids of litchi Seed alleviates schistosomiasis liver fibrosis in mice by suppressing hepatic stellate cells activation and modulating the gut microbiomes. Biomed Pharmacother 2024; 178:117240. [PMID: 39094546 DOI: 10.1016/j.biopha.2024.117240] [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: 05/01/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
Infection with Schistosoma japonicum (S. japonicum) is an important zoonotic parasitic disease that causes liver fibrosis in both human and domestic animals. The activation of hepatic stellate cells (HSCs) is a crucial phase in the development of liver fibrosis, and inhibiting their activation can alleviate this progression. Total flavonoids of litchi seed (TFL) is a naturally extracted drug, and modern pharmacological studies have shown its anti-fibrotic and liver-protective effects. However, the role of TFL in schistosomiasis liver fibrosis is still unclear. This study investigated the therapeutic effects of TFL on liver fibrosis in S. japonicum infected mice and explored its potential mechanisms. Animal study results showed that TFL significantly reduced the levels of Interleukin-1β (IL-1β), Tumor Necrosis Factor-α (TNF-α), Interleukin-4 (IL-4), and Interleukin-6 (IL-6) in the serum of S. japonicum infected mice. TFL reduced the spleen index of mice and markedly improved the pathological changes in liver tissues induced by S. japonicum infection, decreasing the expression of alpha-smooth muscle actin (α-SMA), Collagen I and Collagen III protein in liver tissues. In vitro studies indicated that TFL also inhibited the activation of HCSs induced by Transforming Growth Factor-β1 (TGF-β1) and reduced the levels of α-SMA. Gut microbes metagenomics study revealed that the composition, abundance, and functions of the mice gut microbiomes changed significantly after S. japonicum infection, and TLF treatment reversed these changes. Therefore, our study indicated that TFL alleviated granulomatous lesions and improved S. japonicum induced liver fibrosis in mice by inhibiting the activation of HSCs and by improving the gut microbiomes.
Collapse
Affiliation(s)
- Qing Li
- Department of Cell Biology and Genetics, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Jilong Wang
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiahui Lv
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Dengyu Liu
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Suyu Xiao
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Jingquan Mo
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Zuochao Lu
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Ran Qiu
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Caiqi Li
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Lili Tang
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Shanshan He
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Zeli Tang
- Department of Cell Biology and Genetics, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| | - Qiuchen Cheng
- Department of Gastroenterology, the People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi, China.
| | - Tingzheng Zhan
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China; Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
8
|
Liu MK, Chen XY, Tang JJ, Liu ZP, Lin GY, Cai JL, Chen ZM, Yan YY, Ji XF, Yang ZJ, Li Z. Artemisitene shows superiority over artemisinin in preventing Schistosoma japonica-induced liver disease. Parasit Vectors 2024; 17:342. [PMID: 39148111 PMCID: PMC11328484 DOI: 10.1186/s13071-024-06426-y] [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: 05/07/2024] [Accepted: 07/27/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Artemisinin (ART) analogs, such as dihydroartemisinin, arteether, artemether, and artesunate, all featuring an endoperoxide bridge, have demonstrated efficacy against schistosomiasis. Artemisitene (ATT), which contains an additional α, β-unsaturated carbonyl structure, has shown enhanced biological activities. This study aims to evaluate the anti-schistosomaiasis japonica activity of ATT and compare it with ART. METHODS We assessed liver inflammation and fibrosis in mice using hematoxylin and eosin staining and Sirius red staining, respectively. RNA sequencing analyzed transcriptomics in female and male Schistosoma japonicum (S. japonicum) adult worms and mice livers, with cytokine profiling and flow cytometry to study immune responses under ART or ATT treatment. RESULTS ATT exhibits a marked reduction in female S. japonicum adult worms and egg numbers, damaging the adult worms' surface. It also influences the transcription of genes related to cellular anatomical structures. Notably, ATT treatment resulted in significant reductions in liver granuloma size and collagen area, alongside lowering serum levels of glutamic pyruvic and glutamic oxaloacetic transaminase more effectively than ART. Both ART and ATT markedly decreased neutrophil frequency in the liver and elevated eosinophil counts. However, only ATT treatment significantly reduced the M1/M2 and Th1/Th2 indices, indicating a pronounced shift in immune response profiles. ATT-affected host immunity correlated with the extent of liver fibrosis and the count of single males more strongly than ART. CONCLUSION ATT, as a novel preventive strategy for schistosomiasis japonica in mice, significantly outperforms ART.
