Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Mol Biol Cell ; 32(4): 314-330, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33378226

ABSTRACT

TRIM9 and TRIM67 are neuronally enriched E3 ubiquitin ligases essential for appropriate morphogenesis of cortical and hippocampal neurons and fidelitous responses to the axon guidance cue netrin-1. Deletion of murine Trim9 or Trim67 results in neuroanatomical defects and striking behavioral deficits, particularly in spatial learning and memory. TRIM9 and TRIM67 interact with cytoskeletal and exocytic proteins, but the full interactome is not known. Here we performed the unbiased proximity-dependent biotin identification (BioID) approach to define TRIM9 and TRIM67 protein-protein proximity network in developing cortical neurons and identified putative neuronal TRIM interaction partners. Candidates included cytoskeletal regulators, cytosolic protein transporters, exocytosis and endocytosis regulators, and proteins necessary for synaptic regulation. A subset of high-priority candidates was validated, including Myo16, Coro1A, MAP1B, ExoC1, GRIP1, PRG-1, and KIF1A. For a subset of validated candidates, we utilized total internal reflection fluorescence microscopy to demonstrate dynamic colocalization with TRIM proteins at the axonal periphery, including at the tips of filopodia. Further analysis demonstrated that the RNA interference-based knockdown of the unconventional myosin Myo16 in cortical neurons altered growth cone filopodia density and axonal branching patterns in a TRIM9- and netrin-1-dependent manner. Future analysis of other validated candidates will likely identify novel proteins and mechanisms by which TRIM9 and TRIM67 regulate neuronal form and function. [Media: see text].


Subject(s)
Cytoskeletal Proteins/metabolism , Morphogenesis/physiology , Nerve Tissue Proteins/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Axons/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Female , Growth Cones/metabolism , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neurons/metabolism , Protein Interaction Mapping/methods , Protein Interaction Maps , Pseudopodia/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/physiology
2.
Sci Rep ; 10(1): 16947, 2020 10 12.
Article in English | MEDLINE | ID: mdl-33046791

ABSTRACT

Protein 4.1N, a member of the protein 4.1 family, is highly expressed in the brain. But its function remains to be fully defined. Using 4.1N-/- mice, we explored the function of 4.1N in vivo. We show that 4.1N-/- mice were born at a significantly reduced Mendelian ratio and exhibited high mortality between 3 to 5 weeks of age. Live 4.1N-/- mice were smaller than 4.1N+/+ mice. Notably, while there were no significant differences in organ/body weight ratio for most of the organs, the testis/body and ovary/body ratio were dramatically decreased in 4.1N-/- mice, demonstrating selective effects of 4.1N deficiency on the development of the reproductive systems. Histopathology of the reproductive organs showed atrophy of both testis and ovary. Specifically, in the testis there is a lack of spermatogenesis, lack of leydig cells and lack of mature sperm. Similarly, in the ovary there is a lack of follicular development and lack of corpora lutea formation, as well as lack of secretory changes in the endometrium. Examination of pituitary glands revealed that the secretory granules were significantly decreased in pituitary glands of 4.1N-/- compared to 4.1N+/+. Moreover, while GnRH was expressed in both neuronal cell body and axons in the hypothalamus of 4.1N+/+ mice, it was only expressed in the cell body but not the axons of 4.1N-/- mice. Our findings uncover a novel role for 4.1N in the axis of hypothalamus-pituitary gland-reproductive system.


Subject(s)
Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/physiology , Genitalia/metabolism , Genitalia/pathology , Membrane Proteins/deficiency , Membrane Proteins/physiology , Neuropeptides/deficiency , Neuropeptides/physiology , Neurosecretory Systems/metabolism , Neurosecretory Systems/pathology , Animals , Cytoskeletal Proteins/genetics , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Membrane Proteins/genetics , Mice, Knockout , Neuropeptides/genetics , Organ Size , Ovary/pathology , Pituitary Gland/metabolism , Pituitary Gland/pathology , Spermatogenesis/genetics , Testis/pathology
3.
Sci Rep ; 10(1): 15472, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32963272

ABSTRACT

Neuronal injury leads to rapid, programmed disintegration of axons distal to the site of lesion. Much like other forms of axon degeneration (e.g. developmental pruning, toxic insult from neurodegenerative disorder), Wallerian degeneration associated with injury is preceded by spheroid formation along axons. The mechanisms by which injury leads to formation of spheroids and whether these spheroids have a functional role in degeneration remain elusive. Here, using neonatal mouse primary sympathetic neurons, we investigate the roles of players previously implicated in the progression of Wallerian degeneration in injury-induced spheroid formation. We find that intra-axonal calcium flux is accompanied by actin-Rho dependent growth of calcium rich axonal spheroids that eventually rupture, releasing material to the extracellular space prior to catastrophic axon degeneration. Importantly, after injury, Sarm1-/- and DR6-/-, but not Wlds (excess NAD+) neurons, are capable of forming spheroids that eventually rupture, releasing their contents to the extracellular space to promote degeneration. Supplementation of exogenous NAD+ or expressing WLDs suppresses Rho-dependent spheroid formation and degeneration in response to injury. Moreover, injured or trophically deprived Sarm1-/- and DR6-/-, but not Wlds neurons, are resistant to degeneration induced by conditioned media collected from wild-type axons after spheroid rupture. Taken together, these findings place Rho-actin and NAD+ upstream of spheroid formation and may suggest that other mediators of degeneration, such as DR6 and SARM1, mediate post-spheroid rupture events that lead to catastrophic axon disassembly.


