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










Publication year range
1.
Front Vet Sci ; 10: 1241215, 2023.
Article in English | MEDLINE | ID: mdl-37691637

ABSTRACT

Introduction: Gut dysbiosis has been noted in humans and animals with chronic kidney disease (CKD). However, little is known about the gut microbiome in canine patients with CKD. This study aimed to analyze and compare the gut microbiome profiles of healthy and CKD dogs, including differences in the gut microbiome between each CKD stage. Methods: The study was conducted on 29 client-owned dogs who underwent physical examination, complete blood count (CBC), serum biochemistry, and urinalysis. The gut microbiome profile of healthy dogs (n = 10) and dogs with CKD (n = 19) was analyzed employing 16S rRNA sequencing. Results: Significant differences were seen in the composition of the gut microbiome, with increased operational taxonomic units from the phylum Proteobacteria (p = 0.035), family Enterobacteriaceae (p < 0.001), and genus Enterococcus (p = 0.002) in dogs with CKD, and a decrease in the genus Ruminococcus (p = 0.007). Furthermore, an increase in both the progression of CKD and abundance of genus Klebsiella (Jonckheere-Terpstra test statistic value (JT) = 2.852, p = 0.004) and Clostridium (JT = 2.018, p = 0.044) was observed. Discussion: Our study demonstrated that in dogs with CKD, the composition of the gut microbiome varied depending on the stage of CKD. Alterations in gut microbiome composition observed in CKD patients are characterized by an increase in proteolytic bacteria and a decrease in saccharolytic bacteria. These findings suggest specific gut microbiota could be targeted for clinical management of uremic dogs with CKD.

2.
Biochem Pharmacol ; 151: 59-68, 2018 05.
Article in English | MEDLINE | ID: mdl-29522713

ABSTRACT

Exendin-4, a 39 amino acid peptide isolated from the saliva of the Gila monster, plays an important role in regulating glucose homeostasis, and is used clinically for the treatment of type 2 diabetes. Exendin-4 shares 53% sequence identity with the incretin hormone glucagon-like peptide 1 (GLP-1) but, unlike GLP-1, is highly resistant to proteolytic enzymes such as dipeptidyl peptidase IV (DPP-IV) and neutral endopeptidase 24.11 (NEP 24.11). Herein, we focused on the structure and function of the C-terminal Trp-cage of exendin-4, and suggest that it may be structurally required for resistance to proteolysis by NEP 24.11. Using a series of substitutions and truncations of the C-terminal Trp-cage, we found that residues 1-33, including the N-terminal and helical regions of wild-type (WT) exendin-4, is the minimum motif required for both high peptidase resistance and potent activity toward the GLP-1 receptor comparable to WT exendin-4. To improve the therapeutic utility of C-terminally truncated exendin-4, we incorporated various fatty acids into exendin-4(1-33) in which Ser33 was substituted with Lys for acylation. Exendin-4(1-32)K-capric acid exhibited the most well balanced activity, with much improved therapeutic utility for regulating blood glucose and body weight relative to WT exendin-4.


Subject(s)
Exenatide/chemistry , Exenatide/therapeutic use , Fatty Acids/chemistry , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Peptide Fragments/chemistry , Animals , Diabetes Mellitus, Experimental/drug therapy , Dipeptidyl Peptidase 4/chemistry , Drug Stability , Exenatide/blood , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide-1 Receptor/chemistry , Hypoglycemic Agents/blood , Male , Mice , Mice, Inbred C57BL , Neprilysin/chemistry , Peptide Hydrolases , Protein Conformation , Proteolysis
3.
Cancer Chemother Pharmacol ; 80(2): 363-369, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28660432

