1
|
Abdel-Razek O, Marzouk A, MacKinnon M, Guy ET, Pohar SA, Zhushma E, Liu J, Sia I, Gokey JJ, Tay HG, Amack JD. Calcium signaling mediates proliferation of the precursor cells that give rise to the ciliated left-right organizer in the zebrafish embryo. Front Mol Biosci 2023; 10:1292076. [PMID: 38152112 PMCID: PMC10751931 DOI: 10.3389/fmolb.2023.1292076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/23/2023] [Indexed: 12/29/2023] Open
Abstract
Several of our internal organs, including heart, lungs, stomach, and spleen, develop asymmetrically along the left-right (LR) body axis. Errors in establishing LR asymmetry, or laterality, of internal organs during early embryonic development can result in birth defects. In several vertebrates-including humans, mice, frogs, and fish-cilia play a central role in establishing organ laterality. Motile cilia in a transient embryonic structure called the "left-right organizer" (LRO) generate a directional fluid flow that has been proposed to be detected by mechanosensory cilia to trigger asymmetric signaling pathways that orient the LR axis. However, the mechanisms that control the form and function of the ciliated LRO remain poorly understood. In the zebrafish embryo, precursor cells called dorsal forerunner cells (DFCs) develop into a transient ciliated structure called Kupffer's vesicle (KV) that functions as the LRO. DFCs can be visualized and tracked in the embryo, thereby providing an opportunity to investigate mechanisms that control LRO development. Previous work revealed that proliferation of DFCs via mitosis is a critical step for developing a functional KV. Here, we conducted a targeted pharmacological screen to identify mechanisms that control DFC proliferation. Small molecule inhibitors of the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) were found to reduce DFC mitosis. The SERCA pump is involved in regulating intracellular calcium ion (Ca2+) concentration. To visualize Ca2+ in living embryos, we generated transgenic zebrafish using the fluorescent Ca2+ biosensor GCaMP6f. Live imaging identified dynamic cytoplasmic Ca2+ transients ("flux") that occur unambiguously in DFCs. In addition, we report Ca2+ flux events that occur in the nucleus of DFCs. Nuclear Ca2+ flux occurred in DFCs that were about to undergo mitosis. We find that SERCA inhibitor treatments during DFC proliferation stages alters Ca2+ dynamics, reduces the number of ciliated cells in KV, and alters embryo laterality. Mechanistically, SERCA inhibitor treatments eliminated both cytoplasmic and nuclear Ca2+ flux events, and reduced progression of DFCs through the S/G2 phases of the cell cycle. These results identify SERCA-mediated Ca2+ signaling as a mitotic regulator of the precursor cells that give rise to the ciliated LRO.
Collapse
Affiliation(s)
- Osama Abdel-Razek
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Amanda Marzouk
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Madison MacKinnon
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Edward T. Guy
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Sonny A. Pohar
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Emily Zhushma
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Junjie Liu
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Isabel Sia
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Jason J. Gokey
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Hwee Goon Tay
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Jeffrey D. Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, NY, United States
| |
Collapse
|
2
|
Vanneste M, Venzke A, Guin S, Fuller AJ, Jezewski AJ, Beattie SR, Krysan DJ, Meyers MJ, Henry MD. The anti-cancer efficacy of a novel phenothiazine derivative is independent of dopamine and serotonin receptor inhibition. Front Oncol 2023; 13:1295185. [PMID: 37909019 PMCID: PMC10613967 DOI: 10.3389/fonc.2023.1295185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
Introduction An attractive, yet unrealized, goal in cancer therapy is repurposing psychiatric drugs that can readily penetrate the blood-brain barrier for the treatment of primary brain tumors and brain metastases. Phenothiazines (PTZs) have demonstrated anti-cancer properties through a variety of mechanisms. However, it remains unclear whether these effects are entirely separate from their activity as dopamine and serotonin receptor (DR/5-HTR) antagonists. Methods In this study, we evaluated the anti-cancer efficacy of a novel PTZ analog, CWHM-974, that was shown to be 100-1000-fold less potent against DR/5-HTR than its analog fluphenazine (FLU). Results CWHM-974 was more potent than FLU against a panel of cancer cell lines, thus clearly demonstrating that its anti-cancer effects were independent of DR/5-HTR signaling. Our results further suggested that calmodulin (CaM) binding may be necessary, but not sufficient, to explain the anti-cancer effects of CWHM-974. While both FLU and CWHM-974 induced apoptosis, they induced distinct effects on the cell cycle (G0/G1 and mitotic arrest respectively) suggesting that they may have differential effects on CaM-binding proteins involved in cell cycle regulation. Discussion Altogether, our findings indicated that the anti-cancer efficacy of the CWHM-974 is separable from DR/5-HTR antagonism. Thus, reducing the toxicity associated with phenothiazines related to DR/5-HTR antagonism may improve the potential to repurpose this class of drugs to treat brain tumors and/or brain metastasis.
Collapse
Affiliation(s)
- Marion Vanneste
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Anita Venzke
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Soumitra Guin
- Department of Chemistry, Saint Louis University, Saint Louis, MO, United States
| | - Andrew J. Fuller
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Andrew J. Jezewski
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Sarah R. Beattie
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Damian J. Krysan
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City IA, United States
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Marvin J. Meyers
- Department of Chemistry, Saint Louis University, Saint Louis, MO, United States
| | - Michael D. Henry
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
3
|
Essegian DJ, Chavez V, Khurshid R, Merchan JR, Schürer SC. AI-Assisted chemical probe discovery for the understudied Calcium-Calmodulin Dependent Kinase, PNCK. PLoS Comput Biol 2023; 19:e1010263. [PMID: 37235579 PMCID: PMC10249896 DOI: 10.1371/journal.pcbi.1010263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/08/2023] [Accepted: 04/13/2023] [Indexed: 05/28/2023] Open
Abstract
PNCK, or CAMK1b, is an understudied kinase of the calcium-calmodulin dependent kinase family which recently has been identified as a marker of cancer progression and survival in several large-scale multi-omics studies. The biology of PNCK and its relation to oncogenesis has also begun to be elucidated, with data suggesting various roles in DNA damage response, cell cycle control, apoptosis and HIF-1-alpha related pathways. To further explore PNCK as a clinical target, potent small-molecule molecular probes must be developed. Currently, there are no targeted small molecule inhibitors in pre-clinical or clinical studies for the CAMK family. Additionally, there exists no experimentally derived crystal structure for PNCK. We herein report a three-pronged chemical probe discovery campaign which utilized homology modeling, machine learning, virtual screening and molecular dynamics to identify small molecules with low-micromolar potency against PNCK activity from commercially available compound libraries. We report the discovery of a hit-series for the first targeted effort towards discovering PNCK inhibitors that will serve as the starting point for future medicinal chemistry efforts for hit-to-lead optimization of potent chemical probes.
Collapse
Affiliation(s)
- Derek J. Essegian
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Valery Chavez
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Rabia Khurshid
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Jaime R. Merchan
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Stephan C. Schürer
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| |
Collapse
|
4
|
Xie T, Chen S, Hao J, Wu P, Gu X, Wei H, Li Z, Xiao J. Roles of calcium signaling in cancer metastasis to bone. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:445-462. [PMID: 36071984 PMCID: PMC9446157 DOI: 10.37349/etat.2022.00094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/16/2022] [Indexed: 11/19/2022] Open
Abstract
Bone metastasis is a frequent complication for cancers and an important reason for the mortality in cancer patients. After surviving in bone, cancer cells can cause severe pain, life-threatening hypercalcemia, pathologic fractures, spinal cord compression, and even death. However, the underlying mechanisms of bone metastasis were not clear. The role of calcium (Ca2+) in cancer cell proliferation, migration, and invasion has been well established. Interestingly, emerging evidence indicates that Ca2+ signaling played a key role in bone metastasis, for it not only promotes cancer progression but also mediates osteoclasts and osteoblasts differentiation. Therefore, Ca2+ signaling has emerged as a novel therapeutical target for cancer bone metastasis treatments. Here, the role of Ca2+ channels and Ca2+-binding proteins including calmodulin and Ca2+-sensing receptor in bone metastasis, and the perspective of anti-cancer bone metastasis therapeutics via targeting the Ca2+ signaling pathway are summarized.
Collapse
Affiliation(s)
- Tianying Xie
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Sitong Chen
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jiang Hao
- Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Pengfei Wu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, Hunan, China
| | - Xuelian Gu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Haifeng Wei
- Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Zhenxi Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Shanghai 200003, China
| | - Jianru Xiao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Shanghai 200003, China
| |
Collapse
|
5
|
Abstract
In 2011, CAMKK2, the gene encoding calcium/calmodulin-dependent kinase kinase 2 (CAMKK2), was demonstrated to be a direct target of the androgen receptor and a driver of prostate cancer progression. Results from multiple independent studies have confirmed these findings and demonstrated the potential role of CAMKK2 as a clinical biomarker and therapeutic target in advanced prostate cancer using a variety of preclinical models. Drug development efforts targeting CAMKK2 have begun accordingly. CAMKK2 regulation can vary across disease stages, which might have important implications in the use of CAMKK2 as a biomarker. Moreover, new non-cell-autonomous roles for CAMKK2 that could affect tumorigenesis, metastasis and possible comorbidities linked to disease and treatment have emerged and could present novel treatment opportunities for prostate cancer.
Collapse
|
6
|
Fu D, Pfannenstiel L, Demelash A, Phoon YP, Mayell C, Cabrera C, Liu C, Zhao J, Dermawan J, Patil D, DeVecchio J, Kalady M, Souers AJ, Phillips DC, Li X, Gastman B. MCL1 nuclear translocation induces chemoresistance in colorectal carcinoma. Cell Death Dis 2022; 13:63. [PMID: 35042842 PMCID: PMC8766550 DOI: 10.1038/s41419-021-04334-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/21/2021] [Accepted: 10/19/2021] [Indexed: 11/09/2022]
Abstract
AbstractColorectal cancer (CRC) is one of the most common and deadliest forms of cancer. Myeloid Cell Leukemia 1 (MCL1), a pro-survival member of the Bcl-2 protein family is associated with chemo-resistance in CRC. The ability of MCL1 to inhibit apoptosis by binding to the BH3 domains of pro-apoptotic Bcl-2 family members is a well-studied means by which this protein confers resistance to multiple anti-cancer therapies. We found that specific DNA damaging chemotherapies promote nuclear MCL1 translocation in CRC models. In p53null CRC, this process is associated with resistance to chemotherapeutic agents, the mechanism of which is distinct from the classical mitochondrial protection. We previously reported that MCL1 has a noncanonical chemoresistance capability, which requires a novel loop domain that is distinct from the BH3-binding domain associated with anti-apoptotic function. Herein we disclose that upon treatment with specific DNA-damaging chemotherapy, this loop domain binds directly to alpha-enolase which in turn binds to calmodulin; we further show these protein−protein interactions are critical in MCL1’s nuclear import and chemoresistance. We additionally observed that in chemotherapy-treated p53−/− CRC models, MCL1 nuclear translocation confers sensitivity to Bcl-xL inhibitors, which has significant translational relevance given the co-expression of these proteins in CRC patient samples. Together these findings indicate that chemotherapy-induced MCL1 translocation represents a novel resistance mechanism in CRC, while also exposing an inherent and targetable Bcl-xL co-dependency in these cancers. The combination of chemotherapy and Bcl-xL inhibitors may thus represent a rational means of treating p53−/− CRC via exploitation of this unique MCL1-based chemoresistance mechanism.
