1
|
Ren Y, Anderson AT, Meyer G, Lauber KM, Gallucci JC, Douglas Kinghorn A. Digoxin and its Na +/K +-ATPase-targeted actions on cardiovascular diseases and cancer. Bioorg Med Chem 2024; 114:117939. [PMID: 39396465 PMCID: PMC11527570 DOI: 10.1016/j.bmc.2024.117939] [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: 08/16/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Na+/K+-ATPase (NKA) is a plasma membrane ion-transporting protein involved in the generation and maintenance of Na+ and K+ gradients across the cell membrane, which can produce a driving force for the secondary transport of metabolic substrates. NKA also regulates intracellular calcium that is responsible for modulating numerous cellular processes, while it interacts with many other proteins and functions as a signal transducer, with several signaling pathways being involved. Thus, NKA has become an important target for the treatment of human diseases. Cardiac glycosides are well-known NKA inhibitors, of which (+)-digoxin or digoxin has been long used for the treatment of congestive heart failure. Also, digoxin has exhibited potential antitumor activity, by targeting directly HIF-1α, NKA, and NF-κB. Thus, the function of NKA in human cardiovascular diseases and cancer and the therapeutic effects of digoxin on these diseases are summarized in the present review, with the correlations among digoxin, NKA, cardiovascular diseases, and cancer being discussed. Presented herein are also the antitumor potential of monosaccharide cardiac glycoside analogues of digoxin, including (-)-cryptanoside A, (-)-oleandrin, (-)-ouabain, and (+)-strebloside. It is hoped that this contribution will provide some helpful information for the design and discovery of new cardiac glycoside-type therapeutic agents for the treatment of cardiovascular diseases and cancer.
Collapse
Affiliation(s)
- Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| | - Andrew T Anderson
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Gunnar Meyer
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Kaitlyn M Lauber
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Judith C Gallucci
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
2
|
Oliveira AGS, Rocha MA, de Azevedo LS, Coelho ATDM, Chagas RCR, Santos HB, Thomé RG, Samuel P, Wolfram E, Kim B, Reis RM, Ribeiro RIMA. Tapirira guianensis is Selectively Cytotoxic, Induces Apoptosis to the Glioblastoma and Decreases Tumor Growth and Angiogenesis in vivo. PLANTA MEDICA 2024; 90:13-24. [PMID: 37832581 DOI: 10.1055/a-2181-2569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Glioblastoma is the most frequent primary malignant brain tumor without effective treatment, which makes this work extremely relevant. The study of the bioactive compounds from medicinal plants plays an important role in the discovery of new drugs.This research investigated the constituents of Tapirira guianensis and its antitumor potential (in vitro and in vivo) in glioblastoma. The T. guianensis extracts were characterized by mass spectrometry. The ethyl acetate partition (01ID) and its fractions 01ID-F2 and 01ID-F4 from T. guianensis showed potential antitumor treatment evidenced by selective cytotoxicity for GAMG with IC50 14.1 µg/mL, 83.07 µg/mL, 59.27 µg/mL and U251 with IC50 25.92 µg/mL, 37.3 µg/mL and 18.84 µg/mL. Fractions 01ID-F2 and 01ID-F4 were 10 times more selective when compared to TMZ and 01ID for the two evaluated cell lines. T. guianensis also reduced matrix metalloproteinases 2 - 01ID-F2 (21.84%), 01ID-F4 (29.6%) and 9 - 01ID-F4 (73.42%), ID-F4 (53.84%) activities, and induced apoptosis mainly through the extrinsic pathway. Furthermore, all treatments significantly reduced tumor size (01ID p < 0,01, 01ID-F2 p < 0,01 and 01ID-F4 p < 0,0001) and caused blood vessels to shrink in vivo. The present findings highlight that T. guianensis exhibits considerable antitumor potential in preclinical studies of glioblastoma. This ability may be related to the phenolic compounds and sesquiterpene derivatives identified in the extracts. This study deserves further in vivo research, followed by clinical investigation.