ABSTRACT

PURPOSE: AGM-130 is a cyclin-dependent kinase inhibitor that exhibits dose-dependent efficacy in xenograft mouse models. During preclinical pharmacokinetic (PK) studies, mice and rats showed comparable PK parameters while dogs showed unusually high clearance (CL), which has made human PK prediction challenging. To address this discrepancy, we performed a human microdosing PK and developed a mouse PK/PD model in order to guide the first-in-human studies. METHODS: A microdose of AGM-130 was given via intravenous injection to healthy subjects. Efficacy data obtained using MCF-7 breast cancer cells implanted in mice was analyzed using pre-existing tumor growth inhibition models. We simulated a human PK/PD profile with the PK parameters obtained from the microdose study and the PD parameters estimated from the xenograft PK/PD model. RESULTS: The human CL of AGM-130 was 3.08 L/h/kg, which was comparable to CL in mice and rats. The time-courses of tumor growth in xenograft model was well described by a preexisting model. Our simulation indicated that the human doses needed for 50 and 90% inhibition of tumor growth were about 100 and 400 mg, respectively. CONCLUSIONS: This is the first report of using microdose PK and xenograft PK/PD model to predict efficacious doses before the first-in-human trial in cancer patients. In addition, this work highlights the importance of integration of all of information in PK/PD analysis and illustrates how modeling and simulation can be used to add value in the early stages of drug development.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Indoles/administration & dosage , Models, Biological , Oximes/administration & dosage , Adult , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Female , Humans , Indoles/pharmacokinetics , Indoles/pharmacology , MCF-7 Cells , Male , Mice , Mice, Inbred ICR , Mice, Nude , Oximes/pharmacokinetics , Oximes/pharmacology , Species Specificity , Xenograft Model Antitumor Assays , Young Adult
4.
Nat Neurosci ; 19(8): 1010-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27294513

ABSTRACT

Neurotransmission requires precise control of neurotransmitter release from axon terminals. This process is regulated by glial cells; however, the underlying mechanisms are not fully understood. We found that glutamate release in the brain was impaired in mice lacking low-density lipoprotein receptor-related protein 4 (Lrp4), a protein that is critical for neuromuscular junction formation. Electrophysiological studies revealed compromised release probability in astrocyte-specific Lrp4 knockout mice. Lrp4 mutant astrocytes suppressed glutamatergic transmission by enhancing the release of ATP, whose level was elevated in the hippocampus of Lrp4 mutant mice. Consequently, the mutant mice were impaired in locomotor activity and spatial memory and were resistant to seizure induction. These impairments could be ameliorated by blocking the adenosine A1 receptor. The results reveal a critical role for Lrp4, in response to agrin, in modulating astrocytic ATP release and synaptic transmission. Our findings provide insight into the interaction between neurons and astrocytes for synaptic homeostasis and/or plasticity.


Subject(s)
Astrocytes/metabolism , Hippocampus/metabolism , Receptors, LDL/metabolism , Synaptic Transmission/physiology , Adenosine Triphosphate/metabolism , Agrin/genetics , Agrin/metabolism , Animals , LDL-Receptor Related Proteins , Mice, Knockout , Neuromuscular Junction/metabolism , Neuronal Plasticity/physiology , Presynaptic Terminals/metabolism , Receptors, Cholinergic/metabolism , Receptors, LDL/genetics
5.
J Neurosci ; 33(44): 17398-403, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24174672

ABSTRACT

Increased cranial pressure due to development of edema contributes significantly to the pathology of traumatic brain injury (TBI). Induction of an astrocytic water channel protein, Aquaporin 4 (AQP4), is known to predominantly contribute to cytotoxic edema following TBI. However, the mechanism for the increase in AQP4 following 24 h of TBI is poorly understood. Here we show that transcriptional activation of a ubiquitously expressed mammalian forkhead transcription factor, Foxo3a, induces cerebral edema by increasing the AQP4 level in the controlled cortical impact model of TBI in mice. TBI stimulates nuclear translocation of Foxo3a in astrocytes and subsequently augments its binding to AQP4 promoter in pericontusional cortex. Nuclear accumulation of Foxo3a is augmented by a decrease in phosphorylation at its Ser256 residue due to inactivation of Akt after TBI. Depletion of Foxo3a in mice rescues cytotoxic edema by preventing induction of AQP4 as well as attenuates memory impairment after TBI in mice.


Subject(s)
Aquaporin 4/biosynthesis , Brain Edema/etiology , Brain Injuries/metabolism , Forkhead Transcription Factors/physiology , Up-Regulation/physiology , Active Transport, Cell Nucleus/genetics , Animals , Aquaporin 4/genetics , Base Sequence , Brain Edema/genetics , Brain Edema/pathology , Brain Injuries/genetics , Brain Injuries/pathology , Cells, Cultured , Disease Models, Animal , Down-Regulation/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phosphorylation/genetics , Transcription, Genetic , Up-Regulation/genetics
6.
Cell Mol Life Sci ; 70(22): 4369-83, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23765104