Collapse
|
7
|
Zhang H, Chen B, Zou Z, Feng J, Li Y, Wang Y, He X, Xu C, Wang H, Guo S, Jin L, Li Q, Wang J, Xiao M, Li F, Wu J. Associations Between CAMKK1 Polymorphism rs7214723 and the Prognosis of Patients With Lung Cancer. Front Oncol 2021; 11:757484. [PMID: 34868969 PMCID: PMC8640188 DOI: 10.3389/fonc.2021.757484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/26/2021] [Indexed: 11/18/2022] Open
Abstract
Background The 5-year survival rate of patients with lung cancer in China is less than 20% and predicting their prognosis is challenging. We investigated the association between a common non-synonymous single nucleotide polymorphism (SNP), rs7214723, in the Ca2+/calmodulin-dependent protein kinase kinase 1 (CAMKK1) gene and the prognosis of patients with lung cancer. Methods Genomic DNA was extracted from the blood samples of 839 patients with lung cancer, recruited from Changhai Hospital (n = 536) and Taizhou Institute of Health Sciences (n = 352), and genotyped using the SNPscan technique. The association between patient prognosis and the genotypic data for CAMKK1 was analyzed using a multivariate Cox proportional hazards model adjusted for multiple potential confounders. The CRISPR/Cas9 gene-editing system was used to introduce point mutations in the CAMKK1 rs7214723 of A549 and NCI-H358 cells. Subsequently, Cell proliferation and migration ability were assessed with the Cell Counting Kit-8 and scratch assay. The Annexin V-FITC apoptosis detection kit was used to detect cell apoptosis. Results The CAMKK1 rs7214723 recessive CC genotype conferred significantly better overall survival (CC vs. TT + TC: adjusted hazard ratio = 0.78, 95% confidence interval [CI], 0.61-1.00, P = 0.049) than the TT + TC genotypes. Stratified analysis showed that the CAMKK1 rs7214723 CC genotype and recessive CC genotype conferred a significantly decreased risk of death in patients who were male, had a smoking history, or had stage III + IV cancer, compared with the TT and TT + TC genotypes. Relative to the TT + TC genotypes, the rs7214723 recessive CC genotype was also associated with a decreased risk of death in patients aged < 60 years (CC vs. TT + TC: adjusted hazard ratio = 0.59, 95% CI, 0.37-0.93, P = 0.024) and patients with squamous cell carcinoma (CC vs. TT + TC: adjusted hazard ratio = 0.65, 95% CI, 0.44-0.98, P = 0.038). Remarkably, CRISPR/Cas9-guided single nucleotide editing demonstrated that CAMKK1 rs7214723 T > C mutation significantly inhibits cell proliferation and migration and promotes cell apoptosis. Conclusions CAMKK1 SNP rs7214723 may be a significant prognostic factor for the risk of death among patients with lung cancer.
Collapse
Affiliation(s)
- Haorui Zhang
- Department of Ophthalmology, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Bocen Chen
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, China
| | - Zixiu Zou
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jian Feng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yutao Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yi Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xing He
- Department of Urology, Navy Military Medical University Affiliated Changhai Hospital, Shanghai, China
| | - Chang Xu
- Clinical College, Xiangnan University, Chenzhou, China
| | - Haijian Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Shicheng Guo
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Human Phenome Institute, Fudan University, Shanghai, China
| | - Qiang Li
- Department of Respiratory and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Human Phenome Institute, Fudan University, Shanghai, China
| | - Man Xiao
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, China
| | - Feng Li
- Department of Respiratory Disease, Shanghai Public Health Clinical Center, Shanghai, China
| | - Junjie Wu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Department of Infectious Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Respiratory and Critical Care Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, China
| |
Collapse
|
8
|
Ventura E, Iannuzzi CA, Pentimalli F, Giordano A, Morrione A. RBL1/p107 Expression Levels Are Modulated by Multiple Signaling Pathways. Cancers (Basel) 2021; 13:cancers13195025. [PMID: 34638509 PMCID: PMC8507926 DOI: 10.3390/cancers13195025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 11/16/2022] Open
Abstract
The members of the retinoblastoma (RB) protein family, RB1/p105, retinoblastoma-like (RBL)1/p107 and RBL2/p130 are critical modulators of the cell cycle and their dysregulation has been associated with tumor initiation and progression. The activity of RB proteins is regulated by numerous pathways including oncogenic signaling, but the molecular mechanisms of these functional interactions are not fully defined. We previously demonstrated that RBL2/p130 is a direct target of AKT and it is a key mediator of the apoptotic process induced by AKT inhibition. Here we demonstrated that RBL1/p107 levels are only minorly modulated by the AKT signaling pathway. In contrast, we discovered that RBL1/p107 levels are regulated by multiple pathways linked directly or indirectly to Ca2+-dependent signaling. Inhibition of the multifunctional calcium/calmodulin-dependent kinases (CaMKs) significantly reduced RBL1/p107 expression levels and phosphorylation, increased RBL1/p107 nuclear localization and led to cell cycle arrest in G0/G1. Targeting the Ca2+-dependent endopeptidase calpain stabilized RBL1/p107 levels and counteracted the reduction of RBL1/p107 levels associated with CaMKs inhibition. Thus, these novel observations suggest a complex regulation of RBL1/p107 expression involving different components of signaling pathways controlled by Ca2+ levels, including CaMKs and calpain, pointing out a significant difference with the mechanisms modulating the close family member RBL2/p130.
Collapse
Affiliation(s)
- Elisa Ventura
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (E.V.); (A.G.)
| | - Carmelina Antonella Iannuzzi
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, I-80131 Napoli, Italy; (C.A.I.); (F.P.)
| | - Francesca Pentimalli
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, I-80131 Napoli, Italy; (C.A.I.); (F.P.)
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (E.V.); (A.G.)
- Department of Medical Biotechnologies, University of Siena, I-53100 Siena, Italy
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (E.V.); (A.G.)
- Correspondence: ; Tel.: +215-204-2450
| |
Collapse
|
9
|
Eduful B, O’Byrne SN, Temme L, Asquith CR, Liang Y, Picado A, Pilotte JR, Hossain MA, Wells CI, Zuercher WJ, Catta-Preta CMC, Zonzini Ramos P, Santiago AD, Couñago RM, Langendorf CG, Nay K, Oakhill JS, Pulliam TL, Lin C, Awad D, Willson TM, Frigo DE, Scott JW, Drewry DH. Hinge Binder Scaffold Hopping Identifies Potent Calcium/Calmodulin-Dependent Protein Kinase Kinase 2 (CAMKK2) Inhibitor Chemotypes. J Med Chem 2021; 64:10849-10877. [PMID: 34264658 PMCID: PMC8365604 DOI: 10.1021/acs.jmedchem.0c02274] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Indexed: 12/18/2022]
Abstract
CAMKK2 is a serine/threonine kinase and an activator of AMPK whose dysregulation is linked with multiple diseases. Unfortunately, STO-609, the tool inhibitor commonly used to probe CAMKK2 signaling, has limitations. To identify promising scaffolds as starting points for the development of high-quality CAMKK2 chemical probes, we utilized a hinge-binding scaffold hopping strategy to design new CAMKK2 inhibitors. Starting from the potent but promiscuous disubstituted 7-azaindole GSK650934, a total of 32 compounds, composed of single-ring, 5,6-, and 6,6-fused heteroaromatic cores, were synthesized. The compound set was specifically designed to probe interactions with the kinase hinge-binding residues. Compared to GSK650394 and STO-609, 13 compounds displayed similar or better CAMKK2 inhibitory potency in vitro, while compounds 13g and 45 had improved selectivity for CAMKK2 across the kinome. Our systematic survey of hinge-binding chemotypes identified several potent and selective inhibitors of CAMKK2 to serve as starting points for medicinal chemistry programs.
Collapse
Affiliation(s)
- Benjamin
J. Eduful
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Sean N. O’Byrne
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Louisa Temme
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Christopher R.
M. Asquith
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department
of Pharmacology, School of Medicine, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yi Liang
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Alfredo Picado
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Joseph R. Pilotte
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mohammad Anwar Hossain
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Carrow I. Wells
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - William J. Zuercher
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Carolina M. C. Catta-Preta
- Centro
de Química Medicinal (CQMED), Centro de Biologia Molecular
e Engenharia Genética (CBMEG), Universidade
Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-875, Brazil
- Structural
Genomics Consortium, Departamento de Genética e Evolução,
Instituto de Biologia, UNICAMP, Campinas, São Paulo 13083-886, Brazil
| | - Priscila Zonzini Ramos
- Centro
de Química Medicinal (CQMED), Centro de Biologia Molecular
e Engenharia Genética (CBMEG), Universidade
Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-875, Brazil
- Structural
Genomics Consortium, Departamento de Genética e Evolução,
Instituto de Biologia, UNICAMP, Campinas, São Paulo 13083-886, Brazil
| | - André de
S. Santiago
- Centro
de Química Medicinal (CQMED), Centro de Biologia Molecular
e Engenharia Genética (CBMEG), Universidade
Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-875, Brazil
- Structural
Genomics Consortium, Departamento de Genética e Evolução,
Instituto de Biologia, UNICAMP, Campinas, São Paulo 13083-886, Brazil
| | - Rafael M. Couñago
- Centro
de Química Medicinal (CQMED), Centro de Biologia Molecular
e Engenharia Genética (CBMEG), Universidade
Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-875, Brazil
- Structural
Genomics Consortium, Departamento de Genética e Evolução,
Instituto de Biologia, UNICAMP, Campinas, São Paulo 13083-886, Brazil
| | - Christopher G. Langendorf
- St
Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia
| | - Kévin Nay
- St
Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia
- Mary MacKillop
Institute for Health Research, Australian
Catholic University, 215 Spring Street, Melbourne 3000, Australia
| | - Jonathan S. Oakhill
- St
Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia
- Mary MacKillop
Institute for Health Research, Australian
Catholic University, 215 Spring Street, Melbourne 3000, Australia
| | - Thomas L. Pulliam
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Center
for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas 77204, United States
- Department
of Biology and Biochemistry, University
of Houston, Houston, Texas 77204, United
States
| | - Chenchu Lin
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- The University of Texas MD Anderson Cancer Center UTHealth
Graduate
School of Biomedical Sciences, Houston, Texas 77030, United States
| | - Dominik Awad
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- The University of Texas MD Anderson Cancer Center UTHealth
Graduate
School of Biomedical Sciences, Houston, Texas 77030, United States
| | - Timothy M. Willson
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Daniel E. Frigo
- Department
of Cancer Systems Imaging, University of
Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Center
for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas 77204, United States
- Department
of Biology and Biochemistry, University
of Houston, Houston, Texas 77204, United
States
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
- The Methodist Hospital Research Institute, Houston, Texas 77030, United States
| | - John W. Scott
- St
Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia
- Mary MacKillop
Institute for Health Research, Australian
Catholic University, 215 Spring Street, Melbourne 3000, Australia
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville 3052, Australia
| | - David H. Drewry
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center,
UNC Eshelman School of
Pharmacy, University of North Carolina at
Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
10
|
Essegian D, Khurana R, Stathias V, Schürer SC. The Clinical Kinase Index: A Method to Prioritize Understudied Kinases as Drug Targets for the Treatment of Cancer. CELL REPORTS MEDICINE 2020; 1:100128. [PMID: 33205077 PMCID: PMC7659504 DOI: 10.1016/j.xcrm.2020.100128] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/25/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
Abstract
The approval of the first kinase inhibitor, Gleevec, ushered in a paradigm shift for oncological treatment-the use of genomic data for targeted, efficacious therapies. Since then, over 48 additional small-molecule kinase inhibitors have been approved, solidifying the case for kinases as a highly druggable and attractive target class. Despite the role deregulated kinase activity plays in cancer, only 8% of the kinome has been effectively "drugged." Moreover, 24% of the 634 human kinases are understudied. We have developed a comprehensive scoring system that utilizes differential gene expression, pathological parameters, overall survival, and mutational hotspot analysis to rank and prioritize clinically relevant kinases across 17 solid tumor cancers from The Cancer Genome Atlas. We have developed the clinical kinase index (CKI) app (http://cki.ccs.miami.edu) to facilitate interactive analysis of all kinases in each cancer. Collectively, we report that understudied kinases have potential clinical value as biomarkers or drug targets that warrant further study.
