1
|
Gong X, Liu Y, Liang K, Chen Z, Ding K, Qiu L, Wei J, Du H. Cucurbitacin I exerts its anticancer effects by inducing cell cycle arrest via the KAT2a-ube2C/E2F1 pathway and inhibiting HepG2-induced macrophage M2 polarization. Biochem Biophys Res Commun 2024; 738:150508. [PMID: 39151295 DOI: 10.1016/j.bbrc.2024.150508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies globally, particularly prevalent in China, where it accounts for nearly half of the world's new cases and deaths each year, but has limited therapeutic options. This study systematically investigated the impact of cucurbitacin I on HCC cell lines including SK-Hep-1, Huh-7, and HepG2. The results revealed that cucurbitacin I not only inhibited cell proliferation, cell migration and colony formation, but also induced apoptosis in HCC cells. The apoptotic induction was accompanied by a decrease in the expression of the anti-apoptotic factor B-cell lymphoma 2 (Bcl2), and an elevation in the expression levels of pro-apoptotic factors, including tumor protein p53 (P53), bcl2 associated X-apoptosis regulator (Bax), and caspase3 (Cas3). Additionally, cucurbitacin I caused cell cycle arrest by modulating the lysine acetyltransferase 2A (KAT2A)-E2F transcription factor 1 (E2F1)/Ubiquitin-conjugating enzyme E2 C (UBE2C) signaling axis. In terms of regulation on tumor microenvironment, cucurbitacin I was demonstrated the ability to inhibit HCC cell-induced M2 polarization of macrophages. This comprehensive study unveils the multifaceted anti-cancer mechanisms of cucurbitacin I, providing robust support for its potential application in the treatment of HCC, offering new avenues for the future development of HCC treatment strategies.
Collapse
Affiliation(s)
- Xiaocheng Gong
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PR China
| | - Yunfei Liu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PR China
| | - Keying Liang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PR China
| | - Zixi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PR China
| | - Ke Ding
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PR China
| | - Li Qiu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PR China
| | - Jinfen Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PR China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PR China.
| |
Collapse
|
2
|
Üremiş MM, Türköz Y, Üremiş N. Investigation of apoptotic effects of Cucurbitacin D, I, and E mediated by Bax/Bcl-xL, caspase-3/9, and oxidative stress modulators in HepG2 cell line. Drug Dev Res 2024; 85:e22174. [PMID: 38494997 DOI: 10.1002/ddr.22174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Cucurbitacins, natural compounds highly abundant in the Cucurbitaceae plant family, are characterized by their anticancer, anti-inflammatory, and hepatoprotective properties. These compounds have potential as therapeutic agents in the treatment of liver cancer. This study investigated the association of cucurbitacin D, I, and E (CuD, CuI, and CuE) with the caspase cascade, Bcl-2 family, and oxidative stress modulators in the HepG2 cell line. We evaluated the antiproliferative effects of CuD, CuI, and CuE using the MTT assay. We analyzed Annexin V/PI double staining, cell cycle, mitochondrial membrane potential, and wound healing assays at different doses of the three compounds. To examine the modulation of the caspase cascade, we determined the protein and gene expression levels of Bax, Bcl-xL, caspase-3, and caspase-9. We evaluated the total antioxidant status (TAS), total oxidant status (TOS), superoxide dismutase (SOD), glutathione (GSH), Total, and Native Thiol levels to measure cellular redox status. CuD, CuI, and CuE suppressed the proliferation of HepG2 cells in a dose-dependent manner. The cucurbitacins induced apoptosis by increasing caspase-3, caspase-9, and Bax activity, inhibiting Bcl-xL activation, causing loss of ΔΨm, and suppressing cell migration. Furthermore, cucurbitacins modulated oxidative stress by increasing TOS levels and decreasing SOD, GSH, TAS, and total and native Thiol levels. Our findings suggest that CuD, CuI, and CuE exert apoptotic effects on the hepatocellular carcinoma cell line by regulating Bax/Bcl-xL, caspase-3/9 signaling, and causing intracellular ROS increase in HepG2 cells.
Collapse
Affiliation(s)
- Muhammed Mehdi Üremiş
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Yusuf Türköz
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Nuray Üremiş
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| |
Collapse
|
3
|
Vidal-Gutiérrez M, Torres-Moreno H, Arenas-Luna V, Loredo-Mendoza ML, Tejeda-Dominguez F, Velazquez C, Vilegas W, Hernández-Gutiérrez S, Robles-Zepeda RE. Standardized phytopreparations and cucurbitacin IIb from Ibervillea sonorae (S. Watson) greene induce apoptosis in cervical cancer cells by Nrf2 inhibition. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115606. [PMID: 35944738 DOI: 10.1016/j.jep.2022.115606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/23/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Ibervillea sonorae (S. Watson) Greene is a plant from northwestern Mexico, known as "Wereke" or "Guareque", used by the Mayo ethnic group to treat diabetes and cancer. Cucurbitacin IIb (CIIb), isolated from I. sonorae has apoptotic and antitumor activity in a model of cervical cancer with the HeLa cell line. One pathway affected by cucurbitacins is Nrf2, a glutathione transferase (GST) transcription factor, important in the regulation of mitochondrial oxidative stress (MOS). A signal of MOS is the change in the mitochondrial membrane potential (ΔΨm), which has been detected in HeLa in the presence of CIIb. Fito-Ison-EtOH (Etanison) and Fito-Ison-EtOAc (Acetison) are phytopreparations from I. sonorae standardized according to their CIIb content (6.7 mg/g and 18.4 mg/g of CIIb, respectively). Etanison and Acetison have been reported to induce morphological changes in HeLa like those induced by CIIb. AIM OF THE STUDY To evaluate the apoptotic and Nrf2 inhibition activity of the phytopreparations Acetison and Etanison from Ibervillea Sonorae in the HeLa cervical cancer cell line. MATERIALS AND METHODS Antiproliferative activity was evaluated by the MTT method at 24, 48, and 72 h. For Acetison and Etanison, serial concentrations from 6.25 μg/mL to 100 μg/mL were tested, and for CIIb from 1.56 μg/mL to 50 μg/mL. The expression of Nrf2, caspase 3, and caspase 9 was evaluated by western blot, using concentrations of 30 μg/mL for Acetison, 50 μg/mL for Etanison, and 15 μg/mL for CIIb. Cisplatin was used as a positive control. RESULTS AND CONCLUSIONS Apoptotic activity of Etanison and Acetison was demonstrated in HeLa, due to the presence of caspase-9 and caspase-3 in western blot assays. Likewise, both the phytopreparations and CIIb showed inhibition of Nrf2, associating apoptotic activity with the inhibition of the GST transcription factor. In this sense, the phytopreparations of I. sonorae, as well as their derivatives, have the potential to obtain and develop anticancer products.
Collapse
Affiliation(s)
- Max Vidal-Gutiérrez
- Departamento de Ciencias Químico Biológicas y de la Salud, Universidad de Sonora - Blvd. Luis Donaldo Colosio esq. Rosales S/N, Centro, Hermosillo Sonora, CP: 83000, Mexico; Universidade Estadual Paulista (UNESP), Faculdade de Ciências Farmacêuticas, Rodovia Araraquara - Jaú, Km 1, Araraquara, São Paulo, CEP: 14800-903, Brazil
| | - Heriberto Torres-Moreno
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora - Avenida Universidad e Irigoyen, Caborca Sonora, CP:83621, Mexico
| | - Víctor Arenas-Luna
- Facultad de Ciencias de la Salud, Escuela de Medicina, Universidad Panamericana - Augusto Rodin No. 498, Col. Insurgentes Mixcoac, Ciudad de México, CP: 03920, Mexico
| | - María Lilia Loredo-Mendoza
- Facultad de Ciencias de la Salud, Escuela de Medicina, Universidad Panamericana - Augusto Rodin No. 498, Col. Insurgentes Mixcoac, Ciudad de México, CP: 03920, Mexico
| | - Farid Tejeda-Dominguez
- Facultad de Ciencias de la Salud, Escuela de Medicina, Universidad Panamericana - Augusto Rodin No. 498, Col. Insurgentes Mixcoac, Ciudad de México, CP: 03920, Mexico
| | - Carlos Velazquez
- Departamento de Ciencias Químico Biológicas y de la Salud, Universidad de Sonora - Blvd. Luis Donaldo Colosio esq. Rosales S/N, Centro, Hermosillo Sonora, CP: 83000, Mexico
| | - Wagner Vilegas
- Universidade Estadual Paulista (UNESP), Faculdade de Ciências Farmacêuticas, Rodovia Araraquara - Jaú, Km 1, Araraquara, São Paulo, CEP: 14800-903, Brazil; Universidade Estadual Paulista (UNESP), Coastal Campus of São Vicente, Praça Infante Dom Henrique s/n, São Vicente, São Paulo, CEP 11330-205, Brazil
| | - Salomón Hernández-Gutiérrez
- Facultad de Ciencias de la Salud, Escuela de Medicina, Universidad Panamericana - Augusto Rodin No. 498, Col. Insurgentes Mixcoac, Ciudad de México, CP: 03920, Mexico.
