1
|
Bae S, Bae S, Kim HS, Lim YJ, Kim G, Park IC, So KA, Kim TJ, Lee JH. Deguelin Restores Paclitaxel Sensitivity in Paclitaxel-Resistant Ovarian Cancer Cells via Inhibition of the EGFR Signaling Pathway. Cancer Manag Res 2024; 16:507-525. [PMID: 38827785 PMCID: PMC11144006 DOI: 10.2147/cmar.s457221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
Background Ovarian cancer is one of women's malignancies with the highest mortality among gynecological cancers. Paclitaxel is used in first-line ovarian cancer chemotherapy. Research on paclitaxel-resistant ovarian cancer holds significant clinical importance. Methods Cell viability and flow cytometric assays were conducted at different time and concentration points of deguelin and paclitaxel treatment. Immunoblotting was performed to assess the activation status of key signaling molecules important for cell survival and proliferation following treatment with deguelin and paclitaxel. The fluo-3 acetoxymethyl assay for P-glycoprotein transport activity assay and cell viability assay in the presence of N-acetyl-L-cysteine were also conducted. Results Cell viability and flow cytometric assays demonstrated that deguelin resensitized paclitaxel in a dose- and time-dependent manner. Cotreatment with deguelin and paclitaxel inhibited EGFR and its downstream signaling molecules, including AKT, ERK, STAT3, and p38 MAPK, in SKOV3-TR cells. Interestingly, cotreatment with deguelin and paclitaxel suppressed the expression level of EGFR via the lysosomal degradation pathway. Cotreatment did not affect the expression and function of P-glycoprotein. N-acetyl-L-cysteine failed to restore cell cytotoxicity when used in combination with deguelin and paclitaxel in SKOV3-TR cells. The expression of BCL-2, MCL-1, and the phosphorylation of the S155 residue of BAD were downregulated. Moreover, inhibition of paclitaxel resistance by deguelin was also observed in HeyA8-MDR cells. Conclusion Our research showed that deguelin effectively suppresses paclitaxel resistance in SKOV3-TR ovarian cancer cells by downregulating the EGFR and its downstream signaling pathway and modulating the BCL-2 family proteins. Furthermore, deguelin exhibits inhibitory effects on paclitaxel resistance in HeyA8-MDR ovarian cancer cells, suggesting a potential mechanism for paclitaxel resensitization that may not be cell-specific. These findings suggest that deguelin holds promise as an anticancer therapeutic agent for overcoming chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Seunghee Bae
- Department of Cosmetics Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Sowon Bae
- Department of Cosmetics Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hee Su Kim
- Department of Cosmetics Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Ye Jin Lim
- Department of Cosmetics Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Gyeongmi Kim
- Division of Fusion Radiology Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Republic of Korea
| | - In-Chul Park
- Division of Fusion Radiology Research, Korea Institute of Radiological & Medical Sciences, Seoul, 01812, Republic of Korea
| | - Kyeong A So
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul, 05030, Republic of Korea
| | - Tae Jin Kim
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul, 05030, Republic of Korea
| | - Jae Ho Lee
- Department of Cosmetics Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| |
Collapse
|
2
|
Zhao M, Wen Y, Yang Y, Pan H, Xie S, Shen C, Liao W, Chen N, Zheng Q, Zhang G, Li Y, Gong D, Tang J, Zhao Z, Zeng J. (-)-Asarinin alleviates gastric precancerous lesions by promoting mitochondrial ROS accumulation and inhibiting the STAT3 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155348. [PMID: 38335913 DOI: 10.1016/j.phymed.2024.155348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 12/20/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND (-)-Asarinin (Asarinin) is the primary component in the extract of the herb Asarum sieboldii Miq. It possesses various functions, including pain relief, anti-viral and anti-tuberculous bacilli effects, and inhibition of tumor growth. Gastric precancerous lesion (GPL) is a common but potentially carcinogenic chronic gastrointestinal disease, and its progression can lead to gastric dysfunction and cancer development. However, the protective effects of asarinin against GPL and the underlying mechanisms remain unexplored. METHODS A premalignant cell model (methylnitronitrosoguanidine-induced malignant transformation of human gastric epithelial cell strain, MC cells) and a GPL animal model were established and then were treated with asarinin. The cytotoxic effect of asarinin was assessed using a CCK8 assay. Detection of intracellular reactive oxygen species (ROS) using DCFH-DA. Apoptosis in MC cells was evaluated using an annexin V-FITC/PI assay. We performed western blot analysis and immunohistochemistry (IHC) to analyze relevant markers, investigating the in vitro and in vivo therapeutic effects of asarinin on GPL and its intrinsic mechanisms. RESULTS Our findings showed that asarinin inhibited MC cell proliferation, enhanced intracellular ROS levels, and induced cell apoptosis. Further investigations revealed that the pharmacological effects of asarinin on MC cells were blocked by the ROS scavenger N-acetylcysteine. IHC revealed a significant upregulation of phospho-signal transducer and activator of transcription 3 (p-STAT3) protein expression in human GPL tissues. In vitro, asarinin exerted its pro-apoptotic effects in MC cells by modulating the STAT3 signaling pathway. Agonists of STAT3 were able to abolish the effects of asarinin on MC cells. In vivo, asarinin induced ROS accumulation and inhibited the STAT3 pathway in gastric mucosa of mice, thereby halting and even reversing the development of GPL. CONCLUSION Asarinin induces apoptosis and delays the progression of GPL by promoting mitochondrial ROS production, decreasing mitochondrial membrane potential (MMP), and inhibiting the STAT3 pathway.