Subject(s)
Armadillo Domain Proteins/physiology , Cytoskeletal Proteins/physiology , Nerve Tissue Proteins/physiology , Neurodegenerative Diseases/pathology , Neurons/pathology , Receptors, Tumor Necrosis Factor/physiology , Spheroids, Cellular/pathology , Wallerian Degeneration/physiopathology , Animals , Axons/pathology , Axotomy , Calcium/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurons/metabolism
4.
Neuron ; 92(1): 126-142, 2016 Oct 05.
Article in English | MEDLINE | ID: mdl-27641493

ABSTRACT

Precise connection of thalamic barreloids with their corresponding cortical barrels is critical for processing of vibrissal sensory information. Here, we show that PRG-2, a phospholipid-interacting molecule, is important for thalamocortical axon guidance. Developing thalamocortical fibers both in PRG-2 full knockout (KO) and in thalamus-specific KO mice prematurely entered the cortical plate, eventually innervating non-corresponding barrels. This misrouting relied on lost axonal sensitivity toward lysophosphatidic acid (LPA), which failed to repel PRG-2-deficient thalamocortical fibers. PRG-2 electroporation in the PRG-2-/- thalamus restored the aberrant cortical innervation. We identified radixin as a PRG-2 interaction partner and showed that radixin accumulation in growth cones and its LPA-dependent phosphorylation depend on its binding to specific regions within the C-terminal region of PRG-2. In vivo recordings and whisker-specific behavioral tests demonstrated sensory discrimination deficits in PRG-2-/- animals. Our data show that bioactive phospholipids and PRG-2 are critical for guiding thalamic axons to their proper cortical targets.


Subject(s)
Axon Guidance/physiology , Cerebral Cortex/growth & development , Cytoskeletal Proteins/physiology , Lysophospholipids/physiology , Membrane Proteins/physiology , Signal Transduction/physiology , Thalamus/growth & development , Animals , Cerebral Cortex/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Discrimination, Psychological/physiology , Growth Cones/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Neural Pathways/metabolism , Neural Pathways/physiology , Phosphorylation , Thalamus/metabolism
5.
Cell Biochem Biophys ; 62(1): 193-201, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21913004

ABSTRACT

We previously isolated a sulfated polysaccharide-protein complex from Gekko swinhonis Guenther, a traditional Chinese medicine, and have demonstrated its direct anti-cancer effect on human hepatocellular carcinoma cell line SMMC-7721. Here we investigated the effects of Gekko sulfated polysaccharide-protein complex (GSPP) on the defective biorheological characters of dendritic cells (DCs) under SMMC-7721 microenvironment. Our findings have shown that the biorheological properties of DCs were severely impaired by SMMC-7721 microenvironment, including decreased cell deformability, migration, and electrophoresis mobility, increased osmotic fragilities, and changed organizations of cytoskeletal proteins. We also found decreased secretion of interleukin (IL)-12 and increased secretion of IL-10 in DCs. However, supernatant collected from nonmalignant liver cells had no effect on these parameters. SMMC-7721 cells were treated with GSPP and the supernatant was used to culture DCs. We found that the defective biorheological parameters of DCs, except for osmotic fragility, were partially or completely improved. The secretion of IL-12 did not change as compared with that of DCs in SMMC-7721 microenvironment, but the secretion of IL-10 was resumed to the control level. Our results indicate that GSPP could partially restore the defective biorheological characteristics of DCs via modifying the tumor microenvironment and decreasing the secretion of IL-10 of DCs.


Subject(s)
Antineoplastic Agents/pharmacology , Dendritic Cells/drug effects , Polysaccharides/pharmacology , Tumor Microenvironment , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Cell Shape , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/physiology , Dendritic Cells/cytology , Humans , Interleukin-10/metabolism , Interleukin-12/metabolism , Liver Neoplasms/metabolism , Medicine, Chinese Traditional
6.
Exp Dermatol ; 20(8): 653-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21569101

