1
|
Alaei A, Solali S, Mirzapour MM. Study of TRAIL and SAHA Co-Treatment on Leukemia K562 Cell Line. Cell Biochem Biophys 2024:10.1007/s12013-024-01543-y. [PMID: 39392552 DOI: 10.1007/s12013-024-01543-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 10/12/2024]
Abstract
TRAIL (Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand) is an attractive agent being considered a potential cancer treatment. It attaches to its death receptors, leading many cancer cells to apoptosis. However, some malignancies indicate substantial resistance to TRAIL, challenging anticancer scientists. Herein, combination therapy with TRAIL plus SAHA (Suberoyl Anilide Hydroxamic Acid) was conducted to evaluate the capability of SAHA to overcome TRAIL resistance in the leukemia K562 cell line. First, the IC50 for SAHA was calculated (2 µM) at 12, 24, 48, and 72 h of treatment using MTT assay. Second, the K562 cells were treated with concentrations of 50 and 100 nM of TRAIL and 2 μM of SAHA separately and together for 24, 48, and 72 h and the survival of these cells was evaluated by Flowcytometry following the annexin-V and PI staining. To demonstrate the non-toxicity of the combined treatment for normal cells, the HEK-293 cell line was treated with the TRAIL 100 nM and SAHA 2 μM combined and separated at the same periods. In the end, by performing real-time PCR, the amount of candidate genes' expression implicated in TRAIL resistance, and the levels of BCR-ABL expression was measured. The drug dosages were not toxic to normal cells. SAHA plus TRAIL strongly triggered apoptosis in K562 cells after 24, 48, and 72 h of exposure. Furthermore, it was shown that DR4, DR5, and CHOP expressions were enhanced, and PI3K, Akt, ERK, STAT3, c-FLIPL, NF-κB, and BCR-ABL expressions were decreased by SAHA in K562 cells. Our study indicated that SAHA combined with TRAIL can increase the sensitivity of K562 leukemic cells to TRAIL by suppressing intracellular anti-apoptotic molecules and augmenting the expressions of DR4/DR5 and CHOP.
Collapse
Affiliation(s)
- Amirarsalan Alaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | |
Collapse
|
2
|
Chang HY, Hsu HC, Fang YH, Liu PY, Liu YW. Empagliflozin attenuates doxorubicin-induced cardiotoxicity by inhibiting the JNK signaling pathway. Biomed Pharmacother 2024; 176:116759. [PMID: 38788603 DOI: 10.1016/j.biopha.2024.116759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Sodium-glucose cotransporter-2 inhibitors, such as empagliflozin, are pivotal therapies for heart failure. However, the effect of empagliflozin on doxorubicin-related cardiac dysfunction remains unclear. METHODS Human induced pluripotent stem cell- and embryonic stem cell-derived cardiomyocytes were used to investigate the direct effect of empagliflozin on human cardiomyocytes. Then, the c-Jun amino-terminal kinases (JNK) inhibitor SP600125 was administered to the doxorubicin cardiotoxicity model in vitro and in vivo to investigate the role of JNK in empagliflozin. RESULTS In human stem cell-derived cardiomyocytes, pretreatment with empagliflozin attenuated doxorubicin-induced cleavage of caspase 3 and other apoptosis markers. Empagliflozin significantly attenuated doxorubicin-induced phosphorylation of JNK and p38. Inhibiting the phosphorylation of JNK (SP600125) or STAT3 attenuated doxorubicin-induced apoptosis, but inhibiting the phosphorylation of p38 did not. SP600125 inhibits the phosphorylation of STAT3 (S727), and a STAT3 (Y705) inhibitor also inhibits the phosphorylation of JNK. Empagliflozin and SP600125 attenuated doxorubicin-induced increases in reactive oxygen species (ROS) and decreases in oxidized nicotinamide adenine dinucleotide (NAD+). In animal studies, empagliflozin and SP600125 attenuated doxorubicin-induced cardiac dysfunction and fibrosis. CONCLUSIONS Empagliflozin attenuated doxorubicin-induced apoptosis by inhibiting the phosphorylation of JNK and its downstream signaling pathways, including ROS and NAD+.
Collapse
Affiliation(s)
- Hsien-Yuan Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Chun Hsu
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Hsien Fang
- Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ping-Yen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Yen-Wen Liu
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Cell Therapy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
3
|
Angulo-Elizari E, Henriquez-Figuereo A, Morán-Serradilla C, Plano D, Sanmartín C. Unlocking the potential of 1,4-naphthoquinones: A comprehensive review of their anticancer properties. Eur J Med Chem 2024; 268:116249. [PMID: 38458106 DOI: 10.1016/j.ejmech.2024.116249] [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: 01/15/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/10/2024]
Abstract
Cancer encompasses a group of pathologies with common characteristics, high incidence, and prevalence in all countries. Although there are treatments available for this disease, they are not always effective or safe, often failing to achieve the desired results. This is why it is necessary to continue the search for new therapies. One of the strategies for obtaining new antitumor drugs is the use of 1,4-naphthoquinone as a scaffold in synthetic or natural products with antitumor activity. This review focuses on compiling studies related to the antitumor activity of 1,4-naphthoquinone and its natural and synthetic derivatives over the last 10 years. The work describes the main natural naphthoquinones with antitumor activity and classifies the synthetic naphthoquinones based on the structural modifications made to the scaffold. Additionally, the formation of metal complexes using naphthoquinones as a ligand is considered. After a thorough review, 197 synthetic compounds with potent biological activity against cancer have been classified according to their chemical structures and their mechanisms of action have been described.
Collapse
Affiliation(s)
- Eduardo Angulo-Elizari
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Andreina Henriquez-Figuereo
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Cristina Morán-Serradilla
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain
| | - Daniel Plano
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain; Navarra Institute for Health Research (IdisNA), 31008, Pamplona, Spain.
| | - Carmen Sanmartín
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Sciences, Irunlarrea 1, 31008, Pamplona, Spain; Navarra Institute for Health Research (IdisNA), 31008, Pamplona, Spain.
| |
Collapse
|
4
|
Zarezadeh SM, Sharafi AM, Erabi G, Tabashiri A, Teymouri N, Mehrabi H, Golzan SA, Faridzadeh A, Abdollahifar Z, Sami N, Arabpour J, Rahimi Z, Ansari A, Abbasi MR, Azizi N, Tamimi A, Poudineh M, Deravi N. Natural STAT3 Inhibitors for Cancer Treatment: A Comprehensive Literature Review. Recent Pat Anticancer Drug Discov 2024; 19:403-502. [PMID: 37534488 DOI: 10.2174/1574892818666230803100554] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 08/04/2023]
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide, affecting millions of people physically and financially every year. Over time, many anticancer treatments have been proposed and studied, including synthetic compound consumption, surgical procedures, or grueling chemotherapy. Although these treatments have improved the daily life quality of patients and increased their survival rate and life expectancy, they have also shown significant drawbacks, including staggering costs, multiple side effects, and difficulty in compliance and adherence to treatment. Therefore, natural compounds have been considered a possible key to overcoming these problems in recent years, and thorough research has been done to assess their effectiveness. In these studies, scientists have discovered a meaningful interaction between several natural materials and signal transducer and activator of transcription 3 molecules. STAT3 is a transcriptional protein that is vital for cell growth and survival. Mechanistic studies have established that activated STAT3 can increase cancer cell proliferation and invasion while reducing anticancer immunity. Thus, inhibiting STAT3 signaling by natural compounds has become one of the favorite research topics and an attractive target for developing novel cancer treatments. In the present article, we intend to comprehensively review the latest knowledge about the effects of various organic compounds on inhibiting the STAT3 signaling pathway to cure different cancer diseases.