Collapse
Affiliation(s)
- Maoyuan Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi Yang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shunkai Xie
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Caifei Shen
- Department of Endoscopy Center, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Wenhao Liao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China
| | - Nianzhi Chen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China
| | - Qiao Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China
| | - Gang Zhang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China
| | - Yuchen Li
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China
| | - Daoyin Gong
- Department of Pathology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China.
| | - Ziyi Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China.
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-Qiao Road, Chengdu, Sichuan 610072, China; Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
3
|
Çomaklı S, Özdemir S, Güloğlu M. Chrysin attenuates paclitaxel-induced hepatorenal toxicity in rats by suppressing oxidative damage, inflammation, and apoptosis. Life Sci 2023; 332:122096. [PMID: 37716503 DOI: 10.1016/j.lfs.2023.122096] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/18/2023]
Abstract
AIMS Paclitaxel (Pax) is a chemotherapeutic drug from the taxane family that is used in the treatment of human cancer, including ovarian, breast, and non-small cell lung carcinoma. Chrysin (CR) has antioxidant, anti-inflammatory, anti-apoptotic, anti-diabetic, and anti-carcinogenic properties, as well as hepatoprotective and renoprotective activities. In the present study, we evaluated the protective effect of CR against Pax-induced hepatorenal toxicity on inflammation, apoptosis, antioxidant levels, oxidative DNA damage, and histopathology in rats. MATERIAL AND METHODS Thirty-five male Sprague-Dawley rats were divided into five groups (n = 7): Group I (normal control), Group II (CR alone at a dose of 50 mg/kg), Group III (Pax at a dose of 2 mg/kg), Group IV (Pax+CR 25), and Group V (Pax+CR 50). The expressions of apoptotic (Bax and Bcl-2) and antioxidant genes (SOD1, CAT, GPx3, and GST) were evaluated using RT-PCR from paraffin sections. Caspase 3, KIM-1, NF-kB, COX-2, and 8-OHdG were also determined by immunohistochemical examination. KEY FINDINGS The results revealed that Pax exposure caused hepatic and renal damage in rats, which was indicated by a significant elevation of caspase 3, Bax, KIM-1, NF-kB, COX-2, and 8-OHdG. However, there was a marked downregulation in the expressions of the Bcl-2, SOD1, CAT, GPx3, and GST genes. In contrast, rats given CR in combination showed better gene expression, histological structure, and immunohistochemical staining results. SIGNIFICANCE Consequently, CR exhibited the ability to reduce oxidative DNA damage, exert anti-apoptotic and anti-inflammatory properties, and mitigate the toxic effects of Pax-induced hepatorenal toxicity.
Collapse
Affiliation(s)
- Selim Çomaklı
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey; German Center for Neurodegenerative Diseases, DZNE, Bonn, Germany.
| | - Meryem Güloğlu
- Veterinary Control Institute, Republic of Turkey Ministry of Agriculture and Forestry, Erzurum, Turkey.
| |
Collapse
|
4
|
Tseng YC, Shu CW, Chang HM, Lin YH, Tseng YH, Hsu HS, Goan YG, Tseng CJ. Assessment of Early Growth Response 1 in Tumor Suppression of Esophageal Squamous Cell Carcinoma. J Clin Med 2022; 11:jcm11195792. [PMID: 36233659 PMCID: PMC9572560 DOI: 10.3390/jcm11195792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/08/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Esophageal squamous cell carcinoma (ESCC) is associated with poor survival despite surgical resection, and its pathogenesis has been broadly investigated in the past decade. Early growth response 1 (EGR-1) could involve regulating tumor development in ESCC cells. Methods: An attempt was made to examine the molecular and cellular influence of EGR-1 in esophageal cancer cells by RNA extraction, real-time PCR (qRT-PCR), cell culture, small interfering RNA (siRNA) knockdown, western blot, migration assay, and cell viability assay. One hundred and forty-four samples of ESCC were collected from our hospital and analyzed. Significantly higher EGR-1 expression was noted in tumor-adjacent normal tissue compared with tumor lesions. Results: The univariate analysis showed no significant impacts of EGR-1 expression on patients’ survival. However, after adjusting for the pathological stage, patients with EGR-1 expression > 68th percentile had lower risks of cancer-related death. Moreover, knockdown of EGR-1 significantly enhanced cell migration, invasion, and resistance to chemotherapeutic agents in two ESCC cell lines. Conclusions: EGR-1 plays a key role in tumor suppression involving tumor viability suppression and reflects the treatment effect of current chemotherapy for ESCC.
