1
|
Su Q, Pan H, Hong P, You Y, Wu Y, Zou J, Sun J, Rao G, Liao J, Tang Z, Hu L. Protective effect of curcumin against endoplasmic reticulum stress and lipid metabolism disorders in AFB1-intoxicated duck liver. Mycotoxin Res 2025:10.1007/s12550-025-00586-1. [PMID: 40085329 DOI: 10.1007/s12550-025-00586-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Aflatoxin B1 (AFB1) is a stable and highly toxic toxin that causes multi-organ toxicity with sustained ingestion, most typically in the duck liver. Previous research has shown that AFB1 can bring about endoplasmic reticulum stress (ERS) in animals, and ERS is strongly associated with lipid metabolism. However, the relationship between AFB1-induced duck liver toxicity and ERS and lipid metabolism is currently unclear. Great attention has been paid to the prevention and treatment of AFB1 because of its great harm. Curcumin, a natural polyphenol, is notable for its powerful anti-inflammatory and antioxidant properties. Studies have shown curcumin to be protective against afb1-induced avian multi-organ toxicity. However, the effects of curcumin on the liver of ducks exposed to AFB1 are largely unknown. In the present study, we aimed to investigate whether AFB1 exposure induces ERS and lipid metabolism disorders in duck liver, while exploring the positive role of curcumin in it. One-day-old ducks (n = 80) were randomly divided in four groups: control group, AFB1 group (0.1 mg / kg.bw AFB1), Cur group (400 mg/kg curcumin), and AFB1 + Cur group (0.1 mg/kg.bw AFB1 + 400 mg/kg curcumin), and blood and liver were collected for the study after 21 days of continuous administration. Our research has found that AFB1 exposure significantly increases the levels of liver function indicators ALP, AST, and ALT in ducks' serum (P < 0.05). Duck liver undergoes fatty degeneration under the influence of AFB1. Under the effect of curcumin, AFB1-induced structural damage in duck liver was somewhat controlled. Further experimental results showed that AFB1 treatment significantly increased the expression of glucose-regulated protein 78 (P < 0.001), and activated the endoplasmic reticulum stress pathway. Meanwhile, AFB1 inhibited the LKB1-AMPK signaling pathway and disrupted lipid metabolic homeostasis. And curcumin treatment effectively reversed these changes. Overall, our results suggest that curcumin attenuates AFB1-induced hepatotoxicity in ducks by inhibiting ERS and lipid metabolism disorders.
Collapse
Affiliation(s)
- Qian Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Panjing Hong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yanli You
- College of Life Science, Yantai University, Yantai City, 264005, Shandong Province, China
| | - Yuhan Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Junbo Zou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jingping Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Gan Rao
- Guangzhou General Pharmaceutical Research Institute Co., Ltd, Guangzhou, China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
2
|
Hao Z, Wang Y, Chen L, Zhou Y, Fang D, Yao W, Xiao L, Zhang Y. TRIM21 Promotes Endothelial Cell Activation via Accelerating SOCS3 Ubiquitination Degradation in Atherosclerosis. Cardiovasc Toxicol 2025; 25:395-410. [PMID: 39921798 DOI: 10.1007/s12012-025-09965-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/19/2025] [Indexed: 02/10/2025]
Abstract
Activated endothelial cells play an important role in the beginning of atherosclerotic disease by secreting various proteins and inflammatory cytokines. Ubiquitination is one of the most common post-translational changes in cells. However, the role and mechanisms of ubiquitination in endothelial cell activation remain poorly understood. In this study, we identified TRIM21 as an E3 ubiquitin ligase with increased expression in atherosclerotic disease and activated endothelial cells. Knockdown of TRIM21 resulted in reduced secretion of inflammatory factors and attenuated the pyroptosis of endothelial cells, inhibiting the progression of atherosclerosis. Mechanistically, TRIM21 could bind and ubiquitinate SOCS3, thereby enhancing NLRP3-mediated pyroptosis. Taken together, we found that endothelial TRIM21 activated the JAK/STAT3 pathway by degrading SOCS3, which in turn promoted NLRP3-mediated pyroptosis and aggravated atherosclerosis, revealing that TRIM21 may be a promising treatment target for the medical management of atherosclerosis.
