1
|
Demirbakan B. A highly sensitive creatine kinase detection in human serum using 11-mercaptoundecanoic acid modified ITO-PET electrodes. Anal Biochem 2025; 700:115768. [PMID: 39800130 DOI: 10.1016/j.ab.2025.115768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/28/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
The enzyme creatine kinase (CK) is a biomarker that plays an extremely significant role in the early detection of cardiovascular disorders. Serum levels of CK are regularly monitored in patients with heart attacks, one of the most critical cardiovascular illnesses. In this study, a highly sensitive electrochemical immunosensor system was designed for the importance of early diagnosis of CK. This immunosensor system was developed by immobilizing 11- mercaptoundecanoic acid (11-MuA) on disposable indium tin oxide-polyethylene terephthalate (ITO-PET) electrodes. Electrochemical impedance spectroscopy (EIS), cyclic voltammetry (CV) and single frequency impedance (SFI) techniques were utilized throughout the immobilization process during the construction of the immunosensor. In addition, the proposed CK immunosensor system involves thorough analytical research, which may include linear determination range, repeatability, reproducibility, square wave voltammetry, storage capability, and regeneration. The suggested immunosensor was also characterized using scanning electron microscopy (SEM). The proposed immunosensor system demonstrated a broad dynamic range (0.1 pg/mL - 100 pg/mL), as well as a low limit of detection (LOD) and a low limit of quantification (LOQ) of 0.018 pg/mL and 0.0394 pg/mL, respectively. Finally, the immunosensor was tested on human serum samples, proving that it could be utilized in clinical situations.
Collapse
Affiliation(s)
- Burçak Demirbakan
- Çanakkale Onsekiz Mart University, Faculty of Engineering, Bioengineering Department, Çanakkale, Turkey.
| |
Collapse
|
2
|
Fam96b recruits brain-type creatine kinase to fuel mitotic spindle formation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119410. [PMID: 36503010 DOI: 10.1016/j.bbamcr.2022.119410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
Mitosis is a complicated and ordered process with high energy demands and metabolite fluxes. Cytosolic creatine kinase (CK), an enzyme involved in ATP homeostasis, has been shown to be essential to chromosome movement during mitotic anaphase in sea urchin. However, it remains elusive for the molecular mechanism underlying the recruitment of cytosolic CK by the mitotic apparatus. In this study, Fam96b/MIP18, a component of the MMXD complex with a function in Fe/S cluster supply, was identified as a brain-type CK (CKB)-binding protein. The binding of Fam96b with CKB was independent of the presence of CKB substrates and did not interfere with CKB activity. Fam96b was prone to oligomerize via the formation of intermolecular disulfide bonds, while the binding of enzymatically active CKB could modulate Fam96b oligomerization. Oligomerized Fam96b recruited CKB and the MMXD complex to associate with the mitotic spindle. Depletion of Fam96b or CKB by siRNA in the HeLa cells led to mitotic defects, which further resulted in retarded cell proliferation, increased cell death and aberrant cell cycle progression. Rescue experiments indicated that both Fam96b oligomerization and CKB activity were essential to the proper formation of mitotic spindle. These findings suggest that Fam96b may act as a scaffold protein to coordinate the supply and homeostasis of ATP and Fe/S clusters during mitosis.
Collapse
|
3
|
Liu S, Ding Y, Yu Q, Wang X, Cheng D. Comparative study of aluminum speciation on brain-type creatine kinase: Enzyme kinetic, molecular docking, cellular experiment, and mouse model study. J Inorg Biochem 2023; 238:112032. [PMID: 36327498 DOI: 10.1016/j.jinorgbio.2022.112032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/17/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022]
Abstract
Brain-type Creatine kinase (CK-BB), which has a high affinity for Aluminum (Al), and its abnormality is closely related to neurodegenerative diseases. In this study, the comparative effect of Al speciation on the bioactivity of CK-BB has been studied by the inhibition kinetics method, molecular docking, cellular experiment, and mouse model study. Results showed that the half-inhibitory concentration of AlCl3 was 0.67 mM, while Al(mal)3 was 3.81 mM. Fluorescence spectra showed that Al(mal)3 had a more substantial effect on the endogenous fluorescence of CK-BB than AlCl3. Molecular docking showed that AlCl3 was closer to the active site of CK-BB. C6 cells were used to explore the enzyme activity and intracellular distribution of CK-BB by AlCl3 or Al(mal)3. AlCl3 treatment may directly affect CK-BB activity and cause insufficient local ATP supply in cells which affected the formation of F-actin and cell morphology. The change in the hydrophobicity of CK-BB induced by Al(mal)3 affected the movement of CK-BB, which subsequently activated thecytochrome C (Cyt C)/Caspase 9/Caspase 3 pathway. Similar results have been found in vivo experiments. This study demonstrated that interaction between Al and CK-BB might be related to the process of Al-induced energy metabolism disorders, in which the Al speciation revealed differentiated toxicity mechanisms.
Collapse
Affiliation(s)
- Sijia Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yixin Ding
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Qianqian Yu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Xuerui Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Dai Cheng
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China.
| |
Collapse
|
4
|
CKB inhibits epithelial-mesenchymal transition and prostate cancer progression by sequestering and inhibiting AKT activation. Neoplasia 2021; 23:1147-1165. [PMID: 34706306 PMCID: PMC8551525 DOI: 10.1016/j.neo.2021.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 09/17/2021] [Indexed: 12/26/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) contributes to tumor invasion, metastasis and drug resistance. AKT activation is key in a number of cellular processes. While many positive regulators for either EMT or AKT activation have been reported, few negative regulators are established. Through kinase cDNA screen, we identified brain-type creatine kinase (CKB or BCK) as a potent suppressor for both. As a ubiquitously expressed kinase in normal tissues, CKB is significantly downregulated in several solid cancer types. Lower CKB expression is significantly associated with worse prognosis. Phenotypically, CKB overexpression suppresses, while its silencing promotes, EMT and cell migration, xenograft tumor growth and metastasis of prostate cancer cells. AKT activation is one of the most prominent signaling events upon CKB silencing in prostate cancer cells, which is in line with prostate cancer TCGA data. EMT enhanced by CKB silencing is abolished by AKT inhibition. Mechanistically, CKB interacts with AKT and sequestrates it from activation by mTOR. We further elucidated that an 84aa fragment at C-terminus of CKB protein interacts with AKT's PH domain. Ectopic expression of the 84aa CKB fragment inhibits AKT activation, EMT and cell proliferation. Interestingly, molecular dynamics simulation on crystal structures of AKT and CKB independently demonstrates that AKT's PH domain and CKB's 84aa fragment establish their major interaction interface. In summary, we have discovered CKB as a negative regulator of EMT and AKT activation, revealing a new mode of their regulation . We have also demonstrated that CKB downregulation is a poor prognosticator, which is sufficient to promote prostate cancer progression.
Collapse
|
5
|
Kothari A, Flick MJ. Coagulation Signaling through PAR1 as a Therapeutic Target in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2021; 22:ijms22105138. [PMID: 34066284 PMCID: PMC8152032 DOI: 10.3390/ijms22105138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal disease with a 5-year survival rate of less than 10% following diagnosis. The aggressive and invasive properties of pancreatic cancer tumors coupled with poor diagnostic options contribute to the high mortality rate since most patients present with late-stage disease. Accordingly, PDAC is linked to the highest rate of cancer-associated venous thromboembolic disease of all solid tumor malignancies. However, in addition to promoting clot formation, recent studies suggest that the coagulation system in PDAC mediates a reciprocal relationship, whereby coagulation proteases and receptors promote PDAC tumor progression and dissemination. Here, upregulation of tissue factor (TF) by tumor cells can drive local generation of the central coagulation protease thrombin that promotes cell signaling activity through protease-activated receptors (PARs) expressed by both tumor cells and multiple stromal cell subsets. Moreover, the TF-thrombin-PAR1 signaling axis appears to be a major mechanism of cancer progression in general and PDAC in particular. Here, we summarize the current literature regarding the role of PAR1 in PDAC and review possibilities for pharmacologically targeting PAR1 as a PDAC therapeutic approach.
Collapse
|
6
|
Bonilla DA, Kreider RB, Stout JR, Forero DA, Kerksick CM, Roberts MD, Rawson ES. Metabolic Basis of Creatine in Health and Disease: A Bioinformatics-Assisted Review. Nutrients 2021; 13:nu13041238. [PMID: 33918657 PMCID: PMC8070484 DOI: 10.3390/nu13041238] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Creatine (Cr) is a ubiquitous molecule that is synthesized mainly in the liver, kidneys, and pancreas. Most of the Cr pool is found in tissues with high-energy demands. Cr enters target cells through a specific symporter called Na+/Cl−-dependent Cr transporter (CRT). Once within cells, creatine kinase (CK) catalyzes the reversible transphosphorylation reaction between [Mg2+:ATP4−]2− and Cr to produce phosphocreatine (PCr) and [Mg2+:ADP3−]−. We aimed to perform a comprehensive and bioinformatics-assisted review of the most recent research findings regarding Cr metabolism. Specifically, several public databases, repositories, and bioinformatics tools were utilized for this endeavor. Topics of biological complexity ranging from structural biology to cellular dynamics were addressed herein. In this sense, we sought to address certain pre-specified questions including: (i) What happens when creatine is transported into cells? (ii) How is the CK/PCr system involved in cellular bioenergetics? (iii) How is the CK/PCr system compartmentalized throughout the cell? (iv) What is the role of creatine amongst different tissues? and (v) What is the basis of creatine transport? Under the cellular allostasis paradigm, the CK/PCr system is physiologically essential for life (cell survival, growth, proliferation, differentiation, and migration/motility) by providing an evolutionary advantage for rapid, local, and temporal support of energy- and mechanical-dependent processes. Thus, we suggest the CK/PCr system acts as a dynamic biosensor based on chemo-mechanical energy transduction, which might explain why dysregulation in Cr metabolism contributes to a wide range of diseases besides the mitigating effect that Cr supplementation may have in some of these disease states.
