Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Sci Rep ; 8(1): 4670, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29549309

ABSTRACT

Small-conductance Ca2+-activated K+ (SK) channels regulate the excitability of cardiomyocytes by integrating intracellular Ca2+ and membrane potentials on a beat-to-beat basis. The inextricable interplay between activation of SK channels and Ca2+ dynamics suggests the pathology of one begets another. Yet, the exact mechanistic underpinning for the activation of cardiac SK channels remains unaddressed. Here, we investigated the intracellular Ca2+ microdomains necessary for SK channel activation. SK currents coupled with Ca2+ influx via L-type Ca2+ channels (LTCCs) continued to be elicited after application of caffeine, ryanodine or thapsigargin to deplete SR Ca2+ store, suggesting that LTCCs provide the immediate Ca2+ microdomain for the activation of SK channels in cardiomyocytes. Super-resolution imaging of SK2, Cav1.2 Ca2+ channel, and ryanodine receptor 2 (RyR2) was performed to quantify the nearest neighbor distances (NND) and localized the three molecules within hundreds of nanometers. The distribution of NND between SK2 and RyR2 as well as SK2 and Cav1.2 was bimodal, suggesting a spatial relationship between the channels. The activation mechanism revealed by our study paved the way for the understanding of the roles of SK channels on the feedback mechanism to regulate the activities of LTCCs and RyR2 to influence local and global Ca2+ signaling.


Subject(s)
Calcium Channels, L-Type/metabolism , Myocytes, Cardiac/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Caffeine/pharmacology , Calcium Signaling , Cells, Cultured , HEK293 Cells , Humans , Male , Membrane Potentials , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rabbits , Ryanodine/pharmacology , Thapsigargin/pharmacology
2.
Article in English | MEDLINE | ID: mdl-29025768

ABSTRACT

BACKGROUND: Intracellular pH (pHi) is critical to cardiac excitation and contraction; uncompensated changes in pHi impair cardiac function and trigger arrhythmia. Several ion transporters participate in cardiac pHi regulation. Our previous studies identified several isoforms of a solute carrier Slc26a6 to be highly expressed in cardiomyocytes. We show that Slc26a6 mediates electrogenic Cl-/HCO3- exchange activities in cardiomyocytes, suggesting the potential role of Slc26a6 in regulation of not only pHi, but also cardiac excitability. METHODS AND RESULTS: To test the mechanistic role of Slc26a6 in the heart, we took advantage of Slc26a6 knockout (Slc26a6-/- ) mice using both in vivo and in vitro analyses. Consistent with our prediction of its electrogenic activities, ablation of Slc26a6 results in action potential shortening. There are reduced Ca2+ transient and sarcoplasmic reticulum Ca2+ load, together with decreased sarcomere shortening in Slc26a6-/- cardiomyocytes. These abnormalities translate into reduced fractional shortening and cardiac contractility at the in vivo level. Additionally, pHi is elevated in Slc26a6-/- cardiomyocytes with slower recovery kinetics from intracellular alkalization, consistent with the Cl-/HCO3- exchange activities of Slc26a6. Moreover, Slc26a6-/- mice show evidence of sinus bradycardia and fragmented QRS complex, supporting the critical role of Slc26a6 in cardiac conduction system. CONCLUSIONS: Our study provides mechanistic insights into Slc26a6, a unique cardiac electrogenic Cl-/HCO3- transporter in ventricular myocytes, linking the critical roles of Slc26a6 in regulation of pHi, excitability, and contractility. pHi is a critical regulator of other membrane and contractile proteins. Future studies are needed to investigate possible changes in these proteins in Slc26a6-/- mice.


