Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Physiol Genomics ; 49(3): 115-126, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28039430

ABSTRACT

Following vascular injury medial smooth muscle cells dedifferentiate and migrate through the internal elastic lamina where they form a neointima. The goal of the current study was to identify changes in gene expression that occur before the development of neointima and are associated with the early response to injury. Vascular injury was induced in C57BL/6 mice and in Myh11-creER(T2) mTmG reporter mice by complete ligation of the left carotid artery. Reporter mice were used to visualize cellular changes in the injured vessels. Total RNA was isolated from control carotid arteries or from carotid arteries 3 days following ligation of C57BL/6 mice and analyzed by Affymetrix microarray and quantitative RT-PCR. This analysis revealed decreased expression of mRNAs encoding smooth muscle-specific contractile proteins that was accompanied by a marked increase in a host of mRNAs encoding inflammatory cytokines following injury. There was also marked decrease in molecules associated with BMP, Wnt, and Hedgehog signaling and an increase in those associated with B cell, T cell, and macrophage signaling. Expression of a number of noncoding RNAs were also altered following injury with microRNAs 143/145 being dramatically downregulated and microRNAs 1949 and 142 upregulated. Several long noncoding RNAs showed altered expression that mirrored the expression of their nearest coding genes. These data demonstrate that following carotid artery ligation an inflammatory cascade is initiated that is associated with the downregulation of coding and noncoding RNAs that are normally required to maintain smooth muscle cells in a differentiated state.


Subject(s)
Carotid Arteries/pathology , Cell Dedifferentiation , Inflammation/pathology , Muscle, Smooth, Vascular/pathology , Animals , Cytokines/metabolism , Down-Regulation/genetics , Inflammation/genetics , Inflammation Mediators/metabolism , Ligation , Male , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Muscle Contraction/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Signal Transduction/genetics , Up-Regulation/genetics
2.
Apoptosis ; 19(2): 306-15, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24185832

ABSTRACT

Death associated protein kinase (DAPK) is a large, multi-domain ser/thr kinase whose activities converge upon multiple signaling pathways that regulate autophagy, caspase-dependent cell death, cell adhesion and migration. The cellular levels of DAPK are post-translationally regulated by the combined activities of two degradation systems, including the ubiquitin proteasome and an extra-lysosomal proteolysis pathway. At least three distinct E3 ubiquitin ligases target DAPK, including mindbomb1, the chaperone dependent ligase, CHIP (carboxy terminus of Hsp70-interacting protein) and a cullin RING ligase complex, KLHL20-Cul3-RBX1. In addition, it appears that the cellular levels of DAPK are also regulated by an extra-lysosomal protease, cathepsin B. While protein quality control and recycling clearly benefit cells by removal of misfolded or toxic proteins and recycling of their components, the finding that multiple surveillance systems target DAPK suggests that these protein degradation systems also act to fine tune DAPK expression levels in response to specific signaling pathways.


Subject(s)
Death-Associated Protein Kinases/metabolism , Protein Processing, Post-Translational , Animals , Cathepsin B/metabolism , Humans , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism
3.
Biochim Biophys Acta ; 1813(12): 2036-43, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21985982

ABSTRACT

Delta ligands are important for regulating Notch signaling through transcellular stimulation of Notch receptors. The cytoplasmic tails of Delta ligands have multiple potential regulatory sites including several lysine residues that are putative targets for ubiquitination by the E3 ubiquitin ligases, Mind Bomb and Neuralized. To identify possible roles for specific lysine residues in the cytoplasmic tail of the Notch ligand Dll1 a mutational and functional analysis was performed. Examination of a panel of individual or clustered lysine mutants demonstrated that lysine 613 (K613) in the cytoplasmic tail of Dll1 is a key residue necessary for transcellular activation of Notch signaling. Multi-ubiquitination of the Dll1 mutant Dll1-K613R was altered compared to wild type Dll1, and the K613R mutation blocked the ability of Dll1 to interact with Notch1. Finally, mutation of K613 did not affect the stability of Dll1 or its ability to traffic to recycle to the plasma membrane, but did enhance the fraction associated with lipid rafts. Collectively these results suggest that the transcellular defect in Notch signaling attributed to residue K613 in cytoplasmic tail of Dll1 may result from altering its multi-ubiquitination and increasing its retention in lipid rafts.


