1
|
Zheng Q, Yang SJ, Yi EJ, Park SJ, Jin X, Nguyen TTM, Yi GS, Jeon YJ, Yi TH. Enzyme-assisted Rosa davurica mitigates UV-induced skin photodamage by modulating apoptosis through Nrf2/ARE and MAPK/NF-κB pathways. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 263:113098. [PMID: 39787977 DOI: 10.1016/j.jphotobiol.2025.113098] [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: 07/18/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Exposure to UV irradiation results in abnormal, extensive apoptosis of skin cells. This excessive cell death can promote inflammation and alter the microenvironment, increasing the risk of skin cancer. Despite extensive research, few materials are effective at simultaneously protecting against both UVA and UVB irradiation. This study aims to develop dual-action material using enzyme-assisted extraction of Rosa davurica Pall (RD) to prevent skin photodamage caused by UVA and UVB irradiation. Three different enzymes were used to assist the extraction of RD, followed by an analysis of the changes in active component levels. Skin photodamage models were established by exposing Normal Human Dermal Fibroblasts (NHDF) and HaCaT cells to UVA and UVB irradiation. The impact of enzyme-assisted extracted RD (ERD) on Reactive Oxygen Species (ROS) production and cell apoptosis was assessed using Flow Cytometry. The effects of ERDs on inflammatory cytokines were measured using ELISA, and RT-PCR was used to evaluate its impact on apoptotic gene expression in photodamaged cells. Furthermore, the impact of ERDs on the Nuclear factor erythroid 2-related factor 2 (Nrf2)/Antioxidant response element (ARE) and Mitogen-activated protein kinases (MAPK)/Nuclear factor-κB (NF-κB) signaling pathways was assessed through Western blot analysis. Finally, the impact of ERDs on full-thickness artificial skin tissue after UV irradiation was assessed using hematoxylin and eosin (H&E) staining. Furthermore, leveraging the experimental results, network pharmacology was utilized to explore the potential of ERDs in preventing skin cancer. Enzyme-assisted extraction enhanced the bioactive components of RD. ERDs effectively reduced ROS levels and suppressed the secretion of Tumor necrosis factor (TNF)-α, Interleukin (IL)-1β, and IL-6 by modulating the Nrf2/ARE and inhibiting the MAPK/NF-κB signaling pathways. This mechanism promoted the expression of the anti-apoptotic gene Bcl-2 and decreased the activity of proapoptotic genes BAX, caspase-3, and caspase-9, thereby countering UV-induced apoptosis. Additionally, staining results demonstrated that ERDs effectively repaired UV-induced photodamage and maintained the integrity of skin structure. ERDs provides comprehensive protection against photodamage induced by UVA and UVB irradiation, demonstrating its potential as an effective photoprotective material and possibly in preventing skin cancer.
Collapse
Affiliation(s)
- Qiwen Zheng
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung, Yongin 17104, Republic of Korea
| | - Su-Jin Yang
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung, Yongin 17104, Republic of Korea
| | - Eun-Ji Yi
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung, Yongin 17104, Republic of Korea; Snowwhitefactory Co., Ltd., 184, Jungbu-daero, Giheung, Yongin 17905, Republic of Korea
| | - Se-Jig Park
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung, Yongin 17104, Republic of Korea
| | - Xiangji Jin
- Department of Dermatology, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dong-daemun, Seoul 02447, Republic of Korea
| | - Trang Thi Minh Nguyen
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung, Yongin 17104, Republic of Korea
| | - Gyeong-Seon Yi
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung, Yongin 17104, Republic of Korea
| | - Yong-Jun Jeon
- Department of Oriental Medicine Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung, Yongin 17104, Republic of Korea
| | - Tae-Hoo Yi
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung, Yongin 17104, Republic of Korea.
| |
Collapse
|
2
|
Podyacheva E, Snezhkova J, Onopchenko A, Dyachuk V, Toropova Y. The Role of MicroRNAs in the Pathogenesis of Doxorubicin-Induced Vascular Remodeling. Int J Mol Sci 2024; 25:13335. [PMID: 39769102 PMCID: PMC11728060 DOI: 10.3390/ijms252413335] [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: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 01/14/2025] Open
Abstract
Doxorubicin (DOX), a cornerstone chemotherapeutic agent, effectively combats various malignancies but is marred by significant cardiovascular toxicity, including endothelial damage, chronic heart failure, and vascular remodeling. These adverse effects, mediated by oxidative stress, mitochondrial dysfunction, inflammatory pathways, and dysregulated autophagy, underscore the need for precise therapeutic strategies. Emerging research highlights the critical role of microRNAs (miRNAs) in DOX-induced vascular remodeling and cardiotoxicity. miRNAs, such as miR-21, miR-22, miR-25, miR-126, miR-140-5p, miR-330-5p, miR-146, miR-143, miR-375, miR-125b, miR-451, miR-34a-5p, and miR-9, influence signaling pathways like TGF-β/Smad, AMPKa/SIRT, NF-κB, mTOR, VEGF, and PI3K/AKT/Nrf2, impacting vascular homeostasis, angiogenesis, and endothelial-to-mesenchymal transition. Despite existing studies, gaps remain in understanding the full spectrum of miRNAs involved and their downstream effects on vascular remodeling. This review synthesizes the current knowledge on miRNA dysregulation during DOX exposure, focusing on their dual roles in cardiovascular pathology and tumor progression. Strategies to reduce DOX cardiotoxicity include modulating miRNA expression to restore signaling balance, targeting pro-inflammatory and pro-fibrotic pathways, and leveraging miRNA inhibitors or mimics. This review aims to organize and integrate the existing knowledge on the role of miRNAs in vascular remodeling, particularly in the contexts of DOX treatment and the progression of various cardiovascular diseases, including their potential involvement in tumor growth.