Collapse
Affiliation(s)
- Yen-Chiang Tseng
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Chih-Wen Shu
- Institute of Biopharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (C.-W.S.); (C.-J.T.); Tel.: +886-7-3422121 (ext. 1505) (C.-J.T.); Fax: +886-7-3422288 (C.-J.T.)
| | - Hui-Min Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
| | - Yi-Hsuan Lin
- Department of Family Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei 100, Taiwan
| | - Yen-Han Tseng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Han-Shui Hsu
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yih-Gang Goan
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ching-Jiunn Tseng
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Correspondence: (C.-W.S.); (C.-J.T.); Tel.: +886-7-3422121 (ext. 1505) (C.-J.T.); Fax: +886-7-3422288 (C.-J.T.)
| |
Collapse
|
5
|
Zhao S, Tang Y, Wang R, Najafi M. Mechanisms of cancer cell death induction by paclitaxel: an updated review. Apoptosis 2022; 27:647-667. [PMID: 35849264 DOI: 10.1007/s10495-022-01750-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2022] [Indexed: 02/07/2023]
Abstract
Chemoresistance of cancer cells is a major problem in treating cancer. Knowledge of how cancer cells may die or resist cancer drugs is critical to providing certain strategies to overcome tumour resistance to treatment. Paclitaxel is known as a chemotherapy drug that can suppress the proliferation of cancer cells by inducing cell cycle arrest and induction of mitotic catastrophe. However, today, it is well known that paclitaxel can induce multiple kinds of cell death in cancers. Besides the induction of mitotic catastrophe that occurs during mitosis, paclitaxel has been shown to induce the expression of several pro-apoptosis mediators. It also can modulate the activity of anti-apoptosis mediators. However, certain cell-killing mechanisms such as senescence and autophagy can increase resistance to paclitaxel. This review focuses on the mechanisms of cell death, including apoptosis, mitotic catastrophe, senescence, autophagic cell death, pyroptosis, etc., following paclitaxel treatment. In addition, mechanisms of resistance to cell death due to exposure to paclitaxel and the use of combinations to overcome drug resistance will be discussed.
Collapse
Affiliation(s)
- Shuang Zhao
- School of Basic Medicine, Shaoyang University, Shaoyang, 422000, Hunan, China.
| | - Yufei Tang
- College of Medical Technology, Shaoyang University, Shaoyang, 422000, Hunan, China
| | - Ruohan Wang
- School of Nursing, Shaoyang University, Shaoyang, 422000, Hunan, China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
6
|
Ma RJ, Ma C, Hu K, Zhao MM, Zhang N, Sun ZG. Molecular mechanism, regulation, and therapeutic targeting of the STAT3 signaling pathway in esophageal cancer (Review). Int J Oncol 2022; 61:105. [PMID: 35856449 PMCID: PMC9339493 DOI: 10.3892/ijo.2022.5395] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/05/2022] [Indexed: 12/24/2022] Open
Abstract
Esophageal cancer (EC) is the seventh most common cancer globally, and the overall 5-year survival rate is only 20%. Signal transducer and activator of transcription 3 (STAT3) is aberrantly activated in EC, and its activation is associated with a poor prognosis. STAT3 can be activated by canonical pathways such as the JAK/STAT3 pathway as well as non-canonical pathways including the Wnt/STAT3 and COX2/PGE2/STAT3 pathways. Activated STAT3, present as phosphorylated STAT3 (p-STAT3), can be transported into the nucleus to regulate downstream genes, including VEGF, cyclin D1, Bcl-xL, and matrix metalloproteinases (MMPs), to promote cancer cell proliferation and induce resistance to therapy. Non-coding RNAs, including microRNAs (miRNAs/miRs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs), play a vital role in regulating the STAT3 signaling pathway in EC. Several miRNAs promote or suppress the function of STAT3 in EC, while lncRNAs and circRNAs primarily promote the effects of STAT3 and the progression of cancer. Additionally, various drugs and natural compounds can target STAT3 to suppress the malignant behavior of EC cells, providing novel insights into potential EC therapies.
