1
|
Yue AC, Zhou XD, Song HP, Liu XH, Bi MJ, Han W, Li Q. Effect and molecular mechanism of Sulforaphane alleviates brain damage caused by acute carbon monoxide poisoning:Network pharmacology analysis, molecular docking, and experimental evidence. ENVIRONMENTAL TOXICOLOGY 2024; 39:1140-1162. [PMID: 37860845 DOI: 10.1002/tox.24000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 09/24/2023] [Accepted: 10/07/2023] [Indexed: 10/21/2023]
Abstract
Sulforaphane (SFN) has attracted much attention due to its ability on antioxidant, anti-inflammatory, and anti-apoptotic properties, while its functional targets and underlying mechanism of action on brain injury caused by acute carbon monoxide poisoning (ACOP) have not been fully elucidated. Herein, we used a systematic network pharmacology approach to explore the mechanism of SFN in the treatment of brain damage after ACOP. In this study, the results of network pharmacology demonstrated that there were a total of 81 effective target genes of SFN and 36 drug-disease targets, which were strongly in connection with autophagy-animal signaling pathway, drug metabolism, and transcription disorders in cancer. Upon the further biological function and KEGG signaling pathway enrichment analysis, a large number of them were involved in neuronal death, reactive oxygen metabolic processes and immune functions. Moreover, based on the results of bioinformatics prediction associated with multiple potential targets and pathways, the AMP-activated protein kinase (AMPK) signaling pathway was selected to elucidate the molecular mechanism of SFN in the treatment of brain injury caused by ACOP. The following molecular docking analysis also confirmed that SFN can bind to AMPKα well through chemical bonds. In addition, an animal model of ACOP was established by exposure to carbon monoxide in a hyperbaric oxygen chamber to verify the predicted results of network pharmacology. We found that the mitochondrial ultrastructure of neurons in rats with ACOP was seriously damaged, and apoptotic cells increased significantly. The histopathological changes were obviously alleviated, apoptosis of cortical neurons was inhibited, and the number of Nissl bodies was increased in the SFN group as compared with the ACOP group (p < .05). Besides, the administration of SFN could increase the expressions of phosphorylated P-AMPK and MFN2 proteins and decrease the levels of DRP1, Caspase3, and Casapase9 proteins in the brain tissue of ACOP rats. These findings suggest that network pharmacology is a useful tool for traditional Chinese medicine (TCM) research, SFN can effectively inhibit apoptosis, protect cortical neurons from the toxicity of carbon monoxide through activating the AMPK pathway and may become a potential therapeutic strategy for brain injury after ACOP.
Collapse
Affiliation(s)
- Ao-Chun Yue
- Emergency Department, Shenzhen University General Hospital, Shenzhen, People's Republic of China
- Centre of Integrated Chinese and Western Medicine, School of Clinical Medicine, Qingdao University, Qingdao, People's Republic of China
| | - Xu-Dong Zhou
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Hui-Ping Song
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Xu-Han Liu
- Emergency Department, Shenzhen University General Hospital, Shenzhen, People's Republic of China
| | - Ming-Jun Bi
- Physical Examination Centre, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, People's Republic of China
| | - Wei Han
- Emergency Department, Shenzhen University General Hospital, Shenzhen, People's Republic of China
| | - Qin Li
- Emergency Department, Shenzhen University General Hospital, Shenzhen, People's Republic of China
| |
Collapse
|
2
|