Collapse
Affiliation(s)
- Zhenxuan Hao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yihuan Wang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Linlin Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yanjun Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Dezhou Fang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Wenxiang Yao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Lili Xiao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yanzhou Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
3
|
Fan X, Liu S, Yu J, Hua J, Feng Y, Wang Z, Shen Y, Lan W, Wang J. Puerarin Ameliorates the Ferroptosis in Diabetic Liver Injure Through the JAK2/STAT3 Pathway Inhibition Based on Network Pharmacology and Experimental Validation. Drug Des Devel Ther 2025; 19:737-757. [PMID: 39911447 PMCID: PMC11796443 DOI: 10.2147/dddt.s487496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
Background Diabetic liver injury (DLI) is a common complication of diabetes mellitus (DM), which seriously endangers the health of diabetic patients. Puerarin, the main active component of Pueraria lobata, has shown positive effects in lowering blood glucose and lipids, resisting oxidative stress, and protecting the liver. However, the mechanism of protective effect of Puerarin on DLI remains unclear. Methods Various databases were used to screen for targets of Puerarin, ferroptosis and DLI. Protein-protein interaction (PPI) network and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were used to predict key targets and pathways. Molecular docking was used to predict the interactions between Puerarin and core targets. KK/Upj-Ay/J (KKAy) mice and high glucose (HG)-induced AML12 cells were used to study the protective effect of Puerarin on DLI. The molecular mechanisms by which Puerarin acts were further verified by in vivo and in vitro experiments. Results KEGG analysis indicated that the JAK/STAT pathway might be related to the anti-DLI effect of Puerarin. Molecular docking revealed that Puerarin has good affinity for JAK2 and STAT3. In vivo, Puerarin (80 mg/kg) reduced body weight, blood glucose, blood lipids and liver function in KKAy mice fed a high-sugar, high-fat diet. Puerarin also ameliorated hepatic pathological changes and inflammatory responses, and attenuated oxidative stress and iron overload in KKAy mice. Western blotting results showed that Puerarin could regulate the expression of proteins related to JAK2/STAT3 pathway and ferroptosis pathway. In vitro, Puerarin (25, 50, 100 μM) increased cell viability and decreased steatosis and liver function indexes in AML12 cells induced by HG (30 mm) to varying degrees. More importantly, AG490 blocker experiments showed that the regulation of ferroptosis process by Puerarin was dependent on the JAK2/STAT3 pathway. Conclusion In conclusion, this study revealed Puerarin may regulate the ferroptosis process by inhibiting the JAK2/STAT3 pathway for the treatment of DLI.
Collapse
Affiliation(s)
- Xiaoxu Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Shuangqiao Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Jing Yu
- School of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, People’s Republic of China
| | - Jian Hua
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Yingtong Feng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Zhen Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Yiwei Shen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Wei Lan
- School of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, People’s Republic of China
| | - Jingxia Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| |
Collapse
|
4
|
Yu S, Pei S, Zhang M, Gao S, Chen J, Duan L, Hu E, Wang Y, Huang Y. PKM2-mediated STAT3 phosphorylation promotes acute liver failure via regulating NLRP3-dependent pyroptosis. Commun Biol 2024; 7:1694. [PMID: 39722076 DOI: 10.1038/s42003-024-07227-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 11/07/2024] [Indexed: 12/28/2024] Open
Abstract
Acute liver failure (ALF) is a life-threatening clinical syndrome characterized by high-grade inflammation and multi-organ failure. Our previous study shows that targeting the M2 isoform of pyruvate kinase (PKM2) to inhibit macrophage inflammation may be a promising strategy for ALF treatment. however, the mechanism by which PKM2 regulates the inflammatory response is unclear. Here we demonstrate that PKM2 contributes to ALF by modulating NLRP3-mediated pyroptosis activation in liver macrophages. The specific knockout of PKM2 in myeloid cells reduces mortality and alleviates hepatic injury in D-galactosamine/LPS-induced ALF mice. Single-cell transcriptome analysis suggests that NLRP3 inflammasome activation of macrophages involves in ALF, knockout of PKM2 in macrophages reduces the expression of NLRP3, and activation of pyroptosis. Pharmacological inhibition of the PKM2 nuclear translocation, but not glycolytic activity, protects mice from ALF. Pharmacological and genetic inhibition of PKM2 attenuates NLRP3-mediated pyroptosis activation and consequently reduces the release of IL-1β and IL-18 by macrophages. Mechanistically, PKM2 translocates into the nucleus and combines with STAT3, enhancing its phosphorylation and recruitment to the NLRP3 promoter region, thereby increasing NLRP3 expression. This work defines PKM2 acts as an important nonmetabolic regulator of NLRP3 that modulates pyroptosis activation in macrophages and guides future therapeutic strategies development for ALF.