Subject(s)
Action Potentials , Antiporters/deficiency , Excitation Contraction Coupling , Heart Rate , Myocardial Contraction , Myocytes, Cardiac/metabolism , Animals , Antiporters/genetics , Bradycardia/genetics , Bradycardia/metabolism , Bradycardia/physiopathology , CHO Cells , Cricetulus , Genotype , Hydrogen-Ion Concentration , Kinetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice, 129 Strain , Mice, Knockout , Phenotype , Sarcomeres/metabolism , Sarcoplasmic Reticulum/metabolism , Sulfate Transporters , Transfection
3.
Biochem Cell Biol ; 91(5): 287-94, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24032677

ABSTRACT

Polo-like kinase 1 (Plk1) is a conserved key regulator of the G2/M transition, but its upstream spatiotemporal regulators remain unknown. With the help of immunofluorescence, co-immunoprecipitation, and glutathione S-transferase (GST) pull-down assay, we found that calmodulin (CaM) is one such regulatory molecule that associates with Plk1 from G2 to metaphase. More importantly, this interaction results in considerable stimulation of Plk1 kinase activity leading to hyperphosphorylation of Cdc25C. Our results provide new insight into the role of CaM as an upstream regulator of Plk1 activation during mitotic entry.


Subject(s)
Calmodulin/metabolism , Cell Cycle Proteins/metabolism , Cell Division/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , cdc25 Phosphatases/metabolism , Cell Line , Centrosome/metabolism , Enzyme Activation , G2 Phase , HEK293 Cells , HeLa Cells , Humans , Mitosis , Phosphorylation , Signal Transduction/genetics , Polo-Like Kinase 1
4.
Cell Stress Chaperones ; 14(4): 343-53, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18989758

ABSTRACT

The cell cycle is controlled by regulators functioning at the right time and at the right place. We have found that calmodulin (CaM) has specific distribution patterns during different cell-cycle stages. Here, we identify cell-cycle-specific binding proteins of CaM and examine their function during cell-cycle progression. We first applied immunoprecipitation methods to isolate CaM-binding proteins from cell lysates obtained at different cell-cycle phases and then identified these proteins using mass spectrometry methods. A total of 41 proteins were identified including zinc finger proteins, ribosomal proteins, and heat shock proteins operating in a Ca(2+)-dependent or independent manner. Fifteen proteins were shown to interact with CaM in a cell-phase-specific manner. The association of the selected proteins and CaM were confirmed with in vitro immunoprecipitation and immunostaining methods. One of the identified proteins, heat shock protein 70 (Hsp70), was further studied with respect to its cell-cycle-related function. In vivo fluorescence resonance energy transfer (FRET) analysis showed that the interaction of CaM and Hsp70 was found in the nucleus during the S phase. Overexpression of Hsp70 is shown to arrest cells at S phase and, thus, induce cell apoptosis. When we disrupted the CaM-Hsp70 association with HSP70 truncation without the CaM-binding domain, we found that S-phase arrest and apoptosis could be rescued. The results suggest that the spatial and temporal association of CaM and Hsp70 can regulate cell-cycle progression and cell apoptosis.


Subject(s)
Apoptosis , Calmodulin/metabolism , HSP70 Heat-Shock Proteins/metabolism , Calmodulin/analysis , Cell Line , Fluorescence Resonance Energy Transfer , HSP70 Heat-Shock Proteins/analysis , Humans , Immunoprecipitation , Protein Binding , Protein Structure, Tertiary , S Phase
5.
Int J Biochem Cell Biol ; 40(2): 284-93, 2008.
Article in English | MEDLINE | ID: mdl-17884685

ABSTRACT

Calmodulin (CaM) is a major cytoplasmic calcium receptor that performs multiple functions including cell motility. To investigate the mechanism of the regulation of CaM on cell morphology and motility, first we checked the distribution of CaM in the living cells using GFP-CaM as an indicator. We found that GFP-CaM showed a fiber-like distribution pattern in the cytosol of living Potorous tridactylis kidney (PtK2) cells but not in living HeLa cells. The endogenous CaM in heavily permeabilized HeLa was also found to display a fiber-like distribution pattern. Further examination showed that the distribution pattern of GFP-CaM was same as that of stress fibers, but not microtubules. Co-immunoprecipitation also showed that CaM can interact with actin directly or indirectly. The microinjection of trp peptide, a specific inhibitor of CaM, attenuated the polymerization of stress fibers and induced the alteration of cell morphology. A wound-healing assay and a single cell tracking experiment showed that CaM in PtK2 cells could increase cell motility. The data we have got from living cells suggested that CaM affect cell morphology and motility through binding to stress fibers and regulate f-actin polymerization.