Xue L, Jiang S, Wan XY. Protective Effects of Sesamol on Renal Ischemia-Reperfusion Injury Via Regulation of Nuclear Factor Erythroid 2-Related Factor 2 Pathway. Transplant Proc 2024; 56:290-296. [PMID: 38350822 DOI: 10.1016/j.transproceed.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/28/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND Sesamol is a natural antioxidant known for its potent antioxidant and free radical scavenging properties. This study aimed to explore the therapeutic effects and underlying mechanisms of sesamol in the development of renal ischemia-reperfusion injury (IRI) in mice. METHODS C57BL/6J wild-type mice were divided into 3 groups: IR group, treated with normal saline after undergoing the IRI procedure; Sesamol + IR group, treated with 30 mg/kg/d of sesamol after the IRI procedure; and Sham group, treated with normal saline but not subjected to the IRI process. Renal IRI was induced by performing a right kidney nephrectomy and subjecting the left kidney to 30-minute ischemia, followed by 24-hour reperfusion. Kidney tissues and serum were collected 24 hours post-IRI to assess the impact of sesamol on renal function after IRI. Serum creatinine and blood urea nitrogen levels were assessed, and renal cell apoptosis was detected through terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. The levels of interleukin 1β and interleukin 18 in kidney tissues, as well as indicators of oxidative stress, were also measured. Furthermore, Nrf2-deficient mice were used to examine the protective function of the nuclear factor erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) and NAD(P)H dehydrogenase quinone 1 (NQO1) signaling pathways induced by sesamol, as determined by western blot assay. RESULTS Sesamol demonstrated significant improvement in renal function, along with reductions in renal tubular injury, cell necrosis, and apoptosis in mice. It also effectively lowered key inflammatory mediator levels. Sesamol exhibited antioxidant properties by reducing malondialdehyde levels and enhancing superoxide dismutase activities 24 hours after IRI. Western blot assay revealed increased Nrf2, HO-1, and NQO-1 protein levels with sesamol treatment. Notably, Nrf2-deficient mice did not exhibit the beneficial effects of sesamol. CONCLUSIONS This study demonstrates that sesamol effectively alleviates renal IRI by enhancing antioxidant defenses and reducing inflammation potentially through the Nrf2/HO-1 and NQO1 signaling pathways.
Collapse
Affiliation(s)
- Lu Xue
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Su Jiang
- Department of Rehabilitation Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, People's Republic of China
| | - Xian-Yao Wan
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China.
| |
Collapse
|
3
|
Zhang Q, Dang YY, Luo X, Fu JJ, Zou ZC, Jia XJ, Zheng GD, Li CW. Kazinol B protects H9c2 cardiomyocytes from hypoxia/reoxygenation-induced cardiac injury by modulating the AKT/AMPK/Nrf2 signalling pathway. PHARMACEUTICAL BIOLOGY 2023; 61:362-371. [PMID: 36740871 PMCID: PMC9904293 DOI: 10.1080/13880209.2023.2173247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/07/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT Kazinol B (KB), an isoprenylated flavan derived from Broussonetia kazinoki Sieb. (Moraceae) root, has long been used in folk medicine. OBJECTIVE This study examines the protective effects of KB and its underlying mechanisms in hypoxia and reoxygenation (H/R)-induced cardiac injury in H9c2 rat cardiac myoblasts. MATERIALS AND METHODS H9c2 cells were incubated with various concentrations of KB (0, 0.3, 1, 3, 10 and 30 μM) for 2 h and then subjected to H/R insults. The protective effects of KB and its underlying mechanisms were explored. RESULTS KB significantly elevated cell viability (1 μM, 1.21-fold; 3 μM, 1.36-fold, and 10 μM, 1.47-fold) and suppressed LDH release (1 μM, 0.77-fold; 3 μM, 0.68-fold, and 10 μM, 0.59-fold) in H/R-induced H9c2 cells. Further, 10 μM KB blocked apoptotic cascades, as shown by the Annexin-V/PI (0.41-fold), DNA fragmentation (0.51-fold), caspase-3 (0.52-fold), PARP activation (0.27-fold) and Bax/Bcl-2 expression (0.28-fold) assays. KB (10 μM) downregulated reactive oxygen species production (0.51-fold) and lipid peroxidation (0.48-fold); it upregulated the activities of GSH-Px (2.08-fold) and SOD (1.72-fold). KB (10 μM) induced Nrf2 nuclear accumulation (1.94-fold) and increased ARE promoter activity (2.15-fold), HO-1 expression (3.07-fold), AKT (3.07-fold) and AMPK (3.07-fold) phosphorylation. Nrf2 knockdown via using Nrf2 siRNA abrogated KB-mediated protective effects against H/R insults. Moreover, pharmacological inhibitors of AKT and AMPK also abrogated KB-induced Nrf2 activation and its protective function. DISCUSSION AND CONCLUSIONS KB prevented H/R-induced cardiomyocyte injury via modulating the AKT and AMPK-mediated Nrf2 induction. KB might be a promising drug candidate for managing ischemic cardiac disorders.