ABSTRACT

Photodynamic therapy was found to be an effective therapy for local malignant tumors. This study demonstrated that 80 µg/ml Hedyotis corymbosa extracts with 0.8 J/cm(2) fluence dose caused M21 skin cancer cell death. Photoactivated H. corymbosa-induced M21 cell death is a typical apoptosis that is accompanied by nuclear condensation, externalization of phosphatidylserine and the changes in protein expression of apoptosis-related proteins, such as Bcl-2 and caspase family members. This study applied 2D electrophoresis to analyse the proteins involved in the photoactivated H. corymbosa-induced M21 cell apoptosis. We found 12 proteins to be markedly changed. According to the results of protein sequence analysis of these altered protein spots, we identified that the expression of cytoskeletal proteins and chaperones were involved in the photoactivated H. corymbosa-induced M21 cell apoptosis. We further demonstrated that photoactivated H. corymbosa caused a significant effect on the cytoskeleton distribution and mitochondrial activity in M21 cells. Based on the above findings, this study characterized the effects and mechanisms of the photoactivated H. corymbosa-induced apoptosis in M21 skin cancer cells.


Subject(s)
Cytoskeletal Proteins/physiology , Drugs, Chinese Herbal/therapeutic use , Hedyotis , Melanoma/drug therapy , Molecular Chaperones/physiology , Photochemotherapy , Proteomics , Skin Neoplasms/drug therapy , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/physiology , Apoptosis/drug effects , Apoptosis/physiology , Caspases/physiology , Cell Line, Tumor , Cytochromes c/physiology , Humans , Melanoma/pathology , Melanoma/physiopathology , Mitochondria/drug effects , Mitochondria/physiology , Phalloidine/pharmacology , Proto-Oncogene Proteins c-bcl-2/physiology , Skin Neoplasms/pathology , Skin Neoplasms/physiopathology , bcl-2-Associated X Protein/physiology
7.
Proc Natl Acad Sci U S A ; 107(33): 14828-32, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20675582

ABSTRACT

Models of learning-dependent sensory cortex plasticity require local activity and reinforcement. An alternative proposes that neural activity involved in anticipation of a sensory stimulus, or the preparatory set, can direct plasticity so that changes could occur in regions of sensory cortex lacking activity. To test the necessity of target-induced activity for initial sensory learning, we trained rats to detect a low-frequency sound. After learning, Arc expression and physiologically measured neuroplasticity were strong in a high-frequency auditory cortex region with very weak target-induced activity in control animals. After 14 sessions, Arc and neuroplasticity were aligned with target-induced activity. The temporal and topographic correspondence between Arc and neuroplasticity suggests Arc may be intrinsic to the neuroplasticity underlying perceptual learning. Furthermore, not all neuroplasticity could be explained by activity-dependent models but can be explained if the neural activity involved in the preparatory set directs plasticity.


Subject(s)
Auditory Cortex/physiology , Cytoskeletal Proteins/physiology , Learning/physiology , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Acoustic Stimulation , Analysis of Variance , Animals , Auditory Cortex/metabolism , Auditory Perception/physiology , Brain Mapping , Cytoskeletal Proteins/genetics , Evoked Potentials, Auditory/physiology , Gene Expression , In Situ Hybridization, Fluorescence , Male , Nerve Tissue Proteins/genetics , Rats , Rats, Sprague-Dawley , Reaction Time/physiology , Time Factors
8.
J Immunol ; 183(6): 4003-12, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717512

ABSTRACT

Because IL-1beta plays an important role in inflammation in human and murine arthritis, we investigated the contribution of the inflammasome components ASC, NALP-3, IPAF, and caspase-1 to inflammatory arthritis. We first studied the phenotype of ASC-deficient and wild-type mice during Ag-induced arthritis (AIA). ASC(-/-) mice showed reduced severity of AIA, decreased levels of synovial IL-1beta, and diminished serum amyloid A levels. In contrast, mice deficient in NALP-3, IPAF, or caspase-1 did not show any alteration of joint inflammation, thus indicating that ASC associated effects on AIA are independent of the classical NALP-3 or IPAF inflammasomes. Because ASC is a ubiquitous cytoplasmic protein that has been implicated in multiple cellular processes, we explored other pathways through which ASC may modulate inflammation. Ag-specific proliferation of lymph node and spleen cells from ASC-deficient mice was significantly decreased in vitro, as was the production of IFN-gamma, whereas IL-10 production was enhanced. TCR ligation by anti-CD3 Abs in the presence or absence of anti-CD28 Abs induced a reduction in T cell proliferation in ASC(-/-) T cells compared with wild-type ones. In vivo lymph node cell proliferation was also significantly decreased in ASC(-/-) mice, but no effects on apoptosis were observed either in vitro or in vivo in these mice. In conclusion, these results strongly suggest that ASC modulates joint inflammation in AIA through its effects on cell-mediated immune responses but not via its implication in inflammasome formation.


