201
|
Hisamatsu Y, Suzuki N, Masum AA, Shibuya A, Abe R, Sato A, Tanuma SI, Aoki S. Cationic Amphiphilic Tris-Cyclometalated Iridium(III) Complexes Induce Cancer Cell Death via Interaction with Ca2+-Calmodulin Complex. Bioconjug Chem 2016; 28:507-523. [DOI: 10.1021/acs.bioconjchem.6b00627] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Yosuke Hisamatsu
- Faculty of Pharmaceutical Sciences, ‡Research Institute for Biomedical Sciences, §Division of Medical-Science-Engineering
Cooperation and ∥Imaging Frontier Center, Research Institute for Science
and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Nozomi Suzuki
- Faculty of Pharmaceutical Sciences, ‡Research Institute for Biomedical Sciences, §Division of Medical-Science-Engineering
Cooperation and ∥Imaging Frontier Center, Research Institute for Science
and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Abdullah-Al Masum
- Faculty of Pharmaceutical Sciences, ‡Research Institute for Biomedical Sciences, §Division of Medical-Science-Engineering
Cooperation and ∥Imaging Frontier Center, Research Institute for Science
and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Ai Shibuya
- Faculty of Pharmaceutical Sciences, ‡Research Institute for Biomedical Sciences, §Division of Medical-Science-Engineering
Cooperation and ∥Imaging Frontier Center, Research Institute for Science
and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Ryo Abe
- Faculty of Pharmaceutical Sciences, ‡Research Institute for Biomedical Sciences, §Division of Medical-Science-Engineering
Cooperation and ∥Imaging Frontier Center, Research Institute for Science
and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Akira Sato
- Faculty of Pharmaceutical Sciences, ‡Research Institute for Biomedical Sciences, §Division of Medical-Science-Engineering
Cooperation and ∥Imaging Frontier Center, Research Institute for Science
and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Sei-ichi Tanuma
- Faculty of Pharmaceutical Sciences, ‡Research Institute for Biomedical Sciences, §Division of Medical-Science-Engineering
Cooperation and ∥Imaging Frontier Center, Research Institute for Science
and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Shin Aoki
- Faculty of Pharmaceutical Sciences, ‡Research Institute for Biomedical Sciences, §Division of Medical-Science-Engineering
Cooperation and ∥Imaging Frontier Center, Research Institute for Science
and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| |
Collapse
|
202
|
Anguita E, Villalobo A. Src-family tyrosine kinases and the Ca 2+ signal. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:915-932. [PMID: 27818271 DOI: 10.1016/j.bbamcr.2016.10.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/25/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023]
Abstract
In this review, we shall describe the rich crosstalk between non-receptor Src-family kinases (SFKs) and the Ca2+ transient generated in activated cells by a variety of extracellular and intracellular stimuli, resulting in diverse signaling events. The exchange of information between SFKs and Ca2+ is reciprocal, as it flows in both directions. These kinases are main actors in pathways leading to the generation of the Ca2+ signal, and reciprocally, the Ca2+ signal modulates SFKs activity and functions. We will cover how SFKs participate in the generation of the cytosolic Ca2+ rise upon activation of a series of receptors and the mechanism of clearance of this Ca2+ signal. The role of SFKs modulating Ca2+-translocating channels participating in these events will be amply discussed. Finally, the role of the Ca2+ sensor protein calmodulin on the activity of c-Src, and potentially on other SFKs, will be outlined as well. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Estefanía Anguita
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
203
|
Berchtold MW, Zacharias T, Kulej K, Wang K, Torggler R, Jespersen T, Chen JN, Larsen MR, la Cour JM. The Arrhythmogenic Calmodulin Mutation D129G Dysregulates Cell Growth, Calmodulin-dependent Kinase II Activity, and Cardiac Function in Zebrafish. J Biol Chem 2016; 291:26636-26646. [PMID: 27815504 DOI: 10.1074/jbc.m116.758680] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/04/2016] [Indexed: 11/06/2022] Open
Abstract
Calmodulin (CaM) is a Ca2+ binding protein modulating multiple targets, several of which are associated with cardiac pathophysiology. Recently, CaM mutations were linked to heart arrhythmia. CaM is crucial for cell growth and viability, yet the effect of the arrhythmogenic CaM mutations on cell viability, as well as heart rhythm, remains unknown, and only a few targets with relevance for heart physiology have been analyzed for their response to mutant CaM. We show that the arrhythmia-associated CaM mutants support growth and viability of DT40 cells in the absence of WT CaM except for the long QT syndrome mutant CaM D129G. Of the six CaM mutants tested (N53I, F89L, D95V, N97S, D129G, and F141L), three showed a decreased activation of Ca2+/CaM-dependent kinase II, most prominently the D129G CaM mutation, which was incapable of stimulating Thr286 autophosphorylation. Furthermore, the CaM D129G mutation led to bradycardia in zebrafish and an arrhythmic phenotype in a subset of the analyzed zebrafish.
Collapse
Affiliation(s)
| | | | - Katarzyna Kulej
- the Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark, and
| | - Kevin Wang
- the Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095
| | | | - Thomas Jespersen
- the Danish Arrhythmia Research Centre, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jau-Nian Chen
- the Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095
| | - Martin R Larsen
- the Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark, and
| | | |
Collapse
|
204
|
Genetic variation in the insulin, insulin-like growth factor, growth hormone, and leptin pathways in relation to breast cancer in African-American women: the AMBER consortium. NPJ Breast Cancer 2016; 2. [PMID: 27942580 PMCID: PMC5142758 DOI: 10.1038/npjbcancer.2016.34] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The insulin/insulin-like growth factor (IGF) system and related pathways such as growth hormone, and leptin signaling have a key role in cancer development. It is unclear how germline variation in these pathways affects breast cancer risk. We conducted gene-based analyses of 184 genes in the insulin/IGF, growth hormone, and leptin pathways to identify genetic variation associated with risk of breast cancer overall, and for estrogen receptor (ER) subtypes. Tag single-nucleotide polymorphisms (SNPs) for each gene were selected and genotyped on a customized Illumina SNP array. Imputation was carried out using 1000 Genomes haplotypes. The analysis included 91,627 SNPs genotyped or imputed in 3,663 breast cancer cases, (1,983 ER-positive and 1,098 ER-negative) and 4,687 controls from the African American Breast Cancer Epidemiology and Risk consortium, a collaborative project of four large studies of breast cancer in African-American women (Carolina Breast Cancer Study, Black Women's Health Study, Women's Circle of Health Study, and Multiethnic Cohort). We used a multi-locus adaptive joint test to determine the association of each gene with overall breast cancer and ER subtypes. The most significant gene associations (P⩽0.01) were BAIAP2 and CALM2 for overall breast cancer; BAIAP2 and CSNK2A1 for ER+ breast cancer; and BRAF, BAD, and MAPK3 for ER− breast cancer. The association of BAD with ER− breast cancer was explained by a two-SNP risk model; all other associations were best explained by one-SNP risk models. In total, six genes and seven SNPs had suggestive associations with overall breast cancer or ER subtypes in African-American women.
Collapse
|
205
|
Calcium-induced calmodulin conformational change. Electrochemical evaluation. Bioelectrochemistry 2016; 113:69-78. [PMID: 27768936 DOI: 10.1016/j.bioelechem.2016.10.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/27/2016] [Accepted: 10/04/2016] [Indexed: 01/08/2023]
Abstract
Calmodulin (CaM) is an essential protein present in all eukaryote cells, ranging from vertebrates to unicellular organisms. CaM is the most important Ca2+ signalling protein, composed of two domains, N- and C-terminal domains, linked by a flexible central α-helix, and is responsible for the regulation of numerous calcium-mediated signalling pathways. Four calcium ions bind to CaM, changing its conformation and determining how it recognizes and regulates its cellular targets. The oxidation mechanism of native and denatured CaM, at a glassy carbon electrode, was investigated using differential pulse voltammetry and electrochemical impedance spectroscopy. Native and denatured CaM presented only one oxidation peak, related to the tyrosine amino acid residue oxidation. Calcium-induced calmodulin conformational change and the influence of Ca2+ concentration on the electrochemical behaviour of CaM were evaluated, and significant differences, in the tyrosine amino acid residue peak potential and current, in the absence and in the presence of calcium ions, were observed. Gravimetric measurements were performed with a graphite coated piezoelectric quartz crystal with adsorbed CaM, and calcium aggregation by CaM was demonstrated.