Collapse
Affiliation(s)
- Qian Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuan-Ye Dang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiu Luo
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ji-Jun Fu
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhi-Cong Zou
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xue-Jing Jia
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Guo-Dong Zheng
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Chu-Wen Li
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
Luo J, Huang Y, Chen Y, Yuan Y, Li G, Cai S, Jian J, Yang S. Heme Oxygenase-1 Is Involved in the Repair of Oxidative Damage Induced by Oxidized Fish Oil in Litopenaeus vannamei by Sulforaphane. Mar Drugs 2023; 21:548. [PMID: 37888483 PMCID: PMC10607972 DOI: 10.3390/md21100548] [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: 08/31/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/28/2023] Open
Abstract
Heme oxygenase-1 (HO-1), which could be highly induced under the stimulation of oxidative stress, functions in reducing the damage caused by oxidative stress, and sulforaphane (SFN) is an antioxidant. This study aims to investigate whether HO-1 is involved in the repair of oxidative damage induced by oxidized fish oil (OFO) in Litopenaeus vannamei by sulforaphane (SFN). The oxidative stress model of L. vannamei was established by feeding OFO feed (OFO accounts for 6%), and they were divided into the following four groups: control group (injected with dsRNA-EGFP and fed with common feed), dsRNA-HO-1 group (dsRNA-HO-1, common feed), dsRNA-HO-1 + SFN group (dsRNA-HO-1, supplement 50 mg kg-1 SFN feed), and SFN group (dsRNA-EGFP, supplement 50 mg kg-1 SFN feed). The results showed that the expression level of HO-1 in the dsRNA-HO-1 + SFN group was significantly increased compared with the dsRNA-HO-1 group (p < 0.05). The activities of SOD in muscle and GPX in hepatopancreas and serum of the dsRNA-HO-1 group were significantly lower than those of the control group, and MDA content in the dsRNA-HO-1 group was the highest among the four groups. However, SFN treatment increased the activities of GPX and SOD in hepatopancreas, muscle, and serum and significantly reduced the content of MDA (p < 0.05). SFN activated HO-1, upregulated the expression of antioxidant-related genes (CAT, SOD, GST, GPX, Trx, HIF-1α, Nrf2, prx 2, Hsp 70), and autophagy genes (ATG 3, ATG 5), and stabilized the expression of apoptosis genes (caspase 2, caspase 3) in the hepatopancreas (p < 0.05). In addition, knocking down HO-1 aggravated the vacuolation of hepatopancreas and increased the apoptosis of hepatopancreas, while the supplement of SFN could repair the vacuolation of hepatopancreas and reduce the apoptosis signal. In summary, HO-1 is involved in the repair of the oxidative damage induced by OFO in L. vannamei by SFN.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shiping Yang
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals, Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (Y.H.); (Y.C.); (Y.Y.); (G.L.); (S.C.); (J.J.)
