1
|
Rao X, Xue J, Du Y, Zhou Z, Lu Y. Prognosis Prediction of Lung Adenocarcinoma Patients Based on Molecular Subgroups of DNA Methylation. Appl Immunohistochem Mol Morphol 2023; 31:255-265. [PMID: 36877181 DOI: 10.1097/pai.0000000000001114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 11/13/2022] [Indexed: 03/07/2023]
Abstract
Lung adenocarcinoma (LUAD) is a malignant tumor with high mortality. At present, the clinicopathologic feature is the main breakthrough to assess the prognosis of LUAD patients. However, in most cases, the results are less than satisfactory. Cox regression analysis was conducted in this study to obtain methylation sites with significant prognostic relevance based on mRNA expression, DNA methylation data, and clinical data of LUAD from The Cancer Genome Atlas Program database. LUAD patients were grouped into 4 subtypes according to different methylation levels using K-means consensus cluster analysis. By survival analysis, patients were grouped into high-methylation and low-methylation groups. Later, 895 differentially expressed genes (DEGs) were obtained. Eight optimal methylation signature genes associated with prognosis were screened by Cox regression analysis, and a risk assessment model was constructed based on these genes. Samples were then classified into high-risk and low-risk groups depending on the risk assessment model, and prognostic, predictive ability was assessed using survival and receiver operating characteristic (ROC) curves. The results showed that this risk model had a great efficacy in predicting the prognosis of patients, and it was, therefore, able to be an independent prognostic factor. At last, the enrichment analysis demonstrated that the signaling pathways, including cell cycle, homologous recombination, P53 signaling pathway, DNA replication, pentose phosphate pathway, and glycolysis gluconeogenesis were remarkably activated in the high-risk group. In general, we construct an 8-gene model based on DNA methylation molecular subtypes by a series of bioinformatics methods, which can provide new insights for predicting the prognosis of patients with LUAD.
Collapse
Affiliation(s)
- Xiao Rao
- Department of Cardio-Thoracic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | | | | | | | | |
Collapse
|
2
|
Romito O, Guéguinou M, Raoul W, Champion O, Robert A, Trebak M, Goupille C, Potier-Cartereau M. Calcium signaling: A therapeutic target to overcome resistance to therapies in cancer. Cell Calcium 2022; 108:102673. [PMID: 36410063 DOI: 10.1016/j.ceca.2022.102673] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Innate and acquired resistances to therapeutic agents are responsible for the failure of cancer treatments. Due to the multifactorial nature of resistance, the identification of new therapeutic targets is required to improve cancer treatment. Calcium is a universal second messenger that regulates many cellular functions such as proliferation, migration, and survival. Calcium channels, pumps and exchangers tightly regulate the duration, location and magnitude of calcium signals. Many studies have implicated dysregulation of calcium signaling in several pathologies, including cancer. Abnormal calcium fluxes due to altered channel expression or activation contribute to carcinogenesis and promote tumor development. However, there is limited information on the role of calcium signaling in cancer resistance to therapeutic drugs. This review discusses the role of calcium signaling as a mediator of cancer resistance, and assesses the potential value of combining anticancer therapy with calcium signaling modulators to improve the effectiveness of current treatments.
Collapse
Affiliation(s)
- Olivier Romito
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Maxime Guéguinou
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - William Raoul
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Ophélie Champion
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Alison Robert
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Mohamed Trebak
- Vascular Medicine Institute, Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Caroline Goupille
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France; CHRU de Tours, hôpital Bretonneau, Tours, France.
| | - Marie Potier-Cartereau
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| |
Collapse
|
3
|
Xiong J, He HT, Yang HY, Zeng ZG, Zhong CR, Shi H, Ouyang ML, Tao YY, Pang YL, Zhang YH, Hu B, Fu ZX, Miao XL, Zhu HL, Yao G. Synthesis of 4-Tetrazolyl-Substituted 3,4-Dihydroquinazoline Derivatives with Anticancer Activity via a One-Pot Sequential Ugi-Azide/Palladium-Catalyzed Azide-Isocyanide Cross-Coupling/Cyclization Reaction. J Org Chem 2022; 87:9488-9496. [PMID: 35881945 DOI: 10.1021/acs.joc.2c00382] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A new one-pot preparation of 4-tetrazolyl-3,4-dihydroquinazolines has been reported. The Ugi-azide reactions of 2-azidobenzaldehydes, amines, trimethylsilyl azide, and isocyanides produced azide intermediates without separation, which were treated with isocyanides to give 4-tetrazolyl-3,4-dihydroquinazoline derivatives through a sequential Palladium-catalyzed azide-isocyanide cross-coupling/cyclization reaction in moderate to good yields. The biological evaluation demonstrated that compound 6c inhibited breast cancer cells well and displayed broad applications for synthesis and medicinal chemistry.
