1
|
Qi K, Li J, Hu Y, Qiao Y, Mu Y. Research progress in mechanism of anticancer action of shikonin targeting reactive oxygen species. Front Pharmacol 2024; 15:1416781. [PMID: 39076592 PMCID: PMC11284502 DOI: 10.3389/fphar.2024.1416781] [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: 04/13/2024] [Accepted: 06/13/2024] [Indexed: 07/31/2024] Open
Abstract
Excessive buildup of highly reactive molecules can occur due to the generation and dysregulation of reactive oxygen species (ROS) and their associated signaling pathways. ROS have a dual function in cancer development, either leading to DNA mutations that promote the growth and dissemination of cancer cells, or triggering the death of cancer cells. Cancer cells strategically balance their fate by modulating ROS levels, activating pro-cancer signaling pathways, and suppressing antioxidant defenses. Consequently, targeting ROS has emerged as a promising strategy in cancer therapy. Shikonin and its derivatives, along with related drug carriers, can impact several signaling pathways by targeting components involved with oxidative stress to induce processes such as apoptosis, necroptosis, cell cycle arrest, autophagy, as well as modulation of ferroptosis. Moreover, they can increase the responsiveness of drug-resistant cells to chemotherapy drugs, based on the specific characteristics of ROS, as well as the kind and stage of cancer. This research explores the pro-cancer and anti-cancer impacts of ROS, summarize the mechanisms and research achievements of shikonin-targeted ROS in anti-cancer effects and provide suggestions for designing further anti-tumor experiments and undertaking further experimental and practical research.
Collapse
Affiliation(s)
- Ke Qi
- Department of Diagnostic Clinical Laboratory Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Jiayi Li
- Department of Clinical Test Center, Medical Laboratory, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yang Hu
- Department of Diagnostic Clinical Laboratory Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yiyun Qiao
- Department of Clinical Test Center, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yongping Mu
- Department of Clinical Test Center, Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
2
|
Han L, Xiang X, Fu Y, Wei S, Zhang C, Li L, Liu Y, Lv H, Shan B, Zhao L. Periplcymarin targets glycolysis and mitochondrial oxidative phosphorylation of esophageal squamous cell carcinoma: Implication in anti-cancer therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155539. [PMID: 38522311 DOI: 10.1016/j.phymed.2024.155539] [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: 10/16/2023] [Revised: 01/28/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is the predominant histological subtype of esophageal cancer (EC) in China, and demonstrates varying levels of resistance to multiple chemotherapeutic agents. Our previous studies have proved that periplocin (CPP), derived from the extract of cortex periplocae, exhibiting the capacity to hinder proliferation and induce apoptosis in ESCC cells. Several studies have identified additional anti-cancer constituents in the extract of cortex periplocae, named periplcymarin (PPM), sharing similar compound structure with CPP. Nevertheless, the inhibitory effects of PPM on ESCC and their underlying mechanisms remain to be further elucidated. PURPOSE The aim of this study was to investigate function of PPM inhibiting the growth of ESCC in vivo and in vitro and to explore its underlying mechanism, providing the potential anti-tumor drug for ESCC. METHODS Initially, a comparative analysis was conducted on the inhibitory activity of three naturally compounds obtained from the extract of cortex periplocae on ESCC cells. Among these compounds, PPM was chosen for subsequent investigation owing to its comparatively structure and anti-tumor activity simultaneously. Subsequently, a series of biological functional experiments were carried out to assess the impact of PPM on the proliferation, apoptosis and cell cycle arrest of ESCC cells in vitro. In order to elucidate the molecular mechanism of PPM, various methodologies were employed, including bioinformatics analyses and mechanistic experiments such as high-performance liquid chromatography combined with mass spectrometry (HPLC-MS), cell glycolysis pressure and mitochondrial pressure test. Additionally, the anti-tumor effects of PPM on ESCC cells and potential toxic side effects were evaluated in vivo using the nude mice xenograft assay. RESULTS Our study revealed that PPM possesses the ability to impede the proliferation of ESCC cells, induce apoptosis, and arrest the cell cycle of ESCC cells in the G2/M phase in vitro. Mechanistically, PPM exerted its effects by modulating glycolysis and mitochondrial oxidative phosphorylation (OXPHOS), as confirmed by glycolysis pressure and mitochondrial pressure tests. Moreover, rescue assays demonstrated that PPM inhibits glycolysis and OXPHOS in ESCC cells through the PI3K/AKT and MAPK/ERK signaling pathways. Additionally, we substantiated that PPM effectively suppresses the growth of ESCC cells in vivo, with only modest potential toxic side effects. CONCLUSION Our study provides novel evidence that PPM has the potential to simultaneously target glycolysis and mitochondrial OXPHOS in ESCC cells. This finding highlights the need for further investigation into PPM as a promising therapeutic agent that targets the tumor glucose metabolism pathway in ESCC.