Collapse
Affiliation(s)
- Songman Yu
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Siya Pei
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, China
| | - Min Zhang
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Shang Gao
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Jun Chen
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Duan
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - En Hu
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Wang
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China.
| | - Yan Huang
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
5
|
Ding J, Cheng X, Zeng C, Zhao Q, Xing C, Zhang C, Cao H, Guo X, Hu G, Zhuang Y. Aflatoxin B1 Promotes Pyroptosis in IPEC-J2 Cells by Disrupting Mitochondrial Dynamics through the AMPK/NLRP3 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:28093-28108. [PMID: 39630575 DOI: 10.1021/acs.jafc.4c05876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Aflatoxin B1 (AFB1) is one of the most toxic mycotoxins in food and feed, seriously jeopardizing the intestinal health, while the effects of AFB1 on intestinal damage remain to be well understood. This study aims to evaluate the effect of AFB1 on intestinal injury by regulating AMP-activated protein kinase (AMPK)-mediated pyroptosis in vitro. The present study showed that AFB1 led to the formation of large number of bubble-like protrusions on the cell membrane, releasing lactate dehydrogenase (LDH) and interleukin-1β (IL-1β). Stimulation with AFB1 resulted in the activation of the NOD-like receptor protein 3 (NLRP3) pathway, as indicated by the increased expression of pyroptosis-associated factor mRNAs and proteins, which ultimately led to a significant upregulation of the pyroptosis rate. Meanwhile, AFB1 caused dysfunction of mitochondrial dynamics by activating the AMPK signaling pathway as mainly evidenced by upregulating dynamin-1-like protein 1 (Drp1) mRNA and protein expression. Moreover, inhibition of NLRP3 and AMPK pathways by MCC950 and compound C, respectively, significantly alleviated AFB1-induced damage in IPEC-J2 cells, evidenced by suppressed NLRP3-mediated pyroptosis, and ameliorated AMPK-mediated mitochondrial dynamics imbalance. In conclusion, these results demonstrated that AFB1 promoted pyroptosis of IPEC-J2 cells by interfering with mitochondrial dynamics by activating the AMPK/NRLP3 pathway.
Collapse
Affiliation(s)
- Jiayi Ding
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| | - Xinyi Cheng
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| | - Chun Zeng
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| | - Qintao Zhao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| | - Chenghong Xing
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| | - Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang, Jiangxi 330045, P. R. China
| |
Collapse
|
6
|
Dai C, Li D, Velkov T, Shen J, Hao Z. The Detoxification Effects of Melatonin on Aflatoxin-Caused Toxic Effects and Underlying Molecular Mechanisms. Antioxidants (Basel) 2024; 13:1528. [PMID: 39765856 PMCID: PMC11726890 DOI: 10.3390/antiox13121528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
Aflatoxins (AFTs) are a form of mycotoxins mainly produced by Aspergillus flavus and Aspergillus parasiticus, which are common contaminants in various agricultural sources such as feed, milk, food, and grain crops. Aflatoxin B1 (AFB1) is the most toxic one among all AFTs. AFB1 undergoes bioactivation into AFB1-8,9-epoxide, then leads to diverse harmful effects such as neurotoxicity, carcinogenicity, hepatotoxicity, reproductive toxicity, nephrotoxicity, and immunotoxicity, with specific molecular mechanisms varying in different pathologies. The detoxification of AFB1 is of great importance for safeguarding the health of animals and humans and has increasingly attracted global attention. Recent research has shown that melatonin supplementation can effectively mitigate AFB1-induced multiple toxic effects. The protection mechanisms of melatonin involve the inhibition of oxidative stress, the upregulation of antioxidant enzyme activity, the reduction of mitochondrial dysfunction, the inactivation of the mitochondrial apoptotic pathway, the blockade of inflammatory responses, and the attenuation of cytochrome P450 enzymes' expression and activities. In summary, this review sheds new light on the potential role of melatonin as a potential detoxifying agent against AFB1. Further exploration of the precise molecular mechanisms and clinical efficacy of this promising treatment is urgently needed.
