1
|
Wang F, Wang X, Xiao Y, Liu R, Li X, Hu J, Song W, Feng K, Yuan Y, Yue T. Selenium-enriched Kazachstania unispora KU2 ameliorates patulin-induced intestinal injury in mice by mediating the gut microbiota and selenoprotein P synthesis. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138129. [PMID: 40179778 DOI: 10.1016/j.jhazmat.2025.138129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Patulin (PAT) is a foodborne mycotoxin that causes intestinal injury. Selenium (Se)-enriched Kazachstania unispora (K. unispora) KU2 is a novel dietary Se carrier, and Se exerts important roles in intestinal homeostasis. Here, we examined the ameliorative effects of K. unispora KU2 and Se-enriched K. unispora KU2 against PAT-induced intestinal injury. Results indicated that both K. unispora KU2 and Se-enriched K. unispora KU2 alleviated PAT-induced inflammatory infiltration, disrupted gut microbiota, and associated metabolic imbalances, indicating the probiotic potential of this strain. Se-enriched K. unispora KU2 exhibited more pronounced remediation comparable to K. unispora KU2, revealing the promoting effect of Se. Furthermore, Se-enriched K. unispora KU2 restored intestinal Se homeostasis by upregulating SEPP1 levels to mitigate intestinal injury. Using pseudo germ-free mouse models, we confirmed that gut microbiota was required for the improvement in SEPP1 synthesis and intestinal transport mediated by Se-enriched K. unispora KU2. These findings elucidate a mechanism whereby the alleviation of PAT-induced intestinal injury by Se-enriched K. unispora KU2 is linked to upregulation of SEPP1 by the gut microbiota, suggesting its potential therapeutic relevance for intestinal diseases.
Collapse
Affiliation(s)
- Furong Wang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Xian Wang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Yilei Xiao
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Ruixin Liu
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Xiaoben Li
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Jinpeng Hu
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Wei Song
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Kewei Feng
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China.
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
2
|
Zhang T, Chang M, Hou X, Yan M, Zhang S, Song W, Sheng Q, Yuan Y, Yue T. Apple polyphenols prevent patulin-induced intestinal damage by modulating the gut microbiota and metabolism of the gut-liver axis. Food Chem 2025; 463:141049. [PMID: 39260178 DOI: 10.1016/j.foodchem.2024.141049] [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: 05/23/2024] [Revised: 08/06/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
Patulin (PAT), a foodborne toxin, causes severe intestinal damage. To mitigate this health threat, mice were pretreated with apple polyphenols (AP) in their drinking water (0.01 % and 0.05 %) for eight weeks, followed by exposure to PAT during the last two weeks. Subsequently, histopathological and biochemical evaluations of intestinal tissues were conducted, alongside assessments of alterations in gut microbiota, colonic content metabolome, and hepatic metabolome. Consequently, AP alleviated PAT-induced villus and crypt injury, mucus depletion, GSH level decline, GSH-Px and SOD activity reduction, and MPO activity elevation. Notably, AP counteracted PAT-mediated microbiota disruptions and promoted the abundance of beneficial bacteria (Dubosiella, Akkermansia, Lachnospiraceae, and Lactobacillus). Furthermore, AP counteracted PAT-induced metabolic disorders in the colonic contents and liver. Ultimately, AP prevented intestinal injury by regulating the gut microbiota and amino acid, purine, butanoate, and glycerophospholipid metabolism in the gut-liver axis. These results underscore the potential of AP to prevent foodborne toxin-induced intestinal damage.
Collapse
Affiliation(s)
- Ting Zhang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Min Chang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Xiaohui Hou
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Min Yan
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Shirui Zhang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Wei Song
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Qinglin Sheng
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China.
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China.
| |
Collapse
|
3
|
Lemos FS, Prins CA, Martinez AMB, Carpi-Santos R, Neumann AS, Meireles-da-Costa N, Luisetto R, de Mello-Coelho V, Oliveira FL. UHT Cow's Milk Supplementation Affects Cell Niches and Functions of the Gut-Brain Axis in BALB/c Mice. Biomedicines 2024; 12:2448. [PMID: 39595015 PMCID: PMC11591918 DOI: 10.3390/biomedicines12112448] [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: 10/05/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Cow's milk is a bioactive cocktail with essential nutritional factors that is widely consumed during early childhood development. However, it has been associated with allergic responses and immune cell activation. Here, we investigate whether cow's milk consumption regulates gut-brain axis functions and affects patterns of behaviors in BALB/c mice, previously described by present low sociability, significant stereotypes, and restricted interest features. The major objectives consist of to investigate cow's milk supplementation as possible triggers interfering with cellular niches of the gut-brain axis and behavioral patterns. METHODS Male BALB/c at 6 weeks were randomly divided into two groups, one supplemented with cow's milk processed at ultra-high temperature (UHT) and another group receiving water (controls) three times per day (200 μL per dose) for one week. RESULTS Milk consumption disturbed histological compartments of the small intestine, including niches of KI67+-proliferating cells and CD138+ Ig-secreting plasma cells. In the liver, milk intake was associated with pro-inflammatory responses, oxidative stress, and atypical glycogen distribution. Milk-supplemented mice showed significant increase in granulocytes (CD11b+SSChigh cells) and CD4+ T cells in the blood. These mice also had neuroinflammatory signals, including an enhanced number of cortical Iba-1+ microglial cells in the brain and significant cerebellar expression of nitric oxide synthase 2 by Purkinje cells. These phenotypes and tissue disorders in milk-supplemented mice were associated with atypical behaviors, including low sociability, high restricted interest, and severe stereotypies. Moreover, synaptic niches were also disturbed after milk consumption, and Shank-3+ and Drebrin+ post-synaptic cells were significantly reduced in the brain of these mice. CONCLUSIONS Together, these data suggest that milk consumption interfered with the gut-brain axis in BALB/c mice and increased atypical behaviors, at least in part, linked to synapse dysfunctions, neuroinflammation, and oxidative stress regulation.