Subject(s)
Cell Membrane/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lysine/metabolism , Membrane Microdomains/metabolism , Receptors, Notch/metabolism , Animals , Blotting, Western , Calcium-Binding Proteins , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endocytosis , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunoenzyme Techniques , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/genetics , Kidney/cytology , Kidney/metabolism , Lysine/genetics , Mice , Mutation/genetics , Protein Binding , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
4.
J Biol Chem ; 285(18): 13827-38, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20220139

ABSTRACT

The tumor suppressor, death-associated protein kinase (DAPK), is a Ca(2+)/calmodulin-regulated Ser/Thr kinase with an important role in regulating cytoskeletal dynamics. Autophosphorylation within the calmodulin-binding domain at Ser-308 inhibits DAPK catalytic activity. Dephosphorylation of Ser-308 by a previously unknown phosphatase enhances kinase activity and proteasome-mediated degradation of DAPK. In these studies, we identified two holoenzyme forms of protein phosphatase 2A (PP2A), ABalphaC and ABdeltaC, as DAPK-interacting proteins. These phosphatase holoenzymes dephosphorylate DAPK at Ser-308 in vitro and in vivo resulting in enhanced kinase activity of DAPK. The enzymatic activity of PP2A also negatively regulates DAPK levels by enhancing proteasome-mediated degradation of the kinase. Overexpression of wild type DAPK induces cell rounding and detachment in HEK293 cells; however, this effect is not observed following expression of an inactive DAPK S308E mutant. Finally, activation of DAPK by PP2A was found to be required for ceramide-induced anoikis. Together, our results provide a mechanism by which PP2A and DAPK activities control cell adhesion and anoikis.


Subject(s)
Anoikis/drug effects , Ceramides/pharmacology , Protein Phosphatase 2/metabolism , Amino Acid Substitution , Anoikis/genetics , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , Death-Associated Protein Kinases , HeLa Cells , Holoenzymes/genetics , Holoenzymes/metabolism , Humans , Mutation, Missense , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Phosphatase 2/genetics , Tumor Suppressor Proteins
5.
Am J Physiol Cell Physiol ; 297(5): C1275-83, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19710364

ABSTRACT

Mind bomb 1 (Mib1) is a multidomain E3 ligase that directs ubiquitination of the Notch ligands Delta and Jagged to promote their endocytosis. Here we examine Notch-independent functions of Mib1 and find that its activities are linked to the initiation of the extrinsic cell death pathway. Expression of Mib1 induces a spontaneous, caspase-dependent cell death. Consistent with this, depletion of endogenous Mib1 decreases tumor-necrosis factor (TNF)-induced cell death. Mib1 was found to bind to cellular Fas-associated death domain (FADD)-like IL-1b converting enzyme (FLICE)-like inhibitory proteins (cFLIP-L and cFLIP-S), whereas only cFLIP-s can inhibit Mib1-induced cell death. The interaction between Mib1 and cFLIP decreases the association of caspase-8 with cFLIP, which activates caspase-8 and induces cell death. Collectively, these results suggest that in addition to a central role in Notch signaling, Mib1 has an important role in regulating the extrinsic cell death pathway.


Subject(s)
Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Blotting, Western , Cell Line , Humans , Immunoprecipitation , Ubiquitination
6.
Dev Biol ; 325(2): 374-85, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19027000