| | - Ramón E Robles-Zepeda
- Departamento de Ciencias Químico Biológicas y de la Salud, Universidad de Sonora - Blvd. Luis Donaldo Colosio esq. Rosales S/N, Centro, Hermosillo Sonora, CP: 83000, Mexico.
| |
Collapse
|
4
|
Xu D, Shen H, Tian M, Chen W, Zhang X. Cucurbitacin I inhibits the proliferation of pancreatic cancer through the JAK2/STAT3 signalling pathway in vivo and in vitro. J Cancer 2022; 13:2050-2060. [PMID: 35517401 PMCID: PMC9066209 DOI: 10.7150/jca.65875] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 01/30/2022] [Indexed: 01/08/2023] Open
Abstract
Pancreatic cancer is one of the most aggressive solid malignancies, as it has a 5-year survival rate of less than 10%. The growth and invasion of pancreatic cancer cells into normal tissues and organs make resection and treatment difficult. Finding an effective chemotherapy drug for this disease is crucial. In this study, we selected the tetracyclic triterpenoid compound cucurbitacin I, which may be used as a potential therapeutic drug for treating pancreatic cancer. First, we found that cucurbitacin I inhibited pancreatic cancer proliferation in a dose-time dependent manner. Further studies have shown that cucurbitacin I blocks the cell cycle of pancreatic cancer in the G2/M phase and induces cell apoptosis. In addition, under the action of the compound, the invasion ability of cells was greatly reduced and markedly impaired the growth of pancreatic tumour xenografts in nude mice. Furthermore, the decrease in pancreatic cancer cell proliferation caused by cucurbitacin I appeared to involve JAK2/STAT3 signalling pathway inhibition, and the use of JAK2/STAT3 activators effectively restored the inhibition. In conclusion, our research may provide a basis for the further development of pancreatic cancer treatment drugs.
Collapse
Affiliation(s)
- Dongchao Xu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou310000, China.,Hangzhou Institute of Digestive Diseases, Hangzhou310000, China.,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou310000, China
| | - Hongzhang Shen
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou310000, China.,Hangzhou Institute of Digestive Diseases, Hangzhou310000, China.,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou310000, China
| | - Mengyao Tian
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou310000, China
| | - Wangyang Chen
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou310000, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou310000, China.,Hangzhou Institute of Digestive Diseases, Hangzhou310000, China.,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou310000, China
| |
Collapse
|
5
|
Ju H, Liu C, Zhang G, Xu C, Wang H, Fan H. Neuroprotective potential of nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element signaling modulator cucurbitacin I upon glucose and oxygen deprivation/reperfusion (OGD/RP). Hum Exp Toxicol 2022; 41:9603271221104450. [PMID: 35632987 DOI: 10.1177/09603271221104450] [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: 11/17/2022]
Abstract
This study aimed to investigate the inhibitory effect and mechanism of Cucurbitacin I (Cu I) on apoptosis, oxidative stress, and mitophagy in PC12 cells with glucose and oxygen deprivation/reperfusion (OGD/RP) injury. OGD/RP cell injury model was established by gas anoxic cell incubator and glucose-free medium. The cells were divided into the control group, OGD/RP group, OGD/RP + Cu I group, and OGD/RP + Cu I + 2 µM nuclear factor erythroid 2-related factor 2 (Nrf2) inhibitor ML385 group. The results showed that apoptotic rate and reactive oxygen species (ROS) production were significantly increased in OGD/RP group, which were reversed by Cu I pretreatment. Meanwhile, western blot analysis proved that Cu I inhibited OGD/RP-induced mitophagy, manifested as the decreased expression of PTEN-induced kinase 1 (PINK1) and parkin RBR E3 ubiquitin-protein ligase (Parkin), and light chain 3 (LC3) Ⅱ∕LC3 I, as well as the increased expression of P62. Furthermore, immunofluorescence (IF) staining showed that Cu I reduced the co-localized puncta of LC3 with TOM20 in OGD/RP-induced PC12 cells. Similarly, transmission electron microscope finding is consistent with the IF results. Mechanically, after Cu I and OGD/RP treatments, nuclear Nrf2 expression and the levels of downstream target genes were significantly upregulated compared with OGD/RP alone treatment. Nrf2 inhibition reversed the protective effects of Cu I on OGD/RP-induced injury in PC12 cells. The present study provides evidence of the neuroprotective effect of Cu I unraveling its potential as a potential therapeutic candidate for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Hu Ju
- Department of Neurosurgery, Qinghai University Affiliated Hospital, Chengxi District, Xining, Qinghai, China
| | - Chuanchuan Liu
- Key Laboratory of Hydatid Research, Qinghai University Affiliated Hospital, Qinghai Province Key Laboratory of Hydatid Disease Research, Chengxi District, Xining, Qinghai, China
| | - Guanghua Zhang
- Department of Neurosurgery, Qinghai University Affiliated Hospital, Chengxi District, Xining, Qinghai, China
| | - Changlin Xu
- Department of Neurosurgery, Qinghai University Affiliated Hospital, Chengxi District, Xining, Qinghai, China
| | - Hu Wang
- 599265Health Commission of Qinghai Province, Chengxi District, Xining, Qinghai, China
| | - Haining Fan
- Department of Hepatopancreatobiliary Surgery, Qinghai University Affiliated Hospital, Qinghai Province Key Laboratory of Hydatid Disease Research, Chengxi District, Xining, Qinghai, China
| |
Collapse
|
6
|
Drissi F, Lahfa F, Gonzalez T, Peiretti F, Tanti JF, Haddad M, Fabre N, Govers R. A Citrullus colocynthis fruit extract acutely enhances insulin-induced GLUT4 translocation and glucose uptake in adipocytes by increasing PKB phosphorylation. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113772. [PMID: 33418030 DOI: 10.1016/j.jep.2020.113772] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/14/2020] [Accepted: 12/24/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Citrullus colocynthis (L.) Schrad is a common fruit in traditional medicine and used as remedy against various diseases, especially diabetes. Up to now, its anti-diabetic effects have been fully attributed to its enhancement of pancreatic insulin secretion. Whether C. colocynthis also ameliorates insulin action in peripheral tissues has not been investigated. AIM OF THE STUDY In the present study, using 3T3-L1 adipocytes as cell model, we have investigated whether colocynth fruit extracts affect insulin action. MATERIALS AND METHODS Various extracts were prepared from the C. colocynthis fruit and screened using a cell-based 96 well plate GLUT4 translocation assay. Promising extracts were further studied for their effects on glucose uptake and cell viability. The effect on insulin signal transduction was determined by Western blot and the molecular composition was established by LC-MS. RESULTS The ethyl acetate fractions of aqueous non-defatted extracts of seed and pulp, designated Sna1 and Pna1, acutely enhanced insulin-induced GLUT4 translocation. In accordance, both extracts increased insulin-stimulated cellular glucose uptake. Pna1, which displayed greater effects on GLUT4 and glucose uptake than Sna1, was further investigated and was demonstrated to increase GLUT4 translocation without changing the half-maximum dose (ED50) of insulin, nor changing GLUT4 translocation kinetics. At the molecular level, Pna1 was found to enhance insulin-induced PKB phosphorylation without changing phosphorylation of the insulin receptor. Pna1 appeared not to be toxic to cells and, like insulin, restored cell viability during serum starvation. By investigating the molecular composition of Pna1, nine compounds were identified that made up 87% of the mass of the extract, one of which is likely to be responsible for the insulin-enhancing effects of Pna1. CONCLUSIONS The C. colocynthis fruit possesses insulin-enhancing activity. This activity may explain in part its anti-diabetic effects in traditional medicine. It also identifies the C. colocynthis as a source of a potential novel insulin enhancer that may prove to be useful to reduce hyperglycemia in type 2 diabetes.