ABSTRACT

Actin plays a fundamental role in the regulation of spine morphology (both shrinkage and enlargement) upon synaptic activation. In particular, actin depolymerization is crucial for the spine shrinkage in NMDAR-mediated synaptic depression. Here, we define the role of SPIN90 phosphorylation/dephosphorylation in regulating actin depolymerization via modulation of cofilin activity. When neurons were treated with NMDA, SPIN90 was dephosphorylated by STEP61 (striatal-enriched protein tyrosine phosphatase) and translocated from the spines to the dendritic shafts. In addition, phosphorylated SPIN90 bound cofilin and then inhibited cofilin activity, suggesting that SPIN90 dephosphorylation is a prerequisite step for releasing cofilin so that cofilin can adequately sever actin filaments into monomeric form. We found that SPIN90 YE, a phosphomimetic mutant, remained in the spines after NMDAR activation where it bound cofilin, thereby effectively preventing actin depolymerization. This led to inhibition of the activity-dependent redistribution of cortactin and drebrin A, as well as of the morphological changes in the spines that underlie synaptic plasticity. These findings indicate that NMDA-induced SPIN90 dephosphorylation and translocation initiates cofilin-mediated actin dynamics and spine shrinkage within dendritic spines, thereby modulating synaptic activity.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cofilin 1/metabolism , Hippocampus/metabolism , Muscle Proteins/metabolism , N-Methylaspartate/pharmacology , Neurons/drug effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Dendritic Spines/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Muscle Proteins/genetics , Mutation , Neurons/metabolism , Phosphorylation/drug effects , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Rats , Transfection
7.
PLoS One ; 7(4): e34677, 2012.
Article in English | MEDLINE | ID: mdl-22485184

ABSTRACT

BACKGROUND: Dendritic spines are small membranous protrusions on the neuronal dendrites that receive synaptic input from axon terminals. Despite their importance for integrating the enormous information flow in the brain, the molecular mechanisms regulating spine morphogenesis are not well understood. NESH/Abi-3 is a member of the Abl interactor (Abi) protein family, and its overexpression is known to reduce cell motility and tumor metastasis. NESH is prominently expressed in the brain, but its function there remains unknown. METHODOLOGY/PRINCIPAL FINDINGS: NESH was strongly expressed in the hippocampus and moderately expressed in the cerebral cortex, cerebellum and striatum, where it co-localized with the postsynaptic proteins PSD95, SPIN90 and F-actin in dendritic spines. Overexpression of NESH reduced numbers of mushroom-type spines and synapse density but increased thin, filopodia-like spines and had no effect on spine density. siRNA knockdown of NESH also reduced mushroom spine numbers and inhibited synapse formation but it increased spine density. The N-terminal region of NESH co-sedimented with filamentous actin (F-actin), which is an essential component of dendritic spines, suggesting this interaction is important for the maturation of dendritic spines. CONCLUSIONS/SIGNIFICANCE: NESH is a novel F-actin binding protein that likely plays important roles in the regulation of dendritic spine morphogenesis and synapse formation.


Subject(s)
Dendritic Spines/metabolism , Microfilament Proteins/physiology , Nerve Tissue Proteins/physiology , Synapses/physiology , Actin Cytoskeleton/metabolism , Animals , Cell Shape , Gene Expression , Gene Knockdown Techniques , HEK293 Cells , Hippocampus/cytology , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Protein Binding , Protein Interaction Domains and Motifs , RNA Interference , Rats , Rats, Sprague-Dawley , Synapses/metabolism
8.
Exp Cell Res ; 317(16): 2276-87, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21763308

ABSTRACT

SPIN90 is an F-actin binding protein thought to play important roles in regulating cytoskeletal dynamics. It is known that SPIN90 is expressed during the early stages of neuronal development, but details of its localization and function in growth cones have not been fully investigated. Our immunocytochemical data show that SPIN90 is enriched throughout growth cones and neuronal shafts in young hippocampal neurons. We also found that its localization correlates with and depends upon the presence of F-actin. Detailed observation of primary cultures of hippocampal neurons revealed that SPIN90 knockout reduces both growth cone areas and in the numbers of filopodia, as compared to wild-type neurons. In addition, total neurite length, the combined lengths of the longest (axonal) and shorter (dendritic) neurites, was smaller in SPIN90 knockout neurons than wild-type neurons. Finally, Cdc42 activity was down-regulated in SPIN90 knockout neurons. Taken together, our findings suggest that SPIN90 plays critical roles in controlling growth cone dynamics and neurite outgrowth.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Growth Cones/physiology , Nerve Tissue Proteins/metabolism , Neurites/physiology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Differentiation/physiology , Cells, Cultured , Cytochalasin D/pharmacology , Embryo, Mammalian/cytology , Female , Growth Cones/pathology , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubules/metabolism , Nerve Tissue Proteins/genetics , Neurites/pathology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Pseudopodia/metabolism , Pseudopodia/pathology , Rats , Rats, Inbred Strains , Thiazolidines/pharmacology , Tubulin/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , cdc42 GTP-Binding Protein/metabolism
9.
Exp Cell Res ; 315(14): 2410-9, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19460367