Subject(s)
Apoptosis Regulatory Proteins/deficiency , Arthritis, Experimental/etiology , Calcium-Binding Proteins/deficiency , Carrier Proteins/genetics , Caspase 1/deficiency , Cytoskeletal Proteins/physiology , Inflammation/etiology , Animals , Antigens/toxicity , Arthritis, Experimental/pathology , CARD Signaling Adaptor Proteins , Cell Proliferation , Joint Diseases/pathology , Lymph Nodes/pathology , Mice , Mice, Knockout , Multiprotein Complexes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Spleen/pathology
9.
Brain Res ; 1228: 97-106, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18634768

ABSTRACT

beta-Adducin is a cytoskeletal protein that interacts with the actin filaments to suppress actin polymerization and facilitate actin-spectrin binding. We have previously shown that beta-adducin is phosphorylated by Fyn at tyrosine489 in the rat brain and bound to its Src-homology 2 domain. In the present study, we examined the immunohistochemical localization of the tyrosine489-phosphorylated form of beta-adducin (pY489-beta-adducin) in the rat brain. Among brain regions, highest immunoreactivity was located in the hypothalamic tanycytes that are of glial origin lining around the third cerebral ventricle. Their immunoreactive processes extended into the arcuate nucleus, ventromedial hypothalamus and the median eminence. In addition, the pY489-beta-adducin immunoreactivity in the tanycytes was enhanced after fasting for 36-48 h, being associated with a morphological change of the DARPP-32-immunoreactivity. Intraperitoneal injection of 2-deoxy-d-glucose also enhances pY489-beta-adducin immunoreactivity in the tanycytes, along with increased food intake. These results suggest that tyrosine phosphorylation of beta-adducin in the tanycytes is involved in hypothalamic regulation of food intake and energy homeostasis.


Subject(s)
Calmodulin-Binding Proteins/physiology , Cytoskeletal Proteins/physiology , Energy Metabolism/physiology , Hypothalamus/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Calmodulin-Binding Proteins/metabolism , Cerebral Ventricles/metabolism , Cytoskeletal Proteins/metabolism , Deoxyglucose/administration & dosage , Deoxyglucose/pharmacology , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Eating/drug effects , Eating/physiology , Fasting , Homeostasis/drug effects , Hypothalamus/cytology , Hypothalamus/drug effects , Immunoblotting , Immunohistochemistry , Injections, Intraperitoneal , Injections, Intraventricular , Male , Median Eminence/metabolism , Phosphoproteins/metabolism , Phosphoproteins/physiology , Phosphorylation , Rats , Rats, Wistar , Tyrosine/metabolism
10.
J Cell Biol ; 179(4): 733-46, 2007 Nov 19.
Article in English | MEDLINE | ID: mdl-18025306

ABSTRACT

Immunological synapse (IS) formation involves receptor-ligand pair clustering and intracellular signaling molecule recruitment with a coincident removal of other membrane proteins away from the IS. As microfilament-membrane linkage is critical to this process, we investigated the involvement of ezrin and moesin, the two ezrin/radixin/moesin proteins expressed in T cells. We demonstrate that ezrin and moesin, which are generally believed to be functionally redundant, are differentially localized and have important and complementary functions in IS formation. Specifically, we find that ezrin directly interacts with and recruits the signaling kinase ZAP-70 to the IS. Furthermore, the activation of ezrin by phosphorylation is essential for this process. In contrast, moesin dephosphorylation and removal, along with CD43, are necessary to prepare a region of the cell cortex for IS. Thus, ezrin and moesin have distinct and critical functions in the T cell cortex during IS formation.


Subject(s)
Cytoskeletal Proteins/physiology , Leukosialin/physiology , Microfilament Proteins/physiology , ZAP-70 Protein-Tyrosine Kinase/physiology , Animals , Calcium/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Humans , Jurkat Cells , Leukosialin/metabolism , Lymphocyte Activation/immunology , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation , Phosphorylation , Precipitin Tests , RNA, Small Interfering/metabolism , Recombinant Proteins/metabolism , Spodoptera/cytology , Spodoptera/metabolism , T-Lymphocytes/physiology , Transfection , ZAP-70 Protein-Tyrosine Kinase/metabolism
11.
J Biol Chem ; 280(48): 40012-24, 2005 Dec 02.
Article in English | MEDLINE | ID: mdl-16204241