Collapse
Affiliation(s)
- Diego A. Bonilla
- Research Division, Dynamical Business & Science Society–DBSS International SAS, Bogotá 110861, Colombia
- Research Group in Biochemistry and Molecular Biology, Universidad Distrital Francisco José de Caldas, Bogotá 110311, Colombia
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
- kDNA Genomics, Joxe Mari Korta Research Center, University of the Basque Country UPV/EHU, 20018 Donostia-San Sebastián, Spain
- Correspondence: ; Tel.: +57-320-335-2050
| | - Richard B. Kreider
- Exercise & Sport Nutrition Laboratory, Human Clinical Research Facility, Texas A&M University, College Station, TX 77843, USA;
| | - Jeffrey R. Stout
- Physiology of Work and Exercise Response (POWER) Laboratory, Institute of Exercise Physiology and Rehabilitation Science, University of Central Florida, Orlando, FL 32816, USA;
| | - Diego A. Forero
- Professional Program in Sport Training, School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá 111221, Colombia;
| | - Chad M. Kerksick
- Exercise and Performance Nutrition Laboratory, School of Health Sciences, Lindenwood University, Saint Charles, MO 63301, USA;
| | - Michael D. Roberts
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA;
- Edward via College of Osteopathic Medicine, Auburn, AL 36849, USA
| | - Eric S. Rawson
- Department of Health, Nutrition and Exercise Science, Messiah University, Mechanicsburg, PA 17055, USA;
| |
Collapse
|
7
|
Gallo G. The bioenergetics of neuronal morphogenesis and regeneration: Frontiers beyond the mitochondrion. Dev Neurobiol 2020; 80:263-276. [PMID: 32750228 DOI: 10.1002/dneu.22776] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/27/2022]
Abstract
The formation of axons and dendrites during development, and their regeneration following injury, are energy intensive processes. The underlying assembly and dynamics of the cytoskeleton, axonal transport mechanisms, and extensive signaling networks all rely on ATP and GTP consumption. Cellular ATP is generated through oxidative phosphorylation (OxP) in mitochondria, glycolysis and "regenerative" kinase systems. Recent investigations have focused on the role of the mitochondrion in axonal development and regeneration emphasizing the importance of this organelle and OxP in axon development and regeneration. In contrast, the understanding of alternative sources of ATP in neuronal morphogenesis and regeneration remains largely unexplored. This review focuses on the current state of the field of neuronal bioenergetics underlying morphogenesis and regeneration and considers the literature on the bioenergetics of non-neuronal cell motility to emphasize the potential contributions of non-mitochondrial energy sources.
Collapse
Affiliation(s)
- Gianluca Gallo
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, PA, USA
| |
Collapse
|
8
|
Gabr MT, Balupuri A, Kang NS. High-Throughput Platform for Real-Time Monitoring of ATP-Generating Enzymes in Living Cells Based on a Lanthanide Probe. ACS Sens 2020; 5:1872-1876. [PMID: 32610895 DOI: 10.1021/acssensors.0c00897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Remarkable variation between cell-free and cellular measurements of enzyme activity triggered the unmet need to develop tools for monitoring enzyme activity in living cells. Such tools will advance our understanding of the biological functions of enzymes and their potential impact on drug discovery. We report in this study a universal assay for monitoring ATP-generating enzymes in living cells using a self-assembled Tb3+ complex probe. Modulation of the rheological properties of cell culture media enabled shifting the lifetime of the Tb3+ complex in the presence of ATP from micro-to-millisecond range. Based on the response of the Tb3+ complex to ATP, cellular assays for 5 ATP-generating enzymes were developed. Remarkably, assessment of the activity of these enzymes in living cells is made possible for the first time. The pyruvate kinase M2 (PKM2) assay has been optimized for high-throughput screening (HTS) and further implemented in the identification of novel scaffolds as PKM2 inhibitors.
Collapse
Affiliation(s)
- Moustafa T. Gabr
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Anand Balupuri
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Nam Sook Kang
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 305-764, Republic of Korea
| |
Collapse
|
9
|
Gonadotropin regulation of ankyrin-repeat and SOCS-box protein 9 (ASB9) in ovarian follicles and identification of binding partners. PLoS One 2019; 14:e0212571. [PMID: 30811458 PMCID: PMC6392328 DOI: 10.1371/journal.pone.0212571] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 02/05/2019] [Indexed: 11/23/2022] Open
Abstract
Ankyrin-repeat and SOCS-box protein 9 (ASB9) is a member of the large SOCS-box containing proteins family and acts as the specific substrate recognition component of E3 ubiquitin ligases in the process of ubiquitination and proteasomal degradation. We previously identified ASB9 as a differentially expressed gene in granulosa cells (GC) of bovine ovulatory follicles. This study aimed to further investigate ASB9 mRNA and protein regulation, identify binding partners in GC of bovine ovulatory follicles, and study its function. GC were obtained from small follicles (SF: 2–4 mm), dominant follicles at day 5 of the estrous cycle (DF), and ovulatory follicles, 24 hours following hCG injection (OF). Analyses by RT-PCR showed a 104-fold greater expression of ASB9 in GC of OF than in DF. Steady-state levels of ASB9 in follicular walls (granulosa and theca cells) analyzed at 0, 6, 12, 18 and 24 hours after hCG injection showed a significant induction of ASB9 expression at 12 and 18 hours, reaching a maximum induction of 10.2-fold at 24 hours post-hCG as compared to 0 hour. These results were confirmed in western blot analysis showing strongest ASB9 protein amounts in OF. Yeast two-hybrid screening of OF-cDNAs library resulted in the identification of 10 potential ASB9 binding partners in GC but no interaction was found between ASB9 and creatine kinase B (CKB) in these GC. Functional studies using CRISPR-Cas9 approach revealed that ASB9 inhibition led to increased GC proliferation and modulation of target genes expression. Overall, these results support a physiologically relevant role of ASB9 in the ovulatory follicle by targeting specific proteins likely for degradation, contributing to reduced GC proliferation, and could be involved in the final GC differentiation into luteal cells.
Collapse
|
10
|
Liu X, Yu J, Song S, Yue X, Li Q. Protease-activated receptor-1 (PAR-1): a promising molecular target for cancer. Oncotarget 2017; 8:107334-107345. [PMID: 29291033 PMCID: PMC5739818 DOI: 10.18632/oncotarget.21015] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 08/27/2017] [Indexed: 12/18/2022] Open
Abstract
PAR-1 is expressed not only in epithelium, neurons, astrocytes, immune cells, but also in cancer-associated fibroblasts, ECs (epithelial cells), myocytes of blood vessels, mast cells, and macrophages in tumor microenvironment, whereas PAR-1 stimulates macrophages to synthesize and secrete thrombin as well as other growth factors, resulting in enhanced cell proliferation, tumor growth and metastasis. Therefore, considerable effort has been devoted to the development of inhibitors targeting PAR-1. Here, we provide a comprehensive review of PAR-1’s role in cancer invasiveness and dissemination, as well as potential therapeutic strategies targeting PAR-1 signaling.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.,Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Jiahui Yu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Shangjin Song
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xiaoqiang Yue
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Qi Li
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
11
|
Abstract
Although many studies have demonstrated that components of the hemostatic system may be involved in signaling leading to cancer progression, the potential mechanisms by which they contribute to cancer dissemination are not yet precisely understood. Among known coagulant factors, tissue factor (TF) and thrombin play a pivotal role in cancer invasion. They may be generated in the tumor microenvironment independently of blood coagulation and can induce cell signaling through activation of protease-activated receptors (PARs). PARs are transmembrane G-protein-coupled receptors (GPCRs) that are activated by a unique proteolytic mechanism. They play important roles in vascular physiology, neural tube closure, hemostasis, and inflammation. All of these agents (TF, thrombin, PARs—mainly PAR-1 and PAR-2) are thought to promote cancer invasion and metastasis at least in part by facilitating tumor cell migration, angiogenesis, and interactions with host vascular cells, including platelets, fibroblasts, and endothelial cells lining blood vessels. Here, we discuss the role of PARs and their activators in cancer progression, focusing on TF- and thrombin-mediated actions. Therapeutic options tailored specifically to inhibit PAR-induced signaling in cancer patients are presented as well.
Collapse
|
12
|
Kitzenberg D, Colgan SP, Glover LE. Creatine kinase in ischemic and inflammatory disorders. Clin Transl Med 2016; 5:31. [PMID: 27527620 PMCID: PMC4987751 DOI: 10.1186/s40169-016-0114-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/02/2016] [Indexed: 12/20/2022] Open
Abstract
The creatine/phosphocreatine pathway plays a conserved and central role in energy metabolism. Compartmentalization of specific creatine kinase enzymes permits buffering of local high energy phosphates in a thermodynamically favorable manner, enabling both rapid energy storage and energy transfer within the cell. Augmentation of this metabolic pathway by nutritional creatine supplementation has been shown to elicit beneficial effects in a number of diverse pathologies, particularly those that incur tissue ischemia, hypoxia or oxidative stress. In these settings, creatine and phosphocreatine prevent depletion of intracellular ATP and internal acidification, enhance post-ischemic recovery of protein synthesis and promote free radical scavenging and stabilization of cellular membranes. The creatine kinase energy system is itself further regulated by hypoxic signaling, highlighting the existence of endogenous mechanisms in mammals that can enhance creatine metabolism during oxygen deprivation to promote tissue resolution and homeostasis. Here, we review recent insights into the creatine kinase pathway, and provide rationale for dietary creatine supplementation in human ischemic and inflammatory pathologies.
Collapse
Affiliation(s)
- David Kitzenberg
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, 12700 East 19th Ave. MS B-146, Aurora, CO, 80045, USA.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Sean P Colgan
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, 12700 East 19th Ave. MS B-146, Aurora, CO, 80045, USA.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Louise E Glover
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, 12700 East 19th Ave. MS B-146, Aurora, CO, 80045, USA. .,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
13
|
Rohatgi T, Sedehizade F, Reymann KG, Reiser G. Protease-Activated Receptors in Neuronal Development, Neurodegeneration, and Neuroprotection: Thrombin as Signaling Molecule in the Brain. Neuroscientist 2016; 10:501-12. [PMID: 15534036 DOI: 10.1177/1073858404269955] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Protease-activated receptors (PARs) belong to the superfamily of seven transmembrane domain G protein-coupled receptors. Four PAR subtypes are known, PAR-1 to -4. PARs are highly homologous between the species and are expressed in a wide variety of tissues and cell types. Of particular interest is the role which these receptors play in the brain, with regard to neuroprotection or degeneration under pathological conditions. The main agonist of PARs is thrombin, a multifunctional serine protease, known to be present not only in blood plasma but also in the brain. PARs possess an irreversible activation mechanism. Binding of agonist and subsequent cleavage of the extracellular N-terminus of the receptor results in exposure of a so-called tethered ligand domain, which then binds to extracellular loop 2 of the receptor leading to receptor activation. PARs exhibit an extensive expression pattern in both the central and the peripheral nervous system. PARs participate in several mechanisms important for normal cellular functioning and during critical situations involving cellular survival and death. In the last few years, research on Alzheimer’s disease and stroke has linked PARs to the pathophysiology of these neurodegenerative disorders. Actions of thrombin are concentration-dependent, and therefore, depending on cellular function and environment, serve as a double-edged sword. Thrombin can be neuroprotective during stress conditions, whereas under normal conditions high concentrations of thrombin are toxic to cells.
