1
|
Mao T, Jiang K, Pang Y, Pan Y, Jia W, Gao Q, Lin Q. Hydroxysafflor yellow A for ischemic heart diseases: a systematic review and meta-analysis of animal experiments. Front Pharmacol 2025; 16:1510657. [PMID: 40271057 PMCID: PMC12014549 DOI: 10.3389/fphar.2025.1510657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
Background Hydroxysafflor yellow A (HSYA) possesses a variety of pharmacological activities which has been demonstrated to be effective against ischemic heart disease (IHD). This study aimed to comprehensively examine the efficacy and summarize the potential mechanisms of HSYA against IHD in animal models. Methods We conducted electronic searches for preclinical studies on PubMed, Embase, Web of Science, Cochrane Library, CNKI, SinoMed, Wanfang, and Chinese VIP databases from inception to 31 January 2024. The CAMARADES checklist was chosen to assess the quality of evidence. STATA 14.0 software was utilized to analyze the data. The underlying mechanisms were categorized and summarized. Results Twenty-eight studies involving 686 rodents were included and the mean score of methodology quality was 5.04 (range from 4 to 7). Meta-analysis observed that HSYA could decrease myocardial infarction size (SMD: -2.82, 95%CI: -3.56 to -2.08, p < 0.001) and reduce the levels of biomarkers of myocardial injury including cTnI (SMD: -3.82, 95%CI: -5.20 to -2.44, p < 0.001) and CK-MB (SMD: -2.74, 95%CI: -3.58 to -1.91, p < 0.001). HSYA displayed an improvement in cardiac function indicators including LVEF, LVSP, +dp/dt max and -dp/dt max. Furthermore, HSYA was able to reduce the levels of MDA, TNF-α and IL-6, while increasing SOD and NO levels. Mechanistically, the protective effect of HSYA in alleviating myocardial injury after ischemia may be associated with NLRP3 inflammasome, Bcl-2, Bax, caspase-3, eNOS proteins, and TLR/NF-κB, Nrf2/HO-1, JAK/STAT, PI3K/Akt, AMPK/mTOR, VEGFA pathways. Conclusion This study demonstrates that HSYA exerts cardioprotective effects in decreasing infarct size, reducing myocardial enzymes and improving cardiac function, which may be mediated by anti-inflammatory, antioxidant, anti-apoptotic, regulation of autophagy, improvement of microcirculation and promotion of angiogenesis. However, the absence of safety assessment, lack of animal models of co-morbidities, and inconsistency between timing of administration and clinical practice are limitations of preclinical studies. Systematic Review Registration clinicaltrials.gov, Identifier, CRD42023460790.
Collapse
Affiliation(s)
- Tianshi Mao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kaixin Jiang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanting Pang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Pan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wenhao Jia
- Department of Cardiology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Qun Gao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Lin
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Ren Z, Su R, Liu D, Wang Q, Liu S, Kong D, Qiu Y. Yes-associated protein indispensably mediates hirsutine-induced inhibition on cell growth and Wnt/β-catenin signaling in colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156156. [PMID: 39437684 DOI: 10.1016/j.phymed.2024.156156] [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: 07/04/2024] [Revised: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND PURPOSE Targeting Wnt/β-catenin signaling emerges as one of the promising strategies for colorectal cancer (CRC) treatment, as this signaling is highly activated in CRC progression. Despite reports on the cytotoxic effects of hirsutine (HT), an indole alkaloid found in herbal medicines from the genus Uncaria, its therapeutic potential for CRC and the involved mechanisms are poorly understood. This study investigates the anticancer efficacy and the probable mechanisms of HT against CRC. METHODS To evaluate in vitro anticancer activity of HT, cell growth examined by MTT and colony formation assay, and apoptosis examined by flow cytometry were analyzed. To explore the mechanisms, RNA-sequencing, western blotting, dual-luciferase reporter assays, immunofluorescence, and co-immunoprecipitation were performed. Mouse model of azoxymethane/dextran sodium sulfate (AOM/DSS)-induced colon cancer was utilized to assess HT's in vivo anticancer efficacy. RESULTS HT significantly inhibited CRC cell proliferation with IC50 values of 22.25 ± 3.27 μM for SW620 cells and 22.24 ± 2.36 μM for HCT116 cells, and induced apoptosis. HT decreased protein levels of Wnt3a and β-catenin dose- and time-dependently, and inhibited TOP/FOP FLASH reporter activity, nuclear travel of β-catenin, and downstream targets like c-Myc, Cyclin D1, VEGF. HT reduced β-catenin protein half-life, and the reversal of this effect by MG132 indicated that HT facilitated proteasome-dependent degradation of β-catenin in these two cell lines. HT also increased β-catenin ubiquitination without affecting Axin and β-TrCP levels. HT treatment for 24 h induced YAP cytoplasmic retention, enhanced YAP interacting with β-catenin and β-TrCP, triggering destruction complex formation and β-catenin ubiquitination and degradation, while YAP siRNA impaired these effects. Additionally, β-catenin overexpression and LiCl treatment counteracted HT-induced inhibition on cell growth and Wnt/β-catenin cascade. In model of AOM/DSS-induced mouse colon cancer, compared with AOM/DSS treatment group, HT recovered colon length, reduced tumor numbers and radius, and downregulated β-catenin and Ki-67, while upregulated cleaved PARP in the colorectal tissue with tumors. CONCLUSION HT exhibits anticancer activity against CRC probably by inhibiting Wnt/β-catenin signaling, with YAP playing an indispensible role during the process, highlighting HT as a potential novel candidate drug for CRC therapy.
Collapse
Affiliation(s)
- Zehao Ren
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ruixin Su
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Donghui Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Qian Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Shanshan Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Yuling Qiu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
3
|
Xu Y, Shao L, Zhou Z, Zhao L, Wan S, Sun W, Wanyan W, Yuan Y. ARG2 knockdown promotes G0/G1 cell cycle arrest and mitochondrial dysfunction in adenomyosis via regulation NF-κB and Wnt/Β-catenin signaling cascades. Int Immunopharmacol 2024; 140:112817. [PMID: 39116499 DOI: 10.1016/j.intimp.2024.112817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/28/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Adenomyosis is a common gynecological disease, characterized by overgrowth of endometrial glands and stroma in the myometrium, however its exact pathophysiology still remains uncertain. Emerging evidence has demonstrated the elevated level of arginase 2 (ARG2) in endometriosis and adenomyosis. This study aimed to determine whether ARG2 involved in mitochondrial function and epithelial to mesenchymal transition (EMT) in adenomyosis and its potential underlying mechanisms. MATERIALS AND METHODS RNA interference was used to inhibit ARG2 gene, and then Cell Counting Kit (CCK-8) assay and flow cytometery were performed to detect the cell proliferation capacity, cell cycle, and apoptosis progression, respectively. The mouse adenomyosis model was established and RT-PCR, Western blot analysis, mitochondrial membrane potential (Δψm) detection and mPTP opening evaluation were conducted. RESULTS Silencing ARG2 effectively down-regulated its expression at the mRNA and protein levels in endometrial cells, leading to decreased enzyme activity and inhibition of cell viability. Additionally, ARG2 knockdown induced G0/G1 cell cycle arrest, promoted apoptosis, and modulated the expression of cell cycle- and apoptosis-related regulators. Notably, the interference with ARG2 induces apoptosis by mitochondrial dysfunction, ROS production, ATP depletion, decreasing the Bcl-2/Bax ratio, releasing Cytochrome c, and increasing the expression of Caspase-9/-3 and PARP. In vivo study in a mouse model of adenomyosis demonstrated also elevated levels of ARG2 and EMT markers, while siARG2 treatment reversed EMT and modulated inflammatory cytokines. Furthermore, ARG2 knockdown was found to modulate the NF-κB and Wnt/β-catenin signaling pathways in mouse adenomyosis. CONCLUSION Consequently, ARG2 silencing could induce apoptosis through a mitochondria-dependent pathway mediated by ROS, and G0/G1 cell cycle arrest via suppressing NF-κB and Wnt/β-catenin signaling pathways in Ishikawa cells. These findings collectively suggest that ARG2 plays a crucial role in the pathogenesis of adenomyosis and may serve as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Yaping Xu
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China; State Key Laboratory of Ultrasound in Medicine and Engineering, No.1 Medical College Road, Yuzhong District, Chongqing, China
| | - Lin Shao
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China; State Key Laboratory of Ultrasound in Medicine and Engineering, No.1 Medical College Road, Yuzhong District, Chongqing, China
| | - Zhan Zhou
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Liying Zhao
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Shuquan Wan
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Wenjing Sun
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Wenya Wanyan
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Yinping Yuan
- State Key Laboratory of Ultrasound in Medicine and Engineering, No.1 Medical College Road, Yuzhong District, Chongqing, China; Department of Pathology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250031, China.
| |
Collapse
|
4
|
Letafati A, Bahavar A, Norouzi M, Rasouli A, Hedayatyaghoubi M, Molaverdi G, Mozhgani SH, Siami Z. Effects of HTLV-1 on leukocyte trafficking and migration in ACs compared to healthy individuals. BMC Res Notes 2024; 17:222. [PMID: 39127702 DOI: 10.1186/s13104-024-06887-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) is a RNA virus belonging to Retroviridae family and is associated with the development of various diseases, including adult T-cell leukemia/lymphoma (ATLL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Aside from HAM/TSP, HTLV-1 has been implicated in the development of several disorders that mimic auto-inflammation. T-cell migration is important topic in the context of HTLV-1 associated diseases progression. The primary objective of this case-control study was to assess the relationship between increased mRNA expression in virus migration following HTLV-1 infection. PBMCs from 20 asymptomatic patients and 20 healthy subjects were analyzed using real-time PCR to measure mRNA expression of LFA1, MLCK, RAC1, RAPL, ROCK1, VAV1 and CXCR4. Also, mRNA expression of Tax and HBZ were evaluated. Mean expression of Tax and HBZ in ACs (asymptomatic carriers) was 0.7218 and 0.6517 respectively. The results revealed a noteworthy upregulation of these genes involved in T-cell migration among ACs patients in comparison to healthy individuals. Considering the pivotal role of gene expression alterations associated with the progression into two major diseases (ATLL or HAM/TSP), analyzing the expression of these genes in the ACs group can offer probable potential diagnostic markers and aid in monitoring the condition of ACs.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Atefeh Bahavar
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehdi Norouzi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Aziz Rasouli
- Department of Emergency Medicine, School of Medicine, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Hedayatyaghoubi
- Department of Infectious Diseases, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Ghazale Molaverdi
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Sayed-Hamidreza Mozhgani
- Non-Communicable Disease Research Center, Alborz University of Medical Sciences, Karaj, Iran.
- Department of Microbiology and Virology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| | - Zeinab Siami
- Department of Infectious Diseases, School of Medicine, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Zhang Y, Wang Y, Zhang W, Feng S, Xing Y, Wang T, Huang N, Li K, Zhang A. Comprehensive transcriptomic analysis identifies SLC25A4 as a key predictor of prognosis in osteosarcoma. Front Genet 2024; 15:1410145. [PMID: 38957810 PMCID: PMC11217516 DOI: 10.3389/fgene.2024.1410145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024] Open
Abstract
Background Osteosarcoma (OS) is highly malignant and prone to local infiltration and distant metastasis. Due to the poor outcomes of OS patients, the study aimed to identify differentially expressed genes (DEGs) in OS and explore their role in the carcinogenesis and progression of OS. Methods RNA sequencing was performed to identify DEGs in OS. The functions of the DEGs in OS were investigated using bioinformatics analysis, and DEG expression was verified using RT-qPCR and Western blotting. The role of SLC25A4 was evaluated using gene set enrichment analysis (GSEA) and then investigated using functional assays in OS cells. Results In all, 8353 DEGs were screened. GO and KEGG enrichment analyses indicated these DEGs showed strong enrichment in the calcium signaling pathway and pathways in cancer. Moreover, the Kaplan-Meier survival analysis showed ten hub genes were related to the outcomes of OS patients. Both SLC25A4 transcript and protein expression were significantly reduced in OS, and GSEA suggested that SLC25A4 was associated with cell cycle, apoptosis and inflammation. SLC25A4-overexpressing OS cells exhibited suppressed proliferation, migration, invasion and enhanced apoptosis. Conclusion SLC25A4 was found to be significantly downregulated in OS patients, which was associated with poor prognosis. Modulation of SLC25A4 expression levels may be beneficial in OS treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yinghui Wang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Wenyan Zhang
- School of Food Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Shaojie Feng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tianjiao Wang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Nana Huang
- Department of Neurology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ka Li
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Aijun Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
6
|
Chen S, Zhai D, Li Y, Tan Y, Tang X, Pu X, Chai Y, Li L. Study on the mechanism of inhibition of Escherichia coli by Polygonum capitatum based on network pharmacology and molecular docking technology: A review. Medicine (Baltimore) 2024; 103:e38536. [PMID: 38875382 PMCID: PMC11175921 DOI: 10.1097/md.0000000000038536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 06/16/2024] Open
Abstract
This study aims to analyze the effective components of Polygonum capitatum (PC) inhibiting Escherichia coli based on network pharmacology methods and predict its molecular mechanism of action. PC compounds and targets were collected from the TCMSP database, Swiss Target Prediction, and the literature. E coli targets were searched using the GeneCards database. The targets of E coli and the targets of the active ingredients of PC were taken as intersections to obtain the intersecting targets. The resulting overlapping targets were uploaded to the STRING database to construct the protein interaction network diagram of E coli target inhibition. The key targets for the inhibitory effect of PC on E coli were obtained. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed by uploading key targets into the DAVID database. The results showed that there were 50 targets for PC to inhibit E coli. Among them, there are 5 core targets, mainly including AKT1, TNF, EGFR, JUN, and ESR1. A total of 196 gene ontology functional analysis results and 126 Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis results were obtained. These include cellular response to cadmium-ion, cellular response to reactive oxygen species, pathways in cancer, prostate cancer, and PI3K-Akt signaling pathway. Molecular docking results indicate that Lutedin, Hirsutin, Flazin, and Ellagic acid in PC have high affinity for the target genes AKT1, TNF, MAPK3 and EGFR. PC exerts its inhibitory effect on E coli through multi-targets and multi-pathways, which provides a new basis for the new use of PC as an old medicine.