ABSTRACT

SPIN90 is a key regulator of actin cytoskeletal organization. Using the BioGRID(beta) database (General Repository for Interaction Datasets), we identified IRSp53 as a binding partner of SPIN90, and confirmed the in vivo formation of a SPIN90-IRSp53 complex mediated through direct association of the proline-rich domain (PRD) of SPIN90 with the SH3 domain of IRSp53. SPIN90 and IRSp53 positively cooperated to mediate Rac activation, and co-expression of SPIN90 and IRSp53 in COS-7 cells led to the complex formation of SPIN90-IRSp53 in the leading edge of cells. PDGF treatment induced strong colocalization of SPIN90 and IRSp53 at membrane protrusions. Within such PDGF-induced protrusions, knockdown of SPIN90 protein using siRNA significantly reduced lamellipodia-like protrusions as well as localization of IRSp53 at those sites. Finally, competitive inhibition of SPIN90-IRSp53 binding by SPIN90 PRD dramatically reduced ruffle formation, further suggesting that SPIN90 plays a key role in the formation of the membrane protrusions associated with cell motility.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/metabolism , Muscle Proteins/metabolism , Nerve Tissue Proteins/metabolism , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , COS Cells , Cell Membrane/ultrastructure , Cell Movement/physiology , Cell Surface Extensions , Chlorocebus aethiops , Gene Knockdown Techniques , Genetic Vectors/metabolism , Humans , Muscle Proteins/genetics , Platelet-Derived Growth Factor/pharmacology , RNA, Small Interfering/metabolism , Transfection
10.
J Neurochem ; 109(4): 1106-17, 2009 May.
Article in English | MEDLINE | ID: mdl-19302483

ABSTRACT

Dendritic spines are highly specialized actin-rich structures on which the majority of excitatory synapses are formed in the mammalian CNS. SPIN90 is an actin-binding protein known to be highly enriched in postsynaptic densities (PSDs), though little is known about its function there. Here, we show that SPIN90 is a novel binding partner for Shank proteins in the PSD. SPIN90 and Shank co-immunoprecipitate from brain lysates and co-localize in postsynaptic dendrites and act synergistically to mediate spine maturation and spine head enlargement. At the same time, SPIN90 causes accumulation of Shank and PSD-95 within dendritic spines. In addition, we found that the protein composition of PSDs in SPIN90 knockout mice is altered as is the actin cytoskeleton of cultured hippocampal SPIN90 knockout neurons. Taken together, these findings demonstrate that SPIN90 is a Shank1b binding partner and a key contributor to the regulation of dendritic spine morphogenesis and brain function.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Membrane Proteins/physiology , Nerve Tissue Proteins/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites , Blotting, Western , Brain Chemistry/physiology , Cell Line, Tumor , Cells, Cultured , Cytoskeleton/chemistry , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Glutathione Transferase/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Plasmids/genetics , Protein Binding , Synapses/physiology , Transfection
11.
Cell Commun Adhes ; 14(1): 33-43, 2007.
Article in English | MEDLINE | ID: mdl-17453829

ABSTRACT

We recently reported that SPIN90 is able to bind with several proteins involved in regulating actin cytoskeleton networks, including dynamin, WASP, beta PIX, and Nck. Based on these findings, we investigated how SPIN90 regulates the actin cytoskeleton and promotes actin assembly. This study demonstrated that aluminium fluoride-induced localization of SPIN90 to lamellipodia requires amino acids 582-722 at the SPIN90 C-terminus, which is also essential for F-actin binding and Arp2/3 complex mediated polymerization of actin into branched actin filaments. Furthermore, after deletion of the F-actin binding region (582-722 SPIN90) failed to localize at the membrane edge and was unable to promote lamellipodia formation, suggesting that the F-actin binding region in the SPIN90 C-terminus is essential for the formation of branched actin networks and regulation of the actin cytoskeleton at the leading edge of cells.