Collapse
Affiliation(s)
- Tanuja Rohatgi
- Institut für Neurobiochemie, Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Magdeburg, Germany
| | | | | | | |
Collapse
|
14
|
Cellular compartmentation of energy metabolism: creatine kinase microcompartments and recruitment of B-type creatine kinase to specific subcellular sites. Amino Acids 2016; 48:1751-74. [DOI: 10.1007/s00726-016-2267-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/27/2016] [Indexed: 12/13/2022]
|
15
|
Creatine kinase in cell cycle regulation and cancer. Amino Acids 2016; 48:1775-84. [PMID: 27020776 DOI: 10.1007/s00726-016-2217-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/14/2016] [Indexed: 02/05/2023]
Abstract
The phosphocreatine-creatine kinase (CK) shuttle system is increasingly recognized as a fundamental mechanism for ATP homeostasis in both excitable and non-excitable cells. Many intracellular processes are ATP dependent. Cell division is a process requiring a rapid rate of energy turnover. Cell cycle regulation is also a key point to understanding the mechanisms underlying cancer progression. It has been known for about 40 years that aberrant CK levels are associated with various cancers and for over 30 years that CK is involved in mitosis regulation. However, the underlying molecular mechanisms have not been investigated sufficiently until recently. By maintaining ATP at sites of high-energy demand, CK can regulate cell cycle progression by affecting the intracellular energy status as well as by influencing signaling pathways that are essential to activate cell division and cytoskeleton reorganization. Aberrant CK levels may impair cell viability under normal or stressed conditions and induce cell death. The involvement of CK in cell cycle regulation and cellular energy metabolism makes it a potential diagnostic biomarker and therapeutic target in cancer. To understand the multiple physiological/pathological functions of CK, it is necessary to identify CK-binding partners and regulators including proteins, non-coding RNAs and participating endogenous small molecular weight chemical compounds. This review will focus on molecular mechanisms of CK in cell cycle regulation and cancer progression. It will also discuss the implications of recent mechanistic studies, the emerging problems and future challenges of the multifunctional enzyme CK.
Collapse
|
16
|
Thrombin decreases expression of the glutamate transporter GLAST and inhibits glutamate uptake in primary cortical astrocytes via the Rho kinase pathway. Exp Neurol 2015; 273:288-300. [PMID: 26391563 DOI: 10.1016/j.expneurol.2015.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/17/2015] [Indexed: 01/08/2023]
Abstract
Astrocyte glutamate transporters GLAST and GLT1 play a key role in regulating neuronal excitation and their levels are altered in patients with epilepsy, and after traumatic brain injury. The mechanisms which regulate their expression are not well understood. We tested the hypothesis that exposure of astrocytes to high levels of thrombin, as may occur after a compromise of the blood-brain barrier, would reduce astrocyte glutamate transporter levels. In isolated rat cortical astrocytes we examined the effects of thrombin on the expression and function of glutamate transporters, and the signaling pathways involved in these responses by using Western blotting and selective inhibitors. Thrombin induced a selective decrease in the expression of GLAST but not GLT1, with a corresponding decrease in the capacity of astrocytes to take up glutamate. Activation of the thrombin receptor PAR-1 with an activating peptide induced a similar decrease in the expression of GLAST and compromise of glutamate uptake. The downregulation of GLAST induced by thrombin was mediated by the mitogen activated protein kinases p38 MAPK, ERK and JNK, but inhibition of these kinases did not prevent the decrease in glutamate uptake induced by thrombin. In contrast, inhibition of the Rho kinase pathway using the specific inhibitor, Y27632, suppressed both the decrease in the expression of GLAST and the decrease in glutamate uptake induced by thrombin. In hippocampal astrocyte cultures, thrombin caused a decrease in both GLAST and GLT1. In tissue resected from brains of children with intractable epilepsy, we found a decrease in the integrity of the blood-brain barrier along with a reduction in immunoreactivity for both transporters which was associated with an increase in cleaved thrombin and reactive astrogliosis. The in vitro results suggest a specific mechanism by which thrombin may lead to a compromise of astrocyte function and enhanced synaptic excitability after the blood-brain barrier is compromised. The human in vivo results provide indirect support evidence linking the compromise of the blood-brain barrier to thrombin-induced reduction in glutamate transporter expression and an increase in neuronal excitation.
Collapse
|
17
|
Skeie JM, Roybal CN, Mahajan VB. Proteomic insight into the molecular function of the vitreous. PLoS One 2015; 10:e0127567. [PMID: 26020955 PMCID: PMC4447289 DOI: 10.1371/journal.pone.0127567] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 04/16/2015] [Indexed: 02/06/2023] Open
Abstract
The human vitreous contains primarily water, but also contains proteins which have yet to be fully characterized. To gain insight into the four vitreous substructures and their potential functions, we isolated and analyzed the vitreous protein profiles of three non-diseased human eyes. The four analyzed substructures were the anterior hyaloid, the vitreous cortex, the vitreous core, and the vitreous base. Proteins were separated by multidimensional liquid chromatography and identified by tandem mass spectrometry. Bioinformatics tools then extracted the expression profiles, signaling pathways, and interactomes unique to each tissue. From each substructure, a mean of 2,062 unique proteins were identified, with many being differentially expressed in a specific substructure: 278 proteins were unique to the anterior hyaloid, 322 to the vitreous cortex, 128 to the vitreous base, and 136 to the vitreous core. When the identified proteins were organized according to relevant functional pathways and networks, key patterns appeared. The blood coagulation pathway and extracellular matrix turnover networks were highly represented. Oxidative stress regulation and energy metabolism proteins were distributed throughout the vitreous. Immune functions were represented by high levels of immunoglobulin, the complement pathway, damage-associated molecular patterns (DAMPs), and evolutionarily conserved antimicrobial proteins. The majority of vitreous proteins detected were intracellular proteins, some of which originate from the retina, including rhodopsin (RHO), phosphodiesterase 6 (PDE6), and glial fibrillary acidic protein (GFAP). This comprehensive analysis uncovers a picture of the vitreous as a biologically active tissue, where proteins localize to distinct substructures to protect the intraocular tissues from infection, oxidative stress, and energy disequilibrium. It also reveals the retina as a potential source of inflammatory mediators. The vitreous proteome catalogues the dynamic interactions between the vitreous and surrounding tissues. It therefore could be an indirect and effective method for surveying vitreoretinal disease for specific biomarkers.
Collapse
Affiliation(s)
- Jessica M. Skeie
- Omics Laboratory, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States of America
| | - C. Nathaniel Roybal
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States of America
| | - Vinit B. Mahajan
- Omics Laboratory, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States of America
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States of America
- * E-mail:
| |
Collapse
|
18
|
Ramírez Ríos S, Lamarche F, Cottet-Rousselle C, Klaus A, Tuerk R, Thali R, Auchli Y, Brunisholz R, Neumann D, Barret L, Tokarska-Schlattner M, Schlattner U. Regulation of brain-type creatine kinase by AMP-activated protein kinase: interaction, phosphorylation and ER localization. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2014; 1837:1271-83. [PMID: 24727412 DOI: 10.1016/j.bbabio.2014.03.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/25/2014] [Accepted: 03/31/2014] [Indexed: 12/25/2022]
Abstract
AMP-activated protein kinase (AMPK) and cytosolic brain-type creatine kinase (BCK) cooperate under energy stress to compensate for loss of adenosine triphosphate (ATP) by either stimulating ATP-generating and inhibiting ATP-consuming pathways, or by direct ATP regeneration from phosphocreatine, respectively. Here we report on AMPK-dependent phosphorylation of BCK from different species identified by in vitro screening for AMPK substrates in mouse brain. Mass spectrometry, protein sequencing, and site-directed mutagenesis identified Ser6 as a relevant residue with one site phosphorylated per BCK dimer. Yeast two-hybrid analysis revealed interaction of active AMPK specifically with non-phosphorylated BCK. Pharmacological activation of AMPK mimicking energy stress led to BCK phosphorylation in astrocytes and fibroblasts, as evidenced with a highly specific phospho-Ser6 antibody. BCK phosphorylation at Ser6 did not affect its enzymatic activity, but led to the appearance of the phosphorylated enzyme at the endoplasmic reticulum (ER), close to the ER calcium pump, a location known for muscle-type cytosolic creatine kinase (CK) to support Ca²⁺-pumping.
Collapse
Affiliation(s)
- Sacnicte Ramírez Ríos
- Univ. Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France; Inserm, U1055, Grenoble, France
| | - Frédéric Lamarche
- Univ. Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France; Inserm, U1055, Grenoble, France
| | - Cécile Cottet-Rousselle
- Univ. Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France; Inserm, U1055, Grenoble, France
| | - Anna Klaus
- Univ. Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France; Inserm, U1055, Grenoble, France
| | - Roland Tuerk
- Institute of Cell Biology, ETH Zurich, Switzerland
| | - Ramon Thali
- Institute of Cell Biology, ETH Zurich, Switzerland
| | - Yolanda Auchli
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Switzerland
| | - René Brunisholz
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Switzerland
| | | | - Luc Barret
- Univ. Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France; Inserm, U1055, Grenoble, France
| | - Malgorzata Tokarska-Schlattner
- Univ. Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France; Inserm, U1055, Grenoble, France
| | - Uwe Schlattner
- Univ. Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France; Inserm, U1055, Grenoble, France.
| |
Collapse
|
19
|
Yang J, Hu L, Wu Q, Liu L, Zhao L, Zhao X, Song T, Huang C. A terrified-sound stress induced proteomic changes in adult male rat hippocampus. Physiol Behav 2014; 128:32-8. [PMID: 24518870 DOI: 10.1016/j.physbeh.2014.01.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 01/19/2014] [Accepted: 01/25/2014] [Indexed: 01/24/2023]
Abstract
In this study, we investigated the biochemical mechanisms in the adult rat hippocampus underlying the relationship between a terrified-sound induced psychological stress and spatial learning. Adult male rats were exposed to a terrified-sound stress, and the Morris water maze (MWM) has been used to evaluate changes in spatial learning and memory. The protein expression profile of the hippocampus was examined using two-dimensional gel electrophoresis (2DE), matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, and bioinformatics analysis. The data from the MWM tests suggested that a terrified-sound stress improved spatial learning. The proteomic analysis revealed that the expression of 52 proteins was down-regulated, while that of 35 proteins were up-regulated, in the hippocampus of the stressed rats. We identified and validated six of the most significant differentially expressed proteins that demonstrated the greatest stress-induced changes. Our study provides the first evidence that a terrified-sound stress improves spatial learning in rats, and that the enhanced spatial learning coincides with changes in protein expression in rat hippocampus.