ABSTRACT

The tumor necrosis factor family member Fas ligand (FasL) induces apoptosis in Fas receptor-expressing target cells and is an important cytotoxic effector molecule used by CTL- and NK-cells. In these hematopoietic cells, newly synthesized FasL is stored in specialized secretory lysosomes and only delivered to the cell surface upon activation and target cell recognition. FasL contains an 80-amino acid-long cytoplasmic tail, which includes a proline-rich domain as a bona fide Src homology 3 domain-binding site. This proline-rich domain has been implicated in FasL sorting to secretory lysosomes, and it may also be important for reverse signaling via FasL, which has been described to influence T-cell activation. Here we report the identification of the Src homology 3 domain-containing adaptor protein PSTPIP as a FasL-interacting partner, which binds to the proline-rich domain. PSTPIP co-expression leads to an increased intracellular localization of Fas ligand, thereby regulating extracellular availability and cytotoxic activity of the molecule. In addition, we demonstrate recruitment of the tyrosine phosphatase PTP-PEST by PSTPIP into FasL.PSTPIP.PTP-PEST complexes which may contribute to FasL reverse signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cytoskeletal Proteins/physiology , Membrane Glycoproteins/metabolism , Tumor Necrosis Factors/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Animals , Apoptosis , Binding Sites , COS Cells , Cell Line , Cell Line, Tumor , Cell Separation , Chlorocebus aethiops , Coculture Techniques , Cytoplasm/metabolism , Cytoskeletal Proteins/chemistry , DNA, Complementary/metabolism , Fas Ligand Protein , Flow Cytometry , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoprecipitation , Lysosomes/chemistry , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Models, Biological , Plasmids/metabolism , Proline/chemistry , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 12 , Protein Tyrosine Phosphatases/chemistry , Rats , Signal Transduction , Spleen/cytology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transfection , Two-Hybrid System Techniques , Tyrosine/chemistry , src Homology Domains
12.
Circ Res ; 96(5): e25-34, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15731462

ABSTRACT

I(Ks), the slowly activating component of the delayed rectifier current, plays a major role in repolarization of the cardiac action potential (AP). Genetic mutations in the alpha- (KCNQ1) and beta- (KCNE1) subunits of I(Ks) underlie Long QT Syndrome type 1 and 5 (LQT-1 and LQT-5), respectively, and predispose carriers to the development of polymorphic ventricular arrhythmias and sudden cardiac death. beta-adrenergic stimulation increases I(Ks) and results in rate dependent AP shortening, a control system that can be disrupted by some mutations linked to LQT-1 and LQT-5. The mechanisms by which I(Ks) regulates action potential duration (APD) during beta-adrenergic stimulation at different heart rates are not known, nor are the consequences of mutation induced disruption of this regulation. Here we develop a complementary experimental and theoretical approach to address these questions. We reconstituted I(Ks) in CHO cells (ie, KCNQ1 coexpressed with KCNE1 and the adaptator protein Yotiao) and quantitatively examined the effects of beta-adrenergic stimulation on channel kinetics. We then developed theoretical models of I(Ks) in the absence and presence of beta-adrenergic stimulation. We simulated the effects of sympathetic stimulation on channel activation (speeding) and deactivation (slowing) kinetics on the whole cell action potential under different pacing conditions. The model suggests these kinetic effects are critically important in rate-dependent control of action potential duration. We also investigate the effects of two LQT-5 mutations that alter kinetics and impair sympathetic stimulation of I(Ks) and show the likely mechanism by which they lead to tachyarrhythmias and indicate a distinct role of I(KS) kinetics in this electrical dysfunction. The full text of this article is available online at http://circres.ahajournals.org.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cytoskeletal Proteins/physiology , Myocytes, Cardiac/physiology , Potassium Channels, Voltage-Gated/physiology , Sympathetic Nervous System/physiology , A Kinase Anchor Proteins , Action Potentials/physiology , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Substitution , Animals , CHO Cells , Computer Simulation , Cricetinae , Cricetulus , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeletal Proteins/genetics , Delayed Rectifier Potassium Channels , Humans , Ion Channel Gating/physiology , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Kinetics , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Models, Cardiovascular , Mutation, Missense , Patch-Clamp Techniques , Phosphorylation , Point Mutation , Potassium/metabolism , Potassium Channels, Voltage-Gated/genetics , Protein Processing, Post-Translational , Receptors, Adrenergic, beta/physiology , Recombinant Fusion Proteins/physiology , Second Messenger Systems/physiology , Tachycardia/physiopathology , Transfection
13.
J Neurosci ; 24(35): 7632-9, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15342729

ABSTRACT

Mice with mutations in the Wnt coreceptor low-density lipoprotein receptor-related protein-6 (LRP6) have a smaller and severely disorganized dorsal thalamus and lack thalamocortical projections. Using molecular markers, we showed that most dorsal thalamic and epithalamic neurons were missing, and most of the major dorsal thalamic nuclei were not identifiable. However, the ventral thalamus was essentially unaffected, although the dorsal thalamic defect leads to rostral displacement of portions of the ventral thalamus. Analysis of younger embryos showed that epithalamic and dorsal thalamic neurons were not produced at early stages of development, whereas ventral thalamic neurons were still produced. These defects were accompanied by improper formation of the boundary between dorsal and ventral thalamus, the zona limitans interthalamica (ZLI). Furthermore, the expression of an early marker of posterior forebrain development that marks the compartment from the midbrain-hindbrain junction to the ZLI (including the future dorsal thalamus, pretectum, and midbrain) was disrupted, supporting the idea that diencephalic development is abnormal from very early in embryogenesis. This study provides compelling in vivo evidence that thalamic development requires normal activity of the LRP6-mediated canonical Wnt signaling pathway.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Receptors, LDL/physiology , Thalamus/embryology , Animals , Cytoskeletal Proteins/physiology , Diencephalon/abnormalities , Diencephalon/embryology , Gestational Age , Hedgehog Proteins , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , Mice, Knockout , Morphogenesis , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/physiology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction/physiology , Thalamic Nuclei/abnormalities , Thalamic Nuclei/embryology , Thalamus/abnormalities , Trans-Activators/analysis , Trans-Activators/deficiency , Trans-Activators/physiology , Wnt Proteins , Wnt-5a Protein , beta Catenin
14.
Nat Rev Drug Discov ; 2(5): 379-90, 2003 May.
Article in English | MEDLINE | ID: mdl-12750741