Collapse
Affiliation(s)
- Jun Xiong
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Hui-Ting He
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - He-Yu Yang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Zhi-Gang Zeng
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning 437100, P. R. China
| | - Cheng-Ran Zhong
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Hang Shi
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Meng-Ling Ouyang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Yuan-Yuan Tao
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Yong-Long Pang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Yang-Hong Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Bo Hu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Zi-Xiang Fu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Xiao-Lei Miao
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| | - Hai-Li Zhu
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, P. R. China
| | - Gang Yao
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, Hubei, P. R. China
| |
Collapse
|
4
|
Zhang L, Bing S, Dong M, Lu X, Xiong Y. Targeting ion channels for the treatment of lung cancer. Biochim Biophys Acta Rev Cancer 2021; 1876:188629. [PMID: 34610420 DOI: 10.1016/j.bbcan.2021.188629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/02/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023]
Abstract
Lung cancer is caused by several environmental and genetic variables and is globally associated with elevated morbidity and mortality. Among these variables, membrane-bound ion channels have a key role in regulating multiple signaling pathways in tumor cells and dysregulation of ion channel expression and function is closely related to proliferation, migration, and metastasis of lung cancer. This work reviews and summarizes current knowledge about the role of ion channels in lung cancer, focusing on the changes in the expression and function of various ion channels in lung cancer and how these changes affect lung cancer cell biology both in vitro and in vivo as evidenced by both genetic and pharmacological studies. It can help understand the molecular mechanisms of various ion channels influencing the initiation and progression of lung cancer and shed new insights into their roles in the development and treatment of this deadly disease.
Collapse
Affiliation(s)
- Liqin Zhang
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China.
| | - Shuya Bing
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China
| | - Mo Dong
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China
| | - Xiaoqiu Lu
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China
| | - Yuancheng Xiong
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China
| |
Collapse
|
5
|
Sharma A, Ramena GT, Elble RC. Advances in Intracellular Calcium Signaling Reveal Untapped Targets for Cancer Therapy. Biomedicines 2021; 9:1077. [PMID: 34572262 PMCID: PMC8466575 DOI: 10.3390/biomedicines9091077] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/15/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023] Open
Abstract
Intracellular Ca2+ distribution is a tightly regulated process. Numerous Ca2+ chelating, storage, and transport mechanisms are required to maintain normal cellular physiology. Ca2+-binding proteins, mainly calmodulin and calbindins, sequester free intracellular Ca2+ ions and apportion or transport them to signaling hubs needing the cations. Ca2+ channels, ATP-driven pumps, and exchangers assist the binding proteins in transferring the ions to and from appropriate cellular compartments. Some, such as the endoplasmic reticulum, mitochondria, and lysosomes, act as Ca2+ repositories. Cellular Ca2+ homeostasis is inefficient without the active contribution of these organelles. Moreover, certain key cellular processes also rely on inter-organellar Ca2+ signaling. This review attempts to encapsulate the structure, function, and regulation of major intracellular Ca2+ buffers, sensors, channels, and signaling molecules before highlighting how cancer cells manipulate them to survive and thrive. The spotlight is then shifted to the slow pace of translating such research findings into anticancer therapeutics. We use the PubMed database to highlight current clinical studies that target intracellular Ca2+ signaling. Drug repurposing and improving the delivery of small molecule therapeutics are further discussed as promising strategies for speeding therapeutic development in this area.
Collapse
Affiliation(s)
- Aarushi Sharma
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| | - Grace T. Ramena
- Department of Aquaculture, University of Arkansas, Pine Bluff, AR 71601, USA;
| | - Randolph C. Elble
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| |
Collapse
|
6
|
Overexpression of T-type calcium channel Cav3.1 in oral squamous cell carcinoma: association with proliferation and anti-apoptotic activity. J Mol Histol 2021; 52:511-520. [PMID: 33394292 DOI: 10.1007/s10735-020-09937-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022]
Abstract
Cav3.1, a subfamily of T-type calcium channel, is overexpressed in various human cancers and exerts important functions in tumor progression. This study is to identify the expression pattern and clinical significance of Cav3.1 in oral squamous cell carcinoma (OSCC). Firstly, the expression levels of Cav3.1 in oral mucosa (OM), dysplasia and oral squamous cell carcinoma (OSCC) were determined and compared by real-time quantitative PCR and Western blot analysis. After that, human tissue microarrays, containing 29 OM, 23 dysplasia and 122 primary OSCC samples, were applied to investigate the expression levels of Cav3.1, proliferation markers [Ki-67, proliferating cell nuclear antigen (PCNA)] and cellular anti-apoptosis markers [B cell lymphoma 2 (Bcl-2)] by immunohistochemistry and digital pathology analysis. In addition, we determined the function of Cav3.1 using knockdown assays of Cav3.1 in vitro. The results demonstrated that the mRNA and protein expression of Cav3.1 were significantly higher in OSCC specimens, and Cav3.1 expression in primary OSCCs was correlated with tumor size and pathological grade. Statistical analysis of immunohistochemical staining showed that Cav3.1 was closely correlated with Ki-67, PCNA and Bcl-2. Functional studies showed that the knockdown of Cav3.1 in OSCC cell lines using RNA interference influenced cell proliferation and apoptosis in vitro. Taken together, these findings suggested that Cav3.1 is overexpressed in OSCC tissues, also associated with proliferative and anti-apoptotic activity in oral squamous cell carcinoma.
Collapse
|
7
|
Analysis of gene expression profiles of lung cancer subtypes with machine learning algorithms. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165822. [PMID: 32360590 DOI: 10.1016/j.bbadis.2020.165822] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/13/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
Lung cancer is one of the most common cancer types worldwide and causes more than one million deaths annually. Lung adenocarcinoma (AC) and lung squamous cell cancer (SCC) are two major lung cancer subtypes and have different characteristics in several aspects. Identifying their differentially expressed genes and different gene expression patterns can deepen our understanding of these two subtypes at the transcriptomic level. In this work, we used several machine learning algorithms to investigate the gene expression profiles of lung AC and lung SCC samples retrieved from Gene Expression Omnibus. First, the profiles were analyzed by using a powerful feature selection method, namely, Monte Carlo feature selection. A feature list, ranking all features according to their importance, and some informative features were obtained. Then, the feature list was used in the incremental feature selection method to extract optimal features, which can allow the support vector machine (SVM) to yield the best performance for classifying lung AC and lung SCC samples. Some top genes (CSTA, TP63, SERPINB13, CLCA2, BICD2, PERP, FAT2, BNC1, ATP11B, FAM83B, KRT5, PARD6G, PKP1) were extensively analyzed to prove that they can be differentially expressed genes between lung AC and lung SCC. Meanwhile, a rule learning procedure was applied on informative features to construct the classification rules. These rules provide a clear procedure of classification and show some different gene expression patterns between lung AC and lung SCC.