| |
Collapse
|
5
|
Zhang J, Dong Y, Liu X, Jin H, Wang S, An N, Wang L. Effective myocardial infarction treatment by targeted accumulation of Sulforaphane using porous magnetic silica nanoparticles. Int J Pharm 2023; 645:123389. [PMID: 37714315 DOI: 10.1016/j.ijpharm.2023.123389] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/17/2023]
Abstract
Myocardial infarction (MI) is a common cardiovascular pathology that induces extensive sterile inflammation during its early stages, posing a severe threat to human health. Effectively modulating cardiac inflammation may improve post-MI outcomes. Unfortunately, owing to the side effects of therapeutic drugs and cardiac coronary artery occlusion, current MI drugs are sub-optimal for the clinical management of ischemic myocardia. Sulforaphane (SFN) has been adopted for MI treatment due to its myocardial protective effects and low toxicity. However, the targeted accumulation of SFN in infarcted areas remains challenging. Herein, porous magnetic silica nanoparticles (PMSNs) were synthesized and loaded with SFN to improve the specificity of targeted SFN delivery to infarcted areas in mouse models of MI. PMSNs loaded with SFN (PMSNs + SFN) decreased the levels of pro-inflammatory cytokines, thus leading to the improvement of cardiac function and cell survival without adverse effects. To further explore SFN's mechanisms of action in MI, a cellular (in vitro) model was established via oxygen and glucose deprivation (OGD). HSF1 and Nrf2 knockdown resulted in a decrease of SFN-induced HSP70 expression in OGD cells. Moreover, as a result of HSP70 knockdown, the pro-survival and anti-inflammatory effects of SFN were blocked in OGD cells. The level of pro-inflammatory cytokines decreased upon HSP70 overexpression, and cell survival rate increased under OGD conditions. In summary, the results confirm that PMSNs are capable of transporting SFN to infarcted areas in the myocardium, where the drug exerts cardioprotective effects against myocardial injury by up-regulating HSP70 through Nrf2/HSF1.
Collapse
Affiliation(s)
- Jian Zhang
- Biofunctional Experiment Teaching Center, Harbin Medical University, Harbin 150081, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Yanyan Dong
- Department of Cell Biology, Harbin Medical University, Harbin 150081, China
| | - Xue Liu
- Department of Pharmacology, Harbin Medical University, Harbin 150081, China
| | - Hongbo Jin
- Biofunctional Experiment Teaching Center, Harbin Medical University, Harbin 150081, China
| | - Shuyuan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Na An
- Heilongjiang Medical Academy, Harbin Medical University, Harbin 150081, China.
| | - Lei Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China.
| |
Collapse
|
6
|
Zhang L, Wang S, Zhang Y, Li F, Yu C. Sulforaphane alleviates lung ischemia‑reperfusion injury through activating Nrf‑2/HO‑1 signaling. Exp Ther Med 2023; 25:265. [PMID: 37206558 PMCID: PMC10189751 DOI: 10.3892/etm.2023.11964] [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: 08/22/2022] [Accepted: 03/10/2023] [Indexed: 05/21/2023] Open
Abstract
Oxidative stress and inflammation are both involved in the pathogenesis of lung ischemia-reperfusion (I/R) injury. Sulforaphane (SFN) is a natural product with cytoprotective, anti-inflammatory, and antioxidant properties. The present study hypothesized that SFN may protect against lung I/R injury via the regulation of antioxidant and anti-inflammatory-related pathways. A rat model of lung I/R injury was established, and rats were randomly divided into 3 groups: Sham group, I/R group, and SFN group. It was shown that SFN protected against a pathological inflammatory response via inhibition of neutrophil accumulation and in the reduction of the serum levels of the pro-inflammatory cytokines, IL-6, IL-1β, and TNF-α. SFN treatment also significantly inhibited lung reactive oxygen species production, decreased the levels of 8-OH-dG and malondialdehyde, and reversed the decrease