Collapse
|
206
|
Cellular Stress and p53-Associated Apoptosis by Juniperus communis L. Berry Extract Treatment in the Human SH-SY5Y Neuroblastoma Cells. Int J Mol Sci 2016; 17:ijms17071113. [PMID: 27420050 PMCID: PMC4964488 DOI: 10.3390/ijms17071113] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/19/2022] Open
Abstract
Plant phenolics have shown to activate apoptotic cell death in different tumourigenic cell lines. In this study, we evaluated the effects of juniper berry extract (Juniperus communis L.) on p53 protein, gene expression and DNA fragmentation in human neuroblastoma SH-SY5Y cells. In addition, we analyzed the phenolic composition of the extract. We found that juniper berry extract activated cellular relocalization of p53 and DNA fragmentation-dependent cell death. Differentially expressed genes between treated and non-treated cells were evaluated with the cDNA-RDA (representational difference analysis) method at the early time point of apoptotic process when p53 started to be activated and no caspase activity was detected. Twenty one overexpressed genes related to cellular stress, protein synthesis, cell survival and death were detected. Interestingly, they included endoplasmic reticulum (ER) stress inducer and sensor HSPA5 and other ER stress-related genes CALM2 and YKT6 indicating that ER stress response was involved in juniper berry extract mediated cell death. In composition analysis, we identified and quantified low concentrations of fifteen phenolic compounds. The main groups of them were flavones, flavonols, phenolic acids, flavanol and biflavonoid including glycosides of quercetin, apigenin, isoscutellarein and hypolaetin. It is suggested that juniper berry extract induced the p53-associated apoptosis through the potentiation and synergism by several phenolic compounds.
Collapse
|
207
|
Kim DI, Kweon HJ, Park Y, Jang DJ, Suh BC. Ca2+ controls gating of voltage-gated calcium channels by releasing the β2e subunit from the plasma membrane. Sci Signal 2016; 9:ra67. [PMID: 27382026 DOI: 10.1126/scisignal.aad7247] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Voltage-gated calcium (Cav) channels, which are regulated by membrane potential, cytosolic Ca(2+), phosphorylation, and membrane phospholipids, govern Ca(2+) entry into excitable cells. Cav channels contain a pore-forming α1 subunit, an auxiliary α2δ subunit, and a regulatory β subunit, each encoded by several genes in mammals. In addition to a domain that interacts with the α1 subunit, β2e and β2a also interact with the cytoplasmic face of the plasma membrane through an electrostatic interaction for β2e and posttranslational acylation for β2a. We found that an increase in cytosolic Ca(2+) promoted the release of β2e from the membrane without requiring substantial depletion of the anionic phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) from the plasma membrane. Experiments with liposomes indicated that Ca(2+) disrupted the interaction of the β2e amino-terminal peptide with membranes containing PIP2 Ca(2+) binding to calmodulin (CaM) leads to CaM-mediated inactivation of Cav currents. Although Cav2.2 coexpressed with β2a required Ca(2+)-dependent activation of CaM for Ca(2+)-mediated reduction in channel activity, Cav2.2 coexpressed with β2e exhibited Ca(2+)-dependent inactivation of the channel even in the presence of Ca(2+)-insensitive CaM. Inducible depletion of PIP2 reduced Cav2.2 currents, and in cells coexpressing β2e, but not a form that lacks the polybasic region, increased intracellular Ca(2+) further reduced Cav2.2 currents. Many hormone- or neurotransmitter-activated receptors stimulate PIP2 hydrolysis and increase cytosolic Ca(2+); thus, our findings suggest that β2e may integrate such receptor-mediated signals to limit Cav activity.
Collapse
Affiliation(s)
- Dong-Il Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Hae-Jin Kweon
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Yongsoo Park
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Deok-Jin Jang
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Kyungbuk 742-711, Korea
| | - Byung-Chang Suh
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea.
| |
Collapse
|
208
|
Xing L, Martyniuk CJ, Esau C, Da Fonte DF, Trudeau VL. Proteomic profiling reveals dopaminergic regulation of progenitor cell functions of goldfish radial glial cells in vitro. J Proteomics 2016; 144:123-32. [DOI: 10.1016/j.jprot.2016.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 01/03/2023]
|
209
|
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
|
210
|
Vrenken KS, Jalink K, van Leeuwen FN, Middelbeek J. Beyond ion-conduction: Channel-dependent and -independent roles of TRP channels during development and tissue homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1436-46. [DOI: 10.1016/j.bbamcr.2015.11.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/26/2015] [Accepted: 11/11/2015] [Indexed: 01/09/2023]
|
211
|
Zhang DX, Zhang JP, Hu JY, Huang YS. The potential regulatory roles of NAD(+) and its metabolism in autophagy. Metabolism 2016; 65:454-62. [PMID: 26975537 DOI: 10.1016/j.metabol.2015.11.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/29/2015] [Accepted: 11/25/2015] [Indexed: 02/02/2023]
Abstract
(Macro)autophagy mediates the bulk degradation of defective organelles, long-lived proteins and protein aggregates in lysosomes and plays a critical role in cellular and tissue homeostasis. Defective autophagy processes have been found to contribute to a variety of metabolic diseases. However, the regulatory mechanisms of autophagy are not fully understood. Increasing data indicate that nicotinamide adenine nucleotide (NAD(+)) homeostasis correlates intimately with autophagy. NAD(+) is a ubiquitous coenzyme that functions primarily as an electron carrier of oxidoreductase in multiple redox reactions. Both NAD(+) homeostasis and its metabolism are thought to play critical roles in regulating autophagy. In this review, we discuss how the regulation of NAD(+) and its metabolism can influence autophagy. We focus on the regulation of NAD(+)/NADH homeostasis and the effects of NAD(+) consumption by poly(ADP-ribose) (PAR) polymerase-1 (PARP-1), NAD(+)-dependent deacetylation by sirtuins and NAD(+) metabolites on autophagy processes and the underlying mechanisms. Future studies should provide more direct evidence for the regulation of autophagy processes by NAD(+). A better understanding of the critical roles of NAD(+) and its metabolites on autophagy will shed light on the complexity of autophagy regulation, which is essential for the discovery of new therapeutic tools for autophagy-related diseases.
Collapse
Affiliation(s)
- Dong-Xia Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, PR China, 400038
| | - Jia-Ping Zhang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, PR China, 400038
| | - Jiong-Yu Hu
- Endocrinology Department, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, PR China, 400038
| | - Yue-Sheng Huang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, PR China, 400038.
| |
Collapse
|
212
|
Kibble M, Khan SA, Saarinen N, Iorio F, Saez-Rodriguez J, Mäkelä S, Aittokallio T. Transcriptional response networks for elucidating mechanisms of action of multitargeted agents. Drug Discov Today 2016; 21:1063-75. [PMID: 26979547 DOI: 10.1016/j.drudis.2016.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/15/2016] [Accepted: 03/04/2016] [Indexed: 01/08/2023]
Abstract
Drug discovery is moving away from the single target-based approach towards harnessing the potential of polypharmacological agents that modulate the activity of multiple nodes in the complex networks of deregulations underlying disease phenotypes. Computational network pharmacology methods that use systems-level drug-response phenotypes, such as those originating from genome-wide transcriptomic profiles, have proved particularly effective for elucidating the mechanisms of action of multitargeted compounds. Here, we show, via the case study of the natural product pinosylvin, how the combination of two complementary network-based methods can provide novel, unexpected mechanistic insights. This case study also illustrates that elucidating the mechanism of action of multitargeted natural products through transcriptional response-based approaches is a challenging endeavor, often requiring multiple computational-experimental iterations.
