Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neurosci ; 40(8): 1596-1605, 2020 02 19.
Article in English | MEDLINE | ID: mdl-32075947

ABSTRACT

SynGAP is a potent regulator of biochemical signaling in neurons and plays critical roles in neuronal function. It was first identified in 1998, and has since been extensively characterized as a mediator of synaptic plasticity. Because of its involvement in synaptic plasticity, SynGAP has emerged as a critical protein for normal cognitive function. In recent years, mutations in the SYNGAP1 gene have been shown to cause intellectual disability in humans and have been linked to other neurodevelopmental disorders, such as autism spectrum disorders and schizophrenia. While the structure and biochemical function of SynGAP have been well characterized, a unified understanding of the various roles of SynGAP at the synapse and its contributions to neuronal function remains to be achieved. In this review, we summarize and discuss the current understanding of the multifactorial role of SynGAP in regulating neuronal function gathered over the last two decades.


Subject(s)
Brain/physiology , Cognition/physiology , Neurons/physiology , Synapses/physiology , ras GTPase-Activating Proteins/physiology , Animals , Humans , Neuronal Plasticity/physiology , Synaptic Transmission/physiology
2.
J Neurosci ; 40(41): 7980-7994, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32887745

ABSTRACT

SYNGAP1 is a major genetic risk factor for global developmental delay, autism spectrum disorder, and epileptic encephalopathy. De novo loss-of-function variants in this gene cause a neurodevelopmental disorder defined by cognitive impairment, social-communication disorder, and early-onset seizures. Cell biological studies in mouse and rat neurons have shown that Syngap1 regulates developing excitatory synapse structure and function, with loss-of-function variants driving formation of larger dendritic spines and stronger glutamatergic transmission. However, studies to date have been limited to mouse and rat neurons. Therefore, it remains unknown how SYNGAP1 loss of function impacts the development and function of human neurons. To address this, we used CRISPR/Cas9 technology to ablate SYNGAP1 protein expression in neurons derived from a commercially available induced pluripotent stem cell line (hiPSC) obtained from a human female donor. Reducing SynGAP protein expression in developing hiPSC-derived neurons enhanced dendritic morphogenesis, leading to larger neurons compared with those derived from isogenic controls. Consistent with larger dendritic fields, we also observed a greater number of morphologically defined excitatory synapses in cultures containing these neurons. Moreover, neurons with reduced SynGAP protein had stronger excitatory synapses and expressed synaptic activity earlier in development. Finally, distributed network spiking activity appeared earlier, was substantially elevated, and exhibited greater bursting behavior in SYNGAP1 null neurons. We conclude that SYNGAP1 regulates the postmitotic maturation of human neurons made from hiPSCs, which influences how activity develops within nascent neural networks. Alterations to this fundamental neurodevelopmental process may contribute to the etiology of SYNGAP1-related disorders.SIGNIFICANCE STATEMENTSYNGAP1 is a major genetic risk factor for global developmental delay, autism spectrum disorder, and epileptic encephalopathy. While this gene is well studied in rodent neurons, its function in human neurons remains unknown. We used CRISPR/Cas9 technology to disrupt SYNGAP1 protein expression in neurons derived from an induced pluripotent stem cell line. We found that induced neurons lacking SynGAP expression exhibited accelerated dendritic morphogenesis, increased accumulation of postsynaptic markers, early expression of synapse activity, enhanced excitatory synaptic strength, and early onset of neural network activity. We conclude that SYNGAP1 regulates the postmitotic differentiation rate of developing human neurons and disrupting this process impacts the function of nascent neural networks. These altered developmental processes may contribute to the etiology of SYNGAP1 disorders.


Subject(s)
Dendrites/physiology , Nerve Net/physiology , Nervous System/growth & development , Synapses/physiology , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/physiology , CRISPR-Cas Systems , Cell Differentiation/genetics , Cell Size , Cells, Cultured , Excitatory Postsynaptic Potentials/genetics , Female , Gene Deletion , Humans , Neurodevelopmental Disorders/genetics , Pluripotent Stem Cells
3.
FASEB J ; 34(1): 540-554, 2020 01.
Article in English | MEDLINE | ID: mdl-31914585