Collapse
|
8
|
Nam Y, Ryu KD, Jang C, Moon YH, Kim M, Ko D, Chung KS, Gandini MA, Lee KT, Zamponi GW, Lee JY. Synthesis and cytotoxic effects of 2-thio-3,4-dihydroquinazoline derivatives as novel T-type calcium channel blockers. Bioorg Med Chem 2020; 28:115491. [PMID: 32327350 DOI: 10.1016/j.bmc.2020.115491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 10/24/2022]
Abstract
In our previous work, a series of 2-amino-3,4-dihydroquinazoline derivativesusing an electron acceptor group was reported to be potent T-type calcium channel blockers and exhibit strong cytotoxic effects against various cancerous cell lines. To investigate the role of the guanidine moiety in the 2-amino-3,4-dihydroquinazoline scaffold as a pharmacophore for dual biological activity, a new series of 2-thio-3,4-dihydroquniazoline derivatives using an electron donor group at the C2-position was synthesized and evaluated for T-type calcium channel blocking activity and cytotoxic effects against two human cancerous cell lines (lung cancer A549 and colon cancer HCT-116). Among them, compound 6g showed potent inhibition of Cav3.2 currents (83% inhibition) at 10 µM concentrations. The compound also exhibited IC50 values of 5.0 and 6.4 µM against A549 and HCT-116 cell lines, respectively, which are comparable to the parental lead compound KYS05090. These results indicate that the isothiourea moiety similar to the guanidine moiety of 2-amino-3,4-dihydroquinazoline derivatives may be an essential pharmacophore for the desired biological activities. Therefore, our preliminary work can provide the opportunity to expand a chemical repertoire to improve affinity and selectivity for T-type calcium channels.
Collapse
Affiliation(s)
- Yunchan Nam
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ki Deok Ryu
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Changyoung Jang
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoon Hyoung Moon
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Misong Kim
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dohyeong Ko
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sook Chung
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Maria A Gandini
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada
| | - Kyung-Tae Lee
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada.
| | - Jae Yeol Lee
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
9
|
KCP10043F Represses the Proliferation of Human Non-Small Cell Lung Cancer Cells by Caspase-Mediated Apoptosis via STAT3 Inactivation. J Clin Med 2020; 9:jcm9030704. [PMID: 32150979 PMCID: PMC7141374 DOI: 10.3390/jcm9030704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
We previously reported that 4-(4-fluorobenzylcarbamoylmethyl)-3-(4-cyclohexylphenyl)-2-[3-(N,N-dimethylureido)-N'-methylpropylamino]-3,4-dihydroquinazoline (KCP10043F) can induce G1-phase arrest and synergistic cell death in combination with etoposide in lung cancer cells. Here, we investigated the underlying mechanism by which KCP10043F induces cell death in non-small cell lung cancer (NSCLC). Propidium iodide (PI) and annexin V staining revealed that KCP10043F-induced cytotoxicity was caused by apoptosis. KCP10043F induced a series of intracellular events: (1) downregulation of Bcl-2 and Bcl-xL and upregulation of Bax and cleaved Bid; (2) loss of mitochondrial membrane potential; (3) increase of cytochrome c release; (4) cleavage of procaspase-8, procaspase-9, procaspase-3, and poly (ADP-ribose) polymerase (PARP). In addition, KCP10043F exhibited potent inhibitory effects on constitutive or interleukin-6 (IL-6)-induced signal transducer and activator of transcription (STAT3) phosphorylation and STAT3-regulated genes including survivin, Mcl-1, and cyclin D1. Furthermore, STAT3 overexpression attenuated KCP10043F-induced apoptosis and the cleavage of caspase-9, caspase-3, and PARP. Docking analysis disclosed that KCP10043F could bind to a pocket in the SH2 domain of STAT3 and prevent STAT3 phosphorylation. The oral administration of KCP10043F decreased tumor growth in an A549 xenograft mouse model, as associated with the reduced phosphorylated STAT3, survivin, Mcl-1, and Bcl-2 expression and increased TUNEL staining and PARP cleavage in tumor tissues. Collectively, our data suggest that KCP10043F suppresses NSCLC cell growth through apoptosis induction via STAT3 inactivation.