Collapse
Affiliation(s)
| | | | | | | | - Yana Toropova
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, 197341 Saint-Petersburg, Russia or (E.P.); (J.S.); (A.O.); (V.D.)
| |
Collapse
|
3
|
Shi C, Li Y, You Z, Tian Y, Zhu X, Xu H, Yang M, Zhang Y, Dong R, Quan H, Shang Y, Li X. Mangiferin Ameliorates CCl 4-Triggered Acute Liver Injury by Inhibiting Inflammatory Response and Oxidative Stress: Involving the Nrf2-ARE Pathway. J Inflamm Res 2024; 17:7081-7097. [PMID: 39380664 PMCID: PMC11460351 DOI: 10.2147/jir.s476288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
Purpose Acute liver injury (ALI) is characterized by inflammation and oxidative stress (OS). Although mangiferin (MGF) has antioxidant and anti-inflammatory effects, its role in ALI remains unclear. Accordingly, we investigated the MGF molecular mechanism in carbon tetrachloride (CCl4)-induced ALI in vivo and in vitro. Materials and Methods The CCl4 was utilized to induce ALI in mice. In vivo, the therapeutic effects of MGF on CCl4-induced liver injury were evaluated through biochemical assays and histomorphological analysis. Additionally, immunohistochemistry, immunofluorescence, ELISA and Western blotting were further applied to explore the mechanism. In vitro, The CCK-8 assay and flow cytometry were employed to investigate the protective effects of MGF against CCl4-induced toxicity in HepG2 cells, while mitochondrial reactive oxygen species levels and Western blotting were used to explore the biological effects and molecular mechanisms. Results MGF treatment resulted in a reduction in serum levels of AST and ALT, diminished concentrations of TNF-α, IL-6, and IL-1β in liver tissue, and concurrently decreased cellular apoptosis. Furthermore, MGF pretreatment enhanced the activity of SOD and GSH while concurrently diminishing the MDA production. This study further demonstrated the upregulation of Nrf2, NQO1, and HO-1 protein expression levels, as well as the downregulation of p-p65 protein expression levels. In vitro investigations revealed that the mitigation of CCl4-induced inflammation and OS by MGF was mediated via the Nrf2- antioxidant response element (ARE) pathway, which was disrupted by ML385 in HepG2 cells. Conclusion CCl4 can induce liver injury, while treatment with MGF mitigates ALI by inhibiting oxidative stress, inflammation, and apoptosis. The protective mechanism of MGF is mediated by the Nrf2-ARE pathway activation.
Collapse
Affiliation(s)
- Caixing Shi
- School of Basic Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yueyao Li
- College of Integrated Chinese and Western Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Zhidong You
- School of Nursing, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yiran Tian
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Xiaoyu Zhu
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Hao Xu
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Menghan Yang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yutong Zhang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Rui Dong
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Huirong Quan
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Yongyi Shang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| | - Xiaojin Li
- School of Basic Medicine, Jining Medical University, Jining, 272067, People’s Republic of China
| |
Collapse
|
4
|
Wen X, Tang S, Wan F, Zhong R, Chen L, Zhang H. The PI3K/Akt-Nrf2 Signaling Pathway and Mitophagy Synergistically Mediate Hydroxytyrosol to Alleviate Intestinal Oxidative Damage. Int J Biol Sci 2024; 20:4258-4276. [PMID: 39247828 PMCID: PMC11379072 DOI: 10.7150/ijbs.97263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/24/2024] [Indexed: 09/10/2024] Open
Abstract
Oxidative stress is a major pathogenic factor in many intestinal diseases, such as inflammatory bowel disease (IBD) and colorectal cancer (CRC). The Nrf2 signaling pathway and mitophagy can reduce reactive oxygen species (ROS) and alleviate oxidative stress, but their relationship is unclear. Hydroxytyrosol (HT), a polyphenolic compound abundant in olive oil, has strong antioxidant activity and may help treat these diseases. We used pigs as a model to investigate HT's effect on intestinal oxidative damage and its mechanisms. Diquat (DQ) induced oxidative stress and impaired intestinal barrier function, which HT mitigated. Mechanistic studies in IPEC-J2 cells showed that HT protected against oxidative damage by activating the PI3K/Akt-Nrf2 signaling pathway and promoting mitophagy. Our study highlighted the synergistic relationship between Nrf2 and mitophagy in mediating HT's antioxidant effects. Inhibition studies confirmed that disrupting either pathway compromised HT's protective effects. Maintaining redox balance through Nrf2 and mitophagy is important for eliminating excess ROS. Nrf2 increases antioxidant enzymes to clear existing ROS, while mitophagy removes damaged mitochondria and reduces ROS generation. This study demonstrates that these pathways collaboratively modulate the antioxidant effects of HT, with neither being dispensable. Targeting Nrf2 and mitophagy could be a promising strategy for treating oxidative stress-related intestinal diseases, with HT as a potential treatment.