Subject(s)
Calmodulin/metabolism , Calmodulin/physiology , Cell Movement/physiology , Cell Shape/physiology , Microtubules/metabolism , Stress Fibers/metabolism , Animals , Calmodulin/genetics , Cells, Cultured , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Kidney/cytology , Myosin-Light-Chain Kinase/physiology , Potoroidae , Protein Binding , Recombinant Fusion Proteins/metabolism , Tissue Distribution , Transfection
6.
Cell Biol Int ; 31(2): 126-34, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17081777

ABSTRACT

Angiogenesis, the formation of new blood vessels that is regulated by hypoxia, is a critical process for the growth and spread of tumors. Multiple phases of this process, including migration, adhesion, and formation of new capillary tubes, are needed for optimal tumor growth. Here, a new regulatory function for Ca2+-CaM in the vascular endothelium is described. Ca2+-CaM activation induced by hypoxia in endothelial cells is essential for angiogenic cellular responses. Inhibition of Ca2+-CaM activity suppressed endothelial cell migration, adhesion on collagen I substrate, invasion and impaired in vitro endothelial cell differentiation into tube-like structures. We also reported that CaM is co-distributed with the actin structures in the lamellipodia in migrating cells, whereas the actin cytoskeleton rearrangement induced by hypoxia was disrupted and HIF-1 transcriptional activity was decreased when treated with CaM antagonists into cultures. These data indicate that Ca2+-CaM activation is more closely associated with the regulation of angiogenic key events, especially in response to hypoxic stress.


Subject(s)
Calmodulin/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Neovascularization, Physiologic , Actins/metabolism , Calcium Signaling , Cell Adhesion , Cell Hypoxia , Cell Movement , Collagen Type I/metabolism , Gene Expression , Genes, Reporter , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Protein Transport , Pseudopodia/metabolism
7.
FEBS Lett ; 580(15): 3624-30, 2006 Jun 26.
Article in English | MEDLINE | ID: mdl-16753148

ABSTRACT

Ultraviolet (UV) irradiation can result in cell cycle arrest. The reactivation of Polo-like kinase 1 (Plk1) is necessary for cell cycle reentry. But the mechanism of how Plk1 regulates p53 in UV-induced mitotic arrest cells remained elusive. Here we find that UV treatment leads HEK293 cells to inverse changes of Plk1 and p53. Over-expression of Plk1 rescue UV-induced mitotic arrest cells by inhibiting p53 activation. Plk1 could also inhibit p53 phosphorylation at Ser15, thus facilitates its nuclear export and degradation. Further examination shows that Plk1, p53 and Cdc25C can form a large complex. Plk1 could bind to the sequence-specific DNA-binding domain of p53 and active Cdc25C by hyperphosphorylation. These results hypothesize that Plk1 and Cdc25C participate in recovery the mitotic arrest through binding to the different domain of p53. Cdc25C may first be actived by Plk1, and then its phosphatase activity makes p53 dephosphorylated at Ser15.


Subject(s)
Cell Cycle Proteins/metabolism , Mitosis/radiation effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle Proteins/genetics , Cell Line , DNA Damage/genetics , Humans , Phosphorylation/radiation effects , Phosphoserine/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Protein Transport , Proto-Oncogene Proteins/genetics , cdc25 Phosphatases/metabolism , Polo-Like Kinase 1
8.
Mol Cancer Ther ; 4(10): 1577-84, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16227408