Collapse
Affiliation(s)
- Chongshan Dai
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
| | - Daowen Li
- College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300392, China
| | - Tony Velkov
- Department of Pharmacology, Biodiscovery Institute, Monash University, Parkville, VIC 3052, Australia
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
| | - Zhihui Hao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, China
| |
Collapse
|
7
|
Wang Q, Wang F, Zhou Y, Li X, Xu S, Tang L, Jin Q, Fu A, Yang R, Li W. Bacillus amyloliquefaciens SC06 Attenuated Lipopolysaccharide-Induced acute liver injury by suppressing bile acid-associated NLRP3 inflammasome activation. Int Immunopharmacol 2024; 142:113129. [PMID: 39293317 DOI: 10.1016/j.intimp.2024.113129] [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: 02/19/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 09/20/2024]
Abstract
The involvement of the inflammatory response has been linked to the development of liver illnesses. As medications with the potential to prevent and cure liver illness, probiotics have garnered an increasing amount of interest in recent years. The present study used a piglet model with acute liver injury (ALI) induced by lipopolysaccharides (LPS) to investigate the regulatory mechanisms of Bacillus amyloliquefaciens SC06. Our findings indicated that SC06 mitigated the liver structural damage caused by LPS, as shown by the decreased infiltration of inflammatory cells and the enhanced structural integrity. In addition, After the administration of SC06, there was a reduction in the increased levels of the liver damage markers. In the LPS group, there was an increase in the mRNA expression of inflammatory cytokines, apoptosis cell rate, and genes associated with apoptosis, while these alterations were mitigated by SC06 administration. Furthermore, SC06 prevented pigs from suffering liver damage by preventing the activation of the NLRP3 inflammasome, which was normally triggered by LPS. The examination of serum metabolic pathways found that ALI was related to several metabolic processes, including primary bile acid biosynthesis, pentose and glucuronate interconversions and the metabolism of phenylalanine. Significantly, our research revealed that the administration of SC06 effectively controlled the concentrations of bile acids in the serum. The correlation results also revealed clear relationships between bile acids and liver characteristics and NLRP3 inflammasome-related genes. However, in vitro experiments revealed that SC06 could not directly inhibit NLRP3 activation under ATP, monosodium urate, and nigericin stimulation, while taurochenodeoxycholic acid (TCDCA) activated NLRP3 inflammasome related genes. In conclusion, our study proved that the hepaprotective effect of SC06 on liver injury, which was closely associated with the restoration of bile acids homeostasis and NLRP3 inflammasome inhibition.
Collapse
Affiliation(s)
- Qi Wang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fei Wang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanhao Zhou
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiang Li
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shujie Xu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Li Tang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qian Jin
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Aikun Fu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Rongchang Yang
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, Western Australia, 6150, Australia; Zhejiang Youheyhey Biotechnology Co., LTD, Huzhou 313000, Zhejiang Province, China.
| | - Weifen Li
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry, National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
8
|
Ma M, Wang Q, Liu Y, Li G, Liu L, Wang G, Guo Y, Huang S, Ma Q, Ji C, Zhao L. Bacillus CotA laccase improved the intestinal health, amino acid metabolism and hepatic metabolic capacity of Pekin ducks fed naturally contaminated AFB 1 diet. J Anim Sci Biotechnol 2024; 15:138. [PMID: 39385285 PMCID: PMC11465776 DOI: 10.1186/s40104-024-01091-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Aflatoxin B1 (AFB1) is a prevalent contaminant in agricultural products, presenting significant risks to animal health. CotA laccase from Bacillus licheniformis has shown significant efficacy in degrading mycotoxins in vitro test. The efficacy of Bacillus CotA laccase in animals, however, remains to be confirmed. A 2 × 2 factorial design was used to investigate the effects of Bacillus CotA laccase level (0 or 1 U/kg), AFB1 challenge (challenged or unchallenged) and their interactions on ducks. The purpose of this study was to evaluate the efficacy of Bacillus CotA laccase in alleviating AFB1 toxicosis of ducks. RESULTS Bacillus CotA laccase alleviated AFB1-induced declines in growth performance of ducks accompanied by improved average daily gain (ADG) and lower feed/gain ratio (F/G). Bacillus CotA laccase ameliorated AFB1-induced gut barrier dysfunctions and inflammation testified by increasing the jejunal villi height/crypt depth ratio (VH/CD) and the mRNA expression of tight junction protein 1 (TJP1) and zonula occluden-1 (ZO-1) as well as decreasing the expression of inflammation-related genes in the jejunum of ducks. Amino acid metabolome showed that Bacillus CotA laccase ameliorated AFB1-induced amino acid metabolism disorders evidenced by increasing the level of glutamic acid in serum and upregulating the expression of amino acid transport related genes in jejunum of ducks. Bacillus CotA laccase ameliorated AFB1-induced liver injury testified by suppressing oxidative stress, inhibiting apoptosis, and downregulating the expression of hepatic metabolic enzyme related genes of ducks. Moreover, Bacillus CotA laccase degraded AFB1 in digestive tract of ducks, resulting in the reduced absorption level of AFB1 across intestinal epithelium testified by the decreased level of AFB1-DNA adduct in the liver, and the reduced content of AFB1 residues in liver and feces of ducks. CONCLUSIONS Bacillus CotA laccase effectively improved the growth performance, intestinal health, amino acid metabolism and hepatic aflatoxin metabolism of ducks fed AFB1 diets, highlighting its potential as an efficient and safe feed enzyme for AFB1 degradation in animal production.