in the antioxidant activities of the enzymes catalase, superoxide dismutase, and glutathione peroxidase in the lungs of the I/R treated rats. In addition, SFN ameliorated I/R-induced lung apoptosis in rats by suppressing Bax and cleaved caspase-3 levels and increased Bcl-2 expression. Furthermore, SFN treatment activated an Nrf2-related antioxidant pathway, as indicated by the increased nuclear transfer of Nrf2 and the downstream HO-1 and NADPH quinone oxidoreductase-1. In conclusion, these findings suggested that SFN protected against I/R-induced lung lesions in rats via activation of the Nrf2/HO-1 pathway and the accompanied anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Respiratory and Critical Care Medicine, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Shuxian Wang
- Department of Respiratory, Yantai Beihai Hospital, Yantai, Shandong 265701, P.R. China
| | - Ying Zhang
- Department of Emergency, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Fenghuan Li
- Department of Respiratory and Critical Care Medicine, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Chaoxiao Yu
- Department of Respiratory and Critical Care Medicine, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
- Correspondence to: Dr Chaoxiao Yu, Department of Respiratory and Critical Care Medicine, Yantaishan Hospital, 10,087 Keji Road, Laishan, Yantai, Shandong 264001, P.R. China
| |
Collapse
|
7
|
Zhang J, Li D, Zhong D, Zhou Q, Yin Y, Gao J, Peng C. Processed lateral root of Aconitum carmichaelii Debx.: A review of cardiotonic effects and cardiotoxicity on molecular mechanisms. Front Pharmacol 2022; 13:1026219. [PMID: 36324672 PMCID: PMC9618827 DOI: 10.3389/fphar.2022.1026219] [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: 08/23/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Fuzi, the lateral root of A. carmichaelii Debx., is a typical traditional herbal medicine with both poisonousness and effectiveness, and often used in the treatment of heart failure and other heart diseases. In this review, we searched domestic and foreign literature to sort out the molecular mechanisms of cardiotonic and cardiotoxicity of Fuzi, also including its components. The major bioactive components of Fuzi for cardiotonic are total alkaloids, polysaccharide and the water-soluble alkaloids, with specific mechanisms manifested in the inhibition of myocardial fibrosis, apoptosis and autophagy, and improvement of mitochondrial energy metabolism, which involves RAAS system, PI3K/AKT, JAK/STAT, AMPK/mTOR signaling pathway, etc. Diester-diterpenoid alkaloids in Fuzi can produce cardiotoxic effects by over-activating Na+ and Ca2+ ion channels, over-activating NLRP3/ASC/caspase-3 inflammatory pathway and mitochondria mediated apoptosis pathway. And three clinically used preparations containing Fuzi are also used as representatives to summarize their cardiac-strengthening molecular mechanisms. To sum up, Fuzi has shown valuable cardiotonic effects due to extensive basic and clinical studies, but its cardiotonic mechanisms have not been systematically sorted out. Therefore, it is a need for deeper investigation in the mechanisms of water-soluble alkaloids with low content but obvious therapeutic effect, as well as polysaccharide.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Li
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Zhong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinmei Zhou
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanpeng Yin
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jihai Gao
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Jihai Gao, ; Cheng Peng,
| | - Cheng Peng
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Jihai Gao, ; Cheng Peng,
| |
Collapse
|
8
|
Lysophosphatidic acid protects cervical cancer HeLa cells from apoptosis induced by doxorubicin hydrochloride. Oncol Lett 2022; 24:267. [PMID: 35782896 PMCID: PMC9247665 DOI: 10.3892/ol.2022.13387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 05/27/2022] [Indexed: 11/11/2022] Open