Collapse
Affiliation(s)
- Farah Drissi
- Department of Synthesis and Biological Activities, University of Abou Bekr Belkaïd, 119 13000, Tlemcen, Algeria.
| | - Farid Lahfa
- Department of Synthesis and Biological Activities, University of Abou Bekr Belkaïd, 119 13000, Tlemcen, Algeria.
| | - Teresa Gonzalez
- Aix Marseille Université, INSERM, INRAE, C2VN, 13385, Marseille, France.
| | - Franck Peiretti
- Aix Marseille Université, INSERM, INRAE, C2VN, 13385, Marseille, France.
| | - Jean-François Tanti
- Université Côte D'Azur, INSERM, C3M, Team "Cellular and Molecular Physiopathology of Obesity", 06204, Nice, France.
| | - Mohamed Haddad
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31400, Toulouse, France.
| | - Nicolas Fabre
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31400, Toulouse, France.
| | - Roland Govers
- Aix Marseille Université, INSERM, INRAE, C2VN, 13385, Marseille, France.
| |
Collapse
|
7
|
Liang Y, Zhang T, Ren L, Jing S, Li Z, Zuo P, Li T, Wang Y, Zhang J, Wei Z. Cucurbitacin IIb induces apoptosis and cell cycle arrest through regulating EGFR/MAPK pathway. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 81:103542. [PMID: 33161110 DOI: 10.1016/j.etap.2020.103542] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/23/2020] [Accepted: 10/30/2020] [Indexed: 05/06/2023]
Abstract
Epidermal growth factor receptor (EGFR) is considered as a valid target in the clinical trials of anticancer therapy and tyrosine kinase inhibitors (TKIs) of EGFR are approved for cancer treatments. In present work, cucurbitacin IIb (CuIIb) was confirmed to exhibit the proliferation inhibitory activity in A549 cells. CuIIb induced apoptosis via STAT3 pathway, which was mitochondria-mediated and caspase-dependent. CuIIb also suppressed the cell cycle and induced G2/M phase cell cycle arrest. CuIIb was capable of suppressing the signal transmitting of the EGFR/mitogen-activated protein kinase (MAPK) pathway which was responsible for the apoptosis and cell cycle arrest. Homogeneous time-resolved fluorescence (HTRF) analysis demonstrated that the kinase activity of EGFR was inhibited by CuIIb. Molecular docking suggested that the CuIIb-EGFR binding fundamentally depends on the contribution of both hydrophobic and hydrogen-bonding interactions. Hence CuIIb may serve as a potential EGFR TKI.
Collapse
Affiliation(s)
- Yuan Liang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Siyuan Jing
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Zhuolin Li
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Peng Zuo
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Tiezhu Li
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Yongjun Wang
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China.
| | - Zhengyi Wei
- Institute of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China.
| |
Collapse
|
8
|
Lin X, Farooqi AA. Cucurbitacin mediated regulation of deregulated oncogenic signaling cascades and non-coding RNAs in different cancers: Spotlight on JAK/STAT, Wnt/β-catenin, mTOR, TRAIL-mediated pathways. Semin Cancer Biol 2020; 73:302-309. [PMID: 33152487 DOI: 10.1016/j.semcancer.2020.10.012] [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: 09/18/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 01/03/2023]
Abstract
Research over decades has enabled us in developing a better understanding of the multifaceted and heterogeneous nature of cancer. High-throughput technologies have helped the researchers in unraveling of the underlying mechanisms which centrally regulate cancer onset, metastasis and drug resistance. Our rapidly expanding knowledge about signal transduction cascade has added another layer of complexity to already complicated nature of cancer. Deregulation of cell signaling pathways played a linchpin role in carcinogenesis and metastasis. Cucurbitacins have gained tremendous attention because of their remarkable pharmacological properties and considerable ability to mechanistically modulate myriad of cell signaling pathways in different cancers. In this review, we have attempted to provide a mechanistic and comprehensive analysis of regulation of oncogenic pathways by cucurbitacins in different cancers. We have partitioned this review into separate sections for exclusive analysis of each signaling pathway and critical assessment of the knowledge gaps. In this review, we will summarize most recent and landmark developments related to regulation of Wnt/β-catenin, JAK/STAT, mTOR, VEGFR, EGFR and Hippo pathway by cucurbitacins. Moreover, we will also address how cucurbitacins regulate DNA damage repair pathway and TRAIL-driven signaling in various cancers. However, there are still outstanding questions related to regulation of SHH/GLI, TGF/SMAD and Notch-driven pathway by cucurbitacins in different cancers. Future studies must converge on the analysis of full-fledge potential of cucurbitacins by in-depth analysis of these pathways and how these pathways can be therapeutically targeted by cucurbitacins.
Collapse
Affiliation(s)
- Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan.
| |
Collapse
|
9
|
Jing S, Zou H, Wu Z, Ren L, Zhang T, Zhang J, Wei Z. Cucurbitacins: Bioactivities and synergistic effect with small-molecule drugs. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
10
|
Baker MJ, Cooke M, Kreider-Letterman G, Garcia-Mata R, Janmey PA, Kazanietz MG. Evaluation of active Rac1 levels in cancer cells: A case of misleading conclusions from immunofluorescence analysis. J Biol Chem 2020; 295:13698-13710. [PMID: 32817335 DOI: 10.1074/jbc.ra120.013919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/23/2020] [Indexed: 12/16/2022] Open
Abstract
A large number of aggressive cancer cell lines display elevated levels of activated Rac1, a small GTPase widely implicated in cytoskeleton reorganization, cell motility, and metastatic dissemination. A commonly accepted methodological approach for detecting Rac1 activation in cancer cells involves the use of a conformation-sensitive antibody that detects the active (GTP-bound) Rac1 without interacting with the GDP-bound inactive form. This antibody has been extensively used in fixed cell immunofluorescence and immunohistochemistry. Taking advantage of prostate and pancreatic cancer cell models known to have high basal Rac1-GTP levels, here we have established that this antibody does not recognize Rac1 but rather detects the intermediate filament protein vimentin. Indeed, Rac1-null PC3 prostate cancer cells or cancer models with low levels of Rac1 activation still show a high signal with the anti-Rac1-GTP antibody, which is lost upon silencing of vimentin expression. Moreover, this antibody was unable to detect activated Rac1 in membrane ruffles induced by epidermal growth factor stimulation. These results have profound implications for the study of this key GTPase in cancer, particularly because a large number of cancer cell lines with characteristic mesenchymal features show simultaneous up-regulation of vimentin and high basal Rac1-GTP levels when measured biochemically. This misleading correlation can lead to assumptions about the validity of this antibody and inaccurate conclusions that may affect the development of appropriate therapeutic approaches for targeting the Rac1 pathway.
Collapse
Affiliation(s)
- Martin J Baker
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Medicine, Einstein Medical Center Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | - Paul A Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
11
|
In vitro antitumor effect of cucurbitacin E on human lung cancer cell line and its molecular mechanism. Chin J Nat Med 2020; 18:483-490. [DOI: 10.1016/s1875-5364(20)30058-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 11/22/2022]
|
12
|
P-REX1-Independent, Calcium-Dependent RAC1 Hyperactivation in Prostate Cancer. Cancers (Basel) 2020; 12:cancers12020480. [PMID: 32092966 PMCID: PMC7072377 DOI: 10.3390/cancers12020480] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
The GTPase Rac1 is a well-established master regulator of cell motility and invasiveness contributing to cancer metastasis. Dysregulation of the Rac1 signaling pathway, resulting in elevated motile and invasive potential, has been reported in multiple cancers. However, there are limited studies on the regulation of Rac1 in prostate cancer. Here, we demonstrate that aggressive androgen-independent prostate cancer cells display marked hyperactivation of Rac1. This hyperactivation is independent of P-Rex1 activity or its direct activators, the PI3K product PIP3 and Gβγ subunits. Furthermore, we demonstrate that the motility and invasiveness of PC3 prostate cancer cells is independent of P-Rex1, supporting the analysis of publicly available datasets indicating no correlation between high P-Rex1 expression and cancer progression in patients. Rac1 hyperactivation was not related to the presence of activating Rac1 mutations and was insensitive to overexpression of a Rac-GAP or the silencing of specific Rac-GEFs expressed in prostate cancer cells. Interestingly, active Rac1 levels in these cells were markedly reduced by elevations in intracellular calcium or by serum stimulation, suggesting the presence of an alternative means of Rac1 regulation in prostate cancer that does not involve previously established paradigms.