Collapse
Affiliation(s)
- Juan Yang
- Department of Genetics and Molecular Biology, Medical School of Xi'an Jiaotong University, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Disease-Related Gene, Ministry of Education, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China
| | - Lili Hu
- Department of Genetics and Molecular Biology, Medical School of Xi'an Jiaotong University, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Disease-Related Gene, Ministry of Education, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China
| | - Qiuhua Wu
- Department of Genetics and Molecular Biology, Medical School of Xi'an Jiaotong University, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Disease-Related Gene, Ministry of Education, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China
| | - Liying Liu
- Department of Genetics and Molecular Biology, Medical School of Xi'an Jiaotong University, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Disease-Related Gene, Ministry of Education, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China
| | - Lingyu Zhao
- Department of Genetics and Molecular Biology, Medical School of Xi'an Jiaotong University, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Disease-Related Gene, Ministry of Education, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China
| | - Xiaoge Zhao
- Department of Genetics and Molecular Biology, Medical School of Xi'an Jiaotong University, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Disease-Related Gene, Ministry of Education, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China
| | - Tusheng Song
- Department of Genetics and Molecular Biology, Medical School of Xi'an Jiaotong University, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Disease-Related Gene, Ministry of Education, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China
| | - Chen Huang
- Department of Genetics and Molecular Biology, Medical School of Xi'an Jiaotong University, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China; Key Laboratory of Environment and Disease-Related Gene, Ministry of Education, 76 Western Yanta Road, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
20
|
Lin YS, Cheng TH, Chang CP, Chen HM, Chern Y. Enhancement of brain-type creatine kinase activity ameliorates neuronal deficits in Huntington's disease. Biochim Biophys Acta Mol Basis Dis 2013; 1832:742-53. [DOI: 10.1016/j.bbadis.2013.02.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 01/22/2013] [Accepted: 02/05/2013] [Indexed: 12/27/2022]
|
21
|
Li XH, Chen XJ, Ou WB, Zhang Q, Lv ZR, Zhan Y, Ma L, Huang T, Yan YB, Zhou HM. Knockdown of creatine kinase B inhibits ovarian cancer progression by decreasing glycolysis. Int J Biochem Cell Biol 2013; 45:979-86. [PMID: 23416112 DOI: 10.1016/j.biocel.2013.02.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/31/2013] [Accepted: 02/03/2013] [Indexed: 01/28/2023]
Abstract
Creatine kinase plays a key role in the energy homeostasis of vertebrate cells. Creatine kinase B (CKB), a cytosolic isoform of creatine kinase, shows upregulated expression in a variety of cancers. In this research, we confirmed that some ovarian cancer tissues had elevated CKB expression at the protein level. The functions of CKB in ovarian cancer progression were investigated in the ovarian cancer cell line Skov3, which has a high CKB expression. It was found that CKB knockdown inhibited Skov3 cell proliferation and induced apoptosis under hypoxia or hypoglycemia conditions. CKB depletion also sensitized Skov3 to chemotherapeutic agents. Furthermore, the CKB knockdown reduced glucose consumption and lactate production, and increased ROS production and oxygen consumption. This suggested that CKB knockdown decreased cytosolic glycolysis and resulted in a tumor suppressive metabolic state in Skov3 cells. Consequently, we found that the knockdown of CKB induced G2 arrest in cell cycle by elevating p21 expression and affected the PI3K/Akt and AMPK pathways. These findings provide new insights in the role of CKB in cancer cell survival and tumor progression. Our results also suggest that CKB depletion/inhibition in combination with chemotherapeutic agents might have synergistic effects in ovarian cancer therapy.
Collapse
Affiliation(s)
- Xu-Hui Li
- Beijing Key Laboratory of Protein Therapeutics, School of Life Sciences, Tsinghua University, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ju TC, Lin YS, Chern Y. Energy dysfunction in Huntington's disease: insights from PGC-1α, AMPK, and CKB. Cell Mol Life Sci 2012; 69:4107-20. [PMID: 22627493 PMCID: PMC11115139 DOI: 10.1007/s00018-012-1025-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 04/16/2012] [Accepted: 05/02/2012] [Indexed: 12/23/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by a CAG trinucleotide expansion in the Huntingtin (Htt) gene. When the number of CAG repeats exceeds 36, the translated polyglutamine-expanded Htt protein interferes with the normal functions of many types of cellular machinery and causes cytotoxicity. Clinical symptoms include progressive involuntary movement disorders, psychiatric signs, cognitive decline, dementia, and a shortened lifespan. The most severe brain atrophy is observed in the striatum and cortex. Besides the well-characterized neuronal defects, recent studies showed that the functions of mitochondria and several key players in energy homeostasis are abnormally regulated during HD progression. Energy dysregulation thus is now recognized as an important pathogenic pathway of HD. This review focuses on the importance of three key molecular determinants (peroxisome proliferator-activated receptor-γ coactivator-1α, AMP-activated protein kinase, and creatine kinase B) of cellular energy homeostasis and their possible involvement in HD pathogenesis.
Collapse
Affiliation(s)
- Tz-Chuen Ju
- Division of Neuroscience, Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 11529 Taiwan
| | - Yow-Sien Lin
- Division of Neuroscience, Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 11529 Taiwan
| | - Yijuang Chern
- Division of Neuroscience, Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 11529 Taiwan
| |
Collapse
|
23
|
Lemire J, Appanna VD. Aluminum toxicity and astrocyte dysfunction: a metabolic link to neurological disorders. J Inorg Biochem 2011; 105:1513-7. [PMID: 22099161 DOI: 10.1016/j.jinorgbio.2011.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 06/21/2011] [Accepted: 07/08/2011] [Indexed: 12/21/2022]
Abstract
Aluminum (Al) has been implicated in a variety of neurological diseases. However, the molecular mechanisms that enable Al to be involved in these disorders have yet to be fully delineated. Using astrocytes as a model of the cerebral cellular system, we have uncovered the biochemical networks that are affected by Al toxicity. In this review, we reveal how the inhibitory influence of Al on ATP production and on mitochondrial functions help generate globular astrocytes that are fat producing machines. These biological events may be the contributing factors to Al-triggered brain disorders.
Collapse
Affiliation(s)
- Joseph Lemire
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada P3E 2C6
| | | |
Collapse
|
24
|
Cruz-Topete D, List EO, Okada S, Kelder B, Kopchick JJ. Proteomic changes in the heart of diet-induced pre-diabetic mice. J Proteomics 2011; 74:716-27. [PMID: 21354350 PMCID: PMC3081671 DOI: 10.1016/j.jprot.2011.02.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 02/10/2011] [Accepted: 02/15/2011] [Indexed: 01/18/2023]
Abstract
The development of type 2 diabetes (T2D) is strongly associated with obesity. In humans, T2D increases the risk for end organ complications. Among these, heart disease has been ranked as the leading cause of death. We used a proteomic methodology to test the hypothesis that a pre-diabetic state generated by high-fat diet leads to changes in proteins related to heart function and structure. Over 300 protein spots were resolved by two-dimensional gel electrophoresis (2-DE). Fifteen protein spots were found to be altered (7 decreased and 8 increased) in pre-diabetic hearts. The protein spots were then identified by mass spectrometry and immunoblots. Among the decreased proteins, 3 are involved in heart structure (one isoform of desmin, troponin T2 and α-cardiac actin), 3 are involved in energy metabolism (mitochondrial ATP synthase β subunit, adenylate kinase and creatine kinase) and one is a component of the citric acid cycle (isocitrate dehydrogenase 3). In contrast, proteins involved in fatty acid oxidation (two isoforms of peroxisomal enoyl-CoA hydratase) and the citric acid cycle (three isoforms of malate dehydrogenase) were increased in pre-diabetic hearts. The results suggest that changes in the levels of several heart proteins may have implications in the development of the cardiac phenotype associated to T2D.
Collapse
Affiliation(s)
| | - Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens, OH
| | - Shigeru Okada
- Edison Biotechnology Institute, Ohio University, Athens, OH
- Department of Pediatrics, College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Bruce Kelder
- Edison Biotechnology Institute, Ohio University, Athens, OH
- Department of Pediatrics, College of Osteopathic Medicine, Ohio University, Athens, OH
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH
- Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, OH
- Molecular and Cellular Biology Program, Ohio University, Athens, OH
| |
Collapse
|
25
|
Menon KN, Steer DL, Short M, Petratos S, Smith I, Bernard CCA. A novel unbiased proteomic approach to detect the reactivity of cerebrospinal fluid in neurological diseases. Mol Cell Proteomics 2011; 10:M110.000042. [PMID: 21421798 DOI: 10.1074/mcp.m110.000042] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neurodegenerative diseases, such as multiple sclerosis represent global health issues. Accordingly, there is an urgent need to understand the pathogenesis of this and other central nervous system disorders, so that more effective therapeutics can be developed. Cerebrospinal fluid is a potential source of important reporter molecules released from various cell types as a result of central nervous system pathology. Here, we report the development of an unbiased approach for the detection of reactive cerebrospinal fluid molecules and target brain proteins from patients with multiple sclerosis. To help identify molecules that may serve as clinical biomarkers for multiple sclerosis, we have biotinylated proteins present in the cerebrospinal fluid and tested their reactivity against brain homogenate as well as myelin and myelin-axolemmal complexes. Proteins were separated by two-dimensional gel electrophoresis, blotted onto membranes and probed separately with biotinylated unprocessed cerebrospinal fluid samples. Protein spots that reacted to two or more multiple sclerosis-cerebrospinal fluids were further analyzed by matrix assisted laser desorption ionization-time-of-flight time-of-flight mass spectrometry. In addition to previously reported proteins found in multiple sclerosis cerebrospinal fluid, such as αβ crystallin, enolase, and 14-3-3-protein, we have identified several additional molecules involved in mitochondrial and energy metabolism, myelin gene expression and/or cytoskeletal organization. These include aspartate aminotransferase, cyclophilin-A, quaking protein, collapsin response mediator protein-2, ubiquitin carboxy-terminal hydrolase L1, and cofilin. To further validate these findings, the cellular expression pattern of collapsin response mediator protein-2 and ubiquitin carboxy-terminal hydrolase L1 were investigated in human chronic-active MS lesions by immunohistochemistry. The observation that in multiple sclerosis lesions phosphorylated collapsin response mediator protein-2 was increased, whereas Ubiquitin carboxy-terminal hydrolase L1 was down-regulated, not only highlights the importance of these molecules in the pathology of this disease, but also illustrates the use of our approach in attempting to decipher the complex pathological processes leading to multiple sclerosis and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Krishnakumar N Menon
- Monash Immunology and Stem Cell Laboratories, Monash University, Clayton, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
26
|
Adams MN, Ramachandran R, Yau MK, Suen JY, Fairlie DP, Hollenberg MD, Hooper JD. Structure, function and pathophysiology of protease activated receptors. Pharmacol Ther 2011; 130:248-82. [PMID: 21277892 DOI: 10.1016/j.pharmthera.2011.01.003] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 01/03/2011] [Indexed: 12/18/2022]
Abstract
Discovered in the 1990s, protease activated receptors(1) (PARs) are membrane-spanning cell surface proteins that belong to the G protein coupled receptor (GPCR) family. A defining feature of these receptors is their irreversible activation by proteases; mainly serine. Proteolytic agonists remove the PAR extracellular amino terminal pro-domain to expose a new amino terminus, or tethered ligand, that binds intramolecularly to induce intracellular signal transduction via a number of molecular pathways that regulate a variety of cellular responses. By these mechanisms PARs function as cell surface sensors of extracellular and cell surface associated proteases, contributing extensively to regulation of homeostasis, as well as to dysfunctional responses required for progression of a number of diseases. This review examines common and distinguishing structural features of PARs, mechanisms of receptor activation, trafficking and signal termination, and discusses the physiological and pathological roles of these receptors and emerging approaches for modulating PAR-mediated signaling in disease.