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal, genetic disorder whose relentless progression underscores the urgency for developing a cure. Although Duchenne initiated clinical trials roughly 150 years ago, therapies for DMD remain supportive rather than curative. A paradigm shift towards developing rational therapeutic strategies occurred with identification of the DMD gene. Gene- and cell-based therapies designed to replace the missing gene and/or dystrophin protein have achieved varying degrees of success. However, pharmacological strategies not designed to replace dystrophin per se appear promising, and can circumvent many hurdles hampering gene- and cell-based therapy. Here, we will review present pharmacological strategies, in particular those dealing with functional substitution of dystrophin by utrophin and enhancing muscle progenitor commitment by myostatin blockade, with a view toward facilitating drug discovery for DMD.


Subject(s)
Drug Design , Muscular Dystrophies/drug therapy , Animals , Calcium/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Disease Models, Animal , Drug Evaluation, Preclinical , Dystrophin/genetics , Homeostasis , Humans , Membrane Proteins/genetics , Membrane Proteins/physiology , Promoter Regions, Genetic , Utrophin
15.
Biochem Biophys Res Commun ; 296(1): 221-6, 2002 Aug 09.
Article in English | MEDLINE | ID: mdl-12147254

ABSTRACT

Pro-inflammatory cytokine such as interleukin (IL)-1beta causes inflammation of articular cartilage via induction of cyclooxygenase (COX)-2 expression. We investigated in this study the role of beta-catenin in the IL-1beta regulation of COX-2 expression in articular chondrocytes. IL-1beta increased expression of COX-2 and induced accumulation and nuclear translocation of transcriptionally competent beta-catenin. Inhibition of beta-catenin degradation by the treatment of cells with LiCl or proteasome inhibitor stimulated expression of COX-2, indicating that transcriptionally active beta-catenin is sufficient to induce COX-2 expression. This was demonstrated further by the observation that ectopic expression of transcriptionally competent beta-catenin stimulated expression of COX-2. Levels of beta-catenin and COX-2 protein were increased in osteoarthritic and rheumatoid arthritic cartilage, suggesting that beta-catenin may play a role in the inflammatory responses of arthritic cartilage. Taken together, our data suggest that accumulation of transcriptionally active beta-catenin contributes to the expression of COX-2 in articular chondrocytes.


Subject(s)
Cartilage, Articular/enzymology , Cytoskeletal Proteins/physiology , Gene Expression Regulation, Enzymologic/physiology , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Trans-Activators/physiology , Animals , Arthritis/enzymology , Arthritis/metabolism , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cells, Cultured , Cyclooxygenase 2 , Cytoskeletal Proteins/metabolism , Interleukin-1/pharmacology , Rabbits , Trans-Activators/metabolism , Transcriptional Activation/drug effects , beta Catenin
16.
Curr Opin Cell Biol ; 14(1): 104-9, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11792551

ABSTRACT

The ERM (ezrin, radixin and moesin) family of proteins are linkers that tether actin microfilaments to the plasma membrane. Merlin, the NF2 tumor suppressor gene product, is highly homologous to ERM proteins. In ERM proteins and merlin, interdomain binding promotes auto-inhibition and homo-oligomerization or hetero-oligomerization. Recent studies have revealed that ERM proteins transduce growth signals, and have shed new light on how merlin links cell growth to the cytoskeleton.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Microfilament Proteins/physiology , Neurofibromin 2/physiology , Animals , Blood Proteins/physiology , Cell Division , Cytoskeletal Proteins/physiology , Membrane Proteins/physiology , Microvilli/ultrastructure , Models, Biological , Neoplasms/etiology , Phosphoproteins/physiology , Signal Transduction
17.
J Neuroendocrinol ; 13(3): 249-60, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11207939