Collapse
Affiliation(s)
- Rui-Jie Ma
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Chao Ma
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Kang Hu
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Meng-Meng Zhao
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Nan Zhang
- Department of Breast Disease Center, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Zhi-Gang Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
7
|
Sun L, Zhao P, Chen M, Leng J, Luan Y, Du B, Yang J, Yang Y, Rong R. Taxanes prodrug-based nanomedicines for cancer therapy. J Control Release 2022; 348:672-691. [PMID: 35691501 DOI: 10.1016/j.jconrel.2022.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/04/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022]
Abstract
Malignant tumor remains a huge threat to human health and chemotherapy still occupies an important place in clinical tumor treatment. As a kind of potent antimitotic agent, taxanes act as the first-line broad-spectrum cancer drug in clinical use. However, disadvantages such as prominent hydrophobicity, severe off-target toxicity or multidrug resistance lead to unsatisfactory therapeutic effects, which restricts its wider usage. The efficient delivery of taxanes is still quite a challenge despite the rapid developments in biomaterials and nanotechnology. Great progress has been made in prodrug-based nanomedicines (PNS) for cancer therapy due to their outstanding advantages such as high drug loading efficiency, low carrier induced immunogenicity, tumor stimuli-responsive drug release, combinational therapy and so on. Based on the numerous developments in this filed, this review summarized latest updates of taxanes prodrugs-based nanomedicines (TPNS), focusing on polymer-drug conjugate-based nanoformulations, small molecular prodrug-based self-assembled nanoparticles and prodrug-encapsulated nanosystems. In addition, the new trends of tumor stimuli-responsive TPNS were also discussed. Moreover, the future challenges of TPNS for clinical translation were highlighted. We here expect this review will inspire researchers to explore more practical taxanes prodrug-based nano-delivery systems for clinical use.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Menghan Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jiayi Leng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yixin Luan
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jia Yang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
8
|
Español A, Salem A, Sanchez Y, Sales ME. Breast cancer: Muscarinic receptors as new targets for tumor therapy. World J Clin Oncol 2021; 12:404-428. [PMID: 34189066 PMCID: PMC8223712 DOI: 10.5306/wjco.v12.i6.404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/26/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
The development of breast cancer is a complex process that involves the participation of different factors. Several authors have demonstrated the overexpression of muscarinic acetylcholine receptors (mAChRs) in different tumor tissues and their role in the modulation of tumor biology, positioning them as therapeutic targets in cancer. The conventional treatment for breast cancer involves surgery, radiotherapy, and/or chemotherapy. The latter presents disadvantages such as limited specificity, the appearance of resistance to treatment and other side effects. To prevent these side effects, several schedules of drug administration, like metronomic therapy, have been developed. Metronomic therapy is a type of chemotherapy in which one or more drugs are administered at low concentrations repetitively. Recently, two chemotherapeutic agents usually used to treat breast cancer have been considered able to activate mAChRs. The combination of low concentrations of these chemotherapeutic agents with muscarinic agonists could be a useful option to be applied in breast cancer treatment, since this combination not only reduces tumor cell survival without affecting normal cells, but also decreases pathological neo-angiogenesis, the expression of drug extrusion proteins and the cancer stem cell fraction. In this review, we focus on the previous evidences that have positioned mAChRs as relevant therapeutic targets in breast cancer and analyze the effects of administering muscarinic agonists in combination with conventional chemotherapeutic agents in a metronomic schedule.
Collapse
Affiliation(s)
- Alejandro Español
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Agustina Salem
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Yamila Sanchez
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - María Elena Sales
- Laboratory of Immunopharmacology and Tumor Biology, CEFYBO CONICET University of Buenos Aires, Buenos Aires C1121ABG, Argentina
| |
Collapse
|
9
|