Collapse
Affiliation(s)
- Shunhuan Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Dongyan Zhai
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yuan Li
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yong Tan
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xiaoke Tang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xiang Pu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yihui Chai
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Lailai Li
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
7
|
Shen X, Ma M, Mi R, Zhuang J, Song Y, Yang W, Li H, Lu Y, Yang B, Liu Y, Wu Y, Shen H. EFHD1 promotes osteosarcoma proliferation and drug resistance by inhibiting the opening of the mitochondrial membrane permeability transition pore (mPTP) by binding to ANT3. Cell Mol Life Sci 2024; 81:236. [PMID: 38795203 PMCID: PMC11127909 DOI: 10.1007/s00018-024-05254-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/27/2024]
Abstract
Chemoresistance is the main obstacle in the clinical treatment of osteosarcoma (OS). In this study, we investigated the role of EF-hand domain-containing protein 1 (EFHD1) in OS chemotherapy resistance. We found that the expression of EFHD1 was highly correlated with the clinical outcome after chemotherapy. We overexpressed EFHD1 in 143B cells and found that it increased their resistance to cell death after drug treatment. Conversely, knockdown of EFHD1 in 143BR cells (a cisplatin-less-sensitive OS cell line derived from 143B cells) increased their sensitivity to treatment. Mechanistically, EFHD1 bound to adenine nucleotide translocase-3 (ANT3) and inhibited its conformational change, thereby inhibiting the opening of the mitochondrial membrane permeability transition pore (mPTP). This effect could maintain mitochondrial function, thereby favoring OS cell survival. The ANT3 conformational inhibitor carboxyatractyloside (CATR), which can promote mPTP opening, enhanced the chemosensitivity of EFHD1-overexpressing cells when combined with cisplatin. The ANT3 conformational inhibitor bongkrekic acid (BKA), which can inhibit mPTP opening, restored the resistance of EFHD1 knockdown cells. In conclusion, our results suggest that EFHD1-ANT3-mPTP might be a promising target for OS therapy in the future.
Collapse
Affiliation(s)
- Xin Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Mengjun Ma
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Rujia Mi
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Jiahao Zhuang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Yihui Song
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Wen Yang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Hongyu Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Yixuan Lu
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Biao Yang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Yinliang Liu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China
| | - Yanfeng Wu
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, No. 3025 Shennan Zhong Road, Shenzhen, 518033, Guangdong, China.
| |
Collapse
|
8
|
Zhang JW, Huang LY, Li YN, Tian Y, Yu J, Wang XF. Mitochondrial carrier homolog 2 increases malignant phenotype of human gastric epithelial cells and promotes proliferation, invasion, and migration of gastric cancer cells. World J Gastrointest Oncol 2024; 16:991-1005. [PMID: 38577443 PMCID: PMC10989370 DOI: 10.4251/wjgo.v16.i3.991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 01/19/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND The precise role of mitochondrial carrier homolog 2 (MTCH2) in promoting malignancy in gastric mucosal cells and its involvement in gastric cancer cell metastasis have not been fully elucidated. AIM To determine the role of MTCH2 in gastric cancer. METHODS We collected 65 samples of poorly differentiated gastric cancer tissue and adjacent tissues, constructed MTCH2-overexpressing and MTCH2-knockdown cell models, and evaluated the proliferation, migration, and invasion of human gastric epithelial cells (GES-1) and human gastric cancer cells (AGS) cells. The mitochondrial membrane potential (MMP), mitochondrial permeability transformation pore (mPTP) and ATP fluorescence probe were used to detect mitochondrial function. Mitochondrial function and ATP synthase protein levels were detected via Western blotting. RESULTS The expression of MTCH2 and ATP2A2 in gastric cancer tissues was significantly greater than that in adjacent tissues. Overexpression of MTCH2 promoted colony formation, invasion, migration, MMP expression and ATP production in GES-1 and AGS cells while upregulating ATP2A2 expression and inhibiting cell apoptosis; knockdown of MTCH2 had the opposite effect, promoting overactivation of the mPTP and promoting apoptosis. CONCLUSION MTCH2 can increase the malignant phenotype of GES-1 cells and promote the proliferation, invasion, and migration of gastric cancer cells by regulating mitochondrial function, providing a basis for targeted therapy for gastric cancer cells.
Collapse
Affiliation(s)
- Jing-Wen Zhang
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling-Yan Huang
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ya-Ning Li
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ying Tian
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jia Yu
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Xiao-Fei Wang
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei Province, China
| |
Collapse
|
9
|
Wang H, Liu J, Zhang Z, Peng J, Wang Z, Yang L, Wang X, Hu S, Hong L. β-Sitosterol targets ASS1 for Nrf2 ubiquitin-dependent degradation, inducing ROS-mediated apoptosis via the PTEN/PI3K/AKT signaling pathway in ovarian cancer. Free Radic Biol Med 2024; 214:137-157. [PMID: 38364944 DOI: 10.1016/j.freeradbiomed.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 02/18/2024]
Abstract
The exploration of drugs derived from natural sources holds significant promise in addressing current limitations in ovarian cancer (OC) treatments. While previous studies have highlighted the remarkable anti-cancer properties of the natural compound β-sitosterol (SIT) across various tumors, its specific role in OC treatment remains unexplored. This study aims to investigate the anti-tumor activity of SIT in OC using in vitro and in vivo models, delineate potential mechanisms, and establish a preclinical theoretical foundation for future clinical trials, thus fostering further research. Utilizing network pharmacology, we pinpoint SIT as a promising candidate for OC treatment and predict its potential targets and pathways. Through a series of in vitro and in vivo experiments, we unveil a novel mechanism through which SIT mitigates the malignant biological behaviors of OC cells by modulating redox status. Specifically, SIT selectively targets argininosuccinate synthetase 1 (ASS1), a protein markedly overexpressed in OC tissues and cells. Inhibiting ASS1, SIT enhances the interaction between Nrf2 and Keap1, instigating the ubiquitin-dependent degradation of Nrf2, subsequently diminishing the transcriptional activation of downstream antioxidant genes HO-1 and NQO1. The interruption of the antioxidant program by SIT results in the substantial accumulation of reactive oxygen species (ROS) in OC cells. This, in turn, upregulates PTEN, exerting negative regulation on the phosphorylation activation of AKT. The suppression of AKT signaling disrupted downstream pathways associated with cell cycle, cell survival, apoptosis, migration, and invasion, ultimately culminating in the death of OC cells. Our research uncovers new targets and mechanisms of SIT against OC, contributing to the existing knowledge on the anti-tumor effects of natural products in the context of OC. Additionally, this research unveils a novel role of ASS1 in regulating the Nrf2-mediated antioxidant program and governing redox homeostasis in OC, providing a deeper understanding of this complex disease.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Jingchun Liu
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Zihui Zhang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Jiaxin Peng
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Zhi Wang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Lian Yang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Xinqi Wang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Siyuan Hu
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| | - Li Hong
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Jiefang Road NO.238, Wuhan, 430060, PR China.
| |
Collapse
|
10
|
Gao R, Zhou D, Qiu X, Zhang J, Luo D, Yang X, Qian C, Liu Z. Cancer Therapeutic Potential and Prognostic Value of the SLC25 Mitochondrial Carrier Family: A Review. Cancer Control 2024; 31:10732748241287905. [PMID: 39313442 PMCID: PMC11439189 DOI: 10.1177/10732748241287905] [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: 05/23/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Transporters of the solute carrier family 25 (SLC25) regulate the intracellular distribution and concentration of nucleotides, amino acids, dicarboxylates, and vitamins within the mitochondrial and cytoplasmic matrices. This mechanism involves changes in mitochondrial function, regulation of cellular metabolism, and the ability to provide energy. In this review, important members of the SLC25 family and their pathways affecting tumorigenesis and progression are elucidated, highlighting the diversity and complexity of these pathways. Furthermore, the significant potential of the members of SLC25 as both cancer therapeutic targets and biomarkers will be emphasized.
Collapse
Affiliation(s)
- Renzhuo Gao
- School of Queen Mary, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dan Zhou
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xingpeng Qiu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiayi Zhang
- School of Queen Mary, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Daya Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaohong Yang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Caiyun Qian
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhuoqi Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
11
|
Yang Y, An Y, Ren M, Wang H, Bai J, Du W, Kong D. The mechanisms of action of mitochondrial targeting agents in cancer: inhibiting oxidative phosphorylation and inducing apoptosis. Front Pharmacol 2023; 14:1243613. [PMID: 37954849 PMCID: PMC10635426 DOI: 10.3389/fphar.2023.1243613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
The tumor microenvironment affects the structure and metabolic function of mitochondria in tumor cells. This process involves changes in metabolic activity, an increase in the amount of reactive oxygen species (ROS) in tumor cells compared to normal cells, the production of more intracellular free radicals, and the activation of oxidative pathways. From a practical perspective, it is advantageous to develop drugs that target mitochondria for the treatment of malignant tumors. Such drugs can enhance the selectivity of treatments for specific cell groups, minimize toxic effects on normal tissues, and improve combinational treatments. Mitochondrial targeting agents typically rely on small molecule medications (such as synthetic small molecules agents, active ingredients of plants, mitochondrial inhibitors or autophagy inhibitors, and others), modified mitochondrial delivery system agents (such as lipophilic cation modification or combining other molecules to form targeted mitochondrial agents), and a few mitochondrial complex inhibitors. This article will review these compounds in three main areas: oxidative phosphorylation (OXPHOS), changes in ROS levels, and endogenous oxidative and apoptotic processes.
Collapse
Affiliation(s)
- Yi Yang
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yahui An
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Mingli Ren
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haijiao Wang
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Bai
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenli Du
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
12
|
Niu J, Guo W, Lu A, Han G, Wang G, Peng B, Zhao J. Comparison with gastric cancer-associated genes reveals the role of ferroptosis-related genes in eosinophils of asthma patients: A bioinformatic study. Medicine (Baltimore) 2023; 102:e35002. [PMID: 37832131 PMCID: PMC10578675 DOI: 10.1097/md.0000000000035002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/08/2023] [Indexed: 10/15/2023] Open
Abstract
Ferroptosis-inducing agents (FIAs) induced lipid-peroxidation-independent ferroptosis in eosinophils, thus ameliorating airway inflammation in asthmatic mice. Differences in ferroptosis-related genes (FerrGs) between eosinophils and cells in which FIAs induce canonical ferroptosis are supposed to contribute to this noncanonical ferroptosis but remain unclear. This study aims to explore these differences. This study used gastric cancer cells (GCCs) in stomach adenocarcinoma as the representative of cells in which FIAs induce canonical ferroptosis. FerrGs in Ferroptosis Database V2 respectively intersected with differentially expressed genes (DEGs) of eosinophils (E-MTAB-4660 dataset) and GCCs (GEPIA2 Stomach adenocarcinoma dataset) to obtain original ferroptosis DEGs (FerrDEGs). Then, they were subjected to Venn analysis to identify FerrDEGs shared by them and FerrDEGs exclusively expressed in eosinophils or GCCs. Identified genes were subjected to functional enrichment analysis, protein-protein interactions analysis, Hub genes analysis, and construction of the LncRNA-mediated ceRNA network. Sixty-six original FerrDEGs in eosinophils and 110 original FerrDEGs in GCCs were obtained. Venn analysis identified that eosinophils and GCCs shared 19 FerrDEGs that presented opposite expression directions and were involved in the ferroptosis pathway. Four upregulated and 20 downregulated FerrDEGs were exclusively expressed in eosinophils and GCCs, respectively. The former were enriched only in glycerolipid metabolism, while the latter were not enriched in pathways. Forty downregulated and 68 upregulated FerrDEGs were solely expressed in eosinophils and GCCs, respectively. The former was associated with the FoxO signaling pathway; the latter was related to glutathione metabolism and they were all implicated in autophagy. PPI analysis shows that the top 10 Hub genes of 66 original FerrDEGs and 44 exclusive FerrDEGs in eosinophils shared 9 genes (STAT3, NFE2L2, MAPK8, PTEN, MAPK3, TLR4, SIRT1, BECN1, and PTGS2) and they were also involved in the FoxO signaling pathway and autophagy pathway. Among them, PTEN is involved in forming a ceRNA network containing 3 LncRNAs, 3 miRNAs and 3 mRNAs. In contrast to FerrGs in cells in which FIAs induce canonical ferroptosis, the FerrGs in eosinophils differ in expression and in the regulation of ferroptosis, FoxO signaling pathway, and autophagy. It lays the groundwork for targeted induction of eosinophils lipid-peroxidation-independent ferroptosis in asthma.