Collapse
Affiliation(s)
- Derek Essegian
- Department of Pharmacology, Miller School of Medicine, University of Miami, Miami, USA
| | - Rimpi Khurana
- Department of Pharmacology, Miller School of Medicine, University of Miami, Miami, USA
| | - Vasileios Stathias
- Department of Pharmacology, Miller School of Medicine, University of Miami, Miami, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, USA
| | - Stephan C Schürer
- Department of Pharmacology, Miller School of Medicine, University of Miami, Miami, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, USA.,Institute for Data Science & Computing, University of Miami, Miami, USA
| |
Collapse
|
11
|
O’Byrne SN, Scott JW, Pilotte JR, Santiago ADS, Langendorf CG, Oakhill JS, Eduful BJ, Couñago RM, Wells CI, Zuercher WJ, Willson TM, Drewry DH. In Depth Analysis of Kinase Cross Screening Data to Identify CAMKK2 Inhibitory Scaffolds. Molecules 2020; 25:E325. [PMID: 31941153 PMCID: PMC7024175 DOI: 10.3390/molecules25020325] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/25/2022] Open
Abstract
The calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) activates CAMK1, CAMK4, AMPK, and AKT, leading to numerous physiological responses. The deregulation of CAMKK2 is linked to several diseases, suggesting the utility of CAMKK2 inhibitors for oncological, metabolic and inflammatory indications. In this work, we demonstrate that STO-609, frequently described as a selective inhibitor for CAMKK2, potently inhibits a significant number of other kinases. Through an analysis of literature and public databases, we have identified other potent CAMKK2 inhibitors and verified their activities in differential scanning fluorimetry and enzyme inhibition assays. These inhibitors are potential starting points for the development of selective CAMKK2 inhibitors and will lead to tools that delineate the roles of this kinase in disease biology.
Collapse
Affiliation(s)
- Sean N. O’Byrne
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - John W. Scott
- St Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia; (J.W.S.); (C.G.L.); (J.S.O.)
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville 3052, Australia
| | - Joseph R. Pilotte
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - André da S. Santiago
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas SP 13083-875, Brazil; (A.d.S.S.); (R.M.C.)
- Structural Genomics Consortium, Departamento de Genética e Evolução, Instituto de Biologia, UNICAMP, Campinas SP 13083-886, Brazil
| | - Christopher G. Langendorf
- St Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia; (J.W.S.); (C.G.L.); (J.S.O.)
| | - Jonathan S. Oakhill
- St Vincent’s Institute and Department of Medicine, The University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia; (J.W.S.); (C.G.L.); (J.S.O.)
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia
| | - Benjamin J. Eduful
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - Rafael M. Couñago
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), Universidade Estadual de Campinas (UNICAMP), Campinas SP 13083-875, Brazil; (A.d.S.S.); (R.M.C.)
- Structural Genomics Consortium, Departamento de Genética e Evolução, Instituto de Biologia, UNICAMP, Campinas SP 13083-886, Brazil
| | - Carrow I. Wells
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - William J. Zuercher
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| | - David H. Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.N.O.); (J.R.P.); (B.J.E.); (C.I.W.); (W.J.Z.); (T.M.W.)
| |
Collapse
|
12
|
Zhang Y, Wang LN, Lin YN, Xing YX, Shi Y, Zhao J, Chen WW, Han B. The novel long noncoding RNA LOC283070 is involved in the transition of LNCaP cells into androgen-independent cells via its interaction with PHB2. Asian J Androl 2019; 20:511-517. [PMID: 29956684 PMCID: PMC6116685 DOI: 10.4103/aja.aja_36_18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We sought to investigate the underlying mechanism of action of the long noncoding RNA (lncRNA) LOC283070 in the development of androgen independence in prostate cancer. The interactions between LOC283070 and target proteins were investigated by RNA pull-down and RNA-binding protein immunoprecipitation (RIP) assays. Subcellular fractionation and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) were used to detect the subcellular localization of LOC283070. Western blotting was performed to detect the expression of prohibitin 2 (PHB2). Luciferase activity assays were performed to evaluate the effects of LOC283070 and PHB2 on the androgen receptor (AR) signaling pathway. A methyl thiazolyl tetrazolium (MTT) assay and a growth curve assay were used to test cell viability. Flow cytometry was performed to analyze cell cycles. A transwell assay was employed to test cell migration. We identified PHB2 as an interaction partner of LOC283070 in the pull-down and RIP experiments. Furthermore, we confirmed that the enrichment of LOC283070 with PHB2 in androgen-independent LNCaP (LNCaP-AI) cells was much greater than that in LNCaP cells. Moreover, the expression of PHB2 was not significantly different between the two cell lines, and the expression of LOC283070 in the nuclei of the LNCaP-AI cells was significantly greater than that in the LNCaP cells. In vitro data revealed that PHB2 overexpression significantly inhibited AR activity and cell proliferation and migration and induced accumulation of prostate cancer cells in G0/G1 phase. Moreover, the overexpression of LOC283070 fully abrogated the effects of PHB2 overexpression. In conclusion, we found that LOC283070 can bind to PHB2 located in the nucleus and inhibit its effect, and this is one of the mechanisms by which LOC283070 is involved in the transition of LNCaP cells into androgen-independent cells.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Li-Na Wang
- Department of Clinical Laboratory Medicine, The Second Hospital of Shandong University, Jinan 250033, China
| | - Ya-Ni Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yuan-Xin Xing
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yu Shi
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jian Zhao
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Wei-Wen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Bo Han
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University; Department of Pathology, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
13
|
Brzozowski JS, Skelding KA. The Multi-Functional Calcium/Calmodulin Stimulated Protein Kinase (CaMK) Family: Emerging Targets for Anti-Cancer Therapeutic Intervention. Pharmaceuticals (Basel) 2019; 12:ph12010008. [PMID: 30621060 PMCID: PMC6469190 DOI: 10.3390/ph12010008] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 01/25/2023] Open
Abstract
The importance of Ca2+ signalling in key events of cancer cell function and tumour progression, such as proliferation, migration, invasion and survival, has recently begun to be appreciated. Many cellular Ca2+-stimulated signalling cascades utilise the intermediate, calmodulin (CaM). The Ca2+/CaM complex binds and activates a variety of enzymes, including members of the multifunctional Ca2+/calmodulin-stimulated protein kinase (CaMK) family. These enzymes control a broad range of cancer-related functions in a multitude of tumour types. Herein, we explore the cancer-related functions of these kinases and discuss their potential as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Joshua S Brzozowski
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute (HMRI) and University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Kathryn A Skelding
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute (HMRI) and University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
14
|
Gocher AM, Azabdaftari G, Euscher LM, Dai S, Karacosta LG, Franke TF, Edelman AM. Akt activation by Ca 2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2) in ovarian cancer cells. J Biol Chem 2017. [PMID: 28634229 DOI: 10.1074/jbc.m117.778464] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Hyperactivation of Akt is associated with oncogenic changes in the growth, survival, and chemoresistance of cancer cells. The PI3K/phosphoinositide-dependent kinase (PDK) 1 pathway represents the canonical mechanism for phosphorylation of Akt at its primary activation site, Thr-308. We observed that Ca2+/calmodulin (CaM)-dependent protein kinase kinase 2 (β) (CaMKK2) is highly expressed in high-grade serous ovarian cancer, and we investigated its role in Akt activation in ovarian cancer (OVCa) cell lines (OVCAR-3, SKOV-3, and Caov-3). Knockdown or pharmacological inhibition of CaMKK2 produced phenotypes expected of Akt inhibition, including reductions in cell growth and cell viability and in the regulation of Akt downstream targets involved in G1/S transition and apoptosis. CaMKK2 knockdown or inhibition decreased Akt phosphorylation at Thr-308 and Ser-473 to extents similar to those of PDK1 knockdown or PI3K inhibition. Combined CaMKK2 and PDK1 knockdown or CaMKK and PI3K inhibition, respectively, produced additive effects on p-Akt and cell growth, consistent with direct Akt phosphorylation by CaMKK2. This conclusion was supported by the absence of effects of CaMKK2 knockdown/inhibition on alternative means of activating Akt via p-Akt Thr-450, p-PDK1 Ser-241, or p-IRS1 Ser-636/639. Recombinant CaMKK2 directly activated recombinant Akt by phosphorylation at Thr-308 in a Ca2+/CaM-dependent manner. In OVCa cells, p-Akt Thr-308 was significantly inhibited by intracellular Ca2+i chelation or CaM inhibition. Ionomycin-induced Ca2+ influx promoted p-Akt, an effect blocked by PDK1, and/or CaMKK2, siRNAs, and by PI3K and/or CaMKK inhibitors. CaMKK2 knockdown potentiated the effects of the chemotherapeutic drugs carboplatin and PX-866 to reduce proliferation and survival of OVCa cells.
Collapse
Affiliation(s)
- Angela M Gocher
- From the Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York 14214
| | - Gissou Azabdaftari
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Lindsey M Euscher
- From the Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York 14214
| | - Shuhang Dai
- From the Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York 14214
| | - Loukia G Karacosta
- From the Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York 14214
| | - Thomas F Franke
- Department of Psychiatry, Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016
| | - Arthur M Edelman
- From the Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, New York 14214,.
| |
Collapse
|
15
|
Jiang M, Qiu J, Zhang L, Lü D, Long M, Chen L, Luo X. Changes in tension regulates proliferation and migration of fibroblasts by remodeling expression of ECM proteins. Exp Ther Med 2016; 12:1542-1550. [PMID: 27588075 DOI: 10.3892/etm.2016.3497] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 05/18/2016] [Indexed: 12/27/2022] Open
Abstract
Wound healing is a complicated but highly organized process in which cell migration and proliferation are actively involved. However, the process by which mechanical stretch regulates the proliferation and migration of human skin fibroblasts (HFs) and keratinocytes is poorly understood. Using a house built mechanical stretch device, we examined the HFs extracellular matrix (ECM) components changes under non-stretch, static stretch or cyclic stretch conditions. We further investigated the changes in ECM component protein expression levels in keratinocytes and analyzed the effects of individual ECM component on keratinocyte proliferation and migration. Particularly, the roles of calcium/calmodulin-dependent serine protein kinase (CASK) in the HF proliferation under cyclic stretch were investigated. Cyclic stretch suppressed HF proliferation compared with HFs without stretch or with static stretch. Cyclic stretch also led to a significant reduction in the levels of collagen I and a marked increase of fibronectin in HFs ECM. By contrast, collagen I levels increased and fibronectin levels decreased in response to non-stretch and static stretch conditions. After cyclic stretch, the proliferation of keratinocytes was inhibited by the cyclic stretch-induced ECM in HFs. The inoculation of keratinocytes with single ECM component suggested that collagen I was more capable of inducing cell proliferation than fibronectin, while it had less impact on cell migration compared with fibronectin. Furthermore, cyclic stretch induced by proliferation inhibition was associated with altered integrin β1-CASK signal pathway. The present results demonstrated the existence of HF-ECM-keratinocyte 'cross-talk' in cutaneous tissues. Thus, the integrin β1-CASK signal pathway in HFs may be involved in the outside-in signal transduction of extracellular stretch and the altered ECM component expression.
Collapse
Affiliation(s)
- Minmin Jiang
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; Central Laboratory, The People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| | - Juhui Qiu
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; National Key Laboratory of Trauma and Burns and Chongqing Key Laboratory of Disease Proteomics, Chongqing 400038, P.R. China
| | - Lingling Zhang
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; National Key Laboratory of Trauma and Burns and Chongqing Key Laboratory of Disease Proteomics, Chongqing 400038, P.R. China
| | - Dongyuan Lü
- Key Laboratory of Microgravity (National Microgravity Laboratory) and Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Mian Long
- Key Laboratory of Microgravity (National Microgravity Laboratory) and Center for Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Li Chen
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; National Key Laboratory of Trauma and Burns and Chongqing Key Laboratory of Disease Proteomics, Chongqing 400038, P.R. China; Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xiangdong Luo
- Burn Research Institute, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China; National Key Laboratory of Trauma and Burns and Chongqing Key Laboratory of Disease Proteomics, Chongqing 400038, P.R. China
| |
Collapse
|
16
|
Pharmacological targeting of ion channels for cancer therapy: In vivo evidences. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1385-97. [DOI: 10.1016/j.bbamcr.2015.11.032] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/29/2022]
|
17
|
Hamada T, Souda M, Yoshimura T, Sasaguri S, Hatanaka K, Tasaki T, Yoshioka T, Ohi Y, Yamada S, Tsutsui M, Umekita Y, Tanimoto A. Anti-apoptotic effects of PCP4/PEP19 in human breast cancer cell lines: a novel oncotarget. Oncotarget 2015; 5:6076-86. [PMID: 25153723 PMCID: PMC4171614 DOI: 10.18632/oncotarget.2161] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The PCP4/PEP19 is a calmodulin-binding anti-apoptotic peptide in neural cells but its potential role in human cancer has largely been unknown. We investigated the expression of PCP4/PEP19 in human breast cancer cell lines MCF-7, SK-BR-3, and MDA-MB-231 cells, and found that estrogen receptor (ER)-positive MCF-7 and ER-negative SK-BR-3 cells expressed PCP4/PEP19. In the MCF-7 cells, cell proliferation was estrogen-dependent, and PCP4/PEP19 expression was induced by estrogen. In both cell lines, PCP4/PEP19 knockdown induced apoptosis and slightly decreased Akt phosphorylation. Knockdown of calcium/calmodulin-dependent protein kinase kinase 1 (CaMKK1), resulting in decreased phospho-AktThr308, enhanced apoptosis in SK-BR-3 but not in MCF-7 cells. CaMKK2 knockdown moderately decreased phospho-AktThr308 and increased apoptosis in MCF-7 cells but not in SK-BR-3 cells. These data indicated that PCP4/PEP19 regulates apoptosis but exact mechanism is still unknown. PCP4/PEP19 can therefore potentially serve as independent oncotarget for therapy of PCP4/PEP19-positive breast cancers irrespective of ER expression.