Collapse
|
13
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 331] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
14
|
Zhang J, Song Y, Liang Y, Zou H, Zuo P, Yan M, Jing S, Li T, Wang Y, Li D, Zhang T, Wei Z. Cucurbitacin IIa interferes with EGFR-MAPK signaling pathway leads to proliferation inhibition in A549 cells. Food Chem Toxicol 2019; 132:110654. [DOI: 10.1016/j.fct.2019.110654] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/25/2019] [Accepted: 06/29/2019] [Indexed: 12/20/2022]
|
15
|
Wang W, Yang H, Li Y, Zheng Z, Liu Y, Wang H, Mu Y, Yao Q. Identification of 16,25- O-diacetyl-cucurbitane F and 25- O-acetyl-23,24-dihydrocucurbitacin F as novel anti-cancer chemicals. ROYAL SOCIETY OPEN SCIENCE 2018; 5:180723. [PMID: 30225067 PMCID: PMC6124052 DOI: 10.1098/rsos.180723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/11/2018] [Indexed: 05/14/2023]
Abstract
Seven new cucurbitane glucosides, hemslepensides J-P (1-7), and two known compounds, 16,25-O-diacetyl-cucurbitane F (8) and 25-O-acetyl-23,24-dihydrocucurbitacin F (9), were isolated from the tubers of Hemsleya pengxianensis var. jinfushanensis. The structures of 1-7 were elucidated using infrared absorption spectroscopy, nuclear magnetic resonance spectroscopy and high-resolution electrospray ionization mass spectrometry. The treatment of HT29 cells, human colon cancer cells, with compounds 8 and 9 inhibited cell proliferation. Further study demonstrated that compounds 8 and 9 induced F-actin aggregation, G2/M phase cell cycle arrest and cell apoptosis in HT29 cells. In summary, the present study enriched the chemical composition research of H. pengxianensis, and suggested that the compounds 8/9 treatment may be a potentially useful therapeutic option for colon cancer.
Collapse
Affiliation(s)
- Wenxue Wang
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan 250200, Shandong, People's Republic of China
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, People's Republic of China
| | - Haoran Yang
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan 250200, Shandong, People's Republic of China
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, People's Republic of China
| | - Ying Li
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, People's Republic of China
| | - Zhongfei Zheng
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, People's Republic of China
| | - Yongjun Liu
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, People's Republic of China
| | - Haiyang Wang
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, People's Republic of China
| | - Yanling Mu
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, People's Republic of China
- Authors for correspondence: Yanling Mu e-mail:
| | - Qingqiang Yao
- Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Biotech-Drugs Ministry of Health, Jinan 250062, Shandong, People's Republic of China
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan 250062, Shandong, People's Republic of China
- Authors for correspondence: Qingqiang Yao e-mail:
| |
Collapse
|
16
|
Barrio-Real L, Wertheimer E, Garg R, Abba MC, Kazanietz MG. Characterization of a P-Rex1 gene signature in breast cancer cells. Oncotarget 2018; 7:51335-51348. [PMID: 27351228 PMCID: PMC5239479 DOI: 10.18632/oncotarget.10285] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/12/2016] [Indexed: 01/29/2023] Open
Abstract
The Rac nucleotide Exchange Factor (Rac-GEF) P-Rex1 is highly expressed in breast cancer, specifically in the luminal subtype, and is an essential mediator of actin cytoskeleton reorganization and cell migratory responses induced by stimulation of ErbB and other tyrosine-kinase receptors. Heregulin (HRG), a growth factor highly expressed in mammary tumors, causes the activation of P-Rex1 and Rac1 in breast cancer cells via ErbB3, leading to a motile response. Since there is limited information about P-Rex1 downstream effectors, we carried out a microarray analysis to identify genes regulated by this Rac-GEF after stimulation of ErbB3 with HRG. In T-47D breast cancer cells, HRG treatment caused major changes in gene expression, including genes associated with motility, adhesion, invasiveness and metastasis. Silencing P-Rex1 expression from T-47D cells using RNAi altered the induction and repression of a subset of HRG-regulated genes, among them genes associated with extracellular matrix organization, migration, and chemotaxis. HRG induction of MMP10 (matrix metalloproteinase 10) was found to be highly sensitive both to P-Rex1 depletion and inhibition of Rac1 function by the GTPase Activating Protein (GAP) β2-chimaerin, suggesting the dependence of the P-Rex1/Rac1 pathway for the induction of genes critical for breast cancer invasiveness. Notably, there is a significant association in the expression of P-Rex1 and MMP10 in human luminal breast cancer, and their co-expression is indicative of poor prognosis.
Collapse
Affiliation(s)
- Laura Barrio-Real
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Eva Wertheimer
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rachana Garg
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Martin C Abba
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
17
|
Prophetic medicine as potential functional food elements in the intervention of cancer: A review. Biomed Pharmacother 2017; 95:614-648. [DOI: 10.1016/j.biopha.2017.08.043] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/05/2017] [Accepted: 08/07/2017] [Indexed: 01/01/2023] Open
|
18
|
From bench (laboratory) to bed (hospital/home): How to explore effective natural and synthetic PAK1-blockers/longevity-promoters for cancer therapy. Eur J Med Chem 2017; 142:229-243. [PMID: 28814374 DOI: 10.1016/j.ejmech.2017.07.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/19/2017] [Accepted: 07/21/2017] [Indexed: 12/19/2022]
Abstract
PAK family kinases are RAC/CDC42-activated kinases that were first found in a soil amoeba 4 decades ago, and 2 decades later, were discovered in mammals as well. Since then at least 6 members of this family have been identified in mammals. One of them called PAK1 has been best studied so far, mainly because it is essential not only for malignant cell growth and metastasis, but also for many other diseases/disorders such as diabetes (type 2), AD (Alzheimer's disease), hypertension, and a variety of inflammatory or infectious diseases, which definitely shorten our lifespan. Moreover, PAK1-deficient mutant of C. elegans lives longer than the wild-type by 60%, clearly indicating that PAK1 is not only an oncogenic but also ageing kinase. Thus, in theory, both anti-oncogenic and longevity-promoting activities are among the "intrinsic" properties or criteria of "clinically useful" PAK1-blockers. There are a variety of PAK1-blocking natural products such as propolis and curcumin which indeed extend the healthy lifespan of small animals such as C. elegans by inducing the autophagy. Recently, we managed to synthesize a series of potent water-soluble and highly cell-permeable triazolyl esters of COOH-bearing PAK1-blockers such as Ketorolac, ARC (artepillin C) and CA (caffeic acid) via "Click Chemistry" that boosts their anti-cancer activity over 500-fold, mainly by increasing their cell-permeability, and one of them called 15K indeed extends the lifespan of C. elegans. In this mini-review we shall discuss both synthetic and natural PAK1-blockers, some of which would be potentially useful for cancer therapy with least side effect (rather promoting the longevity as well).
Collapse
|
19
|
Xu Q, Huff LP, Fujii M, Griendling KK. Redox regulation of the actin cytoskeleton and its role in the vascular system. Free Radic Biol Med 2017; 109:84-107. [PMID: 28285002 PMCID: PMC5497502 DOI: 10.1016/j.freeradbiomed.2017.03.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/17/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
The actin cytoskeleton is critical for form and function of vascular cells, serving mechanical, organizational and signaling roles. Because many cytoskeletal proteins are sensitive to reactive oxygen species, redox regulation has emerged as a pivotal modulator of the actin cytoskeleton and its associated proteins. Here, we summarize work implicating oxidants in altering actin cytoskeletal proteins and focus on how these alterations affect cell migration, proliferation and contraction of vascular cells. Finally, we discuss the role of oxidative modification of the actin cytoskeleton in vivo and highlight its importance for vascular diseases.
Collapse
Affiliation(s)
- Qian Xu
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States; Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Lauren P Huff
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States
| | - Masakazu Fujii
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States.
| |
Collapse
|
20
|
Fluctuation of ROS regulates proliferation and mediates inhibition of migration by reducing the interaction between DLC1 and CAV-1 in breast cancer cells. In Vitro Cell Dev Biol Anim 2017; 53:354-362. [PMID: 28130753 DOI: 10.1007/s11626-016-0123-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/14/2016] [Indexed: 12/30/2022]
Abstract
The aim of our present study was to elucidate the effects of up-regulation and down-regulation of intracellular reactive oxygen species (ROS) level on proliferation, migration, and related molecular mechanism. Breast cancer cells were treated by catalase or H2O2. MTT, colony formation assay, and Hoechst/PI staining were used to evaluate proliferation and apoptosis. The level of intracellular ROS was measured by dichlorodihydrofluorescein diacetate probes. The ability of migration was detected by wound healing. Western blotting and coimmunoprecipitation (co-IP) were used to determine the expression of DLC1 and CAV-1 and their interaction. Our data indicated that up-regulation of intracellular ROS induced by H2O2 significantly inhibited proliferation and induced apoptosis accompanying G1 cell cycle arrest and elevated expression of p53. For cell migration, either up-regulation or down-regulation of ROS induced migration inhibition with reduction of interaction between DLC1 and CAV-1. Our results suggested that up-regulation of intracellular ROS inhibited proliferation by promoting expression of p53 and induced G1 cycle arrest and apoptosis. Fluctuation of ROS inhibited migration through reducing the interaction between DLC1 and CAV-1.