Abstract
Cervical cancer is one of the most common types of gynecological tumors. Lysophosphatidic acid (LPA), as a bioactive lipid medium, plays an important role in numerous physiological and pathophysiological processes, including the stimulation of cell migration and tumor cell invasion. LPA is increased in the plasma of patients with cervical cancer. Doxorubicin hydrochloride (DOX) is used as a first-line drug in the treatment of cervical cancer in clinics, however, the effect and molecular mechanism of LPA on DOX-induced apoptosis in cervical cancer cells remain unclear. Therefore, the present study aimed to explore the effect and underlying molecular mechanism of LPA on DOX-induced apoptosis in cervical cancer cells. HeLa cells were treated as a control group or with LPA (10 µmol/l), DOX (4 µmol/l) or LPA (10 µmol/l) + DOX (4 µmol/l) for 24 h. Using transmission electron microscopy the results demonstrated that LPA reduced cell death and the degree of chromatin aggregation in DOX-induced HeLa cells. Reverse transcription-quantitative PCR demonstrated that LPA significantly downregulated caspase-3 mRNA expression levels in DOX-induced HeLa cells. Moreover, western blotting demonstrated that LPA significantly reduced caspase-3 and cleaved caspase-3 protein expression levels in DOX-induced HeLa, C33A and SiHa cells. Furthermore, flow cytometry demonstrated that LPA may prevent apoptosis in DOX-induced HeLa cells (P<0.05). Using dichloro-dihydro-fluorescein diacetate assay, it was demonstrated that LPA significantly reduced the intracellular ROS levels induced by DOX. In summary, the present study indicated that LPA may protect HeLa cells from apoptosis induced by DOX. These findings have provided experimental evidence that LPA may be a potential therapeutic target for the treatment of cervical cancer.
Collapse
|
9
|
Zhang J, Dong Y, Zhou M, Wo X, Niu S, Shao E, Liu X, Jin H, Zhao W. Sulforaphane protects myocardium from ischemia-reperfusion injury by regulating CaMKIIN2 and CaMKIIδ. Biochem Biophys Res Commun 2022; 605:119-126. [DOI: 10.1016/j.bbrc.2022.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 11/02/2022]
|
10
|
Dana AH, Alejandro SP. Role of sulforaphane in endoplasmic reticulum homeostasis through regulation of the antioxidant response. Life Sci 2022; 299:120554. [PMID: 35452639 DOI: 10.1016/j.lfs.2022.120554] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 02/09/2023]
Abstract
Nowadays, the nutraceutical agent sulforaphane (SFN) shows great versatility in turning on different cellular responses. Mainly, this isothiocyanate acts as a master regulator of cellular homeostasis due to its antioxidant response and cytoplasmic, mitochondrial, and endoplasmic reticulum (ER) protein modulation. Even more, SFN acts as an effective strategy to counteract oxidative stress, apoptosis, and ER stress, among others as seen in different injury models. Particularly, ER stress is buffered by the unfolded protein response (UPR) activation, which is the first instance in orchestrating the recovery of ER function. Interestingly, different studies highlight a close interrelationship between ER stress and oxidative stress, two events driven by the accumulation of reactive oxygen species (ROS). This response inevitably perpetuates itself and acts as a vicious cycle that triggers the development of different pathologies, such as cardiovascular diseases, neurodegenerative diseases, and others. Accordingly, it is vital to target ER stress and oxidative stress to increase the effectiveness of clinical therapies used to treat these diseases. Therefore, our study is focused on the role of SFN in preserving cellular homeostasis balance by regulating the ER stress response through the Nrf2-modulated antioxidant pathway.
Collapse
Affiliation(s)
- Arana-Hidalgo Dana
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Silva-Palacios Alejandro
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico.
