1
|
Qaed E, Liu W, Almoiliqy M, Mohamed R, Tang Z. Unleashing the potential of Genistein and its derivatives as effective therapeutic agents for breast cancer treatment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03579-6. [PMID: 39549063 DOI: 10.1007/s00210-024-03579-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024]
Abstract
Breast cancer remains one of the leading causes of cancer-related deaths among women worldwide. Genistein (Gen), a phytoestrogen soy isoflavone, has emerged as a promising agent in the prevention and treatment of breast cancer due to its ability to function as a natural selective estrogen receptor modulator (SERM). This review explores the multifaceted mechanisms through which Gen and its derivatives exert their anticancer effects, including modulation of the PI3K/Akt signaling pathway, regulation of apoptosis, inhibition of angiogenesis, and impacts on DNA methylation and enzyme functions. We discuss the dual roles of Gen in both enhancing and inhibiting estrogen receptor (ER)-dependent pathways., highlighting its complex interactions with ERα and ERβ. Furthermore, the review examines the synergistic effect of combining Gen with conventional chemotherapeutic agents such as doxorubicin, cisplatin, and selenium, as well as other natural compounds like lycopene. Clinical studies suggest that while isoflavones may not significantly influence breast cancer progression in general, the high consumption of soy isoflavones is associated with reduced recurrence rates in breast cancer survivors. Importantly, Gen's ability to modulate key signaling pathways and enhance the efficacy of existing treatments improves its potential as a valuable adjunct in breast cancer therapy. In conclusion, Gen and its derivatives offer a novel and promising approach for treatment of breast cancer. Continued research into their mechanisms of action and clinical applications will be essential in optimizing their therapeutic potential and translating these findings into effective clinical interventions.
Collapse
Affiliation(s)
- Eskandar Qaed
- Collage of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun Dalian, Dalian, 116044, China.
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, P. R. China.
| | - Wu Liu
- Collage of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun Dalian, Dalian, 116044, China
| | - Marwan Almoiliqy
- Collage of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun Dalian, Dalian, 116044, China
| | - Rawan Mohamed
- College of Clinical Pharmacy, Mansoura University, Mansoura, Egypt
| | - Zeyao Tang
- Collage of Pharmacy, Department of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun Dalian, Dalian, 116044, China.
| |
Collapse
|
2
|
Bakhsh MR, Rouhi L, Ghaedi K, Hashemi M, Peymani M, Samarghandian S. Therapeutic effects of guanidine hydrochloride on breast cancer through targeting KCNG1 gene. Biomed Pharmacother 2023; 164:114982. [PMID: 37311278 DOI: 10.1016/j.biopha.2023.114982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/28/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is one of the subtypes of breast cancer (BC) that is associated with poor survival rates and failure to respond to hormonal and targeted therapies. OBJECTIVE The aim of this study was to identify a specific gene at the expression level for TNBC and targeting of this type of breast cancer based on it. Using TCGA database, genes that are particularly high expression in TNBC subtypes compared to other BC subtypes (in terms of receptor status) and normal samples were identified and their sensitivity and specificity were evaluated. Using PharmacoGX and Drug Bank data, drug sensitivity and drug-appropriate genes were identified, respectively. The effects of the identified drug on triple-negative cell lines (MDA-MB-468) were evaluated in comparison with the cell line of other subtypes (MCF7) by apoptosis and MTS tests. RESULTS Data analyzes showed that the expression level of KCNG1 gene in the TNBC subgroup was significantly higher compared to other BC subtypes from the KCN gene family and ROC results showed that this gene had highest sensitivity and specificity in TNBC subtype. The results of drug resistance and sensitivity showed that an increase in the expression level of KCNG1 was associated with sensitivity to Cisplatin and Oxaliplatin. Moreover, Drug Bank results showed that Guanidine hydrochloride (GuHCl) was a suitable inhibitor for KCNG1. In vitro results showed that the expression level of KCNG1 was higher in MDA-MB-468 compared to MCF7. In addition, the rate of apoptosis in response to GuHCl treatment in MDA-MB-468 cell line as TNBC cell model was higher than MCF7 in the same concentration. CONCLUSION This study revealed that GuHCl could be a suitable treatment for TNBC subtype by targeting of KCNG1.