ABSTRACT

Cell-cell fusion is a fundamental cellular process that is essential for development as well as fertilization. Myoblast fusion to form multinucleated skeletal muscle myotubes is a well studied, yet incompletely understood example of cell-cell fusion that is essential for formation of contractile skeletal muscle tissue. Studies in this report identify several novel cytoskeletal events essential to an early phase of myoblast fusion among cultured murine myoblasts. During myoblast pairing and alignment, cortical actin filaments organize into a dense actin wall structure that parallels and extends the length of the plasma membrane of the bipolar, aligned cells. As fusion progresses, gaps appear within the actin wall at sites of vesicle accumulation, the vesicles pair across the aligned myoblasts, cell-cell contacts and fusion pores form. Inhibition of nonmuscle myosin IIA (NM-MHC-IIA) motor activity prevents formation of this cortical actin wall, as well as the appearance of vesicles at a membrane proximal location, and myoblast fusion. These results suggest that early formation of a subplasmalemmal actin wall during myoblast alignment is a critical event for myoblast fusion that supports bipolar membrane alignment and temporally regulates trafficking of vesicles to the nascent fusion sites during skeletal muscle myoblast differentiation.


Subject(s)
Actin Cytoskeleton/physiology , Membrane Fusion/physiology , Myoblasts/physiology , Nonmuscle Myosin Type IIA/metabolism , Animals , Biological Transport , Cell Differentiation , Cell Fusion , Cell Membrane/physiology , Cells, Cultured , Myoblasts/ultrastructure , Rats , Secretory Vesicles/physiology
7.
J Biol Chem ; 282(16): 11795-804, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17324930

ABSTRACT

Death-associated protein kinase (DAPK) has been found associated with HSP90, and inhibition of HSP90 with 17-alkylamino-17-demethoxygeldanamycin reduced expression of DAPK. These results were extended to determine whether the degradation of DAPK in the absence of HSP90 activity is dependent on the ubiquitin-proteasome pathway. Our results show that treatment of cells with geldanamycin (GA) leads to degradation of DAPK, and this degradation is attenuated by the proteasome inhibitor, lactacystin. GA-induced DAPK degradation is also dependent on phosphorylation of DAPK at Ser(308), and the cellular levels of phospho(Ser(308))-DAPK dramatically increase in response to GA treatment. Expression of two distinct ubiquitin E3 ligases, carboxyl terminus of HSC70-interacting protein (CHIP) or DIP1/Mib1, enhanced DAPK degradation, and conversely, short interfering RNA depletion of either CHIP or DIP1/Mib1 attenuated DAPK degradation. In vitro ubiquitination assays confirmed that DAPK is targeted for ubiquitination by both CHIP and DIP. Consistent with these results, DAPK is found in two distinct immune complexes, one containing HSP90 and CHIP and a second complex containing only DIP1/Mib. Collectively, these results indicate that strict modulation of DAPK activities is critical for regulation of apoptosis and cellular homeostasis.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Gene Expression Regulation, Enzymologic , HSP90 Heat-Shock Proteins/chemistry , Apoptosis , Benzoquinones/pharmacology , Catalysis , Death-Associated Protein Kinases , HSP70 Heat-Shock Proteins/chemistry , HeLa Cells , Humans , Lactams, Macrocyclic/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Serine/chemistry , Ubiquitin/chemistry , Ubiquitin/metabolism
8.
J Biol Chem ; 281(51): 39033-40, 2006 Dec 22.
Article in English | MEDLINE | ID: mdl-17056602

ABSTRACT

Activation of death-associated protein kinase (DAPK) occurs via dephosphorylation of Ser-308 and subsequent association of calcium/calmodulin. In this study, we confirmed the existence of the alternatively spliced human DAPK-beta, and we examined the levels of DAPK autophosphorylation and DAPK catalytic activity in response to tumor necrosis factor or ceramide. It was found that DAPK is rapidly dephosphorylated in response to tumor necrosis factor or ceramide and then subsequently degraded via proteasome activity. Dephosphorylation and activation of DAPK are shown to temporally precede its subsequent degradation. This results in an initial increase in kinase activity followed by a decrease in DAPK expression and activity. The decline in DAPK expression is paralleled with increased caspase activity and cell apoptosis. These results suggest that the apoptosis regulatory activities mediated by DAPK are controlled both by phosphorylation status and protein stability.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/physiology , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Proteasome Endopeptidase Complex/metabolism , Alkaline Phosphatase/metabolism , Alternative Splicing , Animals , Apoptosis , Caspases/metabolism , Cell Death , Death-Associated Protein Kinases , HeLa Cells , Humans , Mice , Molecular Sequence Data , Phosphorylation , Serine/chemistry , Tumor Necrosis Factors/metabolism
9.
Am J Physiol Cell Physiol ; 291(5): C817-27, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16774989