Collapse
Affiliation(s)
- Mark N Adams
- Mater Medical Research Institute, Aubigny Place, Raymond Terrace, South Brisbane Qld 4101, Australia
| | | | | | | | | | | | | |
Collapse
|
27
|
Chen J, Sun Y, Mao X, Liu Q, Wu H, Chen Y. RANKL up-regulates brain-type creatine kinase via poly(ADP-ribose) polymerase-1 during osteoclastogenesis. J Biol Chem 2010; 285:36315-21. [PMID: 20837480 DOI: 10.1074/jbc.m110.157743] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Receptor activator of nuclear factor κB ligand (RANKL) is the key regulator for osteoclast formation and function. During osteoclastogenesis, RANKL-stimulated signals differentially modulate expression of a large number of proteins. Using proteomics approaches, we identified that brain-type cytoplasmic creatine kinase (Ckb) was greatly induced in mature osteoclasts. Ckb has been shown to contribute to osteoclast function. However, the mechanisms of Ckb regulation and the contribution of other isoforms of creatine kinase during RANKL-induced osteoclastogenesis are unknown. We found that Ckb was the predominant isoform of creatine kinase during osteoclastogenesis. Real-time PCR confirmed that RANKL induced ckb mRNA expression by over 40-fold in primary mouse bone marrow macrophages and Raw 264.7 cells. The RANKL-responsive region was identified within the -0.4- to -0.2-kb 5'-flanking region of the ckb gene. Affinity binding purification followed by mass spectrometry analysis revealed that poly(ADP-ribose) polymerase-1 (PARP-1) bound to the -0.4/-0.2-kb fragment that negatively regulated expression of ckb in response to RANKL stimulation. Electrophoretic mobility shift assays with PARP-1-specific antibody located the binding site of PARP-1 to the TTCCCA consensus sequence. The expression of PARP-1 was reduced during RANKL-induced osteoclastogenesis, concurrently with increased expression of Ckb. Consistently, knockdown of PARP-1 by lentivirus-delivered shRNA enhanced ckb mRNA expression. The activity of PARP-1 was determined to be required for its inhibitory effect on the ckb expression. In summary, we have demonstrated that PARP-1 is a negative regulator of the ckb expression. Down-regulation of PARP-1 is responsible for the up-regulation of ckb during RANKL-induced osteoclastogenesis.
Collapse
Affiliation(s)
- Jianfeng Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
28
|
Stellas D, El Hamidieh A, Patsavoudi E. Monoclonal antibody 4C5 prevents activation of MMP2 and MMP9 by disrupting their interaction with extracellular HSP90 and inhibits formation of metastatic breast cancer cell deposits. BMC Cell Biol 2010; 11:51. [PMID: 20602761 PMCID: PMC2914660 DOI: 10.1186/1471-2121-11-51] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Accepted: 07/05/2010] [Indexed: 12/23/2022] Open
Abstract
Background Heat shock protein 90 (HSP90) is a molecular chaperone that is considered a new target for the treatment of cancer. Increasing data reveal an extracellular chaperoning activity for HSP90. Here we investigate the interaction of the secreted isoforms of HSP90 with matrix metalloproteinases (MMP) MMP2 and MMP9. Moreover we examine the role of a monoclonal antibody (mAb) against HSP90, mAb 4C5, regarding these interactions and its value as a potential inhibitor of human breast cancer cell invasion and metastasis. Results Our results showed that both HSP90α and HSP90β are secreted by MDAMB453 human breast cancer cells and interact with MMP2 and MMP9. MAb 4C5, while not affecting the secretion of inactive MMPs, inhibits their activation by disrupting their interaction extracellularly with both isoforms of HSP90. The in vivo studies revealed that mAb 4C5 significantly inhibits the metastatic deposit formation of MDAMB453 cells into the lungs of SCID mice. Conclusion Both isoforms of HSP90 are secreted by MDAMB453 cells and interact with MMP2 and MMP9. MAb 4C5 prevents MMP2 and MMP9 activation, by disrupting their interaction with HSP90. Finally mAb 4C5 significantly inhibits the metastatic deposit formation of MDAMB453 cells, by preventing their extravasation and infiltration in the lung tissue and therefore it could be used as a potential therapeutic agent for cancer metastasis.
Collapse
Affiliation(s)
- Dimitris Stellas
- Department of Biochemistry, Hellenic Pasteur Institute, 127 Vasilissis Sofias Ave, 11521 Athens, Greece
| | | | | |
Collapse
|
29
|
Soh UJK, Dores MR, Chen B, Trejo J. Signal transduction by protease-activated receptors. Br J Pharmacol 2010; 160:191-203. [PMID: 20423334 PMCID: PMC2874842 DOI: 10.1111/j.1476-5381.2010.00705.x] [Citation(s) in RCA: 227] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 01/25/2010] [Accepted: 02/02/2010] [Indexed: 12/11/2022] Open
Abstract
The family of G protein-coupled receptors (GPCRs) constitutes the largest class of signalling receptors in the human genome, controlling vast physiological responses and are the target of many drugs. After activation, GPCRs are rapidly desensitized by phosphorylation and beta-arrestin binding. Most classic GPCRs are internalized through a clathrin, dynamin and beta-arrestin-dependent pathway and then recycled back to the cell surface or sorted to lysosomes for degradation. Given the vast number and diversity of GPCRs, different mechanisms are likely to exist to precisely regulate the magnitude, duration and spatial aspects of receptor signalling. The G protein-coupled protease-activated receptors (PARs) provide elegant examples of GPCRs that are regulated by distinct desensitization and endocytic sorting mechanisms, processes that are critically important for the spatial and temporal fidelity of PAR signalling. PARs are irreversibly activated through proteolytic cleavage and transmit cellular responses to extracellular proteases. Activated PAR(1) internalizes through a clathrin- and dynamin-dependent pathway independent of beta-arrestins. Interestingly, PAR(1) is basally ubiquitinated and deubiquitinated after activation and traffics from endosomes to lysosomes independent of ubiquitination. In contrast, beta-arrestins mediate activated PAR(2) internalization and function as scaffolds that promote signalling from endocytic vesicles. Moreover, activated PAR(2) is modified with ubiquitin, which facilitates lysosomal degradation. Activated PARs also adopt distinct active conformations that signal to diverse effectors and are likely regulated by different mechanisms. Thus, the identification of the molecular machinery important for PAR signal regulation will enable the development of new strategies to manipulate receptor signalling and will provide novel targets for the development of drugs.
Collapse
Affiliation(s)
- Unice J K Soh
- Department of Pharmacology, University of California, San Diego, 92093-0636, USA
| | | | | | | |
Collapse
|
30
|
Han J, Liu G, Profirovic J, Niu J, Voyno-Yasenetskaya T. Zyxin is involved in thrombin signaling via interaction with PAR-1 receptor. FASEB J 2009; 23:4193-206. [PMID: 19690217 DOI: 10.1096/fj.09-131862] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Protease-activated receptor 1 (PAR-1) mediates thrombin signaling in human endothelial cells. As a G-protein-coupled receptor, PAR-1 transmits thrombin signal through activation of the heterotrimeric G proteins, Gi, Gq, and G12/13. In this study, we demonstrated that zyxin, a LIM-domain-containing protein, is involved in thrombin-mediated actin cytoskeleton remodeling and serum response element (SRE)-dependent gene transcription. We determined that zyxin binds to the C-terminal domain of PAR-1, providing a possible mechanism of involvement of zyxin as a signal transducer in PAR-1 signaling. Data showing that disruption of PAR-1-zyxin interaction inhibited thrombin-induced stress fiber formation and SRE activation supports this hypothesis. Similarly, depletion of zyxin using siRNA inhibited thrombin-induced actin stress fiber formation and SRE-dependent gene transcription. In addition, depletion of zyxin resulted in delay of endothelial barrier restoration after thrombin treatment. Notably, down-regulation of zyxin did not affect thrombin-induced activation of RhoA or Gi, Gq, and G12/13 heterotrimeric G proteins, implicating a novel signaling pathway regulated by PAR-1 that is not mediated by G-proteins. The observation that zyxin targets VASP, a partner of zyxin in regulation of actin assembly and dynamics, to focal adhesions and along stress fibers on thrombin stimulation suggests that zyxin may participate in thrombin-induced cytoskeletal remodeling through recruitment of VASP. In summary, this study establishes a crucial role of zyxin in thrombin signaling in endothelial cells and provides evidence for a novel PAR-1 signaling pathway mediated by zyxin.
Collapse
Affiliation(s)
- Jingyan Han
- Department of Pharmacology (MC 868), University of Illinois at Chicago, 909 S. Wolcott Ave., Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
31
|
Dzeja P, Terzic A. Adenylate kinase and AMP signaling networks: metabolic monitoring, signal communication and body energy sensing. Int J Mol Sci 2009; 10:1729-1772. [PMID: 19468337 PMCID: PMC2680645 DOI: 10.3390/ijms10041729] [Citation(s) in RCA: 326] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 03/26/2009] [Accepted: 04/02/2009] [Indexed: 12/20/2022] Open
Abstract
Adenylate kinase and downstream AMP signaling is an integrated metabolic monitoring system which reads the cellular energy state in order to tune and report signals to metabolic sensors. A network of adenylate kinase isoforms (AK1-AK7) are distributed throughout intracellular compartments, interstitial space and body fluids to regulate energetic and metabolic signaling circuits, securing efficient cell energy economy, signal communication and stress response. The dynamics of adenylate kinase-catalyzed phosphotransfer regulates multiple intracellular and extracellular energy-dependent and nucleotide signaling processes, including excitation-contraction coupling, hormone secretion, cell and ciliary motility, nuclear transport, energetics of cell cycle, DNA synthesis and repair, and developmental programming. Metabolomic analyses indicate that cellular, interstitial and blood AMP levels are potential metabolic signals associated with vital functions including body energy sensing, sleep, hibernation and food intake. Either low or excess AMP signaling has been linked to human disease such as diabetes, obesity and hypertrophic cardiomyopathy. Recent studies indicate that derangements in adenylate kinase-mediated energetic signaling due to mutations in AK1, AK2 or AK7 isoforms are associated with hemolytic anemia, reticular dysgenesis and ciliary dyskinesia. Moreover, hormonal, food and antidiabetic drug actions are frequently coupled to alterations of cellular AMP levels and associated signaling. Thus, by monitoring energy state and generating and distributing AMP metabolic signals adenylate kinase represents a unique hub within the cellular homeostatic network.