Collapse
Affiliation(s)
- Ana Gabriela Silva Oliveira
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Marina Andrade Rocha
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Lucas Santos de Azevedo
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | | | - Rafael César Russo Chagas
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Hélio Batista Santos
- Tissue Processing Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Ralph Gruppi Thomé
- Tissue Processing Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Peter Samuel
- Zurich University of Applied Sciences, Department of Life Sciences and Facility Management, Wädenswil, Switzerland
| | - Evelyn Wolfram
- Zurich University of Applied Sciences, Department of Life Sciences and Facility Management, Wädenswil, Switzerland
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Portugal
| | | |
Collapse
|
3
|
de Oliveira GC, Rocha SC, da Silva Lopes MA, Paixão N, Alves SLG, Pessoa MTC, Noël F, Quintas LEM, Barbosa LA, Villar JAFP, Cortes VF. Implications of Synthetic Modifications of the Cardiotonic Steroid Lactone Ring on Cytotoxicity. J Membr Biol 2021; 254:487-497. [PMID: 34128090 DOI: 10.1007/s00232-021-00186-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/08/2021] [Indexed: 01/24/2023]
Abstract
Na,K-ATPase (NKA) and cardiotonic steroids (CTS) have shown potent cytotoxic and anticancer effects. Here, we have synthesized a series of CTS digoxin derivatives (γ-benzylidene) with substitutions in the lactone ring and evaluated the cytotoxicity caused by digoxin derivatives in tumor and non-tumor cells lines, as well as their effects on NKA. The cytotoxicity assay was determined in HeLa, A549, and WI-26 VA4 after they were treated for 48 h with increased concentrations of CTS. The effects of CTS on NKA activity and immunoblotting of α1 and β1 isoforms were evaluated at IC50 concentrations in A549 cell membrane. NKA activity from mouse brain cortex was also measured. The majority of CTS exhibited low cytotoxicity in tumor and non-tumor cells, presenting IC50 values at micromolar concentrations, while digoxin showed cytotoxicity at nanomolar concentrations. BD-15 presented the lowest IC50 value (8 µM) in A549 and reduced its NKA activity in 28%. In contrast, BD-7 was the compound that most inhibited NKA (56% inhibition) and presented high IC50 value for A549. In mouse cortex, only BD-15 modulated the enzyme activity in a concentration-dependent inhibition curve. These results demonstrate that the cytotoxicity of these compounds is not related to NKA inhibition. The substitutions in the lactone ring of digoxin led to an increase in the cytotoxic concentration in tumor cells, which may not be interesting for cancer, but it has the advantage of increasing the therapeutic margin of these molecules when compared to classic CTS, and can be used safely in research for other diseases.
Collapse
Affiliation(s)
- Gisele Capanema de Oliveira
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Bairro Chanadour, Divinópolis, MG, 35501-296, Brazil
| | - Sayonarah Carvalho Rocha
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Bairro Chanadour, Divinópolis, MG, 35501-296, Brazil
| | - Miliane Alves da Silva Lopes
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Av Carlos Chagas, 373, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Natasha Paixão
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Av Carlos Chagas, 373, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Silmara Lúcia Grego Alves
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del Rei, Campus Centro Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Bairro Chanadour, Divinópolis, MG, 35501-296, Brazil
| | - Marco Túlio Corrêa Pessoa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Bairro Chanadour, Divinópolis, MG, 35501-296, Brazil.,Marshall Institute for Interdisciplinary Research, Huntington, WV, USA
| | - François Noël
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Av Carlos Chagas, 373, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Luis Eduardo M Quintas
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Av Carlos Chagas, 373, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Leandro Augusto Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Bairro Chanadour, Divinópolis, MG, 35501-296, Brazil
| | - José Augusto Ferreira Perez Villar
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del Rei, Campus Centro Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Bairro Chanadour, Divinópolis, MG, 35501-296, Brazil.
| | - Vanessa Faria Cortes
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro Oeste Dona Lindu, Av Sebastião Gonçalves Coelho, 400, Bairro Chanadour, Divinópolis, MG, 35501-296, Brazil.