ABSTRACT

The mylk1 gene is a large gene spanning approximately 250 kb and comprising at least 31 exons. The mylk1 gene encodes at least four protein products: two isoforms of the 220-kDa myosin light chain kinase (MLCK), a 130-kDa MLCK, and telokin. Transcripts encoding these products are derived from four independent promoters within the mylk1 gene. The kinases expressed from the mylk1 gene have been extensively characterized and function to regulate the activity of nonmuscle and smooth muscle myosin II. Activation of these myosin motors by MLCK modulates a variety of contractile processes, including smooth muscle contraction, cell adhesion, migration, and proliferation. Dysregulation of these processes contributes to a number of diseases. The noncatalytic gene product telokin also has been shown to modulate contraction in smooth muscle cells through its ability to inhibit myosin light chain phosphatase. Given the crucial role of the products of the mylk1 gene in regulating numerous contractile processes, it seems intuitive that alterations in the transcriptional activity of the mylk1 gene also will have a significant impact on many physiological and pathological processes. In this review we highlight some of the recent studies that have described the transcriptional regulation of mylk1 gene products in smooth muscle tissues and discuss the implications of these findings for regulation of expression of other smooth muscle-specific genes.


Subject(s)
Gene Expression Regulation, Enzymologic , Muscle, Smooth/enzymology , Myosin-Light-Chain Kinase/genetics , Peptides/genetics , AT Rich Sequence/genetics , Animals , Humans , Peptide Fragments , Promoter Regions, Genetic/genetics
10.
Am J Physiol Cell Physiol ; 286(1): C8-21, 2004 Jan.
Article in English | MEDLINE | ID: mdl-12967916

ABSTRACT

Thus far, determining the relative contribution of Ca2+/calmodulin-dependent myosin light chain kinase (MLCK) and Ca2+-independent Rho-kinase pathways to myosin II activation and contraction has been difficult. In this study, we characterize the role of Rho-kinase in a rat embryo fibroblast cell line (REF-52), which contains no detectable MLCK. No endogenous MLCK could be detected in REF-52 cells by either Western or Northern blot analysis. In the presence or absence of Ca2+, thrombin or lysophosphatidic acid (LPA) increased RhoA activity and Rhokinase activity, correlating with isometric tension development and myosin II regulatory light chain (RLC) phosphorylation. Resting tension is associated with a basal phosphorylation of 0.31 +/- 0.02 mol PO4/mol RLC, whereas upon LPA or thrombin treatment myosin II RLC phosphorylation increases to 1.08 +/- 0.05 and 0.82 +/- 0.05 mol PO4/mol RLC, respectively, within 2.5 min. Ca2+ chelation has minimal effect on the kinetics and magnitude of isometric tension development and RLC phosphorylation. Treatment of REF-52 cells with the Rho-kinase-specific inhibitor Y-27632 abolished thrombin- and LPA-stimulated contraction and RLC phosphorylation. These results suggest that Rho-kinase is sufficient to activate myosin II motor activity and contraction in REF-52 cells.


Subject(s)
Calcium/physiology , Fibroblasts/physiology , Protein Serine-Threonine Kinases/physiology , Amides/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Intracellular Signaling Peptides and Proteins , Lysophospholipids/pharmacology , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , Rats , Thrombin/pharmacology , rho-Associated Kinases , rhoA GTP-Binding Protein/metabolism
11.
J Biol Chem ; 278(51): 51587-93, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14530257