Collapse
Affiliation(s)
- Mehdi Roshanian Bakhsh
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, the Islamic Republic of Iran
| | - Leila Rouhi
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, the Islamic Republic of Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, the Islamic Republic of Iran; Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, the Islamic Republic of Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran.
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, the Islamic Republic of Iran.
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, the Islamic Republic of Iran.
| |
Collapse
|
3
|
Zou Y, Xie J, Tian W, Wu L, Xie Y, Huang S, Tang Y, Deng X, Wu H, Xie X. Integrative Analysis of KCNK Genes and Establishment of a Specific Prognostic Signature for Breast Cancer. Front Cell Dev Biol 2022; 10:839986. [PMID: 35656548 PMCID: PMC9152175 DOI: 10.3389/fcell.2022.839986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/29/2022] [Indexed: 12/19/2022] Open
Abstract
Two-pore domains potassium channel subunits, encoded by KCNK genes, play vital roles in breast cancer progression. However, the characteristics of most KCNK genes in breast cancer has yet to be clarified. In this study, we comprehensively analyzed the expression, alteration, prognosis, and biological functions of various KCNKs in breast cancer. The expression of KCNK1/4/6/9/10/13 were significantly upregulated, while KCNK2/3/5/7/17 were downregulated in breast cancer tissues compared to normal mammary tissues. Increased expression of KCNK1/3/4/9 was correlated with poor overall survival, while high expression of KCNK2/7/17 predicted better overall survival in breast cancer. Eight KCNK genes were altered in breast cancer patients with a genomic mutation rate ranged from 1.9% to 21%. KCNK1 and KCNK9 were the two most common mutations in breast cancer, occurred in 21% and 18% patients, respectively. Alteration of KCNK genes was associated with the worse clinical characteristics and higher TMB, MSI, and hypoxia score. Using machine learning method, a specific prognostic signature with seven KCNK genes was established, which manifested accuracy in predicting the prognosis of breast cancer in both training and validation cohorts. A nomogram with great predictive performance was afterwards constructed through incorporating KCNK-based risk score with clinical features. Furthermore, KCNKs were correlated with the activation of several tumor microenvironment cells, including T cells, mast cells, macrophages, and platelets. Presentation of antigen, stimulation of G protein signaling and toll-like receptor cascaded were regulated by KCNKs family. Taken together, KCNKs may regulate breast cancer progression via modulating immune response which can serve as ideal prognostic biomarkers for breast cancer patients. Our study provides novel insight for future studies evaluating their usefulness as therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hao Wu
- *Correspondence: Hao Wu, ; Xinhua Xie,
| | | |
Collapse
|
4
|
Romero AH, Sojo F, Arvelo F, Calderón C, Morales A, López SE. Anticancer potential of new 3-nitroaryl-6-(N-methyl)piperazin-1,2,4-triazolo[3,4-a]phthalazines targeting voltage-gated K + channel: Copper-catalyzed one-pot synthesis from 4-chloro-1-phthalazinyl-arylhydrazones. Bioorg Chem 2020; 101:104031. [PMID: 32629281 DOI: 10.1016/j.bioorg.2020.104031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 11/28/2022]
Abstract
A series of six 3-aryl-6-(N-methylpiperazin)-1,2,4-triazolo[3,4-a]phthalazines were prepared through a facile and efficient one-pot copper-catalyzed procedure from 4-chloro-1-phthalazinyl-arylhydrazones with relatively good yields (62-83%). The one-pot copper-catalytic procedure consists of two simultaneous reactions: (i) a direct intramolecular dehydrogentaive cyclization between ylidenic carbon and adjacent pyrazine nitrogen to form 1,2,4-triazolo ring and, (ii) a direct N-amination on carbon-chlorine bond. Then, an in vitro anticancer evaluation was performed for the synthesized compounds against five selected human cancer cells (A549, MCF-7, SKBr3, PC-3 and HeLa). The nitro-derivatives were significantly more active against cancer strains than against the rest of tested compounds. Specifically, compound 8d was identified as the most promising anticancer agent with significant biological responses and low relative toxicities on human dermis fibroblast. The cytotoxic effect of compound 8d was more significant on PC3, MCF-7 and SKBr3 cancer cells with low-micromolar IC50 value ranging from 0.11 to 0.59 μM, superior to Adriamycin drug. Mechanistic experimental and theoretical studies demonstrated that compounds 8d act as a K+ channel inhibitor in cancer models. Further molecular docking studies suggest that the EGFR Tyrosine Kinase enzyme may be a potential target for the most active 3-aryl-6-(N-methylpiperazin)-1,2,4-triazolo[3,4-a]phthalazines.