Collapse
Affiliation(s)
- Lujuan Han
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Department of Pathogenic Biology, Hebei Medical University, Zhongshan Road 361, Shijiazhuang, 050017, PR China
| | - Xiaohan Xiang
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Yuhui Fu
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Sisi Wei
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Cong Zhang
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Lei Li
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Yueping Liu
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China
| | - Huilai Lv
- Department of Thoracic Surgery, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China
| | - Baoen Shan
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China.
| | - Lianmei Zhao
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China.
| |
Collapse
|
3
|
Cho KH, Kim JE, Bahuguna A, Kang DJ. Long-Term Supplementation of Ozonated Sunflower Oil Improves Dyslipidemia and Hepatic Inflammation in Hyperlipidemic Zebrafish: Suppression of Oxidative Stress and Inflammation against Carboxymethyllysine Toxicity. Antioxidants (Basel) 2023; 12:1240. [PMID: 37371970 DOI: 10.3390/antiox12061240] [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: 05/11/2023] [Revised: 05/30/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Ozonated sunflower oil (OSO) is a well-known functional oil with antioxidant, antimicrobial, anti-allergic, and skin-moisturizing properties. However, studies on the effects of OSO on high-cholesterol diet (HCD)-induced metabolic disorders have been scarce. In the current study, we aimed to determine the anti-inflammatory effects of OSO on lipid metabolism in adult hypercholesterolemic zebrafish and its embryos. Microinjection of OSO (final 2%, 10 nL) into zebrafish embryos under the presence of carboxymethyllysine (CML, 500 ng) protected acute embryo death up to 61% survival, while sunflower oil (final 2%) showed much less protection at around 42% survival. The microinjection of OSO was more effective than SO to inhibit reactive oxygen species (ROS) production and apoptosis in the CML induced embryo toxicity. Intraperitoneal injection of OSO under the presence of CML protected acute death from CML-induced neurotoxicity with improved hepatic inflammation, less detection of ROS and interleukin (IL)-6, and lowering blood total cholesterol (TC) and triglyceride (TG), while the SO-injected group did not protect the CML-toxicity. Long-term supplementation of OSO (final 20%, wt/wt) with HCD for 6 months resulted in higher survivability than the HCD alone group or HCD + SO group (final 20%, wt/wt) with significant lowering of plasma TC and TG levels. The HCD + OSO group showed the least hepatic inflammation, fatty liver change, ROS, and IL-6 production. In conclusion, short-term treatment of OSO by injection exhibited potent anti-inflammatory activity against acute neurotoxicity of CML in zebrafish and their embryo. Long-term supplementation of OSO in the diet also revealed the highest survivability and blood lipid-lowering effect through potent antioxidant and anti-inflammatory activity.
Collapse
Affiliation(s)
- Kyung-Hyun Cho
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
- LipoLab, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Ji-Eun Kim
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
| | - Ashutosh Bahuguna
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
| | - Dae-Jin Kang
- Raydel Research Institute, Medical Innovation Complex, Daegu 41061, Republic of Korea
| |
Collapse
|
4
|
Seo J, Lee DE, Kim SM, Kim E, Kim JK. Licochalcone A Exerts Anti-Cancer Activity by Inhibiting STAT3 in SKOV3 Human Ovarian Cancer Cells. Biomedicines 2023; 11:biomedicines11051264. [PMID: 37238935 DOI: 10.3390/biomedicines11051264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/31/2023] [Accepted: 04/18/2023] [Indexed: 05/28/2023] Open
Abstract
Licochalcone A (LicA), a major active component of licorice, has been reported to exhibit various pharmacological actions. The purpose of this study was to investigate the anticancer activity of LicA and detail its molecular mechanisms against ovarian cancer. SKOV3 human ovarian cancer cells were used in this study. Cell viability was measured using a cell counting kit-8 assay. The percentages of apoptotic cells and cell cycle arrest were determined by flow cytometry and Muse flow cytometry. The expression levels of proteins regulating cell apoptosis, cell cycle, and the signal transducer and activator of transcription 3 (STAT3) signaling pathways were examined using Western blotting analysis. The results indicated that LicA treatment inhibited the cell viability of SKOV3 cells and induced G2/M phase arrest. Furthermore, LicA induced an increase in ROS levels, a reduction in mitochondrial membrane potential, and apoptosis accompanied by an increase in cleaved caspases and cytoplasmic cytochrome c. Additionally, LicA caused a dramatic decrease in STAT3 protein levels, but not mRNA levels, in SKOV3 cells. Treatment with LicA also reduced phosphorylation of the mammalian target of rapamycin and eukaryotic translation initiation factor 4E-binding protein in SKOV3 cells. The anti-cancer effects of LicA on SKOV3 cells might be mediated by reduced STAT3 translation and activation.
Collapse
Affiliation(s)
- Jeonghyeon Seo
- Department of Biomedical Science, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| | - Da Eun Lee
- Department of Biomedical Science, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| | - Seong Mi Kim
- Department of Biomedical Science, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| | - Eunjung Kim
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| | - Jin-Kyung Kim
- Department of Biomedical Science, Daegu Catholic University, Gyeongsan-si 38430, Republic of Korea
| |
Collapse
|
5
|
Boonnate P, Kariya R, Okada S. Shikonin Induces ROS-Dependent Apoptosis Via Mitochondria Depolarization and ER Stress in Adult T Cell Leukemia/Lymphoma. Antioxidants (Basel) 2023; 12:antiox12040864. [PMID: 37107239 PMCID: PMC10135058 DOI: 10.3390/antiox12040864] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/18/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Adult T cell leukemia/lymphoma (ATLL) is an aggressive T-cell malignancy that develops in some elderly human T-cell leukemia virus (HTVL-1) carriers. ATLL has a poor prognosis despite conventional and targeted therapies, and a new safe and efficient therapy is required. Here, we examined the anti-ATLL effect of Shikonin (SHK), a naphthoquinone derivative that has shown several anti-cancer activities. SHK induced apoptosis of ATLL cells accompanied by generation of reactive oxygen species (ROS), loss of mitochondrial membrane potential, and induction of endoplasmic reticulum (ER) stress. Treatment with a ROS scavenger, N-acetylcysteine (NAC), blocked both loss of mitochondrial membrane potential and ER stress, and prevented apoptosis of ATLL cells, indicating that ROS is an upstream trigger of SHK-induced apoptosis of ATLL cells through disruption of the mitochondrial membrane potential and ER stress. In an ATLL xenografted mouse model, SHK treatment suppressed tumor growth without significant adverse effects. These results suggest that SHK could be a potent anti-reagent against ATLL.
Collapse
Affiliation(s)
- Piyanard Boonnate
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| | - Ryusho Kariya
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
6
|
Li S, Li Z, Li Y, Zhu Y, Han J, Li W, Jin N, Fang J, Li X, Zhu G. A comparative study of the ability of recombinant oncolytic adenovirus, doxorubicin and tamoxifen to inhibit the proliferation of breast cancer cells. J Cell Mol Med 2022; 26:5222-5234. [PMID: 36148613 PMCID: PMC9575116 DOI: 10.1111/jcmm.17549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/31/2022] [Accepted: 08/13/2022] [Indexed: 11/29/2022] Open
Abstract
In this study, we compared the inhibitory effects of recombinant oncolytic adenovirus (Ad‐apoptin‐hTERTp‐E1a, Ad‐VT) with that of doxorubicin (DOX), a first‐line chemotherapy drug, and tamoxifen (TAM), an endocrine therapy drug, on the proliferation of breast cancer cells. We found that Ad‐VT could effectively inhibit the proliferation of breast cancer cells (p < 0.01); the inhibition rate of Ad‐VT on normal mammary epithelial MCF‐10A cells was less than 20%. DOX can effectively inhibit the proliferation of breast cancer cells and also has a strong inhibitory effect on MCF‐10A cells (p < 0.01). TAM also has a strong inhibitory effect on breast cancer cells, among which the oestrogen‐dependent MCF‐7 cell inhibition was stronger (p < 0.01), At higher concentrations, TAM also had a high rate of inhibition (>70%) on the proliferation of MCF‐10A cells. We also found that both recombinant adenovirus and both drugs could successfully induce tumour cell apoptosis. Further Western blot results showed that the recombinant adenovirus killed breast cancer cells through the endogenous apoptotic pathway. Analysis of the nude mouse subcutaneous breast cancer model showed that Ad‐VT significantly inhibited tumour growth (the luminescence rate of cancer cells was reduced by more than 90%) and improved the survival rate of tumour‐bearing mice (p < 0.01). Compared with DOX and TAM, Ad‐VT has a significant inhibitory effect on breast cancer cells, but almost no inhibitory effect on normal breast epithelial cells, and this inhibitory effect is mainly through the endogenous apoptotic pathway. These results indicate that Ad‐VT has significant potential as a drug for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shanzhi Li
- Academiciann Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Zhuoxin Li
- Academiciann Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.,College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yiquan Li
- Academiciann Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Yilong Zhu
- Academiciann Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Jicheng Han
- Academiciann Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Wenjie Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ningyi Jin
- Academiciann Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jinbo Fang
- Academiciann Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | - Xiao Li
- Academiciann Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China.,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Guangze Zhu
- Academiciann Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
7
|
Valipour M. Recent advances of antitumor shikonin/alkannin derivatives: A comprehensive overview focusing on structural classification, synthetic approaches, and mechanisms of action. Eur J Med Chem 2022; 235:114314. [DOI: 10.1016/j.ejmech.2022.114314] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/20/2022] [Accepted: 03/20/2022] [Indexed: 12/22/2022]
|
8
|
Qi H, Zhang X, Liu H, Han M, Tang X, Qu S, Wang X, Yang Y. Shikonin induced Apoptosis Mediated by Endoplasmic Reticulum Stress in Colorectal Cancer Cells. J Cancer 2022; 13:243-252. [PMID: 34976186 PMCID: PMC8692675 DOI: 10.7150/jca.65297] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
Shikonin is a naphthoquinone pigment isolated from the root of Lithospermum erythrorhizon, which has displayed potent anti-tumor properties. However, the effects of shikonin in colorectal cancer cells have not been yet fully investigated. In this study, we demonstrated that shikonin significantly inhibited the activity of colorectal cancer cells in a time- and dose-dependent manner. The flow cytometry and western blot results indicated that shikonin induced cell apoptosis by down-regulating BCL-2 and activating caspase-3/9 and the cleavage of PARP. The expression of BiP and the PERK/elF2α/ATF4/CHOP and IRE1α /JNK signaling pathways were upregulated after shikonin treatment. The pre-treatment with N-acetyl cysteine significantly reduced the cytotoxicity of shikonin. Taken together, shikonin could inhibit proliferation of the colorectal cancer cell through the activation of ROS mediated-ER stress. The in vivo results showed that shikonin effectively inhibited tumor growth in the HCT-116 and HCT-15 xenograft models. In conclusion, shikonin inhibited the proliferation of colorectal cancer cells in vitro and in vivo and warrants future investigation.
Collapse
Affiliation(s)
- Hui Qi
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.,Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Xing Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Huanhuan Liu
- Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Meng Han
- Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Xuzhen Tang
- Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Shulan Qu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Xiaoyu Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Yifu Yang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| |
Collapse
|
9
|
ROS mediated apoptotic pathways in primary effusion lymphoma: Comment on induction of apoptosis by Shikonin through ROS-mediated intrinsic and extrinsic pathways in primary effusion lymphoma. Transl Oncol 2021; 14:101061. [PMID: 33857745 PMCID: PMC8050935 DOI: 10.1016/j.tranon.2021.101061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 11/21/2022] Open
|