ABSTRACT

Death-associated protein kinases (DAPK) are serine/threonine protein kinases that have an important role in regulating cell death. In this study two antisense approaches were employed to down-regulate expression of the endogenous DAPK-alpha and DAPK-beta proteins. Transient expression of an antisense DAPK cDNA or antisense morpholino oligonucleotides in HeLa, 3T3, or primary human vascular smooth muscle cells demonstrate that decreased DAPK expression promotes a spontaneous, caspase-mediated apoptosis as evidenced by increased activities of caspases-3 and -9. Clonal HeLa cell lines with attenuated levels of DAPK expression, obtained following selection in the presence of antisense DAPK cDNA, are more sensitive to tumor necrosis factor-induced caspase-mediated apoptosis, and their sensitivity is inversely related to DAPK expression. In contrast, HeLa cells with reduced DAPK expression are moderately resistant to cell death induced by interferon-gamma. This finding is consistent with previous studies showing that DAPK has a role in promoting caspase-independent cell death. Together, these studies demonstrate that the cellular activities of DAPK are critical for antagonizing caspase-dependent apoptosis to promote cell survival under normal cell growth conditions.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/physiology , DNA, Antisense/pharmacology , Animals , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Caspases/analysis , Cell Survival , Death-Associated Protein Kinases , Down-Regulation/drug effects , HeLa Cells , Humans , Interferon-gamma/pharmacology , Mice , Muscle, Smooth, Vascular/cytology , NIH 3T3 Cells , Protein Isoforms/drug effects , Protein Isoforms/physiology , Transfection
12.
J Cell Physiol ; 194(2): 194-205, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12494458

ABSTRACT

In bone, a large proportion of osteoblasts, the cells responsible for deposition of new bone, normally undergo programmed cell death (apoptosis). Because mechanical loading of bone increases the rate of new bone formation, we hypothesized that mechanical stimulation of osteoblasts might increase their survival. To test this hypothesis, we investigated the effects of fluid shear stress (FSS) on osteoblast apoptosis using three osteoblast cell types: primary rat calvarial osteoblasts (RCOB), MC3T3-E1 osteoblastic cells, and UMR106 osteosarcoma cells. Cells were treated with TNF-alpha in the presence of cyclohexamide (CHX) to rapidly induce apoptosis. Osteoblasts showed significant signs of apoptosis within 4-6 h of exposure to TNF-alpha and CHX, and application of FSS (12 dyne/cm(2)) significantly attenuated this TNF-alpha-induced apoptosis. FSS activated PI3-kinase signaling, induced phosphorylation of Akt, and inhibited TNF-alpha-induced activation of caspase-3. Inhibition of PI3-kinase, using LY294002, blocked the ability of FSS to rescue osteoblasts from TNF-alpha-induced apoptosis and blocked FSS-induced inhibition of caspase-3 activation in osteoblasts treated with TNF-alpha. LY294002 did not, however, prevent FSS-induced phosphorylation of Akt suggesting that activation of Akt alone is not sufficient to rescue cells from apoptosis. This result also suggests that FSS can activate Akt via a PI3-kinase-independent pathway. These studies demonstrate for the first time that application of FSS to osteoblasts in vitro results in inhibition of TNF-alpha-induced apoptosis through a mechanism involving activation of PI3-kinase signaling and inhibition of caspases. FSS-induced activation of PI3-kinase may promote cell survival through a mechanism that is distinct from the Akt-mediated survival pathway.


Subject(s)
Apoptosis/physiology , Caspase Inhibitors , Osteoblasts/drug effects , Osteoblasts/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Tumor Necrosis Factor-alpha/pharmacology , Animals , Caspase 3 , Cell Survival/physiology , Cells, Cultured , Enzyme Activation/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Rats , Stress, Mechanical
13.
J Biol Chem ; 277(49): 46980-6, 2002 Dec 06.
Article in English | MEDLINE | ID: mdl-12351649