Collapse
Affiliation(s)
- Taiji Hamada
- Department of Molecular and Cellular Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Masakazu Souda
- Department of Molecular and Cellular Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takuya Yoshimura
- Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shoko Sasaguri
- Department of Molecular and Cellular Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kazuhito Hatanaka
- Department of Molecular and Cellular Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takashi Tasaki
- Department of Molecular and Cellular Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takako Yoshioka
- Department of Molecular and Cellular Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yasuyo Ohi
- Department of Pathology, Sagara Hospital, Social Medical Corporation Hakuaikai, Kagoshima, Japan
| | - Sohsuke Yamada
- Department of Pathology and Cell Biology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masato Tsutsui
- Department of Pharmacology, Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yoshihisa Umekita
- Division of Organ Pathology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Akihide Tanimoto
- Department of Molecular and Cellular Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
18
|
Roles of NGAL and MMP-9 in the tumor microenvironment and sensitivity to targeted therapy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:438-448. [PMID: 26278055 DOI: 10.1016/j.bbamcr.2015.08.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/08/2015] [Accepted: 08/10/2015] [Indexed: 12/15/2022]
Abstract
Various, diverse molecules contribute to the tumor microenvironment and influence invasion and metastasis. In this review, the roles of neutrophil gelatinase-associated lipocalin (NGAL) and matrix metalloproteinase-9 (MMP-9) in the tumor microenvironment and sensitivity to therapy will be discussed. The lipocalin family of proteins has many important functions. For example when NGAL forms a complex with MMP-9 it increases its stability which is important in cancer metastasis. Small hydrophobic molecules are bound by NGAL which can alter their entry into and efflux from cells. Iron transport and storage are also influenced by NGAL activity. Regulation of iron levels is important for survival in the tumor microenvironment as well as metastasis. Innate immunity is also regulated by NGAL as it can have bacteriostatic properties. NGAL and MMP-9 expression may also affect the sensitivity of cancer cells to chemotherapy as well as targeted therapy. Thus NGAL and MMP-9 play important roles in key processes involved in metastasis as well as response to therapy. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis, Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.
Collapse
|
19
|
Di J, Huang H, Qu D, Tang J, Cao W, Lu Z, Cheng Q, Yang J, Bai J, Zhang Y, Zheng J. Rap2B promotes proliferation, migration, and invasion of human breast cancer through calcium-related ERK1/2 signaling pathway. Sci Rep 2015. [PMID: 26201295 PMCID: PMC4512009 DOI: 10.1038/srep12363] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Rap2B, a member of GTP-binding proteins, is widely upregulated in many types of tumors and promotes migration and invasion of human suprarenal epithelioma. However, the function of Rap2B in breast cancer is unknown. Expression of Rap2B was examined in breast cancer cell lines and human normal breast cell line using Western blot analysis. Using the CCK-8 cell proliferation assay, cell cycle analysis, and transwell migration assay, we also elucidated the role of Rap2B in breast cancer cell proliferation, migration, and invasion. Results showed that the expression of Rap2B is higher in tumor cells than in normal cells. Flow cytometry and Western blot analysis revealed that Rap2B elevates the intracellular calcium level and further promotes extracellular signal-related kinase (ERK) 1/2 phosphorylation. By contrast, calcium chelator BAPTM/AM and MEK inhibitor (U0126) can reverse Rap2B-induced ERK1/2 phosphorylation. Furthermore, Rap2B knockdown inhibits cell proliferation, migration, and invasion abilities via calcium related-ERK1/2 signaling. In addition, overexpression of Rap2B promotes cell proliferation, migration and invasion abilities, which could be neutralized by BAPTM/AM and U0126. Taken together, these findings shed light on Rap2B as a therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Jiehui Di
- 1] Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China [2] Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, School of Medicine, the University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27514, USA
| | - Hui Huang
- 1] Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China [2] Department of Oncology, the People's Hospital of Kaixian, Kaixian 405400, Chongqing, P.R. China
| | - Debao Qu
- Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China
| | - Juangjuan Tang
- Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China
| | - Wenjia Cao
- Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China
| | - Zheng Lu
- Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China
| | - Qian Cheng
- Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China
| | - Jing Yang
- Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China
| | - Yanping Zhang
- 1] Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China [2] Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, School of Medicine, the University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27514, USA
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou 221002, Jiangsu, P.R. China
| |
Collapse
|
20
|
Berchtold MW, Villalobo A. The many faces of calmodulin in cell proliferation, programmed cell death, autophagy, and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:398-435. [PMID: 24188867 DOI: 10.1016/j.bbamcr.2013.10.021] [Citation(s) in RCA: 236] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/24/2013] [Accepted: 10/26/2013] [Indexed: 12/21/2022]
Abstract
Calmodulin (CaM) is a ubiquitous Ca(2+) receptor protein mediating a large number of signaling processes in all eukaryotic cells. CaM plays a central role in regulating a myriad of cellular functions via interaction with multiple target proteins. This review focuses on the action of CaM and CaM-dependent signaling systems in the control of vertebrate cell proliferation, programmed cell death and autophagy. The significance of CaM and interconnected CaM-regulated systems for the physiology of cancer cells including tumor stem cells, and processes required for tumor progression such as growth, tumor-associated angiogenesis and metastasis are highlighted. Furthermore, the potential targeting of CaM-dependent signaling processes for therapeutic use is discussed.
Collapse
Key Words
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-ethyl]-4,5-dihydro-pyrazol-1-yl]-benzoic acid
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-vinyl]-4,5-dihydro-pyrazol-1-yl]-phenyl)-(4-methyl-piperazin-1-yl)-methanone
- (−) enantiomer of dihydropyrine 3-methyl-5-3-(4,4-diphenyl-1-piperidinyl)-propyl-1,4-dihydro-2,6-dimethyl-4-(3-nitrophenyl)-piridine-3,5-dicarboxylate-hydrochloride (niguldipine)
- 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine
- 12-O-tetradecanoyl-phorbol-13-acetate
- 2-chloro-(ε-amino-Lys(75))-[6-(4-(N,N′-diethylaminophenyl)-1,3,5-triazin-4-yl]-CaM adduct
- 3′-(β-chloroethyl)-2′,4′-dioxo-3,5′-spiro-oxazolidino-4-deacetoxy-vinblastine
- 7,12-dimethylbenz[a]anthracene
- Apoptosis
- Autophagy
- B859-35
- CAPP(1)-CaM
- Ca(2+) binding protein
- Calmodulin
- Cancer biology
- Cell proliferation
- DMBA
- EBB
- FL-CaM
- FPCE
- HBC
- HBCP
- J-8
- KAR-2
- KN-62
- KN-93
- N-(4-aminobutyl)-2-naphthalenesulfonamide
- N-(4-aminobutyl)-5-chloro-2-naphthalenesulfonamide
- N-(6-aminohexyl)-1-naphthalenesulfonamide
- N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide
- N-8-aminooctyl-5-iodo-naphthalenesulfonamide
- N-[2-[N-(4-chlorocinnamyl)-N-methylaminomethyl]phenyl]-N-(2-hydroxyethyl)-4-methoxybenzenesulfonamide
- O-(4-ethoxyl-butyl)-berbamine
- RITC-CaM
- TA-CaM
- TFP
- TPA
- W-12
- W-13
- W-5
- W-7
- fluorescein-CaM adduct
- fluphenazine-N-2-chloroethane
- norchlorpromazine-CaM adduct
- rhodamine isothiocyanate-CaM adduct
- trifluoperazine
Collapse
Affiliation(s)
- Martin W Berchtold
- Department of Biology, University of Copenhagen, Copenhagen Biocenter 4-2-09 Ole Maaløes Vej 5, DK-2200 Copenhagen N, Denmark.
| | - Antonio Villalobo
- Instituto de Investigaciones Biomédicas, Department of Cancer Biology, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
21
|
Rohban R, Reinisch A, Etchart N, Schallmoser K, Hofmann NA, Szoke K, Brinchmann JE, Rad EB, Rohde E, Strunk D. Identification of an effective early signaling signature during neo-vasculogenesis in vivo by ex vivo proteomic profiling. PLoS One 2013; 8:e66909. [PMID: 23826172 PMCID: PMC3691264 DOI: 10.1371/journal.pone.0066909] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 05/11/2013] [Indexed: 12/20/2022] Open
Abstract
Therapeutic neo-vasculogenesis in vivo can be achieved by the co-transplantation of human endothelial colony-forming progenitor cells (ECFCs) with mesenchymal stem/progenitor cells (MSPCs). The underlying mechanism is not completely understood thus hampering the development of novel stem cell therapies. We hypothesized that proteomic profiling could be used to retrieve the in vivo signaling signature during the initial phase of human neo-vasculogenesis. ECFCs and MSPCs were therefore either transplanted alone or co-transplanted subcutaneously into immune deficient mice. Early cell signaling, occurring within the first 24 hours in vivo, was analyzed using antibody microarray proteomic profiling. Vessel formation and persistence were verified in parallel transplants for up to 24 weeks. Proteomic analysis revealed significant alteration of regulatory components including caspases, calcium/calmodulin-dependent protein kinase, DNA protein kinase, human ErbB2 receptor-tyrosine kinase as well as mitogen-activated protein kinases. Caspase-4 was selected from array results as one therapeutic candidate for targeting vascular network formation in vitro as well as modulating therapeutic vasculogenesis in vivo. As a proof-of-principle, caspase-4 and general caspase-blocking led to diminished endothelial network formation in vitro and significantly decreased vasculogenesis in vivo. Proteomic profiling ex vivo thus unraveled a signaling signature which can be used for target selection to modulate neo-vasculogenesis in vivo.
Collapse
Affiliation(s)
- Rokhsareh Rohban
- Stem Cell Research Unit, Medical University of Graz, Graz, Austria
| | - Andreas Reinisch
- Stem Cell Research Unit, Medical University of Graz, Graz, Austria
- Division of Hematology and Stem Cell Transplantation, Medical University of Graz, Graz, Austria
| | - Nathalie Etchart
- Stem Cell Research Unit, Medical University of Graz, Graz, Austria
- Division of Hematology and Stem Cell Transplantation, Medical University of Graz, Graz, Austria
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Katharina Schallmoser
- Stem Cell Research Unit, Medical University of Graz, Graz, Austria
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
- Department of Blood Group Serology and Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria
| | | | - Krisztina Szoke
- Norwegian Center for Stem Cell Research, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jan E. Brinchmann
- Norwegian Center for Stem Cell Research, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ehsan Bonyadi Rad
- Cancer Biology Unit, Department of Dermatology, Medical University of Graz, Graz, Austria
- Department of Pediatric and Adolescence Surgery, Medical University of Graz, Graz, Austria
| | - Eva Rohde
- Department of Blood Group Serology and Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Dirk Strunk
- Stem Cell Research Unit, Medical University of Graz, Graz, Austria
- Division of Hematology and Stem Cell Transplantation, Medical University of Graz, Graz, Austria
- Institute of Experimental and Clinical Cell Therapy, Paracelsus Medical University, Salzburg, Austria
- * E-mail:
| |
Collapse
|
22
|
Tenga MJ, Lazar IM. Proteomic snapshot of breast cancer cell cycle: G1/S transition point. Proteomics 2013; 13:48-60. [PMID: 23152136 DOI: 10.1002/pmic.201200188] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 09/22/2012] [Accepted: 10/25/2012] [Indexed: 01/16/2023]
Abstract
The biological processes that unfold during the G1-phase of the cell cycle are dependent on extracellular mitogenic factors that signal the cell to enter a state of quiescence, or commit to a cell-cycle round by passing the restriction point (R-point) and enter the S-phase. Unlike normal cells, cancer cells evolved the ability to evade the R-point and continue through the cell cycle even in the presence of extensive DNA damage or absence of mitogenic signals. The purpose of this study was to perform a quantitative proteomic evaluation of the biological processes that are responsible for driving MCF-7 breast cancer cells into division even when molecular checkpoints such as the G1/S R-point are in place. Nuclear and cytoplasmic fractions of the G1 and S cell-cycle phases were analyzed by LC-MS/MS to result in the confident identification of more than 2700 proteins. Statistical evaluation of the normalized data resulted in the selection of proteins that displayed twofold or more change in spectral counts in each cell state. Pathway mapping, functional annotation clustering, and protein interaction network analysis revealed that the top-scoring clusters that could play a role in overriding the G1/S transition point included DNA damage response, chromatin remodeling, transcription/translation regulation, and signaling proteins.