Collapse
Affiliation(s)
- Seyed Mahdi Zarezadeh
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Mohammad Sharafi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arefeh Tabashiri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Teymouri
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hoda Mehrabi
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Amirhossein Golzan
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Abdollahifar
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nafiseh Sami
- Student Research Committee, Tehran Medical Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of New Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Zahra Rahimi
- School of Medicine, Zanjan University of Medical Sciences Zanjan, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Nima Azizi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Lohberger B, Kaltenegger H, Eck N, Glänzer D, Leithner A, Kretschmer N. The Biological Assessment of Shikonin and β,β-dimethylacrylshikonin Using a Cellular Myxofibrosarcoma Tumor Heterogeneity Model. Int J Mol Sci 2023; 24:15910. [PMID: 37958891 PMCID: PMC10650664 DOI: 10.3390/ijms242115910] [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: 10/06/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Myxofibrosarcoma (MFS) is a subtype of soft tissue sarcoma of connective tissue, which is characterized by large intra-tumor heterogeneity. Therapy includes surgical resection. Additional chemotherapy is of limited effect. In this study, we demonstrated the potent anticancer activity of shikonin derivatives in our MFS cellular model of tumor heterogeneity for developing a new therapeutic approach. The impact of shikonin and β,β-dimethylacrylshikonin (DMAS) on viability, apoptotic induction, MAPK phosphorylation, and DNA damage response were analyzed by means of two human MFS cell lines, MUG-Myx2a and MUG-Myx2b, derived from a singular tumor tissue specimen. MFS cells showed a dose-dependent inhibition of cell viability and a significant induction of apoptosis. Treatment with shikonin derivatives caused an inhibition of pSTAT3 and an increase in pAKT, pERK, pJNK, and pp38. DMAS and shikonin inhibited the activation of the two master upstream regulators of the DNA damage response, ATR and ATM. MUG-Myx2b, which contains an additional PTEN mutation, was more sensitive in some targets. These data demonstrate the significant antitumorigenic effect of shikonin derivatives in MFS and highlight the importance of intra-tumor heterogeneity in treatment planning.
Collapse
Affiliation(s)
- Birgit Lohberger
- Department of Orthopedics and Trauma, Medical University of Graz, 8036 Graz, Austria; (H.K.); (N.E.); (D.G.); (A.L.)
| | - Heike Kaltenegger
- Department of Orthopedics and Trauma, Medical University of Graz, 8036 Graz, Austria; (H.K.); (N.E.); (D.G.); (A.L.)
| | - Nicole Eck
- Department of Orthopedics and Trauma, Medical University of Graz, 8036 Graz, Austria; (H.K.); (N.E.); (D.G.); (A.L.)
| | - Dietmar Glänzer
- Department of Orthopedics and Trauma, Medical University of Graz, 8036 Graz, Austria; (H.K.); (N.E.); (D.G.); (A.L.)
| | - Andreas Leithner
- Department of Orthopedics and Trauma, Medical University of Graz, 8036 Graz, Austria; (H.K.); (N.E.); (D.G.); (A.L.)
| | - Nadine Kretschmer
- Institute of Pharmaceutical Sciences, Pharmacognosy, University of Graz, 8010 Graz, Austria
| |
Collapse
|
6
|
Hua Z, Yang W, Li D, Cui Y, Shen L, Rao L, Zheng Y, Zhang Q, Zeng W, Gong Y, Yuan L. Metformin regulates the LIN28B‑mediated JNK/STAT3 signaling pathway through miR‑140‑3p in subretinal fibrosis. Exp Ther Med 2023; 26:528. [PMID: 37869644 PMCID: PMC10587880 DOI: 10.3892/etm.2023.12227] [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: 01/13/2023] [Accepted: 08/30/2023] [Indexed: 10/24/2023] Open
Abstract
Subretinal fibrosis (SF) is an important cause of submacular neovascularization that leads to permanent vision loss, but has no effective clinical treatment. The present study examined the influence of metformin on SF, and investigated whether the mechanism involves the microRNA (miR)-140-3p/LIN28B/JNK/STAT3-mediated regulation of oxidative stress, angiogenesis and fibrosis-associated indicators. A mouse model of laser-induced SF was established. In addition, an ARPE-19 fibrotic cell model was established using TGF-β1. A Cell Counting Kit-8 assay was used to examine cell viability. Flow cytometry was used to measure reactive oxygen species levels, and western blotting was used to detect the levels of proteins associated with epithelial-mesenchymal transition (EMT), signaling and fibrosis. The levels of superoxide dismutase, malondialdehyde, glutathione-peroxidase and catalase were measured using kits. Scratch assays and Transwell assays were used to assess cell migration and invasion, respectively, and reverse transcription-quantitative PCR was used to determine the levels of miR-140-3p and LIN28B. Dual-luciferase assays were used to verify the targeting relationship between miR-140-3p and LIN28B, and coimmunoprecipitation was used to confirm the interaction between LIN28B and JNK. Masson staining and hematoxylin and eosin staining were used to examine collagenous fibers and the histopathology of eye tissue. In ARPE-19 cells induced by TGF-β1, metformin promoted miR-140-3p expression and inhibited LIN28B expression and JNK/STAT3 pathway activation, thereby inhibiting oxidative stress, EMT and fibrosis in ARPE-19 cells. The overexpression of LIN28B or treatment with the JNK/STAT3 agonist anisomycin partially reversed the inhibitory effect of metformin on oxidative stress and fibrosis in ARPE-19 cells. The dual-luciferase reporter assay and coimmunoprecipitation assay showed that miR-140-3p targeted the 3' untranslated region of LIN28B mRNA and inhibited LIN28B expression. LIN28B targeted and bound to JNK and regulated the JNK/STAT3 pathway. Therefore, it may be concluded that metformin can promote miR-140-3p expression, inhibit LIN28B and then inhibit the JNK/STAT3 pathway to alleviate SF.
Collapse
Affiliation(s)
- Zhijuan Hua
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Department of Pediatric Ophthalmology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan 650021, P.R. China
| | - Wenchang Yang
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Dongli Li
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yixin Cui
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Lu Shen
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Lingna Rao
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yuxiang Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Qiying Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Wenyi Zeng
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yi Gong
- Department of Physiology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Ling Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
7
|
Zinnah KMA, Munna AN, Seol JW, Park BY, Park SY. An Antidepressant Drug Increased TRAIL Receptor-2 Expression and Sensitized Lung Cancer Cells to TRAIL-induced Apoptosis. Anticancer Agents Med Chem 2023; 23:2225-2236. [PMID: 37859313 PMCID: PMC10788920 DOI: 10.2174/0118715206262252231004110310] [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: 07/14/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND TRAIL has emerged as a promising therapeutic target due to its ability to selectively induce apoptosis in cancer cells while sparing normal cells. Autophagy, a highly regulated cellular recycling mechanism, is known to play a cell survival role by providing a required environment for the cell. Recent studies suggest that autophagy plays a significant role in increasing TRAIL resistance in certain cancer cells. Thus, regulating autophagy in TRAIL-mediated cancer therapy is crucial for its role in cancer treatment. OBJECTIVE Our study explored whether the antidepressant drug desipramine could enhance the ability of TRAIL to kill cancer cells by inhibiting autophagy. METHODS The effect of desipramine on TRAIL sensitivity was examined in various lung cancer cell lines. Cell viability was measured by morphological analysis, trypan blue exclusion, and crystal violet staining. Flow cytometry analysis was carried out to measure apoptosis with annexin V-PI stained cells. Western blotting, rtPCR, and immunocytochemistry were carried out to measure autophagy and death receptor expression. TEM was carried out to detect autophagy inhibition. RESULTS Desipramine treatment increased the TRAIL sensitivity in all lung cancer cell lines. Mechanistically, desipramine treatment induced death receptor expression to increase TRAIL sensitivity. This effect was confirmed when the genetic blockade of DR5 reduced the effect of desipramine in enhanced TRAIL-mediated cell death. Further investigation revealed that desipramine treatment increased the LC3 and p62 levels, indicating the inhibition of lysosomal degradation of autophagy. Notably, TRAIL, in combination with either desipramine or the autophagy inhibitor chloroquine, exhibited enhanced cytotoxicity compared to TRAIL treatment alone. CONCLUSION Our findings revealed the potential of desipramine to induce TRAIL-mediated cell death by autophagy impairment. This discovery suggests its therapeutic potential for inducing TRAIL-mediated cell death by increasing the expression of death receptors, which is caused by impairing autophagy.