ABSTRACT

c-Met is highly expressed and constitutively activated in various human tumors. We employed adenovirus-mediated RNA interference technique to knock down c-Met expression in hepatocellular carcinoma cells and observed its effects on hepatocellular carcinoma cell growth in vitro and in vivo. Among the five hepatocellular carcinoma and one normal human liver cell lines we analyzed, c-Met was highly expressed and constitutively tyrosine phosphorylated in only MHCC97-L and HCCLM3 hepatocellular carcinoma cells. Knockdown of c-Met could inhibit MHCC97-L cells proliferation by arresting cells at G0-G1 phase. Soft agar colony formation assay indicated that the colony forming ability of MHCC97-L cells decreased by approximately 70% after adenovirus AdH1-small interfering RNA (siRNA)/met infection. In vivo experiments showed that adenovirus AdH1-siRNA/met inhibited the tumorigenicity of MHCC97-L cells and significantly suppressed tumor growth when injected directly into tumors. These results suggest that knockdown of c-Met by adenovirus-delivered siRNA may be a potential therapeutic strategy for treatment of hepatocellular carcinoma in which c-Met is overexpressed.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , RNA, Small Interfering/genetics , Adenoviridae/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line , Cell Line, Tumor , Flow Cytometry , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation , Proto-Oncogene Proteins c-met/biosynthesis , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
9.
Cell Res ; 15(7): 548-52, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16045818

ABSTRACT

A transient postanaphase repositioning of the centriole is found to control the completion of cytokinesis. Using a green fluorescent protein-calmodulin fusion protein as a living cell probe, we have previously found that calmodulin is associated with the initiation and progression of cytokinesis. In this study, we further studied the effect of calmodulin on the repositioning of the centriole and subsequent cell cycle progression. When activity of calmodulin is inhibited, the regression of the centriole from the intercellular bridge to the cell center is blocked, and thus the completion of cell division is repressed and two daughter cells are linked by longer cell bridge in perturbed cells. W7 treatment during cytokinesis also results in unfinished cytokinesis and stopped G1 phase. These results suggest that calmodulin activity is required for centriole repositioning and can affect the completion of cytokinesis and cell cycle progression.


Subject(s)
Anaphase/physiology , Calmodulin/pharmacology , Centrioles/physiology , Cytokinesis/physiology , Anaphase/drug effects , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Line , Centrioles/drug effects , Cytokinesis/drug effects , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/pharmacology , HeLa Cells , Humans , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Sulfonamides/pharmacology , Time Factors
10.
Int J Biochem Cell Biol ; 36(8): 1562-72, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15147735

ABSTRACT

Calmodulin is a major cytoplasmic calcium receptor that performs multiple functions in the cell including cytokinesis. Central spindle appears between separating chromatin masses after metaphase-anaphase transition. The interaction of microtubules from central spindle with cell cortex regulates the cleavage furrow formation. In this paper, we use green fluorescence protein (GFP)-tagged calmodulin as a living cell probe to examine the detailed dynamic redistribution and co-localization of calmodulin with central spindle during cytokinesis and the function of this distribution pattern in a tripolar HeLa cell model. We found that calmodulin is associated with spindle microtubules during mitosis and begins to aggregate with central spindle after anaphase initiation. The absence of either central spindle or central spindle-distributed calmodulin is correlated with the defect in the formation of cleavage furrow, where contractile ring-distributed CaM is also extinct. Further analysis found that both the assembly of central spindle and the formation of cleavage furrow are affected by the W7 treatment. The microtubule density of central spindle was decreased after the treatment. Only less than 10% of the synchronized cells enter cytokinesis when treated with 25 microM W7, and the completion time of furrow regression is also delayed from 10 min to at least 40 min. It is suggested that calmodulin plays a significant role in cytokinesis including furrow formation and regression, The understanding of the interaction between calmodulin and microtubules may give us insight into the mechanism through which calmodulin regulates cytokinesis.