Collapse
Affiliation(s)
- Jianfei Niu
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Wei Guo
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Aiyangzi Lu
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Guanxiong Han
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Guanqun Wang
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Bihui Peng
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Jiping Zhao
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
13
|
Deng RM, Zhou J. The role of PI3K/AKT signaling pathway in myocardial ischemia-reperfusion injury. Int Immunopharmacol 2023; 123:110714. [PMID: 37523969 DOI: 10.1016/j.intimp.2023.110714] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Myocardial ischemia has a high incidence and mortality rate, and reperfusion is currently the standard intervention. However, reperfusion may lead to further myocardial damage, known as myocardial ischemia/reperfusion injury (MIRI). There are currently no effective clinical treatments for MIRI. The PI3K/Akt signaling pathway is involved in cardiovascular health and disease and plays an important role in reducing myocardial infarct size and restoring cardiac function after MIRI. Activation of the PI3K/Akt pathway provides myocardial protection through synergistic upregulation of antioxidant, anti-inflammatory, and autophagy activities and inhibition of mitochondrial dysfunction and cardiomyocyte apoptosis. Many studies have shown that PI3K/Akt has a significant protective effect against MIRI. Here, we reviewed the molecular regulation of PI3K/Akt in MIRI and summarized the molecular mechanism by which PI3K/Akt affects MIRI, the effects of ischemic preconditioning and ischemic postconditioning, and the role of related drugs or activators targeting PI3K/Akt in MIRI, providing novel insights for the formulation of myocardial protection strategies. This review provides evidence of the role of PI3K/Akt activation in MIRI and supports its use as a therapeutic target.
Collapse
Affiliation(s)
- Rui-Ming Deng
- Department of Anesthesiology, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; The Affiliated Ganzhou Hospital of Nanchang University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China
| | - Juan Zhou
- Department of thyroid and Breast Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; The Affiliated Ganzhou Hospital of Nanchang University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| |
Collapse
|
14
|
Bhuia MS, Wilairatana P, Ferdous J, Chowdhury R, Bappi MH, Rahman MA, Mubarak MS, Islam MT. Hirsutine, an Emerging Natural Product with Promising Therapeutic Benefits: A Systematic Review. Molecules 2023; 28:6141. [PMID: 37630393 PMCID: PMC10458569 DOI: 10.3390/molecules28166141] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Fruits and vegetables are used not only for nutritional purposes but also as therapeutics to treat various diseases and ailments. These food items are prominent sources of phytochemicals that exhibit chemopreventive and therapeutic effects against several diseases. Hirsutine (HSN) is a naturally occurring indole alkaloid found in various Uncaria species and has a multitude of therapeutic benefits. It is found in foodstuffs such as fish, seafood, meat, poultry, dairy, and some grain products among other things. In addition, it is present in fruits and vegetables including corn, cauliflower, mushrooms, potatoes, bamboo shoots, bananas, cantaloupe, and citrus fruits. The primary emphasis of this study is to summarize the pharmacological activities and the underlying mechanisms of HSN against different diseases, as well as the biopharmaceutical features. For this, data were collected (up to date as of 1 July 2023) from various reliable and authentic literature by searching different academic search engines, including PubMed, Springer Link, Scopus, Wiley Online, Web of Science, ScienceDirect, and Google Scholar. Findings indicated that HSN exerts several effects in various preclinical and pharmacological experimental systems. It exhibits anti-inflammatory, antiviral, anti-diabetic, and antioxidant activities with beneficial effects in neurological and cardiovascular diseases. Our findings also indicate that HSN exerts promising anticancer potentials via several molecular mechanisms, including apoptotic cell death, induction of oxidative stress, cytotoxic effect, anti-proliferative effect, genotoxic effect, and inhibition of cancer cell migration and invasion against various cancers such as lung, breast, and antitumor effects in human T-cell leukemia. Taken all together, findings from this study show that HSN can be a promising therapeutic agent to treat various diseases including cancer.
Collapse
Affiliation(s)
- Md. Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (R.C.); (M.H.B.)
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman, Science and Technology University, Gopalganj 8100, Bangladesh;
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (R.C.); (M.H.B.)
| | - Mehedi Hasan Bappi
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (R.C.); (M.H.B.)
| | - Md Anisur Rahman
- Department of Pharmacy, Islamic University, Kushtia 7003, Bangladesh;
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (R.C.); (M.H.B.)
| |
Collapse
|
15
|
Waseem M, Wang BD. Promising Strategy of mPTP Modulation in Cancer Therapy: An Emerging Progress and Future Insight. Int J Mol Sci 2023; 24:5564. [PMID: 36982637 PMCID: PMC10051994 DOI: 10.3390/ijms24065564] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Cancer has been progressively a major global health concern. With this developing global concern, cancer determent is one of the most significant public health challenges of this era. To date, the scientific community undoubtedly highlights mitochondrial dysfunction as a hallmark of cancer cells. Permeabilization of the mitochondrial membranes has been implicated as the most considerable footprint in apoptosis-mediated cancer cell death. Under the condition of mitochondrial calcium overload, exclusively mediated by oxidative stress, an opening of a nonspecific channel with a well-defined diameter in mitochondrial membrane allows free exchange between the mitochondrial matrix and the extra mitochondrial cytosol of solutes and proteins up to 1.5 kDa. Such a channel/nonspecific pore is recognized as the mitochondrial permeability transition pore (mPTP). mPTP has been established for regulating apoptosis-mediated cancer cell death. It has been evident that mPTP is critically linked with the glycolytic enzyme hexokinase II to defend cellular death and reduce cytochrome c release. However, elevated mitochondrial Ca2+ loading, oxidative stress, and mitochondrial depolarization are critical factors leading to mPTP opening/activation. Although the exact mechanism underlying mPTP-mediated cell death remains elusive, mPTP-mediated apoptosis machinery has been considered as an important clamp and plays a critical role in the pathogenesis of several types of cancers. In this review, we focus on structure and regulation of the mPTP complex-mediated apoptosis mechanisms and follow with a comprehensive discussion addressing the development of novel mPTP-targeting drugs/molecules in cancer treatment.
Collapse
Affiliation(s)
- Mohammad Waseem
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA;
| | - Bi-Dar Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA;
- Hormone Related Cancers Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| |
Collapse
|
16
|
Fang S, Zheng L, Shen L, Su Y, Ding J, Chen W, Chen X, Chen W, Shu G, Chen M, Zhao Z, Tu J, Ji J. Inactivation of KDM5A suppresses growth and enhances chemosensitivity in liver cancer by modulating ROCK1/PTEN/AKT pathway. Eur J Pharmacol 2023; 940:175465. [PMID: 36566915 DOI: 10.1016/j.ejphar.2022.175465] [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: 07/19/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Liver cancer is a kind of malignant tumor with poor sensitivity to chemotherapy. It is urgent to investigate approaches to improve the outcome of chemotherapy. KDM5A has been reported to be an oncogene in various cancers and is associated with drug resistance. However, the functions of KDM5A in chemotherapeutic sensitivity of liver cancer not been well illustrated. In this study, we found that KDM5A was upregulated in liver cancer tissue and cell lines. KDM5A knockdown using a gene interference strategy suppressed the growth of liver cancer in vitro and in vivo. CPI-455, a pharmacological inactivation of KDM5A enhanced the cytotoxicity of cisplatin (CDDP) in liver cells. CPI-455 and CDDP cotreatment resulted in apoptosis and mitochondrial dysfunction. We also found that knockdown or inactivation of KDM5A resulted in the downregulation of ROCK1, an oncogene regulating the activation of the PTEN/AKT signaling pathway. In particular, overexpression of ROCK1 or SF1670, a pharmacological inhibitor of PTEN, alleviated the cytotoxicity of CPI-455 and CDDP cotreatment. In HCCLM3 xenografts, CPI-455 and CDDP cotreatment dramatically inhibited the growth of xenograft tumor compared to CPI-455 or CDDP treatment alone. In conclusion, this study suggested that targeting the inactivation of KDM5A is an efficient strategy to enhance the chemosensitivity of liver cancer cells to CDDP by modulating the ROCK1/PTEN/AKT signaling pathway.
Collapse
Affiliation(s)
- Shiji Fang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China; Interventional Diagnosis and Treatment Center, Lishui Hospital of Zhejiang University, Lishui, 323000, China
| | - Liyun Zheng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China; Interventional Diagnosis and Treatment Center, Lishui Hospital of Zhejiang University, Lishui, 323000, China
| | - Lin Shen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China; Interventional Diagnosis and Treatment Center, Lishui Hospital of Zhejiang University, Lishui, 323000, China
| | - Yanping Su
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Jiayi Ding
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Weiyue Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Xiaoxiao Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Weiqian Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China; Interventional Diagnosis and Treatment Center, Lishui Hospital of Zhejiang University, Lishui, 323000, China
| | - Gaofeng Shu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China; Interventional Diagnosis and Treatment Center, Lishui Hospital of Zhejiang University, Lishui, 323000, China
| | - Jianfei Tu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China; Interventional Diagnosis and Treatment Center, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China; Interventional Diagnosis and Treatment Center, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
| |
Collapse
|
17
|
Zhang ZL, Li YZ, Wu GQ, Li YM, Zhang DD, Wang R. A comprehensive review of phytochemistry, pharmacology and clinical applications of Uncariae Ramulus Cum Uncis. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
18
|
Li Y, Xiong Z, Jiang Y, Zhou H, Yi L, Hu Y, Zhai X, Liu J, Tian F, Chen Y. Klf4 deficiency exacerbates myocardial ischemia/reperfusion injury in mice via enhancing ROCK1/DRP1 pathway-dependent mitochondrial fission. J Mol Cell Cardiol 2023; 174:115-132. [PMID: 36509022 DOI: 10.1016/j.yjmcc.2022.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022]
Abstract
RATIONAL Excessive mitochondrial fission is considered key process involved in myocardial ischemia/reperfusion (I/R) injury. However, the upstream mechanism remains largely unclear. Decreased level of Kruppel Like Factor 4 (KLF4) has been implicated in the pathogenesis of mitochondrial dysfunction and heart's adaption to stress. However, the role of Klf4 in I/R process is not fully elucidated. This study aims to investigate how Klf4 regulates mitochondrial dynamics and further clarify its underlying mechanism during cardiac I/R injury. METHODS Loss-of-function and gain-of-function strategies were applied to investigate the role of Klf4 in cardiac I/R injury via genetic ablation or intra-myocardial adenovirus injection. Mitochondrial dynamics was analyzed by confocal microscopy in vitro and transmission electron microscopy in vivo. Chromatin immunoprecipitation and luciferase reporter assay were performed to explore the underlying mechanisms. RESULTS KLF4 was downregulated in I/R heart. Cardiac-specific Klf4 knockout significantly exacerbated cardiac dysfunction in I/R mice. Mechanistically, Klf4 deficiency aggravated mitochondrial apoptosis, reduced ATP generation and boosted ROS overproduction via enhancing DRP1-dependent mitochondrial fission. ROCK1 was identified as a kinase regulating DRP1 activity at Ser616. Klf4 deficiency upregulated the expression of ROCK1 at transcriptional level, thus increasing S616-DRP1-mediated mitochondrial fission during I/R. Finally, reconstitution of Klf4 inhibited mitochondrial fission, restored mitochondrial function and alleviated I/R injury. CONCLUSION Our study provides the first evidence that Klf4 deficiency exacerbates myocardial I/R injury through regulating ROCK1 expression at transcriptional level to induce DRP1-mediated mitochondrial fission. Targeting mitochondrial dynamics by restoring Klf4 might be potentially cardio-protective strategies attenuating I/R injury.
Collapse
Affiliation(s)
- Yueyang Li
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhenyu Xiong
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yufan Jiang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Hao Zhou
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Li Yi
- Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yingyun Hu
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaofeng Zhai
- The Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jie Liu
- Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Feng Tian
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yundai Chen
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
19
|
Ghareghomi S, Atabaki V, Abdollahzadeh N, Ahmadian S, Hafez Ghoran S. Bioactive PI3-kinase/Akt/mTOR Inhibitors in Targeted Lung Cancer Therapy. Adv Pharm Bull 2023; 13:24-35. [PMID: 36721812 PMCID: PMC9871280 DOI: 10.34172/apb.2023.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/27/2021] [Accepted: 09/28/2021] [Indexed: 02/03/2023] Open
Abstract
One of the central signaling pathways with a regulatory effect on cell proliferation and survival is Akt/mTOR. In many human cancer types, for instance, lung cancer, the overexpression of Akt/mTOR has been reported. For this reason, either targeting cancer cells by synthetic or natural products affecting the Akt/mTOR pathway down-regulation is a useful strategy in cancer therapy. Direct inhibition of the signaling pathway or modulation of each related molecule could have significant feedback on the growth and proliferation of cancer cells. A variety of secondary metabolites has been identified to directly inhibit the AKT/mTOR signaling, which is important in the field of drug discovery. Naturally occurring nitrogenous and phenolic compounds can emerge as two pivotal classes of natural products possessing anticancer abilities. Herein, we have summarized the alkaloids and flavonoids for lung cancer treatment together with all the possible mechanisms of action relying on the Akt/mTOR pathway down-regulation. This review suggested that in search of new drugs, phytochemicals could be considered as promising scaffolds to be developed into efficient drugs for the treatment of cancer. In this review, the terms "Akt/mTOR", "Alkaloid", "flavonoid", and "lung cancer" were searched without any limitation in search criteria in Scopus, PubMed, Web of Science, and Google scholar engines.