Collapse
Affiliation(s)
- Xiaobin Wen
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Shanlong Tang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Fan Wan
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| |
Collapse
|
5
|
Shi P, Lin Z, Song Y, Li Z, Zeng M, Luo L, Cao Y, Zhu X. Chemotherapy-initiated cysteine-rich protein 61 decreases acute B-lymphoblastic leukemia chemosensitivity. J Cancer Res Clin Oncol 2024; 150:159. [PMID: 38530432 PMCID: PMC10965586 DOI: 10.1007/s00432-024-05692-8] [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: 10/24/2023] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
PURPOSE Chemoresistance is a major challenge for acute lymphoblastic leukemia (ALL) treatment. Cysteine-rich protein 61 (Cyr61) plays an important role in drug resistance modulation of tumor cells, and Cyr61 levels are increased in the bone marrow of patients with ALL and contribute to ALL cell survival. However, the effect of Cyr61 on B cell acute lymphoblastic leukemia (B-ALL) cell chemosensitivity and the regulatory mechanisms underlying Cyr61 production in bone marrow remain unknown. METHODS Nalm-6 and Reh human B-ALL cell lines were used in this study. Cyr61 levels were assessed using quantitative real-time PCR (qRT-PCR), western blot analysis, and enzyme-linked immunosorbent assay. The effect of Cyr61 on B-ALL cell chemosensitivity to daunorubicin (DNR) was evaluated using cell viability and flow cytometry analyses. The regulatory mechanisms of Cyr61 production in bone marrow were examined using qRT-PCR and western blot analysis. RESULTS Cyr61 knockdown and overexpression increased and decreased the chemosensitivity of B-ALL cells to DNR, respectively. Cyr61 attenuated chemotherapeutic drug-induced apoptosis by upregulating B cell lymphoma-2. Notably, DNR induced DNA damage response and increased Cyr61 secretion in B-ALL cells through the ataxia telangiectasia mutated (ATM)-dependent nuclear factor kappa B pathway. CONCLUSION DNR induces Cyr61 production in B-ALL cells, and increased Cyr61 levels reduce the chemosensitivity of B-ALL cells. Consequently, targeting Cyr61 or related ATM signaling pathway may present a promising treatment strategy to enhance the chemosensitivity of patients with B-ALL.
Collapse
Affiliation(s)
- Pengchong Shi
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Zhen Lin
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Yanfang Song
- Department of Clinical Laboratory, Affiliated People Hospital of Fujian University of Traditional Chinese Medicine, 602 Bayiqi Road, Fuzhou, 350001, Fujian, China
| | - Zhaozhong Li
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Menglu Zeng
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Li Luo
- Department of Clinical Laboratory, Affiliated People Hospital of Fujian University of Traditional Chinese Medicine, 602 Bayiqi Road, Fuzhou, 350001, Fujian, China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China.
| | - Xianjin Zhu
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China.
| |
Collapse
|
6
|
Xiang D, Zhou L, Yang R, Yuan F, Xu Y, Yang Y, Qiao Y, Li X. Advances in Ferroptosis-Inducing Agents by Targeted Delivery System in Cancer Therapy. Int J Nanomedicine 2024; 19:2091-2112. [PMID: 38476278 PMCID: PMC10929151 DOI: 10.2147/ijn.s448715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Currently, cancer remains one of the most significant threats to human health. Treatment of most cancers remains challenging, despite the implementation of diverse therapies in clinical practice. In recent years, research on the mechanism of ferroptosis has presented novel perspectives for cancer treatment. Ferroptosis is a regulated cell death process caused by lipid peroxidation of membrane unsaturated fatty acids catalyzed by iron ions. The rapid development of bio-nanotechnology has generated considerable interest in exploiting iron-induced cell death as a new therapeutic target against cancer. This article provides a comprehensive overview of recent advancements at the intersection of iron-induced cell death and bionanotechnology. In this respect, the mechanism of iron-induced cell death and its relation to cancer are summarized. Furthermore, the feasibility of a nano-drug delivery system based on iron-induced cell death for cancer treatment is introduced and analyzed. Secondly, strategies for inducing iron-induced cell death using nanodrug delivery technology are discussed, including promoting Fenton reactions, inhibiting glutathione peroxidase 4, reducing low glutathione levels, and inhibiting system Xc-. Additionally, the article explores the potential of combined treatment strategies involving iron-induced cell death and bionanotechnology. Finally, the application prospects and challenges of iron-induced nanoagents for cancer treatment are discussed.