| |
Collapse
|
4
|
Honda T, Motoyoshi K, Kasahara J, Yamagata K, Takahashi H, Nakamura H, Murayama T. Tyrosine-phosphorylation and activation of glucosylceramide synthase by v-Src: Its role in survival of HeLa cells against ceramide. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1866:158817. [PMID: 32980536 DOI: 10.1016/j.bbalip.2020.158817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 10/23/2022]
Abstract
Sphingolipids represent a family of cellular lipid-molecules that regulate physiological and pathophysiological processes. Glucosylceramide (GlcCer), the simplest glycosphingolipid (GSL), is synthesized from ceramide and UDP-glucose by GlcCer synthase (GCS). Both GlcCer (and resulting GSLs) and ceramide regulate various cellular functions including cell death and multiple drug resistance. Src family tyrosine kinases are up-regulated in various human cancer cells. We examined the effect of v-Src expression on GCS activity, the formation of 4-nitrobenzo-2-oxa-1,3-diazole (NBD)-labeled GlcCer from NBD-ceramide, and the effect of tyrosine132 mutation in GCS on ceramide-induced cytotoxicity in HeLa cells. Expression of v-Src increased the formation of NBD-GlcCer in both intact cells without marked changes in other sphingolipid metabolites and cell homogenates without changing affinities of NBD-ceramide and UDP-glucose. Expression of v-Src also increased tyrosine-phosphorylated levels in GCS proteins in HeLa and HEK293T cells. In HEK293T cells transiently expressing the GCS mutant, GCS-Y132F-HA, showing replacement of the tyrosine132 residue with phenylalanine, tyrosine-phosphorylated levels in GCS proteins were significantly lower than those in control cells expressing the GCS-wild-type-HA. The formation of NBD-GlcCer in HeLa cells stably expressing GCS-Y132F-HA was significantly lower than that in the control. Ceramide-induced cytotoxicity in HeLa-GCS-Y132F-HA cells was significantly greater than in the control. In this study, we showed for the first time that expression of v-Src up-regulated GCS activity via tyrosine phosphorylation of the enzyme in a post-translational manner. Mechanisms of Src-induced resistance to ceramide-induced cytotoxicity are discussed in relation to the Src-induced up-regulation of GCS activity.
Collapse
Affiliation(s)
- Takuya Honda
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan.
| | - Kaisei Motoyoshi
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Junya Kasahara
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Kazuyuki Yamagata
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; Laboratory of International Scholars in Pharmaceuticals/Systems Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Hiromasa Takahashi
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| |
Collapse
|
5
|
Implications for glycosylated compounds and their anti-cancer effects. Int J Biol Macromol 2020; 163:1323-1332. [PMID: 32622770 DOI: 10.1016/j.ijbiomac.2020.06.281] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 06/20/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
Glycosylated compounds are major secondary metabolites of plants, which have various therapeutic effects on human diseases, by acting as anti-cancer, antioxidant, and anti-inflammatory agents. Glycosylation increases stability, bioactivity, and solubility of compounds and improves their pharmacological properties. Two well-known examples of glycosylated compounds include cardiac and flavonoid, the anti-tumor activities of which have been emphasized by several studies. However, little is known about their role in the treatment or prevention of cancer. In this review, recent studies on anti-tumor properties of cardiac and flavonoid glycosides, and their mechanisms of action, have been investigated. More specifically, this review is aimed at focusing on the multifactorial properties of cardiac and flavonoid compounds as well as their correlation with signaling pathways in the treatment of cancer.