Collapse
Affiliation(s)
- Petras Dzeja
- Division of Cardiovascular Disease, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; E-Mail:
(A.T.)
| | - Andre Terzic
- Division of Cardiovascular Disease, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; E-Mail:
(A.T.)
| |
Collapse
|
32
|
Zhang Y, Li H, Wang X, Gao X, Liu X. Regulation of T cell development and activation by creatine kinase B. PLoS One 2009; 4:e5000. [PMID: 19337362 PMCID: PMC2659424 DOI: 10.1371/journal.pone.0005000] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Accepted: 03/04/2009] [Indexed: 01/19/2023] Open
Abstract
Creatine kinase catalyzes the reversible transfer of the N-phosphoryl group from phosphocreatine to ADP to generate ATP and plays a key role in highly energy-demanding processes such as muscle contraction and flagellar motility; however, its role in signal transduction (which frequently involves ATP-consuming phosphorylation) and consequent cell-fate decisions remains largely unknown. Here we report that creatine kinase B was significantly up-regulated during the differentiation of double-positive thymocytes into single-positive thymocytes. Ectopic expression of creatine kinase B led to increased ATP level and enhanced phosphorylation of the TCR signaling proteins. Consequentially, transgenic expression of creatine kinase B promoted the expression of Nur77 and Bim proteins and the cell death of TCR signaled thymocyte. In addition, the activation, proliferation and cytokine secretion of T cells were also enhanced by the expression of creatine kinase B transgene. In contrast, treatment of T cells with specific creatine kinase inhibitor or creatine kinase B shRNA resulted in severely impaired T cell activation. Taken together, our results indicate that creatine kinase B plays an unexpected role in modulating TCR-mediated signaling and critically regulates thymocyte selection and T cell activation.
Collapse
Affiliation(s)
- Yafeng Zhang
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hai Li
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoming Wang
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiang Gao
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Xiaolong Liu
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- * E-mail:
| |
Collapse
|
33
|
Kuiper JWP, van Horssen R, Oerlemans F, Peters W, van Dommelen MMT, te Lindert MM, ten Hagen TLM, Janssen E, Fransen JAM, Wieringa B. Local ATP generation by brain-type creatine kinase (CK-B) facilitates cell motility. PLoS One 2009; 4:e5030. [PMID: 19333390 PMCID: PMC2659440 DOI: 10.1371/journal.pone.0005030] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 03/03/2009] [Indexed: 01/01/2023] Open
Abstract
Background Creatine Kinases (CK) catalyze the reversible transfer of high-energy phosphate groups between ATP and phosphocreatine, thereby playing a storage and distribution role in cellular energetics. Brain-type CK (CK-B) deficiency is coupled to loss of function in neural cell circuits, altered bone-remodeling by osteoclasts and complement-mediated phagocytotic activity of macrophages, processes sharing dependency on actomyosin dynamics. Methodology/Principal Findings Here, we provide evidence for direct coupling between CK-B and actomyosin activities in cortical microdomains of astrocytes and fibroblasts during spreading and migration. CK-B transiently accumulates in membrane ruffles and ablation of CK-B activity affects spreading and migration performance. Complementation experiments in CK-B-deficient fibroblasts, using new strategies to force protein relocalization from cytosol to cortical sites at membranes, confirmed the contribution of compartmentalized CK-B to cell morphogenetic dynamics. Conclusion/Significance Our results provide evidence that local cytoskeletal dynamics during cell motility is coupled to on-site availability of ATP generated by CK-B.
Collapse
Affiliation(s)
- Jan W. P. Kuiper
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Remco van Horssen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Frank Oerlemans
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Wilma Peters
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Michiel M. T. van Dommelen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Mariska M. te Lindert
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | - Edwin Janssen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jack A. M. Fransen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
34
|
Involvement of creatine kinase B in hepatitis C virus genome replication through interaction with the viral NS4A protein. J Virol 2009; 83:5137-47. [PMID: 19264780 DOI: 10.1128/jvi.02179-08] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Persistent infection with hepatitis C virus (HCV) is a major cause of chronic liver diseases. The aim of this study was to identify host cell factor(s) participating in the HCV replication complex (RC) and to clarify the regulatory mechanisms of viral genome replication dependent on the host-derived factor(s) identified. By comparative proteome analysis of RC-rich membrane fractions and subsequent gene silencing mediated by RNA interference, we identified several candidates for RC components involved in HCV replication. We found that one of these candidates, creatine kinase B (CKB), a key ATP-generating enzyme that regulates ATP in subcellular compartments of nonmuscle cells, is important for efficient replication of the HCV genome and propagation of infectious virus. CKB interacts with HCV NS4A protein and forms a complex with NS3-4A, which possesses multiple enzyme activities. CKB upregulates both NS3-4A-mediated unwinding of RNA and DNA in vitro and replicase activity in permeabilized HCV replicating cells. Our results support a model in which recruitment of CKB to the HCV RC compartment, which has high and fluctuating energy demands, through its interaction with NS4A is important for efficient replication of the viral genome. The CKB-NS4A association is a potential target for the development of a new type of antiviral therapeutic strategy.
Collapse
|
35
|
Aboghe DH, Yoshioka M, Phaneuf D, St-Amand J. Regulation of gene expression by estrogen in mammary gland of wild type and estrogen receptor alpha knockout mice. J Steroid Biochem Mol Biol 2009; 113:116-26. [PMID: 19126430 DOI: 10.1016/j.jsbmb.2008.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 12/02/2008] [Accepted: 12/08/2008] [Indexed: 01/20/2023]
Abstract
Using serial analysis of gene expression, we examined the effects of estrogen (E2) replacement in gonadectomized wild type (WT) and E2 receptor alpha knockout (ERalphaKO) mice on global gene expression in mammary gland. In WT mice, a total of 429,302 tags were sequenced, representing the expression level of 99,854 tag species. A total of ten transcripts were found to be modulated by E2, such as sorting nexin 5 and two no match tags. In the ERalphaKO mice, a total of 459,439 tags were sequenced, representing the expression level of 120,149 tag species. Interestingly, the same three transcripts were inversely regulated by E2 in ERalphaKO mice. In total, 78 transcripts were upregulated by E2, while 29 transcripts were downregulated. In contrast to WT mice, the majority of transcripts related to immunity were repressed in ERalphaKO mice. Moreover, induction of transcripts involved in cell differentiation, Ca2+ response, cytoskeleton, protein biosynthesis and secretion, glycolysis, and oxidative phosphorylation were seen only in ERalphaKO mice. The current study will provide useful information to understand the cellular mechanisms of E(2)-mediated gene regulation in tissues in vivo for the development of novel drugs targeting specific ER action in pathological conditions.
Collapse
Affiliation(s)
- David Hyacinthe Aboghe
- Functional Genomics Laboratory, Molecular Endocrinology and Oncology Research Center, Laval University Medical Center, Department of Anatomy and Physiology, Laval University, Québec, Canada
| | | | | | | |
Collapse
|
36
|
Chang EJ, Ha J, Oerlemans F, Lee YJ, Lee SW, Ryu J, Kim HJ, Lee Y, Kim HM, Choi JY, Kim JY, Shin CS, Pak YK, Tanaka S, Wieringa B, Lee ZH, Kim HH. Brain-type creatine kinase has a crucial role in osteoclast-mediated bone resorption. Nat Med 2008; 14:966-72. [PMID: 18724377 DOI: 10.1038/nm.1860] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Accepted: 07/23/2008] [Indexed: 11/09/2022]
Abstract
Osteoclasts differentiate from precursor cells of the monocyte-macrophage lineage and subsequently become activated to be competent for bone resorption through programs primarily governed by receptor activator of nuclear factor-kappaB ligand in cooperation with macrophage colony-stimulating factor. Proteins prominently expressed at late phases of osteoclastogenesis and with a supportive role in osteoclast function are potential therapeutic targets for bone-remodeling disorders. In this study, we used a proteomics approach to show that abundance of the brain-type cytoplasmic creatine kinase (Ckb) is greatly increased during osteoclastogenesis. Decreasing Ckb abundance by RNA interference or blocking its enzymatic activity with a pharmacological inhibitor, cyclocreatine, suppressed the bone-resorbing activity of osteoclasts grown in vitro via combined effects on actin ring formation, RhoA GTPase activity and vacuolar ATPase function. Activities of osteoclasts derived from Ckb-/- mice were similarly affected. In vivo studies showed that Ckb-/- mice were better protected against bone loss induced by ovariectomy, lipopolysaccharide challenge or interleukin-1 treatment than wild-type controls. Furthermore, administration of cyclocreatine or adenoviruses harboring Ckb small hairpin RNA attenuated bone loss in rat and mouse models. Our findings establish an important role for Ckb in the bone-resorbing function of osteoclasts and underscore its potential as a new molecular target for antiresorptive drug development.