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is a pivotal neuroendocrine regulator controlling reproductive functions. However, the scattered distribution of GnRH neurones in the mammalian brain has hindered studies on the development and differentiation of GnRH neurones. In the present study, we used the immortalized GnRH-producing GT1-1 cells to examine whether activation of protein kinase C (PKC) pathway with 12-O-tetradecanoyl-13-acetate (TPA) induces morphological and functional differentiation of GnRH neurones. TPA induced neurite outgrowth and inhibited proliferation of GT1-1 cells that were specifically antagonized by cotreatment of PKC inhibitor, calphostin C. The functional significance of TPA-induced differentiation of GT1-1 cells was manifested in part by the changes in the effects of gamma-aminobutyric acid (GABA) on intracellular Ca2+ levels. In untreated GT1-1 cells, activation of GABA-A receptor with 10 microM muscimol increased intracellular Ca2+ levels, whereas such stimulatory effects disappeared in GT1-1 cells bearing neurites. Accordingly, muscimol could not stimulate GnRH release in TPA-treated GT1-1 cells. To elucidate the molecular mechanism underlying TPA-induced neurite outgrowth, we performed differential display reverse transcription-polymerase chain reaction. Among several genes that are affected by TPA treatment, we found a significant induction of beta-catenin mRNA expression. Along with the rapid induction of beta-catenin protein levels, we observed that beta-catenin was reallocated from cell-cell adhesion sites to the growth cones within 3 h of TPA treatment. Transient transfection studies with green fluorescent protein as a reporter gene demonstrated that beta-catenin overexpression alone can promote neurite outgrowth in GT1-1 cells. Moreover, TPA was found to increase the transcription-activational roles of beta-catenin. Together, these data provide evidence that beta-catenin is involved in the TPA-induced functional differentiation of immortalized GnRH neurones.


Subject(s)
Cytoskeletal Proteins/physiology , Gonadotropin-Releasing Hormone/blood , Hypothalamus/ultrastructure , Neurites/drug effects , Neurites/physiology , Neurons/ultrastructure , Tetradecanoylphorbol Acetate/pharmacology , Trans-Activators , Calcium/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Transformed , Cytoskeletal Proteins/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Green Fluorescent Proteins , Luminescent Proteins/genetics , Muscimol/pharmacology , Naphthalenes/pharmacology , Protein Kinase C/antagonists & inhibitors , RNA, Messenger/analysis , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , beta Catenin , gamma-Aminobutyric Acid/pharmacology
18.
J Biol Chem ; 276(10): 7366-75, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11104755

ABSTRACT

The majority of cases with early onset familial Alzheimer's disease have been attributed to mutations in the presenilin 1 (PS1) gene. PS1 protein is a component of a high molecular weight membrane-bound complex that also contains beta-catenin. The physiological relevance of the association between PS1 and beta-catenin remains controversial. In this study, we report the identification and functional characterization of a highly conserved glycogen synthase kinase-3beta consensus phosphorylation site within the hydrophilic loop domain of PS1. Site-directed mutagenesis, together with in vitro and in vivo phosphorylation assays, indicates that PS1 residues Ser(353) and Ser(357) are glycogen synthase kinase-3beta targets. Substitution of one or both of these residues greatly reduces the ability of PS1 to associate with beta-catenin. By disrupting this interaction, we demonstrate that the association between PS1 and beta-catenin has no effect on Abeta peptide production, beta-catenin stability, or cellular susceptibility to apoptosis. Significantly, in the absence of PS1/beta-catenin association, we found no alteration in beta-catenin signaling using induction of this pathway by exogenous expression of Wnt-1 or beta-catenin and a Tcf/Lef transcriptional assay. These results argue against a pathologically relevant role for the association between PS1 and beta-catenin in familial Alzheimer's disease.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Cytoskeletal Proteins/chemistry , Membrane Proteins/chemistry , Signal Transduction , Trans-Activators , Alzheimer Disease/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Apoptosis , Binding Sites , Blotting, Western , Cell Death , Cell Line , Cell Nucleus/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/physiology , Cytosol/metabolism , DNA, Complementary/metabolism , Genetic Vectors , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Luciferases/metabolism , Membrane Proteins/genetics , Membrane Proteins/physiology , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Peptide Fragments/chemistry , Phosphorylation , Precipitin Tests , Presenilin-1 , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Serine/chemistry , Transfection , beta Catenin
19.
Adv Cancer Res ; 77: 1-24, 2000.
Article in English | MEDLINE | ID: mdl-10549354