Collapse
|
6
|
Ren Y, Ribas HT, Heath K, Wu S, Ren J, Shriwas P, Chen X, Johnson ME, Cheng X, Burdette JE, Kinghorn AD. Na +/K +-ATPase-Targeted Cytotoxicity of (+)-Digoxin and Several Semisynthetic Derivatives. JOURNAL OF NATURAL PRODUCTS 2020; 83:638-648. [PMID: 32096998 PMCID: PMC7243443 DOI: 10.1021/acs.jnatprod.9b01060] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
(+)-Digoxin (1) is a well-known cardiac glycoside long used to treat congestive heart failure and found more recently to show anticancer activity. Several known cardenolides (2-5) and two new analogues, (+)-8(9)-β-anhydrodigoxigenin (6) and (+)-17-epi-20,22-dihydro-21α-hydroxydigoxin (7), were synthesized from 1 and evaluated for their cytotoxicity toward a small panel of human cancer cell lines. A preliminary structure-activity relationship investigation conducted indicated that the C-12 and C-14 hydroxy groups and the C-17 unsaturated lactone unit are important for 1 to mediate its cytotoxicity toward human cancer cells, but the C-3 glycosyl residue seems to be less critical for such an effect. Molecular docking profiles showed that the cytotoxic 1 and the noncytotoxic derivative 7 bind differentially to Na+/K+-ATPase. The HO-12β, HO-14β, and HO-3'aα hydroxy groups of (+)-digoxin (1) may form hydrogen bonds with the side-chains of Asp121 and Asn122, Thr797, and Arg880 of Na+/K+-ATPase, respectively, but the altered lactone unit of 7 results in a rotation of its steroid core, which depotentiates the binding between this compound and Na+/K+-ATPase. Thus, 1 was found to inhibit Na+/K+-ATPase, but 7 did not. In addition, the cytotoxic 1 did not affect glucose uptake in human cancer cells, indicating that this cardiac glycoside mediates its cytotoxicity by targeting Na+/K+-ATPase but not by interacting with glucose transporters.
Collapse
Affiliation(s)
- Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Hennrique T. Ribas
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Kimberly Heath
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Sijin Wu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Jinhong Ren
- Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Pratik Shriwas
- Department of Biological Sciences, Edison Biotechnology Institute, and Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, United States
| | - Xiaozhuo Chen
- Department of Biological Sciences, Edison Biotechnology Institute, Molecular and Cellular Biology Program, and Department of Biomedical Sciences, Ohio University, Athens, OH 45701, United States
| | - Michael E. Johnson
- Department of Pharmaceutical Sciences and Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Xiaolin Cheng
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Joanna E. Burdette
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, United States
| | | |
Collapse
|
7
|
Garcia IJP, de Oliveira GC, de Moura Valadares JM, Banfi FF, Andrade SN, Freitas TR, Dos Santos Monção Filho E, Lima Santos HD, Júnior GMV, Chaves MH, de Jesus Rodrigues D, Sanchez BAM, Varotti FP, Barbosa LA. New bufadienolides extracted from Rhinella marina inhibit Na,K-ATPase and induce apoptosis by activating caspases 3 and 9 in human breast and ovarian cancer cells. Steroids 2019; 152:108490. [PMID: 31499071 DOI: 10.1016/j.steroids.2019.108490] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/24/2022]
Abstract
Bufadienolide compounds have been used for growth inhibition and apoptosis induction in tumor cells. Those families of cardiotonic steroids can bind the Na,K-ATPase, causing its inhibition. The use of bufadienolides is widely described in the literature as an anticancer function. The aim of this study was to evaluate the effects of bufadienolides and alkaloid isolated from venom samples from R. marina on tumor cells. We performed cytotoxicity assay in MDA-MB-231 and TOV-21G cells and evaluated the activity of Caspases (3 and 9), Na, K-ATPase, PMCA and SERCA. Four compounds were extrated from the venom of R. marina. The compound 1 showed higher cytotoxicity in MDA-MB-231cells. Compound 1 also showed activation of Caspase 3 and 9. This compound caused an inhibition of the activity and expression of Na, K-ATPase, and also showed activation of both caspase-9 and caspase-3 in MDA-MB-231 cells. We also observed that Compound 1 had a direct effect on some ATPases, such as Na, K-ATPase, PMCA and SERCA. Compound 1 was able to inhibit the activity of the purified Na, K-ATPase enzyme from the concentration of 5 µM. It also caused inhibition of PMCA at all concentrations tested (1 nM-30 µM). However, the compound 1 led to an increase of the activity of purified SERCA between the concentrations of 7.5-30 µM. Thus, we present a Na, K-ATPase and PMCA inhibitor, which may lead to the activation of caspases 3 and 9, causing the cells to enter into apoptosis. Our study suggests that compound 1 may be an interesting molecule as an anticancer agent.