Collapse
Affiliation(s)
- Eun-Ju Chang
- Department of Cell and Developmental Biology, Brain Korea 21, School of Dentistry, Seoul National University, 28 Yeongon-Dong, Jongno-gu, Seoul 110-749, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Salin-Cantegrel A, Shekarabi M, Holbert S, Dion P, Rochefort D, Laganière J, Dacal S, Hince P, Karemera L, Gaspar C, Lapointe JY, Rouleau GA. HMSN/ACC truncation mutations disrupt brain-type creatine kinase-dependant activation of K+/Cl− co-transporter 3. Hum Mol Genet 2008; 17:2703-11. [DOI: 10.1093/hmg/ddn172] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
38
|
Kuiper JWP, Pluk H, Oerlemans F, van Leeuwen FN, de Lange F, Fransen J, Wieringa B. Creatine kinase-mediated ATP supply fuels actin-based events in phagocytosis. PLoS Biol 2008; 6:e51. [PMID: 18336068 PMCID: PMC2265766 DOI: 10.1371/journal.pbio.0060051] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Accepted: 01/18/2008] [Indexed: 11/23/2022] Open
Abstract
Phagocytosis requires locally coordinated cytoskeletal rearrangements driven by actin polymerization and myosin motor activity. How this actomyosin dynamics is dependent upon systems that provide access to ATP at phagosome microdomains has not been determined. We analyzed the role of brain-type creatine kinase (CK-B), an enzyme involved in high-energy phosphoryl transfer. We demonstrate that endogenous CK-B in macrophages is mobilized from the cytosolic pool and coaccumulates with F-actin at nascent phagosomes. Live cell imaging with XFP-tagged CK-B and β-actin revealed the transient and specific nature of this partitioning process. Overexpression of a catalytic dead CK-B or CK-specific cyclocreatine inhibition caused a significant reduction of actin accumulation in the phagocytic cup area, and reduced complement receptor–mediated, but not Fc-γR–mediated, ingestion capacity of macrophages. Finally, we found that inhibition of CK-B affected phagocytosis already at the stage of particle adhesion, most likely via effects on actin polymerization behavior. We propose that CK-B activity in macrophages contributes to complement-induced F-actin assembly events in early phagocytosis by providing local ATP supply. To do work, cells need energy in the form of ATP. High and sudden energy demand is seen during cell-shape change, a process in which ATP fuels the cytoskeletal machinery that drives cell-morphology alteration. How a cell organizes high-energy surges without disrupting global ATP homeostasis remains an important research question. One view proposes that ATP is heterogeneously distributed, but the cytoskeletal proteins actin and myosin receive regional and preferential access to ATP. Yet this model raises another question: how is ATP funneled to these proteins from distant sources? To address some of these questions, we studied the highly localized molecular events controlling actin dynamics around phagocytic activity of macrophages. We demonstrate that actin and creatine kinase-B (CK-B), a long-known enzyme involved in ATP supply, are simultaneously recruited into the sites of action during the early phases of particle ingestion. Local availability of CK-activity and local generation of ATP promotes on-site actin remodeling and particle capture efficiency, and thus supports successful initiation of the first phases of phagocytosis. Interestingly, this coupling between local CK-activity and actin regulation is only relevant for complement-mediated phagocytosis (used by immune cells to target specific particles for ingestion). We predict that our findings may also shed light on how shape dynamics is energized in other cell types. A tight connection exists between local recruitment of creatine kinase-B (CK-B) and actin remodeling activity in phagocytic cups of macrophages. Complement-mediated phagocytosis is stimulated in the presence of enzymatic-active CK-B, indicating that local ATP supply stimulates actin-driven particle uptake events.
Collapse
Affiliation(s)
- Jan W. P Kuiper
- Department of Cell Biology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Helma Pluk
- Department of Cell Biology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Frank Oerlemans
- Department of Cell Biology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Frank de Lange
- Department of Cell Biology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jack Fransen
- Department of Cell Biology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
39
|
O'Connor RS, Steeds CM, Wiseman RW, Pavlath GK. Phosphocreatine as an energy source for actin cytoskeletal rearrangements during myoblast fusion. J Physiol 2008; 586:2841-53. [PMID: 18420707 DOI: 10.1113/jphysiol.2008.151027] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Myoblast fusion is essential for muscle development, postnatal growth and muscle repair after injury. Recent studies have demonstrated roles for actin polymerization during myoblast fusion. Dynamic cytoskeletal assemblies directing cell-cell contact, membrane coalescence and ultimately fusion require substantial cellular energy demands. Various energy generating systems exist in cells but the partitioning of energy sources during myoblast fusion is unknown. Here, we demonstrate a novel role for phosphocreatine (PCr) as a spatiotemporal energy buffer during primary mouse myoblast fusion with nascent myotubes. Creatine treatment enhanced cell fusion in a creatine kinase (CK)-dependent manner suggesting that ATP-consuming reactions are replenished through the PCr/CK system. Furthermore, selective inhibition of actin polymerization prevented myonuclear addition following creatine treatment. As myotube formation is dependent on cytoskeletal reorganization, our findings suggest that PCr hydrolysis is coupled to actin dynamics during myoblast fusion. We conclude that myoblast fusion is a high-energy process, and can be enhanced by PCr buffering of energy demands during actin cytoskeletal rearrangements in myoblast fusion. These findings implicate roles for PCr as a high-energy phosphate buffer in the fusion of multiple cell types including sperm/oocyte, trophoblasts and macrophages. Furthermore, our results suggest the observed beneficial effects of oral creatine supplementation in humans may result in part from enhanced myoblast fusion.
Collapse
Affiliation(s)
- Roddy S O'Connor
- Emory University, Department of Pharmacology, 1510 Clifton Rd, Room 5027, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
40
|
Sidera K, Gaitanou M, Stellas D, Matsas R, Patsavoudi E. A critical role for HSP90 in cancer cell invasion involves interaction with the extracellular domain of HER-2. J Biol Chem 2008; 283:2031-2041. [PMID: 18056992 DOI: 10.1074/jbc.m701803200] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
HSP90 is a ubiquitously expressed molecular chaperone that controls the folding, assembly, intracellular disposition, and proteolytic turnover of many proteins, most of which are involved in signal transduction processes. Recently, a surface form of HSP90 has been identified and associated with cell migration events. In this paper, we explore the interaction of surface HSP90 with HER-2, a receptor-like glycoprotein and member of the ErbB family of receptor tyrosine kinases that play central roles in cellular proliferation, differentiation, and migration as well as in cancer progress. The involvement of HSP90 in the regulation of HER-2 has been attributed so far to receptor stabilization via interaction with its cytoplasmic kinase domain. Here we present evidence, using glutathione S-transferase pull-down and transfection assays, for a novel interaction between surface HSP90 and the extracellular domain of HER-2. Specific disruption of this interaction using mAb 4C5, a function-blocking monoclonal antibody against HSP90, inhibits cell invasion accompanied by altered actin dynamics in human breast cancer cells under ligand stimulation conditions with heregulin. Additionally, disruption of surface HSP90/HER-2 interaction leads to inhibition of heregulin-induced HER-2-HER-3 heterodimer formation, reduced HER-2 phosphorylation, and impaired downstream kinase signaling. Interestingly, this disruption does not affect HER-2 internalization. Our data suggest that surface HSP90 is involved in heregulin-induced HER-2 activation and signaling, leading to cytoskeletal rearrangement, essential for cell invasion.
Collapse
Affiliation(s)
- Katerina Sidera
- Department of Biochemistry, Hellenic Pasteur Institute, 127 Vasilissis Sofias Ave., 11521 Athens, Greece
| | | | | | | | | |
Collapse
|
41
|
Coleman LS. A hypothesis: factor VII governs clot formation, tissue repair and apoptosis. Med Hypotheses 2007; 69:903-7. [PMID: 17383108 DOI: 10.1016/j.mehy.2007.01.063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Accepted: 01/24/2007] [Indexed: 11/30/2022]
Abstract
A hypothesis: thrombin is a "Universal Enzyme of Energy Transduction" that employs ATP energy in flowing blood to activate biochemical reactions and cell effects in both hemostasis and tissue repair. All cells possess PAR-1 (thrombin) receptors and are affected by thrombin elevations, and thrombin effects on individual cell types are determined by their unique complement of PAR-1 receptors. Disruption of the vascular endothelium (VE) activates a tissue repair mechanism (TRM) consisting of the VE, tissue factor (TF), and circulating Factors VII, IX and X that governs localized thrombin elevations to activate clot formation and cellular effects that repair tissue damage. The culmination of the repair process occurs with the restoration of the VE followed by declines in thrombin production that causes Apoptosis ("programmed cell death") in wound-healing fibroblasts, which functions as a mechanism to draw wound edges together. The location and magnitude of TRM activity governs the location and magnitude of Factor VIII activity and clot formation, but the large size of Factor VIII prevents it from penetrating the clot formed by its activity, so that its effects are self-limiting. Factors VII, IX and X function primarily as tissue repair enzymes, while Factor VIII and Factor XIII are the only serine protease enzymes in the "Coagulation Cascade" that are exclusively associated with hemostasis.
Collapse
|
42
|
Bürklen TS, Hirschy A, Wallimann T. Brain-type creatine kinase BB-CK interacts with the Golgi Matrix Protein GM130 in early prophase. Mol Cell Biochem 2006; 297:53-64. [PMID: 17036164 DOI: 10.1007/s11010-006-9322-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2006] [Accepted: 08/29/2006] [Indexed: 11/29/2022]
Abstract
Creatine kinase (CK) isoenzymes are essential for storing, buffering and intracellular transport of "energy-rich" phosphate compounds in tissues with fluctuating high energy demand such as muscle, brain and other tissues and cells where CK is expressed. In brain and many non-muscle cells, ubiquitous cytosolic "brain-type" BB-CK and ubiquitous mitochondrial CK (uMtCK) act as components of a phosphocreatine shuttle to maintain cellular energy pools and distribute energy flux. To date, still relatively little is known about direct coupling of functional dimeric BB-CK with other partner proteins or enzymes that are important for cell function. Using a global yeast two-hybrid (Y2H) screen with monomeric B-CK as bait and a representative brain cDNA library to search for interaction partners of B-CK with proteins of the brain, we repeatedly identified the cis-Golgi Matrix protein (GM130) as recurrent interacting partner of B-CK. Since HeLa cells also express both BB-CK and GM130, we subsequently used this cellular model system to verify and characterize the BB-CK-GM130 complex by GST-pulldown experiments, as well as by in vivo co-localization studies with confocal microscopy. Using dividing HeLa cells, we report here for the first time that GM130 and BB-CK co-localize specifically in a transient fashion during early prophase of mitosis, when GM130 plays an important role in Golgi fragmentation that starts also at early prophase. These data may shed new light on BB-CK function for energy provision for Golgi-fragmentation that is initiated by cell signalling cascades in the early phases of mitosis.