Collapse
Affiliation(s)
- Mingxin Ma
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Qianqian Wang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Yanrong Liu
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Guiming Li
- Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Limeng Liu
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Gaigai Wang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Yongpeng Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Cheng Ji
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Lihong Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, People's Republic of China.
| |
Collapse
|
9
|
Cui Y, Wang Q, Shi Y, Dai Y, Liu Y. mtROS-mediated mitophagy is involved in aflatoxin-B 1 induced liver injury in ducks. Comp Biochem Physiol C Toxicol Pharmacol 2024; 283:109942. [PMID: 38810896 DOI: 10.1016/j.cbpc.2024.109942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Aflatoxin B1 (AFB1) is highly toxic to the liver and can cause excessive production of mitochondrial reactive oxygen species (mtROS) in hepatocytes, leading to oxidative stress, inflammation, fibrosis, cirrhosis, and even liver cancer. The overproduction of mtROS can induce mitophagy, but whether mtROS and mitophagy are involved in the liver injury induced by AFB1 in ducks remains unclear. In this study, we first demonstrated that overproduction of mtROS and mitophagy occurred during liver injury induced by AFB1 exposure in ducks. Then, by inhibiting mtROS and mitophagy, we found that the damage caused by AFB1 in ducks was significantly alleviated, and the overproduction of mtROS induced by AFB1 exposure could mediate the occurrence of mitophagy. These results suggested that mtROS-mediated mitophagy is involved in AFB1-induced duck liver injury, and they may be the prevention and treatment targets of AFB1 hepatotoxicity.
Collapse
Affiliation(s)
- Yilong Cui
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Qi Wang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yun Shi
- Tongliao City Animal Quarantine Technical Service Center, Tongliao 028000, China
| | - Yang Dai
- Tongliao Agriculture and Animal Husbandry Bureau, Tongliao 028000, China
| | - Yanfen Liu
- Institute of Animal Husbandry and Veterinary Medicine, Liaoning Agricultural Vocational and Technical College, Yingkou 115009, China.
| |
Collapse
|
10
|
Guo R, Yu Y, Xu C, Ma M, Hou C, Dong X, Wu J, Ouyang C, Ling J, Huang T. Protective effects of curcumin on corneal endothelial cell PANoptosis and monocyte adhesion induced by tumor necrosis factor-alpha and interferon-gamma in rats. Exp Eye Res 2024; 245:109952. [PMID: 38838973 DOI: 10.1016/j.exer.2024.109952] [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: 02/18/2024] [Revised: 04/18/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
Decrease of human corneal endothelial cell (CEC) density leads to corneal edema, progressive corneal opacity, and reduced visual acuity. A reduction in CEC density may be related to elevated levels of inflammatory cytokines, such as tumor necrosis factor (TNF)-α and interferon (INF)-γ. PANoptosis, characterized by the activation of apoptosis, necroptosis, and pyroptosis, could be a factor in the loss of CECs driven by TNF-α and INF-γ. Cytokines also stimulate monocytes adhesion to endothelium. It has been shown in previous research that curcumin plays protective roles against numerous corneal inflammatory diseases. However, it is not determined whether curcumin acts as an anti-PANoptotic agent or if it mitigates monocyte adhesion to CECs. Therefore, this research aimed to explor the potential therapeutic effects of curcumin and its underlying mechanisms in the loss of CECs. CEC injury models were established, and curcumin was injected subconjunctivally. Clinical evaluation of the corneas was conducted using a scoring system and anterior segment photography. Corneal observation was performed with hematoxylin and eosin staining and immunostaining of zona occludens-1(ZO-1). Apoptotic cells within the corneal endothelium were observed using TUNEL staining. The detection of primary proteins expression was accomplished through Western blot analysis. Interleukin (IL)-1β and macrophage chemotactic protein 1 (MCP-1) levels were determined via ELISA, while the expression of cleaved caspase-3, gasdermin-D (GSDMD), phosphor-mixed lineage kinase domain-like protein (p-MLKL) and intercellular cell adhesion molecule-1 were confirmed by immunofluorescence. Myeloperoxidase (MPO) activity was measured in aqueous humors. Curcumin treatment attenuated the loss of CECs and corneal edema caused by TNF-α and IFN-γ. Besides, it decreased the count of TUNEL-positive cells, and inhibited the upregulation of cleaved caspase-3, cleaved caspase-6, cleaved caspase-7, and cleaved poly (ADP-ribose) polymerase. Moreover, both the expression and phosphorylation of MLKL and receptor-interacting protein 3 were decreased in curcumin-treated rats. Furthermore, curcumin also lowered the expression of cleaved caspase-1, diminished the levels of IL1β and MCP-1, and inhibited the activity of MPO. Besides, the expression of intercellular cell adhesion molecule-1, vascular cell adhesion molecule-1, as well as the number of CD11b-positive cells adhered to the CECs decreased for the administration of curcumin.
Collapse
Affiliation(s)
- Ruilin Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yi Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chenjia Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Minglu Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chao Hou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaojuan Dong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jing Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chen Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jie Ling
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ting Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China.
| |
Collapse
|
11
|
Yang D, Zhang S, Cao H, Wu H, Liang Y, Teng CB, Yu HF. Detoxification of Aflatoxin B 1 by Phytochemicals in Agriculture and Food Science. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14481-14497. [PMID: 38897919 DOI: 10.1021/acs.jafc.4c01796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Aflatoxin B1 (AFB1), the most toxic and harmful mycotoxin, has a high likelihood of occurring in animal feed and human food, which seriously affects agriculture and food safety and endangers animal and human health. Recently, natural plant products have attracted widespread attention due to their low toxicity, high biocompatibility, and simple composition, indicating significant potential for resisting AFB1. The mechanisms by which these phytochemicals resist toxins mainly involve antioxidative, anti-inflammatory, and antiapoptotic pathways. Moreover, these substances also inhibit the genotoxicity of AFB1 by directly influencing its metabolism in vivo, which contributes to its elimination. Here, we review various phytochemicals that resist AFB1 and their anti-AFB1 mechanisms in different animals, as well as the common characteristics of phytochemicals with anti-AFB1 function. Additionally, the shortcomings of current research and future research directions will be discussed. Overall, this comprehensive summary contributes to the better application of phytochemicals in agriculture and food safety.
Collapse
Affiliation(s)
- Dian Yang
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Sihua Zhang
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Hongda Cao
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Huan Wu
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Yang Liang
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Chun-Bo Teng
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Hai-Fan Yu
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, College of Life Science, Northeast Forestry University, Harbin 150040, China
| |
Collapse
|
12
|
Liao C, Xu F, Yu Z, Ding K, Jia Y. The Novel Role of the NLRP3 Inflammasome in Mycotoxin-Induced Toxicological Mechanisms. Vet Sci 2024; 11:291. [PMID: 39057975 PMCID: PMC11281663 DOI: 10.3390/vetsci11070291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Mycotoxins are secondary metabolites produced by several fungi and moulds that exert toxicological effects on animals including immunotoxicity, genotoxicity, hepatotoxicity, teratogenicity, and neurotoxicity. However, the toxicological mechanisms of mycotoxins are complex and unclear. The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome is a multimeric cytosolic protein complex composed of the NLRP3 sensor, ASC adapter protein, and caspase-1 effector. Activation of the NLRP3 inflammasome plays a crucial role in innate immune defence and homeostatic maintenance. Recent studies have revealed that NLRP3 inflammasome activation is linked to tissue damage and inflammation induced by mycotoxin exposure. Thus, this review summarises the latest advancements in research on the roles of NLRP3 inflammasome activation in the pathogenesis of mycotoxin exposure. The effects of exposure to multiple mycotoxins, including deoxynivalenol, aflatoxin B1, zearalenone, T-2 toxin, ochratoxin A, and fumonisim B1, on pyroptosis-related factors and inflammation-related factors in vitro and in vivo and the pharmacological inhibition of specific and nonspecific NLRP3 inhibitors are summarized and examined. This comprehensive review contributes to a better understanding of the role of the NLRP3 inflammasome in toxicity induced by mycotoxin exposure and provides novel insights for pharmacologically targeting NLRP3 as a novel anti-inflammatory agent against mycotoxin exposure.
Collapse
Affiliation(s)
- Chengshui Liao
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Fengru Xu
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Zuhua Yu
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Ke Ding
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| | - Yanyan Jia
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China; (C.L.); (F.X.); (Z.Y.); (K.D.)
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471023, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China
| |
Collapse
|
13
|
Dai C, Sharma G, Liu G, Shen J, Shao B, Hao Z. Therapeutic detoxification of quercetin for aflatoxin B1-related toxicity: Roles of oxidative stress, inflammation, and metabolic enzymes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 345:123474. [PMID: 38309422 DOI: 10.1016/j.envpol.2024.123474] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024]
Abstract
Aflatoxins (AFTs), a type of mycotoxin mainly produced by Aspergillus parasiticus and Aspergillus flavus, could be detected in food, feed, Chinese herbal medicine, grain crops and poses a great threat to public health security. Among them, aflatoxin B1 (AFB1) is the most toxic one. Exposure to AFB1 poses various health risks to both humans and animals, including the development of chronic inflammatory diseases, cardiovascular diseases, neurodegenerative diseases, and cancer. The molecular mechanisms underlying these risks are intricate and dependent on specific contexts. This review primarily focuses on summarizing the protective effects of quercetin, a natural phenolic compound, in mitigating the toxic effects induced by AFB1 in both in vitro experiments and animal models. Additionally, the review explores the molecular mechanisms that underlie these protective effects. Quercetin has been demonstrated to not only have the direct inhibitory action on the production of AFTs from Aspergillus, both also possess potent ameliorative effects against AFB1-induced cytotoxicity, hepatotoxicity, and neurotoxicity. These effects are attributed to the inhibition of oxidative stress, mitochondrial dysfunction, mitochondrial apoptotic pathway, and inflammatory response. It could also directly target several metabolic enzymes (i.e., CYP3As and GSTA1) to reduce the production of toxic metabolites of AFB1 within cells, then reduce AFB1-induced cytotoxicity. In conclusion, this review highlights quercetin is a promising detoxification agent for AFB1. By advancing our understanding of the protective mechanisms offered by quercetin, we aim to contribute to the development of effective detoxification agents against AFB1, ultimately promoting better health outcomes.
Collapse
Affiliation(s)
- Chongshan Dai
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, PR China; Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, PR China
| | - Gaurav Sharma
- Cardiovascular and Thoracic Surgery, Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Gaoyi Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, PR China; Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, PR China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, PR China; Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, PR China
| | - Bing Shao
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Centre for Disease Control and Prevention, Beijing, 100013, PR China
| | - Zhihui Hao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, PR China; Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, PR China.
| |
Collapse
|
14
|
Jiang Z, Liu L, Su H, Cao Y, Ma Z, Gao Y, Huang D. Curcumin and analogues in mitigating liver injury and disease consequences: From molecular mechanisms to clinical perspectives. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155234. [PMID: 38042008 DOI: 10.1016/j.phymed.2023.155234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/01/2023] [Accepted: 11/20/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND Liver injury is a prevalent global health concern, impacting a substantial number of individuals and leading to elevated mortality rates and socioeconomic burdens. Traditional primary treatment options encounter resource constraints and high costs, prompting exploration of alternative adjunct therapies, such as phytotherapy. Curcumin demonstrates significant therapeutic potential across various medical conditions, particularly emerging as a promising candidate for liver injury treatment. PURPOSE This study aims to provide current evidence maps of curcumin and its analogs in the context of liver injury, covering aspects of biosafety, toxicology, and clinical trials. Importantly, it seeks to summarize the intricate mechanisms modulated by curcumin. METHODS We conducted a comprehensive search of MEDLINE, Web of Science, and Embase up to July 2023. Titles and abstracts were reviewed to identify studies that met our eligibility criteria. The screening process involved three authors independently assessing the potential of curcumin mitigating liver injury and its disease consequences by reviewing titles, abstracts, and full texts. RESULTS Curcumin and its analogs have demonstrated low toxicity in vitro and in vivo. However, the limited bioavailability has hindered their advanced use in liver injury. This limitation can potentially be addressed by nano-curcumin and emerging drug delivery systems. Curcumin plays a role in alleviating liver injury by modulating the antioxidant system, as well as cellular and molecular pathways. The specific mechanisms involve multiple pathways, such as NF-κB, p38/MAPK, and JAK2/STAT3, and the pro-apoptosis Bcl-2/Bax/caspase-3 axis in damaged cells. Additionally, curcumin targets nutritional metabolism, regulating the substance in liver cells and tissues. The microenvironment associated with liver injury, like extracellular matrix and immune cells and factors, is also regulated by curcumin. Initial evaluation of curcumin and its analogs through 12 clinical trials demonstrates their potential application in liver injury. CONCLUSION Curcumin emerges as a promising phytomedicine for liver injury owing to its effectiveness in hepatoprotection and low toxicity profile. Nevertheless, in-depth investigations are warranted to unravel the complex mechanisms through which curcumin influences liver tissues and overall physiological milieu. Moreover, extensive clinical trials are essential to determine optimal curcumin dosage forms, maximizing its benefits and achieving favorable clinical outcomes.
Collapse
Affiliation(s)
- Zhishen Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Liu Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hengpei Su
- College of Materials Science and Engineering, Sichuan University, Chengdu 610064, China
| | - Yubin Cao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Zhongkai Ma
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yujie Gao
- Department of Stomatology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
David IG, Iorgulescu EE, Popa DE, Buleandra M, Cheregi MC, Noor H. Curcumin Electrochemistry-Antioxidant Activity Assessment, Voltammetric Behavior and Quantitative Determination, Applications as Electrode Modifier. Antioxidants (Basel) 2023; 12:1908. [PMID: 38001760 PMCID: PMC10669510 DOI: 10.3390/antiox12111908] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Curcumin (CU) is a polyphenolic compound extracted from turmeric, a well-known dietary spice. Since it has been shown that CU exerts beneficial effects on human health, interest has increased in its use but also in its analysis in different matrices. CU has an antioxidant character and is electroactive due to the presence of phenolic groups in its molecule. This paper reviews the data reported in the literature regarding the use of electrochemical techniques for the assessment of CU antioxidant activity and the investigation of the voltammetric behavior at different electrodes of free or loaded CU on various carriers. The performance characteristics and the analytical applications of the electrochemical methods developed for CU analysis are compared and critically discussed. Examples of voltammetric investigations of CU interaction with different metallic ions or of CU or CU complexes with DNA as well as the CU applications as electrode modifiers for the enhanced detection of various chemical species are also shown.
Collapse
Affiliation(s)
- Iulia Gabriela David
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Panduri Av. 90-92, District 5, 050663 Bucharest, Romania; (D.E.P.); (M.B.); (M.C.C.)
| | - Emilia Elena Iorgulescu
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Panduri Av. 90-92, District 5, 050663 Bucharest, Romania; (D.E.P.); (M.B.); (M.C.C.)
| | - Dana Elena Popa
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Panduri Av. 90-92, District 5, 050663 Bucharest, Romania; (D.E.P.); (M.B.); (M.C.C.)
| | - Mihaela Buleandra
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Panduri Av. 90-92, District 5, 050663 Bucharest, Romania; (D.E.P.); (M.B.); (M.C.C.)
| | - Mihaela Carmen Cheregi
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Panduri Av. 90-92, District 5, 050663 Bucharest, Romania; (D.E.P.); (M.B.); (M.C.C.)
| | - Hassan Noor
- Department of Surgery, Faculty of Medicine, “Lucian Blaga” University Sibiu, Lucian Blaga Street 25, 550169 Sibiu, Romania;
- Medlife-Polisano Hospital, Strada Izvorului 1A, 550172 Sibiu, Romania
| |
Collapse
|