| |
Collapse
|
11
|
Kamal RM, Abdull Razis AF, Mohd Sukri NS, Perimal EK, Ahmad H, Patrick R, Djedaini-Pilard F, Mazzon E, Rigaud S. Beneficial Health Effects of Glucosinolates-Derived Isothiocyanates on Cardiovascular and Neurodegenerative Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030624. [PMID: 35163897 PMCID: PMC8838317 DOI: 10.3390/molecules27030624] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/17/2022]
Abstract
Neurodegenerative diseases (NDDs) and cardiovascular diseases (CVDs) are illnesses that affect the nervous system and heart, all of which are vital to the human body. To maintain health of the human body, vegetable diets serve as a preventive approach and particularly Brassica vegetables have been associated with lower risks of chronic diseases, especially NDDs and CVDs. Interestingly, glucosinolates (GLs) and isothiocyanates (ITCs) are phytochemicals that are mostly found in the Cruciferae family and they have been largely documented as antioxidants contributing to both cardio- and neuroprotective effects. The hydrolytic breakdown of GLs into ITCs such as sulforaphane (SFN), phenylethyl ITC (PEITC), moringin (MG), erucin (ER), and allyl ITC (AITC) has been recognized to exert significant effects with regards to cardio- and neuroprotection. From past in vivo and/or in vitro studies, those phytochemicals have displayed the ability to mitigate the adverse effects of reactive oxidation species (ROS), inflammation, and apoptosis, which are the primary causes of CVDs and NDDs. This review focuses on the protective effects of those GL-derived ITCs, featuring their beneficial effects and the mechanisms behind those effects in CVDs and NDDs.
Collapse
Affiliation(s)
- Ramla Muhammad Kamal
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Department of Pharmacology, Federal University Dutse, Dutse 720101, Jigawa State, Nigeria
| | - Ahmad Faizal Abdull Razis
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Correspondence:
| | - Nurul Syafuhah Mohd Sukri
- Faculty of Applied Science and Technology, Universiti Tun Hussein Onn Malaysia, Batu Pahat 86400, Johor, Malaysia;
| | - Enoch Kumar Perimal
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Hafandi Ahmad
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Rollin Patrick
- Université d’Orléans et CNRS, ICOA, UMR 7311, BP 6759, CEDEX 02, F-45067 Orléans, France;
| | - Florence Djedaini-Pilard
- LG2A UMR 7378, Université de Picardie Jules Verne, 33 rue Saint Leu—UFR des Sciences, F-80000 Amiens, France; (F.D.-P.); (S.R.)
| | - Emanuela Mazzon
- Laboratorio di Neurologia Sperimentale, IRCCS Centro Neurolesi "Bonino Pulejo", 98124 Messina, Italy;
| | - Sébastien Rigaud
- LG2A UMR 7378, Université de Picardie Jules Verne, 33 rue Saint Leu—UFR des Sciences, F-80000 Amiens, France; (F.D.-P.); (S.R.)
| |
Collapse
|
12
|
Zhen X, Jindong L, Yang Z, Yashi R, Wei G, Wei J, Wei Z, Sudong L. Activation of Nrf2 Pathway by Dimethyl Fumarate Attenuates Renal Ischemia-Reperfusion Injury. Transplant Proc 2021; 53:2133-2139. [PMID: 34426023 DOI: 10.1016/j.transproceed.2021.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/19/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Dimethyl fumarate (DMF) is a novel antioxidant that selectively reduces hydroxyl radicals. This study aimed to investigate the potential role of DMF in the pathogenesis of renal ischemia-reperfusion injury (IRI) and the mechanisms involved. METHODS C57BL/6 wild-type mice were treated with DMF or a vehicle. Subsequently, renal IRI was induced in mice by a model of right kidney nephrectomy and left renal ischemia for 30 minutes followed by reperfusion for 24 hours. Sham operation and phosphate-buffered saline were used as controls. Serum and renal tissues were collected at 24 hours after IRI to evaluate the influence of DMF on the recovery of renal function after IRI. Blood urea nitrogen and serum creatinine levels were measured. Kidney cell apoptosis was evaluated using terminal deoxynucleotidyl transferase dUTP nick end labeling-positive staining. Interleukin 6 and tumor necrosis factor α cytokines in the kidney tissues were measured. Indicators of oxidative stress in the kidneys were detected. Finally, Nrf2-deficient mice were used to determine the protective role of the nuclear factor erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) and NAD(P)H dehydrogenase quinone 1 (NQO1) signaling pathways induced by DMF using western blot assay. RESULTS DMF significantly attenuated renal dysfunction in mice and showed reductions in the severity of renal tubular injury, cell necrosis, and apoptosis. Moreover, DMF significantly reduced the amount of key inflammatory mediators. Additionally, DMF attenuated the malondialdehyde levels 24 hours after IRI but upregulated the superoxide dismutase activities. Western blot assay showed that DMF significantly increased the protein levels of Nrf2, HO-1, and NQO-1. Importantly, these DMF-mediated beneficial effects were not observed in Nrf2-deficient mice. CONCLUSIONS DMF attenuates renal IRI by reducing inflammation and upregulating the antioxidant capacity, which may be through Nrf2/HO-1and NQO1 signaling pathway.