Collapse
|
21
|
Heregulin/ErbB3 Signaling Enhances CXCR4-Driven Rac1 Activation and Breast Cancer Cell Motility via Hypoxia-Inducible Factor 1α. Mol Cell Biol 2016; 36:2011-26. [PMID: 27185877 DOI: 10.1128/mcb.00180-16] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/06/2016] [Indexed: 01/11/2023] Open
Abstract
The growth factor heregulin (HRG), a ligand of ErbB3 and ErbB4 receptors, contributes to breast cancer development and the promotion of metastatic disease, and its expression in breast tumors has been associated with poor clinical outcome and resistance to therapy. In this study, we found that breast cancer cells exposed to sustained HRG treatment show markedly enhanced Rac1 activation and migratory activity in response to the CXCR4 ligand SDF-1/CXCL12, effects mediated by P-Rex1, a Rac-guanine nucleotide exchange factor (GEF) aberrantly expressed in breast cancer. Notably, HRG treatment upregulates surface expression levels of CXCR4, a G protein-coupled receptor (GPCR) implicated in breast cancer metastasis and an indicator of poor prognosis in breast cancer patients. A detailed mechanistic analysis revealed that CXCR4 upregulation and sensitization of the Rac response/motility by HRG are mediated by the transcription factor hypoxia-inducible factor 1α (HIF-1α) via ErbB3 and independently of ErbB4. HRG caused prominent induction in the nuclear expression of HIF-1α, which transcriptionally activates the CXCR4 gene via binding to a responsive element located in positions -1376 to -1372 in the CXCR4 promoter, as revealed by mutagenesis analysis and chromatin immunoprecipitation (ChIP). Our results uncovered a novel function for ErbB3 in enhancing breast cancer cell motility and sensitization of the P-Rex1/Rac1 pathway through HIF-1α-mediated transcriptional induction of CXCR4.
Collapse
|
22
|
Flavone inhibits migration through DLC1/RhoA pathway by decreasing ROS generation in breast cancer cells. In Vitro Cell Dev Biol Anim 2016; 52:589-97. [DOI: 10.1007/s11626-016-0010-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/11/2016] [Indexed: 12/14/2022]
|
23
|
Deng C, Zhang B, Zhang S, Duan C, Cao Y, Kang W, Yan H, Ding X, Zhou F, Wu L, Duan G, Shen S, Xu G, Zhang W, Chen M, Huang S, Zhang X, Lv Y, Ling T, Wang L, Zou X. Low nanomolar concentrations of Cucurbitacin-I induces G2/M phase arrest and apoptosis by perturbing redox homeostasis in gastric cancer cells in vitro and in vivo. Cell Death Dis 2016; 7:e2106. [PMID: 26890145 PMCID: PMC5399186 DOI: 10.1038/cddis.2016.13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 12/20/2022]
Abstract
Cucurbitacin-I (Cu-I, also known as Elatericin B or JSI-124) is developed to inhibit constitutive and abnormal activation of STAT3 in many cancers, demonstrating a potent anticancer activity by targeting disruption of STAT3 function. Here, we for the first time systematically studied the underlying molecular mechanisms of Cu-I-induced gastric cancer cell death both in vitro and in vivo. In our study, we show that Cu-I markedly inhibits gastric cancer cell growth by inducing G2/M phase cell cycle arrest and apoptosis at low nanomolar concentrations via a STAT3-independent mechanism. Notably, Cu-I significantly decreases intracellular GSH/GSSG ratio by inhibiting NRF2 pathway to break cellular redox homeostasis, and subsequently induces the expression of GADD45α in a p53-independent manner, and activates JNK/p38 MAPK signaling. Interestingly, Cu-I-induced GADD45α and JNK/p38 MAPK signaling form a positive feedback loop and can be reciprocally regulated by each other. Therefore, the present study provides new insights into the mechanisms of antitumor effects of Cu-I, supporting Cu-I as an attractive therapeutic drug in gastric cancer by modulating the redox balance.
Collapse
Affiliation(s)
- C Deng
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - B Zhang
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - S Zhang
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - C Duan
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - Y Cao
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - W Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - H Yan
- Department of Laboratory Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - X Ding
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - F Zhou
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - L Wu
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - G Duan
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - S Shen
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - G Xu
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - W Zhang
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - M Chen
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - S Huang
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - X Zhang
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - Y Lv
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - T Ling
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - L Wang
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| | - X Zou
- Department of Gastroenterology, Drum Tower Hospital, Medical School of Nanjing University, Jiangsu Province, China
| |
Collapse
|
24
|
Sari-Hassoun M, Clement MJ, Hamdi I, Bollot G, Bauvais C, Joshi V, Toma F, Burgo A, Cailleret M, Rosales-Hernández MC, Macias Pérez ME, Chabane-Sari D, Curmi PA. Cucurbitacin I elicits the formation of actin/phospho-myosin II co-aggregates by stimulation of the RhoA/ROCK pathway and inhibition of LIM-kinase. Biochem Pharmacol 2015; 102:45-63. [PMID: 26707799 DOI: 10.1016/j.bcp.2015.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/15/2015] [Indexed: 12/11/2022]
Abstract
Cucurbitacins are cytotoxic triterpenoid sterols isolated from plants. One of their earliest cellular effect is the aggregation of actin associated with blockage of cell migration and division that eventually lead to apoptosis. We unravel here that cucurbitacin I actually induces the co-aggregation of actin with phospho-myosin II. This co-aggregation most probably results from the stimulation of the Rho/ROCK pathway and the direct inhibition of the LIMKinase. We further provide data that suggest that the formation of these co-aggregates is independent of a putative pro-oxidant status of cucurbitacin I. The results help to understand the impact of cucurbitacins on signal transduction and actin dynamics and open novel perspectives to use it as drug candidates for cancer research.
Collapse
Affiliation(s)
- Meryem Sari-Hassoun
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France; Laboratoire des Produits Naturels, LAPRONA, Université Abou Bekr Belkaid, Tlemcen 13000, Algeria
| | - Marie-Jeanne Clement
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Imane Hamdi
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | | | | | - Vandana Joshi
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Flavio Toma
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Andrea Burgo
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Michel Cailleret
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatalisis, Sección de Estudios de Posgrado e Investigación de la Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, México D.F. 11340, México
| | - Martha Edith Macias Pérez
- Laboratorio de Biofísica y Biocatalisis, Sección de Estudios de Posgrado e Investigación de la Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, México D.F. 11340, México
| | - Daoudi Chabane-Sari
- Laboratoire des Produits Naturels, LAPRONA, Université Abou Bekr Belkaid, Tlemcen 13000, Algeria
| | - Patrick A Curmi
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France.
| |
Collapse
|
25
|
Nguyen BCQ, Taira N, Maruta H, Tawata S. Artepillin C and Other Herbal PAK1-blockers: Effects on Hair Cell Proliferation and Related PAK1-dependent Biological Function in Cell Culture. Phytother Res 2015; 30:120-7. [PMID: 26537230 DOI: 10.1002/ptr.5510] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/14/2015] [Accepted: 10/14/2015] [Indexed: 12/28/2022]
Abstract
PAK1 (RAC/CDC42-activated kinase 1) is the major oncogenic kinase, and a number of herbal PAK1-blockers such as propolis and curcumin have been shown to be anti-oncogenic and anti-melanogenic as well as anti-alopecia (promoting hair growth). Previously, we found several distinct PAK1-inhibitors in Okinawa plants including Alpinia zerumbet (alpinia). Thus, here, we tested the effects of these herbal compounds and their derivatives on the growth of cancer or normal hair cells, and melanogenesis in cell culture of A549 lung cancer, hair follicle dermal papilla cell, and B16F10 melanoma. Among these herbal PAK1-inhibitors, cucurbitacin I from bitter melon (Goya) turned out to be the most potent to inhibit the growth of human lung cancer cells with the IC50 around 140 nM and to promote the growth of hair cells with the effective dose around 10 nM. Hispidin, a metabolite of 5,6-dehydrokawain from alpinia, inhibited the growth of cancer cells with the IC50 of 25 μM as does artepillin C, the major anti-cancer ingredient in Brazilian green propolis. Mimosine tetrapeptides (MFWY, MFYY, and MFFY) and hispidin derivatives (H1-3) also exhibited a strong anti-cancer activity with the IC50 ranging from 16 to 30 μM. Mimosine tetrapeptides and hispidin derivatives strongly suppressed the melanogenesis in melanoma cells.