Collapse
Affiliation(s)
- Meng-Ke Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xu-Yang Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Juan-Juan Tang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhi-Peng Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Gui-Ying Lin
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jun-Ling Cai
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zuo-Ming Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Yun Yan
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Xiao-Fang Ji
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhong-Jin Yang
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin, 300384, China.
| | - Zi Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
- The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
9
|
Dourson AJ, Darken RS, Baranski TJ, Gereau RW, Ross WT, Nahman-Averbuch H. The role of androgens in migraine pathophysiology. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 16:100171. [PMID: 39498299 PMCID: PMC11532460 DOI: 10.1016/j.ynpai.2024.100171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/07/2024]
Abstract
Migraine affects ∼12 % of the worldwide population and is more prevalent in females, which suggests a role of sex hormones in migraine pathophysiology. Most studies have focused on estrogen and progesterone, and the involvement of androgens has been less studied. However, due to the recent advances in androgen interventions, which could advance new androgen-based migraine treatments, it is critical to better understand the role of androgens in migraine. Testosterone, the most studied androgen, was found to have an antinociceptive effect in various animal and human pain studies. Thus, it could also have a protective effect related to lower migraine severity and prevalence. In this review, we discuss studies examining the role of androgens on migraine-related symptoms in migraine animal models. Additionally, we summarize the results of human studies comparing androgen levels between patients with migraine and healthy controls, studies assessing the relationships between androgen levels and migraine severity, and intervention studies examining the impact of testosterone treatment on migraine severity. Many of the studies have limitations, however, the results suggest that androgens may have a minor effect on migraine. Still, it is possible that androgens are involved in migraine pathophysiology in a sub-group of patients such as in adolescents or postmenopausal women. We discuss potential mechanisms in which testosterone, as the main androgen tested, can impact migraine. These mechanisms range from the cellular level to systems and behavior and include the effect of testosterone on sensory neurons, the immune and vascular systems, the stress response, brain function, and mood. Lastly, we suggest future directions to advance this line of research.
Collapse
Affiliation(s)
- Adam J. Dourson
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel S. Darken
- Department of Neurology, Washington University School of Medicine, St. Louis Missouri, USA
| | - Thomas J. Baranski
- Division of Endocrinology, Diabetes and Metabolism Washington University School of Medicine in St. Louis Missouri, USA
| | - Robert W. Gereau
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Whitney Trotter Ross
- Division of Minimally Invasive Gynecologic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Hadas Nahman-Averbuch
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Huang P, Ma H, Cao Y, Zhan T, Zhang T, Wang X, Zhang Y, Xu J, Xia C. Activation of primary hepatic stellate cells and liver fibrosis induced by targeting TGF-β1/Smad signaling in schistosomiasis in mice. Parasit Vectors 2022; 15:456. [PMID: 36474240 PMCID: PMC9727849 DOI: 10.1186/s13071-022-05584-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In mice, liver fibrosis is the most serious pathologic change during Schistosoma japonicum (S. japonicum) infection. Schistosomiasis is mainly characterized by schistosome egg-induced granulomatous fibrosis. Hepatic stellate cells (HSCs) are mainly responsible for the net accumulation of collagens and fibrosis formation in the liver. Activated HSCs regulated by transforming growth factor-β1 (TGF-β1)/Smad signaling have emerged as the critical regulatory pathway in hepatitis virus or carbon tetrachloride-induced liver fibrosis. However, the detailed mechanism of HSC activation in schistosome-induced liver fibrosis is poorly understood. METHODS Schistosoma japonicum-induced murine models and a control group were generated by abdominal infection with 15 (± 1) cercariae. The purity of cultured primary HSCs was evaluated by immunocytochemistry. The histopathological changes in the livers of infected mice were estimated by hematoxylin-eosin and Masson staining. Dynamic expression of pro-fibrotic molecules and microRNAs was detected by real-time quantitative PCR (RT-qPCR). Mainly members involved in the TGF-β1/Smad signaling pathway were examined via RT-qPCR and Western blot. RESULTS The egg-induced granulomatous inflammation formed at 4 weeks post-infection (wpi) and developed progressively. Alpha-smooth muscle actin (α-SMA), collagen I, collagen III, TGF-β1, Smad2, Smad3, and Smad4 showed a significant increase in mitochondrial RNA (mRNA) and protein expression compared with the control group at 7 and 9 weeks post-infection (wpi), while an opposite effect on Smad7 was observed. In addition, the mRNA expression of microRNA-21 (miRNA-21) was significantly increased at 7 wpi, and the mRNA expression of miRNA-454 was decreased starting from 4 wpi. CONCLUSION Our present findings revealed that HSCs regulated by the TGF-β1/Smad signaling pathway play an important role in liver fibrosis in S. japonicum-infected mice, which may provide proof of concept for liver fibrosis in schistosomiasis.
Collapse
Affiliation(s)
- Ping Huang
- grid.263761.70000 0001 0198 0694Department of Pathogen Biology, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China
| | - Huihui Ma
- grid.263761.70000 0001 0198 0694Department of Pathogen Biology, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China
| | - Yun Cao
- grid.263761.70000 0001 0198 0694Department of Pathogen Biology, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China ,grid.252957.e0000 0001 1484 5512Department of Laboratory Medicine, Bengbu Medical College, 2600 Donghai Road, Bengbu, 23303 Anhui China
| | - Tingzheng Zhan
- grid.263761.70000 0001 0198 0694Department of Pathogen Biology, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China ,grid.256607.00000 0004 1798 2653Department of Pathogen Biology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021 Guangxi China
| | - Tingting Zhang
- grid.263761.70000 0001 0198 0694Department of Pathogen Biology, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China
| | - Xinyi Wang
- grid.263761.70000 0001 0198 0694Department of Pathogen Biology, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China
| | - Yanan Zhang
- grid.263761.70000 0001 0198 0694Department of Pathogen Biology, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China
| | - Jing Xu
- grid.263761.70000 0001 0198 0694Department of Pathogen Biology, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China
| | - Chaoming Xia
- grid.263761.70000 0001 0198 0694Department of Pathogen Biology, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 Jiangsu China
| |
Collapse
|
11
|
He X, Shi Y, Zeng Z, Tang B, Xiao X, Yu J, Zou P, Liu J, Xiao Y, Luo Y, Xiao R. Intimate intertwining of the pathogenesis of hypoxia and systemic sclerosis: A transcriptome integration analysis. Front Immunol 2022; 13:929289. [PMID: 36389675 PMCID: PMC9660309 DOI: 10.3389/fimmu.2022.929289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/13/2022] [Indexed: 03/30/2024] Open
Abstract
OBJECTIVES Systemic sclerosis (SSc) is an autoimmune disease caused by various pathogenic factors, including hypoxia. Hypoxia stimulates the production of the extracellular matrix to promote fibrosis. However, the integrated function and the underlying mechanism of hypoxia in SSc are unclear. METHODS In the present study, we used Agilent SurePrint G3 Human Gene Expression v3 for the transcriptional sequencing of fibroblasts with and without hypoxia to detect differentially expressed genes (DEGs) in hypoxia. We analyzed the results with the transcriptome data of SSc lesions (GSE95065) to select the co-DEGs. Then, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed on the basis of the co-DEGs using the R package ClusterProfiler, which showed that hypoxia and cross talk of hypoxia with other pathogenic factors are involved in the pathogenesis of SSc. Furthermore, we constructed a protein-protein interaction (PPI) network of co-DEGs and screened two significant functional expression modules. RESULTS We identified nine hub genes (ALDH1A1, EGF, NOX4, LYN, DNTT, PTGS2, TKT, ACAA2, and ALDH3A1). These genes affect the pentose phosphate pathway, oxidative stress, and lipolysis. CONCLUSION Our study provides insights into the mechanisms underlying the effects of hypoxia on SSc pathogenesis, which will help to better understand SSc pathogenesis and develop new therapeutic strategies for SSc.
Collapse
Affiliation(s)
- Xinglan He
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yaqian Shi
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhuotong Zeng
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bingsi Tang
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xuan Xiao
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiangfan Yu
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Puyu Zou
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiani Liu
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yangfan Xiao
- Department of Anesthesiology, Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yangyang Luo
- Department of Dermatology, Hunan Children's Hospital, Changsha, China
| | - Rong Xiao
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Liu Z, Zhang L, Liang Y, Lu L. Pathology and molecular mechanisms of Schistosoma japonicum-associated liver fibrosis. Front Cell Infect Microbiol 2022; 12:1035765. [PMID: 36389166 PMCID: PMC9650140 DOI: 10.3389/fcimb.2022.1035765] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/13/2022] [Indexed: 11/23/2022] Open
Abstract
Schistosomiasis has been widely disseminated around the world, and poses a significant threat to human health. Schistosoma eggs and soluble egg antigen (SEA) mediated inflammatory responses promote the formation of egg granulomas and liver fibrosis. With continuous liver injuries and inflammatory stimulation, liver fibrosis can develop into liver cirrhosis and liver cancer. Therefore, anti-fibrotic therapy is crucial to increase the survival rate of patients. However, current research on antifibrotic treatments for schistosomiasis requires further exploration. In the complicated microenvironment of schistosome infections, it is important to understand the mechanism and pathology of schistosomiasis-associated liver fibrosis(SSLF). In this review, we discuss the role of SEA in inhibiting liver fibrosis, describe its mechanism, and comprehensively explore the role of host-derived and schistosome-derived microRNAs (miRNAs) in SSLF. Inflammasomes and cytokines are significant factors in promoting SSLF, and we discuss the mechanisms of some critical inflammatory signals and pro-fibrotic cytokines. Natural killer(NK) cells and Natural killer T(NKT) cells can inhibit SSLF but are rarely described, therefore, we highlight their significance. This summarizes and provides insights into the mechanisms of key molecules involved in SSLF development.
Collapse
Affiliation(s)
- Zhilong Liu
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
| | - Lichen Zhang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Yinming Liang, ; Liaoxun Lu,
| | - Liaoxun Lu
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Yinming Liang, ; Liaoxun Lu,
| |
Collapse
|
13
|
Interleukin-33 deficiency prevents biliary injuries and repairments caused by Clonorchis sinensis via restraining type 2 cytokines. Parasit Vectors 2022; 15:386. [PMID: 36271450 PMCID: PMC9587592 DOI: 10.1186/s13071-022-05490-6] [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: 02/15/2022] [Accepted: 09/16/2022] [Indexed: 11/10/2022] Open
Abstract
Background Clonorchiasis caused by Clonorchis sinensis is a zoonotic parasitic disease characterized by cholangitis, biliary proliferation, biliary fibrosis, and even cholangiocarcinoma. Our previous study showed that the expression of interleukin (IL)-33 is increased in both humans and mice infected by C. sinensis, suggesting that IL-33 is potentially involved in the pathogenesis of clonorchiasis. However, the roles and potential mechanism of IL-33 underlying remain unknown. Methods Wild-type (WT) and IL-33 knockout (KO) mice (BALB/c female mice) were orally infected with 45 metacercariae of C. sinensis for 8 weeks. Biliary injuries and fibrosis were extensively evaluated. Hepatic type II cytokines (IL-4, IL-13, and IL-10) were detected by ELISA. Results For wild-type mice, we found that the mice infected with C. sinensis showed severe biliary injuries and fibrosis compared with the normal mice that were free from worm infection. In addition, the levels of type II cytokines such as IL-4, IL-13, and IL-10 in infected wild-type mice were significantly higher than in the control mice without infection (P < 0.05). However, IL-33 deficiency (IL-33 KO) prevents the augmentation of biliary injuries and fibrosis caused by C. sinensis infection. Furthermore, the increased levels of these type II cytokines induced by worm infection were also reversed in IL-33 KO mice. Conclusion Our present study demonstrates that IL-33 contributes to the pathogenesis of C. sinensis-induced biliary injuries and repair, which can potentially orchestrate type 2 responses. These findings highlight the pathophysiological role of IL-33 in the progression of clonorchiasis. Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05490-6.
Collapse
|
14
|
Dibo N, Liu X, Chang Y, Huang S, Wu X. Pattern recognition receptor signaling and innate immune responses to schistosome infection. Front Cell Infect Microbiol 2022; 12:1040270. [PMID: 36339337 PMCID: PMC9633954 DOI: 10.3389/fcimb.2022.1040270] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/03/2022] [Indexed: 08/22/2023] Open
Abstract
Schistosomiasis remains to be a significant public health problem in tropical and subtropical regions. Despite remarkable progress that has been made in the control of the disease over the past decades, its elimination remains a daunting challenge in many countries. This disease is an inflammatory response-driven, and the positive outcome after infection depends on the regulation of immune responses that efficiently clear worms and allow protective immunity to develop. The innate immune responses play a critical role in host defense against schistosome infection and pathogenesis. Initial pro-inflammatory responses are essential for clearing invading parasites by promoting appropriate cell-mediated and humoral immunity. However, elevated and prolonged inflammatory responses against the eggs trapped in the host tissues contribute to disease progression. A better understanding of the molecular mechanisms of innate immune responses is important for developing effective therapies and vaccines. Here, we update the recent advances in the definitive host innate immune response to schistosome infection, especially highlighting the critical roles of pattern recognition receptors and cytokines. The considerations for further research are also provided.
Collapse
Affiliation(s)
- Nouhoum Dibo
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Xianshu Liu
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Yunfeng Chang
- Department of Forensic Medicine Science, Xiangya School of Basic Medicine, Central South University, Yueyang, China
| | - Shuaiqin Huang
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
- Hunan Provincial Key Lab of Immunology and Transmission Control on Schistosomiasis, Hunan Provincial Institute of Schistosomiasis Control, Yueyang, China
| | - Xiang Wu
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
- Hunan Provincial Key Lab of Immunology and Transmission Control on Schistosomiasis, Hunan Provincial Institute of Schistosomiasis Control, Yueyang, China
| |
Collapse
|
15
|
Tao W, Li L, Hu J, Xu S, Wang B, Ding J, Zhang M, Meng X, Wei X, Shan X, Peng K, Liu H, Ji F. Interaction between COX-2 and ER stress is involved in the apoptosis-induced myocardial ischemia/reperfusion injury. Am J Transl Res 2022; 14:3360-3371. [PMID: 35702111 PMCID: PMC9185046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE Apoptosis induced by excessive endoplasmic reticulum (ER) stress is accompanied by the occurrence and progression of myocardial ischemia/reperfusion (I/R) injury. COX-2 is also known to affect the development of I/R damage in myocardium. However, the interaction between COX-2 and ER stress in aggravating myocardial I/R lesion is not well characterized. Therefore, the purpose of our research was to explore the interaction between COX-2 and ER stress on myocardial apoptosis. METHODS The left anterior descending (LAD) coronary artery was ligatured with a 6-0# suture for 0.5 hours and subsequently subjected to reperfusion for 3 hours to simulate myocardial I/R in mice. Oxygen glucose deprivation/reoxygenation (OGD/R) was performed on H9c2 cells to construct an in vitro model of this experiment. NS398 (COX-2 specific inhibitor) and Salubrinal (Sal, ER stress inhibitor) were administered to assess the function of COX-2 and ER stress in myocardial I/R impairment. CCK-8 assay was used to evaluate the viability of H9c2 cells under different treatment conditions. TUNEL and Hoechst staining were used to detect the occurrence of apoptosis. Infarct area/area at risk and Hematoxylin-eosin stained sections were assessed after I/R. Protein expressions of glucose-regulated protein 78 (GRP78), COX-2, phosphorylation of eukaryotic translation initiation factor 2 alpha (p-eIF2α), CCAAT/enhancer-binding protein homologous protein (CHOP), and Cleaved caspase 3 in the myocardium were examined using Western blotting. Changes in Cleaved caspase 3 expression in myocardial slices were measured by immunohistochemistry. RESULTS Sal or NS398 partly reduced I/R-induced damage as testified by the apparent decrease in infarct size after I/R and reduced cell viability following OGD/R. Sal distinctly increased p-eIF2α, but caused decreased expression of COX-2, Cleaved caspase 3, and ER stress-associated proteins after I/R, suggesting that Sal effectively inhibited ER stress, apoptosis, and COX-2. Pretreatment with NS398 blocked I/R or OGD/R-induced upregulation of COX-2, Cleaved caspase 3, and ER stress-related marker proteins. CONCLUSIONS Interaction of COX-2 and ER stress regulates apoptosis and contributes to Myocardial lesion induced by I/R.
Collapse
Affiliation(s)
- Wenhui Tao
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow UniversitySuzhou, Jiangsu, China
| | - Lingui Li
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow UniversitySuzhou, Jiangsu, China
| | - Junkai Hu
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow UniversitySuzhou, Jiangsu, China
| | - Shangxian Xu
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Biying Wang
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Jun Ding
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Mian Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow UniversitySuzhou, Jiangsu, China
| | - Xiaowen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow UniversitySuzhou, Jiangsu, China
| | - Xiang Wei
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Xisheng Shan
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow UniversitySuzhou, Jiangsu, China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow UniversitySuzhou, Jiangsu, China
| | - Huayue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow UniversitySuzhou, Jiangsu, China
| | - Fuhai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
- Institute of Anesthesiology, Soochow UniversitySuzhou, Jiangsu, China
| |
Collapse
|
16
|
Qi Z, Lan C, Xiaofang J, Juanjuan T, Cheng F, Ting H, Erxia S, Zi L. Inhibition of COX-2 ameliorates murine liver schistosomiasis japonica through splenic cellular immunoregulation. Parasit Vectors 2022; 15:144. [PMID: 35461268 PMCID: PMC9034617 DOI: 10.1186/s13071-022-05201-1] [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: 11/19/2021] [Accepted: 02/12/2022] [Indexed: 11/16/2022] Open
Abstract
Background We have reported the positive association of the cyclooxygenase 2 (COX-2)/prostaglandin E2 (PGE2) axis with liver fibrosis induced by Schistosoma japonicum (Sj) infection, and TLR4 signaling controlled this axis. However, how COX-2 regulates immune response during Sj infection is still unclear. Methods Hematoxylin and eosin staining was used to evaluate the effect of the COX-2-specific inhibitor NS398 on liver granulomatous inflammation and fibrosis. Flow cytometry was used to explore the frequency and amount of different immune cell infiltration in the spleen during Sj infection. Results NS398 significantly reduced the size of liver granuloma, spleen, and mesenteric lymph node (MLN) and alleviated chronic granulomatous inflammation. Mechanically, this might be by decreasing the number of Sj-induced macrophages and T helper type 1 (Th1), Th2, T follicular helper (Tfh), T follicular regulatory (Tfr), and germinal center B (GC B) cells. There were no differences in the number of neutrophils, myeloid-derived suppressor cells, Th17 cells, regulatory T cells (Treg), or total B cells in the spleen of the mice with or without NS398 treatment. Conclusions COX-2/PGE2 inhibition may represent a potential therapeutic approach for schistosomiasis japonica through splenic cellular immunoregulation. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Zhang Qi
- Sino‑French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China.,Immunology Department, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China
| | - Chen Lan
- Sino‑French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China
| | - Ji Xiaofang
- Sino‑French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China
| | - Tang Juanjuan
- Sino‑French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China
| | - Fu Cheng
- Sino‑French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China.,Immunology Department, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China
| | - Huang Ting
- Sino‑French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China.,Immunology Department, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China
| | - Shen Erxia
- Sino‑French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China. .,Immunology Department, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China. .,The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, Guangdong Province, China.