Collapse
Affiliation(s)
- Debiao Xiang
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People’s Republic of China
- The Clinical Application Research Institute of Antibiotics in Changsha, Changsha, Hunan Province, People’s Republic of China
| | - Lili Zhou
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, People’s Republic of China
| | - Rui Yang
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, People’s Republic of China
| | - Fang Yuan
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People’s Republic of China
- The Clinical Application Research Institute of Antibiotics in Changsha, Changsha, Hunan Province, People’s Republic of China
| | - Yilin Xu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, People’s Republic of China
| | - Yuan Yang
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, People’s Republic of China
| | - Yong Qiao
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People’s Republic of China
- The Clinical Application Research Institute of Antibiotics in Changsha, Changsha, Hunan Province, People’s Republic of China
| | - Xin Li
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People’s Republic of China
- The Clinical Application Research Institute of Antibiotics in Changsha, Changsha, Hunan Province, People’s Republic of China
| |
Collapse
|
7
|
Adeyemi DH, Obembe OO, Hamed MA, Akhigbe RE. Sodium acetate ameliorates doxorubicin-induced cardiac injury via upregulation of Nrf2/HO-1 signaling and downregulation of NFkB-mediated apoptotic signaling in Wistar rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:423-435. [PMID: 37458777 DOI: 10.1007/s00210-023-02620-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/10/2023] [Indexed: 01/07/2024]
Abstract
Despite the effectiveness of doxorubicin (DOX) in the management of a wide range of cancers, a major challenge is its cardio-toxic effect. Oxidative stress, inflammation, and apoptosis are major pathways for the cardiotoxic effect of DOX. On the other hand, acetate reportedly exerts antioxidant, anti-inflammatory, and anti-apoptotic activities. This particular research assessed the impact of acetate on cardiotoxicity induced by DOX. Mechanistically, acetate dramatically inhibited DOX-induced upregulation of xanthine oxidase and uric acid pathway as well as downregulation of Nrf2/HO-1 signaling and its upstream proteins (reduced glutathione peroxidase, superoxide dismutase, glutathione-S-transferase, glutathione, and catalase, glutathione reductase). In addition, acetate markedly attenuated DOX-driven rise inTNF-α, NFkB IL-6 and IL-1β expression, and myeloperoxidase activity. Furthermore, acetate significantly ameliorated DOX-led suppression of Bcl-2 and Ca2+-ATPase activity and upregulation of Bax, caspase 3, and caspase 9 actions. Improved body weight, heart structural integrity, and cardiac function as depicted by cardiac injury markers convoyed these cascades of events. Summarily, the present study demonstrated that acetate protects against DOX-induced cardiotoxicity by upregulating Nrf2/HO-1 signaling and downregulating NFkB-mediated activation of Bax/Bcl-2 and caspase signaling.
Collapse
Affiliation(s)
- D H Adeyemi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Osogbo, Osun State, Nigeria
| | - O O Obembe
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Osogbo, Osun State, Nigeria
| | - M A Hamed
- Department of Medical Laboratory Sciences, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria
- The Brainwill Laboratories, Osogbo, Osun State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - R E Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria.
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| |
Collapse
|
8
|
AbuZahra HM. Kirenol protects against oxidized low-density lipoprotein induced damages in endothelial cells. BRAZ J BIOL 2024; 84:e259421. [DOI: 10.1590/1519-6984.259421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/05/2022] [Indexed: 11/22/2022] Open
Abstract
Abstract Kirenol (KNL) has recently been reported to have anti-inflammatory properties. Yet, little is known about the potential mechanisms of its anti-inflammatory properties. In HUVECs, we elucidated the anti-inflammatory mechanisms of kirenol. RT-PCR was used to test mRNA of pro-inflammatory mediators produced by Ox-LDL. The viability of cells was measured using MTT. Western blots analyzed protein levels. On Ox-LDL-stimulated HUVECs, KNL significantly inhibited the production of pro-inflammatory mediators such as NO, IL-1β, iNOS, TNF-α and IL-6. p38, ROS and Nrf2 expression were inhibited by KNL. Inhibition of p38, ROS, and KNL caused nuclear accumulation of Nrf2. KNL attenuated Ox-LDL-induced phosphorylation of ERK1/2 and p38, too. Based on our results, KNL inhibits NF-кB and MAPK signaling in HUVECs by activating Nrf2 signaling. There's a possibility that KNL could be developed into an anti-inflammatory drug.
Collapse
|
9
|
Xia L, Ma W, Afrashteh A, Sajadi MA, Fakheri H, Valilo M. The nuclear factor erythroid 2-related factor 2/p53 axis in breast cancer. Biochem Med (Zagreb) 2023; 33:030504. [PMID: 37841775 PMCID: PMC10564154 DOI: 10.11613/bm.2023.030504] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
One of the most important factors involved in the response to oxidative stress (OS) is the nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates the expression of components such as antioxidative stress proteins and enzymes. Under normal conditions, Kelch-like ECH-associated protein 1 (Keap1) keeps Nrf2 in the cytoplasm, thus preventing its translocation to the nucleus and inhibiting its role. It has been established that Nrf2 has a dual function; on the one hand, it promotes angiogenesis and cancer cell metastasis while causing resistance to drugs and chemotherapy. On the other hand, Nrf2 increases expression and proliferation of glutathione to protect cells against OS. p53 is a tumour suppressor that activates the apoptosis pathway in aging and cancer cells in addition to stimulating the glutaminolysis and antioxidant pathways. Cancer cells use the antioxidant ability of p53 against OS. Therefore, in the present study, we discussed function of Nrf2 and p53 in breast cancer (BC) cells to elucidate their role in protection or destruction of cancer cells as well as their drug resistance or antioxidant properties.