Chen H, Li Y, Li Y, Chen Z, Xie L, Li W, Zhu Y, Xue H, Koeffler HP, Wu W, Hu K, Yin D. PARK2 promotes mitochondrial pathway of apoptosis and antimicrotubule drugs chemosensitivity via degradation of phospho-BCL-2. Am J Cancer Res 2020; 10:9984-10000. [PMID: 32929329 PMCID: PMC7481404 DOI: 10.7150/thno.47044] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Neoadjuvant chemotherapy has become the standard treatment of locally advanced breast cancer. Antimicrotubule drugs and DNA-damaging drugs are the most popular medicines used for neoadjuvant chemotherapy. However, we are unable to predict which chemotherapeutic drug will benefit to an individual patient. PARK2 as a tumor suppressor in breast cancer has been reported. While the role of PARK2 in chemotherapy response remains unknown. In this study, we explore the impact of PARK2 on chemosensitivity in breast cancer. Methods: PARK2 expression in breast cancer patients with different neoadjuvant chemotherapeutic regimens was studied using immunohistochemistry. Data was correlated to disease-free survival (DFS), overall survival and pathologic complete response (pCR). The functional roles of PARK2 were demonstrated by a series of in vitro and in vivo experiments. Including mass spectrometry, Co-immunoprecipitation, isolation of subcellular fractionation, fluorescence microscopy, in vivo ubiquitination assay and luciferase analyses. Results: Highly expressed PARK2 predicted better response to antimicrotubule drugs-containing regimen associated with higher rate of pathologic complete response (pCR). In contrast, PARK2 expression did not predict response to the DNA-damaging drugs regimen. Following antimicrotubule drugs treatment, levels of PARK2 was upregulated due to the repression of STAT3-mediated transcriptional inhibition of PARK2. Moreover, overexpression of PARK2 specifically rendered cells more sensitive to antimicrotubule drugs, but not to DNA-damaging drugs. Depletion of PARK2 enhanced resistance to antimicrotubule drugs. Mechanistically, PARK2 markedly activated the mitochondrial pathway of apoptosis after exposure to antimicrotubule drugs. This occurred through downregulating the antiapoptotic protein, phospho-BCL-2. BCL-2 phosphorylation can be specifically induced by antimicrotubule drugs, whereas DNA-damaging drugs do not. Notably, PARK2 interacted with phospho-BCL-2 (Ser70) and promoted ubiquitination of BCL-2 in an E3 ligase-dependent manner. Hence, PARK2 significantly enhanced the chemosensitivity of antimicrotubule drugs both in vitro and in vivo, while loss-of-function PARK2 mutants did not. Conclusions: Our findings explained why PARK2 selectively confers chemosensitivity to antimicrotubule drugs, but not to DNA-damaging drugs. In addition, we identified PARK2 as a novel mediator of antimicrotubule drugs sensitivity, which can predict response of breast cancer patients to antimicrotubule drugs-containing regime.
Collapse
|
10
|
Cao Y, Wang J, Tian H, Fu GH. Mitochondrial ROS accumulation inhibiting JAK2/STAT3 pathway is a critical modulator of CYT997-induced autophagy and apoptosis in gastric cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:119. [PMID: 32576206 PMCID: PMC7310559 DOI: 10.1186/s13046-020-01621-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
Abstract
Background Gastric cancer (GC) is a common form of malignant cancer in worldwide which has a poor prognosis. Despite recent improvements in the treatment of GC, the prognosis is not yet satisfactory for GC patients. CYT997, a novel microtubule-targeting agent, recently has been identified to be a promising anticancer candidate for the treatment of cancers; however, the effects of CYT997 in GC remain largely unknown. Methods Cell proliferation and apoptosis were detected by CCK8 assay and flow cytometry. The mitochondrial ROS were detected by confocal microscope and flow cytometry. Gastric cancer patient-derived xenograft (PDX) model was used to evaluate its antitumor activity of CYT997 in vivo. Results CYT997 inhibited gastric cancer cell proliferation and induced cell apoptosis and triggered autophagy. CYT997 induced apoptosis through triggering intracellular mitochondrial ROS generation in GC cells. ROS scavengers N-acetylcysteine (NAC) and Mitoquinone (MitoQ) distinctly weakened CYT997-induced cell cycle G2/M arrest and apoptosis in GC cells. Pretreatment with autophagy inhibitor 3-MA promoted the effect of CYT997 on cells apoptosis. Mechanistically, CYT997 performed its function through regulation of Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in GC cells. In addition, CYT997 inhibited growth of gastric cancer patient-derived xenograft (PDX) tumors. Conclusions CYT997 induces autophagy and apoptosis in gastric cancer by triggering mitochondrial ROS accumulation to silence JAK2/STAT3 pathway. CYT997 might be a potential antitumor drug candidate to treat GC.
Collapse
Affiliation(s)
- Ya Cao
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, 25/Ln 2200, Xietu Road, Shanghai, 200032, China
| | - Jinglong Wang
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, 25/Ln 2200, Xietu Road, Shanghai, 200032, China.
| | - Guo-Hui Fu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, China.
| |
Collapse
|
11
|
Cong Y, Cui Y, Zhu S, Cao J, Zou H, Martin TA, Qiao G, Jiang W, Yu Z. Tim-3 promotes cell aggressiveness and paclitaxel resistance through NF-κB/STAT3 signalling pathway in breast cancer cells. Chin J Cancer Res 2020; 32:564-579. [PMID: 33223752 PMCID: PMC7666787 DOI: 10.21147/j.issn.1000-9604.2020.05.02] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective Although T-cell immunoglobulin and mucin-domain containing molecule-3 (Tim-3) has been recognized as a promising target for cancer immunotherapy, its exact role in breast cancer has not been fully elucidated. Methods Tim-3 gene expression in breast cancer and its prognostic significance were analyzed. Associated mechanisms were then explored in vitro by establishing Tim-3-overexpressing breast cancer cells.