Collapse
Affiliation(s)
- Milla Kibble
- Institute for Molecular Medicine Finland (FIMM), Biomedicum Helsinki 2U, Tukholmankatu 8, University of Helsinki, Helsinki 00014, Finland.
| | - Suleiman A Khan
- Institute for Molecular Medicine Finland (FIMM), Biomedicum Helsinki 2U, Tukholmankatu 8, University of Helsinki, Helsinki 00014, Finland
| | - Niina Saarinen
- Institute of Biomedicine, Turku Center for Disease Modeling & Functional Foods Forum, University of Turku, Turku 20014, Finland
| | - Francesco Iorio
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, UK
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SD, UK; Joint Research Centre for Computational Biomedicine (JRC-COMBINE) - RWTH Aachen University, Faculty of Medicine, D-52074 Aachen, Germany
| | - Sari Mäkelä
- Institute of Biomedicine, Turku Center for Disease Modeling & Functional Foods Forum, University of Turku, Turku 20014, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), Biomedicum Helsinki 2U, Tukholmankatu 8, University of Helsinki, Helsinki 00014, Finland; Department of Mathematics and Statistics, Quantum, University of Turku, Turku 20014, Finland
| |
Collapse
|
213
|
Wu M, Kim SH, Datta I, Levin A, Dyson G, Li J, Kaypee S, Swamy MM, Gupta N, Kwon HJ, Menon M, Kundu TK, Reddy GPV. Hydrazinobenzoylcurcumin inhibits androgen receptor activity and growth of castration-resistant prostate cancer in mice. Oncotarget 2016; 6:6136-50. [PMID: 25704883 PMCID: PMC4467427 DOI: 10.18632/oncotarget.3346] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 01/20/2015] [Indexed: 01/09/2023] Open
Abstract
There is a critical need for therapeutic agents that can target the amino-terminal domain (NTD) of androgen receptor (AR) for the treatment of castration-resistant prostate cancer (CRPC). Calmodulin (CaM) binds to the AR NTD and regulates AR activity. We discovered that Hydrazinobenzoylcurcumin (HBC), which binds exclusively to CaM, inhibited AR activity. HBC abrogated AR interaction with CaM, suppressed phosphorylation of AR Serine81, and blocked the binding of AR to androgen-response elements. RNA-Seq analysis identified 57 androgen-regulated genes whose expression was significantly (p ≤ 0.002) altered in HBC treated cells as compared to controls. Oncomine analysis revealed that genes repressed by HBC are those that are usually overexpressed in prostate cancer (PCa) and genes stimulated by HBC are those that are often down-regulated in PCa, suggesting a reversing effect of HBC on androgen-regulated gene expression associated with PCa. Ingenuity Pathway Analysis revealed a role of HBC affected genes in cellular functions associated with proliferation and survival. HBC was readily absorbed into the systemic circulation and inhibited the growth of xenografted CRPC tumors in nude mice. These observations demonstrate that HBC inhibits AR activity by targeting the AR NTD and suggest potential usefulness of HBC for effective treatment of CRPC.
Collapse
Affiliation(s)
- Min Wu
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Sahn-Ho Kim
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Indrani Datta
- Bioinformatics Core, Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | - Albert Levin
- Bioinformatics Core, Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | - Gregory Dyson
- Biostatistics Core, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Jing Li
- Pharmacology Core, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Stephanie Kaypee
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, JNCASR, Bangalore, Karnataka, India
| | - M Mahadeva Swamy
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, JNCASR, Bangalore, Karnataka, India
| | - Nilesh Gupta
- Department of Pathology, Henry Ford Hospital, Detroit, MI, USA
| | - Ho Jeong Kwon
- Department of Biotechnology, Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea
| | - Mani Menon
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, JNCASR, Bangalore, Karnataka, India
| | | |
Collapse
|
214
|
Purification of F plasmid-encoded native TraC from Escherichia coli by affinity chromatography on calmodulin Sepharose. Protein Expr Purif 2016; 122:97-104. [PMID: 26892535 DOI: 10.1016/j.pep.2016.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 01/27/2016] [Accepted: 02/12/2016] [Indexed: 11/21/2022]
Abstract
We have enriched several native bacterial proteins from Escherichia coli by chromatography on the immobilized eukaryotic Ca(2+)-binding protein, calmodulin. These bacterial proteins bound in a Ca(2+)-dependent manner to calmodulin, and were released by the addition of the Ca(2+)-chelator, EGTA, similar to many eukaryotic calmodulin-binding proteins. One of the bacterial proteins, F factor-encoded TraC, was purified to apparent homogeneity by an additional chromatographic step, anion exchange chromatography on MonoQ. Experiments with four chemically distinct calmodulin antagonists (R24571, Compound 48/80, melittin, and W7) showed that all of these substances inhibited the binding of purified TraC to calmodulin at effective concentrations comparable to those required for inhibiting in vitro binding of eukaryotic calmodulin-binding proteins. Three further bacterial proteins were identified as calmodulin-binding proteins: SecA, GlpD, and GlpC. We suggest that also these native bacterial proteins might be isolated by the unusual purification procedure including affinity chromatography on calmodulin Sepharose. Whether the identified proteins bind to, and are regulated by, putative bacterial calmodulin-like proteins in Escherichia coli remains to be established.
Collapse
|
215
|
Nussinov R, Muratcioglu S, Tsai CJ, Jang H, Gursoy A, Keskin O. K-Ras4B/calmodulin/PI3Kα: A promising new adenocarcinoma-specific drug target? Expert Opin Ther Targets 2016; 20:831-42. [DOI: 10.1517/14728222.2016.1135131] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
- 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, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Hyunbum Jang
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, USA
| | - Attila Gursoy
- Department of Computer Engineering, Koc University, Istanbul, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| |
Collapse
|
216
|
Puniya BL, Allen L, Hochfelder C, Majumder M, Helikar T. Systems Perturbation Analysis of a Large-Scale Signal Transduction Model Reveals Potentially Influential Candidates for Cancer Therapeutics. Front Bioeng Biotechnol 2016; 4:10. [PMID: 26904540 PMCID: PMC4750464 DOI: 10.3389/fbioe.2016.00010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/25/2016] [Indexed: 12/20/2022] Open
Abstract
Dysregulation in signal transduction pathways can lead to a variety of complex disorders, including cancer. Computational approaches such as network analysis are important tools to understand system dynamics as well as to identify critical components that could be further explored as therapeutic targets. Here, we performed perturbation analysis of a large-scale signal transduction model in extracellular environments that stimulate cell death, growth, motility, and quiescence. Each of the model’s components was perturbed under both loss-of-function and gain-of-function mutations. Using 1,300 simulations under both types of perturbations across various extracellular conditions, we identified the most and least influential components based on the magnitude of their influence on the rest of the system. Based on the premise that the most influential components might serve as better drug targets, we characterized them for biological functions, housekeeping genes, essential genes, and druggable proteins. The most influential components under all environmental conditions were enriched with several biological processes. The inositol pathway was found as most influential under inactivating perturbations, whereas the kinase and small lung cancer pathways were identified as the most influential under activating perturbations. The most influential components were enriched with essential genes and druggable proteins. Moreover, known cancer drug targets were also classified in influential components based on the affected components in the network. Additionally, the systemic perturbation analysis of the model revealed a network motif of most influential components which affect each other. Furthermore, our analysis predicted novel combinations of cancer drug targets with various effects on other most influential components. We found that the combinatorial perturbation consisting of PI3K inactivation and overactivation of IP3R1 can lead to increased activity levels of apoptosis-related components and tumor-suppressor genes, suggesting that this combinatorial perturbation may lead to a better target for decreasing cell proliferation and inducing apoptosis. Finally, our approach shows a potential to identify and prioritize therapeutic targets through systemic perturbation analysis of large-scale computational models of signal transduction. Although some components of the presented computational results have been validated against independent gene expression data sets, more laboratory experiments are warranted to more comprehensively validate the presented results.