Collapse
Affiliation(s)
- Felipe S. Lemos
- Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-360, Brazil;
| | - Caio A. Prins
- Department of Pathology, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (C.A.P.); (A.M.B.M.)
| | - Ana M. B. Martinez
- Department of Pathology, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (C.A.P.); (A.M.B.M.)
| | - Raul Carpi-Santos
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (R.C.-S.); (A.S.N.); (V.d.M.-C.)
| | - Arthur S. Neumann
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (R.C.-S.); (A.S.N.); (V.d.M.-C.)
| | - Nathalia Meireles-da-Costa
- Molecular Carcinogenesis Program, Brazilian National Cancer Institute, Rio de Janeiro 20230-130, Brazil;
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35124 Padova, Italy;
| | - Valeria de Mello-Coelho
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (R.C.-S.); (A.S.N.); (V.d.M.-C.)
| | - Felipe L. Oliveira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (R.C.-S.); (A.S.N.); (V.d.M.-C.)
| |
Collapse
|
4
|
Fan L, Hu H. Involvement of multiple forms of cell death in patulin-induced toxicities. Toxicon 2024; 244:107768. [PMID: 38768831 DOI: 10.1016/j.toxicon.2024.107768] [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: 01/22/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Patulin (PAT) is the most common mycotoxin found in moldy fruits and their derived products, and is reported to cause diverse toxic effects, including hepatotoxicity, nephrotoxicity, cardiotoxicity, neurotoxicity, immunotoxicity, gastrointestinal toxicity and dermal toxicity. The cell death induction by PAT is suggested to be a key cellular mechanism involved in PAT-induced toxicities. Accumulating evidence indicates that the multiple forms of cell death are induced in response to PAT exposure, including apoptosis, autophagic cell death, pyroptosis and ferroptosis. Mechanistically, the cell death induction by PAT is associated the oxidative stress induction via reducing the antioxidant capacity or inducing pro-oxidant NADPH oxidase, the activation of mitochondrial pathway via regulating BCL-2 family proteins, the disruption of iron metabolism through ferritinophagy-mediated ferritin degradation, and the induction of the NOD-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome/caspase-1/gasdermin D (GSDMD) pathway. In this review article, we summarize the present understanding of the cell death induction by PAT, discuss the potential signaling pathways underlying PAT-induced cell death, and propose the issues that need to be addressed to promote the development of cell death-based approach to counteract PAT-induced toxicities.
Collapse
Affiliation(s)
- Lihong Fan
- College of Veterinary Medicine, China Agricultural University, No.2 Yuanmingyuan West Road, Haidian District, Beijing, 100193, China.
| | - Hongbo Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua East Road, Haidian District, Beijing, 100083, China
| |
Collapse
|
5
|
Zhang T, Yan M, Chang M, Hou X, Wang F, Song W, Wang Y, Feng K, Yuan Y, Yue T. Integrated transcriptomics and metabolomics reveal the mechanism of intestinal damage upon acute patulin exposure in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116270. [PMID: 38574645 DOI: 10.1016/j.ecoenv.2024.116270] [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: 01/03/2024] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
Mycotoxin contamination has become a major food safety issue and greatly threatens human and animal health. Patulin (PAT), a common mycotoxin in the environment, is exposed through the food chain and damages the gastrointestinal tract. However, its mechanism of enterotoxicity at the genetic and metabolic levels remains to be elucidated. Herein, the intestinal histopathological and biochemical indices, transcriptome, and metabolome of C57BL/6 J mice exposed to different doses of PAT were successively assessed, as well as the toxicokinetics of PAT in vivo. The results showed that acute PAT exposure induced damaged villi and crypts, reduced mucus secretion, decreased SOD and GSH-Px activities, and enhanced MPO activity in the small intestine and mild damage in the colon. At the transcriptional level, the genes affected by PAT were dose-dependently altered in the small intestine and fluctuated in the colon. PAT primarily affected inflammation-related signaling pathways and oxidative phosphorylation in the small intestine and immune responses in the colon. At the metabolic level, amino acids decreased, and extensive lipids accumulated in the small intestine and colon. Seven metabolic pathways were jointly affected by PAT in two intestinal sites. Moreover, changes in PAT products and GST activity were detected in the small intestinal tissue but not in the colonic tissue, explaining the different damage degrees of the two sites. Finally, the integrated results collectively explained the toxicological mechanism of PAT, which damaged the small intestine directly and the colon indirectly. These results paint a clear panorama of intestinal changes after PAT exposure and provide valuable information on the exposure risk and toxic mechanism of PAT.
Collapse
Affiliation(s)
- Ting Zhang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Min Yan
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Min Chang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Xiaohui Hou
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Furong Wang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Wei Song
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Yuan Wang
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Kewei Feng
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an, Shaanxi 710069, China; Research Center of Food Safety Risk Assessment and Control, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
6
|
Zeng C, Liu J, Zheng X, Hu X, He Y. Prostaglandin and prostaglandin receptors: present and future promising therapeutic targets for pulmonary arterial hypertension. Respir Res 2023; 24:263. [PMID: 37915044 PMCID: PMC10619262 DOI: 10.1186/s12931-023-02559-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH), Group 1 pulmonary hypertension (PH), is a type of pulmonary vascular disease characterized by abnormal contraction and remodeling of the pulmonary arterioles, manifested by pulmonary vascular resistance (PVR) and increased pulmonary arterial pressure, eventually leading to right heart failure or even death. The mechanisms involved in this process include inflammation, vascular matrix remodeling, endothelial cell apoptosis and proliferation, vasoconstriction, vascular smooth muscle cell proliferation and hypertrophy. In this study, we review the mechanisms of action of prostaglandins and their receptors in PAH. MAIN BODY PAH-targeted therapies, such as endothelin receptor antagonists, phosphodiesterase type 5 inhibitors, activators of soluble guanylate cyclase, prostacyclin, and prostacyclin analogs, improve PVR, mean pulmonary arterial pressure, and the six-minute walk distance, cardiac output and exercise capacity and are licensed for patients with PAH; however, they have not been shown to reduce mortality. Current treatments for PAH primarily focus on inhibiting excessive pulmonary vasoconstriction, however, vascular remodeling is recalcitrant to currently available therapies. Lung transplantation remains the definitive treatment for patients with PAH. Therefore, it is imperative to identify novel targets for improving pulmonary vascular remodeling in PAH. Studies have confirmed that prostaglandins and their receptors play important roles in the occurrence and development of PAH through vasoconstriction, vascular smooth muscle cell proliferation and migration, inflammation, and extracellular matrix remodeling. CONCLUSION Prostacyclin and related drugs have been used in the clinical treatment of PAH. Other prostaglandins also have the potential to treat PAH. This review provides ideas for the treatment of PAH and the discovery of new drug targets.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China
| | - Jing Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China
| | - Xialei Zheng
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China
| | - Xinqun Hu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China.
| | - Yuhu He
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China.
| |
Collapse
|
7
|
Sun WC, Wang NN, Li R, Sun XC, Liao JW, Yang G, Liu S. Ferritinophagy activation and sideroflexin1-dependent mitochondrial iron overload contribute to patulin-induced cardiac inflammation and fibrosis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 892:164472. [PMID: 37257617 DOI: 10.1016/j.scitotenv.2023.164472] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
Patulin (PAT) is a mycotoxin that is commonly present throughout the ecosystem where fungi grow and mainly contaminates food, soil, and water. PAT was found to be cardiotoxic in previous studies. However, the detailed mechanism has not been fully elucidated. The present study aimed to explore the role and underlying mechanism of ferroptosis in PAT-induced cardiac injury. Here, we confirmed in vivo and in vitro that ferroptosis is involved in PAT-induced myocardial inflammation and fibrosis. Mice exposed to PAT (1 and 2 mg/kg body weight/day for 14 days) exhibited myocardial inflammation and fibrosis along with disrupted iron homeostasis, elevated lipid peroxidation, depletion of glutathione peroxidase 4, and abnormal mitochondrial morphology. When primary neonatal rat cardiomyocytes (NRCMs) and H9c2 cells were exposed to PAT, ferroptosis was initiated in a dose-dependent manner, and this process could be significantly attenuated by ferrostatin-1. Mechanistically, we found that nuclear receptor coactivator (NCOA) 4, a master regulator of ferritinophagy, bound to and degraded ferritin in response to PAT treatment, thereby releasing large amounts of ferrous iron and further leading to sideroflexin (SFXN) 1-dependent mitochondrial iron overload. Conversely, knockdown of NCOA4 or SFXN1 with small interfering RNAs could effectively ameliorate ferroptotic cell death, cellular or mitochondrial iron overload and lipid peroxides accumulation. Furthermore, myocardial inflammation and fibrosis in PAT-exposed mice was alleviated by the mitochondrial iron chelator deferiprone. Overall, our findings underscore that ferritinophagy activation and SFXN1-dependent mitochondrial iron overload play critical roles in PAT-induced myocardial ferroptosis and consequent cardiotoxicity.
Collapse
Affiliation(s)
- Wen-Chang Sun
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Ning-Ning Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Ru Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Xian-Ce Sun
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Jia-Wei Liao
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Guang Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Shuang Liu
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
8
|
Chen H, Du G, Yan X, Ye H, Guo Q, Wang Z, Yuan Y, Yue T. Selenium-Enriched Pediococcus acidilactici MRS-7 Alleviates Patulin-Induced Jejunum Injuries in Mice and Its Possible Mechanisms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:4755-4764. [PMID: 35394776 DOI: 10.1021/acs.jafc.2c00949] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Patulin (PAT) is a common mycotoxin. Oral ingestion of PAT could damage the intestinal mucosa. Both selenium and probiotics can alleviate intestinal damage, but there are few reports on selenium-enriched probiotics. Here, we studied the protective effects of a new selenium-enriched Pediococcus acidilactici MRS-7 (SeP) on PAT-induced jejunum injuries in mice. Results show that PAT induced jejunum injuries such as loss of crypts, ulceration of the mucosa, and intestinal epithelial barrier function impairment. However, SeP could protect against PAT-induced jejunum injuries and significantly inhibit the reduction of goblet cell numbers. SeP could not only alleviate PAT-induced oxidative stress by decreasing malondialdehyde (MDA) and increasing superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) levels in the jejunum tissues but also alleviate the inflammatory response caused by PAT by reducing the levels of inflammatory factors (interleukin (IL)-6 snd IL-1β and tumor necrosis factor-α (TNF-α)) in the serum and jejunum tissues. In addition, SeP also inhibited the expression of nuclear factor-κB (NF-κB) and Toll-like receptor 4 (TLR-4), increased the expression of tight junction proteins (occludin, ZO-1, and claudin-1), and increased the selenium content in the jejunum, thereby antagonizing the jejunum injuries caused by PAT exposure. Finally, SeP rebalanced the intestinal microbiota and improved probiotic abundance such as Turicibacter, Bifidobacterium, Ileibacterium, and Pediococcus in PAT-treated mice. These results support the possibility of SeP as a novel protective agent to mitigate the toxicity of PAT.
Collapse
Affiliation(s)
- Hong Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Gengan Du
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Xiaohai Yan
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Huanfeng Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Qi Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Zhouli Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Yahong Yuan
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
| | - Tianli Yue
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Laboratory of Quality & Safety Risk Assessment for Agro-products (Yangling), Ministry of Agriculture, Yangling 712100, China
- College of Food Science and Technology, Northwest University, Xi'an 710000, China
| |
Collapse
|
9
|
Effects of Selenium Nanoparticles on Preventing Patulin-Induced Liver, Kidney and Gastrointestinal Damage. Foods 2022; 11:foods11050749. [PMID: 35267382 PMCID: PMC8909330 DOI: 10.3390/foods11050749] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/26/2022] [Accepted: 03/01/2022] [Indexed: 01/27/2023] Open
Abstract
Patulin (PAT) is a toxic fungal metabolite, and oxidative damage was proved to be its important toxicity mechanism. Selenium nanoparticles (SeNPs) were prepared by reducing sodium selenite with chitosan as a stabilizer and used for preventing PAT-induced liver, kidney and gastrointestinal damage. SeNPs have good dispersibility, in vitro antioxidant activity, and are much less cytotoxic than sodium selenite. Cell culture studies indicated that SeNPs can effectively alleviate PAT-induced excessive production of intracellular ROS, the decline of glutathione peroxidase activity, and the suppression of cell viability. Evaluation of serum biochemical parameters, histopathology, oxidative stress biomarkers and activities of antioxidant enzymes in a mouse model showed that pre-treatment with SeNPs (2 mg Se/kg body weight) could ameliorate PAT-induced oxidative damage to the liver and kidneys of mice, but PAT-induced gastrointestinal oxidative damage and barrier dysfunction were not recovered by SeNPs, possibly because the toxin doses suffered by the gastrointestinal as the first exposed tissues exceeded the regulatory capacity of SeNPs. These results suggested that a combination of other strategies may be required to completely block PAT toxicity.
Collapse
|
10
|
Transcriptomic and proteomic insights into patulin mycotoxin-induced cancer-like phenotypes in normal intestinal epithelial cells. Mol Cell Biochem 2022; 477:1405-1416. [PMID: 35150386 DOI: 10.1007/s11010-022-04387-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/02/2022] [Indexed: 10/19/2022]
Abstract
Patulin (PAT) is a natural contaminant of fruits (primarily apples) and their products. Significantly, high levels of contamination have been found in fruit juices all over the world. Several in vitro studies have demonstrated PAT's ability to alter intestinal structure and function. However, in real life, the probability of low dose long-term exposure to PAT to humans is significantly higher through contaminated food items. Thus, in the present study, we have exposed normal intestinal cells to non-toxic levels of PAT for 16 weeks and observed that PAT had the ability to cause cancer-like properties in normal intestinal epithelial cells after chronic exposure. Here, our results showed that chronic exposure to low doses of PAT caused enhanced proliferation, migration and invasion ability, and the capability to grow in soft agar (anchorage independence). Moreover, an in vivo study showed the appearance of colonic aberrant crypt foci (ACFs) in PAT-exposed Wistar rats, which are well, establish markers for early colon cancer. Furthermore, as these neoplastic changes are consequences of alterations at the molecular level, here, we combined next-generation RNA sequencing with liquid chromatography mass spectrometry-based proteomic analysis to investigate the possible underlying mechanisms involved in PAT-induced neoplastic changes.
Collapse
|
11
|
Pal S, Singh N, Dev I, Sharma V, Jagdale PR, Ayanur A, Ansari KM. TGF-β/Smad signaling pathway plays a crucial role in patulin-induced pro-fibrotic changes in rat kidney via modulation of slug and snail expression. Toxicol Appl Pharmacol 2022; 434:115819. [PMID: 34896196 DOI: 10.1016/j.taap.2021.115819] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 01/12/2023]
Abstract
Patulin (PAT) is a mycotoxin that contaminates a variety of food and foodstuffs. Earlier in vitro and in vivo findings have indicated that kidney is one of the target organs for PAT-induced toxicity. However, no study has evaluated the chronic effects of PAT exposure at environmentally relevant doses or elucidated the detailed mechanism(s) involved. Here, using in vitro and in vivo experimental approaches, we delineated the mechanism/s involved in pro-fibrotic changes in the kidney after low-dose chronic exposure to PAT. We found that non-toxic concentrations (50 nM and 100 nM) of PAT to normal rat kidney cells (NRK52E) caused a higher generation of reactive oxygen species (ROS) (mainly hydroxyl (•OH), peroxynitrite (ONOO-), and hypochlorite radical (ClO-). PAT exposure caused the activation of mitogen-activated protein kinases (MAPKs) and its downstream c-Jun/Fos signaling pathways. Moreover, our chromatin immunoprecipitation (ChIP) analysis suggested that c-Jun/Fos binds to the promoter region of Transforming growth factor beta (TGF-β1) and possibly induces its expression. Results showed that PAT-induced TGF-β1 further activates the TGF-β1/smad signaling pathways. Higher activation of slug and snail transcription factors further modulates the regulation of pro-fibrotic molecules. Similarly, in vivo results showed that PAT exposure to rats through gavage at 25 and 100 μg/kg b. wt had higher levels of kidney injury/toxicity markers namely vascular endothelial growth factor (VEGF), kidney Injury Molecule-1 (Kim-1), tissue inhibitor of metalloproteinase-1 (Timp-1), and clusterin (CLU). Additionally, histopathological analysis indicated significant alterations in renal tubules and glomeruli along with collagen deposition in PAT-treated rat kidneys. Overall, our data provide evidence of the involvement of ROS mediated MAPKs and TGF-β1/smad pathways in PAT-induced pro-fibrotic changes in the kidney via modulation of slug and snail expression.
Collapse
Affiliation(s)
- Saurabh Pal
- Food Toxicology Laboratory, Food, Drug, and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India
| | - Neha Singh
- Food Toxicology Laboratory, Food, Drug, and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India
| | - Indra Dev
- Food Toxicology Laboratory, Food, Drug, and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India
| | - Vineeta Sharma
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Pankaj Ramji Jagdale
- Pathology Laboratory, Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 M. G. Marg, Lucknow 226001, Uttar Pradesh, India
| | - Anjaneya Ayanur
- Academy of Scientific and Innovative Research, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India; Pathology Laboratory, Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 M. G. Marg, Lucknow 226001, Uttar Pradesh, India
| | - Kausar Mahmood Ansari
- Food Toxicology Laboratory, Food, Drug, and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India.
| |
Collapse
|
12
|
Zhang B, Liang H, Huang K, Li J, Xu D, Huang C, Li Y. Cardiotoxicity of patulin was found in H9c2 cells. Toxicon 2021; 207:21-30. [PMID: 34929212 DOI: 10.1016/j.toxicon.2021.12.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/27/2022]
Abstract
Patulin (PAT) is a kind of mycotoxins that is universally found at rotten fruits, especially apples and apple products. Previous studies have shown that PAT has hepatotoxicity and nephrotoxicity. However, cardiotoxicity of PAT is rarely reported. Present study aimed at investigate the cardiotoxicity and relevant mechanisms of PAT on H9c2 cells. Cytotoxicity of PAT were evaluated by MTT assay and LDH. Hoechst 33258 staining was used to examine the nuclear morphology and AV/PI double staining was employed for apoptosis on H9c2 cells. Expression level of Caspase-3, Caspase-9, Bax, Bcl-2 were quantified to verify the potential mechanism of mitochondrial apoptosis pathway. The tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), and interleukin 6 (IL-6) were quantified to determine the inflammatory response by using ELISA assay. ROS, SOD, MDA, GSH levels were measured to determine the oxidative stress status. Results demonstrated that PAT significantly induced cell injury, as evidenced by the down-regulated of cell viability, and the increase of LDH release. Hoesst33258 staining and flow cytometry showed that apoptosis rate was elevated by PAT. PAT treatment up-regulated the expression of Caspase-3, Caspase-9, Bax level and down-regulated the expression of Bcl-2 level. TNF-α, IL-1β, IL-6 levels showed that PAT increased the pro-inflammatory response. As PAT concentration increased, intracellular MDA, ROS content were elevated, while GSH content and the activity of SOD were significantly decreased. Thus, it is concluded that PAT may induce apoptosis of H9c2 cells through oxidative stress.
Collapse
Affiliation(s)
- Baigang Zhang
- Life Science and Engineering, Lanzhou University of Technology, Gansu, Lanzhou, 730050, China
| | - Hairong Liang
- Life Science and Engineering, Lanzhou University of Technology, Gansu, Lanzhou, 730050, China
| | - Ke Huang
- School of Basic Medical Sciences, Lanzhou University, Gansu, Lanzhou, 730050, China; School/Hospital of Stomatology, Lanzhou University, Gansu, Lanzhou, 730050, China
| | - Jinliang Li
- Life Science and Engineering, Lanzhou University of Technology, Gansu, Lanzhou, 730050, China
| | - Dongmei Xu
- Life Science and Engineering, Lanzhou University of Technology, Gansu, Lanzhou, 730050, China
| | - Chenghui Huang
- Life Science and Engineering, Lanzhou University of Technology, Gansu, Lanzhou, 730050, China
| | - Yi Li
- School/Hospital of Stomatology, Lanzhou University, Gansu, Lanzhou, 730050, China.
| |
Collapse
|
13
|
Kozieł MJ, Ziaja M, Piastowska-Ciesielska AW. Intestinal Barrier, Claudins and Mycotoxins. Toxins (Basel) 2021; 13:758. [PMID: 34822542 PMCID: PMC8622050 DOI: 10.3390/toxins13110758] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 02/08/2023] Open
Abstract
The intestinal barrier is the main barrier against all of the substances that enter the body. Proper functioning of this barrier guarantees maintained balance in the organism. Mycotoxins are toxic, secondary fungi metabolites, that have a negative impact both on human and animal health. It was postulated that various mycotoxins may affect homeostasis by disturbing the intestinal barrier. Claudins are proteins that are involved in creating tight junctions between epithelial cells. A growing body of evidence underlines their role in molecular response to mycotoxin-induced cytotoxicity. This review summarizes the information connected with claudins, their association with an intestinal barrier, physiological conditions in general, and with gastrointestinal cancers. Moreover, this review also includes information about the changes in claudin expression upon exposition to various mycotoxins.
Collapse
|
14
|
Sluter MN, Hou R, Li L, Yasmen N, Yu Y, Liu J, Jiang J. EP2 Antagonists (2011-2021): A Decade's Journey from Discovery to Therapeutics. J Med Chem 2021; 64:11816-11836. [PMID: 34352171 PMCID: PMC8455147 DOI: 10.1021/acs.jmedchem.1c00816] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the wake of health disasters associated with the chronic use of cyclooxygenase-2 (COX-2) inhibitor drugs, it has been widely proposed that modulation of downstream prostanoid synthases or receptors might provide more specificity than simply shutting down the entire COX cascade for anti-inflammatory benefits. The pathogenic actions of COX-2 have long been thought attributable to the prostaglandin E2 (PGE2) signaling through its Gαs-coupled EP2 receptor subtype; however, the truly selective EP2 antagonists did not emerge until 2011. These small molecules provide game-changing tools to better understand the EP2 receptor in inflammation-associated conditions. Their applications in preclinical models also reshape our knowledge of PGE2/EP2 signaling as a node of inflammation in health and disease. As we celebrate the 10-year anniversary of this breakthrough, the exploration of their potential as drug candidates for next-generation anti-inflammatory therapies has just begun. The first decade of EP2 antagonists passes, while their future looks brighter than ever.
Collapse
Affiliation(s)
- Madison N Sluter
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Ruida Hou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Lexiao Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Nelufar Yasmen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Ying Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Jiawang Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Medicinal Chemistry Core, Office of Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
15
|
Zhang SL, Chen ZH, Lin DT, Yan Q, Gao F, Lin H. Epigallocatechin gallate regulates inflammatory responses and new bone formation through Wnt/β-Catenin/COX-2 pathway in spondyloarthritis. Int Immunopharmacol 2021; 98:107869. [PMID: 34153673 DOI: 10.1016/j.intimp.2021.107869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/24/2021] [Accepted: 06/06/2021] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Spondyloarthritis (SpA) is mainly characterized by bone erosion, new bone formation, inflammation and potential disability. Epigallocatechin gallate (EGCG) has been proved to be closely related with the regulation of inflammation and bone metabolism. However, whether EGCG could improve SpA remains unclear. METHODS SpA animal model was established using proteoglycan. Cell proliferation were measured by CCK-8 assay. The mRNA expression levels of genes were detected using qRT-PCR, protein levels were assessed via western blotting and immunohistochemistry. ELISA assay was performed to examined the inflammatory cytokine release. Lesions in spine cartilage tissues were observed using hematoxylin-eosin (HE) and Safranin O staining. Alkaline phosphatase (ALP) assay and Alizarin Red S staining was used to investigate osteoblast mineralization. RESULTS We found that EGCG could inhibit inflammation and new bone formation in SpA mice. Besides, inflammatory factor expression and osteogenic differentiation in osteoblasts isolated from SpA mice were also decreased by EGCG. Further, EGCG treatment suppressed the activation of Wnt/β-Catenin/COX-2 pathway and the activator of this pathway partially reversed the effects of EGCG on inflammation and osteoblast differentiation. CONCLUSIONS EGCG repressed inflammatory responses and new bone formation, and further improved SpA through Wnt/β-Catenin/COX-2 pathway. Our findings may provide a new thought for the prevention and treatment of SpA.
Collapse
Affiliation(s)
- Sheng-Li Zhang
- Department of Rheumatology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, PR China; Department of Rheumatology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China.
| | - Zhi-Han Chen
- Department of Rheumatology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, PR China; Department of Rheumatology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China
| | - Dian-Tian Lin
- Department of Rheumatology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, PR China; Department of Rheumatology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China
| | - Qing Yan
- Department of Rheumatology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, PR China; Department of Rheumatology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China
| | - Fei Gao
- Department of Rheumatology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, PR China; Department of Rheumatology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China
| | - He Lin
- Department of Rheumatology, Fujian Provincial Hospital, Fuzhou 350001, Fujian Province, PR China; Department of Rheumatology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian Province, PR China
| |
Collapse
|
16
|
Singh N, Sharma G, Dev I, Shukla SK, Ansari KM. Study of the metabolic alterations in patulin-induced neoplastic transformation in normal intestinal cells. Toxicol Res (Camb) 2021; 10:592-600. [PMID: 34141173 DOI: 10.1093/toxres/tfab023] [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: 11/06/2020] [Revised: 02/01/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
Several surveillance studies have reported significantly high level of patulin (PAT), mycotoxin in fruit juices suggesting the possible exposure to human. In vitro studies have showed that PAT can alter the permeability, ion transport and modulates tight junction of intestine. In real scenario, human can be exposed with low levels of PAT for longer duration through different fruits and their products. Hence, keeping this possibility in view, we conducted a study where normal intestinal cells were exposed with non-toxic levels of PAT for longer duration and found that PAT exposure causes cancer-like properties in normal intestinal cells. It is a well-known fact that cancer cells rewired their metabolism for cell growth and survival and metabolites closely depict the phenotypic properties of cells. Here, metabolomic study was performed in the PAT transformed and passage matched non-transformed cells using 1H HRMAS NMR. We have identified 12 significantly up-regulated metabolites, which, interestingly, were majorly amino acids, suggesting that PAT-induced pre-cancerous cells are involved in acquirement of nutrients for high protein turn-over. Furthermore, pathway analysis of metabolomics data indicated that aminoacyl tRNA biosynthesis, D-glutamate metabolism, glyoxylate and dicarboxylate metabolism and nitrogen metabolism were majorly hampered in PAT-induced pre-cancerous properties in normal intestinal cells.
Collapse
Affiliation(s)
- Neha Singh
- Food Toxicology Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow 226001, India
| | - Gaurav Sharma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Indra Dev
- Food Toxicology Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow 226001, India
| | - Sanjeev K Shukla
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kausar Mahmood Ansari
- Food Toxicology Laboratory, Food, Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow 226001, India
| |
Collapse
|
17
|
Mahato DK, Kamle M, Sharma B, Pandhi S, Devi S, Dhawan K, Selvakumar R, Mishra D, Kumar A, Arora S, Singh NA, Kumar P. Patulin in food: A mycotoxin concern for human health and its management strategies. Toxicon 2021; 198:12-23. [PMID: 33933519 DOI: 10.1016/j.toxicon.2021.04.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/30/2021] [Accepted: 04/27/2021] [Indexed: 01/09/2023]
Abstract
The mycotoxin patulin is primarily produced as a secondary metabolite by numerous fungal species and predominantly by Aspergillus, Byssochlamys, and Penicillium species. It is generally associated with fungal infected food materials. Penicillium expansum is considered the only fungal species liable for patulin contamination in pome fruits, especially in apples and apple-based products. This toxin in food poses serious health concerns and economic threat, which has aroused the need to adopt effective detection and mitigation strategies. Understanding its origin sources and biosynthetic mechanism stands essential for efficiently designing a management strategy against this fungal contamination. This review aims to present an updated outline of the sources of patulin occurrence in different foods and their biosynthetic mechanisms. It further provides information regarding the detrimental effects of patulin on human and agriculture as well as its effective detection, management, and control strategies.
Collapse
Affiliation(s)
- Dipendra Kumar Mahato
- CASS Food Research Centre, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, 3125, Australia.
| | - Madhu Kamle
- Applied Microbiology Lab., Department of Forestry, North Eastern Regional Institute of Science and Technology, Nirjuli, 791109, Arunachal Pradesh, India.
| | - Bharti Sharma
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Shikha Pandhi
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Sheetal Devi
- National Institute of Food Technology Entrepreneurship and Management (NIFTEM), Sonipat, Haryana, 131028, India.
| | - Kajal Dhawan
- Department of Food Technology and Nutrition, School of Agriculture Lovely Professional University, Phagwara, 144411, Punjab, India.
| | - Raman Selvakumar
- ICAR-Indian Agricultural Research Institute, Pusa Campus, New Delhi, 110012, India.
| | - Diwakar Mishra
- Department of Dairy Technology, Birsa Agricultural University, Dumka, 814145, Jharkhand, India.
| | - Arvind Kumar
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Shalini Arora
- Department of Dairy Technology, College of Dairy Science and Technology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, 125004, Haryana, India.
| | - Namita Ashish Singh
- Department of Microbiology, Mohanlal Sukhadia University, Udaipur, 313001, Rajasthan, India.
| | - Pradeep Kumar
- Applied Microbiology Lab., Department of Forestry, North Eastern Regional Institute of Science and Technology, Nirjuli, 791109, Arunachal Pradesh, India.
| |
Collapse
|
18
|
Sohrabi H, Arbabzadeh O, Khaaki P, Khataee A, Majidi MR, Orooji Y. Patulin and Trichothecene: characteristics, occurrence, toxic effects and detection capabilities via clinical, analytical and nanostructured electrochemical sensing/biosensing assays in foodstuffs. Crit Rev Food Sci Nutr 2021; 62:5540-5568. [PMID: 33624529 DOI: 10.1080/10408398.2021.1887077] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Patulin and Trichothecene as the main groups of mycotoxins in significant quantities can cause health risks from allergic reactions to death on both humans and animals. Accordingly, rapid and highly sensitive determination of these toxics agents is of great importance. This review starts with a comprehensive outlook regarding the characteristics, occurrence and toxic effects of Patulin and Trichothecene. In the following, numerous clinical and analytical approaches have been extensively discussed. The main emphasis of this review is placed on the utilization of novel nanomaterial based electrochemical sensing/biosensing tools for highly sensitive determination of Patulin and Trichothecene. Furthermore, a detailed and comprehensive comparison has been performed between clinical, analytical and sensing methods. Subsequently, the nanomaterial based electrochemical sensing platforms have been approved as reliable tools for on-site analysis of Patulin and Trichothecene in food processing and manufacturing industries. Different nanomaterials in improving the performance of detecting assays were investigated and have various benefits toward clinical and analytical methods. This paper would address the limitations in the current developments as well as the future challenges involved in the successful construction of sensing approaches with the functionalized nanomaterials and also allow exploring into core-research works regarding this area.
Collapse
Affiliation(s)
- Hessamaddin Sohrabi
- Department of Analytical Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Omid Arbabzadeh
- Faculty of Chemical and Petroleum Engineering, University of Tabriz, Tabriz, Iran
| | - Pegah Khaaki
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Alireza Khataee
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.,Рeoples' Friendship University of Russia (RUDN University), Moscow, Russian Federation
| | - Mir Reza Majidi
- Department of Analytical Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Yasin Orooji
- College of Materials Science and Engineering, Nanjing Forestry University, Nanjing, China
| |
Collapse
|
19
|
Chen Y, Xu C. The interaction partners of (pro)renin receptor in the distal nephron. FASEB J 2020; 34:14136-14149. [PMID: 32975331 DOI: 10.1096/fj.202001711r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 11/11/2022]
Abstract
The (pro)renin receptor (PRR), a key regulator of intrarenal renin-angiotensin system (RAS), is predominantly presented in podocytes, proximal tubules, distal convoluted tubules, and the apical membrane of collecting duct A-type intercalated cells, and plays a crucial role in hypertension, cardiovascular disease, kidney disease, and fluid homeostasis. In addition to its well-known renin-regulatory function, increasing evidence suggests PRR can also act in a variety of intracellular signaling cascades independently of RAS in the renal medulla, including Wnt/β-catenin signaling, cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2 ) signaling, and the apelinergic system, and work as a component of the vacuolar H+ -ATPase. PRR and these pathways regulate the expression/activity of each other that controlling blood pressure and renal functions. In this review, we highlight recent findings regarding the antagonistic interaction between PRR and ELABELA/apelin, the mutually stimulatory relationship between PRR and COX-2/PGE2 or Wnt/β-catenin signaling in the renal medulla, and their involvement in the regulation of intrarenal RAS thereby control blood pressure, renal injury, and urine concentrating ability in health and patho-physiological conditions. We also highlight the latest progress in the involvement of PRR for the vacuolar H+ -ATPase activity.
Collapse
Affiliation(s)
- Yanting Chen
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China.,Internal Medicine, Division of Nephrology and Hypertension, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Chuanming Xu
- Internal Medicine, Division of Nephrology and Hypertension, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, USA.,Center for Translational Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
20
|
Wei C, Yu L, Qiao N, Zhao J, Zhang H, Zhai Q, Tian F, Chen W. Progress in the distribution, toxicity, control, and detoxification of patulin: A review. Toxicon 2020; 184:83-93. [DOI: 10.1016/j.toxicon.2020.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/24/2020] [Accepted: 05/15/2020] [Indexed: 01/09/2023]
|
21
|
FitzSimons M, Beauchemin M, Smith AM, Stroh EG, Kelpsch DJ, Lamb MC, Tootle TL, Yin VP. Cardiac injury modulates critical components of prostaglandin E 2 signaling during zebrafish heart regeneration. Sci Rep 2020; 10:3095. [PMID: 32080283 PMCID: PMC7033201 DOI: 10.1038/s41598-020-59868-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 01/31/2020] [Indexed: 12/14/2022] Open
Abstract
The inability to effectively stimulate cardiomyocyte proliferation remains a principle barrier to regeneration in the adult human heart. A tightly regulated, acute inflammatory response mediated by a range of cell types is required to initiate regenerative processes. Prostaglandin E2 (PGE2), a potent lipid signaling molecule induced by inflammation, has been shown to promote regeneration and cell proliferation; however, the dynamics of PGE2 signaling in the context of heart regeneration remain underexplored. Here, we employ the regeneration-competent zebrafish to characterize components of the PGE2 signaling circuit following cardiac injury. In the regenerating adult heart, we documented an increase in PGE2 levels, concurrent with upregulation of cox2a and ptges, two genes critical for PGE2 synthesis. Furthermore, we identified the epicardium as the most prominent site for cox2a expression, thereby suggesting a role for this tissue as an inflammatory mediator. Injury also drove the opposing expression of PGE2 receptors, upregulating pro-restorative ptger2a and downregulating the opposing receptor ptger3. Importantly, treatment with pharmacological inhibitors of Cox2 activity suppressed both production of PGE2, and the proliferation of cardiomyocytes. These results suggest that injury-induced PGE2 signaling is key to stimulating cardiomyocyte proliferation during regeneration.
Collapse
Affiliation(s)
- MaryLynn FitzSimons
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME, 04609, US
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, US
| | - Megan Beauchemin
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME, 04609, US
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, US
- The University of New England, Biddeford, ME, 04005, US
| | - Ashley M Smith
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME, 04609, US
| | - Erika G Stroh
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME, 04609, US
| | - Daniel J Kelpsch
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, US
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, US
| | - Maureen C Lamb
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, US
| | - Tina L Tootle
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, US
| | - Viravuth P Yin
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME, 04609, US.
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, US.
| |
Collapse
|
22
|
Zheng L, Li XY, Huang FZ, Zhang XT, Tang HB, Li YS, Zhang WK, Li XJ, Tian GH. Effect of electroacupuncture on relieving central post-stroke pain by inhibiting autophagy in the hippocampus. Brain Res 2020; 1733:146680. [PMID: 31987731 DOI: 10.1016/j.brainres.2020.146680] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 01/01/2023]
Abstract
INTRODUCTION To explore the underlying mechanism of electroacupuncture (EA) treatment on central post-stroke pain (CPSP), and provide basic evidence for the EA treatment on CPSP. METHODS Firstly, 40 male SD rats were successfully established with a model of CPSP, under the intervention of different EA frequencies (2 Hz and 15 Hz) and fluoxetine (5 ml/kg and 0.4 mg/ml), whose brain tissue was then removed for paraffin-embedded sectioning; secondly, LPS induced the primary brain cells in the hippocampus to cause inflammation model which were added NS398 (inhibitor of COX-2) and DKK-1 (inhibitor of β-catenin) later. The lesion sites of brain tissue were observed by Nissl staining and Transmission Electron Microscope (TEM) and autophagy-related proteins (LC3B, p62, LAMP-1), COX-2 and β-catenin were detected by Western Blot and immunohistochemical staining. Finally, the correlation between LC3B, COX-2, and β-catenin was calculated by multispectral quantification. RESULTS (1) In the EA group (15 Hz), the number of Nissl bodies increased, autophagy-related protein LC3B-Ⅱ/Ⅰ, LAMP-1, COX-2, and β-catenin was lowly expressed, p62 was highly expressed; (2) COX-2, β-catenin and LC3B are positively correlated with each other (COX-2 & β-catenin: r = 0.923; COX-2 & LC3B: r = 0.818; β-catenin & LC3B: r = 0.801); (3) Nissl bodies of primary brain cells of the hippocampus under LPS were like animal experiments; after addition of DKK-1, high expression of β-catenin and COX-2 induced by LPS was significantly down-regulated, and LC3B-II/I was significantly down-regulated, and p62 protein only had up-regulation trend; after addition of NS398, COX-2 and LC3B-II/I was significantly down-regulated. CONCLUSION EA may inhibit autophagy in the hippocampus by reducing β-catenin/COX-2 protein expression and effectively alleviating CPSP. SIGNIFICANCE STATEMENT Previous studies have found that EA can reduce the expression of NK-1R in damaged rats by inhibition of COX-2 and β-catenin loops, which controls the activation of glial cells in the damaged area and the apoptosis of neuronal cells, and alleviated pain. In the male SD rat model, we evaluated this effect that EA inhibits autophagy in the hippocampus by reducing β-catenin/COX-2 protein expression in the brain tissue. In addition, we assessed expression levels of autophagy-related proteins and genes on the inflammatory primary brain cells model. From the experiment, we found EA may inhibit autophagy in the hippocampus by reducing β-catenin/COX-2 protein expression. These findings provide a foundation for the interpretation of the mechanism of EA on relieving CPSP in clinical practice.
Collapse
Affiliation(s)
- Ling Zheng
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-central University For Nationalities, Wuhan 430074, China
| | - Xin-Yi Li
- Key Laboratory of Chinese Internal Medicine of MOE and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Feng-Zhen Huang
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-central University For Nationalities, Wuhan 430074, China
| | - Xia-Tian Zhang
- Key Laboratory of Chinese Internal Medicine of MOE and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - He-Bin Tang
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-central University For Nationalities, Wuhan 430074, China.
| | - Yu-Sang Li
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-central University For Nationalities, Wuhan 430074, China
| | - Wei Kevin Zhang
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-central University For Nationalities, Wuhan 430074, China
| | - Xiao-Jun Li
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-central University For Nationalities, Wuhan 430074, China
| | - Gui-Hua Tian
- Key Laboratory of Chinese Internal Medicine of MOE and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
23
|
Connexin43 enhances Wnt and PGE2-dependent activation of β-catenin in osteoblasts. Pflugers Arch 2019; 471:1235-1243. [PMID: 31240382 DOI: 10.1007/s00424-019-02295-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/06/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022]
Abstract
Connexin43 is an important modulator of many signaling pathways in bone. β-Catenin, a key regulator of the osteoblast differentiation and function, is among the pathways downstream of connexin43-dependent intercellular communication. There are striking overlaps between the functions of these two proteins in bone cells. However, differential effects of connexin43 on β-catenin activity have been reported. Here, we examined how connexin43 influenced both Wnt-dependent and Wnt-independent activation of β-catenin in osteoblasts in vitro. Our data show that loss of connexin43 in primary osteoblasts or connexin43 overexpression in UMR106 cells regulated active β-catenin and phospho-Akt levels, with loss of connexin43 inhibiting and connexin43 overexpression increasing the levels of active β-catenin and phospho-Akt. Increasing connexin43 expression synergistically enhanced Wnt3a-dependent activation of β-catenin protein and β-catenin transcriptional activity, as well as Wnt-independent activation of β-catenin by prostaglandin E2 (PGE2). Finally, we show that the activation of β-catenin by PGE2 required signaling through the phosphatidylinositol 3-kinase (PI3K)/Akt/glycogen synthase kinase 3 beta (GSK3β) pathway, as the PI3K inhibitor, LY-294002, disrupted the synergy between connexin43 and PGE2. These data show that connexin43 regulates Akt and β-catenin activity and synergistically enhances both Wnt-dependent and Wnt-independent β-catenin signaling in osteoblasts.
Collapse
|