Collapse
Affiliation(s)
- Milagros J Tenga
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 246021, USA
| | | |
Collapse
|
23
|
Mallampalli RK, Kaercher L, Snavely C, Pulijala R, Chen BB, Coon T, Zhao J, Agassandian M. Fbxl12 triggers G1 arrest by mediating degradation of calmodulin kinase I. Cell Signal 2013; 25:2047-59. [PMID: 23707388 DOI: 10.1016/j.cellsig.2013.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 05/07/2013] [Indexed: 12/18/2022]
Abstract
Cell cycle progression through its regulatory control by changes in intracellular Ca(2+) levels at the G1/S transition mediates cellular proliferation and viability. Ca(2+)/CaM-dependent kinase 1 (CaMKI) appears critical in regulating the assembly of the cyclin D1/cdk4 complex essential for G1 progression, but how this occurs is unknown. Cyclin D1/cdk4 assembly in the early G1 phase is also regulated via binding to p27. Here, we show that a ubiquitin E3 ligase component, F-box protein Fbxl12, mediates CaMKI degradation via a proteasome-directed pathway leading to disruption of cyclin D1/cdk4 complex assembly and resultant G1 arrest in lung epithelia. We also demonstrate that i) CaMKI phosphorylates p27 at Thr(157) and Thr(198) in human cells and at Thr(170) and Thr(197) in mouse cells to modulate its subcellular localization; ii) Fbxl12-induced CaMKI degradation attenuates p27 phosphorylation at these sites in early G1 and iii) activation of CaMKI during G1 transition followed by p27 phosphorylation appears to be upstream to other p27 phosphorylation events, an effect abrogated by Fbxl12 overexpression. Lastly, known inducers of G1 arrest significantly increase Fbxl12 levels in cells. Thus, Fbxl12 may be a previously uncharacterized, functional growth inhibitor regulating cell cycle progression that might be used for mechanism-based therapy.
Collapse
Affiliation(s)
- Rama K Mallampalli
- Department of Medicine, Acute Lung Injury Center of Excellence, The University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Chappell WH, Abrams SL, Franklin RA, LaHair MM, Montalto G, Cervello M, Martelli AM, Nicoletti F, Candido S, Libra M, Polesel J, Talamini R, Milella M, Tafuri A, Steelman LS, McCubrey JA. Ectopic NGAL expression can alter sensitivity of breast cancer cells to EGFR, Bcl-2, CaM-K inhibitors and the plant natural product berberine. Cell Cycle 2012; 11:4447-61. [PMID: 23159854 PMCID: PMC3552927 DOI: 10.4161/cc.22786] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL, a.k.a Lnc2) is a member of the lipocalin family and has diverse roles. NGAL can stabilize matrix metalloproteinase-9 from autodegradation. NGAL is considered as a siderocalin that is important in the transport of iron. NGAL expression has also been associated with certain neoplasias and is implicated in the metastasis of breast cancer. In a previous study, we examined whether ectopic NGAL expression would alter the sensitivity of breast epithelial, breast and colorectal cancer cells to the effects of the chemotherapeutic drug doxorubicin. While abundant NGAL expression was detected in all the cells infected with a retrovirus encoding NGAL, this expression did not alter the sensitivity of these cells to doxorubicin as compared with empty vector-transduced cells. We were also interested in determining the effects of ectopic NGAL expression on the sensitivity to small-molecule inhibitors targeting key signaling molecules. Ectopic NGAL expression increased the sensitivity of MCF-7 breast cancer cells to EGFR, Bcl-2 and calmodulin kinase inhibitors as well as the natural plant product berberine. Furthermore, when suboptimal concentrations of certain inhibitors were combined with doxorubicin, a reduction in the doxorubicin IC 50 was frequently observed. An exception was observed when doxorubicin was combined with rapamycin, as doxorubicin suppressed the sensitivity of the NGAL-transduced MCF-7 cells to rapamycin when compared with the empty vector controls. In contrast, changes in the sensitivities of the NGAL-transduced HT-29 colorectal cancer cell line and the breast epithelial MCF-10A cell line were not detected compared with empty vector-transduced cells. Doxorubicin-resistant MCF-7/Dox (R) cells were examined in these experiments as a control drug-resistant line; it displayed increased sensitivity to EGFR and Bcl-2 inhibitors compared with empty vector transduced MCF-7 cells. These results indicate that NGAL expression can alter the sensitivity of certain cancer cells to small-molecule inhibitors, suggesting that patients whose tumors exhibit elevated NGAL expression or have become drug-resistant may display altered responses to certain small-molecule inhibitors.
Collapse
Affiliation(s)
- William H. Chappell
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Stephen L. Abrams
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Richard A. Franklin
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Michelle M. LaHair
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Giuseppe Montalto
- Department of Internal Medicine and Specialties; University of Palermo; Palermo, Italy
- Consiglio Nazionale delle Ricerche; Istituto di Biomedicina e Immunologia Molecolare “Alberto Monroy”; Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche; Istituto di Biomedicina e Immunologia Molecolare “Alberto Monroy”; Palermo, Italy
| | - Alberto M. Martelli
- Department of Biomedical and Neuromotor Sciences; Università di Bologna; Bologna, Italy
- Institute of Molecular Genetics; National Research Council-Rizzoli Orthopedic Institute; Bologna, Italy
| | | | - Saverio Candido
- Department of Bio-Medical Sciences; University of Catania; Catania, Italy
| | - Massimo Libra
- Department of Bio-Medical Sciences; University of Catania; Catania, Italy
| | - Jerry Polesel
- Unit of Epidemiology and Biostatistics; Centro di Riferimento Oncologico; IRCCS; Aviano, Italy
| | - Renato Talamini
- Unit of Epidemiology and Biostatistics; Centro di Riferimento Oncologico; IRCCS; Aviano, Italy
| | | | - Agostino Tafuri
- Department of Cellular Biotechnology and Hematology; University of Rome, Sapienza; Rome, Italy
| | - Linda S. Steelman
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - James A. McCubrey
- Department of Microbiology & Immunology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| |
Collapse
|
25
|
Rim HK, Lee HW, Choi IS, Park JY, Choi HW, Choi JH, Cho YW, Lee JY, Lee KT. T-type Ca2+ channel blocker, KYS05047 induces G1 phase cell cycle arrest by decreasing intracellular Ca2+ levels in human lung adenocarcinoma A549 cells. Bioorg Med Chem Lett 2012; 22:7123-6. [PMID: 23079520 DOI: 10.1016/j.bmcl.2012.09.076] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 09/04/2012] [Accepted: 09/21/2012] [Indexed: 01/15/2023]
Abstract
In a previous study, we found that the 3,4-dihydroquinazoline derivative, 4-(Benzylcarbamoylmethyl)-2-(biphenyl-4-ylamino)-3-(5-tert-butyloxycarbamoyl-1-pentyl)-3,4-dihydroquinazoline (KYS05047), was a selective T-type Ca(2+) channel blocker with anti-proliferative effects against various cancer cells. However, the mechanism responsible for its effects has not been studied. In this study, we investigated the effect of KYS05047 on cell cycle arrest and the mechanisms involved in human lung adenocarcinoma A549 cells. Among the G(1) phase cell cycle-related proteins examined, the levels of cyclin-dependent protein kinase (Cdk2) and Cdk4 were reduced by KYS05047 (7 μM), whereas the steady-state levels of cyclin D1 and E were unaffected. In addition, KYS05047 increased the protein level of p27(KIP1) and suppressed the kinase activities of Cdk2 and Cdk4. In addition, pretreatment with KCl, which increases intracellular Ca(2+) levels, prevented KYS05047-induced intracellular Ca(2+) decreases and cell cycle arrest. Furthermore, the administration of KYS05047 (2 or 10 mg/kg, po) for 21 days was also found to significantly inhibit tumor growth in an A549 xenograft nude mice model. In conclusion, our results suggested that KYS05047 induced G(1) phase cell cycle arrest in A549 cells associated with a decrease in intracellular Ca(2+) concentrations and inhibited the in vivo tumor growth of A549 xenograft mice.
Collapse
Affiliation(s)
- Hong-Kun Rim
- Department of Pharmaceutical Biochemistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Karacosta LG, Foster BA, Azabdaftari G, Feliciano DM, Edelman AM. A regulatory feedback loop between Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2) and the androgen receptor in prostate cancer progression. J Biol Chem 2012; 287:24832-43. [PMID: 22654108 DOI: 10.1074/jbc.m112.370783] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The androgen receptor (AR) plays a critical role in prostate cancer (PCa) progression, however, the molecular mechanisms by which the AR regulates cell proliferation in androgen-dependent and castration-resistant PCa are incompletely understood. We report that Ca(2+)/calmodulin-dependent kinase kinase 2 (CaMKK2) expression increases and becomes nuclear or perinuclear in advanced PCa. In the TRAMP (transgenic adenocarcinoma of mouse prostate) model of PCa, CaMKK2 expression increases with PCa progression with many cells exhibiting nuclear staining. CaMKK2 expression is higher in human castration-resistant tumor xenografts compared with androgen-responsive xenografts and is markedly higher in the AR-expressing, tumorigenic cell line LNCaP compared with cell lines that are AR-nonexpressing and/or nontumorigenic. In LNCaP cells, dihydrotestosterone induced CaMKK2 mRNA and protein expression and translocation of CaMKK2 to the nucleus. Conversely, androgen withdrawal suppressed CaMKK2 expression. Knockdown of CaMKK2 expression by RNAi reduced LNCaP cell proliferation and increased percentages of cells in G(1) phase, whereas correspondingly reducing percentages in S phase, of the cell cycle. CaMKK2 knockdown reduced expression of the AR target gene prostate-specific antigen at both mRNA and protein levels, AR transcriptional activity driven by androgen responsive elements from the prostate-specific probasin gene promoter and levels of the AR-regulated cell cycle proteins, cyclin D1 and hyperphosphorylated Rb. Our results suggest that in PCa progression, CaMKK2 and the AR are in a feedback loop in which CaMKK2 is induced by the AR to maintain AR activity, AR-dependent cell cycle control, and continued cell proliferation.