Collapse
Affiliation(s)
- Xu Zhen
- Department of Urology, Taizhou People's Hospital, Taizhou, China
| | - Li Jindong
- Department of Pharmacy, Taizhou People's Hospital, Taizhou, China
| | - Zhou Yang
- Department of Pathology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Ruan Yashi
- Department of Urology, Taizhou People's Hospital, Taizhou, China
| | - Guo Wei
- Department of Urology, Taizhou People's Hospital, Taizhou, China
| | - Jiang Wei
- Department of Urology, Taizhou People's Hospital, Taizhou, China
| | - Zhang Wei
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Sudong
- Department of Urology, Taizhou People's Hospital, Taizhou, China.
| |
Collapse
|
13
|
Zheng W, Xie Q, Zhang Z, Li J, Fang L, Li W. Inhibited HDAC3 or Elevated MicroRNA-494-3p Plays a Protective Role in Myocardial Ischemia-Reperfusion Injury via Suppression of BRD4. Mol Neurobiol 2021; 58:4268-4279. [PMID: 33982231 DOI: 10.1007/s12035-021-02369-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/22/2021] [Indexed: 11/29/2022]
Abstract
Increased histone deacetylase 3 (HDAC3) has been demonstrated to contribute to the pathogenesis of myocardial ischemia-reperfusion injury (MI/RI). Therefore, the goal of this study was to investigate how HDAC3 regulated MI/RI by mediating microRNA (miR)-494-3p/dromodomain-containing protein 4 (BRD4) axis. The MI/RI model was established by ligating the right anterior descending coronary artery. Cardiomyocytes from newborn mice were treated with hypoxia/reoxygenation (H/R). Gain-of-function and loss-of-function approaches were implemented to figure out the roles of miR-494-3p and HDAC3 in MI/RI. miR-494-3p, HDAC3, and BRD4 in myocardial tissues of mice with MI/RI and H/R-treated cardiomyocytes were detected. The relationships between miR-494-3p and HDAC3 and BRD4 were identified. Reduced miR-494-3p and upregulated HDAC3 and BRD4 exhibited in myocardial tissues of mice with MI/RI and H/R-treated cardiomyocytes. Inhibited HDAC3 or elevated miR-494-3p repressed the inflammation and apoptosis, improved cardiac function, and ameliorated myocardial injury in myocardial tissues of mice with MI/RI. Suppression of HDAC3 or elevation of miR-494-3p depressed inflammation and apoptosis and promoted cell viability of primary cardiomyocytes. miR-494-3p targeted BRD4. The study concludes that suppressed HDAC3 plays a protective role in MI/RI by upregulation of miR-494-3p and inhibition of BRD4, which could be helpful for MI/RI therapy.
Collapse
Affiliation(s)
- Wuyang Zheng
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Siming District, Xiamen, 361000, Fujian, China
| | - Qiang Xie
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Siming District, Xiamen, 361000, Fujian, China
| | - Ziguan Zhang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Siming District, Xiamen, 361000, Fujian, China
| | - Jun Li
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Siming District, Xiamen, 361000, Fujian, China
| | - Lihuan Fang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Siming District, Xiamen, 361000, Fujian, China
| | - Weihua Li
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Road, Siming District, Xiamen, 361000, Fujian, China.
| |
Collapse
|