Collapse
Affiliation(s)
- Kazi Mohammad Ali Zinnah
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
- Department of Animal and Fish Biotechnology, Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Ali Newaz Munna
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| | - Jae-Won Seol
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| | - Byung-Yong Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| |
Collapse
|
8
|
Mathieu V, Superchi S, Masi M, Scafato P, Kornienko A, Evidente A. In Vitro Effects of Fungal Phytotoxins on Cancer Cell Viability: First Insight into Structure Activity Relationship of a Potent Metabolite of Cochliobolus australiensis Radicinin. Toxins (Basel) 2022; 14:toxins14080517. [PMID: 36006179 PMCID: PMC9415302 DOI: 10.3390/toxins14080517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/15/2022] [Accepted: 07/23/2022] [Indexed: 01/18/2023] Open
Abstract
Natural compounds have always represented an important source for new drugs. Although fungi represent one such viable source, to date, no fungal metabolite has been marketed as an anticancer drug. Based on our work with phytotoxins as potential chemical scaffolds and our recent findings involving three phytopathogenic fungi, i.e., Cochliobolus australiensis, Kalmusia variispora and Hymenoscyphus fraxineus, herein, we evaluate the in vitro anti-cancer activity of the metabolites of these fungi by MTT assays on three cancer cell models harboring various resistance levels to chemotherapeutic drugs. Radicinin, a phytotoxic dihydropyranopyran-4,5-dione produced by Cochliobolus australiensis, with great potential for the biocontrol of the invasive weed buffelgrass (Cenchrus ciliaris), showed significant anticancer activity in the micromolar range. Furthermore, a SAR study was carried out using radicinin, some natural analogues and hemisynthetic derivatives prepared by synthetic methods developed as part of work aimed at the potential application of these molecules as bioherbicides. This investigation opens new avenues for the design and synthesis of novel radicinin analogues as potential anticancer agents.
Collapse
Affiliation(s)
- Veronique Mathieu
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Boulevard du Triomphe, Accès 2, 1050 Ixelles, Belgium
- ULB Cancer Research Center, Université Libre de Bruxelles (ULB), 1050 Bruxelles, Belgium
- Correspondence: (V.M.); (P.S.)
| | - Stefano Superchi
- Department of Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy;
| | - Marco Masi
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Napoli, Italy; (M.M.); (A.E.)
| | - Patrizia Scafato
- Department of Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy;
- Correspondence: (V.M.); (P.S.)
| | - Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA;
| | - Antonio Evidente
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte Sant’Angelo, Via Cintia 4, 80126 Napoli, Italy; (M.M.); (A.E.)
| |
Collapse
|
9
|
Lohberger B, Glänzer D, Kaltenegger H, Eck N, Leithner A, Bauer R, Kretschmer N, Steinecker-Frohnwieser B. Shikonin derivatives cause apoptosis and cell cycle arrest in human chondrosarcoma cells via death receptors and MAPK regulation. BMC Cancer 2022; 22:758. [PMID: 35820864 PMCID: PMC9275282 DOI: 10.1186/s12885-022-09857-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
Background Although chondrosarcoma is the second most common primary malignant bone tumor, treatment options are limited due to its extensive resistance to a chemo- and radiation therapy. Since shikonin has shown potent anticancer activity in various types of cancer cells, it represents a promising compound for the development of a new therapeutic approach. Methods The dose-relationships of shikonin and its derivatives acetylshikonin and cyclopropylshikonin on two human chondrosarcoma cell lines were measured using the CellTiter-Glo®. The changes in the cell cycle were presented by flow cytometry. Protein phosphorylation and expression apoptotic markers, MAPKs and their downstream targets were analyzed using western blotting and gene expression were evaluated using RT-qPCR. Results Chondrosarcoma cells showed a dose-dependent inhibition of cell viability after treatment with shikonin and its derivatives, with the strongest effect for shikonin and IC50 values of 1.3 ± 0.2 µM. Flow cytometric measurements revealed a G2/M arrest of the cells after treatment. Protein and gene expression analysis demonstrated a dose-dependent downregulation of survivin and XIAP, and an upregulation of Noxa, γH2AX, cleaved caspase-8, -9, -3, and -PARP. Furthermore, the expression of various death receptors was modulated. As MAPK signaling pathways play a key role in tumor biology, their phosphorylation pattern and their corresponding downstream gene regulation were analyzed. Treatment with shikonin derivatives caused an inhibition of pSTAT3 and an increase of pAKT and the MAPKs pERK, pJNK, and pp38 in a dose-dependent manner. Conclusions These data demonstrated the significant anti-tumorigenic effect of shikonin derivatives in chondrosarcoma and encourage further research. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09857-x.
Collapse
Affiliation(s)
- Birgit Lohberger
- Department of Orthopedics and Trauma, Medical University of Graz, 8036, Graz, Austria. .,Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Saalfelden, Austria.
| | - Dietmar Glänzer
- Department of Orthopedics and Trauma, Medical University of Graz, 8036, Graz, Austria.,Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Saalfelden, Austria
| | - Heike Kaltenegger
- Department of Orthopedics and Trauma, Medical University of Graz, 8036, Graz, Austria
| | - Nicole Eck
- Department of Orthopedics and Trauma, Medical University of Graz, 8036, Graz, Austria.,Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Saalfelden, Austria
| | - Andreas Leithner
- Department of Orthopedics and Trauma, Medical University of Graz, 8036, Graz, Austria
| | - Rudolf Bauer
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Graz, Austria
| | - Nadine Kretschmer
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, University of Graz, Graz, Austria
| | | |
Collapse
|
10
|
Wang H, Ai J, Shopit A, Niu M, Ahmed N, Tesfaldet T, Tang Z, Li X, Jamalat Y, Chu P, Peng J, Ma X, Qaed E, Han G, Zhang W, Wang J, Tang Z. Protection of pancreatic β-cell by phosphocreatine through mitochondrial improvement via the regulation of dual AKT/IRS-1/GSK-3β and STAT3/Cyp-D signaling pathways. Cell Biol Toxicol 2022; 38:531-551. [PMID: 34455488 DOI: 10.1007/s10565-021-09644-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 08/09/2021] [Indexed: 10/20/2022]
Abstract
Diabetes mellitus (DM) is a metabolic syndrome, caused by insufficient insulin secretion or insulin resistance (IR). DM enhances oxidative stress and induces mitochondrial function in different kinds of cell types, including pancreatic β-cells. Our previous study has showed phosphocreatine (PCr) can advance the mitochondrial function through enhancing the oxidative phosphorylation and electron transport ability in mitochondria damaged by methylglyoxal (MG). Our aim was to explore the potential role of PCr as a molecule to protect mitochondria from diabetes-induced pancreatic β-cell injury with insulin secretion deficiency or IR through dual AKT/IRS-1/GSK-3β and STAT3/Cyclophilin D (Cyp-D) signaling pathways. MG-induced INS-1 cell viability, apoptosis, mitochondrial division and fusion, the morphology, and function of mitochondria were suppressed. Flow cytometry was used to detect the production of intracellular reactive oxygen species (ROS) and the changes of intracellular calcium, and the respiratory function was measured by oxygraph-2k. The expressions of AKT, IRS-1, GSK-3β, STAT3, and Cyp-D were detected using Western blot. The result showed that the oxidative stress-related kinases were significantly restored to the normal level after the pretreatment with PCr. Moreover, PCr pretreatment significantly inhibited cell apoptosis, decreased intracellular calcium, and ROS production, and inhibited mitochondrial division and fusion, and increased ATP synthesis damaged by MG in INS-1 cells. In addition, pretreatment with PCr suppressed Cytochrome C, p-STAT3, and Cyp-D expressions, while increased p-AKT, p-IRS-1, p-GSK-3β, caspase-3, and caspase-9 expressions. In conclusion, PCr has protective effect on INS-1 cells in vitro and in vivo, relying on AKT mediated STAT3/ Cyp-D pathway to inhibit oxidative stress and restore mitochondrial function, signifying that PCr might become an emerging candidate for the cure of diabetic pancreatic cancer β-cell damage.