Collapse
Affiliation(s)
- Bhanwar Lal Puniya
- Department of Biochemistry, University of Nebraska-Lincoln , Lincoln, NE , USA
| | - Laura Allen
- Department of Mathematics, University of Nebraska at Omaha , Omaha, NE , USA
| | | | - Mahbubul Majumder
- Department of Mathematics, University of Nebraska at Omaha , Omaha, NE , USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln , Lincoln, NE , USA
| |
Collapse
|
217
|
McCammick EM, McVeigh P, McCusker P, Timson DJ, Morphew RM, Brophy PM, Marks NJ, Mousley A, Maule AG. Calmodulin disruption impacts growth and motility in juvenile liver fluke. Parasit Vectors 2016; 9:46. [PMID: 26817678 PMCID: PMC4730779 DOI: 10.1186/s13071-016-1324-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/19/2016] [Indexed: 12/15/2022] Open
Abstract
Background Deficiencies in effective flukicide options and growing issues with drug resistance make current strategies for liver fluke control unsustainable, thereby promoting the need to identify and validate new control targets in Fasciola spp. parasites. Calmodulins (CaMs) are small calcium-sensing proteins with ubiquitous expression in all eukaryotic organisms and generally use fluctuations in intracellular calcium levels to modulate cell signalling events. CaMs are essential for fundamental processes including the phosphorylation of protein kinases, gene transcription, calcium transport and smooth muscle contraction. In the blood fluke Schistosoma mansoni, calmodulins have been implicated in egg hatching, miracidial transformation and larval development. Previously, CaMs have been identified amongst liver fluke excretory-secretory products and three CaM-like proteins have been characterised biochemically from adult Fasciola hepatica, although their functions remain unknown. Methods In this study, we set out to investigate the biological function and control target potential of F. hepatica CaMs (FhCaMs) using RNAi methodology alongside novel in vitro bioassays. Results Our results reveal that: (i) FhCaMs are widely expressed in parenchymal cells throughout the forebody region of juvenile fluke; (ii) significant transcriptional knockdown of FhCaM1-3 was inducible by exposure to either long (~200 nt) double stranded (ds) RNAs or 27 nt short interfering (si) RNAs, although siRNAs were less effective than long dsRNAs; (iii) transient long dsRNA exposure-induced RNA interference (RNAi) of FhCaMs triggered transcript knockdown that persisted for ≥ 21 days, and led to detectable suppression of FhCaM proteins; (iv) FhCaM RNAi significantly reduced the growth of juvenile flukes maintained in vitro; (v) FhCaM RNAi juveniles also displayed hyperactivity encompassing significantly increased migration; (vi) both the reduced growth and increased motility phenotypes were recapitulated in juvenile fluke using the CaM inhibitor trifluoperazine hydrochloride, supporting phenotype specificity. Conclusions These data indicate that the Ca2+-modulating functions of FhCaMs are important for juvenile fluke growth and movement and provide the first functional genomics-based example of a growth-defect resulting from gene silencing in liver fluke. Whilst the phenotypic impacts of FhCaM silencing on fluke behaviour do not strongly support their candidature as new flukicide targets, the growth impacts encourage further consideration, especially in light of the speed of juvenile fluke growth in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1324-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erin M McCammick
- Microbes & Pathogen Biology: Institute for Global Food Security, School of Biological Sciences, Queen's, University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Paul McVeigh
- Microbes & Pathogen Biology: Institute for Global Food Security, School of Biological Sciences, Queen's, University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Paul McCusker
- Microbes & Pathogen Biology: Institute for Global Food Security, School of Biological Sciences, Queen's, University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - David J Timson
- Microbes & Pathogen Biology: Institute for Global Food Security, School of Biological Sciences, Queen's, University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Russell M Morphew
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Penglais, Aberystwyth, Ceredigion, SY23 3FL, UK.
| | - Peter M Brophy
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Penglais, Aberystwyth, Ceredigion, SY23 3FL, UK.
| | - Nikki J Marks
- Microbes & Pathogen Biology: Institute for Global Food Security, School of Biological Sciences, Queen's, University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Angela Mousley
- Microbes & Pathogen Biology: Institute for Global Food Security, School of Biological Sciences, Queen's, University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Aaron G Maule
- Microbes & Pathogen Biology: Institute for Global Food Security, School of Biological Sciences, Queen's, University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
218
|
Identification of the Calmodulin-Binding Domains of Fas Death Receptor. PLoS One 2016; 11:e0146493. [PMID: 26735300 PMCID: PMC4703387 DOI: 10.1371/journal.pone.0146493] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/17/2015] [Indexed: 01/25/2023] Open
Abstract
The extrinsic apoptotic pathway is initiated by binding of a Fas ligand to the ectodomain of the surface death receptor Fas protein. Subsequently, the intracellular death domain of Fas (FasDD) and that of the Fas-associated protein (FADD) interact to form the core of the death-inducing signaling complex (DISC), a crucial step for activation of caspases that induce cell death. Previous studies have shown that calmodulin (CaM) is recruited into the DISC in cholangiocarcinoma cells and specifically interacts with FasDD to regulate the apoptotic/survival signaling pathway. Inhibition of CaM activity in DISC stimulates apoptosis significantly. We have recently shown that CaM forms a ternary complex with FasDD (2:1 CaM:FasDD). However, the molecular mechanism by which CaM binds to two distinct FasDD motifs is not fully understood. Here, we employed mass spectrometry, nuclear magnetic resonance (NMR), biophysical, and biochemical methods to identify the binding regions of FasDD and provide a molecular basis for the role of CaM in Fas-mediated apoptosis. Proteolytic digestion and mass spectrometry data revealed that peptides spanning residues 209-239 (Fas-Pep1) and 251-288 (Fas-Pep2) constitute the two CaM-binding regions of FasDD. To determine the molecular mechanism of interaction, we have characterized the binding of recombinant/synthetic Fas-Pep1 and Fas-Pep2 peptides with CaM. Our data show that both peptides engage the N- and C-terminal lobes of CaM simultaneously. Binding of Fas-Pep1 to CaM is entropically driven while that of Fas-Pep2 to CaM is enthalpically driven, indicating that a combination of electrostatic and hydrophobic forces contribute to the stabilization of the FasDD-CaM complex. Our data suggest that because Fas-Pep1 and Fas-Pep2 are involved in extensive intermolecular contacts with the death domain of FADD, binding of CaM to these regions may hinder its ability to bind to FADD, thus greatly inhibiting the initiation of apoptotic signaling pathway.
Collapse
|
219
|
|
220
|
Frandsen SK, Gibot L, Madi M, Gehl J, Rols MP. Calcium Electroporation: Evidence for Differential Effects in Normal and Malignant Cell Lines, Evaluated in a 3D Spheroid Model. PLoS One 2015; 10:e0144028. [PMID: 26633834 PMCID: PMC4669124 DOI: 10.1371/journal.pone.0144028] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/12/2015] [Indexed: 12/21/2022] Open
Abstract
Background Calcium electroporation describes the use of high voltage electric pulses to introduce supraphysiological calcium concentrations into cells. This promising method is currently in clinical trial as an anti-cancer treatment. One very important issue is the relation between tumor cell kill efficacy–and normal cell sensitivity. Methods Using a 3D spheroid cell culture model we have tested the effect of calcium electroporation and electrochemotherapy using bleomycin on three different human cancer cell lines: a colorectal adenocarcinoma (HT29), a bladder transitional cell carcinoma (SW780), and a breast adenocarcinoma (MDA-MB231), as well as on primary normal human dermal fibroblasts (HDF-n). Results The results showed a clear reduction in spheroid size in all three cancer cell spheroids three days after treatment with respectively calcium electroporation (p<0.0001) or electrochemotherapy using bleomycin (p<0.0001). Strikingly, the size of normal fibroblast spheroids was neither affected after calcium electroporation nor electrochemotherapy using bleomycin, indicating that calcium electroporation, like electrochemotherapy, will have limited adverse effects on the surrounding normal tissue when treating with calcium electroporation. The intracellular ATP level, which has previously been shown to be depleted after calcium electroporation, was measured in the spheroids after treatment. The results showed a dramatic decrease in the intracellular ATP level (p<0.01) in all four spheroid types—malignant as well as normal. Conclusion In conclusion, calcium electroporation seems to be more effective in inducing cell death in cancer cell spheroids than in a normal fibroblast spheroid, even though intracellular ATP level is depleted in all spheroid types after treatment. These results may indicate an important therapeutic window for this therapy; although further studies are needed in vivo and in patients to investigate the effect of calcium electroporation on surrounding normal tissue when treating tumors.
Collapse
Affiliation(s)
- Stine Krog Frandsen
- Center for Experimental Drug and Gene Electrotransfer, Department of Oncology, Copenhagen University Hospital Herlev, 2730 Herlev, Denmark
| | - Laure Gibot
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
| | - Moinecha Madi
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
| | - Julie Gehl
- Center for Experimental Drug and Gene Electrotransfer, Department of Oncology, Copenhagen University Hospital Herlev, 2730 Herlev, Denmark
- * E-mail: (JG); (MPR)
| | - Marie-Pierre Rols
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, UPS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- * E-mail: (JG); (MPR)
| |
Collapse
|
221
|
The activating role of phospho-(Tyr)-calmodulin on the epidermal growth factor receptor. Biochem J 2015; 472:195-204. [DOI: 10.1042/bj20150851] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/22/2015] [Indexed: 02/06/2023]
Abstract
The existence of a calmodulin (CaM)/phospho-(Tyr)-CaM cycle involved in the regulation of the epidermal growth factor receptor could have important consequences for the control of cell proliferation, as its alteration could potentially result in uncontrolled tumour growth.