Subject(s)
Actins/chemistry , Actins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Pseudopodia/metabolism , Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Aluminum Compounds/pharmacology , Cell Line, Tumor , Fluorides/pharmacology , Humans , Models, Biological , Protein Binding/drug effects , Protein Transport/drug effects , Pseudopodia/chemistry , Pseudopodia/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
12.
Genes Cells ; 11(10): 1197-211, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16999739

ABSTRACT

SPIN90, a 90-kDa Nck-interacting protein with a SH3 domain, plays a role in sarcomere formation and myofibril assembly, and its phosphorylation is modulated by cell adhesion and Erk activation. Here we demonstrate that SPIN90 participates in receptor-mediated endocytic pathway in fibroblasts. We identified syndapin (synaptic dynamin-binding protein) as a SPIN90 interacting protein using yeast two-hybrid screening. SPIN90 directly binds the SH3 domain of syndapin via its proline rich domain in vitro and in vivo and also associates with clathrin. Over-expression of SPIN90-full length in COS-7 cells inhibited transferrin uptake, a marker of endocytosis. Interestingly, SPIN90-PRD, a syndapin-binding domain, significantly inhibited endocytosis, and the inhibition was reversed by co-expression of syndapin. Depleting SPIN90 through antibody microinjection or Knocking it down using siRNAs also significantly inhibited transferrin internalization. Moreover, early endosomal marker proteins (EEA1 and Rab5) appeared to closely associate or partially co-localize with SPIN90 in endosomes and an internalized FITC-dextran and Texas Red-EGF were found on the endosomes in association with SPIN90. Time-lapse video showed that GFP-SPIN90 travels with moving vesicles within living cells. Taken together, these findings suggest that SPIN90 is implicated in receptor-mediated endocytic pathway in fibroblasts.


Subject(s)
Carrier Proteins/metabolism , Clathrin/metabolism , Endocytosis/physiology , Animals , Base Sequence , COS Cells , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Chlorocebus aethiops , Cytoskeletal Proteins , DNA, Complementary/genetics , Endosomes/metabolism , Fibroblasts/metabolism , In Vitro Techniques , Protein Binding , RNA, Small Interfering/genetics , Rats , Two-Hybrid System Techniques , src Homology Domains
13.
J Biol Chem ; 279(8): 7082-90, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14660602

ABSTRACT

By using transient elevations of cytosolic free calcium levels triggered by integrin antibody or laminin (Kwon, M. S., Park, C. S., Choi, K., Park, C.-S., Ahnn, J., Kim, J. I., Eom, S. H., Kaufman, S. J., and Song, W. K. (2000) Mol. Biol. Cell 11, 1433-1443), we have demonstrated that protein phosphatase 2A (PP2A) is implicated in the regulation of reversible phosphorylation of integrin. In E63 skeletal myoblasts, the treatment of PP2A inhibitors such as okadaic acid and endothall induces an increase of phosphorylation of integrin beta1A and thereby inhibits integrin-induced elevation of cytosolic calcium level and formation of focal adhesions. None of these effects were in differentiated myotubes expressing the alternate beta1D isoform. In the presence of okadaic acid, PP2A in association with integrin beta1A was reduced on myoblasts, whereas beta1D on myotubes remained bound with PP2A. Both co-immunoprecipitation and in vitro phosphatase assays revealed that dephosphorylation of residues Thr788-Thr789 in the integrin beta1A cytoplasmic domain is dependent upon PP2A activity. Mutational analysis of the cytoplasmic domain and confocal microscopy experiments indicated that substitution of Thr788-Thr789 with Asn788-Asn789 is of critical importance for regulating the function of integrin beta1. These results suggest that PP2A may be a primary regulator of threonine phosphorylation of integrin beta1A and subsequent activation of downstream signaling molecules. Taken together, we propose that dephosphorylation of residues Thr788-Thr789 in the cytoplasmic domain of integrin beta1A may contribute to the linkage of integrins to focal adhesion sites and induce the association with cytoskeleton proteins. The switch of integrin beta1A to beta1D isoform in myotubes therefore may be a mechanism to escape from phospho-regulation by PP2A and promotes a more stable association of the cytoskeleton with the extracellular matrix.


Subject(s)
Integrin beta1/metabolism , Muscles/metabolism , Threonine/chemistry , Amino Acid Sequence , Animals , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Cytoplasm/metabolism , Cytoskeleton/metabolism , Cytosol/metabolism , Extracellular Matrix/metabolism , Focal Adhesions , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Mutation , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/chemistry , Phosphorylation , Precipitin Tests , Protein Binding , Protein Isoforms , Protein Phosphatase 2 , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...