Collapse
Affiliation(s)
- Somayyeh Ghareghomi
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Vahideh Atabaki
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Naseh Abdollahzadeh
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Shahin Ahmadian
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.,Corresponding Authors: Salar Hafez Ghoran and Shahin Ahmadian, and
| | - Salar Hafez Ghoran
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Corresponding Authors: Salar Hafez Ghoran and Shahin Ahmadian, and
| |
Collapse
|
20
|
刘 见, 沈 卫, 程 海, 范 旻, 肖 君, 徐 长, 谭 佳, 赖 岳, 余 成, 孙 东, 李 柳. [ Shenbai Jiedu Fang inhibits AOM/DSS-induced colorectal adenoma formation and carcinogenesis in mice via miRNA-22-mediated regulation of the PTEN/PI3K/AKT signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1452-1461. [PMID: 36329578 PMCID: PMC9637489 DOI: 10.12122/j.issn.1673-4254.2022.10.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To observe the inhibitory effect of Shenbai Jiedu Fang (SBJDF, a compound recipe of traditional Chinese herbal drugs) on chemically induced carcinogenesis of colorectal adenoma in mice and explore the role of PTEN/PI3K/AKT signaling pathway in mediating this effect. METHODS Four-week-old male C57BL/6 mice were randomly divided into control group (n=10), AOM/DSS model group (n=20), low-dose (14 g/kg) SBJDF group (n=10) and high-dose (42 g/kg) SBJDF group (n= 10). In the latter 3 groups, the mice were treated with azoxymethane (AOM) and dextran sodium sulphate (DSS) to induce carcinogenesis of colorectal adenoma. In the two SBJDF treatment groups, SBJDF was administered daily by gavage during the modeling. The survival rate, body weight, general condition of the mice, and intestinal adenoma formation and carcinogenesis were observed. The expressions of proteins associated with the PTEN/PI3K/AKT signaling pathway in the intestinal tissue were detected using immunohistochemistry. RESULTS Compared with those in the model group, the mice treated with SBJDF, especially at the high dose, showed a significantly lower incidence of intestinal carcinogenesis and had fewer intestinal tumors with smaller tumor volume. Pathological examination showed the occurrence of adenocarcinoma in the model group, while only low-grade and high-grade neoplasia were found in low-dose SBJDF group; the mice treated with high-dose SBJDF showed mainly normal mucosal tissues in the intestines with only a few lesions of low-grade neoplasia of adenoma. Compared with those in the control group, the mice in the model group had significantly elevated plasma miRNA-222 level (P < 0.05), which was obviously lowered in the two SBJDF groups (P < 0.01). The results of immunohistochemistry revealed that compared with the model group, the two SBJDF groups, especially the high-dose group, had significantly up-regulated expressions of PTEN, P-PTEN and GSK-3β and down-regulated expressions of p-GSK-3 β, PI3K, AKT, P-AKT, β-catenin, c-myc, cyclinD1 and survivin in the intestinal tissues. CONCLUSION SBJDF can significantly inhibit colorectal adenoma formation and carcino-genesis in mice possibly through regulating miRNA-222 and affecting PTEN/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- 见荣 刘
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 南京市中医院,江苏 南京 210022Nanjing Hospital of Chinese Medicine, Nanjing, 210000, China
| | - 卫星 沈
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 江苏省中医药防治肿瘤协同创新中心,江苏 南京 210023Jiangsu Collaborative Innovation Center of TCM Prevention and Treatment of Tumor, Nanjing 210023, China
| | - 海波 程
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 江苏省中医药防治肿瘤协同创新中心,江苏 南京 210023Jiangsu Collaborative Innovation Center of TCM Prevention and Treatment of Tumor, Nanjing 210023, China
| | - 旻旻 范
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 江苏省中医药防治肿瘤协同创新中心,江苏 南京 210023Jiangsu Collaborative Innovation Center of TCM Prevention and Treatment of Tumor, Nanjing 210023, China
| | - 君 肖
- 江苏省中医院,江苏 南京 210004Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| | - 长亮 徐
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 江苏省中医药防治肿瘤协同创新中心,江苏 南京 210023Jiangsu Collaborative Innovation Center of TCM Prevention and Treatment of Tumor, Nanjing 210023, China
| | - 佳妮 谭
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 江苏省中医药防治肿瘤协同创新中心,江苏 南京 210023Jiangsu Collaborative Innovation Center of TCM Prevention and Treatment of Tumor, Nanjing 210023, China
| | - 岳阳 赖
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 江苏省中医药防治肿瘤协同创新中心,江苏 南京 210023Jiangsu Collaborative Innovation Center of TCM Prevention and Treatment of Tumor, Nanjing 210023, China
| | - 成涛 余
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 江苏省中医药防治肿瘤协同创新中心,江苏 南京 210023Jiangsu Collaborative Innovation Center of TCM Prevention and Treatment of Tumor, Nanjing 210023, China
| | - 东东 孙
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 江苏省中医药防治肿瘤协同创新中心,江苏 南京 210023Jiangsu Collaborative Innovation Center of TCM Prevention and Treatment of Tumor, Nanjing 210023, China
| | - 柳 李
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 江苏省中医药防治肿瘤协同创新中心,江苏 南京 210023Jiangsu Collaborative Innovation Center of TCM Prevention and Treatment of Tumor, Nanjing 210023, China
| |
Collapse
|
21
|
Zhang L, Liu Y, Zhou R, He B, Wang W, Zhang B. Cyclophilin D: Guardian or Executioner for Tumor Cells? Front Oncol 2022; 12:939588. [PMID: 35860554 PMCID: PMC9289278 DOI: 10.3389/fonc.2022.939588] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclophilin D (CypD) is a peptide-proline cis-trans isomerase (PPIase) distributed in the mitochondrial matrix. CypD regulates the opening of the mitochondrial permeability conversion pore (mPTP) and mitochondrial bioenergetics through PPIase activity or interaction with multiple binding partners in mitochondria. CypD initially attracted attention due to its regulation of mPTP overopening-mediated cell death. However, recent studies on the effects of CypD on tumors have shown conflicting results. Although CypD has been proven to promote the aerobic glycolysis in tumor cells, its regulation of malignant characteristics such as the survival, invasion and drug resistance of tumor cells remains controversial. Here, we elaborate the main biological functions of CypD and its relationships with tumor progression identified in recent years, focusing on the dual role of CypD in tumors.
Collapse
Affiliation(s)
- Ling Zhang
- School of Nursing, Jining Medical University, Jining, China
- *Correspondence: Bin Zhang, ; Ling Zhang,
| | - Yi Liu
- School of Nursing, Jining Medical University, Jining, China
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Rou Zhou
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Baoyu He
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Wenjun Wang
- School of Nursing, Jining Medical University, Jining, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
- *Correspondence: Bin Zhang, ; Ling Zhang,
| |
Collapse
|
22
|
Gao Y, Zhang D, Wang F, Zhang D, Li P, Wang K. BRAF V600E protect from cell death via inhibition of the mitochondrial permeability transition in papillary and anaplastic thyroid cancers. J Cell Mol Med 2022; 26:4048-4060. [PMID: 35748101 PMCID: PMC9279591 DOI: 10.1111/jcmm.17443] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/08/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022] Open
Abstract
BRAF T1799A mutation is the most common genetic variation in thyroid cancer, resulting in the production of BRAF V600E mutant protein reported to make cells resistant to apoptosis. However, the mechanism by which BRAF V600E regulates cell death remains unknown. We constructed BRAF V600E overexpression and knockdown 8505C and BCPAP papillary and anaplastic thyroid cancer cell to investigate regulatory mechanism of BRAF V600E in cell death induced by staurosporine (STS). Induced BRAF V600E expression attenuated STS-induced papillary and anaplastic thyroid cancer death, while BRAF V600E knockdown aggravated it. TMRM and calcein-AM staining showed that opening of the mitochondrial permeability transition pore (mPTP) during STS-induced cell death could be significantly inhibited by BRAF V600E. Moreover, our study demonstrated that BRAF V600E constitutively activates mitochondrial ERK (mERK) to inhibit GSK-3-dependent CypD phosphorylation, thereby making BRAF V600E mutant tumour cells more resistant to mPTP opening. In the mitochondria of BRAF V600E mutant cells, there was an interaction between ERK1/2 and GSKa/ß, while upon BRAF V600E knockdown, interaction of GSKa/ß to ERK was decreased significantly. These results show that in thyroid cancer, BRAF V600E regulates the mitochondrial permeability transition through the pERK-pGSK-CypD pathway to resist death, providing new intervention targets for BRAF V600E mutant tumours.
Collapse
Affiliation(s)
- Yanyan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China.,Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Deyu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Fei Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Dejiu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
23
|
Zhang S, Wang Y, Cao Y, Wu J, Zhang Z, Ren H, Xu X, Kaznacheyeva E, Li Q, Wang G. Inhibition of the PINK1-Parkin Pathway Enhances the Lethality of Sorafenib and Regorafenib in Hepatocellular Carcinoma. Front Pharmacol 2022; 13:851832. [PMID: 35370635 PMCID: PMC8967359 DOI: 10.3389/fphar.2022.851832] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/28/2022] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common fatal malignancies and the main cause of cancer-related deaths. The multitarget tyrosine kinase inhibitors (TKIs) sorafenib and regorafenib are systemic therapeutic drugs approved for the treatment of HCC. Here, we found that sorafenib and regorafenib injured mitochondria by inducing mitochondrial Ca2+ (mtCa2+) overload and mitochondrial permeability transition pore (mPTP) opening, resulting in mitochondria-mediated cell death, which was alleviated by cyclosporin A (CsA), an inhibitor of mPTP. Meanwhile, mPTP opening caused PINK1 accumulation on damaged mitochondria, which recruited Parkin to mitochondria to induce mitophagy. Inhibition of autophagy by the lysosomal inhibitor chloroquine (CQ) or inhibition of mitochondrial fission by mdivi-1 aggravated sorafenib- and regorafenib-induced cell death. Moreover, knockdown of PINK1 also promotes sorafenib- and regorafenib-induced cell death. An in vivo study showed that sorafenib and regorafenib inhibited HepG2 cell growth more effectively in PINK1 knockdown cells than in shNTC cells in null mice. Thus, our data demonstrate that PINK1-Parkin-mediated mitophagy alleviates sorafenib and regorafenib antitumor effects in vitro and in vivo.
Collapse
Affiliation(s)
- Shun Zhang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yixin Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yifan Cao
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jin Wu
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zubin Zhang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Haigang Ren
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xiaohui Xu
- Department of General Surgery, the First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, China
| | | | - Qing Li
- Department of Gastroenterology, the First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,Center of Translational Medicine, the First People's Hospital of Taicang, Taicang Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Wang H, Fang F, Chen S, Jing X, Zhuang Y, Xie Y. Dual efficacy of Fasudil at improvement of survival and reinnervation of flap through RhoA/ROCK/PI3K/Akt pathway. Int Wound J 2022; 19:2000-2011. [PMID: 35315211 PMCID: PMC9705174 DOI: 10.1111/iwj.13800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/14/2022] [Indexed: 11/27/2022] Open
Abstract
Fasudil is reported to be effective at protecting against ischaemic diseases, and at augmenting axon growth. In this study, we aim to evaluate its efficacy in promoting flap survival and reinnervation. Ninety-two Institute of Cancer Research (ICR) mice were used and divided into the control, Fasudil, LY294002, Fasudil+LY294002 groups, receiving a daily intraperitoneal injection of normal saline, Fasudil (10 mg/kg), LY294002 (5 mg/kg), and Fasudil (10 mg/kg) + LY294002 (5 mg/kg), respectively. On days 0 and 5, the blood perfusion and diameter of the iliolumbar artery in the pedicle of the flaps in the four groups were evaluated using laser speckling contrast imaging (LSCI). On day 5, the flaps were photographed and the necrosis rate of the flaps was calculated using Photoshop CS6. In addition, tissues were harvested from the flaps and divided into two parts. One part underwent routine cryosection and immunofluorescent staining using the antibody against CD31 for evaluation of the microvascular density in the four groups. In the other part, the expression of RhoA, ROCK1+2, p-CPI-17, p-MYPT, p-PTEN, p-PI3K, p-Akt, and vascular endothelial growth factor (VEGF) within the flaps were determined using western blotting. Moreover, at days 0, 7, 15, and 30 after flap surgery, the axons within the flaps were evaluated using immunofluorescent staining with the antibody against Neurofilament-200. It turned out that the necrosis rate was (24.4 ± 7.7)%, (5.2 ± 1.6)%, (29.8 ± 4.2)%, and (30.9 ± 7.1)%, respectively, in the control, Fasudil, LY294002, LY294002+Fasudil groups. There was a significant reduction in the necrosis rate of the flaps in the Fasudil group (P < .001). The LSCI and immunofluorescent staining demonstrated that Fasudil could significantly expand the diameter of the iliolumbar artery in the pedicle, boost the overall blood perfusion, and increase the microvascular density of the flaps in the Fasudil group (P < .05), which could all be abolished by PI3K inhibitor LY294002. On day 5, the expression of p-CPI-17, p-MYPT, and p-PTEN were downregulated, whereas pPI3K, p-Akt, and VEGF were upregulated in the Fasudil group (P < .001). As for reinnervation, Neurofilament-200 fluorescent staining revealed that at days 15 and 30 after flap harvest, only in the Fasudil group could new axons be observed. It can be concluded that Fasudil could simultaneously improve the survival and axon growth after flap harvest, a dual efficacy achieved by inhibition of the RhoA/ROCK pathway, which in turn activates /PI3K/AKT pathway.