Results In a pooled analysis of The Cancer Genome Atlas (TCGA) database, Tim-3 gene expression levels were significantly higher (P<0.001) in breast cancer tissue, compared with normal tissues. Tim-3 was a prognosis indicator in breast cancer patients [relapse-free survival (RFS), P=0.004; overall survival (OS), P=0.099]. Tim-3 overexpression in Tim-3low breast cancer cells promoted aggressiveness of breast cancer cells, as evidenced by enhanced proliferation, migration, invasion, tight junction deterioration and tumor-associated tubal formation. Tim-3 also enhanced cellular resistance to paclitaxel. Furthermore, Tim-3 exerted its function by activating the NF-κB/STAT3 signalling pathway and by regulating gene expression [cyclin D1 (CCND1), C-Myc, matrix metalloproteinase-1(MMP1), TWIST, vascular endothelial growth factor (VEGF) upregulation, concomitant with E-cadherin downregulation). Lastly, Tim-3 downregulated tight junction-associated molecules zona occludens (ZO)-2, ZO-1 and occludin, which may further facilitate tumor progression.
Conclusions Tim-3 plays an oncogenic role in breast cancer and may represent a potential target for antitumor therapy.
Collapse
Affiliation(s)
- Yizi Cong
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China.,Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264001, China
| | - Yuxin Cui
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Shiguang Zhu
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264001, China
| | - Jianqiao Cao
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264001, China
| | - Haidong Zou
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264001, China
| | - Tracey A Martin
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Guangdong Qiao
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264001, China
| | - Wenguo Jiang
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Zhigang Yu
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| |
Collapse
|
12
|
Lee MC, Chen YK, Hsu YJ, Lin BR. Niclosamide inhibits the cell proliferation and enhances the responsiveness of esophageal cancer cells to chemotherapeutic agents. Oncol Rep 2019; 43:549-561. [PMID: 31894334 PMCID: PMC6967135 DOI: 10.3892/or.2019.7449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/20/2019] [Indexed: 12/24/2022] Open
Abstract
Niclosamide is an FDA-approved anthelmintic drug, and may elicit antineoplastic effects through direct STAT3 inhibition, which has been revealed in numerous human cancer cells. Chemotherapy is the standard treatment for advanced esophageal cancers, but also causes severe systemic side effects. The present study represents the first study evaluating the anticancer efficacy of niclosamide in esophageal cancers. Through western blot assay, it was demonstrated that niclosamide suppressed the STAT3 signaling pathway in esophageal adenocarcinoma cells (BE3) and esophageal squamous cell carcinoma cells (CE48T and CE81T). In addition, niclosamide inhibited cell proliferation as determined by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay and soft agar colony forming assay, and induced cell apoptosis as determined by Annexin V and PI staining. The induction of p21 and G1 arrest of the cell cycle also was revealed in niclosamide-treated CE81T cells by qPCR and flow cytometric assays, respectively. Furthermore, in the combination analysis of niclosamide and chemotherapeutic agents by MTS assay, low IC50 values were detected in cells co-treated with niclosamide, with the exception of cisplatin-treated CE81T cells. To confirm the results using an apoptosis assay, the apoptotic enhancement of niclosamide was only demonstrated in CE48T cells co-treated with 5-FU, cisplatin, or paclitaxel, and in BE3 cells co-treated with paclitaxel, but not in CE81T cells. These findings indicate a future clinical application of niclosamide in esophageal cancers.
Collapse
Affiliation(s)
- Ming-Cheng Lee
- Department of Research and Development, DrSignal BioTechnology Ltd., New Taipei City 23143, Taiwan, R.O.C
| | - Yin-Kai Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan, R.O.C
| | - Yih-Jen Hsu
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei 10051, Taiwan, R.O.C
| | - Bor-Ru Lin
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei 10051, Taiwan, R.O.C
| |
Collapse
|
13
|
Jiang H, Zhang XW, Liao QL, Wu WT, Liu YL, Huang WH. Electrochemical Monitoring of Paclitaxel-Induced ROS Release from Mitochondria inside Single Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1901787. [PMID: 31183973 DOI: 10.1002/smll.201901787] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/22/2019] [Indexed: 05/27/2023]
Abstract
Mitochondria are believed to be the major source of intracellular reactive oxygen species (ROS). However, in situ, real-time and quantitative monitoring of ROS release from mitochondria that are present in their cytosolic environment remains a great challenge. In this work, a platinized SiC@C nanowire electrode is placed into a single cell for in situ detection of ROS signals from intracellular mitochondria, and antineoplastic agent (paclitaxel) induced ROS production is successfully recorded. Further investigations indicate that complex IV (cytochrome c oxidase, COX) is the principal site for ROS generation, and significantly more ROS are generated from mitochondria in cancer cells than that from normal cells. This work provides an effective approach to directly monitor intracellular mitochondria by nanowire electrodes, and consequently obtains important physiological evidence on antineoplastic agent-induced ROS generation, which will be of great benefit for better understanding of chemotherapy at subcellular levels.