Collapse
Affiliation(s)
- Angel H Romero
- Cátedra de Química General, Facultad de Farmacia, Universidad Central de Venezuela, Los Chaguaramos, Caracas 1041-A, Venezuela.
| | - Felipe Sojo
- Fundación Institutos de Estudios Avanzados-IDEA, Área Salud, Venezuela; Laboratorio de Cultivo de Tejidos y Biología de Tumores, Instituto de Biología Experimental-IBE, Facultad de Ciencias-UCV, Bello Monte, Caracas, Venezuela
| | - Francisco Arvelo
- Fundación Institutos de Estudios Avanzados-IDEA, Área Salud, Venezuela; Laboratorio de Cultivo de Tejidos y Biología de Tumores, Instituto de Biología Experimental-IBE, Facultad de Ciencias-UCV, Bello Monte, Caracas, Venezuela
| | - Christian Calderón
- Laboratorio de Fisiología y Biofísica, Centro de Biología Celular, Instituto de Biología Experimental-IBE, Facultad de Ciencias, UCV, Bello Monte, Caracas, Venezuela
| | - Alvaro Morales
- Laboratorio de Biotecnología Clínica Santa María, Cevalfes, Valencia, Venezuela
| | - Simón E López
- Department of Chemistry, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
5
|
Boldrini-França J, Pinheiro-Junior EL, Peigneur S, Pucca MB, Cerni FA, Borges RJ, Costa TR, Carone SEI, Fontes MRDM, Sampaio SV, Arantes EC, Tytgat J. Beyond hemostasis: a snake venom serine protease with potassium channel blocking and potential antitumor activities. Sci Rep 2020; 10:4476. [PMID: 32161292 PMCID: PMC7066243 DOI: 10.1038/s41598-020-61258-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/19/2020] [Indexed: 02/07/2023] Open
Abstract
Snake venom serine proteases (SVSPs) are complex and multifunctional enzymes, acting primarily on hemostasis. In this work, we report the hitherto unknown inhibitory effect of a SVSP, named collinein-1, isolated from the venom of Crotalus durissus collilineatus, on a cancer-relevant voltage-gated potassium channel (hEAG1). Among 12 voltage-gated ion channels tested, collinein-1 selectively inhibited hEAG1 currents, with a mechanism independent of its enzymatic activity. Corroboratively, we demonstrated that collinein-1 reduced the viability of human breast cancer cell line MCF7 (high expression of hEAG1), but does not affect the liver carcinoma and the non-tumorigenic epithelial breast cell lines (HepG2 and MCF10A, respectively), which present low expression of hEAG1. In order to obtain both functional and structural validation of this unexpected discovery, where an unusually large ligand acts as an inhibitor of an ion channel, a recombinant and catalytically inactive mutant of collinein-1 (His43Arg) was produced and found to preserve its capability to inhibit hEAG1. A molecular docking model was proposed in which Arg79 of the SVSP 99-loop interacts directly with the potassium selectivity filter of the hEAG1 channel.