Collapse
Affiliation(s)
- Hai Wang
- Orthopedic Department, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Fang Fang
- Department of pharmacology, Fujian Medical University, Fuzhou, China
| | - Shaofeng Chen
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Institute of Clinical Applied Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xing Jing
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Institute of Clinical Applied Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yuehong Zhuang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Institute of Clinical Applied Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yun Xie
- Orthopedic Department, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
25
|
Ai J, Li J, Su Q, Ma H, Wei Q, Li H, Gao G. rAAV-delivered PTEN therapeutics for prostate cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:122-132. [PMID: 34976432 PMCID: PMC8671520 DOI: 10.1016/j.omtn.2021.11.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 11/28/2021] [Indexed: 02/05/2023]
Abstract
Effective treatments for prostate cancer (PCa) require further development, and previous studies have reported that PTEN and its downstream target CDKN1B are significantly downregulated in PCa cells compared with normal cells. Therefore, modulation of PTEN and CDKN1B expression might be a promising therapeutic approach for PCa treatment. Expression of PTEN and CDKN1B was verified in specimens from PCa patients and transgenic adenocarcinoma mouse prostate (TRAMP) mice. The effect of PTEN on PCa cell migration, apoptosis, and the cell cycle was analyzed in vitro using a wound-healing assay and flow cytometry. We assessed the ability of intraprostatic and intratumoral injections of recombinant adeno-associated virus (rAAV) 9 expressing Pten or Cdkn1b into TRAMP mice and a subcutaneous tumor xenograft mouse model, respectively, to inhibit PCa progression. PTEN and CDKN1B were significantly downregulated in human and mouse PCa samples, and CDKN1B expression correlated positively with PTEN expression. PTEN overexpression significantly inhibited cell migration and cell-cycle progression and promoted apoptosis in PCa cells by decreasing Ccnd1 expression and increasing that of Cdkn1b. Importantly, treatment with the rAAV9.Pten or rAAV9.Cdkn1b extended the lifespan of TRAMP mice and inhibited the growth rate of tumor xenografts by regulating downstream gene expression. Moreover, neoplasia in treated prostates was significantly diminished compared with that in control prostates, and apoptosis was markedly observed in xenografts treated with Pten or Cdkn1b. These data indicate that rAAV-based PTEN/CDKN1B delivery is promising for the development of novel therapeutics for PCa.
Collapse
Affiliation(s)
- Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
- Horae Gene Therapy Center, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Jia Li
- Horae Gene Therapy Center, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Qin Su
- Horae Gene Therapy Center, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Hong Ma
- Horae Gene Therapy Center, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Hong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| |
Collapse
|
26
|
Boyenle ID, Oyedele AK, Ogunlana AT, Adeyemo AF, Oyelere FS, Akinola OB, Adelusi TI, Ehigie LO, Ehigie AF. Targeting the mitochondrial permeability transition pore for drug discovery: Challenges and opportunities. Mitochondrion 2022; 63:57-71. [PMID: 35077882 DOI: 10.1016/j.mito.2022.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 12/29/2022]
Abstract
Several drug targets have been amenable to drug discovery pursuit not until the characterization of the mitochondrial permeability transition pore (MPTP), a pore with an undefined molecular identity that forms on the inner mitochondrial membrane upon mitochondrial permeability transition (MPT) under the influence of calcium overload and oxidative stress. The opening of the pore which is presumed to cause cell death in certain human diseases also has implications under physiological parlance. Different models for this pore have been postulated following its first identification in the last six decades. The mitochondrial community has witnessed many protein candidates such as; voltage-dependent anion channel (VDAC), adenine nucleotide translocase (ANT), Mitochondrial phosphate carrier (PiC), Spastic Paralegin (SPG7), disordered proteins, and F1Fo ATPase. However, genetic studies have cast out most of these candidates with only F1Fo ATPase currently under intense argument. Cyclophilin D (CyPD) remains the widely accepted positive regulator of the MPTP known to date, but no drug candidate has emerged as its inhibitor, raising concern issues for therapeutics. Thus, in this review, we discuss various models of MPTP reported with the hope of stimulating further research in this field. We went beyond the classical description of the MPTP to ascribe a 'two-edged sword property' to the pore for therapeutic function in human disease because its inhibition and activation have pharmacological relevance. We suggested putative proteins upstream to CyPD that can regulate its activity and prevent cell deaths in neurodegenerative disease and ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ibrahim Damilare Boyenle
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria; Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abdulquddus Kehinde Oyedele
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abdeen Tunde Ogunlana
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Aishat Folashade Adeyemo
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | | | - Olateju Balikis Akinola
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Temitope Isaac Adelusi
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Leonard Ona Ehigie
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Adeola Folasade Ehigie
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria.
| |
Collapse
|
27
|
Dong L, Liu F, Liu D, Kang S, Yang X, Wang J. Jolkinolide B attenuates laryngeal cancer cell growth and induces apoptosis via PTEN/PI3K/Akt signaling pathway. In Vitro Cell Dev Biol Anim 2021; 57:786-794. [PMID: 34697781 DOI: 10.1007/s11626-021-00612-3] [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: 01/18/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022]
Abstract
Jolkinolide B (JB) is a bioactive diterpenoid, isolated from the root of Euphorbia fischeriana Steud, and has been reported to have anti-tumor and anti-inflammation function by regulation of cell migration, invasion, apoptosis, and cell cycle. We aimed to evaluate the effect of JB on laryngeal cancer cells. Human normal larynx epithelial (HBE) cells and cancer cell lines TU212, TU177, and Hep-2 were cultured; MTT assay was used to assess cell proliferation. LY294002 (a PI3K/Akt inhibitor) and IGF-1 (a PI3K/Akt activator) were employed to investigate the expression of PI3K/Akt pathway. Cell migration and invasion activities were detected by scratch wound healing and transwell assay, respectively. Flow cytometry assay was used to assess cell apoptosis. The expression levels of proteins were assessed by immunofluorescence and Western blotting assay. JB inhibited TU212, TU177, and Hep-2 cell viability with an IC50 value of 54.57 ± 0.53 μg/mL, 44.82 ± 0.32 μg/mL, and 49.63 ± 0.47 μg/mL, respectively. Compared with control group, the proliferation, migration, and invasion of cells significantly decreased after JB and LY294002 treatment, while cell apoptosis increased. In IGF-1 group, the results were opposite compared to the JB and LY294002 groups. Western blotting results showed that JB and LY294002 treatment significantly inhibited the levels of Bcl-2, p-PI3K, and p-Akt while the levels of Bax, cleaved caspase-3, and PTEN protein significantly increased. Our study suggested that JB exhibits an inhibition effect on laryngeal cancer cell growth in vitro.
Collapse
Affiliation(s)
- Lei Dong
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Yantai, 264000, Shandong, China
| | - Feifei Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Yantai, 264000, Shandong, China
| | - Dawei Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Yantai, 264000, Shandong, China
| | - Shasha Kang
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Yantai, 264000, Shandong, China
| | - Xin Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Yantai, 264000, Shandong, China.
| | - Junxia Wang
- Department of Otorhinolaryngology, Yantai Haigang Hospital, No. 100 Xingfu Road, Zhifu District, Yantai, 264000, Shandong, China.
| |
Collapse
|
28
|
Xiaoli F, Yaqing Z, Ruhui L, Xuan L, Aijie C, Yanli Z, Chen H, Lili C, Longquan S. Graphene oxide disrupted mitochondrial homeostasis through inducing intracellular redox deviation and autophagy-lysosomal network dysfunction in SH-SY5Y cells. JOURNAL OF HAZARDOUS MATERIALS 2021; 416:126158. [PMID: 34492938 DOI: 10.1016/j.jhazmat.2021.126158] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/05/2021] [Accepted: 05/16/2021] [Indexed: 06/13/2023]
Abstract
Graphene oxide (GO) nanomaterials have significant advantages for drug delivery and electrode materials in neural science, however, their exposure risks to the central nervous system (CNS) and toxicity concerns are also increased. The current studies of GO-induced neurotoxicity remain still ambiguous, let alone the mechanism of how complicated GO chemistry affects its biological behavior with neural cells. In this study, we characterized the commercially available GO in detail and investigated its biological adverse effects using cultured SH-SY5Y cells. We found that ultrasonic processing in medium changed the oxidation status and surface reactivity on the planar surface of GO due to its hydration activity, causing lipid peroxidation and cell membrane damage. Subsequently, ROS-disrupted mitochondrial homeostasis, resulting from the activation of NOX2 signaling, was observed following GO internalization. The autophagy-lysosomal network was initiated as a defensive reaction to obliterate oxidative damaged mitochondria and foreign nanomaterials, which was ineffective due to reduced lysosomal degradation capacity. These sequential cellular responses exacerbated mitochondrial stress, leading to apoptotic cell death. These data highlight the importance of the structure-related activity of GO on its biological properties and provide an in-depth understanding of how GO-derived cellular redox signaling induces mitochondrion-related cascades that modulate cell functionality and survival.
Collapse
Affiliation(s)
- Feng Xiaoli
- Stomatology Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China
| | - Zhang Yaqing
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Luo Ruhui
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lai Xuan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chen Aijie
- Stomatology Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhang Yanli
- Stomatology Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hu Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chen Lili
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shao Longquan
- Stomatology Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China.
| |
Collapse
|
29
|
Szabo I, Zoratti M, Biasutto L. Targeting mitochondrial ion channels for cancer therapy. Redox Biol 2021; 42:101846. [PMID: 33419703 PMCID: PMC8113036 DOI: 10.1016/j.redox.2020.101846] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Pharmacological targeting of mitochondrial ion channels is emerging as a promising approach to eliminate cancer cells; as most of these channels are differentially expressed and/or regulated in cancer cells in comparison to healthy ones, this strategy may selectively eliminate the former. Perturbation of ion fluxes across the outer and inner membranes is linked to alterations of redox state, membrane potential and bioenergetic efficiency. This leads to indirect modulation of oxidative phosphorylation, which is/may be fundamental for both cancer and cancer stem cell survival. Furthermore, given the crucial contribution of mitochondria to intrinsic apoptosis, modulation of their ion channels leading to cytochrome c release may be of great advantage in case of resistance to drugs triggering apoptotic events upstream of the mitochondrial phase. In the present review, we give an overview of the known mitochondrial ion channels and of their modulators capable of killing cancer cells. In addition, we discuss state-of-the-art strategies using mitochondriotropic drugs or peptide-based approaches allowing a more efficient and selective targeting of mitochondrial ion channel-linked events.
Collapse
Affiliation(s)
- Ildiko Szabo
- Department of Biology, University of Padova, Italy; CNR Institute of Neurosciences, Padova, Italy.
| | | | - Lucia Biasutto
- CNR Institute of Neurosciences, Padova, Italy; Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
30
|
Lica JJ, Wieczór M, Grabe GJ, Heldt M, Jancz M, Misiak M, Gucwa K, Brankiewicz W, Maciejewska N, Stupak A, Bagiński M, Rolka K, Hellmann A, Składanowski A. Effective Drug Concentration and Selectivity Depends on Fraction of Primitive Cells. Int J Mol Sci 2021; 22:ijms22094931. [PMID: 34066491 PMCID: PMC8125035 DOI: 10.3390/ijms22094931] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/15/2021] [Accepted: 04/23/2021] [Indexed: 12/25/2022] Open
Abstract
Poor efficiency of chemotherapeutics in the eradication of Cancer Stem Cells (CSCs) has been driving the search for more active and specific compounds. In this work, we show how cell density-dependent stage culture profiles can be used in drug development workflows to achieve more robust drug activity (IC50 and EC50) results. Using flow cytometry and light microscopy, we characterized the cytological stage profiles of the HL-60-, A-549-, and HEK-293-derived sublines with a focus on their primitive cell content. We then used a range of cytotoxic substances—C-123, bortezomib, idarubicin, C-1305, doxorubicin, DMSO, and ethanol—to highlight typical density-related issues accompanying drug activity determination. We also showed that drug EC50 and selectivity indices normalized to primitive cell content are more accurate activity measurements. We tested our approach by calculating the corrected selectivity index of a novel chemotherapeutic candidate, C-123. Overall, our study highlights the usefulness of accounting for primitive cell fractions in the assessment of drug efficiency.
Collapse
Affiliation(s)
- Jan Jakub Lica
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (K.G.); (K.R.)