| | - Li Zi
- Sino‑French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, China. .,The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
17
|
Lechner A, Bohnacker S, Esser-von Bieren J. Macrophage regulation & function in helminth infection. Semin Immunol 2021; 53:101526. [PMID: 34802871 DOI: 10.1016/j.smim.2021.101526] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/03/2021] [Accepted: 11/06/2021] [Indexed: 12/14/2022]
Abstract
Macrophages are innate immune cells with essential roles in host defense, inflammation, immune regulation and repair. During infection with multicellular helminth parasites, macrophages contribute to pathogen trapping and killing as well as to tissue repair and the resolution of type 2 inflammation. Macrophages produce a broad repertoire of effector molecules, including enzymes, cytokines, chemokines and growth factors that govern anti-helminth immunity and repair of parasite-induced tissue damage. Helminth infection and the associated type 2 immune response induces an alternatively activated macrophage (AAM) phenotype that - beyond driving host defense - prevents aberrant Th2 cell activation and type 2 immunopathology. The immune regulatory potential of macrophages is exploited by helminth parasites that induce the production of anti-inflammatory mediators such as interleukin 10 or prostaglandin E2 to evade host immunity. Here, we summarize current insights into the mechanisms of macrophage-mediated host defense and repair during helminth infection and highlight recent progress on the immune regulatory crosstalk between macrophages and helminth parasites. We also point out important remaining questions such as the translation of findings from murine models to human settings of helminth infection as well as long-term consequences of helminth-induced macrophage reprogramming for subsequent host immunity.
Collapse
Affiliation(s)
- Antonie Lechner
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
| | - Sina Bohnacker
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
| | - Julia Esser-von Bieren
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany.
| |
Collapse
|
18
|
Zhang C, Yan Y, Gao X, Ma Y. [Therapeutic mechanism of the Mongolian medicine Qiwei Qinggan Powder against liver fibrosis based on UHPLC-TOF-MS combined with network pharmacological methods]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1131-1141. [PMID: 34549702 DOI: 10.12122/j.issn.1673-4254.2021.08.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To elucidate the chemical composition of the Mongolian medicine Qiwei Qinggan Powder and explore its key targets, related pathways and its therapeutic mechanism for liver fibrosis. METHODS UHPLC-TOF-MS was used to analyze the composition of Qiwei Qinggan Powder. The therapeutic targets of Qiwei Qinggan Powder were screened in Swiss Target Prediction database, and liver fibrosis-related targets were screened in TTD and GeneCards databases to identify the anti-fibrosis targets of Qiwei Qinggan Powder by intersection using Venny.2.1.0. The protein interaction was analyzed using STRING database, the GO functions and KEGG pathways were analyzed on the Metascape platform, and the core targets and active components were verified by molecular docking using AutoDock software. The therapeutic mechanism of Qiwei Qinggan Powder against liver fibrosis was verified in rat models and cell experiment. RESULTS We identified a total of 45 chemical constituents in Qiwei Qinggan Powder, including flavonoids, alkaloids, coumarins, terpenes, phenols and fatty acids. Network pharmacological analysis identified 62 targets of Qiwei Qinggan Powder, including 10 core targets. GO enrichment analysis suggested that the therapeutic effect of Qiwei Qinggan Powder was mediated by biological processes (BP), cell components (CC) and molecular functions (MF). KEGG enrichment results showed that PI3K/Akt, Rap1, MAPK, AMPK and PPAR were all pathways associated with liver fibrosis. Molecular docking showed that quercetin, luteolin and kaempferol could bind to Akt1, PIK3R1 and MAPK1, respectively. In rat models of liver fibrosis, treatment with Qiwei Qinggan Powder significantly suppressed proliferation of fibrous tissues and inflammatory cell infiltration to improve fibrosis in the liver tissue. Western blotting demonstrated that Qiwei Qinggan Powder significantly decreased the expressions of the Liver fibrosis markers including α-SMA, Collagen1, PI3K and Akt (P < 0.01). In vitro cell experiment, Qiwei Qinggan Powder-containing serum obviously promoted apoptosis of HSC-T6 cells. CONCLUSION The therapeutic effect of Qiwei Qinggan Powder against liver fibrosis is mediated by multiple components, targets and channels, and its mechanism may involve the regulation of PI3K, Akt and other key targets and modulation of cell apoptosis and energy metabolism.
Collapse
Affiliation(s)
- C Zhang
- Department of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010059, China
| | - Y Yan
- Department of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010059, China
| | - X Gao
- Department of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010059, China
| | - Y Ma
- Department of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010059, China
| |
Collapse
|
19
|
Yamaguchi M, Dohi N, Ooka A, Saito SY, Ishikawa T. Caffeine-induced inversion of prostaglandin E 2 effects on hepatic stellate cell activation. Biomed Pharmacother 2021; 142:111989. [PMID: 34388524 DOI: 10.1016/j.biopha.2021.111989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND AIMS Liver inflammation leads to the activation of hepatic stellate cells (HSCs), resulting in the development of liver fibrosis. The present study aimed to investigate the effects of prostaglandin E2 (PGE2), which is biosynthesized by Kupffer cells, hepatocytes, and HSCs during inflammation, on HSC activation, including its combinatory effect with caffeine. METHODS HSCs isolated from mice were activated by culturing in a medium supplemented with 10% fetal bovine serum for 7 days on plastic plates. The activation of HSCs was evaluated by immunofluorescence of α-smooth muscle actin in HSCs. Comprehensive gene expression analysis was performed using mRNA-sequencing to compare HSCs cultured for 1 or 7 days, with or without PGE2, caffeine, or both. RESULTS PGE2 (1 μM) facilitated the activation of HSCs but inhibited the HSC activation in the presence of caffeine (3 mM). Comprehensive gene expression analysis revealed that HSCs treated with PGE2 in the presence of caffeine were classified in the same class as HSCs cultured for 1 day, i.e., quiescent HSCs. In contrast, PGE2 did not exhibit an inhibitory effect on HSC activation when co-treated with any isoform-specific phosphodiesterase inhibitors. Although the adenylate cyclase inhibitor 2',5'-dideoxyadenosine suppressed the elevation of intracellular cAMP level induced by PGE2 in the presence of caffeine, it had no effect on the inhibition of HSC activation by PGE2 plus caffeine. CONCLUSION The effect of PGE2 on HSC activation is changed from facilitatory to inhibitory when combined with caffeine, suggesting that caffeine may effectively suppress liver fibrosis during inflammation.
Collapse
Affiliation(s)
- Momoka Yamaguchi
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan.
| | - Naoki Dohi
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Akira Ooka
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| | - Shin-Ya Saito
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan; Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari City, Ehime 794-8555, Japan
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka City, Shizuoka 422-8526, Japan
| |
Collapse
|
20
|
Biernacki M, Jastrząb A, Skrzydlewska E. Changes in Hepatic Phospholipid Metabolism in Rats under UV Irradiation and Topically Treated with Cannabidiol. Antioxidants (Basel) 2021; 10:1157. [PMID: 34439405 PMCID: PMC8388943 DOI: 10.3390/antiox10081157] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023] Open
Abstract
The liver is a key metabolic organ that is particularly sensitive to environmental factors, including UV radiation. As UV radiation induces oxidative stress and inflammation, natural compounds are under investigation as one method to counteract these consequences. The aim of this study was to assess the effect of topical application of phytocannabinoid-cannabidiol (CBD) on the skin of nude rats chronically irradiated with UVA/UVB, paying particular attention to its impact on the liver antioxidants and phospholipid metabolism. The results of this study indicate that CBD reaches the rat liver where it is then metabolized into decarbonylated cannabidiol, 7-hydroxy-cannabidiol and cannabidiol-glucuronide. CBD increased the levels of GSH and vitamin A after UVB radiation. Moreover, CBD prevents the increase of 4-hydroxynonenal and 8-iso-prostaglandin-F2α levels in UVA-irradiated rats. As a consequence of reductions in phospholipase A2 and cyclooxygenases activity following UV irradiation, CBD upregulates the level of 2-arachidonoylglycerol and downregulates prostaglandin E2 and leukotriene B4. Finally, CBD enhances decreased level of 15-deoxy-Δ-12,14-prostaglandin J2 after UVB radiation and 15-hydroxyeicosatetraenoic acid after UVA radiation. These data show that CBD applied to the skin prevents ROS- and enzyme-dependent phospholipid metabolism in the liver of UV-irradiated rats, suggesting that it may be used as an internal organ protector.
Collapse
Affiliation(s)
| | | | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, A. Mickiewicza 2D, 15-222 Bialystok, Poland; (M.B.); (A.J.)
| |
Collapse
|