ABSTRACT

Death-associated protein kinase (DAPK) is a multi-domain Ser/Thr protein kinase with an important role in apoptosis regulation. In these studies we have identified a DAPK-interacting protein called DIP-1, which is a novel multi-RING finger protein. The RING finger motifs of DIP-1 have E3 ligase activity that can auto-ubiquitinate DIP-1 in vitro. In vivo, DIP-1 is detected as a polyubiquitinated protein, suggesting that the intracellular levels of DIP-1 are regulated by the ubiquitin-proteasome system. Transient expression of DIP-1 in HeLa cells antagonizes the anti-apoptotic function of DAPK to promote a caspase-dependent apoptosis. These studies also demonstrate that DAPK is an in vitro and in vivo target for ubiquitination by DIP-1, thereby providing a mechanism by which DAPK activities can be regulated through proteasomal degradation.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic , Ligases/biosynthesis , Ligases/chemistry , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Blotting, Northern , Blotting, Western , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Cell Line , Cloning, Molecular , Cysteine Endopeptidases/metabolism , Death-Associated Protein Kinases , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Fungal Proteins/metabolism , HeLa Cells , Humans , Mice , Molecular Sequence Data , Multienzyme Complexes/metabolism , Precipitin Tests , Proteasome Endopeptidase Complex , Protein Binding , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Time Factors , Tissue Distribution , Two-Hybrid System Techniques , Ubiquitin/metabolism , Ubiquitin-Protein Ligases
14.
Am J Physiol Cell Physiol ; 282(3): C451-60, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11832329

ABSTRACT

To better understand the distinct functional roles of the 220- and 130-kDa forms of myosin light chain kinase (MLCK), expression and intracellular localization were determined during development and in adult mouse tissues. Northern blot, Western blot, and histochemical studies show that the 220-kDa MLCK is widely expressed during development as well as in several adult smooth muscle and nonmuscle tissues. The 130-kDa MLCK is highly expressed in all adult tissues examined and is also detectable during embryonic development. Colocalization studies examining the distribution of 130- and 220-kDa mouse MLCKs revealed that the 130-kDa MLCK colocalizes with nonmuscle myosin IIA but not with myosin IIB or F-actin. In contrast, the 220-kDa MLCK did not colocalize with either nonmuscle myosin II isoform but instead colocalizes with thick interconnected bundles of F-actin. These results suggest that in vivo, the physiological functions of the 220- and 130-kDa MLCKs are likely to be regulated by their intracellular trafficking and distribution.


Subject(s)
Myosin-Light-Chain Kinase/metabolism , Actins/metabolism , Amino Acid Sequence , Animals , Blotting, Northern , Blotting, Western , Cell Line , Cells, Cultured , Cloning, Molecular , Embryo, Mammalian/enzymology , Embryonic and Fetal Development , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Humans , Immunohistochemistry , Mice , Molecular Sequence Data , Muscle, Smooth/enzymology , Myosin-Light-Chain Kinase/chemistry , Myosin-Light-Chain Kinase/genetics , Myosins/metabolism , Organ Specificity , Protein Isoforms , Sequence Alignment , Tissue Distribution
15.
J Cell Biol ; 156(3): 543-53, 2002 Feb 04.
Article in English | MEDLINE | ID: mdl-11815633

ABSTRACT

Approaches with high spatial and temporal resolution are required to understand the regulation of nonmuscle myosin II in vivo. Using fluorescence resonance energy transfer we have produced a novel biosensor allowing simultaneous determination of myosin light chain kinase (MLCK) localization and its [Ca2+]4/calmodulin-binding state in living cells. We observe transient recruitment of diffuse MLCK to stress fibers and its in situ activation before contraction. MLCK is highly active in the lamella of migrating cells, but not at the retracting tail. This unexpected result highlights a potential role for MLCK-mediated myosin contractility in the lamella as a driving force for migration. During cytokinesis, MLCK was enriched at the spindle equator during late metaphase, and was maximally activated just before cleavage furrow constriction. As furrow contraction was completed, active MLCK was redistributed to the poles of the daughter cells. These results show MLCK is a myosin regulator in the lamella and contractile ring, and pinpoints sites where myosin function may be mediated by other kinases.


Subject(s)
Biosensing Techniques/methods , Cell Movement/physiology , Cytoskeleton/metabolism , Intracellular Membranes/metabolism , Myosin-Light-Chain Kinase/metabolism , Myosins/metabolism , Pseudopodia/metabolism , Animals , Calcium/metabolism , Calcium Signaling/physiology , Calmodulin/metabolism , Cell Division/physiology , Cell Polarity/physiology , Dipodomys , Microscopy, Fluorescence/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...