Collapse
Affiliation(s)
- Loukia G Karacosta
- Department of Pharmacology and Toxicology, State University of New York, Buffalo, New York 14214, USA
| | | | | | | | | |
Collapse
|
27
|
Calcium/calmodulin-dependent kinase inhibitor induces growth inhibition, cell cycle arrest, and apoptosis in human choriocarcinoma cells. Tumour Biol 2012; 33:1053-8. [PMID: 22290426 DOI: 10.1007/s13277-012-0339-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 01/19/2012] [Indexed: 10/14/2022] Open
Abstract
KN-93, a membrane-permeant calcium/calmodulin- dependent kinase-selective inhibitor, induces apoptosis in some lines of human tumor cells. We investigated the effect of KN-93 in the choriocarcinoma cell line, BeWo. BeWo cells were treated with various concentrations of KN-93, and changes in cell growth, the cell cycle, apoptosis, and related parameters were examined. A WST-1 assay showed that BeWo cells were sensitive to the growth inhibitory effect of KN-93. Cell cycle analysis indicated that exposure to KN-93 decreased the proportion of cells in the S phase and increased the proportion in the G0/G1 phases of the cell cycle. Induction of apoptosis was confirmed by Annexin V staining of externalized phosphatidylserine, by the loss of mitochondrial transmembrane potential, and by antibodies directed against histones from fragmented DNA. This induction occurred in conjunction with the altered expression of genes related to cell growth, malignant phenotype, and apoptosis. These results suggest that KN-93 may serve as a therapeutic agent for the treatment of choriocarcinoma.
Collapse
|
28
|
Ramakrishnan R, Rice AP. Cdk9 T-loop phosphorylation is regulated by the calcium signaling pathway. J Cell Physiol 2012; 227:609-17. [PMID: 21448926 DOI: 10.1002/jcp.22760] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Eukaryotic RNA polymerase II transcriptional elongation is a tightly regulated process and is dependent upon positive transcription elongation factor-b (P-TEFb). The core P-TEFb complex is composed of Cdk9 and Cyclin T and is essential for the expression of most protein coding genes. Cdk9 kinase function is dependent upon phosphorylation of Thr186 in its T-loop. In this study, we examined kinases and signaling pathways that influence Cdk9 T-loop phosphorylation. Using an RNAi screen in HeLa cells, we found that Cdk9 T-loop phosphorylation is regulated by Ca(2+)/calmodulin-dependent kinase 1D (CaMK1D). Using small molecules inhibitors in HeLa cells and primary CD4(+) T lymphocytes, we found that the Ca(2+) signaling pathway is required for Cdk9 T-loop phosphorylation. Inhibition of Ca(2+) signaling led to dephosphorylation of Thr186 on Cdk9. In reporter plasmid assays, inhibition of the Ca(2+) signaling pathway repressed the PCNA promoter and HIV-1 Tat transactivation of the HIV-1 LTR, but not HTLV-1 Tax transactivation of the HTLV-1 LTR, suggesting that perturbation of the Ca(2+) pathway and reduction of Cdk9 T-loop phosphorylation inhibits transcription units that have a rigorous requirement for P-TEFb function.
Collapse
Affiliation(s)
- Rajesh Ramakrishnan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|
29
|
Wallace JL, Gow IF, Warnock M. The life and death of breast cancer cells: proposing a role for the effects of phytoestrogens on potassium channels. J Membr Biol 2011; 242:53-67. [PMID: 21728044 DOI: 10.1007/s00232-011-9376-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 06/20/2011] [Indexed: 10/18/2022]
Abstract
Changes in the regulation of potassium channels are increasingly implicated in the altered activity of breast cancer cells. Increased or reduced expression of a number of K(+) channels have been identified in numerous breast cancer cell lines and cancerous tissue biopsy samples, compared to normal tissue, and are associated with tumor formation and spread, enhanced levels of proliferation, and resistance to apoptotic stimuli. Through knockout or silencing of K(+) channel genes, and use of specific or more broad pharmacologic K(+) channel blockers, the growth of numerous cell lines, including breast cancer cells, has been modified. In this manner it has been proposed that in MCF7 breast cancer cells proliferation appears to be regulated by the activity of a number of K(+) channels, including the Ca(2+) activated K(+) channels, and the voltage-gated K(+) channels hEAG and K(v)1.1. The effect of phytoestrogens on K(+) channels has not been extensively studied but yields some interesting results. In a number of cell lines the phytoestrogen genistein inhibits K(+) current through several channels including K(v)1.3 and hERG. Where it has been used, structurally similar daidzein has little or no effect on K(+) channel activity. Since many K(+) channels have roles in proliferation and apoptosis in breast cancer cells, the impact of K(+) channel regulation by phytoestrogens is of potentially great relevance.
Collapse
Affiliation(s)
- Joanne L Wallace
- School of Health Sciences, Queen Margaret University, Musselburgh, Edinburgh, Scotland, UK.
| | | | | |
Collapse
|
30
|
Calmodulin Involved in The Cell Proliferation of Root Apical Meristem and ABA Response in Arabidopsis*. PROG BIOCHEM BIOPHYS 2011. [DOI: 10.3724/sp.j.1206.2010.00441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
31
|
Curry MC, Roberts-Thomson SJ, Monteith GR. Plasma membrane calcium ATPases and cancer. Biofactors 2011; 37:132-8. [PMID: 21674637 DOI: 10.1002/biof.146] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 12/15/2010] [Indexed: 01/12/2023]
Abstract
The plasma membrane calcium ATPases (PMCAs) are vital regulators of basal Ca(2+) and shape the nature of intracellular free Ca(2+) transients after cellular stimuli and are thus regulators of a plethora of cellular processes. Studies spanning many years have identified that at least some cancers are associated with a remodeling of PMCA isoform expression. This alteration in Ca(2+) efflux capacity may have a variety of consequences including reduced sensitivity to apoptosis and increases in the responsiveness of cancer cells to proliferative stimuli. In this review we provide an overview of studies focused on PMCAs in the context of cancer. We discuss how the remodeling of PMCA expression could provide a survival and/or growth advantage to cancer cells, as well as the potential of pharmacological agents that target specific PMCA isoforms to be novel therapies for the treatment of cancer.
Collapse
Affiliation(s)
- Merril C Curry
- The University of Queensland, School of Pharmacy, Brisbane, QLD, Australia
| | | | | |
Collapse
|
32
|
Gilot D, Le Meur N, Giudicelli F, Le Vée M, Lagadic-Gossmann D, Théret N, Fardel O. RNAi-based screening identifies kinases interfering with dioxin-mediated up-regulation of CYP1A1 activity. PLoS One 2011; 6:e18261. [PMID: 21479225 PMCID: PMC3066211 DOI: 10.1371/journal.pone.0018261] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 02/23/2011] [Indexed: 11/18/2022] Open
Abstract
Background The aryl hydrocarbon receptor (AhR) is a transcription factor activated by several environmental pollutants, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), and involved in carcinogenesis and various physiological processes, including immune response and endocrine functions. Characterization of kinases-related AhR transduction pathway remains an important purpose. Results We performed a kinome-wide siRNA screen in human mammary MCF-7 cells to identify non redundant protein kinases implicated in the up-regulation of cytochrome P-450 (CYP) 1A1 activity, an AhR referent target, in response to TCDD exposure. To this aim, we monitored CYP1A1-related ethoxyresorufin-O-deethylase (EROD) activity and quantified cell density. This normalization was crucial since it allowed us to focus only on siRNA affecting EROD activity and discard siRNA affecting cell density. Analyses of the cell density data allowed us to identify several hits already well-characterized as effectors of the cell cycle and original hits. Collectively, these data fully validated the protocol and the siRNA library. Next, 22 novel candidates were identified as kinases potentially implicated in the up-regulation of CYP1A1 in response to TCDD, without alteration of cell survival or cell proliferation. The siRNA library screen gave a limited number of hits (approximately 3%). Interestingly, four of them are able to bind calmodulin among which the IP3 kinase A (ITPKA) and pregnancy up-regulated non-ubiquitously expressed CaM kinase (PNCK, also named CaMKIβ). Remarkably, for both proteins, their kinase activity depends on the calmodulin binding. Involvement of ITPKA and PNCK in TCDD-mediated CYP1A1 up-regulation was further validated by screening-independent expression knock-down. PNCK was finally shown to regulate activation of CaMKIα, a CaMKI isoform previously reported to interplay with the AhR pathway. Conclusions These data fully support a role for both IP3-related kinase and CaMK isoforms in the AhR signaling cascade. More generally, this study also highlights the interest of large scale loss-of-function screens for characterizing the molecular mechanism of action of environmental contaminants.
Collapse
Affiliation(s)
- David Gilot
- EA 4427 Signalisation et Réponse aux Agents Infectieux et Chimiques, Université de Rennes 1, Institut de Recherche Santé, Environnement et Travail, Institut Fédératif de Recherche 140, Rennes, France.
| | | | | | | | | | | | | |
Collapse
|
33
|
Song MY, Makino A, Yuan JXJ. STIM2 Contributes to Enhanced Store-operated Ca Entry in Pulmonary Artery Smooth Muscle Cells from Patients with Idiopathic Pulmonary Arterial Hypertension. Pulm Circ 2011; 1:84-94. [PMID: 21709766 PMCID: PMC3121304 DOI: 10.4103/2045-8932.78106] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pulmonary vasoconstriction and vascular remodeling are two major causes for elevated pulmonary vascular resistance and pulmonary arterial pressure in patients with idiopathic pulmonary arterial hypertension (IPAH). An increase in cytosolic free Ca2+concentration ([Ca2+]cyt) in pulmonary artery smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction and an important stimulus for PASMC proliferation, which causes pulmonary vascular remodeling. Store-operated Ca2+ entry (SOCE), induced by depletion of stored Ca2+ in the sarcoplasmic reticulum (SR), can increase [Ca2+]cyt in PASMC, independent of other means of Ca2+ entry. Stromal interaction molecule (STIM) proteins, STIM1 and STIM2, were both recently identified as sensors for store depletion and also signaling molecules to open store-operated Ca2+ channels. We previously reported that SOCE was significantly enhanced in PASMC from IPAH patients compared to PASMC from normotensive control subjects. Enhanced SOCE plays an important role in the pathophysiological changes in PASMC associated with pulmonary arterial hypertension. In this study, we examine whether the expression levels of STIM1 and STIM2 are altered in IPAH-PASMC compared to control PASMC, and whether these putative changes in the STIM1 and STIM2 expression levels are responsible for enhanced SOCE and proliferation in IPAH-PASMC. Compared to control PASMC, the protein expression level of STIM2 was significantly increased in IPAH-PASMC, whereas STIM1 protein expression was not significantly changed. In IPAH-PASMC, the small interfering RNA (siRNA)-mediated knockdown of STIM2 decreased SOCE and proliferation, while knockdown of STIM2 in control PASMC had no effect on either SOCE or proliferation. Overexpression of STIM2 in the control PASMC failed to enhance SOCE or proliferation. These data indicate that enhanced protein expression of STIM2 is necessary, but not sufficient, for enhanced SOCE and proliferation of IPAH-PASMC.
Collapse
Affiliation(s)
- Michael Y Song
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093-0725
| | | | | |
Collapse
|
34
|
Ding X, He Z, Zhou K, Cheng J, Yao H, Lu D, Cai R, Jin Y, Dong B, Xu Y, Wang Y. Essential role of TRPC6 channels in G2/M phase transition and development of human glioma. J Natl Cancer Inst 2010; 102:1052-68. [PMID: 20554944 DOI: 10.1093/jnci/djq217] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Patients with glioblastoma multiforme, the most aggressive form of glioma, have a median survival of approximately 12 months. Calcium (Ca(2+)) signaling plays an important role in cell proliferation, and some members of the Ca(2+)-permeable transient receptor potential canonical (TRPC) family of channel proteins have demonstrated a role in the proliferation of many types of cancer cells. In this study, we investigated the role of TRPC6 in cell cycle progression and in the development of human glioma. METHODS TRPC6 protein and mRNA expression were assessed in glioma (n = 33) and normal (n = 17) brain tissues from patients and in human glioma cell lines U251, U87, and T98G. Activation of TRPC6 channels was tested by platelet-derived growth factor-induced Ca(2+) imaging. The effect of inhibiting TRPC6 activity or expression using the dominant-negative mutant TRPC6 (DNC6) or RNA interference, respectively, was tested on cell growth, cell cycle progression, radiosensitization of glioma cells, and development of xenografted human gliomas in a mouse model. The green fluorescent protein (GFP) and wild-type TRPC6 (WTC6) were used as controls. Survival of mice bearing xenografted tumors in the GFP, DNC6, and WTC6 groups (n = 13, 15, and 13, respectively) was compared using Kaplan-Meier analysis. All statistical tests were two-sided. RESULTS Functional TRPC6 was overexpressed in human glioma cells. Inhibition of TRPC6 activity or expression attenuated the increase in intracellular Ca(2+) by platelet-derived growth factor, suppressed cell growth and clonogenic ability, induced cell cycle arrest at the G2/M phase, and enhanced the antiproliferative effect of ionizing radiation. Cyclin-dependent kinase 1 activation and cell division cycle 25 homolog C expression regulated the cell cycle arrest. Inhibition of TRPC6 activity also reduced tumor volume in a subcutaneous mouse model of xenografted human tumors (P = .014 vs GFP; P < .001 vs WTC6) and increased mean survival in mice in an intracranial model (P < .001 vs GFP or WTC6). CONCLUSIONS In this preclinical model, TRPC6 channels were essential for glioma development via regulation of G2/M phase transition. This study suggests that TRPC6 might be a new target for therapeutic intervention of human glioma.