Subject(s)
Calmodulin/metabolism , Cytokinesis , Spindle Apparatus/metabolism , Anaphase , Green Fluorescent Proteins , HeLa Cells , Humans , Microtubules/metabolism , Mitosis , Protein Transport , Sulfonamides/pharmacology , Time
11.
Cell Biol Int ; 28(4): 317-22, 2004.
Article in English | MEDLINE | ID: mdl-15109989

ABSTRACT

During meiotic maturation, mammalian oocytes undergo a series of morphological and physiological changes that prepare them for fertilization. Calcium-initiated signaling is thought to trigger these processes. In this study, we examine the spatio-temporal pattern of calcium and calmodulin (CaM), its downstream receptor, in order to investigate their association with meiotic maturation. Intracellular free calcium and activated CaM levels were measured using the fluorescent probes Calcium Green-1 and TA-CaM, respectively. The distribution patterns were examined using confocal microscopy. Both calcium and activated CaM showed a dynamic spatiotemporal distribution during meiotic maturation. After release from IBMX buffer, calcium was found to periodically translocate from the perinuclear region to the germinal vesicle (GV) in 90 s intervals. After 90 min, calcium stopped oscillating and became concentrated within the GV. After a further 60 min, the GV broke down and calcium dispersed into the ooplasm, but calcium levels were slightly lower here than in the original nuclear region. Activated CaM also showed a dynamic patterning process similar to calcium. Taking the data from calcium chelation and CaM inhibition together, our results suggest that the dynamic distribution patterns of calcium and activated CaM are crucial for oocyte maturation.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Cell Nucleus/metabolism , Meiosis/physiology , Oocytes/metabolism , Animals , Calcium Signaling/physiology , Female , Fluorescent Dyes/chemistry , Mice , Microscopy, Confocal , Oocytes/cytology
12.
Shi Yan Sheng Wu Xue Bao ; 36(5): 335-41, 2003 Oct.
Article in Chinese | MEDLINE | ID: mdl-14724944

ABSTRACT

Calmodulin (CaM) is a major cytoplasmic Ca2+ receptor and performs a multiplicity of functions in the cell. By using GFP-CaM fusion protein, we have studied the detailed dynamic redistribution of CaM during cytokinesis in HeLa cells. CaM associates with midbody in late cytokinesis phase. When the cells were treated with Ca2+/CaM inhibitor W7, the dissolving of the midbody was delayed. Moreover, we have found that gamma-tubulin colocalized with CaM at the midbody during cytokinesis. W7 treatment could affect the dissociation of gamma-tubulin from midbody. These results suggest that CaM may involve in the regulation of midbody microtubules disassembly and may thus affect the completion of cytokinesis.


Subject(s)
Calmodulin/metabolism , Centrosome/physiology , Sulfonamides/pharmacology , Tubulin/metabolism , Calmodulin/antagonists & inhibitors , Cell Cycle , Cell Division/physiology , Centrosome/metabolism , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/metabolism , Microtubules/metabolism , Recombinant Fusion Proteins/metabolism
13.
Shi Yan Sheng Wu Xue Bao ; 35(4): 249-56, 2002 Dec.
Article in Chinese | MEDLINE | ID: mdl-15346980

ABSTRACT

A cell-free system including HeLa cell lysate of synchronized metaphase or G2-phase and isolated germinal vesicles (GV) from mouse oocytes was used to study the role of calcium and its downstream mediator during mature resumption. The isolated GVs could resume meiotic maturation in the lysate prepared from M phase HeLa cell, which marked by chromatin condensation. And this process was not affected by calcium chelating agent. But calcium in lysate from G2 phase cells was critical to meiotic maturation. Only in mid-G2 phase cell lysate (released from nocodazole for about 20-23h) chromatin condensation could be induced by calcium. Calcium had no effect on the cell lysate prepared from earlier (18-20h) and later (24h) G2 phase cells. Further studies showed that down stream mediator CaM and CaMKII might also involove in this process. Inhibition the function of CaM and CaMKII could block GVBD and first polar body extrusion of DOs cultured in vitro. The target of calcium signal might be MPF because MPF was existed from mid-G2 phase to metaphase and the tyrosine phosphorylation level of Cdc2 subunit was significantly dephosphorylated in M phase. Our results further confirmed that the resumption of meiosis maturation was promoted in a calcium/CaM depended pathway.


Subject(s)
Calcium/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Cell Differentiation/physiology , Cell-Free System/metabolism , Female , HeLa Cells , Humans , Meiosis/physiology , Mesothelin , Mice , Oocytes/metabolism , Oocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL