Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Diabetes ; 62(4): 1139-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23274900

ABSTRACT

In skeletal muscle, the actin cytoskeleton-regulating GTPase, Rac1, is necessary for insulin-dependent GLUT4 translocation. Muscle contraction increases glucose transport and represents an alternative signaling pathway to insulin. Whether Rac1 is activated by muscle contraction and regulates contraction-induced glucose uptake is unknown. Therefore, we studied the effects of in vivo exercise and ex vivo muscle contractions on Rac1 signaling and its regulatory role in glucose uptake in mice and humans. Muscle Rac1-GTP binding was increased after exercise in mice (~60-100%) and humans (~40%), and this activation was AMP-activated protein kinase independent. Rac1 inhibition reduced contraction-stimulated glucose uptake in mouse muscle by 55% in soleus and by 20-58% in extensor digitorum longus (EDL; P < 0.01). In agreement, the contraction-stimulated increment in glucose uptake was decreased by 27% (P = 0.1) and 40% (P < 0.05) in soleus and EDL muscles, respectively, of muscle-specific inducible Rac1 knockout mice. Furthermore, depolymerization of the actin cytoskeleton decreased contraction-stimulated glucose uptake by 100% and 62% (P < 0.01) in soleus and EDL muscles, respectively. These are the first data to show that Rac1 is activated during muscle contraction in murine and human skeletal muscle and suggest that Rac1 and possibly the actin cytoskeleton are novel regulators of contraction-stimulated glucose uptake.


Subject(s)
Glucose/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Neuropeptides/metabolism , rac GTP-Binding Proteins/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adult , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Electric Stimulation , Exercise Test , Female , Humans , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Muscle, Skeletal/cytology , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Ribonucleotides/pharmacology , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
2.
Mol Biol Cell ; 23(20): 4065-78, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22918957

ABSTRACT

GLUT4-containing vesicles cycle between the plasma membrane and intracellular compartments. Insulin promotes GLUT4 exocytosis by regulating GLUT4 vesicle arrival at the cell periphery and its subsequent tethering, docking, and fusion with the plasma membrane. The molecular machinery involved in GLUT4 vesicle tethering is unknown. We show here that Myo1c, an actin-based motor protein that associates with membranes and actin filaments, is required for insulin-induced vesicle tethering in muscle cells. Myo1c was found to associate with both mobile and tethered GLUT4 vesicles and to be required for vesicle capture in the total internal reflection fluorescence (TIRF) zone beneath the plasma membrane. Myo1c knockdown or overexpression of an actin binding-deficient Myo1c mutant abolished insulin-induced vesicle immobilization, increased GLUT4 vesicle velocity in the TIRF zone, and prevented their externalization. Conversely, Myo1c overexpression immobilized GLUT4 vesicles in the TIRF zone and promoted insulin-induced GLUT4 exposure to the extracellular milieu. Myo1c also contributed to insulin-dependent actin filament remodeling. Thus we propose that interaction of vesicular Myo1c with cortical actin filaments is required for insulin-mediated tethering of GLUT4 vesicles and for efficient GLUT4 surface delivery in muscle cells.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Cytoplasmic Vesicles/metabolism , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Myosin Type I/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Membrane/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoplasmic Vesicles/drug effects , Cytosol/drug effects , Cytosol/metabolism , Endocytosis/drug effects , Exocytosis/drug effects , Gene Knockdown Techniques , Humans , Mice , Microscopy, Fluorescence , Protein Binding/drug effects , Protein Transport/drug effects , Rats
3.
Biochemistry ; 50(15): 3048-61, 2011 Apr 19.
Article in English | MEDLINE | ID: mdl-21405107

ABSTRACT

Glucose transporter 4 (GLUT4) is responsible for the uptake of glucose into muscle and adipose tissues. Under resting conditions, GLUT4 is dynamically retained through idle cycling among selective intracellular compartments, from whence it undergoes slow recycling to the plasma membrane (PM). This dynamic retention can be released by command from intracellular signals elicited by insulin and other stimuli, which result in 2-10-fold increases in the surface level of GLUT4. Insulin-derived signals promote translocation of GLUT4 to the PM from a specialized compartment termed GLUT4 storage vesicles (GSV). Much effort has been devoted to the characterization of the intracellular compartments and dynamics of GLUT4 cycling and to the signals by which GLUT4 is sorted into, and recruited from, GSV. This review summarizes our understanding of intracellular GLUT4 traffic during its internalization from the membrane, its slow, constitutive recycling, and its regulated exocytosis in response to insulin. In spite of specific differences in GLUT4 dynamic behavior in adipose and muscle cells, the generalities of its endocytic and exocytic itineraries are consistent and an array of regulatory proteins that regulate each vesicular traffic event emerges from these cell systems.


Subject(s)
Endocytosis , Exocytosis , Glucose Transporter Type 4/metabolism , Animals , Cell Membrane/metabolism , Humans , Insulin Resistance , Intracellular Space/metabolism
4.
J Cell Physiol ; 226(1): 173-80, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20658540

ABSTRACT

The signaling pathways that stimulate glucose uptake in response to muscle contraction are not well defined. Recently, we showed that carbachol, an acetylcholine analog, stimulates contraction of C2C12 myotube cultures and the rapid arrival of myc-epitope tagged GLUT4 glucose transporters at the cell surface. Here, we explore a role for protein kinase C (PKC) in regulating GLUT4 traffic. Cell surface carbachol-induced GLUT4myc levels were partly inhibited by the conventional/novel PKC inhibitors GF-109203X, Gö6983, and Ro-31-8425 but not by the conventional PKC inhibitor Gö6976. C2C12 myotubes expressed several novel isoforms of PKC mRNA with PKCδ and PKCε in greater abundance. Carbachol stimulated phosphorylation of PKC isoforms and translocation of PKCδ and PKCε to membranes within 5 min. However, only a peptidic inhibitor of PKCε translocation (myristoylated-EAVSLKPT), but not one of PKCδ (myristoylated-SFNSYELGSL), prevented the GLUT4myc response to carbachol. Significant participation of PKCε in the carbachol-induced gain of GLUT4myc at the surface of C2C12 myotubes was further supported through siRNA-mediated PKCε protein knockdown. These findings support a role for novel PKC isoforms, especially PKCε, in contraction-stimulated GLUT4 traffic in muscle cells.


Subject(s)
Gene Expression Regulation/physiology , Glucose Transporter Type 4/metabolism , Muscle Contraction/physiology , Myoblasts/metabolism , Protein Kinase C-epsilon/metabolism , Adenylate Kinase/genetics , Adenylate Kinase/metabolism , Animals , Carbachol/administration & dosage , Carbachol/pharmacology , Dose-Response Relationship, Drug , Glucose Transporter Type 4/genetics , Mice , Myoblasts/drug effects , Protein Kinase C-epsilon/genetics
5.
Am J Physiol Endocrinol Metab ; 298(5): E1058-71, 2010 May.
Article in English | MEDLINE | ID: mdl-20159855

ABSTRACT

Muscle contraction stimulates glucose uptake acutely to increase energy supply, but suitable cellular models that faithfully reproduce this complex phenomenon are lacking. To this end, we have developed a cellular model of contracting C(2)C(12) myotubes overexpressing GLUT4 with an exofacial myc-epitope tag (GLUT4myc) and explored stimulation of GLUT4 traffic by physiologically relevant agents. Carbachol (an acetylcholine receptor agonist) induced a gain in cell surface GLUT4myc that was mediated by nicotinic acetylcholine receptors. Carbachol also activated AMPK, and this response was sensitive to the contractile myosin ATPase inhibitor N-benzyl-p-toluenesulfonamide. The gain in surface GLUT4myc elicited by carbachol or by the AMPK activator 5-amino-4-carboxamide-1 beta-ribose was sensitive to chemical inhibition of AMPK activity by compound C and partially reduced by siRNA-mediated knockdown of AMPK catalytic subunits or LKB1. In addition, the carbachol-induced gain in cell surface GLUT4myc was partially sensitive to chelation of intracellular calcium with BAPTA-AM. However, the carbachol-induced gain in cell surface GLUT4myc was not sensitive to the CaMKK inhibitor STO-609 despite expression of both isoforms of this enzyme and a rise in cytosolic calcium by carbachol. Therefore, separate AMPK- and calcium-dependent signals contribute to mobilizing GLUT4 in response to carbachol, providing an in vitro cell model that recapitulates the two major signals whereby acute contraction regulates glucose uptake in skeletal muscle. This system will be ideal to further analyze the underlying molecular events of contraction-regulated GLUT4 traffic.


Subject(s)
Calcium/metabolism , Glucose Transporter Type 4/metabolism , Muscle Contraction/physiology , Muscle Fibers, Skeletal/metabolism , AMP-Activated Protein Kinases/metabolism , Acetylcholine/metabolism , Animals , Blotting, Western , Calcium Signaling/drug effects , Carbachol/pharmacology , Cell Line , Cells, Cultured , Cholinergic Agonists/pharmacology , Fluorescent Antibody Technique , Glucose/metabolism , Mice , Muscle Contraction/drug effects , Muscle Fibers, Skeletal/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology
6.
Curr Biol ; 19(21): 1788-98, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19836242

ABSTRACT

BACKGROUND: The epidermal growth factor (EGF) stimulates rapid tyrosine phosphorylation of the EGF receptor (EGFR). This event precedes signaling from both the plasma membrane and from endosomes, and it is essential for recruitment of a ubiquitin ligase, CBL, that sorts activated receptors to endosomes and degradation. Because hyperphosphorylation of EGFR is involved in oncogenic pathways, we performed an unbiased screen of small interfering RNA (siRNA) oligonucleotides targeting all human tyrosine phosphatases. RESULTS: We report the identification of PTPRK and PTPRJ (density-enhanced phosphatase-1 [DEP-1]) as EGFR-targeting phosphatases. DEP-1 is a tumor suppressor that dephosphorylates and thereby stabilizes EGFR by hampering its ability to associate with the CBL-GRB2 ubiquitin ligase complex. DEP-1 silencing enhanced tyrosine phosphorylation of endosomal EGFRs and, accordingly, increased cell proliferation. In line with functional interactions, EGFR and DEP-1 form physical associations, and EGFR phosphorylates a substrate-trapping mutant of DEP-1. Interestingly, the interactions of DEP-1 and EGFR are followed by physical segregation: whereas EGFR undergoes endocytosis, DEP-1 remains confined to the cell surface. CONCLUSIONS: EGFR and DEP-1 physically interact at the cell surface and maintain bidirectional enzyme-substrate interactions, which are relevant to their respective oncogenic and tumor-suppressive functions. These observations highlight the emerging roles of vesicular trafficking in malignant processes.


Subject(s)
Endocytosis/physiology , ErbB Receptors/metabolism , Endosomes/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , ErbB Receptors/analysis , HeLa Cells , Humans , Models, Biological , Phosphorylation , RNA Interference , Receptor-Like Protein Tyrosine Phosphatases, Class 3/analysis , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Cell Cycle ; 7(15): 2377-83, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18677116

ABSTRACT

Centrosomes control microtubule dynamics in many cell types, and their removal from the cytoplasm leads to a shift from dynamic instability to treadmilling behavior and to a dramatic decrease of microtubule mass (Rodionov et al., 1999; PNAS 96:115). In cadherin-expressing cells, these effects can be reversed:non-centrosomal cytoplasts that form cadherin-mediated adherens junctions display dense arrays of microtubules (Chausovsky et al., 2000; Nature Cell Biol 2:797). In adherens junctions, cadherin's cytoplasmic domain binds p120 catenin and beta-catenin, which in turn binds alpha-catenin. To elucidate the roles of the cadherin-associated proteins in regulating microtubule dynamics, we prepared GFP-tagged, plasma membrane targeted or untargeted p120 catenin, alpha-catenin and beta-catenin and tested their ability to rescue the loss of microtubule mass caused by centrosomal removal in the poorly adhesive cell line CHO-K1. Only membrane targeting of alpha-catenin led to a significant increase in microtubule length and density in centrosome-free cytoplasts. Expression of non-membrane-targeted alpha-catenin produced only a slight effect, while both membrane-targeted and non-targeted p120 and beta-catenin were ineffective in this assay. Together, these findings suggest that alpha-catenin is able to regulate microtubule dynamics in a centrosome-independent manner.


Subject(s)
Microtubules/metabolism , Signal Transduction/physiology , alpha Catenin/physiology , Animals , CHO Cells , Cadherins/genetics , Cadherins/metabolism , Cell Membrane/metabolism , Centrosome/physiology , Cricetinae , Cricetulus , Cytoplasm/ultrastructure , Microtubules/ultrastructure , Protein Transport , Transfection , alpha Catenin/genetics , alpha Catenin/metabolism
8.
Proc Natl Acad Sci U S A ; 104(26): 10882-7, 2007 Jun 26.
Article in English | MEDLINE | ID: mdl-17576929

ABSTRACT

The armadillo-family protein, p120 catenin (p120), binds to the juxtamembrane domain of classical cadherins and increases cell-cell junction stability. Overexpression of p120 modulates the activity of Rho family GTPases and augments cell migratory ability. Here we show that down-regulation of p120 in epithelial MCF-7 cells by siRNA leads to a striking decrease in lamellipodial persistence and focal adhesion formation. Similar alterations in lamellipodial activity were observed in MCF-7 cells treated with siRNA to cortactin, an activator of Arp2/3-dependent actin polymerization. We found that, in many cell types, p120 is colocalized with cortactin-containing actin structures not only at cell-cell junctions, but also at extrajunctional sites including membrane ruffles and actin-rich halos around endocytotic vesicles. p120 depletion led to dramatic loss of cortactin and its partner, Arp3, from the cell leading edges. Cortactin and p120 are shown to directly interact with each other via the cortactin N-terminal region. We propose that the mechanism underlying p120 functions at the leading edge involves its cooperation with cortactin.


Subject(s)
Actin-Related Protein 3/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/physiology , Cell Adhesion , Cortactin/metabolism , Phosphoproteins/metabolism , Phosphoproteins/physiology , Pseudopodia/metabolism , Catenins , Cell Line, Tumor , Epithelial Cells , Focal Adhesions , Humans , Protein Binding , Delta Catenin
9.
Bone ; 34(2): 246-60, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14962803

ABSTRACT

Microarray gene expression analysis was utilized to identify genes upregulated in primary rat calvaria cultures in response to mechanical force. One of the identified genes designated CMF608 appeared to be novel. The corresponding full-length cDNA was cloned and characterized in more details. It encodes a putative 2597 amino acid protein containing N-terminal signal peptide, six leucine-rich repeats (LRRs), and 12 immunoglobulin-like repeats, 10 of which are clustered within the C-terminus. Expression of CMF608 is bone-specific and the main type of CMF608-positive cells is mesenchymal osteochondroprogenitors with fibroblast-like morphology. These cells reside in the perichondral fibrous ring of La Croix, periosteum, endosteum of normal bone as well as in the activated periosteum and early fibrous callus generated postfracture. Expression of CMF608 is notably absent from the regions of endochondral ossification. Mature bone cell types do not produce CMF608 with the exception of chondrocytes of the tangential layer of the articular cartilage, which are thought to be under constant mechanical loading. Ectopic expression of CMF608 in HEK293T cells shows that the protein is subjected to post-translational processing and its N-terminal approximately 90 kDa polypeptide can be found in the conditioned medium. Ectopic expression of either the full-length cDNA of CMF608 or of its N-terminal region in CMF608-negative ROS17/2.8 rat osteosarcoma cells results in transfected clones displaying increased proliferation rate and the characteristics of less-differentiated osteoblasts compared to the control cells. Our data indicate that CMF608 is a unique marker of early osteochondroprogenitor cells. We propose that it could be functionally involved in maintenance of the osteochondroprogenitor cells pool and its down-regulation precedes terminal differentiation.


Subject(s)
Bone and Bones/physiology , Chondrocytes/physiology , Osteocytes/physiology , Protein Biosynthesis , Stem Cells/physiology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Cells, Cultured , Fractures, Bone/genetics , Humans , In Situ Hybridization , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Skull/physiology , Stress, Mechanical , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...