Collapse
Affiliation(s)
- Hongyan Wang
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Jie Ai
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Abdullah Shopit
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Mengyue Niu
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Nisar Ahmed
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Tsehaye Tesfaldet
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | | | - Xiaodong Li
- Second Clinical College, Dalian Medical University, Dalian, China
| | - Yazeed Jamalat
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Peng Chu
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Jinyong Peng
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Xiaodong Ma
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Eskandar Qaed
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Guozhu Han
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China
| | - Weisheng Zhang
- First Clinical College, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China.
| | - Jun Wang
- Department of Pathophysiology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China.
| | - Zeyao Tang
- Acad Integrated Med & College of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 Western Section, Lvshun South Street, Dalian, 116044, China.
| |
Collapse
|
11
|
Zhang N, Zhou J, Zhou Y, Guan F. MicroRNA-148a Inhibits Hepatocellular Carcinoma Cell Growth via Epithelial-to-Mesenchymal Transition and PI3K/AKT Signaling Pathways by Targeting Death Receptor-5. Appl Biochem Biotechnol 2022; 194:2731-2746. [PMID: 35267120 DOI: 10.1007/s12010-022-03863-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/24/2022] [Indexed: 11/24/2022]
Abstract
The purpose of this study was to investigate the role of microRNA-148a (miR-148a) in hepatocellular carcinoma (HCC) metastasis and explore its potential mechanism in HCC cells. Expression levels of miR-148a were measured using qRT-PCR in 120 HCC tissue samples and two HCC cell lines. Migration and invasion assays were used to determine the role of miR-148a in HCC cells. Flow cytometry was used to access the effect of miR-148a on cell cycle of HCC cells. Western blot was performed to analyze the effect of miR-148a on epithelial-to-mesenchymal transition (EMT) and PI3K/AKT signaling pathways in HCC cells. Luciferase reporter assay was conducted to explore the downstream targets and biological function of miR-148a in HCC cells. The results showed that level of miR-148a was significantly downregulated in both HCC tissue and plasma samples in HCC patients. A higher level of miR-148a was positively correlated with better survival time and prognosis of HCC patients. Transfection of miR-148a inhibited the proliferation, migration and invasion of HCC cell lines. Transfection of miR-148a arrested HCC cells at S phase and promoted apoptosis of HCC cells. Death receptor-5 (DR-5) was identified as a direct target of miR-148a in HCC cell lines. Western blot and qRT-PCR analyses showed that miR-148a upregulated EMT and downregulated PI3K/AKT signaling pathways in HCC cell lines. In conclusion, data in the current study indicate that miR-148a inhibits HCC cells growth via downregulation of EMT and PI3K/AKT signaling pathways by targeting death receptor. These data suggest that miR-148a may serve as a therapeutic target for HCC cancer therapy in the future.
Collapse
Affiliation(s)
- Naipeng Zhang
- Department of General Surgery, Hongqi Hospital affiliated to Mudanjiang Medical University, No.5, Tongxiang Road, Aimin District, Heilongjiang Province, 157000, Mudanjiang City, China
| | - Jian Zhou
- Department of General Surgery, Hongqi Hospital affiliated to Mudanjiang Medical University, No.5, Tongxiang Road, Aimin District, Heilongjiang Province, 157000, Mudanjiang City, China
| | - Yang Zhou
- Department of Stomatology, Hongqi Hospital affiliated to Mudanjiang Medical University, 157000, Mudanjiang City, China
| | - Fulong Guan
- Department of General Surgery, Hongqi Hospital affiliated to Mudanjiang Medical University, No.5, Tongxiang Road, Aimin District, Heilongjiang Province, 157000, Mudanjiang City, China.
| |
Collapse
|
12
|
Sun Q, Gong T, Liu M, Ren S, Yang H, Zeng S, Zhao H, Chen L, Ming T, Meng X, Xu H. Shikonin, a naphthalene ingredient: Therapeutic actions, pharmacokinetics, toxicology, clinical trials and pharmaceutical researches. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 94:153805. [PMID: 34749177 DOI: 10.1016/j.phymed.2021.153805] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/15/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Shikonin is one of the major phytochemical components of Lithospermum erythrorhizon (Purple Cromwell), which is a type of medicinal herb broadly utilized in traditional Chinese medicine. It is well established that shikonin possesses remarkable therapeutic actions on various diseases, with the underlying mechanisms, pharmacokinetics and toxicological effects elusive. Also, the clinical trial and pharmaceutical study of shikonin remain to be comprehensively delineated. PURPOSE The present review aimed to systematically summarize the updated knowledge regarding the therapeutic actions, pharmacokinetics, toxicological effects, clinical trial and pharmaceutical study of shikonin. METHODS The information contained in this review article were retrieved from some authoritative databases including Web of Science, PubMed, Google scholar, Chinese National Knowledge Infrastructure (CNKI), Wanfang Database and so on, till August 2021. RESULTS Shikonin exerts multiple therapeutic efficacies, such as anti-inflammation, anti-cancer, cardiovascular protection, anti-microbiomes, analgesia, anti-obesity, brain protection, and so on, mainly by regulating the NF-κB, PI3K/Akt/MAPKs, Akt/mTOR, TGF-β, GSK3β, TLR4/Akt signaling pathways, NLRP3 inflammasome, reactive oxygen stress, Bax/Bcl-2, etc. In terms of pharmacokinetics, shikonin has an unfavorable oral bioavailability, 64.6% of the binding rate of plasma protein, and enhances some metabolic enzymes, particularly including cytochrome P450. In regard to the toxicological effects, shikonin may potentially cause nephrotoxicity and skin allergy. The above pharmacodynamics and pharmacokinetics of shikonin have been validated by few clinical trials. In addition, pharmaceutical innovation of shikonin with novel drug delivery system such as nanoparticles, liposomes, microemulsions, nanogel, cyclodextrin complexes, micelles and polymers are beneficial to the development of shikonin-based drugs. CONCLUSIONS Shikonin is a promising phytochemical for drug candidates. Extensive and intensive explorations on shikonin are warranted to expedite the utilization of shikonin-based drugs in the clinical setting.
Collapse
Affiliation(s)
- Qiang Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ting Gong
- Department of Ultrasound, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, Sichuan, China
| | - Maolun Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shan Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Han Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Sha Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hui Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tianqi Ming
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
13
|
Wang Q, Wang J, Wang J, Ju X, Zhang H. Molecular mechanism of shikonin inhibiting tumor growth and potential application in cancer treatment. Toxicol Res (Camb) 2021; 10:1077-1084. [PMID: 34956612 PMCID: PMC8692723 DOI: 10.1093/toxres/tfab107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/13/2021] [Accepted: 10/27/2021] [Indexed: 12/20/2022] Open
Abstract
Shikonin is one of the major bioactive components of Lithospermum erythrorhizon. It has a good killing effect in a variety of tumor cells. Its antitumor effect involves multiple targets and pathways and has received extensive attention and study in recent years. In this review, we systematically review recent progress in determining the antitumor mechanism of shikonin and its derivatives, specifically their induction of reactive oxygen species production, inhibition of EGFR and PI3K/AKT signaling pathway activation, inhibition of angiogenesis and induction of apoptosis and necroptosis. We also discuss the application of nanoparticles loaded with shikonin in the targeted therapy of various cancers. Finally, we suggest new strategies for the clinical application of shikonin and its derivatives.
Collapse
Affiliation(s)
- Qiang Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, PR China
| | - Jing Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, PR China
| | - Jiayou Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, PR China
| | - Xiaoli Ju
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Heng Zhang
- Department of General Surgery, Nanjing Lishui District People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| |
Collapse
|
14
|
Liu G, Zeng T. Sporoderm-Removed Ganoderma lucidum Spore Powder May Suppress the Proliferation, Migration, and Invasion of Esophageal Squamous Cell Carcinoma Cells Through PI3K/AKT/mTOR and Erk Pathway. Integr Cancer Ther 2021; 20:15347354211062157. [PMID: 34841952 PMCID: PMC8649442 DOI: 10.1177/15347354211062157] [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] [Indexed: 12/24/2022] Open
Abstract
Tumor metastasis is a key factor of therapeutic failure in tumor patients, but the underlying molecular mechanism remains to be explored and novel effective curative strategies are urgently required. Emerging evidence suggests that sporoderm-removed Ganoderma lucidum spore powder can suppress tumor growth and metastasis. However, the molecular mechanisms of action remain elusive. In the present study, we investigated the effects and mechanisms of sporoderm-removed Ganoderma lucidum spore powder against esophageal squamous cell carcinomas (ESCC). The expression of MCP-1 in esophageal squamous cell carcinoma cells was detected by Western blotting. The MTS assay was used to assess the esophageal squamous cell carcinoma cells viability. The clone formation assay was used to evaluate to the proliferation ability of KYSE140 and KYSE510 cells. Apoptosis and the cell cycle were analyzed by flow cytometry. Wound healing and Transwell assays were used to analyze the migration of KYSE140 and KYSE510 cells. Invasion was also analyzed by the Transwell assay. The expressions of PI3K, AKT/p-AKT, Erk/p-Erk, JNK1, and mTOR were detected by Western blotting. We found that the MCP-1 protein was highly expressed in KYSE140 and KYSE510. In addition, sporoderm-removed Ganoderma lucidum spore powder treatment was found to inhibit esophageal squamous cell carcinoma cell proliferation, to block the cell cycle, to induce cell apoptosis and to inhibit cell migration and invasion. Finally, we found that sporoderm-removed Ganoderma lucidum spore powder decreased the expression of PI3K/AKT/mTOR and Erk signaling pathways. Taken together, these findings demonstrate that sporoderm-removed Ganoderma lucidum spore powder suppresses esophageal squamous cell carcinomas by involving MCP-1, regulated by PI3K/AKT/mTOR and Erk signal pathways.