Collapse
|
10
|
Granados K, Hüser L, Federico A, Sachindra S, Wolff G, Hielscher T, Novak D, Madrigal-Gamboa V, Sun Q, Vierthaler M, Larribère L, Umansky V, Utikal J. T-type calcium channel inhibition restores sensitivity to MAPK inhibitors in de-differentiated and adaptive melanoma cells. Br J Cancer 2020; 122:1023-1036. [PMID: 32063604 PMCID: PMC7109069 DOI: 10.1038/s41416-020-0751-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/19/2019] [Accepted: 01/24/2020] [Indexed: 11/25/2022] Open
Abstract
Background Drug resistance remains as one of the major challenges in melanoma therapy. It is well known that tumour cells undergo phenotypic switching during melanoma progression, increasing melanoma plasticity and resistance to mitogen-activated protein kinase inhibitors (MAPKi). Methods We investigated the melanoma phenotype switching using a partial reprogramming model to de-differentiate murine melanoma cells and target melanoma therapy adaptation against MAPKi. Results Here, we show that partially reprogrammed cells are a less proliferative and more de-differentiated cell population, expressing a gene signature for stemness and suppressing melanocyte-specific markers. To investigate adaptation to MAPKi, cells were exposed to B-Raf Proto-Oncogene (BRAF) and mitogen-activated protein kinase kinase (MEK) inhibitors. De-differentiated cells became less sensitive to MAPKi, showed increased cell viability and decreased apoptosis. Furthermore, T-type calcium channels expression increased in adaptive murine cells and in human adaptive melanoma cells. Treatment with the calcium channel blocker mibefradil induced cell death, differentiation and susceptibility to MAPKi in vitro and in vivo. Conclusion In summary, we show that partial reprogramming of melanoma cells induces de-differentiation and adaptation to MAPKi. Moreover, we postulated a calcium channel blocker such as mibefradil, as a potential candidate to restore sensitivity to MAPKi in adaptive melanoma cells.
Collapse
Affiliation(s)
- Karol Granados
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany.,Department of Biochemistry, School of Medicine, University of Costa Rica (UCR), Rodrigo Facio Campus, San Pedro Montes Oca, San Jose, 2060, Costa Rica
| | - Laura Hüser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Aniello Federico
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Sachindra Sachindra
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany.,Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gretchen Wolff
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Novak
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Verónica Madrigal-Gamboa
- Department of Biochemistry, School of Medicine, University of Costa Rica (UCR), Rodrigo Facio Campus, San Pedro Montes Oca, San Jose, 2060, Costa Rica
| | - Qian Sun
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Marlene Vierthaler
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Lionel Larribère
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, D-68135, Mannheim, Germany.
| |
Collapse
|
11
|
Visa A, Shaikh S, Alza L, Herreros J, Cantí C. The Hard-To-Close Window of T-Type Calcium Channels. Trends Mol Med 2019; 25:571-584. [PMID: 31031178 DOI: 10.1016/j.molmed.2019.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 01/03/2023]
Abstract
T-Type calcium channels (TTCCs) are key regulators of membrane excitability, which is the reason why TTCC pharmacology is subject to intensive research in the neurological and cardiovascular fields. TTCCs also play a role in cancer physiology, and pharmacological blockers such as tetralols and dihydroquinazolines (DHQs) reduce the viability of cancer cells in vitro and slow tumor growth in murine xenografts. However, the available compounds are better suited to blocking TTCCs in excitable membranes rather than TTCCs contributing window currents at steady potentials. Consistently, tetralols and dihydroquinazolines exhibit cytostatic/cytotoxic activities at higher concentrations than those required for TTCC blockade, which may involve off-target effects. Gene silencing experiments highlight the targetability of TTCCs, but further pharmacological research is required for TTCC blockade to become a chemotherapeutic option.
Collapse
Affiliation(s)
- Anna Visa
- Laboratory of Calcium Cell Signaling, IRBLleida-Universitat de Lleida, Rovira Roure, 80, 25198-Lleida, Spain
| | - Soni Shaikh
- Laboratory of Calcium Cell Signaling, IRBLleida-Universitat de Lleida, Rovira Roure, 80, 25198-Lleida, Spain
| | - Lía Alza
- Laboratory of Calcium Cell Signaling, IRBLleida-Universitat de Lleida, Rovira Roure, 80, 25198-Lleida, Spain
| | - Judit Herreros
- Laboratory of Calcium Cell Signaling, IRBLleida-Universitat de Lleida, Rovira Roure, 80, 25198-Lleida, Spain
| | - Carles Cantí
- Laboratory of Calcium Cell Signaling, IRBLleida-Universitat de Lleida, Rovira Roure, 80, 25198-Lleida, Spain.
| |
Collapse
|
12
|
Canella R, Martini M, Cavicchio C, Cervellati F, Benedusi M, Valacchi G. Involvement of the TREK-1 channel in human alveolar cell membrane potential and its regulation by inhibitors of the chloride current. J Cell Physiol 2019; 234:17704-17713. [PMID: 30805940 DOI: 10.1002/jcp.28396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 01/03/2019] [Accepted: 01/10/2019] [Indexed: 12/14/2022]
Abstract
K+ channels of the alveolar epithelium control the driving force acting on the ionic and solvent flow through the cell membrane contributing to the maintenance of cell volume and the constitution of epithelial lining fluid. In the present work, we analyze the effect of the Cl- channel inhibitors: (4-[(2-butyl-6,7-dichloro-2-cyclopentyl-2,3-dihydro-1-oxo-inden-5-yl)oxy] butanoic acid (DCPIB) and 9-anthracenecarboxylic acid (9-AC) on the total current in a type II pneumocytes (A549 cell line) model by patch clamp, immunocytochemical, and gene knockdown techniques. We noted that DCPIB and 9-AC promote the activation of K conductance. In fact, they significantly increase the intensity of the current and shift its reversal potential to values more negative than the control. By silencing outward rectifier channel in its anoctamin 6 portion, we excluded a direct involvement of Cl- ions in modulation of IK and, by means of functional tests with its specific inhibitor spadin, we identified the TREK-1 channel as the presumable target of both drugs. As the activity of TREK-1 has a key role for the correct functioning of the alveolar epithelium, the identification of DCPIB and 9-AC molecules as its activators suggests their possible use to build new pharmacological tools for the modulation of this channel.
Collapse
Affiliation(s)
- Rita Canella
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Marta Martini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Carlotta Cavicchio
- Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina
| | - Franco Cervellati
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Mascia Benedusi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina
| |
Collapse
|
13
|
One-pot and regioselective synthesis of polysubstituted 3,4-dihydroquinazolines and 4,5-dihydro-3H-1,4-benzodiazepin-3-ones by sequential Ugi/Staudinger/aza-Wittig reaction. Tetrahedron 2019. [DOI: 10.1016/j.tet.2019.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
14
|
Long non-coding RNA MT1DP shunts the cellular defense to cytotoxicity through crosstalk with MT1H and RhoC in cadmium stress. Cell Discov 2018; 4:5. [PMID: 29507753 PMCID: PMC5824791 DOI: 10.1038/s41421-017-0005-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Metallothioneins (MTs) are known to protect cells against oxidative stress, especially providing protection against cadmium (Cd) toxicity in hepatocytes. There are various gene variants and pseudogenes for MTs; however, there is little understanding on the functions of those non-coding MT members that are known to be expressed as long non-coding RNAs (lncRNAs) nowadays. Different from most protein-coding MT members, MT1DP was here found that remarkably induced to provoke cytotoxicity in hepatocytes in response to Cd treatment. MT1DP exerted such a pro-apoptotic function in Cd-treated hepatocytes through interacting with two partners: RhoC and MT1H. On one hand, MT1DP interacted with RhoC protein to increase the latter’s stability by preventing lysosome-dependent protein degradation. Therefore, upon Cd stress, MT1DP/RhoC complex was quickly reinforced to activate RhoC-CCN1/2-AKT signaling and potentiate Ca2+ influx, leading to enhanced Cd uptake and elevated Cd toxicity. On the other hand, MT1H, a protein-coding member of the MT family with little known function, was found to quickly respond to Cd exposure along with MT1DP. Mechanistically, MT1H and MT1DP were uncovered to mutually protect each other through a reciprocal ceRNA mechanism, building up a positive feedback loop to enforce MT1DP-conducted signaling upon Cd exposure. Moreover, MT1DP was found to contribute much more to the activation of RhoC-CCN1/2-AKT signaling than MT1H. Considered together, we here unveiled a mystery whether a pseudogene within the MT family, MT1DP, has actual biological functions in regulating Cd-induced cellular defense. Our findings unearthed an important role of pseudogene MT1DP in calibrating the cellular machinery to switch the cellular defense to cytotoxicity through crosslinking an interplay between its two partners, namely MT1H and RhoC, under cadmium stress.
Collapse
|
15
|
Synthesis and biological evaluation of fluoro-substituted 3,4-dihydroquinazoline derivatives for cytotoxic and analgesic effects. Bioorg Med Chem 2017; 25:4656-4664. [DOI: 10.1016/j.bmc.2017.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 01/15/2023]
|
16
|
One-pot and regioselective synthesis of 3,4-dihydroquinazolines by Sequential Ugi/Staudinger/aza-Wittig reaction starting from functionalized isocyanides. Tetrahedron 2017. [DOI: 10.1016/j.tet.2017.08.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
17
|
Singh A, Sharma RK, Chagtoo M, Agarwal G, George N, Sinha N, Godbole MM. 1H NMR Metabolomics Reveals Association of High Expression of Inositol 1, 4, 5 Trisphosphate Receptor and Metabolites in Breast Cancer Patients. PLoS One 2017; 12:e0169330. [PMID: 28072864 PMCID: PMC5225010 DOI: 10.1371/journal.pone.0169330] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 12/15/2016] [Indexed: 01/01/2023] Open
Abstract
1H NMR is used to detect alterations in metabolites and their linkage to metabolic processes in a number of pathological conditions including breast cancer. Inositol 1, 4, 5 trisphosphate (IP3R) receptor is an intracellular calcium channel known to regulate metabolism and cellular bioenergetics. Its expression is up regulated in a number of cancers. However, its linkage to metabolism in disease conditions has not been evaluated. This study was designed to determine the association if any, of these metabolites with altered expression of IP3R in breast cancer. We used 1H NMR to identify metabolites in the serum of breast cancer patients (n = 27) and performed Real-time Polymerase Chain Reaction analysis for quantifying the expression of IP3R type 3 and type 2 in tissues from breast cancer patients (n = 40). Principal Component Analysis (PCA) and Partial Least Square-Discriminant Analysis (PLS-DA) clearly distinguished patients with high/low IP3R expression from healthy subjects. The present study revealed high expression of IP3R type 2 and type 3 in human breast tumor tissue compared to adjacent non-tumorous tissue. Moreover, patients with ≥ 2-fold increase in IP3R (high IP3R group) had significantly higher concentration of metabolic intermediates compared to those with < 2-fold increase in IP3R (low IP3R group). We observed an increase in lipoprotein content and the levels of metabolites like lactate, lysine and alanine and a decrease in the levels of pyruvate and glucose in serum of high IP3R group patients when compared to those in healthy subjects. Receiver operating characteristic (ROC) curve analysis was performed to show the clinical utility of metabolites. In addition to the human studies, functional relevance of IP3Rs in causing metabolic disruption was observed in MCF-7 and MDA MB-231 cells. Results from our studies bring forth the importance of metabolic (or metabolomics) profiling of serum by 1H NMR in conjunction with tissue expression studies for characterizing breast cancer patients. The results from this study provide new insights into relationship of breast cancer metabolites with IP3R.