Collapse
Affiliation(s)
- Israel José Pereira Garcia
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste, Divinópolis, MG, Brazil; Laboratório de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste, Divinópolis, MG, Brazil.
| | - Gisele Capanema de Oliveira
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste, Divinópolis, MG, Brazil
| | | | - Felipe Finger Banfi
- Universidade Federal de Mato Grosso, Instituto de Ciências da Saúde, Sinop, MT, Brazil
| | - Silmara Nunes Andrade
- Núcleo de Pesquisa em Química Biológica, Universidade Federal de São João Del-Rei, Campus Centro Oeste, Divinópolis, MG, Brazil
| | - Túlio Resende Freitas
- Núcleo de Pesquisa em Química Biológica, Universidade Federal de São João Del-Rei, Campus Centro Oeste, Divinópolis, MG, Brazil
| | | | - Hérica de Lima Santos
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste, Divinópolis, MG, Brazil; Laboratório de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste, Divinópolis, MG, Brazil
| | | | | | | | | | - Fernando P Varotti
- Núcleo de Pesquisa em Química Biológica, Universidade Federal de São João Del-Rei, Campus Centro Oeste, Divinópolis, MG, Brazil
| | - Leandro Augusto Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste, Divinópolis, MG, Brazil; Laboratório de Membranas e ATPases, Universidade Federal de São João del Rei, Campus Centro-Oeste, Divinópolis, MG, Brazil.
| |
Collapse
|
8
|
Silva AG, Lopes CFB, Carvalho Júnior CG, Thomé RG, dos Santos HB, Reis R, Ribeiro RIMDA. WIN55,212-2 induces caspase-independent apoptosis on human glioblastoma cells by regulating HSP70, p53 and Cathepsin D. Toxicol In Vitro 2019; 57:233-243. [DOI: 10.1016/j.tiv.2019.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/02/2019] [Accepted: 02/08/2019] [Indexed: 11/28/2022]
|
9
|
Zhao L, Fu L, Xu Z, Fan R, Xu R, Fu R, Zou S, Wang C, Zhang Y, Wang J, Bao J, Wang Z, Hou X, Zheng Y, Dai E, Wang F. The anticancer effects of cinobufagin on hepatocellular carcinoma Huh‑7 cells are associated with activation of the p73 signaling pathway. Mol Med Rep 2019; 19:4119-4128. [PMID: 30942456 PMCID: PMC6471725 DOI: 10.3892/mmr.2019.10108] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 02/14/2019] [Indexed: 12/18/2022] Open
Abstract
The Na+/K+-ATPase inhibitor cinobufagin exhibits numerous anticancer effects on hepatocellular carcinoma (HCC) cells expressing wild-type p53 via inhibition of aurora kinase A (AURKA) and activation of p53 signaling. However, the effects of cinobufagin on HCC cells expressing mutant p53 remain unclear. In the present study, the anticancer effects of cinobufagin were investigated on HCC Huh-7 cells with mutant p53, and the effects of AURKA overexpression or inhibition on the anticancer effects of cinobufagin were analyzed. Viability, cell cycle progression and apoptosis of cells were determined using an MTT assay, flow cytometry and Hoechst 33342 staining, respectively. The expression levels of p53 and p73 signaling-associated proteins were investigated via western blot analysis. The results demonstrated that the expression levels of AURKA, B-cell lymphoma 2 (Bcl-2), cyclin-dependent kinase 1, cyclin B1, proliferating cell nuclear antigen and heterogeneous nuclear ribonucleoprotein K, as well as the phosphorylation of p53 and mouse double minute 2 homolog, were significantly decreased in Huh-7 cells treated with 5 µmol/l cinobufagin for 24 h. Conversely, the expression levels of Bcl-2-associated X protein, p21, p53 upregulated modulator of apoptosis and phorbol-12-myristate-13-acetate-induced protein 1, were significantly increased by cinobufagin treatment. Overexpression or inhibition of AURKA suppressed or promoted the anticancer effects of cinobufagin on Huh-7 cells, respectively. These results indicated that cinobufagin may induce anticancer effects on Huh-7 cells via the inhibition of AURKA and p53 signaling, and via the activation of p73 signaling, in an AURKA-dependent manner.