Collapse
Affiliation(s)
- Guiping Liu
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, P.R. China.,Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, P.R. China
| | - Tao Zeng
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, P.R. China
| |
Collapse
|
15
|
Chemo-Sensitization of CD133+ Cancer Stem Cell Enhances the Effect of Mesenchymal Stem Cell Expressing TRAIL in Non-Small Cell Lung Cancer Cell Lines. BIOLOGY 2021; 10:biology10111103. [PMID: 34827096 PMCID: PMC8614666 DOI: 10.3390/biology10111103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/03/2021] [Accepted: 10/07/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary The anti-tumor properties of mesenchymal stem cell (MSCs) expressing TNF-related apoptosis inducing ligand (TRAIL) or MSC-TRAIL have been well documented by several reports. However, some tumors are resistant to TRAIL due to the existence of cancer stem cells (CSCs). Chemo-sensitization of tumors and their CSCs has been reported to enhance TRAIL-mediated inhibition. In this study, we examined the effect of pre-treatment using first-line chemotherapies on MSC-TRAIL-induced inhibition in non-small cell lung cancers (NSCLCs)–derived CSCs. We found that these chemotherapies were able to induce a chemo-sensitization effect to the CSC, thus improving the MSC-TRAIL-induced inhibition. We also noticed that the effect of chemo-sensitization was cell type specific and selecting chemotherapies for the right NSCLC subtypes might help in inducing a more meaningful combinatory effect. As such, this study has proven that chemo-sensitization of the CSCs was able to enhance the MSC-TRAIL-induced inhibition in NSCLC cell lines. Abstract Pre-clinical studies have demonstrated the efficacy of mesenchymal stem cells (MSCs) expressing tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) or MSC-TRAIL against several tumors. However, due to the existence of cancer stem cells (CSCs), some tumors, including non-small cell lung cancer (NSCLC), exhibit TRAIL resistance. This study was designed to evaluate the capacity of using first-line chemotherapies including cisplatin, 5-fluorouracil (5-FU) and vinorelbine to act as a chemo-sensitizer on CD133+ (prominin-1 positive) CSCs derived from NSCLC cell lines (A549, H460 and H2170) for the purpose of MSC-TRAIL-induced inhibition. We showed that MSC-TRAIL was resistant to all three chemotherapies compared to the NSCLC cell lines, suggesting that the chemotherapies had little effect on MSC-TRAIL viability. Pre-treatment using either cisplatin or 5-FU, but not with vinorelbine, was able to increase the efficacy of MSC-TRAIL to kill the TRAIL-resistant A549-derived CSCs. The study also demonstrated that both 5-FU and vinorelbine were an effective chemo-sensitizer, used to increase the anti-tumor effect of MSC-TRAIL against H460- and H2170-derived CSCs. Furthermore, pre-treatment using cisplatin was noted to enhance the effect of MSC-TRAIL in H460-derived CSCs; however, this effect was not detected in the H2170-derived CSCs. These findings suggest that a pre-treatment using certain chemotherapies in NSCLC could enhance the anti-tumor effect of MSC-TRAIL to target the CSCs, and therefore the combination of chemotherapies and MSC-TRAIL may serve as a novel approach for the treatment of NSCLC.
Collapse
|
16
|
Lee HYJ, Meng M, Liu Y, Su T, Kwan HY. Medicinal herbs and bioactive compounds overcome the drug resistance to epidermal growth factor receptor inhibitors in non-small cell lung cancer. Oncol Lett 2021; 22:646. [PMID: 34386068 DOI: 10.3892/ol.2021.12907] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Non-small cell lung cancer (NSCLC) accounts for ~85% of all lung cancer cases. Patients harboring epidermal growth factor receptor (EGFR) mutations usually develop resistance to treatment with frontline EGFR-tyrosine kinase inhibitors (EGFR-TKIs). The present review summarizes the current findings and delineates the molecular mechanism of action for the therapeutic effects of herbal extracts and phytochemicals in overcoming EGFR-TKI resistance in NSCLC. Novel molecular targets underlying EGFR-TKI resistance in NSCLC are also discussed. This review provides valuable information for the development of herbal bioactive compounds as alternative treatments for EGFR-TKI-resistant NSCLC.
Collapse
Affiliation(s)
- Hiu Yan Jennifer Lee
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P.R. China
| | - Mingjing Meng
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Yulong Liu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P.R. China
| | - Tao Su
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P.R. China
| |
Collapse
|
17
|
Liu Y, Liu X, Yang S. MicroRNA-221 Upregulates the Expression of P-gp and Bcl-2 by Activating the Stat3 Pathway to Promote Doxorubicin Resistance in Osteosarcoma Cells. Biol Pharm Bull 2021; 44:861-868. [PMID: 33828027 DOI: 10.1248/bpb.b21-00163] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
MicroRNA-221 (miRNA-221) is upregulated in several malignant tumors and is associated with poor patient prognosis. Therefore, the present study aimed to investigate the role and underlying mechanism of miRNA-221 in doxorubicin (DOX) resistance in osteosarcoma cells. We constructed DOX-resistant Saos-2/DOX cells and treated them with DOX. Cell viability was determined by performing a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cells were transfected with either miRNA-221 mimic or miRNA-221 inhibitor; quantitative (q)RT-PCR was performed to detect the expression of miRNA-221. Flow cytometry and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-digoxigenin nick-end labeling (TUNEL) staining were used to detect cell apoptosis. The immunofluorescence method was also used to detect cell signal transduction and activator of transcription 3 (Stat3) protein expression distribution. In addition, Western blotting was used to detect changes in the expression of each protein. We found that miRNA-221 was upregulated in Saos-2/DOX cells. Moreover, the miRNA-221 mimic induced DOX resistance in Saos-2 cells, whereas the miRNA-221 inhibitor enhanced DOX sensitivity in Saos-2/DOX cells. The miRNA-221 mimic upregulated the expression of phosphorylated-Stat3, P-glycoprotein (P-gp), and B-cell lymphoma-2 (Bcl-2) proteins in Saos-2 cells and induced the entry of Stat3 into the nucleus, whereas the miRNA-221 inhibitor exerted the opposite effect. Pretreatment with the Stat3 chemical inhibitor, STAT3-IN-3, significantly inhibited the upregulation of P-gp and Bcl-2 protein expression induced by the miRNA-221 mimic in Saos-2 cells; it also caused the Saos-2 cells to overcome DOX resistance induced by the miRNA-221 mimic. Thus, miRNA-221 increased the expression of P-gp and Bcl-2 by activating the Stat3 pathway to promote DOX resistance in osteosarcoma cells, indicating a potential use of miRNA-221 in osteosarcoma treatment.
Collapse
Affiliation(s)
- Yancai Liu
- Department of Pathology, The Fourth People's Hospital of Hengshui
| | - Xuegang Liu
- Department of General Surgery, The Fourth People's Hospital of Hengshui
| | - Shan Yang
- Department of Pain Treatment, The Third Hospital of Hebei Medical University
| |
Collapse
|
18
|
Sun B, Liu Y, He D, Li J, Wang J, Wen W, Hong M. Traditional Chinese medicines and their active ingredients sensitize cancer cells to TRAIL-induced apoptosis. J Zhejiang Univ Sci B 2021; 22:190-203. [PMID: 33719224 DOI: 10.1631/jzus.b2000497] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The rapidly developing resistance of cancers to chemotherapy agents and the severe cytotoxicity of such agents to normal cells are major stumbling blocks in current cancer treatments. Most current chemotherapy agents have significant cytotoxicity, which leads to devastating adverse effects and results in a substandard quality of life, including increased daily morbidity and premature mortality. The death receptor of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can sidestep p53-dependent pathways to induce tumor cell apoptosis without damaging most normal cells. However, various cancer cells can develop resistance to TRAIL-induced apoptosis via different pathways. Therefore, it is critical to find an efficient TRAIL sensitizer to reverse the resistance of tumor cells to TRAIL, and to reinforce TRAIL's ability to induce tumor cell apoptosis. In recent years, traditional Chinese medicines and their active ingredients have shown great potential to trigger apoptotic cell death in TRAIL-resistant cancer cell lines. This review aims to collate information about Chinese medicines that can effectively reverse the resistance of tumor cells to TRAIL and enhance TRAIL's ability to induce apoptosis. We explore the therapeutic potential of TRAIL and provide new ideas for the development of TRAIL therapy and the generation of new anti-cancer drugs for human cancer treatment. This study involved an extensive review of studies obtained from literature searches of electronic databases such as Google Scholar and PubMed. "TRAIL sensitize" and "Chinese medicine" were the search keywords. We then isolated newly published studies on the mechanisms of TRAIL-induced apoptosis. The name of each plant was validated using certified databases such as The Plant List. This study indicates that TRAIL can be combined with different Chinese medicine components through intrinsic or extrinsic pathways to promote cancer cell apoptosis. It also demonstrates that the active ingredients of traditional Chinese medicines enhance the sensitivity of cancer cells to TRAIL-mediated apoptosis. This provides useful information regarding traditional Chinese medicine treatment, the development of TRAIL-based therapies, and the treatment of cancer.
Collapse
Affiliation(s)
- Bingyu Sun
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yongqiang Liu
- Research Center of Chinese Herbal Resource Science and Engineering, Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Danhua He
- Research Center of Chinese Herbal Resource Science and Engineering, Guangzhou University of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Jinke Li
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS 66105, USA
| | - Jiawei Wang
- Zhongshan People's Hospital, Zhongshan 528400, China
| | - Wulin Wen
- ENT & HN Surgery Department, the Second Affiliated Hospital of Ningxia Medical University, Yinchuan 750000, China.
| | - Ming Hong
- Institute of Advanced Diagnostic and Clinical Medicine, Zhongshan People's Hospital, Guangzhou University & Zhongshan People's Hospital Joint Biomedical Institute, Zhongshan 528400, China. .,Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan 523000, China.
| |
Collapse
|
19
|
Chuang HY, Tyan YS, Hwang JJ, Shih KC, Lin WC. A combination of sorafenib and radiotherapy reduces NF-κB activity and growth of hepatocellular carcinoma in an orthotopic mouse model. Oncol Lett 2021; 21:337. [PMID: 33692869 PMCID: PMC7933744 DOI: 10.3892/ol.2021.12598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is difficult to diagnose at an early stage, and its prognosis is generally poor. Sorafenib is the primary treatment for unresectable advanced HCC and targets multiple receptor tyrosine kinases. However, sorafenib only extends the average survival time by 3 months. This observation indicates that sorafenib may need to be combined with other treatments to further improve outcomes. We previously showed that combination of sorafenib with radiotherapy (RT) enhances tumor inhibition in subcutaneous HCC mouse models compared with monotherapy. The present study demonstrated that combining sorafenib and RT could suppress tumor growth in an orthotopic HCC model by regulating apoptosis and NF-κB-related pathways. Moreover, decreased numbers of visible liver tumors and a smaller percentage of spleen metastases were found in the combination group. A transient drop in body weight was initially observed after RT, but progressive recovery of body weight occurred. The current study showed that the combination of sorafenib and RT could be a safe strategy for HCC treatment.
Collapse
Affiliation(s)
- Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan, R.O.C
| | - Yeu-Sheng Tyan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung 402, Taiwan, R.O.C
| | - Jeng-Jong Hwang
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung 402, Taiwan, R.O.C.,Department of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung 402, Taiwan, R.O.C
| | - Kuang-Chung Shih
- Division of Endocrinology and Metabolism, Department of Medicine, Cheng-Hsin General Hospital, Taipei 112, Taiwan, R.O.C.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| | - Wei-Chan Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan, R.O.C.,Department of Radiology, Cathay General Hospital, New Taipei 106, Taiwan, R.O.C.,School of Medicine, Fu-Jen Catholic University, New Taipei 106, Taiwan, R.O.C
| |
Collapse
|
20
|
Tse C, Warner A, Farook R, Cronin JG. Phytochemical Targeting of STAT3 Orchestrated Lipid Metabolism in Therapy-Resistant Cancers. Biomolecules 2020; 10:biom10081118. [PMID: 32731620 PMCID: PMC7464013 DOI: 10.3390/biom10081118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
Lipids are critical for maintaining homeostasis and cellular metabolism. However, the dysregulation of lipid metabolism contributes to the pathogenesis of chronic inflammatory diseases and is a hallmark of several cancer types. Tumours exist in a microenvironment of poor vascularization-depleted oxygen and restricted nutrients. Under these conditions, tumours have been shown to increasingly depend on the metabolism of fatty acids for sustained proliferation and survival. Signal transducer and activator of transcription 3 (STAT3) plays a key role in cellular processes such as cell growth, apoptosis and lipid metabolism. Aberrant STAT3 activity, as seen in several cancer types, is associated with tumour progression and malignancy, in addition to propagating crosstalk between tumour cells and the microenvironment. Furthermore, STAT3-regulated lipid metabolism is critical for cancer stem cell self-renewal and therapy resistance. Plant-derived compounds known as phytochemicals are a potential source for novel cancer therapeutic drugs. Dietary phytochemicals are known to modulate key cellular signalling pathways involved in lipid homeostasis and metabolism, including the STAT3 signalling pathways. Targeting STAT3 orchestrated lipid metabolism has shown therapeutic promise in human cancer models. In this review, we summarize the antitumour activity of phytochemicals with an emphasis placed on their effect on STAT3-regulated lipid metabolism and their role in abrogating therapy resistance.
Collapse
|
21
|
Qiao X, Wang C, Wang W, Shang Y, Li Y, Ni J, Chen SZ. Levamisole enhances DR4-independent apoptosis induced by TRAIL through inhibiting the activation of JNK in lung cancer. Life Sci 2020; 257:118034. [PMID: 32621923 DOI: 10.1016/j.lfs.2020.118034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/24/2020] [Accepted: 06/28/2020] [Indexed: 12/12/2022]
Abstract
THE HEADINGS AIMS Levamisole has anti-parasite and antitumor activities, but the anti-lung cancer mechanism has not been studied. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is regarded as a promising drug because of the ability to selectively target cancer cells. However, the tolerance of cancer cells to TRAIL limits its antitumor activity. Other drugs combined with TRAIL need to be explored to enhance its antitumor activity. Based on the adjuvant anticancer effect of levamisole on anticancer drugs activity, the antitumor activity of levamisole combined with TRAIL will be investigated. MATERIALS AND METHODS In vitro and in vivo experiments were employed to investigate the anti-tumor activity. Flow-cytometry analysis, western blotting and siRNA transfection were used to explore the molecular mechanism. KEY FINDINGS Levamisole decreased the proliferation of lung cancer cells in vitro and in vivo and induced cell cycle arrest in G0/G1 phase. Besides, levamisole also enhanced TRAIL-induced DR4-independent apoptosis by inhibiting the phosphorylation of cJUN. A new cellular protective pathway LC3B-DR4/Erk was also disclosed, in which levamisole only increased the expression of LC3B and then activated the phosphorylation of Erk and increased the expression of DR4, while p-Erk and DR4 inter-regulated. SIGNIFICANCE Levamisole may be used as an adjuvant of TRAIL in treating lung cancer. The discovery of LC3B-DR4/Erk as a new protective pathway provides a new direction for sensitizing lung cancer cells to TRAIL.
Collapse
Affiliation(s)
- Xinran Qiao
- Institute of Medicinal Biotehnology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Chen Wang
- Institute of Medicinal Biotehnology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Wendie Wang
- Institute of Medicinal Biotehnology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Yue Shang
- Institute of Medicinal Biotehnology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Yi Li
- Institute of Medicinal Biotehnology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Jun Ni
- Institute of Medicinal Biotehnology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Shu-Zhen Chen
- Institute of Medicinal Biotehnology, Chinese Academy of Medical Sciences & Peking Union Medical College, China.
| |
Collapse
|
22
|
Cao HH, Liu DY, Lai YC, Chen YY, Yu LZ, Shao M, Liu JS. Inhibition of the STAT3 Signaling Pathway Contributes to the Anti-Melanoma Activities of Shikonin. Front Pharmacol 2020; 11:748. [PMID: 32536866 PMCID: PMC7267064 DOI: 10.3389/fphar.2020.00748] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 05/05/2020] [Indexed: 12/24/2022] Open
Abstract
Background Malignant melanoma is an extremely aggressive and metastatic cancer, and highly resistant to conventional therapies. Signal transducer and activator of transcription 3 (STAT3) signaling promotes melanoma development and progression, which has been validated as an effective target in melanoma treatment. Natural naphthoquinone shikonin is reported to exert anti-melanoma effects. However, the underlying mechanisms have not been fully elucidated. Purpose This study aims to evaluate the anti-melanoma activities of shikonin and explore the involvement of STAT3 signaling in these effects. Methods Zebrafish tumor model was established to evaluate the anti-human melanoma effects of shikonin in vivo. MTT assay and colony formation assay were employed to investigate the anti-proliferative effects of shikonin on human melanoma A375 and A2058 cells. Flow cytometry was used to analyze cell cycle distribution and apoptosis induction. Wound healing assay and Transwell chamber assay were conducted to examine the cell migratory and invasive abilities. Immunofluorescence assay was used to observe F-actin, Tubulin, and STAT3 localization. Western blotting was used to determine the expression levels of proteins associated with apoptosis and key proteins in the STAT3 signaling pathway. Immunoblotting was performed in DSS cross-linked cells to determine the homo-dimerization of STAT3. Gelatin zymography was employed to evaluate the enzymatic activity of MMP-2 and MMP-9. Transient transfection was used to overexpress STAT3 in cell models. Results Shikonin suppressed melanoma growth in cultured cells and in zebrafish xenograft models. Shikonin induced melanoma cells apoptosis, inhibited cell migration and invasion. Mechanistic study indicated that shikonin inhibited the phosphorylation and homo-dimerization of STAT3, thus reduced its nuclear localization. Further study showed that shikonin decreased the levels of STAT3-targeted genes Mcl-1, Bcl-2, MMP-2, vimentin, and Twist, which are involved in melanoma survival, migration, and invasion. More importantly, overexpression of constitutively active STAT3 partially abolished the anti-proliferative, anti-migratory, and anti-invasive effects of shikonin. Conclusion The anti-melanoma activity of shikonin is at least partially attributed to the inhibition on STAT3 signaling. These findings provide new insights into the anti-melanoma molecular mechanisms of shikonin, suggesting its potential in melanoma treatment.
Collapse
Affiliation(s)
- Hui-Hui Cao
- Traditional Chinese Pharmacological, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Dong-Yi Liu
- Traditional Chinese Pharmacological, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Ye-Cai Lai
- Guangzhou BaiYunShan Pharmaceutical General Factory, Guangzhou BaiYunShan Pharmaceutical Holdings Co., Ltd., Guangzhou, China
| | - Yu-Yao Chen
- Traditional Chinese Pharmacological, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lin-Zhong Yu
- Traditional Chinese Pharmacological, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Meng Shao
- Traditional Chinese Pharmacological, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jun-Shan Liu
- Traditional Chinese Pharmacological, Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
23
|
Lin HY, Han HW, Wang YS, He DL, Sun WX, Feng L, Wen ZL, Yang MK, Lu GH, Wang XM, Qi JL, Yang YH. Shikonin and 4-hydroxytamoxifen synergistically inhibit the proliferation of breast cancer cells through activating apoptosis signaling pathway in vitro and in vivo. Chin Med 2020; 15:23. [PMID: 32175001 PMCID: PMC7063777 DOI: 10.1186/s13020-020-00305-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/28/2020] [Indexed: 01/13/2023] Open
Abstract
Background Tamoxifen (TAM) is a cell type-specific anti-estrogen and is applied to improve the survival of patients with estrogen receptor positive (ER +) breast cancer. However, long-term TAM use can induce serious drug resistance, leading to breast cancer recurrence and death in patients. Further, it is almost useless among patients with estrogen receptor negative (ER −) breast cancer. Shikonin (SK) is a natural product broadly explored in cancer therapy. Some studies have demonstrated the combined treatment of SK and clinical anticancer drugs including TAM on various tumors. However, the combined effect of SK and 4-hydroxytamoxifen (4-OHT) on ER- breast cancer is not known. The current study aimed to assess the combination effects of SK and 4-OHT on human breast cancer cells, MCF-7 (ER +) and MDA-MB-435S (ER −), in vitro and in vivo and to investigate the underlying mechanisms. Methods CCK-8 assays and flow cytometry were conducted to determine the cell viability and apoptotic profiles of human breast cancer cell lines (MCF-7 and MDA-MB-435S) treated with SK, 4-OHT, and the combination. ROS and JC-1 assays were used to determine ROS level and mitochondrial membrane potential. Western blot analysis was performed to investigate proteins that are associated with apoptosis. Haematoxylin & Eosin (HE) staining was used to detect the tumor and kidney morphology of mice. TUNEL and immunohistochemical staining were performed to detect Ki67 expression level and cell apoptotic profile in tumor tissues. Results SK and 4-OHT synergistically inhibited MCF-7 and MDA-MB-435S cell proliferation and promoted apoptosis by reducing mitochondrial membrane potential and increasing the intracellular ROS level. The combination of SK and 4-OHT activated the mitochondrial-dependent apoptosis and the death receptor pathways, significantly regulating the PI3K/AKT/Caspase 9 signaling pathway. Compared with SK and 4-OHT alone, the combination of SK and 4-OHT could better inhibit tumor growth in mice. Conclusion The combination of SK and 4-OHT shows highly efficient anticancer effects on breast cancer therapy. SK may be a promising candidate as an adjuvant to 4-OHT for breast cancer treatments, especially for ER- breast cancer.
Collapse
Affiliation(s)
- Hong-Yan Lin
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China.,2Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037 People's Republic of China
| | - Hong-Wei Han
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China.,2Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037 People's Republic of China
| | - Yin-Song Wang
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China
| | - De-Liu He
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China
| | - Wen-Xue Sun
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China
| | - Lu Feng
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China
| | - Zhong-Ling Wen
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China
| | - Min-Kai Yang
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China
| | - Gui-Hua Lu
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China.,3School of Life Sciences, Huaiyin Normal University, Huaian, 223300 China
| | - Xiao-Ming Wang
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China.,2Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037 People's Republic of China
| | - Jin-Liang Qi
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China.,2Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037 People's Republic of China
| | - Yong-Hua Yang
- 1State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023 People's Republic of China.,2Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037 People's Republic of China
| |
Collapse
|
24
|
Zhou H, Liu J, Chen Z. Coronarin D suppresses proliferation, invasion and migration of glioma cells via activating JNK signaling pathway. Pathol Res Pract 2019; 216:152789. [PMID: 31870592 DOI: 10.1016/j.prp.2019.152789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 11/25/2019] [Accepted: 12/12/2019] [Indexed: 01/03/2023]
Abstract
Coronarin D (CD) is one of the primary components of the Hedychium coronarium rhizomes and possesses strong anticancer effects via preventing cell growth in many cancer cells. The study was aimed to explore the molecular mechanisms underlying effects of CD on proliferation, invasion and migration of gliomas cells. Gliomas cell lines U251 was employed for detecting cells viability and proliferation by Cell Counting Kit-8 assay and colony formation assay. In addition, scratch wound healing and transwell assays were performed for the analysis of U251 cells invasion and migration respectively. Furthermore, the expression of p-Akt/Akt, p-p38/p38, p-ERK/ERK, p-JNK/JNK, p-STAT3/STAT3, cyclinE, cyclinD1, CTGF, MMP-2 and MMP-9 were measured by Western blotting. CD could suppress proliferation, invasion and migration of glioma cells and induced reduction of cyclinE, cyclinD1, CTGF, MMP-2 and MMP-9 expression via activating JNK signaling pathway. CD treatment suppressed expression of p-AKT, p38, and ERK and elevated expression of p-JNK in concentration- and time-dependent manners. Moreover, CD significantly induced reduction of phosphorylated STAT3 expression. Exposure of cells to the JNK-specific inhibitor SP600125 reduced the cytotoxicity effects of CD, combination of CD and SP600125 corrected overexpression of phosphorylated JNK and reduction of phosphorylated STAT3. Pretreatment of SP600125 also improves gliomas cells viability and invasion. The results revealed that CD may remarkably suppress gliomas cell growth through JNK and STAT3 signaling. In present study, these findings revealed that CD induces suppression of cell viability in gliomas cells and possesses therapeutic effect on gliomas.
Collapse
Affiliation(s)
- Hongjun Zhou
- Department of Neurosurgery, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), PR China
| | - Jiang Liu
- Department of Neurosurgery, The Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), PR China
| | - Zhongjun Chen
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, PR China.
| |
Collapse
|
25
|
Li X, Zeng X. Shikonin suppresses progression and epithelial-mesenchymal transition in hepatocellular carcinoma (HCC) cells by modulating miR-106b/SMAD7/TGF-β signaling pathway. Cell Biol Int 2019; 44:467-476. [PMID: 31617643 DOI: 10.1002/cbin.11247] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/13/2019] [Indexed: 12/13/2022]
Abstract
Shikonin is a natural naphthoquinone component with antioxidant and anti-tumor function and has been used for hepatocellular carcinoma (HCC) treatment. According to the previous study, many herbs can regulate cancer cell progression by targeting specific microRNA (miRNA) (Liu, 2016). However, the underlying pathological mechanism of shikonin in HCC therapy is still unclear. The detection of cell growth and death rate were performed by hemacytometry and trypan blue staining, respectively. The expression of miR-106b and SMAD7 messenger RNA (mRNA) in HCC cells was evaluated by quantitative real-time polymerase chain reaction. Cell proliferation, apoptosis, and migration ability were measured by cell counting kit-8 (CCK-8), flow cytometry, and transwell assay. The expression of proteins E-cadherin, N-cadherin, vimentin, SMAD7, TGF-β1, p-SMAD3, SMAD3, and GAPDH was examined by western blot. The interaction between SMAD7 and miR-106b was assessed by luciferase reporter system. Shikonin inhibited Huh7 and HepG2 cell growth in a dose-dependent manner while induced cell death in a time-dependent manner. In addition, the expression of miR-106b was reduced after shikonin treatment. Moreover, miR-106b attenuated the suppressive effects of shikonin on HCC cell migration and epithelial-mesenchymal transition (EMT). SMAD7 was predicted as a target of miR-106b and the prediction was confirmed by luciferase reporter system. Additionally, we observed that SMAD7 reversed the promotive effects of miR-106b on HCC cell progression and EMT. The subsequent western blot assay revealed that shikonin could modulate SMAD7/TGF-β signaling pathway by targeting miR-106b. In conclusion, Shikonin suppresses cell progression and EMT and accelerates cell death of HCC cells via modulating miR-106b/SMAD7/TGF-β signaling pathway, suggesting shikonin could be an effective agent for HCC treatment.
Collapse
Affiliation(s)
- Xiaojing Li
- Department of Minimal Invasive Surgery, the Second Xiangya Hospital of Central South University, Changsha, 410000, China
| | - Xianpeng Zeng
- Department of Institute of Hepatobiliary Diseases, the Zhongnan Hospital of Wuhan University, Wuhan, 430000, China
| |
Collapse
|
26
|
Natural Product Mediated Regulation of Death Receptors and Intracellular Machinery: Fresh from the Pipeline about TRAIL-Mediated Signaling and Natural TRAIL Sensitizers. Int J Mol Sci 2019; 20:ijms20082010. [PMID: 31022877 PMCID: PMC6515249 DOI: 10.3390/ijms20082010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/24/2022] Open
Abstract
Rapidly developing resistance against different therapeutics is a major stumbling block in the standardization of therapy. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-mediated signaling has emerged as one of the most highly and extensively studied signal transduction cascade that induces apoptosis in cancer cells. Rapidly emerging cutting-edge research has helped us to develop a better understanding of the signaling machinery involved in inducing apoptotic cell death. However, excitingly, cancer cells develop resistance against TRAIL-induced apoptosis through different modes. Loss of cell surface expression of TRAIL receptors and imbalance of stoichiometric ratios of pro- and anti-apoptotic proteins play instrumental roles in rewiring the machinery of cancer cells to develop resistance against TRAIL-based therapeutics. Natural products have shown excellent potential to restore apoptosis in TRAIL-resistant cancer cell lines and in mice xenografted with TRAIL-resistant cancer cells. Significantly refined information has previously been added and continues to enrich the existing pool of knowledge related to the natural-product-mediated upregulation of death receptors, rebalancing of pro- and anti-apoptotic proteins in different cancers. In this mini review, we will set spotlight on the most recently published high-impact research related to underlying mechanisms of TRAIL resistance and how these deregulations can be targeted by natural products to restore TRAIL-mediated apoptosis in different cancers.
Collapse
|
27
|
Guo N, Miao R, Gao X, Huang D, Hu Z, Ji N, Nan Y, Jiang F, Gou X. Shikonin inhibits proliferation and induces apoptosis in glioma cells via downregulation of CD147. Mol Med Rep 2019; 19:4335-4343. [PMID: 30942433 PMCID: PMC6472142 DOI: 10.3892/mmr.2019.10101] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 02/19/2019] [Indexed: 12/23/2022] Open
Abstract
Shikonin, a traditional Chinese medicine, has been identified as being capable of inducing apoptosis in various tumors, including glioma, and is thus considered to be a promising therapeutic agent for tumor therapy. However, little is known about the molecular mechanism of shikonin in glioma. The present study investigated the influence of shikonin on the proliferation and apoptosis of glioma cells U251 and U87MG and explored the potential molecular mechanisms. It was identified that shikonin was able to induce apoptosis in human glioma cells in a time‑ and dose‑dependent manner, and a decreased expression level of cluster of differentiation (CD)147 was observed in shikonin‑treated U251 and U87MG cells. Knockdown of CD147 inhibited U251 and U87MG cell growth, whereas CD147 overexpression enhanced cell growth and decreased shikonin‑induced apoptosis. Additionally, an increased expression level of CD147 suppressed the elevated production of reactive oxygen species and mitochondrial membrane potential levels induced by shikonin. The data indicated that shikonin‑induced apoptosis in glioma cells was associated with the downregulation of CD147 and the upregulation of oxidative stress. CD147 may be an optional target of shikonin‑induced cell apoptosis in glioma cells.
Collapse
Affiliation(s)
- Na Guo
- Institute of Basic and Translational Medicine, and Department of Immunology, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Rui Miao
- Department of Neurology, Shaanxi Second Provincial People's Hospital, Xi'an, Shaanxi 710005, P.R. China
| | - Xingchun Gao
- Institute of Basic and Translational Medicine, and Department of Immunology, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Dong Huang
- Department of Neurology, Shaanxi Second Provincial People's Hospital, Xi'an, Shaanxi 710005, P.R. China
| | - Zhifang Hu
- Institute of Basic and Translational Medicine, and Department of Immunology, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Naichun Ji
- Department of Sports, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Ying Nan
- Institute of Basic and Translational Medicine, and Department of Immunology, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Fengliang Jiang
- Institute of Basic and Translational Medicine, and Department of Immunology, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Xingchun Gou
- Institute of Basic and Translational Medicine, and Department of Immunology, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| |
Collapse
|
28
|
Prakash O, Nath Dwivedi U. Identification of repurposed protein kinase B binders from FDA-approved drug library: a hybrid-structure activity relationship and systems modeling based approach. J Biomol Struct Dyn 2019; 38:660-672. [PMID: 30806166 DOI: 10.1080/07391102.2019.1585293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Food and Drug Administration (FDA)-approved drugs may be repurposed against those diseases, for which their therapeutic action has not been described. The present study deals with repurposing FDA-approved drugs for selective targeting of protein kinase B (PKB/Akt) for anti-cancer activity, through a two-tier (Cell and Target) model hybridization protocol implemented with support vector machine-based learning method. The hybridization was done as per rules of reaction kinetics. The hybridization process was facilitated as a standalone application for free access at https://github.com/undwivedi/Akt-Selective.git. The selectivity of the ligands for PKB/Akt binding was also evaluated on the basis of mitophagy system model for anti-apoptotic activity. Screening of the FDA-approved drug library, using the developed H- SAR model, led to identification of four compounds (Cas nos. 94749-08-3, 57808-66-9, 62-13-5, 76-43-7), bearing the selectivity for PKB/Akt. Since, the identified compounds have already crossed the barriers of absorption, distribution, metabolism, excretion, toxicity in clinical trials, therefore are safe to be considered for repurposing individually or in combination with other drugs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Om Prakash
- Department of Biochemistry, Bioinformatics Infrastructure Facility, Centre of Excellence in Bioinformatics & Institute for Development of Advanced Computing, ONGC Centre for Advanced Studies University of Lucknow, Lucknow, Uttar Pradesh, India
| | - Upendra Nath Dwivedi
- Department of Biochemistry, Bioinformatics Infrastructure Facility, Centre of Excellence in Bioinformatics & Institute for Development of Advanced Computing, ONGC Centre for Advanced Studies University of Lucknow, Lucknow, Uttar Pradesh, India
| |
Collapse
|