Collapse
Affiliation(s)
- Hong Jiang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Xin-Wei Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Quan-Lan Liao
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Wen-Tao Wu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yan-Ling Liu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Wei-Hua Huang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
14
|
Wang Y, Xie F, Chen D, Wang L. Inhibition of mitochondrial respiration by tigecycline selectively targets thyroid carcinoma and increases chemosensitivity. Clin Exp Pharmacol Physiol 2019; 46:890-897. [PMID: 31209921 DOI: 10.1111/1440-1681.13126] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 05/15/2019] [Accepted: 06/12/2019] [Indexed: 01/07/2023]
Abstract
The role of mitochondria in cancer and mitochondria-targeted therapy has been gaining attention for its effectiveness and selectivity between cancer and normal cells. In line with this notion, our work demonstrates that inducing mitochondrial dysfunction by tigecycline, a FDA-approved antibiotic, selectively targets thyroid cancer and enhances chemosensitivity. We found that tigecycline inhibited proliferation and induced apoptosis in a panel of thyroid cancer cell lines. Consistently, tigecycline inhibited thyroid cancer growth in mice without causing significant toxicity. The combination of tigecycline with paclitaxel achieved greater efficacy than paclitaxel alone in vitro and in vivo. Mechanistically, tigecycline inhibited mitochondrial respiration and ATP reduction through decreasing mitochondrial membrane potential and inhibiting mitochondrial translation, leading to oxidative stress and damage. In contrast, tigecycline was ineffective in mitochondrial respiration-deficient cells, confirming that tigecycline acts on thyroid cancer via inhibiting mitochondrial respiration. Interestingly, although tigecycline inhibited mitochondrial respiration in both thyroid cancer and normal thyroid cells in a similar manner, tigecycline was more effective in thyroid cancer than normal thyroid cells, suggesting that thyroid cancer cells are more dependent on mitochondrial functions than normal thyroid cells. This was supported by our observations that thyroid cancer cells had higher level of mitochondrial biogenesis compared to normal thyroid cells. Our work is the first to demonstrate that the combination of chemotherapy with tigecycline is a potential sensitizing strategy for thyroid cancer treatment. Our findings also highlight the higher dependence of thyroid cancer cells on mitochondrial functions than normal thyroid cells.
Collapse
Affiliation(s)
- Yuehua Wang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang, China
| | - Fei Xie
- Department of General Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang, China
| | - Dejie Chen
- Department of General Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang, China
| | - Ling Wang
- Department of General Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang, China
| |
Collapse
|
15
|
Cai G, Yu W, Song D, Zhang W, Guo J, Zhu J, Ren Y, Kong L. Discovery of fluorescent coumarin-benzo[b]thiophene 1, 1-dioxide conjugates as mitochondria-targeting antitumor STAT3 inhibitors. Eur J Med Chem 2019; 174:236-251. [PMID: 31048139 DOI: 10.1016/j.ejmech.2019.04.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/18/2019] [Accepted: 04/10/2019] [Indexed: 10/27/2022]
Abstract
STAT3 has been extensively studied as a potential antitumor target. Though studies on regulating STAT3 mainly focus on the inhibition of STAT3 phosphorylation at Tyr705 residue, the phosphorylation at Ser727 residue of STAT3 protein is also closely associated with the mitochondrial import of STAT3 protein. N, N-diethyl-7-aminocoumarin is a fluorescent mitochondria-targeting probe. In this study, a series of STAT3 inhibitors were developed by connecting N, N-diethyl-7-aminocoumarin fluorophore with benzo [b]thiophene 1, 1-dioxide moiety. All designed compounds displayed potent anti-proliferative activity against cancer cells. The representative compound 7a was mainly accumulated in mitochondria visualized by its fluorescence. STAT3 phosphorylation was inhibited by compound 7a at both Tyr705 and Ser727 residues. Compound 7a inhibited STAT3 phosphorylation whereas had no influence on the phosphorylation levels of STAT1, JAK2, Src and Erk1/2, indicating good selectivity of compound 7a. Moreover, compound 7a down-regulated the expression of STAT3 target genes Bcl-2 and Cyclin D1, increased ROS production and remarkably reduced the mitochondrial membrane potential to induce mitochondrial apoptotic pathway. Furthermore, compound 7ain vivo suppressed breast cancer 4T1 implanted tumor growth. Taken together, these results highlighted that compound 7a might be a promising mitochondria-targeting STAT3 inhibitor for cancer therapy.