Collapse
Affiliation(s)
- Binh Cao Quan Nguyen
- Department of Bioscience and Biotechnology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-8580, Japan
| | - Nozomi Taira
- Department of Bioscience and Biotechnology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-8580, Japan
| | | | - Shinkichi Tawata
- Department of Bioscience and Biotechnology, Faculty of Agriculture, University of the Ryukyus, Senbaru 1, Nishihara-cho, Okinawa, 903-0213, Japan
| |
Collapse
|
26
|
Cai Y, Fang X, He C, Li P, Xiao F, Wang Y, Chen M. Cucurbitacins: A Systematic Review of the Phytochemistry and Anticancer Activity. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:1331-50. [PMID: 26503558 DOI: 10.1142/s0192415x15500755] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cucurbitacins are highly oxidized tetracyclic triterpenoids that are widely present in traditional Chinese medicines (Cucurbitaceae family), possess strong anticancer activity, and are divided into 12 classes from A to T with over 200 derivatives. The eight most active cucurbitacin components against cancer are cucurbitacin B, D, E, I, IIa, L glucoside, Q, and R. Their mechanisms of action include antiproliferation, inhibition of migration and invasion, proapoptosis, and cell cycle arrest promotion. Cucurbitacins are also found to be the inhibitors of JAK-STAT3, Wnt, PI3K/Akt, and MAPK signaling pathways, which play important roles in the apoptosis and survival of cancer cells. Recently, new studies have discovered synergistic anticancer effects by using cucurbitacins together with clinically approved chemotherapeutic drugs, such as docetaxel and methotrexate. This paper provides a summary of recent research progress on the anticancer property of cucurbitacins and the various intracellular signaling pathways involved in the regulation of cancer cell proliferation, death, invasion, and migration. Therefore, cucurbitacins are a class of promising anticancer drugs to be used alone or be intergraded in current chemotherapies and radiotherapies to treat many types of cancers.
Collapse
Affiliation(s)
- Yuee Cai
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Xiefan Fang
- † Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Chengwei He
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Peng Li
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Fei Xiao
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China.,‡ Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, P.R. China
| | - Yitao Wang
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Meiwan Chen
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| |
Collapse
|
27
|
Gutierrez-Uzquiza A, Lopez-Haber C, Jernigan DL, Fatatis A, Kazanietz MG. PKCε Is an Essential Mediator of Prostate Cancer Bone Metastasis. Mol Cancer Res 2015; 13:1336-46. [PMID: 26023164 DOI: 10.1158/1541-7786.mcr-15-0111] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/20/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED The bone is a preferred site for metastatic homing of prostate cancer cells. Once prostate cancer patients develop skeletal metastases, they eventually succumb to the disease; therefore, it is imperative to identify key molecular drivers of this process. This study examines the involvement of protein kinase C epsilon (PKCε), an oncogenic protein that is abnormally overexpressed in human tumor specimens and cell lines, on prostate cancer cell bone metastasis. PC3-ML cells, a highly invasive prostate cancer PC3 derivative with bone metastatic colonization properties, failed to induce skeletal metastatic foci upon inoculation into nude mice when PKCε expression was silenced using shRNA. Interestingly, while PKCε depletion had only marginal effects on the proliferative, adhesive, and migratory capacities of PC3-ML cells in vitro or in the growth of xenografts upon s.c. inoculation, it caused a significant reduction in cell invasiveness. Notably, PKCε was required for transendothelial cell migration (TEM) as well as for the growth of PC3-ML cells in a bone biomimetic environment. At a mechanistic level, PKCε depletion abrogates the expression of IL1β, a cytokine implicated in skeletal metastasis. Taken together, PKCε is a key factor for driving the formation of bone metastasis by prostate cancer cells and is a potential therapeutic target for advanced stages of the disease. IMPLICATIONS This study uncovers an important new function of PKCε in the dissemination of cancer cells to the bone; thus, highlighting the promising potential of this oncogenic kinase as a therapeutic target for skeletal metastasis.
Collapse
Affiliation(s)
- Alvaro Gutierrez-Uzquiza
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cynthia Lopez-Haber
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Danielle L Jernigan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Alessandro Fatatis
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania. Program in Biology of Prostate Cancer, Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
28
|
Nguyen BCQ, Be Tu PT, Tawata S, Maruta H. Combination of immunoprecipitation (IP)-ATP_Glo kinase assay and melanogenesis for the assessment of potent and safe PAK1-blockers in cell culture. Drug Discov Ther 2015; 9:289-95. [DOI: 10.5582/ddt.2015.01041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Binh Cao Quan Nguyen
- PAK Research Center
- Department of Biochemistry and Applied Bioscience, The United Graduate School of Agricultural Sciences, Kagoshima University
| | - Pham Thi Be Tu
- PAK Research Center
- Department of Biochemistry and Applied Bioscience, The United Graduate School of Agricultural Sciences, Kagoshima University
| | | | | |
Collapse
|
29
|
Zhu H, Chen X, Chen B, Chen B, Fan J, Song W, Xie Z, Jiang D, Li Q, Zhou M, Sun D, Zhao Y. Activating transcription factor 4 mediates a multidrug resistance phenotype of esophageal squamous cell carcinoma cells through transactivation of STAT3 expression. Cancer Lett 2014; 354:142-52. [PMID: 25130172 DOI: 10.1016/j.canlet.2014.07.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 07/31/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022]
Abstract
Multidrug resistance (MDR) is a major challenge to the clinical treatment of esophageal cancer. The stress response gene activating transcription factor 4 (ATF4) is involved in homeostasis and cellular protection. However, relatively little is known about the expression and function of ATF4 in esophageal squamous cell carcinoma (ESCC) MDR. In this study, we investigate the potential role and mechanisms of ATF4 in ESCC MDR. We demonstrated that overexpression of ATF4 promotes the MDR phenotype in ESCC cells, while depletion of ATF4 in the MDR ESCC cell line induces drug re-sensitization. We also demonstrated that ATF4 transactivates STAT3 expression by directly binding to the signal transducers and activators of transcription 3 (STAT3) promoter, resulting in MDR in ESCC cells. Significantly, inhibition of STAT3 by small interfering RNA (siRNA) or a selective inhibitor (JSI-124) reintroduces therapeutic sensitivity. In addition, increased Bcl-2, survivin, and MRP1 expression levels were observed in ATF4-overexpressing cells. In conclusion, ATF4 may promote MDR in ESCC cells through the up-regulation of STAT3 expression, and thus is an attractive therapeutic target to combat therapeutic resistance in ESCC.
Collapse
Affiliation(s)
- Hongwu Zhu
- Department of Gastroenterology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China
| | - Xiong Chen
- Department of Oncology, Fuzhou General Hospital of the Nanjing Military Command of the PLA, Fuzhou, China
| | - Bin Chen
- Department of Oncology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China
| | - Bei Chen
- Department of Oncology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China
| | - Jianyong Fan
- Department of Dermatology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China
| | - Weibing Song
- Department of Gerontology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China
| | - Ziying Xie
- Department of Gastroenterology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China
| | - Dan Jiang
- Department of Gastroenterology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China
| | - Qiuqiong Li
- Department of Gastroenterology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China
| | - Meihua Zhou
- Department of Gastroenterology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China
| | - Dayong Sun
- Department of Gastroenterology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China.
| | - Yagang Zhao
- Department of Gastroenterology, Guangzhou General Hospital of the Guangzhou Military Command of the People's Liberation Army (PLA), Guangzhou, China.