Collapse
Affiliation(s)
- Tanja S Bürklen
- Institute of Cell Biology, HPM D24, ETH ZURICH, Schafmattstr. 18, Zurich 8093, Switzerland
| | | | | |
Collapse
|
43
|
Coleman LS. To the editor: Is von Willebrand factor a hormone that regulates a coagulation mechanism? World J Surg 2006; 30:479-81. [PMID: 16479332 DOI: 10.1007/s00268-005-0583-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
44
|
Inoue K, Yamada J, Ueno S, Fukuda A. Brain-type creatine kinase activates neuron-specific K+-Cl- co-transporter KCC2. J Neurochem 2006; 96:598-608. [PMID: 16336223 DOI: 10.1111/j.1471-4159.2005.03560.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
GABA, a major inhibitory neurotransmitter in the adult CNS, is excitatory at early developmental stages as a result of the elevated intracellular Cl- concentration ([Cl-]i). This functional switch is primarily attributable to the K+-Cl- co-transporter KCC2, the expression of which is developmentally regulated in neurons. Previously, we reported that KCC2 interacts with brain-type creatine kinase (CKB). To elucidate the functional significance of this interaction, HEK293 cells were transfected with KCC2 and glycine receptor alpha2 subunit, and gramicidin-perforated patch-clamp recordings were performed to measure the glycine reversal potential (Egly), giving an estimate of [Cl-]i. KCC2-expressing cells displayed the expected changes in Egly following alterations in the extracellular K+ concentration ([K+]o) or administration of an inhibitor of KCCs, suggesting that the KCC2 function was being properly assessed. When added into KCC2-expressing cells, dominant-negative CKB induced a depolarizing shift in Egly and reduced the hyperpolarizing shift in Egly seen in response to a lowering of [K+]o compared with wild-type CKB. Moreover, 2,4-dinitrofluorobenzene (DNFB), an inhibitor of CKs, shifted Egly in the depolarizing direction. In primary cortical neurons expressing CKB, the GABA reversal potential was also shifted in the depolarizing direction by DNFB. Our findings suggest that, in the cellular micro-environment, CKB activates the KCC2 function.
Collapse
Affiliation(s)
- Koichi Inoue
- Department of Physiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.
| | | | | | | |
Collapse
|
45
|
Cesar JM, Pallares E, Rubí J, Navarro JL. Lactate production by thrombin-activated platelets of patients with primary thrombocythemia. Thromb Res 2006; 118:335-9. [PMID: 16256174 DOI: 10.1016/j.thromres.2005.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 09/16/2005] [Accepted: 09/26/2005] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Platelet activation needs a high energy demand which is supplied by the degradation of glucose into lactate. Platelet response to agonists in patients with primary thrombocythemia is defective. We studied the production of lactate by the platelets of patients with this disease and defective platelet aggregation. MATERIAL AND METHODS Ten patients suffering from primary thrombocythemia and ten controls were included in this study. The lactate generation was measured in resting and thrombin activated platelets in absence or presence of glucose. RESULTS Resting platelets incubated for 30 min in phosphate-buffered saline (PBS) generated the same amount of lactate in patients (44.6+/-21.6 micromol/10(11) cells) and controls (41.0+/-17.3 micromol/10(11) cells). Addition of glucose led to similar increases in lactate formation by platelets in patients (82.2+/-26.4 micromol/10(11) cells) and controls (88.1+/-34.5 micromol/10(11) cells). The addition of thrombin in absence of glucose did not modify the lactate formation respective to PBS. Finally, the incubation of platelets with both glucose and thrombin caused further increases in the generation of lactate in both groups, patients (236.9+/-83.9 micromol/10(11) cells) and controls (228.6+/-63.5 micromol/10(11) cells) without differences between them. The production of lactate in both groups was also similar when platelets were incubated for 10 min or 20 min with both thrombin and glucose. However at 5 min, platelets of patients generated more lactate (97.8+/-23.7 micromol/10(11) cells) than controls (66.5+/-38.7 micromol/10(11) cells, p<0.05). CONCLUSIONS These results suggest that thrombin is able to induce an initial hyperactivity of those pathways involved in the platelet energy production of patients with primary thrombocythemia.
Collapse
Affiliation(s)
- Jesús M Cesar
- Department of Haematology, Hospital Ramón y Cajal, Ctera Colmenar Km 9,1. 28034 Madrid, Spain.
| | | | | | | |
Collapse
|
46
|
Ares IR, Louzao MC, Vieytes MR, Yasumoto T, Botana LM. Actin cytoskeleton of rabbit intestinal cells is a target for potent marine phycotoxins. J Exp Biol 2005; 208:4345-54. [PMID: 16272256 DOI: 10.1242/jeb.01897] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
SUMMARY
Biotoxins produced by harmful marine microalgae (phycotoxins) can be accumulated into seafood, representing a great risk for public health. Some of these phycotoxins are responsible for a variety of gastrointestinal disturbances; however, the relationship between their mechanism of action and toxicity in intestinal cells is still unknown. The actin cytoskeleton is an important and highly complicated structure in intestinal cells, and on that basis our aim has been to investigate the effect of representative phycotoxins on the enterocyte cytoskeleton. We have quantified for the first time the loss of enterocyte microfilament network induced by each toxin and recorded fluorescence images using a laser-scanning cytometer and confocal microscopy. Our data show that pectenotoxin-6, maitotoxin, palytoxin and ostreocin-D cause a significant reduction in the actin cytoskeleton. In addition, we found that the potency of maitotoxin, palytoxin and ostreocin-D to damage filamentous actin is related to Ca2+ influx in enterocytes. Those results identify the cytoskeleton as an early target for the toxic effect of those toxins.
Collapse
Affiliation(s)
- I R Ares
- Departamento de Farmacología, Facultad de Veterinaria de Lugo, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | | | | | | | | |
Collapse
|
47
|
Willis D, Zhang Y, Molloy GR. Transcription of brain creatine kinase in U87-MG glioblastoma is modulated by factor AP2. ACTA ACUST UNITED AC 2005; 1728:18-33. [PMID: 15777731 DOI: 10.1016/j.bbaexp.2005.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2004] [Revised: 01/14/2005] [Accepted: 01/19/2005] [Indexed: 11/28/2022]
Abstract
Our previous studies established in U87-MG glioblastoma cells that elevated cAMP increased transcription of the endogenous as well as a transiently-transfected brain creatine kinase (CKB) gene, despite the absence of a cAMP response element (CRE) in the CKB proximal promoter. This report employed transfection to show that the transcription of CKB in U87 cells is induced by transcription factor AP2alpha, which is known to be activated by cAMP. Dominant-negative forms of AP2alpha not only prevented the AP2alpha-mediated activation of CKB but also blocked the cAMP-mediated increase in CKB transcription caused by forskolin treatment. The mutation of the four potential AP2 elements within the CKB proximal promoter showed that induction of CKB by AP2 was mediated principally through the AP2 element located at -50 bp in the promoter. Electromobility shift assays revealed a protein in U87 nuclear extracts that bound to a consensus AP2alpha element as well as to the (-50) AP2 element in CKB. Interestingly, the CKB (-50) AP2 element contains GCCAATGGG which also bound NF-Y, the CCAAT-binding protein, suggesting that interplay between AP2 and NF-Y may modulate CKB transcription. This is the first report of a role for AP2 in the regulation of CKB transcription and of an AP2 element within which an NF-Y site is located.
Collapse
Affiliation(s)
- Dianna Willis
- Department of Biological Sciences, University of Delaware, 117 Wolf Hall, Newark, DE 19716, USA
| | | | | |
Collapse
|
48
|
Steinhoff M, Buddenkotte J, Shpacovitch V, Rattenholl A, Moormann C, Vergnolle N, Luger TA, Hollenberg MD. Proteinase-activated receptors: transducers of proteinase-mediated signaling in inflammation and immune response. Endocr Rev 2005; 26:1-43. [PMID: 15689571 DOI: 10.1210/er.2003-0025] [Citation(s) in RCA: 369] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Serine proteinases such as thrombin, mast cell tryptase, trypsin, or cathepsin G, for example, are highly active mediators with diverse biological activities. So far, proteinases have been considered to act primarily as degradative enzymes in the extracellular space. However, their biological actions in tissues and cells suggest important roles as a part of the body's hormonal communication system during inflammation and immune response. These effects can be attributed to the activation of a new subfamily of G protein-coupled receptors, termed proteinase-activated receptors (PARs). Four members of the PAR family have been cloned so far. Thus, certain proteinases act as signaling molecules that specifically regulate cells by activating PARs. After stimulation, PARs couple to various G proteins and activate signal transduction pathways resulting in the rapid transcription of genes that are involved in inflammation. For example, PARs are widely expressed by cells involved in immune responses and inflammation, regulate endothelial-leukocyte interactions, and modulate the secretion of inflammatory mediators or neuropeptides. Together, the PAR family necessitates a paradigm shift in thinking about hormone action, to include proteinases as key modulators of biological function. Novel compounds that can modulate PAR function may be potent candidates for the treatment of inflammatory or immune diseases.
Collapse
Affiliation(s)
- Martin Steinhoff
- Department of Dermatology and Boltzmann Institute for Immunobiology of the Skin, University of Münster, von-Esmarch-Strasse 58, 48149 Münster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
G protein-coupled receptors (GPCR) interact not only with heterotrimeric G proteins but also with accessory proteins called GPCR interacting proteins (GIP). These proteins have important functions. They are implicated in GPCR targeting to specific cellular compartments, in their assembling into large functional complexes called "receptosomes," in their trafficking to and from the plasma membrane, and in the fine-tuning of their signaling properties. There are several types of GIPs. Some are transmembrane proteins such as another GPCR (homodimerization and heterodimerization), ionic channels, ionotropic receptors, and single transmembrane proteins. The latter is implicated in the fine-tuning of receptor pharmacology or signaling. Other GIPs are soluble proteins interacting mainly with the "magic" C-terminal tail. Among them, PDZ domain-containing proteins are the most abundant. They generally, but not always, interact with the extreme C-terminal domain of GPCRs. Some GIPs interact with specific sequences of the C-terminal such as the Homer binding sequence (-PPxxFR-), the dopamine receptor interacting protein (DRIP) binding sequence (-FxxxFxxxF-), etc. Finally, only few GIPs have been found thus far to interact with the third intracellular loop of GPCRs. The future will tell us if this situation is only due to technical reasons.
Collapse
Affiliation(s)
- Joël Bockaert
- UPR CNRS 2580, CCIPE, 141 Rue de la Cardonille, 34094 Montpellier Cedex 5, France.
| | | | | | | |
Collapse
|
50
|
Inoue K, Ueno S, Fukuda A. Interaction of neuron-specific K+-Cl- cotransporter, KCC2, with brain-type creatine kinase. FEBS Lett 2004; 564:131-5. [PMID: 15094054 DOI: 10.1016/s0014-5793(04)00328-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Revised: 03/04/2004] [Accepted: 03/05/2004] [Indexed: 11/16/2022]
Abstract
gamma-Aminobutyric acid, a major inhibitory neurotransmitter within the adult central nervous system, is also known to be excitatory at early developmental stages due to the elevated intracellular Cl(-) concentration. This functional change is primarily attributable to a K(+)-Cl(-) cotransporter, KCC2, the expression of which is developmentally regulated in neurons. However, little detail information is available concerning the intracellular regulation of KCC2 function. Here, we identify an interaction between KCC2 and brain-type creatine kinase by means of yeast two-hybrid screening. This interaction, which was also detected in cultured cells and brain extracts, might contribute to KCC2-mediated modulation of Cl(-) homeostasis.
Collapse
Affiliation(s)
- Koichi Inoue
- Department of Physiology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka 431-3192, Japan.
| | | | | |
Collapse
|