Collapse
|
222
|
Szabo M, Dulka K, Gulya K. Calmodulin inhibition regulates morphological and functional changes related to the actin cytoskeleton in pure microglial cells. Brain Res Bull 2015; 120:41-57. [PMID: 26551061 DOI: 10.1016/j.brainresbull.2015.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/26/2015] [Accepted: 11/03/2015] [Indexed: 01/24/2023]
Abstract
The roles of calmodulin (CaM), a multifunctional intracellular calcium receptor protein, as concerns selected morphological and functional characteristics of pure microglial cells derived from mixed primary cultures from embryonal forebrains of rats, were investigated through use of the CaM antagonists calmidazolium (CALMID) and trifluoperazine (TFP). The intracellular localization of the CaM protein relative to phalloidin, a bicyclic heptapeptide that binds only to filamentous actin, and the ionized calcium-binding adaptor molecule 1 (Iba1), a microglia-specific actin-binding protein, was determined by immunocytochemistry, with quantitative analysis by immunoblotting. In unchallenged and untreated (control) microglia, high concentrations of CaM protein were found mainly perinuclearly in ameboid microglia, while the cell cortex had a smaller CaM content that diminished progressively deeper into the branches in the ramified microglia. The amounts and intracellular distributions of both Iba1 and CaM proteins were altered after lipopolysaccharide (LPS) challenge in activated microglia. CALMID and TFP exerted different, sometimes opposing, effects on many morphological, cytoskeletal and functional characteristics of the microglial cells. They affected the CaM and Iba1 protein expressions and their intracellular localizations differently, inhibited cell proliferation, viability and fluid-phase phagocytosis to different degrees both in unchallenged and in LPS-treated (immunologically challenged) cells, and differentially affected the reorganization of the actin cytoskeleton in the microglial cell cortex, influencing lamellipodia, filopodia and podosome formation. In summary, these CaM antagonists altered different aspects of filamentous actin-based cell morphology and related functions with variable efficacy, which could be important in deciphering the roles of CaM in regulating microglial functions in health and disease.
Collapse
Affiliation(s)
- Melinda Szabo
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Karolina Dulka
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Karoly Gulya
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary.
| |
Collapse
|
223
|
Peng H, Wu X, Zhao L, Feng Y. Dynamic analysis of phospholipid metabolism of mouse macrophages treated with common non-steroidal anti-inflammatory drugs. Mol Cell Biochem 2015; 411:161-71. [DOI: 10.1007/s11010-015-2578-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/26/2015] [Indexed: 11/28/2022]
|
224
|
Kulkarni C, Lo M, Fraseur JG, Tirrell DA, Kinzer-Ursem TL. Bioorthogonal Chemoenzymatic Functionalization of Calmodulin for Bioconjugation Applications. Bioconjug Chem 2015; 26:2153-60. [PMID: 26431265 DOI: 10.1021/acs.bioconjchem.5b00449] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Calmodulin (CaM) is a widely studied Ca(2+)-binding protein that is highly conserved across species and involved in many biological processes, including vesicle release, cell proliferation, and apoptosis. To facilitate biophysical studies of CaM, researchers have tagged and mutated CaM at various sites, enabling its conjugation to fluorophores, microarrays, and other reactive partners. However, previous attempts to add a reactive label to CaM for downstream studies have generally employed nonselective labeling methods or resulted in diminished CaM function. Here we report the first engineered CaM protein that undergoes site-specific and bioorthogonal labeling while retaining wild-type activity levels. By employing a chemoenzymatic labeling approach, we achieved selective and quantitative labeling of the engineered CaM protein with an N-terminal 12-azidododecanoic acid tag; notably, addition of the tag did not interfere with the ability of CaM to bind Ca(2+) or a partner protein. The specificity of our chemoenzymatic labeling approach also allowed for selective conjugation of CaM to reactive partners in bacterial cell lysates, without intermediate purification of the engineered protein. Additionally, we prepared CaM-affinity resins that were highly effective in purifying a representative CaM-binding protein, demonstrating that the engineered CaM remains active even after surface capture. Beyond studies of CaM and CaM-binding proteins, the protein engineering and surface capture methods described here should be translatable to other proteins and other bioconjugation applications.
Collapse
Affiliation(s)
- Chethana Kulkarni
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Megan Lo
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Julia G Fraseur
- Weldon School of Biomedical Engineering, Purdue University , 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| | - David A Tirrell
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Tamara L Kinzer-Ursem
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States.,Weldon School of Biomedical Engineering, Purdue University , 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
225
|
Tripathi S, Waxham MN, Cheung MS, Liu Y. Lessons in Protein Design from Combined Evolution and Conformational Dynamics. Sci Rep 2015; 5:14259. [PMID: 26388515 PMCID: PMC4585694 DOI: 10.1038/srep14259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/21/2015] [Indexed: 11/09/2022] Open
Abstract
Protein-protein interactions play important roles in the control of every cellular process. How natural selection has optimized protein design to produce molecules capable of binding to many partner proteins is a fascinating problem but not well understood. Here, we performed a combinatorial analysis of protein sequence evolution and conformational dynamics to study how calmodulin (CaM), which plays essential roles in calcium signaling pathways, has adapted to bind to a large number of partner proteins. We discovered that amino acid residues in CaM can be partitioned into unique classes according to their degree of evolutionary conservation and local stability. Holistically, categorization of CaM residues into these classes reveals enriched physico-chemical interactions required for binding to diverse targets, balanced against the need to maintain the folding and structural modularity of CaM to achieve its overall function. The sequence-structure-function relationship of CaM provides a concrete example of the general principle of protein design. We have demonstrated the synergy between the fields of molecular evolution and protein biophysics and created a generalizable framework broadly applicable to the study of protein-protein interactions.
Collapse
Affiliation(s)
- Swarnendu Tripathi
- Department of Physics, University of Houston, Houston, TX.,Center for Theoretical Biological Physics, Rice University, Houston, TX
| | - M Neal Waxham
- Department of Neurobiology and Anatomy, University of Texas, Health Science Center, Houston, TX
| | - Margaret S Cheung
- Department of Physics, University of Houston, Houston, TX.,Center for Theoretical Biological Physics, Rice University, Houston, TX
| | - Yin Liu
- Department of Neurobiology and Anatomy, University of Texas, Health Science Center, Houston, TX
| |
Collapse
|
226
|
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
|
227
|
Thermodynamics of Calcium binding to the Calmodulin N-terminal domain to evaluate site-specific affinity constants and cooperativity. J Biol Inorg Chem 2015; 20:905-19. [DOI: 10.1007/s00775-015-1275-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 05/31/2015] [Indexed: 10/23/2022]
|
228
|
Stateva SR, Salas V, Anguita E, Benaim G, Villalobo A. Ca2+/Calmodulin and Apo-Calmodulin Both Bind to and Enhance the Tyrosine Kinase Activity of c-Src. PLoS One 2015; 10:e0128783. [PMID: 26058065 PMCID: PMC4461253 DOI: 10.1371/journal.pone.0128783] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/30/2015] [Indexed: 12/19/2022] Open
Abstract
Src family non-receptor tyrosine kinases play a prominent role in multiple cellular processes, including: cell proliferation, differentiation, cell survival, stress response, and cell adhesion and migration, among others. And when deregulated by mutations, overexpression, and/or the arrival of faulty incoming signals, its hyperactivity contributes to the development of hematological and solid tumors. c-Src is a prototypical member of this family of kinases, which is highly regulated by a set of phosphorylation events. Other factor contributing to the regulation of Src activity appears to be mediated by the Ca2+ signal generated in cells by different effectors, where the Ca2+-receptor protein calmodulin (CaM) plays a key role. In this report we demonstrate that CaM directly interacts with Src in both Ca2+-dependent and Ca2+-independent manners in vitro and in living cells, and that the CaM antagonist N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide (W-7) inhibits the activation of this kinase induced by the upstream activation of the epidermal growth factor receptor (EGFR), in human carcinoma epidermoide A431 cells, and by hydrogen peroxide-induced oxidative stress, in both A431 cells and human breast adenocarcinoma SK-BR-3 cells. Furthermore, we show that the Ca2+/CaM complex strongly activates the auto-phosphorylation and tyrosine kinase activity of c-Src toward exogenous substrates, but most relevantly and for the first time, we demonstrate that Ca2+-free CaM (apo-CaM) exerts a far higher activatory action on Src auto-phosphorylation and kinase activity toward exogenous substrates than the one exerted by the Ca2+/CaM complex. This suggests that a transient increase in the cytosolic concentration of free Ca2+ is not an absolute requirement for CaM-mediated activation of Src in living cells, and that a direct regulation of Src by apo-CaM could be inferred.
Collapse
Affiliation(s)
- Silviya R. Stateva
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029, Madrid, Spain
| | - Valentina Salas
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029, Madrid, Spain
- Instituto de Biología Experimental, Facultad de Ciencias, Universidad Central de Venezuela, Caracas, Venezuela
| | - Estefanía Anguita
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029, Madrid, Spain
| | - Gustavo Benaim
- Instituto de Biología Experimental, Facultad de Ciencias, Universidad Central de Venezuela, Caracas, Venezuela
- Instituto de Estudios Avanzados (IDEA), Caracas, Venezuela
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029, Madrid, Spain
- * E-mail:
| |
Collapse
|
229
|
Rai J, Pemmasani JK, Voronovsky A, Jensen IS, Manavalan A, Nyengaard JR, Golas MM, Sander B. Strep-tag II and Twin-Strep based cassettes for protein tagging by homologous recombination and characterization of endogenous macromolecular assemblies in Saccharomyces cerevisiae. Mol Biotechnol 2015; 56:992-1003. [PMID: 24969434 DOI: 10.1007/s12033-014-9778-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Peptide sequences fused to a gene of interest facilitate the isolation of proteins or protein complexes from cell extracts. In the case of fluorescent protein tags, the tagged protein can be visually localized in living cells. To tag endogenous genes, PCR-based homologous recombination is a powerful approach used in the yeast Saccharomyces cerevisiae. This approach uses short, homologous DNA sequences that flank the tagging cassette to direct recombination. Here, we constructed a set of plasmids, whose sequences were optimized for codon usage in yeast, for Strep-tag II and Twin-Strep tagging in S. cerevisiae. Some plasmids also contain sequences encoding for a fluorescent protein followed by the purification tag. We demonstrate using the yeast pyruvate dehydrogenase (PDH) complex that these plasmids can be used to purify large protein complexes efficiently. We furthermore demonstrate that purification from the endogenous pool using the Strep-tag system results in functionally active complexes. Finally, using the fluorescent tags, we show that a kinase and a phosphatase involved in regulating the activity of the PDH complex localize in the cells' mitochondria. In conclusion, our cassettes can be used as tools for biochemical, functional, and structural analyses of endogenous multi-protein assemblies in yeast.
Collapse
Affiliation(s)
- Jay Rai
- Stereology and EM Laboratory, Department of Clinical Medicine, Institute of Clinical Medicine, Aarhus University, c/o Wilhelm Meyers Allé 3, Building 1233/1234, 8000, Aarhus C, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
230
|
The calmodulin inhibitor CGS 9343B inhibits voltage-dependent K + channels in rabbit coronary arterial smooth muscle cells. Toxicol Appl Pharmacol 2015; 285:207-13. [DOI: 10.1016/j.taap.2015.03.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/05/2015] [Accepted: 03/10/2015] [Indexed: 11/18/2022]
|
231
|
Huang YS, Hsieh TJ, Lu CY. Simple analytical strategy for MALDI-TOF-MS and nanoUPLC–MS/MS: Quantitating curcumin in food condiments and dietary supplements and screening of acrylamide-induced ROS protein indicators reduced by curcumin. Food Chem 2015; 174:571-6. [DOI: 10.1016/j.foodchem.2014.11.115] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 08/14/2014] [Accepted: 11/11/2014] [Indexed: 10/24/2022]
|
232
|
Fearnley GW, Bruns AF, Wheatcroft SB, Ponnambalam S. VEGF-A isoform-specific regulation of calcium ion flux, transcriptional activation and endothelial cell migration. Biol Open 2015; 4:731-42. [PMID: 25910937 PMCID: PMC4467193 DOI: 10.1242/bio.201410884] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) regulates many aspects of vascular physiology such as cell migration, proliferation, tubulogenesis and cell-cell interactions. Numerous isoforms of VEGF-A exist but their physiological significance is unclear. Here we evaluated two different VEGF-A isoforms and discovered differential regulation of cytosolic calcium ion flux, transcription factor localisation and endothelial cell response. Analysis of VEGF-A isoform-specific stimulation of VEGFR2-dependent signal transduction revealed differential capabilities for isoform activation of multiple signal transduction pathways. VEGF-A165 treatment promoted increased phospholipase Cγ1 phosphorylation, which was proportional to the subsequent rise in cytosolic calcium ions, in comparison to cells treated with VEGF-A121. A major consequence of this VEGF-A isoform-specific calcium ion flux in endothelial cells is differential dephosphorylation and subsequent nuclear translocation of the transcription factor NFATc2. Using reverse genetics, we discovered that NFATc2 is functionally required for VEGF-A-stimulated endothelial cell migration but not tubulogenesis. This work presents a new mechanism for understanding how VEGF-A isoforms program complex cellular outputs by converting signal transduction pathways into transcription factor redistribution to the nucleus, as well as defining a novel role for NFATc2 in regulating the endothelial cell response.
Collapse
Affiliation(s)
- Gareth W Fearnley
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Alexander F Bruns
- Division of Cardiovascular & Diabetes Research, Faculty of Medicine & Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Stephen B Wheatcroft
- Division of Cardiovascular & Diabetes Research, Faculty of Medicine & Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Sreenivasan Ponnambalam
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
233
|
Bonnefond ML, Lambert B, Giffard F, Abeilard E, Brotin E, Louis MH, Gueye MS, Gauduchon P, Poulain L, N’Diaye M. Calcium signals inhibition sensitizes ovarian carcinoma cells to anti-Bcl-xL strategies through Mcl-1 down-regulation. Apoptosis 2015; 20:535-50. [PMID: 25627260 PMCID: PMC4348506 DOI: 10.1007/s10495-015-1095-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ovarian carcinoma is the leading cause of death from gynecologic cancer in the developed world and is characterized by acquired chemoresistance leading to an overall 5-year survival rate of about 30 %. We previously showed that Bcl-xL and Mcl-1 cooperatively protect platinum-resistant ovarian cancer cells from apoptosis. Despite BH3-mimetics represent promising drugs to target Bcl-xL, anti-Mcl-1 strategies are still in pre-clinical studies and required new investigations. Calcium is a universal second messenger and dysregulation of calcium signal is often observed during carcinogenesis. As change in cytosolic free calcium concentration [Ca(2+)]i is known to control the fate of the cell by regulating Bcl-2 family members, we wonder if calcium signal could impact on Mcl-1 expression and if its pharmacological inhibition could be useful to sensitize ovarian carcinoma cells to anti-Bcl-xL strategies. We therefore studied the effect of different calcium signals inhibitors in ovarian carcinoma cell lines SKOV3 and IGROV1-R10 and analysed their effects on proliferation and Mcl-1 expression. We also exposed these cells to these inhibitors in combination with anti-Bcl-xL strategies (siRNA or BH3-mimetic: ABT-737). We found that calcium signaling regulates Mcl-1 through translational events and a calmodulin-mediated pathway. BAPTA-AM and calmodulin inhibitor combination with ABT-737 leads to apoptosis, a process that is reversed by Mcl-1 enforced expression. As Mcl-1 represents a crucial hurdle to the success of chemotherapy, these results could open to new area of investigation using calcium modulators to directly or indirectly target Mcl-1 and thus efficiently sensitize ovarian carcinoma cells to anti-Bcl-xL strategies.
Collapse
Affiliation(s)
- Marie-Laure Bonnefond
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
| | - Bernard Lambert
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
- CNRS (placed at the disposition of EA4656 by CNRS), Délégation régionale Ile-de-France Est, 94532 Thiais Cedex, France
| | - Florence Giffard
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
| | - Edwige Abeilard
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
| | - Emilie Brotin
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
| | - Marie-Hélène Louis
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
| | - Mor Sény Gueye
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
| | - Pascal Gauduchon
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
| | - Laurent Poulain
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
| | - Monique N’Diaye
- Normandy University, Caen, France
- UNICAEN, INSERM U1199 “Biology and Innovative Therapeutics of Locally Aggressive Cancers” Unit, Caen, France
- François Baclesse Comprehensive Cancer Center, 3 Avenue du Général Harris, BP5026, 14076 Caen Cedex 05, France
| |
Collapse
|
234
|
Stateva SR, Salas V, Benaim G, Menéndez M, Solís D, Villalobo A. Characterization of phospho-(tyrosine)-mimetic calmodulin mutants. PLoS One 2015; 10:e0120798. [PMID: 25830911 PMCID: PMC4382182 DOI: 10.1371/journal.pone.0120798] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/06/2015] [Indexed: 11/18/2022] Open
Abstract
Calmodulin (CaM) phosphorylated at different serine/threonine and tyrosine residues is known to exert differential regulatory effects on a variety of CaM-binding enzymes as compared to non-phosphorylated CaM. In this report we describe the preparation and characterization of a series of phospho-(Y)-mimetic CaM mutants in which either one or the two tyrosine residues present in CaM (Y99 and Y138) were substituted to aspartic acid or glutamic acid. It was expected that the negative charge of the respective carboxyl group of these amino acids mimics the negative charge of phosphate and reproduce the effects that distinct phospho-(Y)-CaM species may have on target proteins. We describe some physicochemical properties of these CaM mutants as compared to wild type CaM, after their expression in Escherichia coli and purification to homogeneity, including: i) changes in their electrophoretic mobility in the absence and presence of Ca2+; ii) ultraviolet (UV) light absorption spectra, far- and near-UV circular dichroism data; iii) thermal stability in the absence and presence of Ca2+; and iv) Tb3+-emitted fluorescence upon tyrosine excitation. We also describe some biochemical properties of these CaM mutants, such as their differential phosphorylation by the tyrosine kinase c-Src, and their action as compared to wild type CaM, on the activity of two CaM-dependent enzymes: cyclic nucleotide phosphodiesterase 1 (PDE1) and endothelial nitric oxide synthase (eNOS) assayed in vitro.
Collapse
Affiliation(s)
- Silviya R. Stateva
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Valentina Salas
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
- Universidad Central de Venezuela, Facultad de Ciencias, Instituto de Biología Experimental, Caracas, Venezuela
| | - Gustavo Benaim
- Universidad Central de Venezuela, Facultad de Ciencias, Instituto de Biología Experimental, Caracas, Venezuela
- Instituto de Estudios Avanzados (IDEA), Caracas, Venezuela
| | - Margarita Menéndez
- Instituto de Química Física Rocasolano, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Dolores Solís
- Instituto de Química Física Rocasolano, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Antonio Villalobo
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
235
|
Mata R, Figueroa M, González-Andrade M, Rivera-Chávez JA, Madariaga-Mazón A, Del Valle P. Calmodulin inhibitors from natural sources: an update. JOURNAL OF NATURAL PRODUCTS 2015; 78:576-586. [PMID: 25536331 DOI: 10.1021/np500954x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Calmodulin (CaM) plays a central role in regulating a myriad of cellular functions in physiological and pathophysiological processes, thus representing an important drug target. In previous reviews, our group has reported relevant information regarding natural anti-CaM compounds up to 2009. Natural sources continue to provide a diverse and unique reservoir of CaM inhibitors for drug and research tool discovery. This review provides an update of natural products with reported CaM inhibitory properties, which includes around 70 natural products and some synthetic analogues, belonging to different structural classes. Most of these natural inhibitors were isolated from fungi and plants and belong to the stilbenoid, polyketide, alkaloid, and peptide structural classes. These products were discovered mainly using a fluorescence-based method on rationally designed biosensors, which are highly specific, low-cost, and selective and have short reaction times. The effect of several antimitotic drugs on Ca(2+)-hCaM is also described.
Collapse
Affiliation(s)
- Rachel Mata
- †Facultad de Química and ‡Facultad de Medicina, Universidad Nacional Autónoma de México, México DF 04510, Mexico
| | - Mario Figueroa
- †Facultad de Química and ‡Facultad de Medicina, Universidad Nacional Autónoma de México, México DF 04510, Mexico
| | - Martín González-Andrade
- †Facultad de Química and ‡Facultad de Medicina, Universidad Nacional Autónoma de México, México DF 04510, Mexico
| | - José Alberto Rivera-Chávez
- †Facultad de Química and ‡Facultad de Medicina, Universidad Nacional Autónoma de México, México DF 04510, Mexico
| | - Abraham Madariaga-Mazón
- †Facultad de Química and ‡Facultad de Medicina, Universidad Nacional Autónoma de México, México DF 04510, Mexico
| | - Paulina Del Valle
- †Facultad de Química and ‡Facultad de Medicina, Universidad Nacional Autónoma de México, México DF 04510, Mexico
| |
Collapse
|
236
|
Li H, Choi IW, Hong DH, Son YK, Na SH, Jung WK, Firth AL, Jung ID, Park YM, Park WS. W-7 inhibits voltage-dependent K(+) channels independent of calmodulin activity in rabbit coronary arterial smooth muscle cells. Eur J Pharmacol 2015; 750:14-9. [PMID: 25617796 DOI: 10.1016/j.ejphar.2014.12.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 12/02/2014] [Accepted: 12/20/2014] [Indexed: 10/24/2022]
Abstract
We investigated the effect of W-7, a calmodulin inhibitor, on voltage-dependent K(+) (Kv) channels in freshly isolated coronary arterial smooth muscle cells using the whole-cell patch clamp technique. The amplitude of Kv currents was inhibited by W-7 in a concentration-dependent manner, with an IC50 value of 3.38±0.47μM and a Hill coefficient of 0.84±0.10. W-7 shifted the activation curve to a more positive potential but had no significant effect on the inactivation curve, which indicated that W-7 inhibited the Kv current in a closed state of the Kv channel. Another calmodulin inhibitor, W-13, had no significant effect on Kv currents and did not change the inhibitory effect of W-7 on Kv channels. From these results, we conclude that W-7 inhibited the Kv current in a dose-dependent manner, but this inhibition occurred independent of calmodulin activity and in a closed (inactivated) state of the Kv channels.
Collapse
Affiliation(s)
- Hongliang Li
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South Korea
| | - Il-Whan Choi
- Department of Microbiology, Inje University College of Medicine, Busan 614-735, South Korea
| | - Da Hye Hong
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South Korea
| | - Youn Kyoung Son
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South Korea
| | - Sung Hun Na
- Institute of Medical Sciences, Department of Obstetrics and Gynecology, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Won-Kyo Jung
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus) Pukyong National University, Busan 608-737, South Korea
| | - Amy L Firth
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - In Duk Jung
- Department of Immunology, Lab of Dendritic Cell Differentiation and Regulation, College of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Yeong-Min Park
- Department of Immunology, Lab of Dendritic Cell Differentiation and Regulation, College of Medicine, Konkuk University, Chungju 380-701, South Korea.
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 200-701, South Korea.
| |
Collapse
|
237
|
|
238
|
Abstract
The small, calcium-sensor protein, calmodulin, is ubiquitously expressed and central to cell function in all cell types. Here the literature linking calmodulin to Alzheimer's disease is reviewed. Several experimentally-verified calmodulin-binding proteins are involved in the formation of amyloid-β plaques including amyloid-β protein precursor, β-secretase, presenilin-1, and ADAM10. Many others possess potential calmodulin-binding domains that remain to be verified. Three calmodulin binding proteins are associated with the formation of neurofibrillary tangles: two kinases (CaMKII, CDK5) and one protein phosphatase (PP2B or calcineurin). Many of the genes recently identified by genome wide association studies and other studies encode proteins that contain putative calmodulin-binding domains but only a couple (e.g., APOE, BIN1) have been experimentally confirmed as calmodulin binding proteins. At least two receptors involved in calcium metabolism and linked to Alzheimer's disease (mAchR; NMDAR) have also been identified as calmodulin-binding proteins. In addition to this, many proteins that are involved in other cellular events intimately associated with Alzheimer's disease including calcium channel function, cholesterol metabolism, neuroinflammation, endocytosis, cell cycle events, and apoptosis have been tentatively or experimentally verified as calmodulin binding proteins. The use of calmodulin as a potential biomarker and as a therapeutic target is discussed.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto at Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Kristeen Eshak
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Michael A. Myre
- Center for Human Genetic Research, Richard B. Simches Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
239
|
Astegno A, Maresi E, Marino V, Dominici P, Pedroni M, Piccinelli F, Dell'Orco D. Structural plasticity of calmodulin on the surface of CaF2 nanoparticles preserves its biological function. NANOSCALE 2014; 6:15037-47. [PMID: 25367003 DOI: 10.1039/c4nr04368e] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Nanoparticles are increasingly used in biomedical applications and are especially attractive as biocompatible and biodegradable protein delivery systems. Herein, the interaction between biocompatible 25 nm CaF2 nanoparticles and the ubiquitous calcium sensor calmodulin has been investigated in order to assess the potential of these particles to serve as suitable surface protein carriers. Calmodulin is a multifunctional messenger protein that activates a wide variety of signaling pathways in eukaryotic cells by changing its conformation in a calcium-dependent manner. Isothermal titration calorimetry and circular dichroism studies have shown that the interaction between calmodulin and CaF2 nanoparticles occurs with physiologically relevant affinity and that the binding process is fully reversible, occurring without significant alterations in protein secondary and tertiary structures. Experiments performed with a mutant form of calmodulin having an impaired Ca(2+)-binding ability in the C-terminal lobe suggest that the EF-hand Ca(2+)-binding motifs are directly involved in the binding of calmodulin to the CaF2 matrix. The residual capability of nanoparticle-bound calmodulin to function as a calcium sensor protein, binding to and altering the activity of a target protein, was successfully probed by biochemical assays. Even if efficiently carried by CaF2 nanoparticles, calmodulin may dissociate, thus retaining the ability to bind the peptide encompassing the putative C-terminal calmodulin-binding domain of glutamate decarboxylase and activate the enzyme. We conclude that the high flexibility and structural plasticity of calmodulin are responsible for the preservation of its function when bound in high amounts to a nanoparticle surface.
Collapse
|
240
|
Yokokura S, Yurimoto S, Matsuoka A, Imataki O, Dobashi H, Bandoh S, Matsunaga T. Calmodulin antagonists induce cell cycle arrest and apoptosis in vitro and inhibit tumor growth in vivo in human multiple myeloma. BMC Cancer 2014; 14:882. [PMID: 25424011 PMCID: PMC4258255 DOI: 10.1186/1471-2407-14-882] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/19/2014] [Indexed: 02/02/2023] Open
Abstract
Background Human multiple myeloma (MM) is an incurable hematological malignancy for which novel therapeutic agents are needed. Calmodulin (CaM) antagonists have been reported to induce apoptosis and inhibit tumor cell invasion and metastasis in various tumor models. However, the antitumor effects of CaM antagonists on MM are poorly understood. In this study, we investigated the antitumor effects of naphthalenesulfonamide derivative selective CaM antagonists W-7 and W-13 on MM cell lines both in vitro and in vivo. Methods The proliferative ability was analyzed by the WST-8 assay. Cell cycle was evaluated by flow cytometry after staining of cells with PI. Apoptosis was quantified by flow cytometry after double-staining of cells by Annexin-V/PI. Molecular changes of cell cycle and apoptosis were determined by Western blot. Intracellular calcium levels and mitochondrial membrane potentials were determined using Fluo-4/AM dye and JC-10 dye, respectively. Moreover, we examined the in vivo anti-MM effects of CaM antagonists using a murine xenograft model of the human MM cell line. Results Treatment with W-7 and W-13 resulted in the dose-dependent inhibition of cell proliferation in various MM cell lines. W-7 and W-13 induced G1 phase cell cycle arrest by downregulating cyclins and upregulating p21cip1. In addition, W-7 and W-13 induced apoptosis via caspase activation; this occurred partly through the elevation of intracellular calcium levels and mitochondrial membrane potential depolarization and through inhibition of the STAT3 phosphorylation and subsequent downregulation of Mcl-1 protein. In tumor xenograft mouse models, tumor growth rates in CaM antagonist-treated groups were significantly reduced compared with those in the vehicle-treated groups. Conclusions Our results demonstrate that CaM antagonists induce cell cycle arrest, induce apoptosis via caspase activation, and inhibit tumor growth in a murine MM model and raise the possibility that inhibition of CaM might be a useful therapeutic strategy for the treatment of MM.
Collapse
Affiliation(s)
- Shigeyuki Yokokura
- Department of Internal Medicine, Division of Hematology, Rheumatology and Respiratory Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | | | | | | | | | | | | |
Collapse
|
241
|
Potent anti-cancer effect of 3'-hydroxypterostilbene in human colon xenograft tumors. PLoS One 2014; 9:e111814. [PMID: 25389774 PMCID: PMC4229093 DOI: 10.1371/journal.pone.0111814] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 10/08/2014] [Indexed: 12/25/2022] Open
Abstract
Here we report that 3′-hydroxypterostilbene (HPSB), a natural pterostilbene analogue, was more potent than pterostilbene against the growth of human cancer cells (COLO 205, HCT-116, and HT-29) with measured IC50 values of 9.0, 40.2, and 70.9 µM, respectively. We found that HPSB effectively inhibited the growth of human colon cancer cells by inducing apoptosis and autophagy. Autophagy occurred at an early stage and was observed through the formation of acidic vesicular organelles and microtubule-associated protein 1 light chain 3-II production. At the molecular levels, the results from western blot analysis showed that HPSB significantly down-regulated phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinases (MAPKs) signalings including decreased the phosphorylation of mammalian target of rapamycin (mTOR). Significant therapeutic effects were demonstrated in vivo by treating nude mice bearing COLO 205 tumor xenografts with HPSB (10 mg/kg i.p.). These inhibitory effects were accompanied by mechanistic down-regulation of the protein levels of cyclooxygenase-2 (COX-2), matrix metallopeptidase-9 (MMP-9), vascular endothelial growth factor (VEGF), and cyclin D1, as well as by the induction of apoptosis in colon tumors. Our findings suggest that HPSB could serve as a novel promising agent for colon cancer treatment.
Collapse
|
242
|
Gérard N, Chanson-Rollé A, Rock E, Brachet P. Proteomic analysis identifies cytoskeleton-interacting proteins as major downstream targets of altered folate status in the aorta of adult rat. Mol Nutr Food Res 2014; 58:2307-19. [PMID: 25266508 DOI: 10.1002/mnfr.201400317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/14/2014] [Accepted: 09/08/2014] [Indexed: 12/30/2022]
Abstract
SCOPE Mild folate deficiency and subsequently elevated plasma level of homocysteine are associated with an increased risk for vascular diseases in adults. Conversely, high intakes of folic acid (FA) may have beneficial effects on vascular function, presumably in part through homocysteine lowering. However, these effects have not yet been translated in terms of prevention or treatment of vascular pathologies. Besides, the complex biologic perturbation induced by variations of the folate supply is still not fully deciphered. We thus carried out a proteomic analysis of the aorta of adult rats after a dietary FA depletion or supplementation. METHODS AND RESULTS Nine month-old rats were fed a FA-depleted, FA-supplemented or control diet for 8 weeks. Total proteins from adventitia-free aortas were separated by 2DE and differentially expressed proteins were identified by MS. FA depletion or supplementation resulted in significantly changed abundance of 29 spots (p < 0.05), of which 20 proteins were identified. Bioinformatic analysis revealed that most of these proteins are involved in cytoskeleton-related processes important to cell function/maintenance, assembly/organization, and movement. CONCLUSION Our proteomic study supports that expression of proteins essential to vascular structure and, presumably, function is modulated by high intake as well as deprivation of FA.
Collapse
Affiliation(s)
- Nicolas Gérard
- Human Nutrition Unit, UMR 1019, National Institute for Agronomic Research, University of Auvergne, Clermont-Ferrand, France
| | | | | | | |
Collapse
|
243
|
Li S, Jia Z, Li X, Geng X, Sun J. Calmodulin is a stress and immune response gene in Chinese mitten crab Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2014; 40:120-128. [PMID: 24997436 DOI: 10.1016/j.fsi.2014.06.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 06/25/2014] [Accepted: 06/25/2014] [Indexed: 06/03/2023]
Abstract
Calmodulin (CaM) is a multifunctional calcium sensor protein that participates in various cellular processes under normal, stress and pathological conditions. In crabs, however, the involvement of CaM in response to environmental stress and immune challenges has not been revealed yet. In the present study, a CaM cDNA (EsCaM) was identified from Chinese mitten crab Eriocheir sinensis and its mRNA expression patterns in response to ambient (salinity and pH) stress and immune challenges was examined. EsCaM encodes a 149-amino-acid protein with a calculated molecular mass of 16.8 kDa and an isoelectric point of 4.09. In unstimulated healthy E. sinensis, EsCaM mRNA transcript was detected in all tested tissues with predominant expression in hepatopancreas and the lowest expression in haemocytes. Ambient salinity (15‰ and 30‰ salinities) and pH (pH 6 and 8.5) stress significantly altered EsCaM mRNA expression in gill, hepatopancreas, haemocytes, intestine and muscle in Chinese mitten crab. In addition, EsCaM gene expression was significantly and rapidly induced as early as 2 h after LPS and Poly(I:C) immune stimulations in haemocytes in vitro. Furthermore, EsCaM expression was significantly up-regulated in E. sinensis haemocytes, gill, hepatopancreas, intestine and muscle in response to Edwardsiella tarda and Vibrio anguillarum challenges. Collectively, our findings suggest that EsCaM is an important stress and immune response gene in Chinese mitten crab.
Collapse
Affiliation(s)
- Shuo Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 Binshuixidao, Xiqing District, Tianjin 300387, China.
| | - Zirui Jia
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 Binshuixidao, Xiqing District, Tianjin 300387, China
| | - Xuejing Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 Binshuixidao, Xiqing District, Tianjin 300387, China
| | - Xuyun Geng
- Tianjin Center for Control and Prevention of Aquatic Animal Infectious Disease, 442 South Jiefang Road, Hexi District, Tianjin 300221, China
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, 393 Binshuixidao, Xiqing District, Tianjin 300387, China.
| |
Collapse
|
244
|
The Ever Changing Moods of Calmodulin: How Structural Plasticity Entails Transductional Adaptability. J Mol Biol 2014; 426:2717-35. [DOI: 10.1016/j.jmb.2014.05.016] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/14/2014] [Accepted: 05/16/2014] [Indexed: 11/20/2022]
|