Collapse
Affiliation(s)
- Xia Ding
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Shanghai Institute for Biological Sciences, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Jung HJ, Kim JH, Shim JS, Kwon HJ. A novel Ca2+/calmodulin antagonist HBC inhibits angiogenesis and down-regulates hypoxia-inducible factor. J Biol Chem 2010; 285:25867-74. [PMID: 20554536 DOI: 10.1074/jbc.m110.135632] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent reports have shown that Ca(2+)/calmodulin (Ca(2+)/CaM) signaling plays a crucial role in angiogenesis. We previously developed a new Ca(2+)/CaM antagonist, HBC (4-{3,5-bis-[2-(4-hydroxy-3-methoxyphenyl)ethyl]-4,5-dihydropyrazol-1-yl}benzoic acid), from a curcumin-based synthetic chemical library. Here, we investigated its anti-angiogenic activity and mode of action. HBC potently inhibited the proliferation of human umbilical vascular endothelial cells with no cytotoxicity. Furthermore, HBC blocked in vitro characteristics of angiogenesis such as tube formation and chemoinvasion, as well as neovascularization of the chorioallantoic membrane of growing chick embryos in vivo. Notably, HBC markedly inhibited expression of hypoxia-inducible factor-1alpha (HIF-1alpha) at the translational level during hypoxia, thereby reducing HIF-1 transcriptional activity and expression of its major target gene, vascular endothelial growth factor. In addition, combination treatment with HBC and various HIF-1 inhibitors, including suberoylanilide hydroxamic acid, rapamycin, and terpestacin, had greater anti-angiogenic activity than treatment with each single agent. Collectively, our findings indicate that HBC is a new anti-angiogenic agent targeting HIF that can be used to explore the biological role of Ca(2+)/CaM in angiogenesis.
Collapse
Affiliation(s)
- Hye Jin Jung
- Chemical Genomics Laboratory, Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | | | | | | |
Collapse
|
36
|
Ding X, He Z, Shi Y, Wang Q, Wang Y. Targeting TRPC6 channels in oesophageal carcinoma growth. Expert Opin Ther Targets 2010; 14:513-27. [DOI: 10.1517/14728221003733602] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
37
|
Ma J, Harnett KM, Behar J, Biancani P, Cao W. Signaling in TRPV1-induced platelet activating factor (PAF) in human esophageal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2010; 298:G233-40. [PMID: 19959817 PMCID: PMC2822503 DOI: 10.1152/ajpgi.00409.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Transient receptor potential channel, vanilloid subfamily member 1 (TRPV1) receptors were identified in human esophageal squamous epithelial cell line HET-1A by RT-PCR and by Western blot. In fura-2 AM-loaded cells, the TRPV1 agonist capsaicin caused a fourfold cytosolic calcium increase, supporting a role of TRPV1 as a capsaicin-activated cation channel. Capsaicin increased production of platelet activating factor (PAF), an important inflammatory mediator that acts as a chemoattractant and activator of immune cells. The increase was reduced by the p38 MAP kinase (p38) inhibitor SB203580, by the cytosolic phospholipase A2 (cPLA(2)) inhibitor AACOCF3, and by the lyso-PAF acetyltransferase inhibitor sanguinarin, indicating that capsaicin-induced PAF production may be mediated by activation of cPLA(2), p38, and lyso-PAF acetyltransferase. To establish a sequential signaling pathway, we examined the phosphorylation of p38 and cPLA(2) by Western blot. Capsaicin induced phosphorylation of p38 and cPLA(2). Capsaicin-induced p38 phosphorylation was not affected by AACOCF3. Conversely, capsaicin-induced cPLA(2) phosphorylation was blocked by SB203580, indicating that capsaicin-induced PAF production depends on sequential activation of p38 and cPLA(2). To investigate how p38 phosphorylation may result from TRPV1-mediated calcium influx, we examined a possible role of calmodulin kinase (CaM-K). p38 phosphorylation was stimulated by the calcium ionophore A23187 and by capsaicin, and the response to both agonists was reduced by a CaM inhibitor and by CaM-KII inhibitors, indicating that calcium induced activation of CaM and CaM-KII results in P38 phosphorylation. Acetyl-CoA transferase activity increased in response to capsaicin and was inhibited by SB203580, indicating that p38 phosphorylation in turn causes activation of acetyl-CoA transferase to produce PAF. Thus epithelial cells produce PAF in response to TRPV1-mediated calcium elevation.
Collapse
Affiliation(s)
- Jie Ma
- 1Department of Medicine, Rhode Island Hospital and Brown University, Providence, Rhode Island; ,2School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Karen M. Harnett
- 1Department of Medicine, Rhode Island Hospital and Brown University, Providence, Rhode Island;
| | - Jose Behar
- 1Department of Medicine, Rhode Island Hospital and Brown University, Providence, Rhode Island;
| | - Piero Biancani
- 1Department of Medicine, Rhode Island Hospital and Brown University, Providence, Rhode Island;
| | - Weibiao Cao
- 1Department of Medicine, Rhode Island Hospital and Brown University, Providence, Rhode Island;
| |
Collapse
|
38
|
McCubrey JA, Abrams SL, Stadelman K, Chappell WH, Lahair M, Ferland RA, Steelman LS. Targeting signal transduction pathways to eliminate chemotherapeutic drug resistance and cancer stem cells. ADVANCES IN ENZYME REGULATION 2009; 50:285-307. [PMID: 19895837 PMCID: PMC2862855 DOI: 10.1016/j.advenzreg.2009.10.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
39
|
Han EH, Kim HG, Im JH, Jeong TC, Jeong HG. Up-regulation of CYP1A1 by rutaecarpine is dependent on aryl hydrocarbon receptor and calcium. Toxicology 2009; 266:38-47. [PMID: 19853001 DOI: 10.1016/j.tox.2009.10.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 10/12/2009] [Accepted: 10/12/2009] [Indexed: 01/31/2023]
Abstract
Rutaecarpine is a quinazolinocarboline alkaloid isolated from a traditional Chinese medicinal fruit, Evodia rutaecarpa. In the present study, we investigated the effect of rutaecarpine on CYP1A1 expression mediated by [Ca(2+)] and the AhR pathway in mouse hepatoma Hepa-1c1c7 cells. Rutaecarpine also significantly increased CYP1A1 enzyme activity and mRNA and protein levels. Rutaecarpine markedly induced XRE and AhR binding activity. CH-223191, an AhR antagonist, blocked the rutaecarpine-induced CYP1A1 enzyme activity and mRNA and protein expression. In addition, rutaecarpine remarkably induced the phosphorylation of Ca(2+)/calmodulin (CaM)-dependent protein kinase (CaMK). W7 and BAPTA/AM, a CaM antagonist and an intracellular Ca(2+) chelator, respectively, blocked the rutaecarpine-induced CYP1A1 enzyme activity and mRNA and protein expression. These results indicate that rutaecarpine induces CYP1A1 expression through AhR- and calcium-dependent mechanisms.
Collapse
Affiliation(s)
- Eun Hee Han
- Department of Toxicology, College of Pharmacy, Chungnam National University, 220 Gung-dong, Yuseong-Gu, Daejeon 305-764, South Korea
| | | | | | | | | |
Collapse
|
40
|
ERK activation and cell growth require CaM kinases in MCF-7 breast cancer cells. Mol Cell Biochem 2009; 335:155-71. [DOI: 10.1007/s11010-009-0252-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 09/02/2009] [Indexed: 10/20/2022]
|
41
|
Gattenlöhner S, Stühmer T, Leich E, Reinhard M, Etschmann B, Völker HU, Rosenwald A, Serfling E, Bargou RC, Ertl G, Einsele H, Müller-Hermelink HK. Specific detection of CD56 (NCAM) isoforms for the identification of aggressive malignant neoplasms with progressive development. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1160-71. [PMID: 19246644 DOI: 10.2353/ajpath.2009.080647] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alternative splicing of transcripts from many cancer-associated genes is believed to play a major role in carcinogenesis as well as in tumor progression. Alternative splicing of one such gene, the neural cell adhesion molecule CD56 (NCAM), impacts the progression, inadequate therapeutic response, and reduced total survival of patients who suffer from numerous malignant neoplasms. Although previous investigations have determined that CD56 exists in three major isoforms (CD56(120kD), CD56(140kD), and CD56(180kD)) with individual structural and functional properties, neither the expression profiles nor the functional relevance of these isoforms in malignant tumors have been consistently investigated. Using new quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) strategies and novel CD56 isoform-specific antibodies, CD56(140kD) was shown to be exclusively expressed in a number of highly malignant CD56(+) neoplasms and was associated with the progression of CD56(+) precursor lesions of unclear malignant potential. Moreover, only CD56(140kD) induced antiapoptotic/proliferative pathways and specifically phosphorylated calcium-dependent kinases that are relevant for tumorigenesis. We conclude, therefore, that the specific detection of CD56 isoforms will help to elucidate their individual functions in the pathogenesis and progression of malignant neoplasms and may have a positive impact on the development of CD56-based immunotherapeutic strategies.
Collapse
Affiliation(s)
- Stefan Gattenlöhner
- Institute of Pathology, University of Würzburg, Josef-Schneiderstr.2, D-97080 Würzburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Takai N, Ueda T, Nasu K, Yamashita S, Toyofuku M, Narahara H. Targeting calcium/calmodulin-dependence kinase I and II as a potential anti-proliferation remedy for endometrial carcinomas. Cancer Lett 2009; 277:235-43. [PMID: 19168280 DOI: 10.1016/j.canlet.2008.12.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 12/09/2008] [Accepted: 12/09/2008] [Indexed: 11/17/2022]
Abstract
Calcium/calmodulin-dependent kinase (CaMK) I and II expression in endometrial cancer cells correlates with the malignant potential of this tumor, and CaMKI and II are potential therapeutic targets in endometrial cancer. CaMKI and II expression was significantly associated with PCNA-labeling index, clinical stage, histological grade, the presence of invasion to greater than one-half the myometrium, and clinical outcome. All endometrial cancer cell lines examined were sensitive to the growth-inhibitory effect of KN-93, a membrane-permeant CaMKs-selective inhibitor that is competitive with calmodulin. KN-93 induced the G0/G1 arrest and apoptosis, rising hopes that KN-93 may become a useful treatment for endometrial cancers.
Collapse
Affiliation(s)
- Noriyuki Takai
- Department of Obstetrics and Gynecology, Oita University Faculty of Medicine, 1-1 Idaigaioka, Hasama-machi, Yufu-shi, Oita, Japan.
| | | | | | | | | | | |
Collapse
|
43
|
Bergamaschi A, Kim YH, Kwei KA, La Choi Y, Bocanegra M, Langerød A, Han W, Noh DY, Huntsman DG, Jeffrey SS, Børresen-Dale AL, Pollack JR. CAMK1D amplification implicated in epithelial-mesenchymal transition in basal-like breast cancer. Mol Oncol 2008; 2:327-39. [PMID: 19383354 DOI: 10.1016/j.molonc.2008.09.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Revised: 09/17/2008] [Accepted: 09/17/2008] [Indexed: 01/27/2023] Open
Abstract
Breast cancer exhibits clinical and molecular heterogeneity, where expression profiling studies have identified five major molecular subtypes. The basal-like subtype, expressing basal epithelial markers and negative for estrogen receptor (ER), progesterone receptor (PR) and HER2, is associated with higher overall levels of DNA copy number alteration (CNA), specific CNAs (like gain on chromosome 10p), and poor prognosis. Discovering the molecular genetic basis of tumor subtypes may provide new opportunities for therapy. To identify the driver oncogene on 10p associated with basal-like tumors, we analyzed genomic profiles of 172 breast carcinomas. The smallest shared region of gain spanned just seven genes at 10p13, including calcium/calmodulin-dependent protein kinase ID (CAMK1D), functioning in intracellular signaling but not previously linked to cancer. By microarray, CAMK1D was overexpressed when amplified, and by immunohistochemistry exhibited elevated expression in invasive carcinomas compared to carcinoma in situ. Engineered overexpression of CAMK1D in non-tumorigenic breast epithelial cells led to increased cell proliferation, and molecular and phenotypic alterations indicative of epithelial-mesenchymal transition (EMT), including loss of cell-cell adhesions and increased cell migration and invasion. Our findings identify CAMK1D as a novel amplified oncogene linked to EMT in breast cancer, and as a potential therapeutic target with particular relevance to clinically unfavorable basal-like tumors.