Collapse
Affiliation(s)
- Guiping Cai
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Wenying Yu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China.
| | - Dongmei Song
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Wenda Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Jianpeng Guo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Jiawen Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Yuhao Ren
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China.
| |
Collapse
|
16
|
Pucci P, Rescigno P, Sumanasuriya S, de Bono J, Crea F. Hypoxia and Noncoding RNAs in Taxane Resistance. Trends Pharmacol Sci 2018; 39:695-709. [PMID: 29891252 DOI: 10.1016/j.tips.2018.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/15/2022]
Abstract
Taxanes are chemotherapeutic drugs employed in the clinic to treat a variety of malignancies. Despite their overall efficacy, cancer cells often display resistance to taxanes. Therefore, new strategies to increase the effectiveness of taxane-based chemotherapeutics are urgently needed. Multiple molecular players are linked to taxane resistance; these include efflux pumps, DNA repair mechanisms, and hypoxia-related pathways. In addition, emerging evidence indicates that both non-coding RNAs and epigenetic effectors might also be implicated in taxane resistance. Here we focus on the causes of taxane resistance, with the aim to envisage an integrated model of the 'taxane resistance phenome'. This model could help the development of novel therapeutic strategies to treat taxane-resistant neoplasms.
Collapse
Affiliation(s)
- Perla Pucci
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Pasquale Rescigno
- Prostate Cancer Targeted Therapy Group, The Institute of Cancer Research, Sutton, UK; Department of Clinical Medicine, University of Naples 'Federico II', Naples, Italy
| | - Semini Sumanasuriya
- Prostate Cancer Targeted Therapy Group, The Institute of Cancer Research, Sutton, UK
| | - Johann de Bono
- Prostate Cancer Targeted Therapy Group, The Institute of Cancer Research, Sutton, UK
| | - Francesco Crea
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK.
| |
Collapse
|
17
|
Meram AT, Chen J, Patel S, Kim DD, Shirley B, Covello P, Coppola D, Wei EX, Ghali G, Kevil CG, Shackelford RE. Hydrogen Sulfide Is Increased in Oral Squamous Cell Carcinoma Compared to Adjacent Benign Oral Mucosae. Anticancer Res 2018; 38:3843-3852. [PMID: 29970504 PMCID: PMC7771275 DOI: 10.21873/anticanres.12668] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/06/2018] [Accepted: 06/08/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND/AIM Hydrogen sulfide (H2S) and the enzymes that synthesize it, cystathionine-b-synthase, cystathionine γ-lyase, and 3-mercaptopyruvate, are increased in different human malignancies. Due to its short half-life, H2S concentrations have not been directly measured in a human malignancy. Here we directly measured in vivo H2S levels within oral squamous cell carcinoma (OSCC). PATIENTS AND METHODS Punch biopsies of OSCC and benign mucosae from 15 patients were analyzed by HPLC, western blotting, and tissue microarray analyses. RESULTS H2S concentrations were significantly higher in OSCC compared to adjacent benign oral mucosae. Western blot and tissue microarray studies revealed significantly increased cystathionine-b-synthase, cystathionine γ-lyase, and 3-mercaptopyruvate, phopho-Stat3, mitoNEET, hTERT, and MAPK protein levels in OSCC. CONCLUSION H2S concentrations and the enzymes that synthesize it are significantly increased in OSCC. Here, for the first time H2S concentrations within a living human malignancy were measured and compared to adjacent counterpart benign tissue.
Collapse
Affiliation(s)
- Andrew T Meram
- Head & Neck Oncologic/Microvascular Reconstructive Surgery Department of Oral & Maxillofacial/Head & Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A
| | - Jie Chen
- Department of Pathology and Translational Pathobiology Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A
| | - Stavan Patel
- Head & Neck Oncologic/Microvascular Reconstructive Surgery Department of Oral & Maxillofacial/Head & Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A
| | - Dongsoo D Kim
- Head & Neck Oncologic/Microvascular Reconstructive Surgery Department of Oral & Maxillofacial/Head & Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A
| | - Brett Shirley
- Head & Neck Oncologic/Microvascular Reconstructive Surgery Department of Oral & Maxillofacial/Head & Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A
| | - Paul Covello
- Head & Neck Oncologic/Microvascular Reconstructive Surgery Department of Oral & Maxillofacial/Head & Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Eric X Wei
- Department of Pathology and Translational Pathobiology Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A
| | - Ghali Ghali
- Head & Neck Oncologic/Microvascular Reconstructive Surgery Department of Oral & Maxillofacial/Head & Neck Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A
| | - Rodney E Shackelford
- Department of Pathology and Translational Pathobiology Louisiana State University Health Sciences Center, Shreveport, LA, U.S.A.
| |
Collapse
|
18
|
Wang Y, Lu HL, Liu YD, Yang LY, Jiang QK, Zhu XJ, Fan HN, Qian Y. Cryptotanshinone sensitizes antitumor effect of paclitaxel on tongue squamous cell carcinoma growth by inhibiting the JAK/STAT3 signaling pathway. Biomed Pharmacother 2017; 95:1388-1396. [DOI: 10.1016/j.biopha.2017.09.062] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/02/2017] [Accepted: 09/13/2017] [Indexed: 10/18/2022] Open
|