ABSTRACT

Wingless/Wnt signaling directs cell-fate choices during embryonic development. In Drosophila, Wingless signaling mediates endoderm induction and the establishment of segment polarity in the developing embryo. The fly Wingless cascade is strikingly similar to the vertebrate Wnt signaling pathway, which controls a number of key developmental decisions such as dorsal-ventral patterning in Xenopus. Factors of the TCF/LEF HMG domain family (Tcfs) have recently been established as the downstream effectors of the Wingless/Wnt signal transduction pathways. Upon Wingless/Wnt signaling, a cascade is initiated that results in the accumulation of cytoplasmic beta-catenin (or its fly homolog, Armadillo). There is also a concomitant translocation of beta-catenin/Armadillo to the nucleus, where it interacts with a specific sequence motif at the N terminus of Tcfs to generate a transcriptionally active complex. This bipartite transcription factor is targeted to the upstream regulatory regions of Tcf target genes including Siamois and Nodal related gene-3 in Xenopus, engrailed and Ultrabithorax in Drosophila via the sequence-specific HMG box, and mediates their transcriptional activation by virtue of transactivation domains contributed by beta-catenin/Armadillo. In the absence of Wingless/Wnt signals, a key negative regulator of the pathway, GSK3 beta, is activated, which mediates the downregulation of cytoplasmic beta-catenin/Armadillo via the ubiquitin-proteasome pathway. In the absence of nuclear beta-catenin, the Tcfs recruit the corepressor protein Groucho to the target gene enhancers and actively repress their transcription. An additional corepressor protein, CREB-binding protein (CBP), may also be involved in this repression of Tcf target gene activity. Several other proteins, including adenomatous polyposis coli (APC), GSK3 beta, and Axin/Conductin, are instrumental in the regulation of beta-catenin/Armadillo. In APC-deficient colon carcinoma cell lines, beta-catenin accumulates and is constitutively complexed with nuclear Tcf-4. A proportion of APC wild-type colon carcinomas and melanomas also contains constitutive nuclear Tcf-4/beta-catenin complexes as a result of dominant mutations in the N terminus of beta-catenin that render it insensitive to downregulation by APC, GSK3 beta, and Axin/Conductin. This results in the unregulated expression of Tcf-4 target genes such as c-myc. Based on the established role for Tcf-4 in maintaining intestinal stem cells it is likely that deregulation of c-myc expression as a result of constitutive Tcf-4/beta-catenin activity promotes uncontrolled intestinal cell proliferation. This would readily explain the formation of intestinal polyps during colon carcinogenesis. Similar mechanisms leading to deregulation of Tcf target gene activity are likely to be involved in melanoma and other forms of cancer.


Subject(s)
Cadherins/physiology , Cytoskeletal Proteins/physiology , High Mobility Group Proteins/metabolism , Proto-Oncogene Proteins/physiology , Signal Transduction , Trans-Activators , Transcription Factors/metabolism , Zebrafish Proteins , Animals , Body Patterning , Colonic Neoplasms/genetics , Colonic Neoplasms/physiopathology , Drosophila/embryology , Humans , Mitogens/physiology , Tumor Cells, Cultured , Wnt Proteins , Xenopus/embryology , Xenopus Proteins , beta Catenin
20.
Wound Repair Regen ; 6(3): 202-12, 1998.
Article in English | MEDLINE | ID: mdl-9776864

ABSTRACT

Scar contraction following the healing of deep partial-thickness or full-thickness dermal injury is a leading cause of functional and cosmetic morbidity. The therapeutic use of interferon for the treatment of fibroproliferative disorders associated with scar contraction, including hypertrophic scar, has been suggested because of its antifibrotic properties. Treatment of fibroblasts with interferon has been shown to reduce the rate and extent of contraction using the in vitro fibroblast-populated collagen lattice model. In order to establish the effect of interferon-alpha2b on full-thickness wound contraction in vivo, osmotic pumps loaded with interferon or sterile saline were implanted intraperitoneally in guinea pigs. Seven days following implantation, six full-thickness punch biopsy wounds were created and were monitored by daily assessment of the wound. There was a significant reduction in the rate of wound contraction in the interferon-treated animals after day 3 (p < 0.01). Western blot analysis was used to quantitate selected cytoskeletal proteins in the normal skin and tissue biopsied from the wound at days 7, 14, and 21 postinjury. The amount of vimentin in the contracted wound increased following injury as compared with the amount present in normal skin (p < 0.0001); however, the relative amounts of the myofibroblast-associated cytoskeletal proteins alpha-smooth muscle actin and smooth muscle myosin were less than those found in normal, uninjured skin. By using vimentin to adjust the levels of cytoskeletal proteins for the increase in cellularity in the wounds, both alpha-smooth muscle actin and smooth muscle myosin significantly increased after closure of the wounds on day 14, as compared with the open-wound stage (day 7), before further reductions occurred with remodeling on day 21. Measurement of apoptotic cells using the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay revealed an increase in apoptosis in the interferon-alpha2b-treated animals at 21 days following wounding (p < 0.001), which did not colocalize with alpha-smooth muscle actin staining. Taken together, these findings suggest that interferon-alpha2b inhibits wound contraction in vivo, not through an appreciable alteration in myofibroblast number or cytoskeletal protein expression, but possibly through a reduction in fibroblast cellularity by the induction of apoptosis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Cicatrix/etiology , Cicatrix/prevention & control , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/physiology , Interferon-alpha/therapeutic use , Wound Healing/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis , Biopsy , Blotting, Western , Cicatrix/pathology , Cicatrix/physiopathology , Cytoskeletal Proteins/analysis , Disease Models, Animal , Drug Evaluation, Preclinical , Guinea Pigs , Immunohistochemistry , In Situ Nick-End Labeling , Interferon alpha-2 , Interferon-alpha/pharmacology , Random Allocation , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL