1
|
Ganjiani V, Meimandi-Parizi A, Ahmadi N, Sharifiyazdi H, Divar MR. Evaluation of effects of Tempol on testicular ischemia/reperfusion injury. Am J Emerg Med 2024; 82:107-116. [PMID: 38901331 DOI: 10.1016/j.ajem.2024.06.009] [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: 03/13/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024] Open
Abstract
AIM Tempol, a synthetic antioxidant compound, has received significant attention for its potential therapeutic applications in recent years, especially against ischemia/reperfusion (I/R) injury. The aim of the present research was to assess the protective effects of Tempol on testicular I/R injury caused by testicular torsion and detorsion (T/D) in rats. METHODS The subjects were divided into five groups: sham, testicular T/D, testicular T/D with Tempol treatment at 50 and 100 mg/kg, and healthy rats treated with Tempol at 100 mg/kg. Testicular torsion was induced by rotating the left testicles for 2 h, followed by detorsion for 24 h. Testicular tissues were evaluated for gene expression, oxidative stress markers, and histopathology, epididymal sperms were stained and analyzed, and blood serum samples were collected to measure the testosterone hormone. RESULTS The results showed that testicular I/R caused a significant decrease in sperm velocity parameters, viability, and count, as well as an increase in abnormal sperms (p < 0.05). However, treatment with Tempol significantly improved these parameters (p < 0.05). Histopathological analysis revealed severe damage to the testicular tissues, but treatment with Tempol improved the structural integrity of the seminiferous tubules. Testicular I/R also resulted in increased oxidative stress index and decreased testosterone levels significantly (p < 0.05), but Tempol administration mitigated these effects significantly (p < 0.05). Furthermore, the expression of Bax and Bcl2, genes associated with apoptosis, were significantly altered by testicular I/R (p < 0.05), but Tempol prevented these changes significantly (p < 0.05). CONCLUSION These findings provide strong evidence that Tempol can effectively prevent testicular I/R injury.
Collapse
Affiliation(s)
- Vahid Ganjiani
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | | | - Nasrollah Ahmadi
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Hassan Sharifiyazdi
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mohammad-Reza Divar
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
2
|
Alves MDR, Nascimento RDP, da Fonseca Machado AP, Dos Santos P, Aledo E, Morandi Vuolo M, Cavalheiro CO, Giaculi VO, Berilli P, Dos Santos NM, Marostica Junior MR. Hop ( Humulus lupulus L.) extract reverts glycaemic imbalance and cognitive impairment in an animal model of obesity. Food Funct 2024; 15:7669-7680. [PMID: 38961720 DOI: 10.1039/d4fo02062f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The rates of overweight and obesity around the world have increased in past years. The body's adipose tissue stimulates the antioxidant and oxidation imbalance capacity at the cellular level. This scenario favors an inflammatory low-grade systemic condition starting with insulin resistance, which in turn may involve diabetes mellitus type 2 and cognitive decline afterward. Neurological diseases have been correlated to senile age diseases over time. This scenario calls for a change in the incidence of obesity in the younger generation. An unhealthy dietary consumption together with sedentary habits might lead to poor gut absorption of nutrients. Several plants and foods have bioactive compounds that can reduce or inhibit radical scavengers, reactive oxygen species, and metal ion complexes that threaten the cerebral defense system. The bitter acids from hops (Humulus lupulus L.) have been demonstrated to have promising effects on lipid and carbohydrate metabolism improvement, reducing inflammatory responses through alpha acids, beta acids, and analogs action. Therefore, the current study aimed to investigate the bioactivity of hop bitter acids in obese and lean mice. For that, a dry hop extract (DHE) was obtained by applying carbon dioxide as the fluid of supercritical extraction. Afterward, seventy-eight male mice of the C57BL/6J strain were weighed and randomly distributed into six groups of 13 animals each according to the diet offered: (NO) normolipidic diet, (NO1) normolipidic diet containing 0.35% alpha acids, (NO2) normolipidic diet containing 3.5% alpha acids, (HP) hyperlipidic diet, (HP1) hyperlipidic diet containing 0.35% alpha acids, and (HP2) hyperlipidic diet containing 3.5% alpha acids. After applying the glycemic tolerance and insulin tolerance tests, a better stabilization of glycemia levels and weight gain among those animals fed with DHE (NO2 and HP2) were observed in comparison to the obese control group (HP) (p < 0.05). There was also an amelioration of antioxidant capacity observed by checking the enzymatic profile by SOD and an apparent mitigation of brain degeneration by checking GSK3β and p-IRS1 proteins expression (p < 0.05). The y-maze cognitive test applied to highlight possible obesity-harmful animal brains did not indicate a statistical difference between the groups. Although the weekly dietary intake between the obese HP2 group (33.32 ± 4.11, p < 0.05) and control HP (42.3 ± 5.88, p < 0.05) was different. The bioactive compounds present in DHE have demonstrated relevant effects on glycemic control, insulin signaling, and the consequent modulatory action of the obesity-related markers with the brain's inflammatory progression.
Collapse
Affiliation(s)
- Mariana da Rocha Alves
- Universidade Estadual de Campinas, School of Food Engineering, Department of Food Science and Nutrition, Laboratory of Nutrition and Metabolism, 80 Rua Monteiro Lobato, 13083-862 Campinas, São Paulo, Brazil.
| | - Roberto de Paula Nascimento
- Universidade Estadual de Campinas, School of Food Engineering, Department of Food Science and Nutrition, Laboratory of Nutrition and Metabolism, 80 Rua Monteiro Lobato, 13083-862 Campinas, São Paulo, Brazil.
| | - Ana Paula da Fonseca Machado
- Universidade Federal da Grande Dourados, Faculdade de Engenharia, Rod. Dourados-Itahum Km 12, C.P.: 79804-970 - Dourados, Mato Grosso do Sul, Brasil
| | - Philipe Dos Santos
- Rubian xtratos LTDA, Rua do Café, 375 Vila Valle, 13174-000, Sumaré, São Paulo, Brazil
| | - Eduardo Aledo
- Rubian xtratos LTDA, Rua do Café, 375 Vila Valle, 13174-000, Sumaré, São Paulo, Brazil
| | - Milena Morandi Vuolo
- Rubian xtratos LTDA, Rua do Café, 375 Vila Valle, 13174-000, Sumaré, São Paulo, Brazil
| | - Carolina Oliveira Cavalheiro
- Universidade Estadual de Campinas, School of Food Engineering, Department of Food Science and Nutrition, Laboratory of Nutrition and Metabolism, 80 Rua Monteiro Lobato, 13083-862 Campinas, São Paulo, Brazil.
| | - Vinícius Oliveira Giaculi
- Universidade Estadual de Campinas, School of Food Engineering, Department of Food Science and Nutrition, Laboratory of Nutrition and Metabolism, 80 Rua Monteiro Lobato, 13083-862 Campinas, São Paulo, Brazil.
| | - Patrícia Berilli
- Universidade Estadual de Campinas, School of Food Engineering, Department of Food Science and Nutrition, Laboratory of Nutrition and Metabolism, 80 Rua Monteiro Lobato, 13083-862 Campinas, São Paulo, Brazil.
| | - Nathália Medina Dos Santos
- Universidade Estadual de Campinas, School of Food Engineering, Department of Food Science and Nutrition, Laboratory of Nutrition and Metabolism, 80 Rua Monteiro Lobato, 13083-862 Campinas, São Paulo, Brazil.
| | - Mario Roberto Marostica Junior
- Universidade Estadual de Campinas, School of Food Engineering, Department of Food Science and Nutrition, Laboratory of Nutrition and Metabolism, 80 Rua Monteiro Lobato, 13083-862 Campinas, São Paulo, Brazil.
| |
Collapse
|
3
|
Alfa R, Considine T, Virani S, Pfeiffer M, Donato A, Dickerson D, Shuster D, Ellis J, Rushton K, Wei H, Gibson C. Clinical pharmacology and tolerability of REC-994, a redox-cycling nitroxide compound, in randomized phase 1 dose-finding studies. Pharmacol Res Perspect 2024; 12:e1200. [PMID: 38655895 PMCID: PMC11040693 DOI: 10.1002/prp2.1200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/28/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
Cerebral cavernous malformation (CCM) has variable clinical symptoms, including potentially fatal hemorrhagic stroke. Treatment options are very limited, presenting a large unmet need. REC-994 (also known as tempol), identified as a potential treatment through an unbiased drug discovery platform, is hypothesized to treat CCMs through a reduction in superoxide, a reactive oxygen species. We investigated the safety, tolerability, and pharmacokinetic profile of REC-994 in healthy volunteers. Single- and multiple-ascending dose (SAD and MAD, respectively) studies were conducted in adult volunteers (ages 18-55). SAD study participants received an oral dose of REC-994 or placebo. MAD study participants were randomized 3:1 to oral doses of REC-994 or matching placebo, once daily for 10 days. Thirty-two healthy volunteers participated in the SAD study and 52 in the MAD study. Systemic exposure increased in proportion to REC-994 dose after single doses of 50-800 mg and after 10 days of dosing over the 16-fold dose range of 50-800 mg. Median Tmax and mean t1/2 were independent of dose in both studies, and the solution formulation was more rapidly absorbed. REC-994 was well tolerated. Treatment-emergent adverse effects across both studies were mild and transient and resolved by the end of the study. REC-994 has a favorable safety profile and was well tolerated in single and multiple doses up to 800 mg with no dose-limiting adverse effects identified. Data support conducting a phase 2 clinical trial in patients with symptomatic CCM.
Collapse
Affiliation(s)
- Ron Alfa
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
- Present address:
NoetikSan FranciscoCaliforniaUSA
| | - Timothy Considine
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
- Present address:
Considine Comprehensive ConsultingSan DiegoCaliforniaUSA
| | | | - Matt Pfeiffer
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
| | - Anthony Donato
- Department of Internal MedicineUniversity of UtahSalt Lake CityUtahUSA
| | | | - Diana Shuster
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
- Present address:
CenExelSalt Lake CityUtahUSA
| | - Joel Ellis
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
- Present address:
Mariner BioPharmaSan JoseCaliforniaUSA
| | | | - Helen Wei
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
| | | |
Collapse
|
4
|
Shan M, Ma Q, Sun Y, Gao F, Cai S. The Protective Effect and Mechanism of a Phytochemical Extract from the Wild Vegetable Shutou ( Crateva unilocularis Buch.) against Acetaminophen-Induced Liver Injury in Mice. Foods 2023; 12:3109. [PMID: 37628108 PMCID: PMC10453156 DOI: 10.3390/foods12163109] [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: 06/23/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Acetaminophen (APAP) abuse is a common public health problem which can cause severe liver damage. However, strategies for dealing with this situation safely and effectively are very limited. The goal of the current work was to evaluate the protection and potential molecular mechanisms of an ethanol extract from shoots of the wild vegetable shutou (Crateva unilocularis Buch.) (ECS) against APAP-induced liver damage in mice. Mice orally received ECS for seven days (300 or 600 mg/kg b.w. per day) before being intraperitoneally injected with APAP (250 mg/kg). Results exhibited that ECS obviously decreased the content of alkaline phosphatase, alanine aminotransferase, aspartate transaminase, and malondialdehyde (p < 0.05). Catalase and superoxide dismutase were notably restored (p < 0.05), and the content of reduced glutathione was obviously increased (p < 0.05). Moreover, ECS significantly inhibited the secretion of interleukin-1β and tumor necrosis factor-α (p < 0.05). Further analyses of the mechanisms showed that ECS may alleviate oxidative stress in the liver by increasing the expression of the nuclear factor erythroid-2-related factor 2 and NADH quinone oxidoreductase 1 proteins, and may suppress liver inflammation by inhibiting the expression of the phosphorylated-inhibitor kappa B alpha/inhibitor kappa B alpha, phosphorylated-nuclear factor κB/nuclear factor κB, and cyclooxygenase-2 proteins. Meanwhile, ECS inhibited hepatocyte apoptosis by enhancing B-cell lymphoma gene 2 and suppressing Bcl-2-associated X protein. In summary, ECS may be used as a dietary supplement to prevent the liver damage caused by APAP abuse.
Collapse
Affiliation(s)
- Meimei Shan
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (M.S.); (Q.M.); (Y.S.)
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China
| | - Qian Ma
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (M.S.); (Q.M.); (Y.S.)
- Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yilin Sun
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (M.S.); (Q.M.); (Y.S.)
- Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Fengyi Gao
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China
| | - Shengbao Cai
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (M.S.); (Q.M.); (Y.S.)
| |
Collapse
|
5
|
Zhang H, Zheng L, Li C, Jing J, Li Z, Sun S, Xue T, Zhang K, Xue M, Cao C, Ouyang L, Qian Z, Xu R, He Z, Ma R, Chen L, Yao B. Effects of gut microbiota on omega-3-mediated ovary and metabolic benefits in polycystic ovary syndrome mice. J Ovarian Res 2023; 16:138. [PMID: 37443082 PMCID: PMC10347784 DOI: 10.1186/s13048-023-01227-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder that frequently exhibits low-grade inflammation, pro-oxidant activity, and gut dysbiosis. PCOS has become one of the leading causes of female infertility worldwide. Recently, omega-3 polyunsaturated fatty acids (PUFAs) have been proven to benefit metabolic disorders in PCOS patients. However, its roles in the regulation of metabolic and endocrinal balances in PCOS pathophysiology are not clear. In the present study, we aimed to explore how omega-3 PUFAs alleviate ovarian dysfunction and insulin resistance in mice with dehydroepiandrosterone (DHEA)-induced PCOS by modulating the gut microbiota. METHODS We induced PCOS in female mice by injecting them with DHEA and then treated them with omega-3 PUFAs. 16S ribosomal DNA (rDNA) amplicon sequencing, fecal microbiota transplantation (FMT) and antibiotic treatment were used to evaluate the role of microbiota in the regulation of ovarian functions and insulin resistance (IR) by omega-3 PUFAs. To further investigate the mechanism of gut microbiota on omega-3-mediated ovarian and metabolic protective effects, inflammatory and oxidative stress markers in ovaries and thermogenic markers in subcutaneous and brown adipose tissues were investigated. RESULTS We found that oral supplementation with omega-3 PUFAs ameliorates the PCOS phenotype. 16S rDNA analysis revealed that omega-3 PUFA treatment increased the abundance of beneficial bacteria in the gut, thereby alleviating DHEA-induced gut dysbiosis. Antibiotic treatment and FMT experiments further demonstrated that the mechanisms underlying omega-3 benefits likely involve direct effects on the ovary to inhibit inflammatory cytokines such as IL-1β, TNF-α and IL-18. In addition, the gut microbiota played a key role in the improvement of adipose tissue morphology and function by decreasing multilocular cells and thermogenic markers such as Ucp1, Pgc1a, Cited and Cox8b within the subcutaneous adipose tissues. CONCLUSION These findings indicate that omega-3 PUFAs ameliorate androgen-induced gut microbiota dysbiosis. The gut microbiota plays a key role in the regulation of omega-3-mediated IR protective effects in polycystic ovary syndrome mice. Moreover, omega-3 PUFA-regulated improvements in the ovarian dysfunction associated with PCOS likely involve direct effects on the ovary to inhibit inflammation. Our findings suggest that omega-3 supplementation may be a promising therapeutic approach for the treatment of PCOS by modulating gut microbiota and alleviating ovarian dysfunction and insulin resistance.