Collapse
Affiliation(s)
- Johara Boldrini-França
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903, Ribeirão Preto, SP, Brazil.,University of Vila Velha, Av. Comissário José Dantas de Melo, 21, Boa Vista II, 29102-920, Vila Velha, ES, Brazil
| | - Ernesto Lopes Pinheiro-Junior
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903, Ribeirão Preto, SP, Brazil.,Toxicology and Pharmacology, KU Leuven, O&N II Herestraat 49, PO 922, 3000, Leuven, Belgium
| | - Steve Peigneur
- Toxicology and Pharmacology, KU Leuven, O&N II Herestraat 49, PO 922, 3000, Leuven, Belgium
| | - Manuela Berto Pucca
- Medical School of Roraima, Federal University of Roraima, Av. Capitão Ene Garcez, 2413, Bairro Aeroporto, 69310-970, Boa Vista, RR, Brazil
| | - Felipe Augusto Cerni
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903, Ribeirão Preto, SP, Brazil
| | - Rafael Junqueira Borges
- Institute of Biosciences, São Paulo State University (UNESP), Rua Prof. Dr. Antonio Celso Wagner Zanin, 250, 18618-689, Botucatu, SP, Brazil
| | - Tássia Rafaella Costa
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903, Ribeirão Preto, SP, Brazil
| | - Sante Emmanuel Imai Carone
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903, Ribeirão Preto, SP, Brazil
| | - Marcos Roberto de Mattos Fontes
- Institute of Biosciences, São Paulo State University (UNESP), Rua Prof. Dr. Antonio Celso Wagner Zanin, 250, 18618-689, Botucatu, SP, Brazil
| | - Suely Vilela Sampaio
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903, Ribeirão Preto, SP, Brazil
| | - Eliane Candiani Arantes
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903, Ribeirão Preto, SP, Brazil.
| | - Jan Tytgat
- Toxicology and Pharmacology, KU Leuven, O&N II Herestraat 49, PO 922, 3000, Leuven, Belgium.
| |
Collapse
|
6
|
He S, Moutaoufik MT, Islam S, Persad A, Wu A, Aly KA, Fonge H, Babu M, Cayabyab FS. HERG channel and cancer: A mechanistic review of carcinogenic processes and therapeutic potential. Biochim Biophys Acta Rev Cancer 2020; 1873:188355. [PMID: 32135169 DOI: 10.1016/j.bbcan.2020.188355] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/21/2022]
Abstract
The human ether-à-go-go related gene (HERG) encodes the alpha subunit of Kv11.1, which is a voltage-gated K+ channel protein mainly expressed in heart and brain tissue. HERG plays critical role in cardiac repolarization, and mutations in HERG can cause long QT syndrome. More recently, evidence has emerged that HERG channels are aberrantly expressed in many kinds of cancer cells and play important roles in cancer progression. HERG could therefore be a potential biomarker for cancer and a possible molecular target for anticancer drug design. HERG affects a number of cellular processes, including cell proliferation, apoptosis, angiogenesis and migration, any of which could be affected by dysregulation of HERG. This review provides an overview of available information on HERG channel as it relates to cancer, with focus on the mechanism by which HERG influences cancer progression. Molecular docking attempts suggest two possible protein-protein interactions of HERG with the ß1-integrin receptor and the transcription factor STAT-1 as novel HERG-directed therapeutic targeting which avoids possible cardiotoxicity. The role of epigenetics in regulating HERG channel expression and activity in cancer will also be discussed. Finally, given its inherent extracellular accessibility as an ion channel, we discuss regulatory roles of this molecule in cancer physiology and therapeutic potential. Future research should be directed to explore the possibilities of therapeutic interventions targeting HERG channels while minding possible complications.
Collapse
Affiliation(s)
- Siyi He
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | | | - Saadul Islam
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Amit Persad
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Adam Wu
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Khaled A Aly
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Humphrey Fonge
- Department of Medical Imaging, University of Saskatchewan, Saskatoon, Saskatchewan S7N 0W8, Canada; Department of Medical Imaging, Royal University Hospital, Saskatoon, Saskatchewan S7N 0W8, Canada
| | - Mohan Babu
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Francisco S Cayabyab
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| |
Collapse
|
7
|
Thillaiyampalam G, Liberante F, Murray L, Cardwell C, Mills K, Zhang SD. An integrated meta-analysis approach to identifying medications with potential to alter breast cancer risk through connectivity mapping. BMC Bioinformatics 2017; 18:581. [PMID: 29268695 PMCID: PMC5740937 DOI: 10.1186/s12859-017-1989-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 12/06/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Gene expression connectivity mapping has gained much popularity in recent years with a number of successful applications in biomedical research testifying its utility and promise. A major application of connectivity mapping is the identification of small molecule compounds capable of inhibiting a disease state. In this study, we are additionally interested in small molecule compounds that may enhance a disease state or increase the risk of developing that disease. Using breast cancer as a case study, we aim to develop and test a methodology for identifying commonly prescribed drugs that may have a suppressing or inducing effect on the target disease (breast cancer). RESULTS We obtained from public data repositories a collection of breast cancer gene expression datasets with over 7000 patients. An integrated meta-analysis approach to gene expression connectivity mapping was developed, which involved unified processing and normalization of raw gene expression data, systematic removal of batch effects, and multiple runs of balanced sampling for differential expression analysis. Differentially expressed genes stringently selected were used to construct multiple non-joint gene signatures representing the same biological state. Remarkably these non-joint gene signatures retrieved from connectivity mapping separate lists of candidate drugs with significant overlaps, providing high confidence in their predicted effects on breast cancers. Of particular note, among the top 26 compounds identified as inversely connected to the breast cancer gene signatures, 14 of them are known anti-cancer drugs. CONCLUSIONS A few candidate drugs with potential to enhance breast cancer or increase the risk of the disease were also identified; further investigation on a large population is required to firmly establish their effects on breast cancer risks. This work thus provides a novel approach and an applicable example for identifying medications with potential to alter cancer risks through gene expression connectivity mapping.
Collapse
Affiliation(s)
| | - Fabio Liberante
- Centre for Cancer Research and Cell Biology (CCRCB), Queen’s University Belfast, Belfast, UK
| | - Liam Murray
- Centre for Public Health, Queen’s University Belfast, Belfast, UK
| | - Chris Cardwell
- Centre for Public Health, Queen’s University Belfast, Belfast, UK
| | - Ken Mills
- Centre for Cancer Research and Cell Biology (CCRCB), Queen’s University Belfast, Belfast, UK
| | - Shu-Dong Zhang
- Centre for Cancer Research and Cell Biology (CCRCB), Queen’s University Belfast, Belfast, UK
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, L/Derry, Northern Ireland, BT47 6SB UK
| |
Collapse
|
8
|
Dookeran KA, Zhang W, Stayner L, Argos M. Associations of two-pore domain potassium channels and triple negative breast cancer subtype in The Cancer Genome Atlas: systematic evaluation of gene expression and methylation. BMC Res Notes 2017; 10:475. [PMID: 28899398 PMCID: PMC5596847 DOI: 10.1186/s13104-017-2777-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022] Open
Abstract
Objectives It is unclear whether 2-pore domain potassium channels are novel molecular markers with differential expression related to biologically aggressive triple-negative type breast tumors. Our objective was to systematically evaluate associations of 2-pore domain potassium channel gene expression and DNA methylation with triple-negative subtype in The Cancer Genome Atlas invasive breast cancer dataset. Methylation and expression data for all fifteen 2-pore domain potassium family genes were examined for 1040 women, and associations with triple-negative subtype (vs. luminal A) were evaluated using age/race adjusted generalized-linear models, with Bonferroni-corrected significance thresholds. Subtype associated CpG loci were evaluated for functionality related to expression using Spearman’s correlation. Results Overexpression of KCNK5, KCNK9 and KCNK12, and underexpression of KCNK6 and KCNK15, were significantly associated with triple-negative subtype (Bonferroni-corrected p < 0.0033). A total of 195 (114 hypomethylated and 81 hypermethylated) CpG loci were found to be significantly associated with triple-negative subtype (Bonferroni-corrected p < 8.22 × 10−8). Significantly negatively correlated expression patterns that were differentially observed in triple-negative vs. luminal A subtype were demonstrated for: KCNK2 (gene body: cg04923840, cg13916421), KCNK5 (gene body: cg05255811, cg18705155, cg09130674, cg21388745, cg00859574) and KCNK9 (TSS1500: cg21415530, cg12175729; KCNK9/TRAPPC9 intergenic region: cg17336929, cg25900813, cg03919980). CpG loci listed for KCNK5 and KCNK9 all showed relative hypomethylation for probability of triple-negative vs. luminal A subtype. Triple-negative subtype was associated with distinct 2-pore domain potassium channel expression patterns. Both KCNK5 and KCNK9 overexpression appeared to be functionally related to CpG loci hypomethylation. Electronic supplementary material The online version of this article (doi:10.1186/s13104-017-2777-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Keith A Dookeran
- Epidemiology, University of Wisconsin-Milwaukee, Joseph J. Zilber School of Public Health, 1240 N. 10th St, Milwaukee, WI, 53205, USA. .,The Cancer Foundation for Minority and Underserved Populations, Chicago, IL, 60614, USA.
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N. Lake Shore Drive, Suite 1400, Chicago, IL, 60611, USA
| | - Leslie Stayner
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago, School of Public Health, 1603 West Taylor Street, MC923, Chicago, IL, 60612, USA
| | - Maria Argos
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago, School of Public Health, 1603 West Taylor Street, MC923, Chicago, IL, 60612, USA
| |
Collapse
|
9
|
He Y, Niu W, Xia C, Cao B. Daidzein reduces the proliferation and adiposeness of 3T3-L1 preadipocytes via regulating adipogenic gene expression. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.01.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
10
|
Perez-Neut M, Shum A, Cuevas BD, Miller R, Gentile S. Stimulation of hERG1 channel activity promotes a calcium-dependent degradation of cyclin E2, but not cyclin E1, in breast cancer cells. Oncotarget 2015; 6:1631-9. [PMID: 25596745 PMCID: PMC4359320 DOI: 10.18632/oncotarget.2829] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/27/2014] [Indexed: 01/28/2023] Open
Abstract
Cyclin E2 gene amplification, but not cyclin E1, has been recently defined as marker for poor prognosis in breast cancer, and appears to play a major role in proliferation and therapeutic resistance in several breast cancer cells. Our laboratory has previously reported that stimulation of the hERG1 potassium channel with selective activators led to down-regulation of cyclin E2 in breast cancer cells. In this work, we demonstrate that stimulation of hERG1 promotes an ubiquitin-proteasome-dependent degradation of cyclin E2 in multiple breast cancer cell lines representing Luminal A, HER2+ and Trastuzumab-resistant breast cancer cells. In addition we have also reveal that hERG1 stimulation induces an increase in intracellular calcium that is required for cyclin E2 degradation. This novel function for hERG1 activity was specific for cyclin E2, as cyclins A, B, D E1 were unaltered by the treatment. Our results reveal a novel mechanism by which hERG1 activation impacts the tumor marker cyclin E2 that is independent of cyclin E1, and suggest a potential therapeutic use for hERG1 channel activators.
Collapse
Affiliation(s)
- Mathew Perez-Neut
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Andrew Shum
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Bruce D Cuevas
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Richard Miller
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Saverio Gentile
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
11
|
Hosseinzadeh Z, Warsi J, Elvira B, Almilaji A, Shumilina E, Lang F. Up-regulation of Kv1.3 Channels by Janus Kinase 2. J Membr Biol 2015; 248:309-17. [DOI: 10.1007/s00232-015-9772-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/14/2015] [Indexed: 01/08/2023]
|
12
|
Hosseinzadeh Z, Almilaji A, Honisch S, Pakladok T, Liu G, Bhavsar SK, Ruth P, Shumilina E, Lang F. Upregulation of the large conductance voltage- and Ca2+-activated K+ channels by Janus kinase 2. Am J Physiol Cell Physiol 2014; 306:C1041-9. [PMID: 24696148 DOI: 10.1152/ajpcell.00209.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The iberiotoxin-sensitive large conductance voltage- and Ca(2+)-activated potassium (BK) channels (maxi-K(+)-channels) hyperpolarize the cell membrane thus supporting Ca(2+) entry through Ca(2+)-release activated Ca(2+) channels. Janus kinase-2 (JAK2) has been identified as novel regulator of ion transport. To explore whether JAK2 participates in the regulation of BK channels, cRNA encoding Ca(2+)-insensitive BK channels (BK(M513I+Δ899-903)) was injected into Xenopus oocytes with or without cRNA encoding wild-type JAK2, gain-of-function (V617F)JAK2, or inactive (K882E)JAK2. K(+) conductance was determined by dual electrode voltage clamp and BK-channel protein abundance by confocal microscopy. In A204 alveolar rhabdomyosarcoma cells, iberiotoxin-sensitive K(+) current was determined utilizing whole cell patch clamp. A204 cells were further transfected with JAK2 and BK-channel transcript, and protein abundance was quantified by RT-PCR and Western blotting, respectively. As a result, the K(+) current in BK(M513I+Δ899-903)-expressing oocytes was significantly increased following coexpression of JAK2 or (V617F)JAK2 but not (K882E)JAK2. Coexpression of the BK channel with (V617F)JAK2 but not (K882E)JAK2 enhanced BK-channel protein abundance in the oocyte cell membrane. Exposure of BK-channel and (V617F)JAK2-expressing oocytes to the JAK2 inhibitor AG490 (40 μM) significantly decreased K(+) current. Inhibition of channel insertion by brefeldin A (5 μM) decreased the K(+) current to a similar extent in oocytes expressing the BK channel alone and in oocytes expressing the BK channel and (V617F)JAK2. The iberiotoxin (50 nM)-sensitive K(+) current in rhabdomyosarcoma cells was significantly decreased by AG490 pretreatment (40 μM, 12 h). Moreover, overexpression of JAK2 in A204 cells significantly enhanced BK channel mRNA and protein abundance. In conclusion, JAK2 upregulates BK channels by increasing channel protein abundance in the cell membrane.
Collapse
Affiliation(s)
| | - Ahmad Almilaji
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - Sabina Honisch
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - Tatsiana Pakladok
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - GuoXing Liu
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - Shefalee K Bhavsar
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - Peter Ruth
- Institute of Pharmacy, Department of Pharmacology and Toxicology, University of Tübingen, Tübingen, Germany
| | | | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| |
Collapse
|
13
|
Calycosin suppresses breast cancer cell growth via ERβ-dependent regulation of IGF-1R, p38 MAPK and PI3K/Akt pathways. PLoS One 2014; 9:e91245. [PMID: 24618835 PMCID: PMC3949755 DOI: 10.1371/journal.pone.0091245] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 02/11/2014] [Indexed: 12/27/2022] Open
Abstract
We previously reported that calycosin, a natural phytoestrogen structurally similar to estrogen, successfully triggered apoptosis of estrogen receptor (ER)-positive breast cancer cell line, MCF-7. To better understand the antitumor activities of calycosin against breast cancer, besides MCF-7 cells, another ER-positive cell line T-47D was analyzed here, with ER-negative cell lines (MDA-231, MDA-435) as control. Notably, calycosin led to inhibited cell proliferation and apoptosis only in ER-positive cells, particularly in MCF-7 cells, whereas no such effect was observed in ER-negative cells. Then we investigated whether regulation of ERβ, a subtype of ER, contributed to calycosin-induced apoptosis in breast cancer cells. The results showed that incubation of calycosin resulted in enhanced expression ERβ in MCF-7 and T-47D cells, rather than MDA-231 and MDA-435 cells. Moreover, with the upregulation of ERβ, successive changes in downstream signaling pathways were found, including inactivation of insulin-like growth factor 1 receptor (IGF-1R), then stimulation of p38 MAPK and suppression of the serine/threonine kinase (Akt), and finally poly(ADP-ribose) polymerase 1 (PARP-1) cleavage. However, the other two members of the mitogen-activated protein kinase (MAPK) family, extracellular signal-regulated kinase (ERK) 1/2 and c-Jun N-terminal kinase (JNK), were not consequently regulated by downregulated IGF-1R, indicating ERK 1/2 and JNK pathways were not necessary to allow proliferation inhibition by calycosin. Taken together, our results indicate that calycosin tends to inhibit growth and induce apoptosis in ER-positive breast cancer cells, which is mediated by ERβ-induced inhibition of IGF-1R, along with the selective regulation of MAPK and phosphatidylinositol 3-kinase (PI3K)/Akt pathways.
Collapse
|
14
|
Huang L, Huang QY, Huang HQ. The evidence of HeLa cell apoptosis induced with tetraethylammonium using proteomics and various analytical methods. J Biol Chem 2013; 289:2217-29. [PMID: 24297172 DOI: 10.1074/jbc.m113.515932] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tetraethylammonium (TEA) is a potassium channel (KCh) blocker applied in the functional and pharmacological studies of the KChs. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, a colorimetric assay to quantitatively measure living cells, demonstrated that TEA reduced the HeLa cell viability dose-dependently. Flow cytometry analysis indicated an increased apoptosis rate of the HeLa cell after exposing to TEA. The patch clamp technique revealed that the K(+) current of the HeLa cell was inhibited up to 80% when exposed to TEA. In addition, quantitative real-time PCR approach set up cross-talk among the cytotoxicity of TEA, 4-aminopyridine, and anti-cancer drug such as cisplatin. Using comparative proteomics combined with MALDI-TOF MS/MS, 33 significantly changed proteins were found from TEA treatment group; among these proteins, 12 were up-regulated, and 21 were down-regulated. Here we indicated that these proteins were closely connected with many biological functions such as oxidative stress response, signal transduction, metabolism, protein synthesis, and degradation. Both Western blotting and quantitative real-time PCR approaches further verified these differential proteins. Ingenuity Pathways Analysis software, a tool to analyze "omics" data and model biological system, was applied to analyze the interaction pathways of these proteins. The subcellular locations of the differential proteins are also predicted from Uniprot. All results above can help in our understanding of the mechanism of TEA-induced cytotoxicity and provide potential cancer biomarkers. Various experimental results in this study (like those for cisplatin) indicated that TEA is not only a KCh blocker but also a potential anti-cancer drug.
Collapse
Affiliation(s)
- Lin Huang
- From the State Key Laboratory of Cellular Stress Biology, School of Life Sciences, State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen 361005, China and
| | | | | |
Collapse
|
15
|
Weiger TM, Hermann A. Cell proliferation, potassium channels, polyamines and their interactions: a mini review. Amino Acids 2013; 46:681-8. [PMID: 23820618 DOI: 10.1007/s00726-013-1536-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/13/2013] [Indexed: 01/18/2023]
Abstract
Polyamines, which are obligatory molecules involved in cell cycling and proliferation, are subject to a change in their free intracellular concentrations during the cell cycle. Potassium (K(+)) channels are also considered, but less well recognized, to be necessary for cell proliferation by either hyperpolarizing or depolarizing cells during the cell cycle. A block of polyamine synthesis as well as block or knockout of K(+) channels can halt cell proliferation. K(+) channels like BK (maxi calcium (Ca(2+))-activated K(+)), Kir (inward rectifier), M-type K(+)-and TASK (two-pore domain K(+)) channels or the delayed rectifier K(+) channels are modulated in their electrical properties by polyamines. Polyamines are most effective in blocking these channels when applied to the intracellular face of these channels except for TASK channels where they act only from the extracellular side. Quinidine, a general K(+) channel blocker, was found to reduce putrescine concentrations, to block the ornithine decarboxylase and halt cell proliferation. From these results, the question arises if there is an interaction between polyamines, K(+) channels and proliferation. It might be speculated that a decrease of intracellular polyamines allows more K(+) channels to be active, thus inducing hyperpolarization, while an increase of the polyamine concentration may block K(+) channel activity leading to depolarization of the membrane potential. On the other hand, a block or a deletion of K(+) channels may cause a decrease of the polyamine concentration in cells. More research is needed to test these hypotheses.
Collapse
Affiliation(s)
- Thomas M Weiger
- Division of Cellular and Molecular Neurobiology, Department of Cell Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria,
| | | |
Collapse
|