- Correspondence:
| | - Miłosz Wieczór
- Department of Physical Chemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland;
| | - Grzegorz Jan Grabe
- Department of Microbiology, Harvard Medical School, 4 Blackfan Circle, Boston, MA 02115, USA;
| | - Mateusz Heldt
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland; (M.H.); (M.J.); (M.M.); (W.B.); (N.M.); (M.B.); (A.S.)
| | - Marta Jancz
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland; (M.H.); (M.J.); (M.M.); (W.B.); (N.M.); (M.B.); (A.S.)
| | - Majus Misiak
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland; (M.H.); (M.J.); (M.M.); (W.B.); (N.M.); (M.B.); (A.S.)
| | - Katarzyna Gucwa
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (K.G.); (K.R.)
| | - Wioletta Brankiewicz
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland; (M.H.); (M.J.); (M.M.); (W.B.); (N.M.); (M.B.); (A.S.)
| | - Natalia Maciejewska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland; (M.H.); (M.J.); (M.M.); (W.B.); (N.M.); (M.B.); (A.S.)
| | - Anna Stupak
- Polpharma Biologics S.A., Gdansk Science & Technology Park, Building A, 80-172 Gdansk, Poland;
| | - Maciej Bagiński
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland; (M.H.); (M.J.); (M.M.); (W.B.); (N.M.); (M.B.); (A.S.)
| | - Krzysztof Rolka
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (K.G.); (K.R.)
| | - Andrzej Hellmann
- Department of Hematology and Transplantology, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Andrzej Składanowski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland; (M.H.); (M.J.); (M.M.); (W.B.); (N.M.); (M.B.); (A.S.)
| |
Collapse
|
31
|
Sappanone A alleviates hypoxia/reoxygenation-induced cardiomyocytes injury through inhibition of mitochondrial apoptosis and activation of PI3K-Akt-Gsk-3β pathway. Biosci Rep 2021; 40:222121. [PMID: 32095825 PMCID: PMC7042124 DOI: 10.1042/bsr20192442] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 01/14/2023] Open
Abstract
Myocardial ischemia reperfusion injury (MIRI) is a complex pathophysiological process involved with the activation of oxidative stress, inflammation and apoptosis. Sappanone A (SA), a homoisoflavanone isolated from the heartwood of Caesalpinia sappan L., could exhibit antioxidant, anti-inflammatory and anti-apoptotic activities. Therefore, we assumed that SA has a potential use for preventing against MIRI. The present study aimed to investigate the effect of SA treatment on MIRI and its mechanism. Cardiomyocytes (H9c2 cells) were treated with SA for 1 h, followed by 6 h of hypoxia/3 h of reoxygenation. Cell viability assay was detected by CCK-8 assay. Apoptosis was measured by flow cytometry and Hoechst staining. Mitochondrial permeability transition pore (mPTP) opening and mitochondrial transmembrane potential (ΔΨm) were measured by spectrophotometry and JC-1 staining. The changes of mitochondrial apoptosis-related proteins and PI3K–Akt–Gsk-3β signaling pathway were evaluated by Western blotting. The results showed that SA pretreatment enhanced the cell viability and decreased the activity of myocardial enzyme in a dose-dependent manner. Moreover, SA pretreatment significantly inhibited apoptosis, blocked mPTP opening, suppressed the release of ΔΨm, prevented the cytochrome c releasing from mitochondria into cytoplasm, and repressed the cleavage of caspase-9 and caspase-3. Furthermore, SA pretreatment increased the phosphorylation levels of Akt and Gsk-3β but not of Stat-3. Meanwhile, the protective effect of SA was abrogated by PI3K inhibitor (LY294002). In conclusion, our results demonstrate that SA could prevent hypoxia/reoxygenation-induced cardiomyocytes injury through inhibition of mitochondrial apoptosis and activation of PI3K–Akt–Gsk-3β pathway. Thus, SA may have a potential use for the prevention of MIRI.
Collapse
|
32
|
Meng J, Su R, Wang L, Yuan B, Li L. Inhibitory effect and mechanism of action (MOA) of hirsutine on the proliferation of T-cell leukemia Jurkat clone E6-1 cells. PeerJ 2021; 9:e10692. [PMID: 33604171 PMCID: PMC7863788 DOI: 10.7717/peerj.10692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/11/2020] [Indexed: 12/24/2022] Open
Abstract
Background The bark of Uncaria rhynchophylla has been traditionally used to treat convulsion, bleeding, hypertension, auto-immune conditions, cancer, and other diseases. The main focus of this research is done for the purpose of exploring the antitumor activity and mechanism of action (MOA) for hirsutine isolated from U. rhynchophylla. Methods Jurkat clone E6-1 cells were treated using 10, 25 and 50 μM for 48 h. Inhibition of cell proliferation due to hirsutine treatment was evaluated by CCK8 assay. Flow cytometry was applied to ascertain Jurkat cell cycle progression and apoptosis after treatment with 10, 25 and 50 μM hirsutine for 48 h. The expression and level of the apoptosis-related genes and proteins was analyzed by Real-time Quantitative polymerase chain reaction (qPCR) and Western blotting method, respectively. Results CCK8 analyses revealed that hirsutine could significantly inhibit the proliferation of Jurkat clone E6-1 cells, in a concentration and time-dependent fashion. Flow cytometry assays revealed that hirsutine could drive apoptotic death and G0/G1 phase arrest in Jurkat cells. Apoptotic cells frequencies were 4.99 ± 0.51%, 13.69 ± 2.00% and 40.21 ± 15.19%, and respective cell cycle arrest in G0/G1 accounted for 34.85 ± 1.81%, 42.83 ± 0.70% and 49.12 ± 4.07%. Simultaneously, compared with the control group, Western blot assays indicated that the up-regulation of pro-apoptotic Bax, cleaved-caspase3, cleaved-caspase9 and Cyto c proteins, as well as the down-regulation of Bcl-2 protein which guards against cell death, might be correlated with cell death induction and inhibition of cell proliferation. QPCR analyses indicated that hirsutine could diminish BCL2 expression and, at the same time, improve Bax, caspase-3 and caspase-9 mRNA levels, thus reiterating a putative correlation of hirsutine treatment in vitro with apoptosis induction and inhibition of cell proliferation (p-value < 0.05). Excessive hirsutine damages the ultrastructure in mitochondria, leading to the release of Cyt c from the mitochondria to cytoplasm in Jurkat clone E6-1 cells, thereby inducing the activated caspase cascade apoptosis process through a mitochondria-mediated pathway. Conclusion An important bioactive constituent-hirsutine-appears to have antitumor effects in human T-cell leukemia, thus enlightening the use of phytomedicines as a novel source for tumor therapy. It is speculated that hirsutine may induce apoptosis of Jurkat Clone E6-1 cells through the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Jie Meng
- Department of Pharmacy, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Hongqiao International Institute of Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Rui Su
- Department of Pharmacy, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Luping Wang
- Department of Pharmacy, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bo Yuan
- Department of Pharmacy, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ling Li
- Department of Pharmacy, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Cui Y, Pan M, Ma J, Song X, Cao W, Zhang P. Recent progress in the use of mitochondrial membrane permeability transition pore in mitochondrial dysfunction-related disease therapies. Mol Cell Biochem 2021; 476:493-506. [PMID: 33000352 DOI: 10.1007/s11010-020-03926-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria have various cellular functions, including ATP synthesis, calcium homeostasis, cell senescence, and death. Mitochondrial dysfunction has been identified in a variety of disorders correlated with human health. Among the many underlying mechanisms of mitochondrial dysfunction, the opening up of the mitochondrial permeability transition pore (mPTP) is one that has drawn increasing interest in recent years. It plays an important role in apoptosis and necrosis; however, the molecular structure and function of the mPTP have still not been fully elucidated. In recent years, the abnormal opening up of the mPTP has been implicated in the development and pathogenesis of diverse diseases including ischemia/reperfusion injury (IRI), neurodegenerative disorders, tumors, and chronic obstructive pulmonary disease (COPD). This review provides a systematic introduction to the possible molecular makeup of the mPTP and summarizes the mitochondrial dysfunction-correlated diseases and highlights possible underlying mechanisms. Since the mPTP is an important target in mitochondrial dysfunction, this review also summarizes potential treatments, which may be used to inhibit pore opening up via the molecules composing mPTP complexes, thus suppressing the progression of mitochondrial dysfunction-related diseases.
Collapse
Affiliation(s)
- Yuting Cui
- School of Life Science, Shandong University of Technology, Zibo, Shandong Province, China
| | - Mingyue Pan
- Department of Pharmacy, Shenzhen Luohu People's Hospital, Shenzhen, Guangdong Province, China
| | - Jing Ma
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong Province, China
| | - Xinhua Song
- School of Life Science, Shandong University of Technology, Zibo, Shandong Province, China
| | - Weiling Cao
- Department of Pharmacy, Shenzhen Luohu People's Hospital, Shenzhen, Guangdong Province, China.
| | - Peng Zhang
- Department of Pharmacy, Shenzhen Luohu People's Hospital, Shenzhen, Guangdong Province, China.
| |
Collapse
|
34
|
Zhao L, Tang M, Bode AM, Liao W, Cao Y. ANTs and cancer: Emerging pathogenesis, mechanisms, and perspectives. Biochim Biophys Acta Rev Cancer 2020; 1875:188485. [PMID: 33309965 DOI: 10.1016/j.bbcan.2020.188485] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/03/2020] [Accepted: 11/21/2020] [Indexed: 12/15/2022]
Abstract
Adenine nucleotide translocases (ANTs) are a class of transporters located in the inner mitochondrial membrane that not only couple processes of cellular productivity and energy expenditure, but are also involved in the composition of the mitochondrial membrane permeability transition pore (mPTP). The function of ANTs has been found to be most closely related to their own conformational changes. Notably, as multifunctional proteins, ANTs play a key role in oncogenesis, which provides building blocks for tumor anabolism, control oxidative phosphorylation and glycolysis homeostasis, and govern cell death. Thus, ANTs constitute promising targets for the development of novel anticancer agents. Here, we review the recent findings regarding ANTs and their important mechanisms in cancer, with a focus on the therapeutic potential of targeting ANTs for cancer therapy.
Collapse
Affiliation(s)
- Lin Zhao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha 410078, China; Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha 410078, China
| | - Min Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha 410078, China; Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha 410078, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, Changsha 410078, China; Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China; Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha 410078, China; Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, China; Research Center for Technologies of Nucleic Acid-Based Diagnostics and Therapeutics Hunan Province, Changsha 410078, China; National Joint Engineering Research Center for Genetic Diagnostics of Infectious Diseases and Cancer, Changsha 410078, China.
| |
Collapse
|
35
|
Liang JH, Wang C, Huo XK, Tian XG, Zhao WY, Wang X, Sun CP, Ma XC. The genus Uncaria: A review on phytochemical metabolites and biological aspects. Fitoterapia 2020; 147:104772. [PMID: 33152463 DOI: 10.1016/j.fitote.2020.104772] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/03/2020] [Accepted: 10/25/2020] [Indexed: 12/14/2022]
Abstract
The genus Uncaira (Rubiaceae) comprises of 34 species, many of which are usually used as traditional Chinese medicines (TCMs) to treat hypertension, fever, headache, gastrointestinal illness, and fungal infection. Over the past twenty years, Uncaira species have been paid the considerable attentions in phytochemical and biological aspects, and about 100 new secondary metabolites, including alkaloids, triterpenes, and flavonoids, have been elucidated. This review aims to present a comprehensive and up-to date overview of the biological source, structures and their biosynthetic pathways, as well as the pharmacological of the compounds reported in the genus Uncaria for the past two decades. It would provide an insight into the emerging pharmacological applications of the genus Uncaria.
Collapse
Affiliation(s)
- Jia-Hao Liang
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Chao Wang
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Xiao-Kui Huo
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Xiang-Ge Tian
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Wen-Yu Zhao
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Xun Wang
- Department of Neurosurgery, The Third People's Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Cheng-Peng Sun
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China.
| | - Xiao-Chi Ma
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China.
| |
Collapse
|
36
|
Qin N, Lu X, Liu Y, Qiao Y, Qu W, Feng F, Sun H. Recent research progress of Uncaria spp. based on alkaloids: phytochemistry, pharmacology and structural chemistry. Eur J Med Chem 2020; 210:112960. [PMID: 33148492 DOI: 10.1016/j.ejmech.2020.112960] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 02/06/2023]
Abstract
Medicinal plants are well-known in affording clinically useful agents, with rich medicinal values by combining with disease targets through various mechanisms. Plant secondary metabolites as lead compounds lay the foundation for the discovery and development of new drugs in disease treatment. Genus Uncaria from Rubiaceae family is a significant plant source of active alkaloids, with anti-hypertensive, sedative, anti-Alzheimer's disease, anti-drug addiction and anti-inflammatory effects. This review summarizes and discuss the research progress of Uncaria based on alkaloids in the past 15 years, mainly in the past 5 years, including biosynthesis, phytochemistry, pharmacology and structural chemistry. Among, focusing on representative compounds rhynchophylline and isorhynchophylline, the pharmacological activities surrounding the central nervous system and cardiovascular system are described in detail. On the basis of case studies, this article provides a brief overview of the synthesis and analogues of representative compounds types. In summary, this review provides an early basis for further searching for new targets and activities, discussing the mechanisms of pharmacological activity and studying the structure-activity relationships of active molecules.
Collapse
Affiliation(s)
- Nan Qin
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Xin Lu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Yuting Qiao
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China; Jiangsu Food and Pharmaceutical Science College, Huaian, 223003, People's Republic of China.