Collapse
Affiliation(s)
- Lei Zhao
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Lina Fu
- Department of Gastroenterology, Tianjin Fourth Central Hospital, Tianjin 300140, P.R. China
| | - Zhongwei Xu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Rong Fan
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Ruicheng Xu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Rong Fu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Shuang Zou
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Congcong Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Yan Zhang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Jiabao Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Jun Bao
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Zhimei Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Xiaojie Hou
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin 300309, P.R. China
| | - Yupiao Zheng
- Department of Gastroenterology and Hepatology, The Third Central Hospital of Tianjin, Tianjin 300170, P.R. China
| | - Erqing Dai
- Hepatology Department of Pingjin Hospital, Logistics University of Chinese People's Armed Police Forces, Tianjin 300162, P.R. China
| | - Fengmei Wang
- Department of Gastroenterology and Hepatology, The Third Central Hospital of Tianjin, Tianjin 300170, P.R. China
| |
Collapse
|
10
|
21‑Benzylidene digoxin, a novel digoxin hemi-synthetic derivative, presents an anti-inflammatory activity through inhibition of edema, tumour necrosis factor alpha production, inducible nitric oxide synthase expression and leucocyte migration. Int Immunopharmacol 2018; 65:174-181. [DOI: 10.1016/j.intimp.2018.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/10/2018] [Accepted: 10/06/2018] [Indexed: 02/03/2023]
|
11
|
Zhang WY, Liu YJ, He Y, Chen P. Suppression of long noncoding RNA NCK1-AS1 increases chemosensitivity to cisplatin in cervical cancer. J Cell Physiol 2018; 234:4302-4313. [PMID: 30221354 DOI: 10.1002/jcp.27198] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/17/2018] [Indexed: 12/30/2022]
Abstract
Cervical cancer remains a serious health problem till now, with nearly 500,000 women cases diagnosed each year around the world. Long noncoding RNA (lncRNA) is a novel class of RNA transcripts (>200 nucleotides in length) participating in gene transcription, cell proliferation, differentiation, and drug resistance. This study aimed to explore the regulatory relationship among lncRNA NCK1-AS1, miR-134-5p, and MutS protein homolog 2 (MSH2), so that the resistance against cisplatin in cervical cancer treatment could be better understood. Comprehensive lncRNA profiling analysis was performed to screen lncRNAs differentially expressed in cervical cancer. The expression patterns of miR-134-5p, NCK1-AS1, and MSH2 were evaluated in cancerous tissues and adjacent normal tissues obtained from 75 cervical cancer patients. Subsequently, anti-NCK1-AS1 small interfering RNA, miR-134-5p mimics, and miR-134-5p inhibitors were transfected into cervical cancer cells, and the effects of these transcripts on cisplatin resistance and cell apoptosis were investigated. The regulatory relationship among NCK1-AS1, miR-134-5p, and MSH2 was identified using a dual-luciferase reporter gene assay, and the results were further validated by RNA pull-down and RNA immunoprecipitation assays. Based on the microarray data of GSE63514 and GSE27678, NCK1-AS1 was upregulated in cervical cancer. Increased expression of NCK1-AS1, MSH2, and decreased expression of miR-134-5p were observed in cervical cancer tissues. In addition, NCK1-AS1 competitively bound to miR-134-5p to regulate MSH2. Therefore, si-NCK1-AS1 and miR-134-5p mimic both reduced MSH2 activity and increased cisplatin-induced apoptosis in cervical cancer cells. Taken together, NCK1-AS1 may become a novel target in improving the chemotherapeutic response and survival of cervical cancer patients.
Collapse
Affiliation(s)
- Wei-Yi Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yin-Jiao Liu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yan He
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Ping Chen
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|