Collapse
Affiliation(s)
- Aru Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, India
| | | | - Megha Chagtoo
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, India
| | - Gaurav Agarwal
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, India
| | - Nelson George
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, India
| | - Neeraj Sinha
- Centre of Biomedical Research, Raebareli Road, Lucknow, India
| | - Madan M. Godbole
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, India
- * E-mail:
| |
Collapse
|
18
|
Fendiline inhibits proliferation and invasion of pancreatic cancer cells by interfering with ADAM10 activation and β-catenin signaling. Oncotarget 2016; 6:35931-48. [PMID: 26440150 PMCID: PMC4742152 DOI: 10.18632/oncotarget.5933] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/14/2015] [Indexed: 12/20/2022] Open
Abstract
ADAM10 (A Disintegrin and Metalloprotease Domain 10) affects the pathophysiology of various cancers, and we had shown that inhibition of ADAM10 sensitizes pancreatic cancer cells to gemcitabine. ADAM10 is activated in response to calcium influx, and here we examined if calcium channel blockers (CCB) would impede ADAM10 activation and affect biology of pancreatic cancer cells. We find that the CCB, fendiline, significantly reduces proliferation, migration, invasion, and anchorage independent growth of pancreatic cancer cells. This was associated with ADAM10 inhibition and its localization at the actin-rich membrane protrusions. Further, fendiline-treated cells formed cadherin-catenin positive tight adherens junctions and elicited defective protein trafficking and recycling. Furthermore, the expression of β-catenin target genes, cyclinD1, c-Myc and CD44, were significantly decreased, suggesting that fendiline might prevent cell proliferation and migration by inhibiting ADAM10 function, cadherin proteolysis and stabilization of cadherin-catenin interaction at the plasma membrane. This will subsequently diminish β-catenin intracellular signaling and repress TCF/LEF target gene expression. Supporting this notion, RNAi-directed downregulation of ADAM10 in cancer cells decreased the expression of cyclinD1, c-Myc and CD44. Furthermore, analysis of human pancreatic tumor tissue microarrays and lysates showed elevated levels of ADAM10, suggesting that aberrant activation of ADAM10 plays a fundamental role in growth and metastasis of PDACs and inhibiting this pathway might be a viable strategy to combat PDACs.
Collapse
|
19
|
Buchanan PJ, McCloskey KD. Ca V channels and cancer: canonical functions indicate benefits of repurposed drugs as cancer therapeutics. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:621-633. [PMID: 27342111 PMCID: PMC5045480 DOI: 10.1007/s00249-016-1144-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/17/2016] [Accepted: 05/23/2016] [Indexed: 01/19/2023]
Abstract
The importance of ion channels in the hallmarks of many cancers is increasingly recognised. This article reviews current knowledge of the expression of members of the voltage-gated calcium channel family (CaV) in cancer at the gene and protein level and discusses their potential functional roles. The ten members of the CaV channel family are classified according to expression of their pore-forming α-subunit; moreover, co-expression of accessory α2δ, β and γ confers a spectrum of biophysical characteristics including voltage dependence of activation and inactivation, current amplitude and activation/inactivation kinetics. CaV channels have traditionally been studied in excitable cells including neurones, smooth muscle, skeletal muscle and cardiac cells, and drugs targeting the channels are used in the treatment of hypertension and epilepsy. There is emerging evidence that several CaV channels are differentially expressed in cancer cells compared to their normal counterparts. Interestingly, a number of CaV channels also have non-canonical functions and are involved in transcriptional regulation of the expression of other proteins including potassium channels. Pharmacological studies show that CaV canonical function contributes to the fundamental biology of proliferation, cell-cycle progression and apoptosis. This raises the intriguing possibility that calcium channel blockers, approved for the treatment of other conditions, could be repurposed to treat particular cancers. Further research will reveal the full extent of both the canonical and non-canonical functions of CaV channels in cancer and whether calcium channel blockers are beneficial in cancer treatment.
Collapse
Affiliation(s)
- Paul J Buchanan
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7AE, UK.,National Institute of Cellular Biotechnology, School of Nursing and Human Science, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Karen D McCloskey
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7AE, UK.
| |
Collapse
|
20
|
Lange I, Moschny J, Tamanyan K, Khutsishvili M, Atha D, Borris RP, Koomoa DL. Scrophularia orientalis extract induces calcium signaling and apoptosis in neuroblastoma cells. Int J Oncol 2016; 48:1608-16. [PMID: 26848085 PMCID: PMC4777595 DOI: 10.3892/ijo.2016.3373] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/15/2015] [Indexed: 12/26/2022] Open
Abstract
Effective neuroblastoma (NB) treatments are still limited despite treatment options available today. Therefore, this study attempted to identify novel plant extracts that have anticancer effects. Cytotoxicity and increased intracellular calcium levels were determined using the Sulforhodamine B (SRB) assay and Fluo4-AM (acetoxymethyl) staining and fluorescence microscopy in NB cells in order to screen a library of plant extracts. The current study examined the anticancer effects of a dichloromethane extract from Scrophularia orientalis L. (Scrophulariaceae), a plant that has been used in Traditional Chinese Medicine. This extract contained highly potent agents that significantly reduced cell survival and increased calcium levels in NB cells. Further analysis revealed that cell death induced by this extract was associated with intracellular calcium release, opening of the MPTP, caspase 3- and PARP-cleavage suggesting that this extract induced aberrant calcium signaling that resulted in apoptosis via the mitochondrial pathway. Therefore, agents from Scrophularia orientalis may have the potential to lead to new chemo therapeutic anticancer drugs. Furthermore, targeting intracellular calcium signaling may be a novel strategy to develop more effective treatments for NB.
Collapse
Affiliation(s)
- Ingo Lange
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, USA
| | - Julia Moschny
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, USA
| | - Kamilla Tamanyan
- Institute of Botany, National Academy of Sciences, Yerevan, Armenia
| | - Manana Khutsishvili
- National Herbarium of Georgia, Institute of Botany, Ilia State University, Tbilisi, Georgia
| | - Daniel Atha
- The New York Botanical Garden, Bronx, NY, USA
| | - Robert P Borris
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Dana-Lynn Koomoa
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, USA
| |
Collapse
|
21
|
Byun JS, Sohn JM, Leem DG, Park B, Nam JH, Shin DH, Shin JS, Kim HJ, Lee KT, Lee JY. In vitro synergistic anticancer activity of the combination of T-type calcium channel blocker and chemotherapeutic agent in A549 cells. Bioorg Med Chem Lett 2015; 26:1073-1079. [PMID: 26739776 DOI: 10.1016/j.bmcl.2015.12.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 11/10/2015] [Accepted: 12/04/2015] [Indexed: 01/15/2023]
Abstract
As a result of our continuous research, new 3,4-dihydroquinazoline derivative containing ureido group, KCP10043F was synthesized and evaluated for T-type Ca(2+) channel (Cav3.1) blockade, cytotoxicity, and cell cycle arrest against human non-small cell lung (A549) cells. KCP10043F showed both weaker T-type Ca(2+) channel blocking activity and less cytotoxicity against A549 cells than parent compound KYS05090S [4-(benzylcarbamoylmethyl)-3-(4-biphenylyl)-2-(N,N',N'-trimethyl-1,5-pentanediamino)-3,4-dihydroquinazoline 2 hydrochloride], but it exhibited more potent G1-phase arrest than KYS05090S in A549 cells. This was found to be accompanied by the downregulations of cyclin-dependent kinase (CDK) 2, CDK4, CDK6, cyclin D2, cyclin D3, and cyclin E at the protein levels. However, p27(KIP1) as a CDK inhibitor was gradually upregulated at the protein levels and increased recruitment to CDK2, CDK4 and CDK6 after KCP10043F treatment. Based on the strong G1-phase cell cycle arrest of KCP10043F in A549 cells, the combination of KCP10043F with etoposide (or cisplatin) resulted in a synergistic cell death (combination index=0.2-0.8) via the induction of apoptosis compared with either agent alone. Taken together with these overall results and the favorable in vitro ADME (absorption, distribution, metabolism, and excretion) profiles of KCP10043F, therefore, it could be used as a potential agent for the combination therapy on human lung cancer.
Collapse
Affiliation(s)
- Joon Seok Byun
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Joo Mi Sohn
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Dong Gyu Leem
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Byeongyeon Park
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Ji Hye Nam
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Dong Hyun Shin
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Ji Sun Shin
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hyoung Ja Kim
- Molecular Recognition Research Center, Future Convergence Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Kyung-Tae Lee
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Jae Yeol Lee
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
22
|
Nussinov R, Tsai CJ, Muratcioglu S, Jang H, Gursoy A, Keskin O. Principles of K-Ras effector organization and the role of oncogenic K-Ras in cancer initiation through G1 cell cycle deregulation. Expert Rev Proteomics 2015; 12:669-82. [DOI: 10.1586/14789450.2015.1100079] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
23
|
Nussinov R, Muratcioglu S, Tsai CJ, Jang H, Gursoy A, Keskin O. The Key Role of Calmodulin in KRAS-Driven Adenocarcinomas. Mol Cancer Res 2015; 13:1265-73. [PMID: 26085527 PMCID: PMC4572916 DOI: 10.1158/1541-7786.mcr-15-0165] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/09/2015] [Indexed: 12/14/2022]
Abstract
KRAS4B is a highly oncogenic splice variant of the KRAS isoform. It is the only isoform associated with initiation of adenocarcinomas. Insight into why and how KRAS4B can mediate ductal adenocarcinomas, particularly of the pancreas, is vastly important for its therapeutics. Here we point out the overlooked critical role of calmodulin (CaM). Calmodulin selectively binds to GTP-bound K-Ras4B; but not to other Ras isoforms. Cell proliferation and growth require the MAPK (Raf/MEK/ERK) and PI3K/Akt pathways. We propose that Ca(2+)/calmodulin promote PI3Kα/Akt signaling, and suggest how. The elevated calcium levels clinically observed in adenocarcinomas may explain calmodulin's involvement in recruiting and stimulating PI3Kα through interaction with its n/cSH2 domains as well as K-Ras4B; importantly, it also explains why K-Ras4B specifically is a key player in ductal carcinomas, such as pancreatic (PDAC), colorectal (CRC), and lung cancers. We hypothesize that calmodulin recruits and helps activate PI3Kα at the membrane, and that this is the likely reason for Ca(2+)/calmodulin dependence in adenocarcinomas. Calmodulin can contribute to initiation/progression of ductal cancers via both PI3Kα/Akt and Raf/MEK/ERK pathways. Blocking the K-Ras4B/MAPK pathway and calmodulin/PI3Kα binding in a K-Ras4B/calmodulin/PI3Kα trimer could be a promising adenocarcinoma-specific therapeutic strategy.
Collapse
Affiliation(s)
- Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NCI at Frederick, Frederick, Maryland. Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Serena Muratcioglu
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| | - Chung-Jung Tsai
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NCI at Frederick, Frederick, Maryland
| | - Hyunbum Jang
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NCI at Frederick, Frederick, Maryland
| | - Attila Gursoy
- Department of Computer Engineering, Koc University, Istanbul, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| |
Collapse
|
24
|
Meta-Analysis of Public Microarray Datasets Reveals Voltage-Gated Calcium Gene Signatures in Clinical Cancer Patients. PLoS One 2015; 10:e0125766. [PMID: 26147197 PMCID: PMC4493072 DOI: 10.1371/journal.pone.0125766] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 03/26/2015] [Indexed: 12/25/2022] Open
Abstract
Voltage-gated calcium channels (VGCCs) are well documented to play roles in cell proliferation, migration, and apoptosis; however, whether VGCCs regulate the onset and progression of cancer is still under investigation. The VGCC family consists of five members, which are L-type, N-type, T-type, R-type and P/Q type. To date, no holistic approach has been used to screen VGCC family genes in different types of cancer. We analyzed the transcript expression of VGCCs in clinical cancer tissue samples by accessing ONCOMINE (www.oncomine.org), a web-based microarray database, to perform a systematic analysis. Every member of the VGCCs was examined across 21 different types of cancer by comparing mRNA expression in cancer to that in normal tissue. A previous study showed that altered expression of mRNA in cancer tissue may play an oncogenic role and promote tumor development; therefore, in the present findings, we focus only on the overexpression of VGCCs in different types of cancer. This bioinformatics analysis revealed that different subtypes of VGCCs (CACNA1C, CACNA1D, CACNA1B, CACNA1G, and CACNA1I) are implicated in the development and progression of diverse types of cancer and show dramatic up-regulation in breast cancer. CACNA1F only showed high expression in testis cancer, whereas CACNA1A, CACNA1C, and CACNA1D were highly expressed in most types of cancer. The current analysis revealed that specific VGCCs likely play essential roles in specific types of cancer. Collectively, we identified several VGCC targets and classified them according to different cancer subtypes for prospective studies on the underlying carcinogenic mechanisms. The present findings suggest that VGCCs are possible targets for prospective investigation in cancer treatment.
Collapse
|
25
|
Cho S, Choi MJ, Kim M, Lee S, Lee J, Lee SJ, Cho H, Lee KT, Lee JY. Three-dimensional quantitative structure–activity relationship study on anti-cancer activity of 3,4-dihydroquinazoline derivatives against human lung cancer A549 cells. J Mol Struct 2015. [DOI: 10.1016/j.molstruc.2014.12.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
26
|
Zhang L, Wang L, Jiang J, Zheng D, Liu S, Liu C. Lipopolysaccharides upregulate calcium concentration in mouse uterine smooth muscle cells through the T-type calcium channels. Int J Mol Med 2014; 35:784-90. [PMID: 25573237 DOI: 10.3892/ijmm.2014.2054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 12/08/2014] [Indexed: 11/06/2022] Open
Abstract
Infection is a significant cause of preterm birth. Abnormal changes in intracellular calcium signals are the ultimate triggers of early uterine contractions that result in preterm birth. T‑type calcium channels play an important role in the pathogenesis of cancer, as well as endocrine and cardiovascular diseases. However, there are limited studies on their role in uterine contractions and parturition. In the present study, mouse uterine smooth muscle cells were isolated and treated with lipopolysaccharides (LPS) to mimic the microenvironment of uterine infection in vitro to investigate the role of T‑type calcium channels in the process of infection‑induced preterm birth. The results from quantitative polymerase chain reaction and western blot analysis showed that LPS significantly induced the expression of the Cav3.1 and Cav3.2 subtypes of T‑type calcium channels. Measurements of intracellular calcium concentration showed a significant increase in response to LPS. However, these effects can be reversed by T‑type calcium channel blockers. Western blot analysis further indicated that LPS induced the activation of the nuclear factor (NF)‑κB signaling pathway, and endothelin‑1 (ET‑1) was significantly upregulated, whereas NF‑κB inhibitors significantly inhibited the LPS‑induced upregulation of Cav3.1, Cav3.2 and ET‑1 expression. In addition, ET‑1 directly induced Cav3.1 and Cav3.2 expression, whereas ET‑1 antagonists inhibited the LPS‑induced upregulation of Cav3.1 and Cav3.2 expression. In conclusion, the present study demonstrates that infection triggers the upregulation of T‑type calcium channels and promotes calcium influx. This process relies on the activation of the NF‑κB/ET‑1 signaling pathway. The T‑type calcium channel is expected to become an effective target for the prevention of infection‑induced preterm birth.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, P.R. China
| | - Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, P.R. China
| | - Jingyi Jiang
- Clinical, Medical and Pharmaceutical College, China Medical University, Shenyang 110002, P.R. China
| | - Dongming Zheng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, P.R. China
| | - Sishi Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, P.R. China
| | - Caixia Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, P.R. China
| |
Collapse
|
27
|
Kwon GH, Cho S, Lee J, Sohn JM, Byun JS, Lee KT, Lee JY. CoMSIA 3D-QSAR Analysis of 3,4-Dihydroquinazoline Derivatives Against Human Colon Cancer HT-29 Cells. B KOREAN CHEM SOC 2014. [DOI: 10.5012/bkcs.2014.35.11.3181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|