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China; Jiangsu Food and Pharmaceutical Science College, Huaian, 223003, People's Republic of China.
| |
Collapse
|
37
|
Galber C, Acosta MJ, Minervini G, Giorgio V. The role of mitochondrial ATP synthase in cancer. Biol Chem 2020; 401:1199-1214. [PMID: 32769215 DOI: 10.1515/hsz-2020-0157] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022]
Abstract
The mitochondrial ATP synthase is a multi-subunit enzyme complex located in the inner mitochondrial membrane which is essential for oxidative phosphorylation under physiological conditions. In this review, we analyse the enzyme functions involved in cancer progression by dissecting specific conditions in which ATP synthase contributes to cancer development or metastasis. Moreover, we propose the role of ATP synthase in the formation of the permeability transition pore (PTP) as an additional mechanism which controls tumour cell death. We further describe transcriptional and translational modifications of the enzyme subunits and of the inhibitor protein IF1 that may promote adaptations leading to cancer metabolism. Finally, we outline ATP synthase gene mutations and epigenetic modifications associated with cancer development or drug resistance, with the aim of highlighting this enzyme complex as a potential novel target for future anti-cancer therapy.
Collapse
Affiliation(s)
- Chiara Galber
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, V.le G. Colombo 3, I-35121, Padova, Italy
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| | - Manuel Jesus Acosta
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, V.le G. Colombo 3, I-35121, Padova, Italy
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| | - Giovanni Minervini
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| | - Valentina Giorgio
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, V.le G. Colombo 3, I-35121, Padova, Italy
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| |
Collapse
|
38
|
The HMGB1-2 Ovarian Cancer Interactome. The Role of HMGB Proteins and Their Interacting Partners MIEN1 and NOP53 in Ovary Cancer and Drug-Response. Cancers (Basel) 2020; 12:cancers12092435. [PMID: 32867128 PMCID: PMC7564582 DOI: 10.3390/cancers12092435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
High mobility group box B (HMGB) proteins are overexpressed in different types of cancers such as epithelial ovarian cancers (EOC). We have determined the first interactome of HMGB1 and HMGB2 in epithelial ovarian cancer (the EOC-HMGB interactome). Libraries from the SKOV-3 cell line and a primary transitional cell carcinoma (TCC) ovarian tumor were tested by the Yeast Two Hybrid (Y2H) approach. The interactome reveals proteins that are related to cancer hallmarks and their expression is altered in EOC. Moreover, some of these proteins have been associated to survival and prognosis of patients. The interaction of MIEN1 and NOP53 with HMGB2 has been validated by co-immunoprecipitation in SKOV-3 and PEO1 cell lines. SKOV-3 cells were treated with different anti-tumoral drugs to evaluate changes in HMGB1, HMGB2, MIEN1 and NOP53 gene expression. Results show that combined treatment of paclitaxel and carboplatin induces a stronger down-regulation of these genes in comparison to individual treatments. Individual treatment with paclitaxel or olaparib up-regulates NOP53, which is expressed at lower levels in EOC than in non-cancerous cells. On the other hand, bevacizumab diminishes the expression of HMGB2 and NOP53. This study also shows that silencing of these genes affects cell-viability after drug exposure. HMGB1 silencing causes loss of response to paclitaxel, whereas silencing of HMGB2 slightly increases sensitivity to olaparib. Silencing of either HMGB1 or HMGB2 increases sensitivity to carboplatin. Lastly, a moderate loss of response to bevacizumab is observed when NOP53 is silenced.
Collapse
|
39
|
Yu H, Zhen J, Yang Y, Du J, Leng J, Tong Q. Rg1 protects H9C2 cells from high glucose-/palmitate-induced injury via activation of AKT/GSK-3β/Nrf2 pathway. J Cell Mol Med 2020; 24:8194-8205. [PMID: 32548942 PMCID: PMC7348154 DOI: 10.1111/jcmm.15486] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/06/2020] [Accepted: 05/24/2020] [Indexed: 12/15/2022] Open
Abstract
Our previous studies have assessed ginsenoside Rg1 (Rg1)‐mediated protection in a type 1 diabetes rat model. To uncover the mechanism through which Rg1 protects against cardiac injury induced by diabetes, we mimicked diabetic conditions by culturing H9C2 cells in high glucose/palmitate. Rg1 had no toxic effect, and it alleviated the high glucose/palmitate damage in a dose‐dependent manner, as indicated by 3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyl tetrazolium bromide assay and lactate dehydrogenase release to the culture medium. Rg1 prevented high glucose/palmitate‐induced cell apoptosis, assessed using cleaved caspase‐3 and terminal deoxynucleotidyl transferase dUTP nick end labelling staining. Rg1 also reduced high glucose‐/palmitate‐induced reactive oxygen species formation and increased intracellular antioxidant enzyme activity. We found that Rg1 activates protein kinase B (AKT)/glycogen synthase kinase‐3 (GSK‐3β) pathway and antioxidant nuclear factor erythroid 2‐related factor 2 (Nrf2) pathway, indicated by increased phosphorylation of AKT and GSK‐3β, and nuclear translocation of Nrf2. We used phosphatidylinositol‐3‐kinase inhibitor Ly294002 to block the activation of the AKT/GSK‐3β pathway and found that it partially reversed the protection by Rg1 and decreased Nrf2 pathway activation. The results suggest that Rg1 exerts a protective effect against high glucose and palmitate damage that is partially AKT/GSK‐3β/Nrf2‐mediated. Further studies are required to validate these findings using primary cardiomyocytes and animal models of diabetes.
Collapse
Affiliation(s)
- Haitao Yu
- The First Hospital of Jilin University, Changchun, China
| | - Juan Zhen
- The First Hospital of Jilin University, Changchun, China
| | - Yang Yang
- The First Hospital of Jilin University, Changchun, China
| | - Jian Du
- The First Hospital of Jilin University, Changchun, China
| | - Jiyan Leng
- The First Hospital of Jilin University, Changchun, China
| | - Qian Tong
- The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
40
|
Meng Y, Xu Q, Chen L, Wang L, Hu X. The function of SOX2 in breast cancer and relevant signaling pathway. Pathol Res Pract 2020; 216:153023. [PMID: 32703490 DOI: 10.1016/j.prp.2020.153023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 04/28/2020] [Accepted: 05/15/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The purpose of this study was to explore the functional roles of SOX2 in the progression of breast cancer and relevant molecular mechanism. METHODS A total of 108 breast cancer patients were included, and breast cancer cell line MDA-MB-231 was selected for this research. Real time-qualitative polymerase chain reaction (RT-qPCR) was conducted to measure the expression level of SOX2 mRNA. MTT and Transwell assays were used to detected the proliferation, migration and invasion of breast cancer cells, respectively. Luciferase reporter assay was conducted to reveal the relationship of SOX2 with PTEN. Western blot was performed to detect the expressions of Wnt/β-catenin pathway-related proteins. RESULTS The expression of SOX2 mRNA was up-regulated in breast cancer tissues and cells (P < 0.001). SOX2 expression was significantly associated with TNM stage and lymph node metastasis of breast cancer patients (P < 0.05). SOX2 knockdown significantly inhibited the proliferation, migration and invasion of breast cancer cells (P < 0.05). PTEN was a direct target of SOX2. The inhibition of PTEN could significantly suppress the progression of breast cancer cells with SOX2 overexpression. SOX2 knockdown also inhibited the expressions of β-catenin, TCP-4, FZD7, C-myc and MMP-7 proteins. Moreover, PTEN knockdown reversed the results caused by SOX2 overexpression, that is, increased expressions of β-catenin, TCP-4, FZD7, C-myc and MMP-7 proteins (P < 0.05). CONCLUSION SOX2 promotes the progression of breast cancer through activating Wnt/β-catenin signaling pathway via regulating PTEN.
Collapse
Affiliation(s)
- Yanchun Meng
- Department of Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Phase I Clinical Trial Center, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Qunfang Xu
- Clinical Laboratory, Capital Medical University Electric Teaching Hospital (State Grid Coporation of China Beijing Electric Power Hospital), Beijing, China
| | - Lin Chen
- Department of Colorectal Surgery, Department of General Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lingfei Wang
- Department of Oncology, the 903rd Hospital of PLA, Hangzhou, 310013, China.
| | - Xichun Hu
- Department of Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Phase I Clinical Trial Center, Shanghai Cancer Center, Fudan University, Shanghai, China.
| |
Collapse
|
41
|
Xu Y, Guo X, Wang G, Zhou C. Vitamin C Inhibits Metastasis of Peritoneal Tumors By Preventing Spheroid Formation in ID8 Murine Epithelial Peritoneal Cancer Model. Front Pharmacol 2020; 11:645. [PMID: 32477126 PMCID: PMC7236773 DOI: 10.3389/fphar.2020.00645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
High mortality is associated with exclusively metastasis within the peritoneal cavity among patients with epithelial ovarian cancer that is the most lethal gynecologic cancer. There is an unmet need to develop more effective therapies to prevent metastasis of peritoneal cancer. Multicellular spheroid formation, during which cancer cells migrate and adhere to tumor-associated macrophages, is a critical step of ovarian cancer metastasis. Here, we showed that vitamin C inhibited spheroid formation and metastasis in ID8 ovarian cancer-bearing mice. We further found that vitamin C treatment decreased the levels of M2 macrophages in tumor nodules and suppressed the epithelial-mesenchymal transition (EMT). In vitro studies revealed that vitamin C inhibited proliferation, arrested cell cycle, attenuated migration, and prevented the spheroid formation of ID8 ovarian cancer cells. Vitamin C induced apoptosis of ID8 cells, which was confirmed by membrane potential collapse, cytosolic calcium overload, ATP depletion, and caspase-3 activation in vitamin C-treated cells. Intriguingly, vitamin C treatment caused striking morphological change and apoptosis of macrophages. The presented proof of concept study strategically identifies new anticancer mechanisms of vitamin C.
Collapse
Affiliation(s)
- Yayun Xu
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, China
| | - Xing Guo
- Department of Pharmacy, People's Hospital of Rizhao, Rizhao, China
| | - Ganyu Wang
- Department of Pediatric Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Changkuo Zhou
- Department of Urology, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
42
|
Zhou M, Li G, Zhu L, Zhou H, Lu L. Arctiin attenuates high glucose-induced human retinal capillary endothelial cell proliferation by regulating ROCK1/PTEN/PI3K/Akt/VEGF pathway in vitro. J Cell Mol Med 2020; 24:5695-5706. [PMID: 32297701 PMCID: PMC7214144 DOI: 10.1111/jcmm.15232] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 03/01/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the most prominent microvascular complications of diabetes, which remains the leading cause of legal blindness in the world. Arctiin, a bioactive compound from Arctium lappa L., has been reported to have antidiabetic activity. In this study, we investigated the effect of arctiin on a human retinal capillary endothelial cell (HRCEC) line and how arctiin inhibits cell proliferation in high glucose (HG)-induced HRCECs. Results showed that arctiin decreased HG-induced HRCECs proliferation in a dose-dependent manner by inducing cell cycle arrest at the G0/G1 phase. Tube formation assay and immunofluorescence staining indicated that arctiin abrogated tube formation induced by HG-induced HRCECs in a dose-dependent manner via down-regulation of VEGF expression. Mechanistic study indicated that perturbation of the ROCK1/PTEN/PI3K/Akt signalling pathway plays a vital role in the arctiin-mediated anti-proliferative effect. Furthermore, pre-incubation of HRCECs with Y-27632 attenuated arctiin-induced cell cycle arrest, cell proliferation and tube formation inhibition. Y-27632 also reversed the activation of PTEN, the inactivation/dephosphorylation of PI3K/Akt and down-regulation of VEGF. Taken together, the results demonstrated that arctiin inhibits the proliferation of HG-induced HRCECs through the activation of ROCK1 and PTEN and inactivation of PI3K and Akt, resulting in down-regulation of VEGF, which inhibits endothelial cell proliferation.
Collapse
Affiliation(s)
- Min Zhou
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqingChina
- Department of PharmacyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Guobing Li
- Department of PharmacyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Liancai Zhu
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqingChina
| | - Huyue Zhou
- Department of PharmacyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Laichun Lu
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqingChina
| |
Collapse
|
43
|
Wang ZJ, Chang LL, Wu J, Pan HM, Zhang QY, Wang MJ, Xin XM, Luo SS, Chen JA, Gu XF, Guo W, Zhu YZ. A Novel Rhynchophylline Analog, Y396, Inhibits Endothelial Dysfunction Induced by Oxidative Stress in Diabetes Through Epidermal Growth Factor Receptor. Antioxid Redox Signal 2020; 32:743-765. [PMID: 31892280 DOI: 10.1089/ars.2018.7721] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Aims: Endothelial dysfunction appears in early diabetes mellitus partially because of epidermal growth factor receptor (EGFR) abnormal activation and downstream oxidative stress. The aim of this study was to determine whether Y396, a synthesized analog of rhynchophylline, could protect against endothelial dysfunction in diabetes and the underlying molecular mechanism. Results: Y396 could directly target the EGFR and inhibit its phosphorylation induced by high glucose and EGF, downstream translocation to the nucleus of E2F1, and its transcriptional activity and expression of Nox4. Diabetes-induced endothelium malfunction was ameliorated by Y396 treatment through EGFR inhibition. Downstream oxidative stress was decreased by Y396 in the aortas of type 1 diabetes mellitus mice and primary rat aorta endothelial cells (RAECs). Y396 could also ameliorate tunicamycin-induced oxidative stress in the aorta and RAECs. In addition, we again determined the protective effects of Y396 on high-fat diet/streptozotocin-induced type 2 diabetes mellitus. Innovation: This is the first study to demonstrate that Y396, a novel rhynchophylline analog, suppressed high-glucose-induced endothelial malfunction both in vivo and in vitro by inhibiting abnormal phosphorylation of EGFR. Our work uncovered EGFR as a novel therapeutic target and Y396 as a potential therapy against diabetes-induced complication. Conclusion: Y396 could directly bind with EGFR, and inhibit its phosphorylation and downstream E2F1 transcriptional activity. It could also preserve tunicamycin-evoked endothelial dysfunction and oxidative stress. It could protect against diabetes-induced endothelium malfunction in vivo through EGFR inhibition and downstream oxidative stress. Antioxid. Redox Signal. 32, 743-765.
Collapse
Affiliation(s)
- Zhi-Jun Wang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| | - Ling-Ling Chang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Jian Wu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Hong-Ming Pan
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Qiu-Yan Zhang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, China
| | - Min-Jun Wang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Xiao-Ming Xin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Shan-Shan Luo
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Ji-An Chen
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Xian-Feng Gu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Wei Guo
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Yi-Zhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, People's Republic of China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
44
|
Hu J, Zhang H, Li J, Jiang X, Zhang Y, Wu Q, Shen L, Shi J, Gao N. ROCK1 activation-mediated mitochondrial translocation of Drp1 and cofilin are required for arnidiol-induced mitochondrial fission and apoptosis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:37. [PMID: 32075676 PMCID: PMC7031977 DOI: 10.1186/s13046-020-01545-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/14/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Arnidiol is a pentacyclic triterpene diol that has multiple pharmacological activities. However, the apoptotic activities of arnidiol in human cancer cells have not yet been explored, nor has the mechanism by which arnidiol induces apoptosis been examined in depth. METHODS MDA-MB-231 cells and xenografted mice were treated with arnidiol. Mitochondrial fission and apoptosis were determined by immunofluorescence, flow cytometry and related molecular biological techniques. The interaction and colocalization of cofilin and Drp1 was determined by immunoprecipitation and immunofluorescence assays. RESULTS Arnidiol induces mitochondrial fission and apoptosis through mitochondrial translocation of Drp1 and cofilin. Importantly, the interaction of Drp1 and cofilin in mitochondria is involved in arnidiol-induced mitochondrial fission and apoptosis. Knockdown of either Drp1 or cofilin abrogated arnidiol-induced mitochondrial translocation, interaction of Drp1 and cofilin, mitochondrial fission and apoptosis. Only dephosphorylated Drp1 (Ser637) and cofilin (Ser3) were translocated to the mitochondria. Mutants of Drp1 S637A and cofilin S3A, which mimic the dephosphorylated forms, enhanced mitochondrial fission and apoptosis induced by arnidiol, whereas mutants of Drp1 S637D and cofilin S3E, which mimic the phosphorylated forms, suppressed mitochondrial fission and apoptosis induced by arnidiol. A mechanistic study revealed that ROCK1 activation plays an important role in the arnidiol-mediated Drp1 and cofilin dephosphorylation and mitochondrial translocation, mitochondrial fission, and apoptosis. CONCLUSIONS Our data reveal a novel role of both Drp1 and cofilin in the regulation of mitochondrial fission and apoptosis and suggest that arnidiol could be developed as a potential agent for the treatment of human cancer.
Collapse
Affiliation(s)
- Jinjiao Hu
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Hongwei Zhang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Jie Li
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xiuxing Jiang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yanhao Zhang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Liwen Shen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Ning Gao
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
45
|
Zhang X, Xu L, Yang T. miR-31 Modulates Liver Cancer HepG2 Cell Apoptosis and Invasion via ROCK1/F-Actin Pathways. Onco Targets Ther 2020; 13:877-888. [PMID: 32099392 PMCID: PMC6996230 DOI: 10.2147/ott.s227467] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/23/2019] [Indexed: 01/27/2023] Open
Abstract
PURPOSE Liver cancer is one of the most common malignant tumor in the world. miR-31 is downregulated in liver cancer and associated with tumor growth and metastasis. However, the underlying mechanism remains unclear. METHODS Cellular apoptosis was detected via MTT, TUNEL assay, LDH release and Annexin V/PI flow-cytometry analysis. Cellular migration and invasion were measured by the Transwell chamber assay. Mitochondrial functions were evaluated via mitochondrial membrane potential JC-1 staining and mPTP opening assessment. The mitophagy activity was examined via Western blots. RESULTS In the present study, our results confirm that miR-31 promotes apoptosis and inhibits proliferation and metastasis in liver cancer HepG2 cells. In vitro, miR-31 promotes HepG2 cell apoptosis through the mitochondrial pathway as indicated by mitochondrial potential reduction, increased mPTP opening time, cty-c release and imbalance of pro- and anti-apoptotic proteins. Furthermore, miR-31 reduces the energy generation by inhibiting mitochondrial respiratory function. At last, it is demonstrated that miR-31 triggers the mitochondrial damage via ROCK1/F-actin pathway. Inhibiting the ROCK1/F-actin pathway abolishes the effects of miR-31 mimic on mitochondrial injury, apoptosis, proliferation arrest and migration inhibition. CONCLUSION Our results reveal that miR-31 can inhibit HepG2 cell survival and metastasis by activating the ROCK1/F-actin pathway.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning110042, People’s Republic of China
| | - Lan Xu
- Department of Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning110042, People’s Republic of China
| | - Ting Yang
- Department of Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning110042, People’s Republic of China
| |
Collapse
|
46
|
Two folate-derived analogues from an aqueous decoction of Uncaria rhynchophylla. Chin J Nat Med 2019; 17:928-934. [DOI: 10.1016/s1875-5364(19)30115-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Indexed: 11/18/2022]
|
47
|
Zheng Z, Xiang S, Wang Y, Dong Y, Li Z, Xiang Y, Bian Y, Feng B, Yang B, Weng X. NR4A1 promotes TNF‑α‑induced chondrocyte death and migration injury via activating the AMPK/Drp1/mitochondrial fission pathway. Int J Mol Med 2019; 45:151-161. [PMID: 31746366 PMCID: PMC6889925 DOI: 10.3892/ijmm.2019.4398] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022] Open
Abstract
Nuclear receptor subfamily 4 group A member 1 (NR4A1)-induced chondrocyte death plays a critical role in the development of osteoarthritis through poorly defined mechanisms. The present study aimed to investigate the role of NR4A1 in regulating chondrocyte death in response to tumor necrosis factor-α (TNF-α) and cycloheximide (CHX) treatment, with a focus on mitochondrial fission and the AMP-activated protein kinase (AMPK) signaling pathway. The results demonstrated that NR4A1 was significantly upregulated in TNF-α and CHX exposed chondrocytes. Increased NR4A1 triggered mitochondrial fission via the AMPK/dynamin-related protein 1 (Drp1) pathway, resulting in mitochondrial dysfunction, and mitochondrial permeability transition pore (mPTP) opening-related cell death. Furthermore, excessive mitochondrial fission impaired chondrocyte migration through imbalance of F-actin homeo-stasis. Inhibiting NR4A1 attenuated TNF-α and CHX-induced mitochondrial fission and, thus, reduced mitochondrial dysfunction in chondrocytes, mPTP opening-related cell death and migration injury. Altogether, the present data confirmed that mitochondrial fission was involved in NR4A1-mediated chondrocyte injury via regulation of mitochondrial dysfunction, mPTP opening-induced cell death and F-actin-related migratory inhibition.
Collapse
Affiliation(s)
- Zhibo Zheng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Shuai Xiang
- Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Yingjie Wang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yulei Dong
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Zeng Li
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yongbo Xiang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yanyan Bian
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Bin Feng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Bo Yang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xisheng Weng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
48
|
Wu N, Zhang X, Bao Y, Yu H, Jia D, Ma C. Down-regulation of GAS5 ameliorates myocardial ischaemia/reperfusion injury via the miR-335/ROCK1/AKT/GSK-3β axis. J Cell Mol Med 2019; 23:8420-8431. [PMID: 31625671 PMCID: PMC6850918 DOI: 10.1111/jcmm.14724] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/19/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022] Open
Abstract
Growth arrest‐specific transcript 5 (GAS5), along non‐coding RNA (LncRNA), is highly expressed in hypoxia/reoxygenation (H/R)‐cardiomyocytes and promotes H/R‐induced apoptosis. In this study, we determined whether down‐regulation of GAS5 ameliorates myocardial ischaemia/reperfusion (I/R) injury and further explored its mechanism. GAS5 expression in cardiomyocytes and rats was knockdown by transfected or injected with GAS5‐specific small interfering RNA or adeno‐associated virus delivering small hairpin RNAs, respectively. The effects of GAS5 knockdown on myocardial I/R injury were detected by CCK‐8, myocardial enzyme test, flow cytometry, TTC and terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) staining. qRT‐PCR and luciferase reporter assay were carried out to analyse the relationship between GAS5 and miR‐335. The regulation of GAS5 on Rho‐associated protein kinase 1 (ROCK1) expression, the activation of PI3K/AKT/GSK‐3β pathway and mitochondrial permeability transition pore (mPTP) opening was further evaluated. The results indicated that GAS5 knockdown enhanced the viability, decreased apoptosis and reduced the levels of lactate dehydrogenase and creatine kinase‐MB in H/R‐treatment cardiomyocytes. Meanwhile, down‐regulation of GAS5 limited myocardial infarct size and reduced apoptosis in I/R‐heart. GAS5 was found to bind to miR‐335 and displayed a reciprocal inhibition between them. Furthermore, GAS5 knockdown repressed ROCK1 expression, activated PI3K/AKT, thereby leading to inhibition of GSK‐3β and mPTP opening. These suppressions were abrogated by miR‐335 inhibitor treatment. Taken together, our results demonstrated that down‐regulation of GAS5 ameliorates myocardial I/R injury via the miR‐335/ROCK1/AKT/GSK‐3β axis. Our findings suggested that GAS5 may be a new therapeutic target for the prevention of myocardial I/R injury.
Collapse
Affiliation(s)
- Nan Wu
- The Central Laboratory of the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaowen Zhang
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yandong Bao
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hang Yu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dalin Jia
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
49
|
Hu C, Zhou H, Liu Y, Huang J, Liu W, Zhang Q, Tang Q, Sheng F, Li G, Zhang R. ROCK1 promotes migration and invasion of non‑small‑cell lung cancer cells through the PTEN/PI3K/FAK pathway. Int J Oncol 2019; 55:833-844. [PMID: 31485605 PMCID: PMC6741846 DOI: 10.3892/ijo.2019.4864] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Rho-associated protein kinase 1 (ROCK1), a member of the ROCK family, serves an important function in cell migration and invasion in neoplasms. ROCK1 has been found to be overexpressed in several types of cancers. However, the role of ROCK1 in non-small-cell lung cancer (NSCLC) is poorly understood. In the present study, ROCK1 was found to be overexpressed in NSCLC cells and tissues, and it was associated with poor survival of NSCLC patients. Subsequently, ROCK1 knockdown NSCLC cell lines were established using shRNA. ROCK1 knockdown significantly reduced the migration and invasion ability in the cell monolayer scratching and Transwell assays. ROCK1 knockdown was also found to markedly inhibit cell adhesion ability. Moreover, the phosphorylation of focal adhesion kinase (FAK) was inhibited by ROCK1 knockdown, reducing NSCLC cell migration and invasion ability. This mechanistic study revealed that ROCK1 significantly enhanced cell migration and invasion by inhibiting the phosphatase and tensin homolog (PTEN)/phosphoinositide 3-kinase (PI3K)/FAK pathway. More importantly, the interruption of the PTEN/PI3K/FAK pathway markedly rescued the inhibition of cell migration and invasion mediated by ROCK1 knockdown. Taken together, these results suggest a novel role for ROCK1 in cell migration and invasion by inhibiting cell adhesion ability, and indicate that ROCK1 may be of value as a therapeutic target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Changpeng Hu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Huyue Zhou
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Yali Liu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Jingbin Huang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Wuyi Liu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Qian Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Qin Tang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Fangfang Sheng
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
50
|
The Role of Herbal Bioactive Components in Mitochondria Function and Cancer Therapy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3868354. [PMID: 31308852 PMCID: PMC6594309 DOI: 10.1155/2019/3868354] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/27/2019] [Accepted: 06/02/2019] [Indexed: 12/13/2022]
Abstract
Mitochondria are highly dynamic double-membrane organelles which play a well-recognized role in ATP production, calcium homeostasis, oxidation-reduction (redox) status, apoptotic cell death, and inflammation. Dysfunction of mitochondria has long been observed in a number of human diseases, including cancer. Targeting mitochondria metabolism in tumors as a cancer therapeutic strategy has attracted much attention for researchers in recent years due to the essential role of mitochondria in cancer cell growth, apoptosis, and progression. On the other hand, a series of studies have indicated that traditional medicinal herbs, including traditional Chinese medicines (TCM), exert their potential anticancer effects as an effective adjunct treatment for alleviating the systemic side effects of conventional cancer therapies, for reducing the risk of recurrence and cancer mortality and for improving the quality of patients' life. An amazing feature of these structurally diverse bioactive components is that majority of them target mitochondria to provoke cancer cell-specific death program. The aim of this review is to summarize the in vitro and in vivo studies about the role of these herbs, especially their bioactive compounds in the modulation of the disturbed mitochondrial function for cancer therapy.
Collapse
|