Collapse
Affiliation(s)
- Hong Zhang
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China
| | - Lu Zheng
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China
| | - Chuwei Li
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China
| | - Jun Jing
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China
| | - Zhou Li
- Center of Reproductive Medicine, Nanjing Jinling Hospital, School of Medicine, Jiangsu University, 212000, Zhenjiang, China
| | - Shanshan Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Jiangsu, 210023, Nanjing, China
| | - Tongmin Xue
- Reproductive Medical Center, Clinical Medical College (Northern Jiangsu People's Hospital), Yangzhou University, 98 Nantong West Road, Jiangsu, 225001, Yangzhou, China
| | - Kemei Zhang
- Reproductive Medical Center, Jinling Hospital Department, Nanjing Medical University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China
| | - Mengqi Xue
- Reproductive Medical Center, Jinling Hospital Department, Nanjing Medical University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China
| | - Chun Cao
- Department of Reproductive Medicine, Affiliated Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, 210002, Nanjing, China
| | - Lei Ouyang
- Department of Reproductive Medicine, Affiliated Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, 210002, Nanjing, China
| | - Zhang Qian
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China
| | - Rui Xu
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China
| | - Zhaowanyue He
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China
| | - Rujun Ma
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China.
| | - Li Chen
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China.
| | - Bing Yao
- Center of Reproductive Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China.
- Center of Reproductive Medicine, Nanjing Jinling Hospital, School of Medicine, Jiangsu University, 212000, Zhenjiang, China.
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wen Yuan Road, Jiangsu, 210023, Nanjing, China.
- Reproductive Medical Center, Jinling Hospital Department, Nanjing Medical University, 305 Zhongshan East Road, Jiangsu, 210002, Nanjing, China.
- Department of Reproductive Medicine, Affiliated Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, 210002, Nanjing, China.
| |
Collapse
|
6
|
Rossetto I, Santos F, Kido L, Lamas C, Montico F, Cagnon V. Tempol differential effect on prostate cancer inflammation: In vitro and in vivo evaluation. Prostate 2023; 83:403-415. [PMID: 36546327 DOI: 10.1002/pros.24473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 11/12/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Tempol is a redox-cycling nitroxide that acts directly on inflammation. However, few studies have reported the use of tempol in prostate cancer (PCa). The present study investigated the effects of tempol on inflammation related to NF-κB signaling, using hormone-dependent or hormone-independent cell lines and the transgenic adenocarcinoma of the mouse prostate PCa animal model in the early and late stages of cancer progression. METHODS PC-3 and LnCaP cells were exposed to different tempol doses in vitro, and cell viability assays were performed. The optimal treatment dose was chosen for subsequent analysis using western blotting. Five experimental groups were evaluated in vivo to test for tempol effects in the early (CT12 and TPL12 groups) and late stages (CT20, TPL20-I, and TLP20-II) of PCa development. The TPL groups were treated with 50 or 100 mg/kg tempol. All control groups received water as the vehicle. The ventral lobe of the prostate was collected and subjected to immunohistochemical and western blot analysis. RESULTS Tempol treatment reduced cellular proliferation in vitro and improved prostatic morphology in vivo, thereby decreasing tumor progression. Tempol reduced inflammation in preclinical models, and downregulated the initial inflammatory signaling through toll-like receptors, not always mediated by the MyD88 pathway. In addition, it upregulated iκB-α and iκB -β levels, leading to a decrease in NF-κB, TNF-α, and other inflammatory markers. Tempol also influenced cell survival markers. CONCLUSIONS Tempol can be considered a beneficial therapy for PCa treatment owing to its anti-inflammatory and antiproliferative effects. Nevertheless, the action of tempol was different depending on the degree of the prostatic lesion in vivo and hormone reliance in vitro. This indicates that tempol plays a multifaceted role in the prostatic tissue environment.
Collapse
Affiliation(s)
- Isabela Rossetto
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Felipe Santos
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Larissa Kido
- Department of Food and Nutrition, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Celina Lamas
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Fábio Montico
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Valéria Cagnon
- Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
7
|
Zhao J, Shi S, Zhang X, Liu Y, Yuan M, Cheng G, Wang Y. Confusoside, a dihydrochalcone glucoside, prevents acetaminophen-induced liver injury by modulating the Nrf2/NF-κB/caspase signaling pathway. Food Funct 2023; 14:2432-2443. [PMID: 36786681 DOI: 10.1039/d2fo03497b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Dihydrochalcones are important bioactive ingredients in plants. Anneslea fragrans is an edible and medicinal plant, and its leaves are rich in dihydrochalcones. Confusoside (CF) is the most abundant dihydrochalcone in A. fragrans leaves, which is traditionally used in the treatment of liver diseases. The aim of this study was to investigate the hepatoprotective effect of CF on acetaminophen (APAP)-induced hepatic injury in mice. CF could reduce the levels of AST, ALT, and LDH in the serum and enhance the antioxidant activity by activating the Nrf2 signaling pathway to increase the activities of antioxidant enzymes (SOD and CAT), and the GSH content but decrease the MDA accumulation in liver tissues. Immunofluorescence assay and western blotting analysis showed that CF can regulate Nrf2 into the cell nucleus, thereby promoting the expression of downstream antioxidant-related proteins, including NQO1 and HO-1. In addition, CF could inhibit the liver inflammatory response by suppressing the activation of the NF-κB signaling pathway to reduce the expressions of TNF-α, IL-1β, IL-6, and NO. Molecular docking results showed that there was good binding between the CF and Keap1-Nrf2 protein. Western blotting and TUNEL analysis also revealed CF-inhibited cell apoptosis-related protein expression (Bcl2 and caspase-3/9 proteins). Thus, the CF from A. fragrans leaves could be served as an alternative hepaprotective agent for the treatment and prevention of APAP-induced liver injury.
Collapse
Affiliation(s)
- Jinghao Zhao
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650500, Yunnan, China.
| | - Shang Shi
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650500, Yunnan, China.
| | - Xiaoyu Zhang
- The faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China.
| | - Yaping Liu
- The faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China.
| | - Minglong Yuan
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650500, Yunnan, China. .,School of Chemistry and Environment, National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, Yunnan Minzu University, Kunming 650500, Yunnan, China
| | - Guiguang Cheng
- The faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China.
| | - Yudan Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650500, Yunnan, China. .,School of Chemistry and Environment, National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, Yunnan Minzu University, Kunming 650500, Yunnan, China
| |
Collapse
|
8
|
Saeedi BJ, Hunter-Chang S, Luo L, Li K, Liu KH, Robinson BS. Oxidative stress mediates end-organ damage in a novel model of acetaminophen-toxicity in Drosophila. Sci Rep 2022; 12:19309. [PMID: 36369211 PMCID: PMC9652370 DOI: 10.1038/s41598-022-21156-w] [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: 02/04/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Acetaminophen is the most common cause of acute drug-induced liver injury in the United States. However, research into the mechanisms of acetaminophen toxicity and the development of novel therapeutics is hampered by the lack of robust, reproducible, and cost-effective model systems. Herein, we characterize a novel Drosophila-based model of acetaminophen toxicity. We demonstrate that acetaminophen treatment of Drosophila results in similar pathophysiologic alterations as those observed in mammalian systems, including a robust production of reactive oxygen species, depletion of glutathione, and dose-dependent mortality. Moreover, these effects are concentrated in the Drosophila fat body, an organ analogous to the mammalian liver. Utilizing this system, we interrogated the influence of environmental factors on acetaminophen toxicity which has proven difficult in vertebrate models due to cost and inter-individual variability. We find that both increasing age and microbial depletion sensitize Drosophila to acetaminophen toxicity. These environmental influences both alter oxidative stress response pathways in metazoans. Indeed, genetic and pharmacologic manipulations of the antioxidant response modify acetaminophen toxicity in our model. Taken together, these data demonstrate the feasibility of Drosophila for the study of acetaminophen toxicity, bringing with it an ease of genetic and microbiome manipulation, high-throughput screening, and availability of transgenic animals.
Collapse
Affiliation(s)
- Bejan J Saeedi
- Department of Pathology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Sarah Hunter-Chang
- Department of Pathology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Liping Luo
- Department of Pathology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Kaiyan Li
- Department of Pathology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Ken H Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Brian S Robinson
- Department of Pathology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA, 30322, USA.
| |
Collapse
|
9
|
Sahin B, Karabulut S, Filiz AK, Özkaraca M, Gezer A, Akpulat HA, Ataseven H. Galium aparine L. protects against acetaminophen-induced hepatotoxicity in rats. Chem Biol Interact 2022; 366:110119. [PMID: 36029804 DOI: 10.1016/j.cbi.2022.110119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/08/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022]
Abstract
The toxicity of acetaminophen (N-acetyl-para-aminophenol (APAP)) is the most frequent cause of drug-induced liver damage. Galium aparine L. (GA) is traditionally used to treat jaundice. We aimed to investigate the hepatoprotective potential of GA in the APAP-induced hepatic encephalopathy (HE) rat model. Qualitative phytochemical characterization of GA was performed by LC/Q-TOF/MS analysis. Wistar rats were pretreated with GA (250 and 500 mg/kg b.wt. per oral) for five days. On the 6th day, the rats were exposed to APAP (1500 mg/kg b.wt. oral gavage) and behavioral tests (open field and passive avoidance tests) were applied on the 7th and 8th days. The animals were killed, and biochemical and histopathological parameters were assessed in blood and hepatic specimens. GA pretreated rats exhibited a significant reduction in APAP-induced liver damage, evidenced by the reduction in liver necrosis and alanine aminotransferase (ALT), aspartate aminotransferase (AST), and bilirubin (BIL). GA demonstrated an anxiolytic effect, as seen in the acquisition trial and grooming behavior. The short-term memory performances of animals were not changed in all groups, suggesting that APAP intoxication did not affect hippocampal function. These results show that GA extract markedly exerts hepatoprotective activity, while its effect on hepatic encephalopathy was limited.
Collapse
Affiliation(s)
- Bilal Sahin
- Department of Medical Physiology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Sebahattin Karabulut
- Department of Medical Services and Techniques, Vocational School of Health Services, Sivas Cumhuriyet University, Sivas, Turkey.
| | - Ahmet Kemal Filiz
- Department of Medical Physiology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Mustafa Özkaraca
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Arzu Gezer
- Department of Health Care Services, Vocational School of Health Services, Atatürk University, Erzurum, Turkey
| | | | - Hilmi Ataseven
- Department of Internal Medicine, Discipline of Gastroenterology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
10
|
Liao Y, Li J, Li S, Han B, Wu P, Deng N, Guo X, Lv Z, Zhang Z. Inorganic mercury induces liver oxidative stress injury in quails by inhibiting Akt/Nrf2 signal pathway. INORG CHEM COMMUN 2022. [DOI: 10.1016/j.inoche.2022.109603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
11
|
Virgana R, Atik N, Gunadi JW, Jonathan E, Ramadhani DE, Soetadji RS, Goenawan H, Lesmana R, Kartasasmita A. MitoTEMPOL Inhibits ROS-Induced Retinal Vascularization Pattern by Modulating Autophagy and Apoptosis in Rat-Injected Streptozotocin Model. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071061. [PMID: 35888150 PMCID: PMC9320075 DOI: 10.3390/life12071061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 12/03/2022]
Abstract
Diabetic retinopathy leads to retinal malfunction, blindness, and reduced quality of life in adult diabetes patients. The involvement of reactive oxygen species (ROS) regulation stimulated by high blood glucose levels opens the opportunity for ROS modulator agents such as MitoTEMPOL. This study aims to explore the effect of MitoTEMPOL on ROS balance that may be correlated with retinal vascularization pattern, autophagy, and apoptosis in a streptozotocin-induced rat model. Four groups of male Wistar rats (i.e., control, TEMPOL (100 mg/kg body weight [BW]), diabetic (streptozotocin, 50 mg/kg BW single dose), and diabetic + TEMPOL; n = 5 for each group) were used in the study. MitoTEMPOL was given for 5 weeks, followed by funduscopy, and gene and protein expression were explored from the rat’s retina. Streptozotocin injection decreased bodyweight and increased food and water intake, as well as fasting blood glucose. The results showed that MitoTEMPOL reduced retinal vascularization pattern and decreased superoxide dismutase gene expression and protein carbonyl, caspase 3, and caspase 9 protein levels. A modulation of autophagy in diabetes that was reversed in the diabetic + TEMPOL group was found. In conclusion, MitoTEMPOL modulation on autophagy and apoptosis contributes to its role as a potent antioxidant to prevent diabetic retinopathy by inhibiting ROS-induced retinal vascularization patterns.
Collapse
Affiliation(s)
- Rova Virgana
- Department of Ophthalmology, Faculty of Medicine, Universitas Padjadjaran, Professor Eyckman 38, Bandung 40161, Indonesia;
- Cicendo National Eye Hospital, Cicendo 4, Bandung 40117, Indonesia
- Correspondence:
| | - Nur Atik
- Biology Cell Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Raya Bandung-Sumedang Km 21, Bandung 45363, Indonesia;
| | - Julia Windi Gunadi
- Department of Physiology, Faculty of Medicine, Maranatha Christian University, Surya Sumantri 65, Bandung 40164, Indonesia;
| | - Evelyn Jonathan
- Faculty of Medicine, Maranatha Christian University, Surya Sumantri 65, Bandung 40164, Indonesia; (E.J.); (D.E.R.); (R.S.S.)
| | - Dona Erisa Ramadhani
- Faculty of Medicine, Maranatha Christian University, Surya Sumantri 65, Bandung 40164, Indonesia; (E.J.); (D.E.R.); (R.S.S.)
| | - Ray Sebastian Soetadji
- Faculty of Medicine, Maranatha Christian University, Surya Sumantri 65, Bandung 40164, Indonesia; (E.J.); (D.E.R.); (R.S.S.)
| | - Hanna Goenawan
- Physiology Cell Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Raya Bandung-Sumedang Km 21, Bandung 45363, Indonesia; (H.G.); (R.L.)
- Physiology Molecular Laboratory, Biological Activity Division, Central Laboratory, Universitas Padjadjaran, Raya Bandung-Sumedang Km 21, Bandung 45363, Indonesia
| | - Ronny Lesmana
- Physiology Cell Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Raya Bandung-Sumedang Km 21, Bandung 45363, Indonesia; (H.G.); (R.L.)
- Physiology Molecular Laboratory, Biological Activity Division, Central Laboratory, Universitas Padjadjaran, Raya Bandung-Sumedang Km 21, Bandung 45363, Indonesia
| | - Arief Kartasasmita
- Department of Ophthalmology, Faculty of Medicine, Universitas Padjadjaran, Professor Eyckman 38, Bandung 40161, Indonesia;
- Cicendo National Eye Hospital, Cicendo 4, Bandung 40117, Indonesia
| |
Collapse
|
12
|
Wang YP, Wang YD, Liu YP, Cao JX, Yang ML, Wang YF, Khan A, Zhao TR, Cheng GG. 6'- O-Caffeoylarbutin from Que Zui tea ameliorates acetaminophen-induced liver injury via enhancing antioxidant ability and regulating the PI3K signaling pathway. Food Funct 2022; 13:5299-5316. [PMID: 35441652 DOI: 10.1039/d2fo00507g] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Que Zui tea (QT), a traditional herbal tea in China, has a significant hepatoprotective effect. 6'-O-Caffeoylarbutin (CA) is the most abundant chemical compound in the QT. However, the hepatoprotective effect of CA has not been investigated. This study is aimed to evaluate the protective effect of CA on acetaminophen (APAP) induced hepatotoxicity in vivo and in vitro and its possible underlying mechanism. In APAP-induced HepG-2 cells, CA inhibited intracellular ROS accumulation and cell apoptosis, and improved the expression of antioxidants including SOD, CAT and GSH. In APAP-administrated mice, CA pretreatment remarkably ameliorated the histopathological damage and inflammatory response, and antioxidant enzyme activity in the serum and liver tissues. Moreover, the immunohistochemistry and immunofluorescence assay results revealed that the CA markedly reduced ROS production and apoptosis, and activated antioxidant transcription factor Nrf2 in the liver. Meanwhile, molecular docking results showed that the strong binding force of CA and PI3K was due to the higher number of hydrogen- and π-bonds with active site residues. Notably, CA pretreatment significantly regulated the expression of PI3K, Akt, Nrf2, NQO1, HO-1, Bcl-2, Bax, caspase-3, and caspase-9 proteins in APAP-treated liver tissues. These data demonstrated that CA had a protective effect against APAP-induced hepatotoxicity via regulating the PI3K/Akt and Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yong-Peng Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Yu-Dan Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China. .,National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, Yunnan Minzu University, Kunming, 650500, China
| | - Ya-Ping Liu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Jian-Xin Cao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Mei-Lian Yang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Yi-Fen Wang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Afsar Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | - Tian-Rui Zhao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Gui-Guang Cheng
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
13
|
Ding Z, Li Y, Tang Z, Song X, Jing F, Wu H, Lu B. Role of gambogenic acid in regulating PI3K/Akt/NF-kβ signaling pathways in rat model of acute hepatotoxicity. Biosci Biotechnol Biochem 2021; 85:520-527. [PMID: 33624779 DOI: 10.1093/bbb/zbaa039] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022]
Abstract
The purpose of this study is to investigate the protective effect of gambogenic acid (GA) in acetaminophen (APAP)-induced hepatotoxicity in rat models. GA (10 mg/kg) was administered intraperitoneal (i.p.) to rats for 7 consecutive days followed by APAP (500 mg/kg) single dose (i.p.) on the final day after GA administration. The levels of MDA, GSH, SOD, CAT, GPx, GST, ALP, AST, ALT, proinflammatory cytokines (TNF-α, IL-1β, IL-6), apoptosis markers (caspase-3 and -9, Bax, Bcl-2), 4-hydroxynonenal (4-HNE), and prostaglandin E2 (PGE2) were evaluated. Results exhibited protective effects of GA by inhibiting inflammation, preventing oxidative stress and apoptosis in APAP-induced liver. Histopathological changes caused by APAP were attenuated, protein expressions of phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) were upregulated, and nuclear factor-kappa β (NF-kβ) was downregulated by GA. In summary, GA significantly exerted anti-inflammatory and antiapoptotic effects against APAP-induced hepatotoxicity potentially through regulation of PI3K/Akt and NF-kβ signaling pathways.
Collapse
Affiliation(s)
- Zhongyang Ding
- Department of General Surgery, Wuxi Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Wuxi, Jiangsu, China
| | - Ying Li
- Department of Emergency, First Teaching Hospital of Tianjin University of TCM, Tianjin, China
| | - Zhangfeng Tang
- Department of General Surgery, Wuxi Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Wuxi, Jiangsu, China
| | - Xiaoyi Song
- Department of General Surgery, Wuxi Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Wuxi, Jiangsu, China
| | - Fa Jing
- Department of General Surgery, Wuxi Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Wuxi, Jiangsu, China
| | - Haotian Wu
- Department of General Surgery, Wuxi Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Wuxi, Jiangsu, China
| | - Bei Lu
- Department of Hepato-pancreato-biliary Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Rosa AC, Corsi D, Cavi N, Bruni N, Dosio F. Superoxide Dismutase Administration: A Review of Proposed Human Uses. Molecules 2021; 26:1844. [PMID: 33805942 PMCID: PMC8037464 DOI: 10.3390/molecules26071844] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Superoxide dismutases (SODs) are metalloenzymes that play a major role in antioxidant defense against oxidative stress in the body. SOD supplementation may therefore trigger the endogenous antioxidant machinery for the neutralization of free-radical excess and be used in a variety of pathological settings. This paper aimed to provide an extensive review of the possible uses of SODs in a range of pathological settings, as well as describe the current pitfalls and the delivery strategies that are in development to solve bioavailability issues. We carried out a PubMed query, using the keywords "SOD", "SOD mimetics", "SOD supplementation", which included papers published in the English language, between 2012 and 2020, on the potential therapeutic applications of SODs, including detoxification strategies. As highlighted in this paper, it can be argued that the generic antioxidant effects of SODs are beneficial under all tested conditions, from ocular and cardiovascular diseases to neurodegenerative disorders and metabolic diseases, including diabetes and its complications and obesity. However, it must be underlined that clinical evidence for its efficacy is limited and consequently, this efficacy is currently far from being demonstrated.
Collapse
Affiliation(s)
- Arianna Carolina Rosa
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| | - Daniele Corsi
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| | - Niccolò Cavi
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| | - Natascia Bruni
- Istituto Farmaceutico Candioli, Strada Comunale di None, 1, 10092 Beinasco, Italy;
| | - Franco Dosio
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 9, 10125 Turin, Italy; (D.C.); (N.C.); (F.D.)
| |
Collapse
|
15
|
Naim S, Fernandez-Marrero Y, de Brot S, Bachmann D, Kaufmann T. Loss of BOK Has a Minor Impact on Acetaminophen Overdose-Induced Liver Damage in Mice. Int J Mol Sci 2021; 22:ijms22063281. [PMID: 33807047 PMCID: PMC8004760 DOI: 10.3390/ijms22063281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/14/2021] [Accepted: 03/20/2021] [Indexed: 12/27/2022] Open
Abstract
Acetaminophen (APAP) is one of the most commonly used analgesic and anti-pyretic drugs, and APAP intoxication is one of the main reasons for liver transplantation following liver failure in the Western world. While APAP poisoning ultimately leads to liver necrosis, various programmed cell death modalities have been implicated, including ER stress-triggered apoptosis. The BCL-2 family member BOK (BCL-2-related ovarian killer) has been described to modulate the unfolded protein response and to promote chemical-induced liver injury. We therefore investigated the impact of the loss of BOK following APAP overdosing in mice. Surprisingly, we observed sex-dependent differences in the activation of the unfolded protein response (UPR) in both wildtype (WT) and Bok-/- mice, with increased activation of JNK in females compared with males. Loss of BOK led to a decrease in JNK activation and a reduced percentage of centrilobular necrosis in both sexes after APAP treatment; however, this protection was more pronounced in Bok-/- females. Nevertheless, serum ALT and AST levels of Bok-/- and WT mice were comparable, indicating that there was no major difference in the overall outcome of liver injury. We conclude that after APAP overdosing, loss of BOK affects initiating signaling steps linked to ER stress, but has a more minor impact on the outcome of liver necrosis. Furthermore, we observed sex-dependent differences that might be worthwhile to investigate.
Collapse
Affiliation(s)
- Samara Naim
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010 Bern, Switzerland; (S.N.); (Y.F.-M.); (D.B.)
| | - Yuniel Fernandez-Marrero
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010 Bern, Switzerland; (S.N.); (Y.F.-M.); (D.B.)
- Sunnybrook Health Sciences Centre, University of Toronto, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Simone de Brot
- COMPATH, Institute of Animal Pathology, University of Bern, Laenggassstrasse 122, CH-3012 Bern, Switzerland;
| | - Daniel Bachmann
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010 Bern, Switzerland; (S.N.); (Y.F.-M.); (D.B.)
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010 Bern, Switzerland; (S.N.); (Y.F.-M.); (D.B.)
- Correspondence:
| |
Collapse
|
16
|
Li T, Zhang T, Gao H, Liu R, Gu M, Yang Y, Cui T, Lu Z, Yin C. Tempol ameliorates polycystic ovary syndrome through attenuating intestinal oxidative stress and modulating of gut microbiota composition-serum metabolites interaction. Redox Biol 2021; 41:101886. [PMID: 33592539 PMCID: PMC7896192 DOI: 10.1016/j.redox.2021.101886] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/08/2020] [Accepted: 01/28/2021] [Indexed: 12/22/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex endocrine and metabolic disorder, which is often accompanied by oxidative stress. Tempol, a superoxide dismutase mimetic, protects against several diseases caused by oxidative stress. However, the effect of tempol on PCOS has not been investigated. The present study demonstrated the alleviation of ovarian dysfunction and glucose tolerance in dehydroepiandrosterone (DHEA)-induced PCOS rats treated with tempol. Tempol significantly reduced the intestinal oxidative stress in PCOS rats without affecting the ovarian redox rate. The 16S rDNA sequencing of the intestinal microbiome and non-targeted metabolomics analysis indicated significant differences in gut microbiota composition and serum metabolite profiles between the control and PCOS rats, and most of these differences were reduced after tempol intervention. Tempol alters the gut microbiome by increasing the abundance of genus Ruminococcus_1 and by decreasing the abundance of Ruminococcus_2, Staphylococcus, Ideonella, and Corynebnacterium genera. Tempol also attenuates the reduction of serum bile acid and stachyose levels in PCOS rats, and the serum stachyose level was significantly correlated with the abundance of 15 genera, particularly Ruminococcus_1 and Ruminococcus_2. Moreover, stachyose administration improved ovarian dysfunction in PCOS rats. Thus, our data indicate that tempol ameliorates PCOS phenotype by reducing intestinal oxidative stress, restoring gut dysbiosis, and modulating the interaction between gut microbiota and host metabolite. Therefore, tempol intervention is a potential therapeutic approach for PCOS. Tempol improved ovarian dysfunction and glucose tolerance in polycystic ovary syndrome rats. Tempol ameliorates intestinal oxidative stress and gut microbiota dysbiosis. The protective effect of tempol is associated alternations in serum bile acid and stachyose levels. Stachyose administration improved ovarian dysfunction in polycystic ovary syndrome rats.
Collapse
Affiliation(s)
- Tianhe Li
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Tingting Zhang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Huimin Gao
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Ruixia Liu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Muqing Gu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Yuxi Yang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Tianyu Cui
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Zhongbing Lu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Chenghong Yin
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| |
Collapse
|
17
|
Ma H, Ren H, Wang J, Yuan X, Wu X, Shi X. Targeting PI3K/Akt/Nrf2 pathway by glabridin alleviates acetaminophen-induced hepatic injury in rats. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2020.102968] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
18
|
Xu Q, Xu J, Zhang K, Zhong M, Cao H, Wei R, Jin L, Gao Y. Study on the protective effect and mechanism of Dicliptera chinensis (L.) Juss (Acanthaceae) polysaccharide on immune liver injury induced by LPS. Biomed Pharmacother 2020; 134:111159. [PMID: 33370627 DOI: 10.1016/j.biopha.2020.111159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
The purpose of this study is to use Dicliptera chinensis (L.) Juss (Acanthaceae) polysaccharide (DCP) to act on the NF-κB inflammatory pathway and Fas/FasL ligand system, in order to find a new method to improve immune liver injury. Lipopolysaccharide (LPS) was used to establish an injury model in vivo (Kunming mice) and in vitro (LO2 cells). In this experiment, hematoxylin-eosin (H&E) staining and related biochemical indicators were used to observe the pathological changes of liver tissues, oxidative stress and inflammatory reactions. Immunohistochemistry, ELISA, RT-PCR and Western blot were used to detect protein or mRNA expressions associated with inflammation response and apoptosis. The experimental results show that the model group has obvious liver cell damage and inflammatory infiltration. After DCP intervention, it could significantly reduce the levels of ALT, AST, ALP, TBIL and MDA in serum, and increase the content of SOD and GSH-Px. In addition, DCP can reduce the expression level of NF-κB in the liver and reduce the release of downstream inflammatory factors TNF-α, IL-6 and IL-1β, thereby reducing the inflammation. At the same time, DCP can significantly inhibit the expression of Fas/FasL ligand system and apoptosis related-proteins and mRNA, which in turn can reduce cell apoptosis. In conclusion, DCP can alleviate liver injury by inhibiting liver inflammation and apoptosis, which provides a new strategy for clinical treatment of immune liver injury.
Collapse
Affiliation(s)
- Qiongmei Xu
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China
| | - Jie Xu
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China
| | - Kefeng Zhang
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China
| | - Mingli Zhong
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China
| | - Houkang Cao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Riming Wei
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China
| | - Ling Jin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China.
| | - Ya Gao
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China; College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
19
|
Koyuncuoğlu T, Yıldırım A, Dertsiz EK, Yüksel M, Ercan F, Yeğen BÇ. Estrogen receptor agonists protect against acetaminophen-induced hepatorenal toxicity in rats. Life Sci 2020; 263:118561. [PMID: 33045213 DOI: 10.1016/j.lfs.2020.118561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/16/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023]
Abstract
AIMS Acetaminophen-induced hepatorenal toxicity varies among sexes with controversial results among species. The aim was to compare the impact of sex and ovarian hormones on hepatorenal toxicity and to elucidate protective effects of estrogen and estrogen receptor (ER) agonists. MAIN METHODS Under anesthesia, female rats underwent ovariectomy (OVX) or sham-OVX. Starting at postsurgical 40th day, OVX-rats received subcutaneously (each, 1 mg/kg/day) 17β-estradiol (E2), ERβ-agonist (DPN) or ERα-agonist (PPT) for 10 days, while male and sham-OVX rats received vehicle for 10 days. Then, rats received either acetaminophen (3 g/kg) or saline by orogastric gavage and were decapitated at 24th h. Blood samples were obtained to measure serum ALT, AST, BUN, creatinine levels. Liver and kidney samples were obtained for histopathologic examination and for analyzing levels of luminol- and lucigenin-chemiluminescence, glutathione and myeloperoxidase activity. KEY FINDINGS Compared to their control groups, levels of AST, ALT, BUN, creatinine, hepatic and renal myeloperoxidase activity and chemiluminescence levels were increased, and hepatic glutathione level was decreased in acetaminophen-administered male groups, while ALT and hepatic chemiluminescence levels were not elevated in sham-OVX-rats. Both ER-agonists and E2 reduced BUN, creatinine and reversed all oxidative parameters in renal tissues of OVX-rats. Additionally, ERα-agonist reversed all hepatic injury parameters, while ERβ-agonist elevated hepatic glutathione level. SIGNIFICANCE Acetaminophen toxicity in female rats presented with a more preserved hepatic function, while renal toxicity was not influenced by sex or by the lack of ovarian hormones. Pretreatment with estrogen or ER agonists, via their antioxidant actions, provided protective effects on acetaminophen-induced hepatorenal toxicity.
Collapse
Affiliation(s)
- Türkan Koyuncuoğlu
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Alper Yıldırım
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Ekin K Dertsiz
- Department of Histology & Embryology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Meral Yüksel
- Department of Medical Laboratory, Vocational School of Health-Related Professions, Marmara University, Istanbul, Turkey
| | - Feriha Ercan
- Department of Histology & Embryology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Berrak Ç Yeğen
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey.
| |
Collapse
|
20
|
Galani BRT, Owona BA, Chuisseu DPD, Machewere E, Ngantchouko CBN, Moundipa PF. Hepatoprotective Activity of Leptadenia hastata ( Asclepiadaceae) on Acetaminophen-Induced Toxicity in Mice: In Vivo Study and Characterization of Bioactive Compounds through Molecular Docking Approaches. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3807234. [PMID: 32953880 PMCID: PMC7482022 DOI: 10.1155/2020/3807234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/18/2020] [Accepted: 08/07/2020] [Indexed: 02/05/2023]
Abstract
MATERIALS AND METHODS Various aqueous extracts were prepared from this plant and preadministered per os to albino mice 3 h before APAP administration, once daily for one week. Animals from the normal group were given only distilled water while those from negative control received only APAP 250 mg/kg. After treatment, mice were sacrificed, the liver was collected for histopathology analysis, and different biochemical markers (alanine aminotransferase (ALT), superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), malondialdehyde (MDA), and tumor necrosis factor-alpha (TNFα)) were measured. The content of the active extract was analyzed by HPLC/UV. Molecular docking was conducted using iGEMDOCK software, and the drug-likeness and pharmacokinetic profiles were evaluated using Swiss ADME. RESULTS APAP administration significantly increased (p < 0.001) ALT in liver homogenates when compared to normal controls whereas the stem decoction at 250 mg/kg significantly (p < 0.001) reduced this activity to a normal value comparable to silymarin 50 mg/kg which is better than leaf and root extracts. Moreover, the stem decoction also significantly reduced the MDA levels (p < 0.05) and increased those of GSH, SOD, and CAT (p < 0.001) at doses of 250 and 500 mg/kg compared to the negative control. A significant (p < 0.001) decrease of TNFα levels and leukocyte infiltration was observed following treatment with this extract. The HPLC/UV analysis of the decoction revealed the presence of dihydroxycoumarin, quinine, and scopoletin with the following retention times: 2.6, 5.1, and 7.01 min, respectively. In silico studies showed that quinine and dihydroxycoumarin had great potentials to be orally administered drugs and possessed strong binding affinities with TNFα, TNF receptor, cyclooxygenase-2, iNOS, cytochrome P450 2E1, and GSH reductase. CONCLUSION Based on these results, L. hastata could be considered a source of promising hepatoprotective compounds with antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Borris R. T. Galani
- Laboratory of Applied Biochemistry, Department of Biological Sciences, Faculty of Science, University of Ngaoundere, P.O. Box 454 Ngaoundere, Cameroon
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty of Science, University of Yaounde I, P.O. Box 812 Yaounde, Cameroon
| | - Brice A. Owona
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty of Science, University of Yaounde I, P.O. Box 812 Yaounde, Cameroon
| | - Dieudonné P. D. Chuisseu
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty of Science, University of Yaounde I, P.O. Box 812 Yaounde, Cameroon
- Department of Medicine, Medical and Biomedical Sciences, Higher Institute of Health Sciences, Université des Montagnes, P.O. Box 208 Bangangte, Cameroon
| | - Esaïe Machewere
- Laboratory of Applied Biochemistry, Department of Biological Sciences, Faculty of Science, University of Ngaoundere, P.O. Box 454 Ngaoundere, Cameroon
| | - Claude B. N. Ngantchouko
- Department of Pharmacy, Higher Institute of Health Sciences, Université des Montagnes, P.O. Box 208 Bangangte, Cameroon
| | - Paul F. Moundipa
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty of Science, University of Yaounde I, P.O. Box 812 Yaounde, Cameroon
| |
Collapse
|
21
|
Jaeschke H, Ramachandran A. Acetaminophen-induced apoptosis: Facts versus fiction. J Clin Transl Res 2020; 6:36-47. [PMID: 33426354 PMCID: PMC7787220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 12/15/2022] Open
Abstract
An overdose of the widely used analgesic acetaminophen (APAP) is the most common cause of acute liver failure in the western world and hence is a clinically significant problem. Thus, mechanisms of APAP-induced hepatotoxicity have been the focus of extensive investigation for decades and it was established that APAP induces hepatocyte cell death by necrosis. Although APAP-induced necrosis shares some features of apoptosis induced by the intrinsic pathway, apoptotic cell death in this context was ruled out due to the absence of caspase activation and lack of protection by caspase inhibitors and missing morphological characteristics of apoptotic cells. Deeper mechanistic understanding of the cell death process after APAP in recent years has now revealed that cells die by programmed necrosis and apoptosis is not a relevant mode of cell death in this context. Hence, it is alarming to note that an increasing number of studies are being published purporting to indicate that APAP induces apoptotic cell death. These papers broadly measure "apoptotic markers" with questionable specificity such as Bax, Bcl-2 and caspase-3 protein expression, or use the terminal deoxynucleotidyl transferase dUTP nick end labeling assay as basis for the conclusion that there is apoptosis after APAP overdose. The misguided use of these apoptosis parameters in correlative studies without context or scientific rationale confuses the field and threatens to undo decades of careful mechanistic investigation into this topic. This review examines this emerging problem in detail and recommends approaches to correct it. RELEVANCE FOR PATIENTS Hepatotoxicity and acute liver failure caused by an acetaminophen overdose is a serious clinical problem in western countries. Understanding the mode of cell death and the signaling pathways involved is critical for developing new therapeutic approaches. Recent trends to claim that apoptosis is a relevant mode of cell death in acetaminophen hepatotoxicity are not justified by sound scientific data and will not lead to effective new drug development.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA,
Corresponding author: Hartmut Jaeschke Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA. Tel. +1 913 588 7969
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
22
|
Xin R, Chen Z, Fu J, Shen F, Zhu Q, Huang F. Xanomeline Protects Cortical Cells From Oxygen-Glucose Deprivation via Inhibiting Oxidative Stress and Apoptosis. Front Physiol 2020; 11:656. [PMID: 32595528 PMCID: PMC7303960 DOI: 10.3389/fphys.2020.00656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Xanomeline, a muscarinic acetylcholine receptor agonist, is one of the first compounds that was found to be effective in the treatment of schizophrenics and attenuating behavioral disturbances of patients with Alzheimer's disease (AD). However, its role in ischemia-induced injury due to oxygen and glucose deprivation (OGD) remains unclear. Primary rat neuronal cells were exposed to OGD and treated with xanomeline. The effects of xanomeline on apoptosis, cell viability, lactate dehydrogenase (LDH) levels, and reactive oxygen species (ROS) were determined using an Annexin V Apoptosis Detection Kit, a non-radioactive cell counting kit-8 (CCK-8) assay, colorimetric LDH cytotoxicity assay kit, and a dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay, respectively, and the expressions of Sirtuin 1, haem oxygenase-1 (HO-1), B-cell lymphoma 2 (Bcl-2), poly ADP-ribose polymerase (PARP), and hypoxia-inducible factor α (HIF-1α) as well as the level of phosphorylated kinase B (p-Akt) were determined by Western blotting. Compared with the control, xanomeline pretreatment increased the viability of isolated cortical neurons and decreased the LDH release induced by OGD. Compared with OGD-treated cells, xanomeline inhibited apoptosis, reduced ROS production, attenuated the OGD-induced HIF-1α increase and partially reversed the reduction of HO-1, Sirtuin-1, Bcl-2, PARP, and p-Akt induced by OGD. In conclusion, xanomeline treatment protects cortical neuronal cells possibly through the inhibition of apoptosis after OGD.
Collapse
Affiliation(s)
- Rujuan Xin
- Department of Pharmacy, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zhongjian Chen
- Department of Pharmacy, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin Fu
- Department of Pharmacy, Ninghai First Hospital, Zhejiang, China
| | - Fuming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Quangang Zhu
- Department of Pharmacy, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fang Huang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
23
|
Zhang K, Xu Q, Gao Y, Cao H, Lian Y, Li Z, Xu J, Zhong M, Li J, Wei R, Dong J, Jin L. Polysaccharides from Dicliptera chinensis ameliorate liver disturbance by regulating TLR-4/NF-κB and AMPK/Nrf2 signalling pathways. J Cell Mol Med 2020; 24:6397-6409. [PMID: 32337831 PMCID: PMC7294158 DOI: 10.1111/jcmm.15286] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022] Open
Abstract
The purpose of this study was to alleviate liver disturbance by applying polysaccharides from Dicliptera chinensis (DCP) to act on the adenosine monophosphate-activated protein kinase/ nuclear factor erythroid 2-related factor 2 (AMPK/ Nrf2) oxidative stress pathway and the Toll-like receptor 4 (TLR-4)/ nuclear factor kappa-B (NF-κB) inflammatory pathway and to establish an in vivo liver disturbance model using male C57BL/6J and TLR-4 knockout (-/- ) mice. For this, we evaluated the expression levels of SREBP-1 and Nrf2 after silencing the expression of AMPK using siRNA technology. Our results show that with regard to the TLR-4/ NF-κB inflammatory pathway, DCP inhibits TLR-4, up-regulates the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ), reduces the expression of phospho(p)-NF-κB and leads to the reduction of downstream inflammatory factors, such as tumour necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1β, thereby inhibiting the inflammatory response. Regarding the AMPK/ Nrf2 oxidative stress pathway, DCP up-regulates the expression of p-AMPK and Nrf2, in addition to regulating glucose and lipid metabolism, oxidative stress and ameliorating liver disturbance symptoms. In summary, our study shows that DCP alleviates liver disturbances by inhibiting mechanisms used during liver inflammation and oxidative stress depression, which provides a new strategy for the clinical treatment of liver disturbance.
Collapse
Affiliation(s)
- Kefeng Zhang
- College of Pharmacy, Guilin Medical University, Guilin, China.,College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qiongmei Xu
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Ya Gao
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Houkang Cao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yuanyu Lian
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Zimeng Li
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Jie Xu
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Mingli Zhong
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Jiani Li
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Riming Wei
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Jianghui Dong
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Ling Jin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
24
|
Ye S, Zeng S, Huang M, Chen J, Chen X, Xu P, Wang Q, Gao W, Yang B, Hao B, Huang W, Liu Q. [Effect of the chemoprotectant tempol on anti-tumor activity of cisplatin]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:883-890. [PMID: 31511206 DOI: 10.12122/j.issn.1673-4254.2019.08.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the effect of the chemoprotectant tempol on the anti-tumor activity of cisplatin (DDP). METHODS The cellular toxicity of tempol in human colon cancer SW480 cells and mouse colon cancer CT26 cells were evaluated using MTT and cell counting kit-8 assays. CalcuSyn software analysis was used to determine the interaction between tempol and DDP in inhibition of the cell viability. A subcutaneous homograft mouse model of colon cancer was established. The mice were randomly divided into control group, tempol group, cisplatin group and tempol + DDP treatment group with intraperitoneal injections of the indicated agents. The tumor size, body weight and lifespan of the mice were measured, and HE staining was used to analyze the cytotoxic effect of the agents on the kidney and liver. Immunohistochemistry and Western blotting were performed to detect the expression of Bax and Bcl2 in the tumor tissue, and TUNEL staining was used to analyze the tumor cell apoptosis. The level of reactive oxygen species (ROS) in the tumor tissue was determined using flow cytometry. RESULTS Tempol showed inhibitory effects on the viability of SW480 and CT26 cells. CalcuSyn software analysis showed that tempol had a synergistic anti-tumor effect with DDP (CI < 1). In the homograft mouse model, tempol treatment alone did not produce obvious anti-tumor effect. HE staining showed that the combined use of tempol and DDP alleviated DDP-induced fibrogenesis in the kidneys, but tempol also reduced the anti-tumor activity of DDP. Compared with the mice treated with DDP alone, the mice treated with both tempol and DDP had a significantly larger tumor size (P < 0.01) and a shorter lifespan (P < 0.05). Tempol significantly reversed DDP-induced expression of Bax and Bcl2 in the tumor tissue and tumor cell apoptosis (P < 0.001), and obviously reduced the elevation of ROS level in the tumor tissue induced by DDP treatment (P < 0.05). CONCLUSIONS Tempol can attenuate the anti-tumor effect of DDP while reducing the side effects of DDP. Caution must be taken and the risks and benefits should be carefully weighed when considering the use of tempol as an anti-oxidant to reduce the toxicities of DDP.
Collapse
Affiliation(s)
- Shuangyan Ye
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Sisi Zeng
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Mengqiu Huang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Jianping Chen
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Xi Chen
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Pengfei Xu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Qianli Wang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Wenwen Gao
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Bingsheng Yang
- Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Bingtao Hao
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China
| | - Wenhuan Huang
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Department of Human Anatomy, School of Basic Medical Sciences, Guangdong Medical University, Zhanjiang 524003, China
| | - Qiuzhen Liu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University/Guangdong Provincial Key Laboratory of Cancer Immunotherapy/Guangzhou Key Laboratory of Tumor Immunology Research, Guangzhou 510515, China.,Center for Medical Transformation, Shunde Hospital, Southern Medical University, Foshan 528300, China
| |
Collapse
|