Collapse
Affiliation(s)
- Anna Bergamaschi
- Department of Genetics, Institute for Cancer Research, Rikshospitalet-Radiumhospitalet Medical Center, Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pedersen ME, Fortunati D, Nielsen M, Brorson SH, Lekva T, Nissen-Meyer LSH, Gautvik VT, Shahdadfar A, Gautvik KM, Jemtland R. Calmodulin-dependent kinase 1beta is expressed in the epiphyseal growth plate and regulates proliferation of mouse calvarial osteoblasts in vitro. Bone 2008; 43:700-7. [PMID: 18620088 DOI: 10.1016/j.bone.2008.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 04/08/2008] [Accepted: 06/02/2008] [Indexed: 11/20/2022]
Abstract
The Ca(2+)/Calmodulin-dependent protein kinase (CaMK) family is activated in response to elevation of intracellular Ca(2+), and includes CaMK1 (as well as CaMK2 and CaMK4), which exists as different isoforms (alpha, beta, gamma and delta). CaMK1 is present in several cell types and may be involved in various cellular processes, but its role in bone is unknown. In situ hybridization was used to determine the spatial and temporal expression of CaMK1beta during endochondral bone development in mouse embryos and newborn pups. The cellular and subcellular distribution of CaMK1 was assessed by quantitative immunogold electron microscopy (EM). The role of CaMK1beta in mouse calvarial osteoblasts was investigated by using small interfering RNA (siRNA) to silence its expression, while in parallel monitoring cell proliferation and levels of skeletogenic transcripts. cRNA in situ hybridization and EM studies show that CaMK1beta is mainly located in developing long bones and vertebrae (from ED14.5 until day 10 after birth), with highest expression in epiphyseal growth plate hypertrophic chondrocytes. By RT-PCR, we show that CaMK1beta2 (but not beta1) is expressed in mouse hind limbs (in vivo) and mouse calvarial osteoblasts (in vitro), and also in primary human articular chondrocyte cultures. Silencing of CaMK1beta in mouse calvarial osteoblasts by siRNA significantly decreases osteoblast proliferation and c-Fos gene expression (approx. 50%), without affecting skeletogenic markers for more differentiated osteoblasts (i.e. Cbfa1/Runx2, Osterix (Osx), Osteocalcin (Oc), Alkaline phosphatase (Alp) and Osteopontin (Opn)). These results identify CaMK1beta as a novel regulator of osteoblast proliferation, via mechanisms that may at least in part involve c-Fos, thus implicating CaMK1beta in the regulation of bone and cartilage development.
Collapse
Affiliation(s)
- Mona E Pedersen
- Institute of Basic Medical Sciences, Department of Biochemistry, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Long JM, Bell CW, Fagg WS, Kushman ME, Becker KG, McCubrey JA, Farwell MA. Microarray and pathway analysis reveals decreased CDC25A and increased CDC42 associated with slow growth of BCL2 overexpressing immortalized breast cell line. Cell Cycle 2008; 7:3062-73. [PMID: 18838868 PMCID: PMC2634598 DOI: 10.4161/cc.7.19.6761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Bcl-2 is an anti-apoptotic protein that is frequently overexpressed in cancer cells but its role in carcinogenesis is not clear. We are interested in how Bcl-2 expression affects non-cancerous breast cells and its role in the cell cycle. We prepared an MCF10A breast epithelial cell line that stably overexpressed Bcl-2. We analyzed the cells by flow cytometry after synchronization, and used cDNA microarrays with quantitative reverse-transcription PCR (qRT-PCR) to determine differences in gene expression. The microarray data was subjected to two pathway analysis tools, parametric analysis of gene set enrichment (PAGE) and ingenuity pathway analysis (IPA), and western analysis was carried out to determine the correlation between mRNA and protein levels. The MCF10A/Bcl-2 cells exhibited a slow-growth phenotype compared to control MCF10A/Neo cells that we attributed to a slowing of the G(1)-S cell cycle transition. A total of 363 genes were differentially expressed by at least two-fold, 307 upregulated and 56 downregulated. PAGE identified 22 significantly changed gene sets. The highest ranked network of genes identified by IPA contained 24 genes. Genes that were chosen for further analysis were confirmed by qRT-PCR, however, the western analysis did not always confirm differential expression of the proteins. Downregulation of the phosphatase CDC25A could solely be responsible for the slow growth phenotype in MCF10A/Bcl-2 cells. Increased levels of GTPase Cdc42 could be adding to this effect. PAGE and IPA are valuable tools for microarray analysis, but protein expression results do not always follow mRNA expression results.
Collapse
Affiliation(s)
- Jacquelyn M Long
- Department of Biology, East Carolina University, Greenville, North Carolina 27858, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Roderick HL, Cook SJ. Ca2+ signalling checkpoints in cancer: remodelling Ca2+ for cancer cell proliferation and survival. Nat Rev Cancer 2008; 8:361-75. [PMID: 18432251 DOI: 10.1038/nrc2374] [Citation(s) in RCA: 542] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increases in cytosolic free Ca2+ ([Ca2+]i) represent a ubiquitous signalling mechanism that controls a variety of cellular processes, including proliferation, metabolism and gene transcription, yet under certain conditions increases in intracellular Ca2+ are cytotoxic. Thus, in using Ca2+ as a messenger, cells walk a tightrope in which [Ca2+]i is strictly maintained within defined boundaries. To adhere to these boundaries and to sustain their modified phenotype, many cancer cells remodel the expression or activity of their Ca2+ signalling apparatus. Here, we review the role of Ca2+ in promoting cell proliferation and cell death, how these processes are remodelled in cancer and the opportunities this might provide for therapeutic intervention.
Collapse
Affiliation(s)
- H Llewelyn Roderick
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK.
| | | |
Collapse
|
47
|
A cascade of Ca(2+)/calmodulin-dependent protein kinases regulates the differentiation and functional activation of murine neutrophils. Exp Hematol 2008; 36:832-44. [PMID: 18400360 DOI: 10.1016/j.exphem.2008.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 01/28/2008] [Accepted: 02/14/2008] [Indexed: 01/21/2023]
Abstract
OBJECTIVE The function of neutrophils as primary mediators of innate immunity depends on the activity of granule proteins and critical components of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex. Expression of their cognate genes is regulated during neutrophil differentiation by a complex network of intracellular signaling pathways. In this study, we have investigated the role of two members of the calcium/calmodulin-dependent protein kinase (CaMK) signaling cascade, CaMK I-like kinase (CKLiK) and CaMKKalpha, in regulating neutrophil differentiation and functional activation. MATERIALS AND METHODS Mouse myeloid cell lines were used to examine the expression of a CaMK cascade in developing neutrophils and to examine the effects of constitutive activation vs inhibition of CaMKs on neutrophil maturation. RESULTS Expression of CaMKKalpha was shown to increase during neutrophil differentiation in multiple cell lines, whereas expression of CKLiK increased as multipotent progenitors committed to promyelocytes, but then decreased as cells differentiated into mature neutrophils. Expression of constitutively active CKLiKs did not affect morphologic maturation, but caused dramatic decreases in both respiratory burst responses and chemotaxis. This loss of neutrophil function was accompanied by reduced secondary granule and gp91(phox) gene expression. The CaMK inhibitor KN-93 attenuated cytokine-stimulated proliferative responses in promyelocytic cell lines, and inhibited the respiratory burst. Similar data were observed with the CaMKKalpha inhibitor, STO-609. CONCLUSIONS Overactivation of a cascade of CaMKs inhibits neutrophil maturation, suggesting that these kinases play an antagonistic role during neutrophil differentiation, but at least one CaMK is required for myeloid cell expansion and functional activation.
Collapse
|
48
|
Colomer J, Means AR. Physiological roles of the Ca2+/CaM-dependent protein kinase cascade in health and disease. Subcell Biochem 2008; 45:169-214. [PMID: 18193638 DOI: 10.1007/978-1-4020-6191-2_7] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Numerous hormones, growth factors and physiological processes cause a rise in cytosolic Ca2+, which is translated into meaningful cellular responses by interacting with a large number of Ca2(+)-binding proteins. The Ca2(+)-binding protein that is most pervasive in mediating these responses is calmodulin (CaM), which acts as a primary receptor for Ca2+ in all eukaryotic cells. In turn, Ca2+/CaM functions as an allosteric activator of a host of enzymatic proteins including a considerable number of protein kinases. The topic of this review is to discuss the physiological roles of a sub-set of these protein kinases which can function in cells as a Ca2+/CaM-dependent kinase signaling cascade. The cascade was originally believed to consist of a CaM kinase kinase that phosphorylates and activates one of two CaM kinases, CaMKI or CaMKIV. The unusual aspect of this cascade is that both the kinase kinase and the kinase require the binding of Ca2+/CaM for activation. More recently, one of the CaM kinase kinases has been found to activate another important enzyme, the AMP-dependent protein kinase so the concept of the CaM kinase cascade must be expanded. A CaM kinase cascade is important for many normal physiological processes that when misregulated can lead to a variety of disease states. These processes include: cell proliferation and apoptosis that may conspire in the genesis of cancer; neuronal growth and function related to brain development, synaptic plasticity as well as memory formation and maintenance; proper function of the immune system including the inflammatory response, activation of T lymphocytes and hematopoietic stem cell maintenance; and the central control of energy balance that, when altered, can lead to obesity and diabetes. Although the study of the CaM-dependent kinase cascades is still in its infancy continued analysis of the pathways regulated by these Ca2(+)-initiated signaling cascades holds considerable promise for the future of disease-related research.
Collapse
Affiliation(s)
- J Colomer
- Department of Pharmacology and Cancer Biology, Duke University Medical Center USA
| | | |
Collapse
|
49
|
Monteiro P, Gilot D, Le Ferrec E, Rauch C, Lagadic-Gossmann D, Fardel O. Dioxin-Mediated Up-Regulation of Aryl Hydrocarbon Receptor Target Genes Is Dependent on the Calcium/Calmodulin/CaMKIα Pathway. Mol Pharmacol 2007; 73:769-77. [DOI: 10.1124/mol.107.043125] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
50
|
K+ channel expression in human breast cancer cells: involvement in cell cycle regulation and carcinogenesis. J Membr Biol 2007; 221:1-6. [PMID: 18060344 DOI: 10.1007/s00232-007-9080-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Accepted: 10/09/2007] [Indexed: 01/22/2023]
Abstract
K+ channels are a most diverse class of ion channels in the plasma membrane and are distributed widely throughout a variety of cells including cancer cells. Evidence has been accumulating from fundamental studies indicating that tumour cells possess various types of K+ channels and that these K+ channels play important roles in regulating tumor cell proliferation, cell cycle progression and apoptosis. Moreover, a significant increase in K+ channel expression has been correlated with tumorigenesis, suggesting the possibility of using these proteins as transformation markers and perhaps reducing the tumor growth rate by selectively inhibiting their functional activity. Significant progress has been made in defining the properties of breast K+ channels, including their biophysical and pharmacological properties and distribution throughout different phases of the cell cycle in breast cell line MCF-7. This review aims to provide a comprehensive overview of the current state of research into K+ channels/currents in breast cancer cells. The possible mechanisms by which K+ channels affect tumor cell proliferation and cell cycle progression are discussed.
Collapse
|