| |
Collapse
|
30
|
Wang H, Gutierrez-Uzquiza A, Garg R, Barrio-Real L, Abera MB, Lopez-Haber C, Rosemblit C, Lu H, Abba M, Kazanietz MG. Transcriptional regulation of oncogenic protein kinase Cϵ (PKCϵ) by STAT1 and Sp1 proteins. J Biol Chem 2014; 289:19823-19838. [PMID: 24825907 PMCID: PMC4094091 DOI: 10.1074/jbc.m114.548446] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 05/05/2014] [Indexed: 02/05/2023] Open
Abstract
Overexpression of PKCϵ, a kinase associated with tumor aggressiveness and widely implicated in malignant transformation and metastasis, is a hallmark of multiple cancers, including mammary, prostate, and lung cancer. To characterize the mechanisms that control PKCϵ expression and its up-regulation in cancer, we cloned an ∼ 1.6-kb promoter segment of the human PKCϵ gene (PRKCE) that displays elevated transcriptional activity in cancer cells. A comprehensive deletional analysis established two regions rich in Sp1 and STAT1 sites located between -777 and -105 bp (region A) and -921 and -796 bp (region B), respectively, as responsible for the high transcriptional activity observed in cancer cells. A more detailed mutagenesis analysis followed by EMSA and ChIP identified Sp1 sites in positions -668/-659 and -269/-247 as well as STAT1 sites in positions -880/-869 and -793/-782 as the elements responsible for elevated promoter activity in breast cancer cells relative to normal mammary epithelial cells. RNAi silencing of Sp1 and STAT1 in breast cancer cells reduced PKCϵ mRNA and protein expression, as well as PRKCE promoter activity. Moreover, a strong correlation was found between PKCϵ and phospho-Ser-727 (active) STAT1 levels in breast cancer cells. Our results may have significant implications for the development of approaches to target PKCϵ and its effectors in cancer therapeutics.
Collapse
Affiliation(s)
- HongBin Wang
- From the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Alvaro Gutierrez-Uzquiza
- From the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Rachana Garg
- From the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Laura Barrio-Real
- From the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Mahlet B Abera
- From the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Cynthia Lopez-Haber
- From the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Cinthia Rosemblit
- From the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Huaisheng Lu
- From the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Martin Abba
- the Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Universidad Nacional de La Plata, CP1900 La Plata, Argentina
| | - Marcelo G Kazanietz
- From the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| |
Collapse
|
31
|
Ren Y, Yu K, Sun S, Li Z, Yuan J, Han XD, Shi J, Zhen L. JSI124 inhibits breast cancer cell growth by suppressing the function of B cells via the downregulation of signal transducer and activator of transcription 3. Oncol Lett 2014; 8:928-932. [PMID: 25013518 PMCID: PMC4081387 DOI: 10.3892/ol.2014.2221] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 05/13/2014] [Indexed: 01/24/2023] Open
Abstract
JSI-124, also known as cucurbitacin I, is a selective inhibitor of Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3), and in vitro and in vivo studies have found that it has anti-tumor and anti-proliferative properties. However, the role of JSI124 in tumor-associated B cells has yet to be elucidated. The present study demonstrated that STAT3 is significantly activated in the B cells of patients with breast cancer. Furthermore, a 4T1 tumor-bearing mouse model revealed that JSI124 effectively inhibited tumor growth. Moreover, the STAT3 levels in the B cells of the JSI124-treated mice were found to be significantly decreased. B cells from normal Balb/c mice, the 4T1-bearing mice and the JSI124-treated 4T1 mice were purified and intravenously injected into the 4T1-bearing Balb/c mice. Tumor growth data showed that the 4T1 tumor mouse-derived B cells, which exhibited a higher level of STAT3, promoted tumor growth, while the JSI124-treated 4T1 mouse-derived B cells had a tumor suppressor function. Furthermore, the B cells from the normal Balb/c mice were treated with phosphate-buffered saline, JSI124 and 4T1 tumor cells, then the B cell STAT3 levels were analyzed. Following injection into the 4T1 mice, the 4T1 cell-treated B cells were observed to enhance tumor growth, while the JSI124-treated B cells were found to inhibit the growth of 4T1 tumors in vivo. These findings show a novel role of JSI124 in tumor suppression through the downregulation of the expression of STAT3 in tumor-associated B cells.
Collapse
Affiliation(s)
- Yi Ren
- Department of Breast and Thyroid Surgery, Huai'an First People's Hospital, Huai'an, Jiangsu 223001, P.R. China
| | - Kun Yu
- Department of Cardiology, Huai'an First People's Hospital, Huai'an, Jiangsu 223001, P.R. China
| | - Su'an Sun
- Department of Pathology, Huai'an First People's Hospital, Huai'an, Jiangsu 223001, P.R. China
| | - Zhi Li
- Department of Cardiology, Huai'an First People's Hospital, Huai'an, Jiangsu 223001, P.R. China
| | - Jin Yuan
- Department of Cardiology, Huai'an First People's Hospital, Huai'an, Jiangsu 223001, P.R. China
| | - Xue Dong Han
- Department of Breast and Thyroid Surgery, Huai'an First People's Hospital, Huai'an, Jiangsu 223001, P.R. China
| | - Jianhua Shi
- Department of Breast and Thyroid Surgery, Huai'an First People's Hospital, Huai'an, Jiangsu 223001, P.R. China
| | - Linlin Zhen
- Department of Breast and Thyroid Surgery, Huai'an First People's Hospital, Huai'an, Jiangsu 223001, P.R. China
| |
Collapse
|
32
|
Liu QR, Liu JM, Chen Y, Xie XQ, Xiong XX, Qiu XY, Pan F, Liu D, Yu SB, Chen XQ. Piperlongumine inhibits migration of glioblastoma cells via activation of ROS-dependent p38 and JNK signaling pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:653732. [PMID: 24967005 PMCID: PMC4055624 DOI: 10.1155/2014/653732] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/21/2014] [Accepted: 04/25/2014] [Indexed: 02/07/2023]
Abstract
Piperlongumine (PL) is recently found to kill cancer cells selectively and effectively via targeting reactive oxygen species (ROS) responses. To further explore the therapeutic effects of PL in cancers, we investigated the role and mechanisms of PL in cancer cell migration. PL effectively inhibited the migration of human glioma (LN229 or U87 MG) cells but not normal astrocytes in the scratch-wound culture model. PL did not alter EdU(+)-cells and cdc2, cdc25c, or cyclin D1 expression in our model. PL increased ROS (measured by DCFH-DA), reduced glutathione, activated p38 and JNK, increased IκBα, and suppressed NFκB in LN229 cells after scratching. All the biological effects of PL in scratched LN229 cells were completely abolished by the antioxidant N-acetyl-L-cysteine (NAC). Pharmacological administration of specific p38 (SB203580) or JNK (SP600125) inhibitors significantly reduced the inhibitory effects of PL on LN229 cell migration and NF κ B activity in scratch-wound and/or transwell models. PL prevented the deformation of migrated LN229 cells while NAC, SB203580, or SP600125 reversed PL-induced morphological changes of migrated cells. These results suggest potential therapeutic effects of PL in the treatment and prevention of highly malignant tumors such as glioblastoma multiforme (GBM) in the brain by suppressing tumor invasion and metastasis.
Collapse
Affiliation(s)
- Qian Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ju Mei Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yong Chen
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao Qiang Xie
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Xin Xiong
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Yao Qiu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feng Pan
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Urology, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Di Liu
- Department of Urology, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shang Bin Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao Qian Chen
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Neurological Diseases, Ministry of Education and Hubei Provincial Key Laboratory of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
33
|
Hobbs GA, Zhou B, Cox AD, Campbell SL. Rho GTPases, oxidation, and cell redox control. Small GTPases 2014; 5:e28579. [PMID: 24809833 DOI: 10.4161/sgtp.28579] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
While numerous studies support regulation of Ras GTPases by reactive oxygen and nitrogen species, the Rho subfamily has received considerably less attention. Over the last few years, increasing evidence is emerging that supports the redox sensitivity of Rho GTPases. Moreover, as Rho GTPases regulate the cellular redox state by controlling enzymes that generate and convert reactive oxygen and nitrogen species, redox feedback loops likely exist. Here, we provide an overview of cellular oxidants, Rho GTPases, and their inter-dependence.
Collapse
Affiliation(s)
- G Aaron Hobbs
- Department of Biochemistry and Biophysics; University of North Carolina; Chapel Hill, NC USA
| | - Bingying Zhou
- Department of Pharmacology; University of North Carolina; Chapel Hill, NC USA
| | - Adrienne D Cox
- Department of Pharmacology; University of North Carolina; Chapel Hill, NC USA; Department of Radiation Oncology, University of North Carolina; Chapel Hill, NC USA; Lineberger Comprehensive Cancer Center; University of North Carolina; Chapel Hill, NC USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics; University of North Carolina; Chapel Hill, NC USA; Lineberger Comprehensive Cancer Center; University of North Carolina; Chapel Hill, NC USA
| |
Collapse
|
34
|
Zhang YT, Xu LH, Lu Q, Liu KP, Liu PY, Ji F, Liu XM, Ouyang DY, He XH. VASP activation via the Gα13/RhoA/PKA pathway mediates cucurbitacin-B-induced actin aggregation and cofilin-actin rod formation. PLoS One 2014; 9:e93547. [PMID: 24691407 PMCID: PMC3972149 DOI: 10.1371/journal.pone.0093547] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 03/06/2014] [Indexed: 11/25/2022] Open
Abstract
Cucurbitacin B (CuB), a potent antineoplastic agent of cucurbitacin triterpenoids, induces rapid disruption of actin cytoskeleton and aberrant cell cycle inhibiting carcinogenesis. However, the underlying molecular mechanism of such anticancer effects remains incompletely understood. In this study, we showed that CuB treatment rapidly induced vasodilator-stimulated phosphoprotein (VASP) phosphorylation (i.e. activation) at the Ser157 residue and generated VASP clumps which were co-localized with amorphous actin aggregates prior to the formation of highly-ordered cofilin-actin rods in melanoma cells. Knockdown of VASP or inhibition of VASP activation using PKA-specific inhibitor H89 suppressed CuB-induced VASP activation, actin aggregation and cofilin-actin rod formation. The VASP activation was mediated by cAMP-independent PKA activation as CuB decreased the levels of cAMP while MDL12330A, an inhibitor of adenylyl cyclase, had weak effect on VASP activation. Knockdown of either Gα13 or RhoA not only suppressed VASP activation, but also ameliorated CuB-induced actin aggregation and abrogated cofilin-actin rod formation. Collectively, our studies highlighted that the CuB-induced actin aggregation and cofilin-actin rod formation was mediated via the Gα13/RhoA/PKA/VASP pathway.
Collapse
Affiliation(s)
- Yan-Ting Zhang
- Department of Immunobiology, Jinan University, Guangzhou, China
| | - Li-Hui Xu
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Qun Lu
- Department of Anatomy and Cell Biology, East Carolina University Brody School of Medicine, Greenville, North Carolina, United States of America
| | - Kun-Peng Liu
- Department of Immunobiology, Jinan University, Guangzhou, China
| | - Pei-Yan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Fang Ji
- Guangdong Entomological Institute, Guangzhou, China
| | - Xiao-Ming Liu
- Southern China Primate Research Center, Guangzhou, China
| | - Dong-Yun Ouyang
- Department of Immunobiology, Jinan University, Guangzhou, China
| | - Xian-Hui He
- Department of Immunobiology, Jinan University, Guangzhou, China
| |
Collapse
|
35
|
Lan T, Wang L, Xu Q, Liu W, Jin H, Mao W, Wang X, Wang X. Growth inhibitory effect of Cucurbitacin E on breast cancer cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:1799-1805. [PMID: 24040444 PMCID: PMC3759486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 08/03/2013] [Indexed: 06/02/2023]
Abstract
OBJECTIVE Due its inhibitory effects on chemical carcinogenesis and inflammation, Cucurbitacins have been proposed as an effective agent for the prevention or treatment of human cancers. In this study, we aimed to explore the effect of Cucurbitacin E (CuE) on human breast cancer cells. METHODS The inhibitory effect of CuE on proliferation of Bcap37 and MDA-MB-231 cells was assessed by MTT assay. The cell cycle distribution and cell apoptosis were determined by flow cytometry (FCM). The expression of pro-caspase 3, cleaved caspase 3, p21, p27 and the phosphorylation of signaling proteins was detected by Western Blotting. RESULTS CuE inhibited the growth of human breast cancer cells in a dose and time-dependent manner. FCM analysis showed that CuE induced G2/M phase arrest and cell apoptosis. CuE treatment promoted the cleavage of caspase 3 and upregulated p21 and p27. In addition, the phosphorylation of STAT3 but not ERK-1/2 was abrogated upon CuE treatment. Interestingly, losedose CuE significantly enhanced the growth inhibition induced by cisplatin. Conclusions Cucurbitacin E (CuE) could inhibit the growth of human breast cancer cells in vitro. CuE induced both apoptosis and cell cycle arrest probably through the inhibition of STAT3 function. Lose-dose CuE significantly enhanced the growth inhibitory effect of cisplatin on breast cancer cells, further indicating the potential clinical values of CuE for the prevention or treatment of human breast cancer.
Collapse
Affiliation(s)
- Tian Lan
- Department of Medical Oncology, the key laboratory of Integrated Chinese and Western Medical Oncology in Zhejiang Province, Zhejiang Cancer HospitalHangZhou, China 310022
- Department of Medical Oncology, Institute of Clinical Science, Sir Runrun Shaw Hospital, Medical College of Zhejiang UniversityHangzhou, China 310003
| | - Linling Wang
- Department of Medical Oncology, Institute of Clinical Science, Sir Runrun Shaw Hospital, Medical College of Zhejiang UniversityHangzhou, China 310003
| | - Qian Xu
- Department of Medical Oncology, the key laboratory of Integrated Chinese and Western Medical Oncology in Zhejiang Province, Zhejiang Cancer HospitalHangZhou, China 310022
| | - Weiguo Liu
- Department of Medical Oncology, the key laboratory of Integrated Chinese and Western Medical Oncology in Zhejiang Province, Zhejiang Cancer HospitalHangZhou, China 310022
| | - Hongchuan Jin
- Department of Medical Oncology, Institute of Clinical Science, Sir Runrun Shaw Hospital, Medical College of Zhejiang UniversityHangzhou, China 310003
| | - Weimin Mao
- Department of Medical Oncology, the key laboratory of Integrated Chinese and Western Medical Oncology in Zhejiang Province, Zhejiang Cancer HospitalHangZhou, China 310022
| | - Xian Wang
- Department of Medical Oncology, Institute of Clinical Science, Sir Runrun Shaw Hospital, Medical College of Zhejiang UniversityHangzhou, China 310003
| | - Xiaojia Wang
- Department of Medical Oncology, the key laboratory of Integrated Chinese and Western Medical Oncology in Zhejiang Province, Zhejiang Cancer HospitalHangZhou, China 310022
| |
Collapse
|
36
|
Gabrielsen M, Schuldt M, Munro J, Borucka D, Cameron J, Baugh M, Mleczak A, Lilla S, Morrice N, Olson MF. Cucurbitacin covalent bonding to cysteine thiols: the filamentous-actin severing protein Cofilin1 as an exemplary target. Cell Commun Signal 2013; 11:58. [PMID: 23945128 PMCID: PMC3751690 DOI: 10.1186/1478-811x-11-58] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/06/2013] [Indexed: 12/03/2022] Open
Abstract
Background Cucurbitacins are a class of triterpenoid natural compounds with potent bioactivities that led to their use as traditional remedies, and which continue to attract considerable attention as chemical biology tools and potential therapeutics. One obvious target is the actin-cytoskeleton; treatment with cucurbitacins results in cytoskeletal rearrangements that impact upon motility and cell morphology. Findings Cucurbitacin reacted with protein cysteine thiols as well as dithiothreitol, and we propose that the cucurbitacin mechanism of action is through broad protein thiol modifications that could result in inhibition of numerous protein targets. An example of such a target protein is Cofilin1, whose filamentous actin severing activity is inhibited by cucurbitacin conjugation. Conclusions The implications of these results are that cucurbitacins are unlikely to be improved for selectivity by medicinal chemistry and that their use as chemical biology probes to analyse the role of specific signalling pathways should be undertaken with caution.
Collapse
Affiliation(s)
- Mads Gabrielsen
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road Glasgow G61 1BD, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Tumorigenesis is the process by which normal cells evolve the capacity to evade and overcome the constraints usually placed upon their growth and survival. To ensure the integrity of organs and tissues, the balance of cell proliferation and cell death is tightly maintained. The proteins controlling this balance are either considered oncogenes, which promote tumorigenesis, or tumor suppressors, which prevent tumorigenesis. Phosphoinositide 3-kinase enhancer (PIKE) is a family of GTP-binding proteins that possess anti-apoptotic functions and play an important role in the central nervous system. Notably, accumulating evidence suggests that PIKE is a proto-oncogene involved in tumor progression. The PIKE gene (CENTG1) is amplified in a variety of human cancers, leading to the resistance against apoptosis and the enhancement of invasion. In this review, we will summarize the functions of PIKE proteins in tumorigenesis and discuss their potential implications in cancer therapy.
Collapse
|