Collapse
Affiliation(s)
- Lei Xia
- Surgical oncology ward 2, Qinghai Provincial People’s Hospital, Xining Qinghai, China
| | - Wenbiao Ma
- Surgical oncology ward 2, Qinghai Provincial People’s Hospital, Xining Qinghai, China
| | - Ahmad Afrashteh
- Department of Periodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hadi Fakheri
- Paramedical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
10
|
He Y, Xi J, Fang J, Zhang B, Cai W. Aloe-emodin alleviates doxorubicin-induced cardiotoxicity via inhibition of ferroptosis. Free Radic Biol Med 2023; 206:13-21. [PMID: 37364691 DOI: 10.1016/j.freeradbiomed.2023.06.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
Aloe-emodin (AE), a novel ferroptosis inhibitor, alleviates the doxorubicin (DOX)-induced cardiotoxicity in H9c2 rat cardiomyocytes. The inhibition of ferroptosis and the protective effect against cardiotoxicity were evaluated via MTT assay in H9c2 cells. The molecular mechanism of action (MOA) of nuclear factor erythroid 2-related factor 2 (Nrf2) activation, including transactivation of multiple downstream cytoprotective genes, were further assessed by Western blot, luciferase reporter assay and qRT-PCR analyses. Fluorescent imaging was performed to detect the change of intracellular reactive oxygen species, mitochondrial membrane potential and lipid peroxidation. In addition, an infrared spectroscopy was employed to detect the AE-Fe (II) complex. AE, alleviates oxidative stress in DOX-induced H9c2 cells by activating Nrf2 and increasing the expression of Nrf2 downstream antioxidant genes, SLC7A11 and GPX4. Furthermore, AE complexes bivalent iron and regulates the intracellular iron-related genes. In conclusion, the discovery of AE as a novel ferroptosis inhibitor and its MOA provides a new perspective for further exploration of cardio-protective agents in cancer patients during chemotherapy.
Collapse
Affiliation(s)
- Ying He
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Junmin Xi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China; School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu, 210094, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China.
| | - Wenqing Cai
- Regor Therapeutics Inc,1206 Zhangjiang Road, Building C, Pu Dong New District, Shanghai, 201210, China.
| |
Collapse
|
11
|
Podyacheva E, Danilchuk M, Toropova Y. Molecular mechanisms of endothelial remodeling under doxorubicin treatment. Biomed Pharmacother 2023; 162:114576. [PMID: 36989721 DOI: 10.1016/j.biopha.2023.114576] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Doxorubicin (DOX) is an effective antineoplastic agent used to treat various types of cancers. However, its use is limited by the development of cardiotoxicity, which may result in heart failure. The exact mechanisms underlying DOX-induced cardiotoxicity are not fully understood, but recent studies have shown that endothelial-mesenchymal transition (EndMT) and endothelial damage play a crucial role in this process. EndMT is a biological process in which endothelial cells lose their characteristics and transform into mesenchymal cells, which have a fibroblast-like phenotype. This process has been shown to contribute to tissue fibrosis and remodeling in various diseases, including cancer and cardiovascular diseases. DOX-induced cardiotoxicity has been demonstrated to increase the expression of EndMT markers, suggesting that EndMT may play a critical role in the development of this condition. Furthermore, DOX-induced cardiotoxicity has been shown to cause endothelial damage, leading to the disruption of the endothelial barrier function and increased vascular permeability. This can result in the leakage of plasma proteins, leading to tissue edema and inflammation. Moreover, DOX can impair the production of nitric oxide, endothelin-1, neuregulin, thrombomodulin, thromboxane B2 etc. by endothelial cells, leading to vasoconstriction, thrombosis and further impairing cardiac function. In this regard, this review is devoted to the generalization and structuring of information about the known molecular mechanisms of endothelial remodeling under the action of DOX.
Collapse
|
12
|
Shi S, Chen Y, Luo Z, Nie G, Dai Y. Role of oxidative stress and inflammation-related signaling pathways in doxorubicin-induced cardiomyopathy. Cell Commun Signal 2023; 21:61. [PMID: 36918950 PMCID: PMC10012797 DOI: 10.1186/s12964-023-01077-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/12/2023] [Indexed: 03/16/2023] Open
Abstract
Doxorubicin (DOX) is a powerful and commonly used chemotherapeutic drug, used alone or in combination in a variety of cancers, while it has been found to cause serious cardiac side effects in clinical application. More and more researchers are trying to explore the molecular mechanisms of DOX-induced cardiomyopathy (DIC), in which oxidative stress and inflammation are considered to play a significant role. This review summarizes signaling pathways related to oxidative stress and inflammation in DIC and compounds that exert cardioprotective effects by acting on relevant signaling pathways, including the role of Nrf2/Keap1/ARE, Sirt1/p66Shc, Sirt1/PPAR/PGC-1α signaling pathways and NOS, NOX, Fe2+ signaling in oxidative stress, as well as the role of NLRP3/caspase-1/GSDMD, HMGB1/TLR4/MAPKs/NF-κB, mTOR/TFEB/NF-κB pathways in DOX-induced inflammation. Hence, we attempt to explain the mechanisms of DIC in terms of oxidative stress and inflammation, and to provide a theoretical basis or new idea for further drug research on reducing DIC. Video Abstract.
Collapse
Affiliation(s)
- Saixian Shi
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, Sichuan Province, China.,School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Ye Chen
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, Sichuan Province, China.,School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Zhijian Luo
- Department of Ultrasound, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Guojun Nie
- The First Outpatient Department of People's Liberation Army Western Theater General Hospital, Chengdu, 610000, Sichuan Province, China
| | - Yan Dai
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, Sichuan Province, China.
| |
Collapse
|
13
|
Lima EA, Wyde RA, Sorace AG, Yankeelov TE. Optimizing combination therapy in a murine model of HER2+ breast cancer. COMPUTER METHODS IN APPLIED MECHANICS AND ENGINEERING 2022; 402:115484. [PMID: 37800167 PMCID: PMC10552906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Human epidermal growth factor receptor 2 positive (HER2+) breast cancer is frequently treated with drugs that target the HER2 receptor, such as trastuzumab, in combination with chemotherapy, such as doxorubicin. However, an open problem in treatment design is to determine the therapeutic regimen that optimally combines these two treatments to yield optimal tumor control. Working with data quantifying temporal changes in tumor volume due to different trastuzumab and doxorubicin treatment protocols in a murine model of human HER2+ breast cancer, we propose a complete framework for model development, calibration, selection, and treatment optimization to find the optimal treatment protocol. Through different assumptions for the drug-tumor interactions, we propose ten different models to characterize the dynamic relationship between tumor volume and drug availability, as well as the drug-drug interaction. Using a Bayesian framework, each of these models are calibrated to the dataset and the model with the highest Bayesian information criterion weight is selected to represent the biological system. The selected model captures the inhibition of trastuzumab due to pre-treatment with doxorubicin, as well as the increase in doxorubicin efficacy due to pre-treatment with trastuzumab. We then apply optimal control theory (OCT) to this model to identify two optimal treatment protocols. In the first optimized protocol, we fix the maximum dosage for doxorubicin and trastuzumab to be the same as the maximum dose delivered experimentally, while trying to minimize tumor burden. Within this constraint, optimal control theory indicates the optimal regimen is to first deliver two doses of trastuzumab on days 35 and 36, followed by two doses of doxorubicin on days 37 and 38. This protocol predicts an additional 45% reduction in tumor burden compared to that achieved with the experimentally delivered regimen. In the second optimized protocol we fix the tumor control to be the same as that obtained experimentally, and attempt to reduce the doxorubicin dose. Within this constraint, the optimal regimen is the same as the first optimized protocol but uses only 43% of the doxorubicin dose used experimentally. This protocol predicts tumor control equivalent to that achieved experimentally. These results strongly suggest the utility of mathematical modeling and optimal control theory for identifying therapeutic regimens maximizing efficacy and minimizing toxicity.
Collapse
Affiliation(s)
- Ernesto A.B.F. Lima
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, United States of America
- Texas Advanced Computing Center, The University of Texas at Austin, United States of America
| | - Reid A.F. Wyde
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, United States of America
| | - Anna G. Sorace
- Department of Radiology, The University of Alabama at Birmingham, United States of America
- Department of Biomedical Engineering, The University of Alabama at Birmingham, United States of America
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, United States of America
| | - Thomas E. Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, United States of America
- Department of Biomedical Engineering, The University of Texas at Austin, United States of America
- Department of Diagnostic Medicine, The University of Texas at Austin, United States of America
- Department of Oncology, The University of Texas at Austin, United States of America
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, United States of America
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, United States of America
| |
Collapse
|
14
|
Alfwuaires M, Elsawy H, Sedky A. Acacetin Inhibits Cell Proliferation and Induces Apoptosis in Human Hepatocellular Carcinoma Cell Lines. Molecules 2022; 27:molecules27175361. [PMID: 36080130 PMCID: PMC9457933 DOI: 10.3390/molecules27175361] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/13/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Human hepatocellular carcinoma (HCC) is the fifth most common cancer and the third leading cause of death across the world. Recent evidence suggests that STAT3 regulates proliferative, survival, metastasis, and angiogenesis genes in HCC. Novel agents that suppress STAT3 activation can be used to prevent or treat HCC. We used a functional proteomics tumor pathway technology platform and multiple HCC cell lines to investigate the effects of acacetin (ACN) on STAT3 activation, protein kinases, phosphatases, products of STAT3-regulated genes, and apoptosis. ACN was found to inhibit STAT3 activation in a dose- and time-dependent manner in HCC cells. Upstream kinases c-Src, Janus-activated kinase 1, and Janus-activated kinase 2 were also inhibited. The ACN inhibition of STAT3 was abolished by vanadate treatment, suggesting the involvement of tyrosine phosphatase activity. ACN was found to suppress the protein expression of genes involved in proliferation, survival, and angiogenesis via STAT3 inhibition. ACN appears to be a novel STAT3 inhibitor and may be a promising therapeutic compound for application in the treatment of HCC and other cancers.
Collapse
Affiliation(s)
- Manal Alfwuaires
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia
- Correspondence: (M.A.); (H.E.); Tel.: +96-61-3589-1008 (M.A.); +96-61-3589-7402 (H.E.)
| | - Hany Elsawy
- Department of Chemistry, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia
- Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
- Correspondence: (M.A.); (H.E.); Tel.: +96-61-3589-1008 (M.A.); +96-61-3589-7402 (H.E.)
| | - Azza Sedky
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia
- Department of Zoology Faculty of Science, Alexandria University, Alexandria 21526, Egypt
| |
Collapse
|
15
|
Kang L, Zhang H, Jia C, Zhang R, Shen C. Targeting Oxidative Stress and Inflammation in Intervertebral Disc Degeneration: Therapeutic Perspectives of Phytochemicals. Front Pharmacol 2022; 13:956355. [PMID: 35903342 PMCID: PMC9315394 DOI: 10.3389/fphar.2022.956355] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Low back pain is a major cause of disability worldwide that declines the quality of life; it poses a substantial economic burden for the patient and society. Intervertebral disc (IVD) degeneration (IDD) is the main cause of low back pain, and it is also the pathological basis of several spinal degenerative diseases, such as intervertebral disc herniation and spinal stenosis. The current clinical drug treatment of IDD focuses on the symptoms and not their pathogenesis, which results in frequent recurrence and gradual aggravation. Moreover, the side effects associated with the long-term use of these drugs further limit their use. The pathological mechanism of IDD is complex, and oxidative stress and inflammation play an important role in promoting IDD. They induce the destruction of the extracellular matrix in IVD and reduce the number of living cells and functional cells, thereby destroying the function of IVD and promoting the occurrence and development of IDD. Phytochemicals from fruits, vegetables, grains, and other herbs play a protective role in the treatment of IDD as they have anti-inflammatory and antioxidant properties. This article reviews the protective effects of phytochemicals on IDD and their regulatory effects on different molecular pathways related to the pathogenesis of IDD. Moreover, the therapeutic limitations and future prospects of IDD treatment have also been reviewed. Phytochemicals are promising candidates for further development and research on IDD treatment.
Collapse
|
16
|
Supercritical Impregnation of Mango Leaf Extract into PLA 3D-Printed Devices and Evaluation of Their Biocompatibility with Endothelial Cell Cultures. Polymers (Basel) 2022; 14:polym14132706. [PMID: 35808751 PMCID: PMC9269286 DOI: 10.3390/polym14132706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
The addition of natural substances with pharmacoactive properties to polymeric biomedical devices would provide beneficial regarding the assimilation of these endoprostheses when implanted into a patient’s body. The added drug would facilitate endothelization by regulating the inflammatory processes that such interventions entail, preventing contamination hazards and favoring the angiogenesis or formation of blood vessels in the tissue. The present work used mango leaf extract (MLE) obtained through pressurized ethanol for this purpose. Polylactic acid (PLA) in the form of filaments or 3D-printed disks was impregnated by means of supercritical technology with MLE for the culture essays. The release kinetics has been studied and the polymer matrices have been examined by scanning electron microscopy (SEM). The impregnated devices were subjected to in vitro culture of colony-forming endothelial cells. The influence of the different impregnation conditions used for the production of the MLE impregnated polymeric devices on the development of the cell culture was determined by fluorescence microscopy. The best results were obtained from the calcein cultures on 35 °C MLE impregnated into 3D-printed polymer disks.
Collapse
|
17
|
Brassinin Enhances Apoptosis in Hepatic Carcinoma by Inducing Reactive Oxygen Species Production and Suppressing the JAK2/STAT3 Pathway. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12094733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Plants from the family Brassicaceae produce brassinin (BSN), which is an essential indole phytoalexin. BSN can kill certain types of cancer cells. Using hepatocarcinoma (HCC) cells, we examined the molecular mechanisms of BSN. We found that HCC cell growth was suppressed and apoptosis was induced by BSN via the downregulation of the JAK/STAT3 pathway. The cytoplasmic latent transcription factor STAT3, belonging to the STAT family, acted as both a signal transducer and an activator and was linked to tumor progression and decreased survival. BSN incubation caused HCC cells to produce reactive oxygen species (ROS). By activating caspase-9/-3 and PARP cleavage, Bcl-2 was reduced, and apoptosis was increased. BSN inhibited constitutive STAT3, JAK2, and Src phosphorylation. The JAK/STAT signaling cascade was confirmed by siRNA silencing STAT3 in HCC cells. BSN also suppressed apoptosis by Z-Val-Ala-Asp-Fluoromethylketone (Z-VAD-FMK), an apoptotic inhibitor. N-acetylcysteine (NAC) inhibited the production of ROS and diminished BSN-induced apoptosis. Our findings suggested that BSN has potential as a treatment for cancer.
Collapse
|
18
|
Rajendran P, AlZahrani A. Fucoxanthin suppresses OxLDL-induced inflammation via activation of Nrf2 and inhibition of NF-κB signaling. Asian Pac J Trop Biomed 2022. [DOI: 10.4103/2221-1691.343388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
19
|
Rajendran P, Elsawy H, Sedky A, Alfwuaires M. Ruscogenin protects against deoxynivalenol-Induced hepatic injury by inhibiting oxidative stress, inflammation, and apoptosis through the Nrf2 signaling pathway: An In vitro study. SAUDI JOURNAL OF MEDICINE AND MEDICAL SCIENCES 2022; 10:207-215. [PMID: 36247053 PMCID: PMC9555037 DOI: 10.4103/sjmms.sjmms_725_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/16/2022] [Accepted: 06/21/2022] [Indexed: 12/05/2022] Open
Abstract
Background Deoxynivalenol (DON) is a trichothecene mycotoxin with demonstrated cytotoxicity in several cell lines and animals, primarily owing to inflammation and reactive oxygen species accumulation. Ruscogenin (RGN), a steroidal sapogenin of Radix Ophiopogon japonicus, has significant anti-thrombotic/anti-inflammatory effects. Objective: The aim of this study was to assess the protective role of RGN against DON-induced oxidative stress, which occurs through the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway and is regulated by phosphoinositide 3-kinases/protein kinase B (PI3K/AKT). Methods: The effects were examined using the HepG2 cell line. RGN and DON were suspended in serum-free medium. Cells were seeded onto plates, and then RGN, DON, or both were added over 24 h in triplicates for each group. Results: RGN conferred protection against DON-exhibited cytotoxicity against HepG2 cells. RGN pretreatment downregulated the expression of DON-induced TNF-α and COX-2 and the formation of reactive oxygen species in a dose-dependent manner. RGN upregulated the expression of Nrf2 and its antioxidant proteins as well as mRNA levels of HO-1/NQO-1/HO-1/Nrf2. Similarly, treatment with DON + RGN resulted in upregulation of the pI3K/pAKT signaling pathway in a dose-dependent manner. Finally, RGN was also found to inhibit the DON-induced apoptosis by upregulating the levels of cleaved proteins and downregulating the expression of Bcl2. Conclusion: The study demonstrates that RGN suppresses hepatic cell injury induced by oxidative stress through Nrf2 via activation of the pI3K/AKT signaling pathway.
Collapse
|
20
|
Protective Effect of Flavonoids from a Deep-Sea-Derived Arthrinium sp. against ox-LDL-Induced Oxidative Injury through Activating the AKT/Nrf2/HO-1 Pathway in Vascular Endothelial Cells. Mar Drugs 2021; 19:md19120712. [PMID: 34940711 PMCID: PMC8707590 DOI: 10.3390/md19120712] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Oxidized low-density lipoprotein (ox-LDL)-induced oxidative injury in vascular endothelial cells is crucial for the progression of cardiovascular diseases, including atherosclerosis. Several flavonoids have been shown cardiovascular protective effects. Recently, our research group confirmed that the novel flavonoids isolated from the deep-sea-derived fungus Arthrinium sp., 2,3,4,6,8-pentahydroxy-1-methylxanthone (compound 1) and arthone C (compound 2) effectively scavenged ROS in vitro. In this study, we further investigated whether these compounds could protect against ox-LDL-induced oxidative injury in endothelial cells and the underlying mechanisms. Our results showed that compounds 1 and 2 inhibited ox-LDL-induced apoptosis and adhesion factors expression in human umbilical vein vascular endothelial cells (HUVECs). Mechanistic studies showed that these compounds significantly inhibited the ROS level increase and the NF-κB nuclear translocation induced by ox-LDL. Moreover, compounds 1 and 2 activated the Nrf2 to transfer into nuclei and increased the expression of its downstream antioxidant gene HO-1 by inducing the phosphorylation of AKT in HUVECs. Importantly, the AKT inhibitor MK-2206 2HCl or knockdown of Nrf2 by RNA interference attenuated the inhibition effects of these compounds on ox-LDL-induced apoptosis in HUVECs. Meanwhile, knockdown of Nrf2 abolished the effects of the compounds on ox-LDL-induced ROS level increase and the translocation of NF-κB to nuclei. Collectively, the data showed that compounds 1 and 2 protected endothelial cells against ox-LDL-induced oxidative stress through activating the AKT/Nrf2/HO-1 pathway. Our study provides new strategies for the design of lead compounds for related cardiovascular diseases treatment.
Collapse
|
21
|
Su CH, Chen SP, Chen LY, Yang JJ, Lee YC, Lee SS, Chen HH, Ng YY, Kuan YH. 3-Bromofluoranthene-induced cardiotoxicity of zebrafish and apoptosis in the vascular endothelial cells via intrinsic and extrinsic caspase-dependent pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:112962. [PMID: 34775346 DOI: 10.1016/j.ecoenv.2021.112962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/28/2021] [Accepted: 10/31/2021] [Indexed: 06/13/2023]
Abstract
Fluoranthene, a high-molecular-weight polycyclic aromatic hydrocarbon (PAH), is widely present in air pollutants, including fine inhalable particulate matter. 3-Bromofluoranthene (3-BrFlu), which is a brominated fluoranthene and halogenated PAH, is generated from waste combustion, metallurgical processes, cement production, e-waste dismantling, and photoreaction. Vascular endothelial cells have key functions in the homeostasis and the development of the cardiovascular system. The zebrafish model has been widely employed to study cardiotoxicity and embryotoxicity. However, no evidence has indicated that 3-BrFlu induces cytotoxicity in vascular endothelial cells, or cardiotoxicity and embryotoxicity in zebrafish. In this study, 3-BrFlu induced concentration-dependent changes in embryo- and cardiotoxicity. Cytotoxicity was also induced by 3-BrFlu in a concentration-dependent manner through apoptosis and necrosis in vascular endothelial cells, SVEC4-10 cells. The activities of caspase-3, -8, and -9 were induced by 3-BrFlu via an intrinsic pathway constituting Bcl-2 downregulation, Bad upregulation, and mitochondrial dysfunction; the extrinsic pathway included the expression of death receptors, including tumour necrosis factor α and Fas receptors. These results indicated that 3-BrFlu caused cardio- and embryotoxicity in zebrafish through vascular endothelial cells cytotoxicity resulting from caspase-dependent apoptosis through intrinsic and extrinsic pathways.
Collapse
Affiliation(s)
- Chun-Hung Su
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC; Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
| | - Shih-Pin Chen
- Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Li-You Chen
- Department of Anatomy, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Jiann-Jou Yang
- Department of BioMedical Sciences, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Yi-Chia Lee
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Shiuan-Shinn Lee
- School of Public Health, Chung Shan Medical University, Taichung, Taiwan, ROC
| | - Hsin-Hung Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asia University Hospital, Taichung, Taiwan, ROC; School of Medicine, Institute of Medicine and public health, Chung Shan Medical University, Taichung, Taiwan, ROC; Chung Sheng Clinic, Nantou, Taiwan, ROC
| | - Yan-Yan Ng
- Department of Pediatric, Chung Kang branch, Cheng Ching Hospital, Taichung City, Taiwan, ROC
| | - Yu-Hsiang Kuan
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC.
| |
Collapse
|