ABSTRACT

A costimulatory signal from the tumor necrosis factor receptor (TNFR) family molecule OX40 (CD134), which is induced on activated T cells, is important for T-cell immunity. Aberrant OX40 cosignaling has been implicated in autoimmune and inflammatory disorders. However, the molecular mechanism by which the OX40 cosignaling regulates the T-cell response remains obscure. We found that OX40 associated with a scaffold protein, IQ motif-containing GTPase-activating protein 1 (IQGAP1) after ligation by its ligand OX40L. Naïve CD4+ T cells from Iqgap1-/- mice displayed enhanced proliferation and cytokine secretion upon receiving OX40 cosignaling. A C-terminal IQGAP1 region was responsible for its association with OX40, and TNFR-associated factor 2 (TRAF2) bridged these two proteins. The enhanced cytokine response in Iqgap1-/- T cells was restored by the expression of the C-terminal IQGAP1. Thus, the IQGAP1 binding limits the OX40 cosignaling. Disease severity of experimental autoimmune encephalomyelitis (EAE) was significantly exacerbated in Iqgap1-/- mice as compared to wild-type mice. Additionally, recipient mice with Iqgap1-/- donor CD4+ T cells exhibited significantly higher EAE scores than those with their wild-type counterparts, and OX40 blockade led to a significant reduction in the EAE severity. Thus, our study defines an important component of the OX40 cosignaling that restricts inflammation driven by antigen-activated T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunologic Memory/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Receptors, OX40/metabolism , ras GTPase-Activating Proteins/physiology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, OX40/genetics , Signal Transduction
4.
J Biol Chem ; 294(44): 16198-16213, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31515267

ABSTRACT

Ligand-dependent differences in the regulation and internalization of the µ-opioid receptor (MOR) have been linked to the severity of adverse effects that limit opiate use in pain management. MOR activation by morphine or [d-Ala2,N-MePhe4, Gly-ol]enkephalin (DAMGO) causes differences in spatiotemporal signaling dependent on MOR distribution at the plasma membrane. Morphine stimulation of MOR activates a Gαi/o-Gßγ-protein kinase C (PKC) α phosphorylation pathway that limits MOR distribution and is associated with a sustained increase in cytosolic extracellular signal-regulated kinase (ERK) activity. In contrast, DAMGO causes a redistribution of the MOR at the plasma membrane (before receptor internalization) that facilitates transient activation of cytosolic and nuclear ERK. Here, we used proximity biotinylation proteomics to dissect the different protein-interaction networks that underlie the spatiotemporal signaling of morphine and DAMGO. We found that DAMGO, but not morphine, activates Ras-related C3 botulinum toxin substrate 1 (Rac1). Both Rac1 and nuclear ERK activity depended on the scaffolding proteins IQ motif-containing GTPase-activating protein-1 (IQGAP1) and Crk-like (CRKL) protein. In contrast, morphine increased the proximity of the MOR to desmosomal proteins, which form specialized and highly-ordered membrane domains. Knockdown of two desmosomal proteins, junction plakoglobin or desmocolin-1, switched the morphine spatiotemporal signaling profile to mimic that of DAMGO, resulting in a transient increase in nuclear ERK activity. The identification of the MOR-interaction networks that control differential spatiotemporal signaling reported here is an important step toward understanding how signal compartmentalization contributes to opioid-induced responses, including anti-nociception and the development of tolerance and dependence.


Subject(s)
Analgesics, Opioid/metabolism , Receptors, Opioid, mu/metabolism , rac1 GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Analgesics, Opioid/pharmacology , Animals , Cell Membrane/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , HEK293 Cells , Humans , Ligands , MAP Kinase Signaling System/physiology , Morphine/metabolism , Morphine/pharmacology , Phosphorylation , Protein Interaction Mapping/methods , Protein Interaction Maps , Receptors, Opioid, mu/genetics , Signal Transduction/physiology , rac1 GTP-Binding Protein/physiology , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/physiology
5.
Br J Cancer ; 123(7): 1154-1163, 2020 09.
Article in English | MEDLINE | ID: mdl-32632148

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV) has a crucial role in the progression of hepatocellular carcinoma (HCC). Tumour cells must develop anoikis resistance in order to survive before metastasis. This study aimed to investigate the mechanism of IQGAP1 in HBV-mediated anoikis evasion and metastasis in HCC cells. METHODS: IQGAP1 expression was detected by immunohistochemistry, real-time PCR and immunoblot analysis. Lentiviral-mediated stable upregulation or knockdown of IGAQP1, immunoprecipitation, etc. were used in function and mechanism study. RESULTS: IQGAP1 was markedly upregulated in HBV-positive compared with HBV-negative HCC cells and tissues. IQGAP1 was positively correlated to poor prognosis of HBV-associated HCC patients. IQGAP1 overexpression significantly enhanced the anchorage-independent growth and metastasis, whereas IQGAP1-deficient HCC cells are more sensitive to anoikis. Mechanistically, we found that HBV-induced ROS enhanced the association of IQGAP1 and Rac1 that activated Rac1, leading to phosphorylation of Src/FAK pathway. Antioxidants efficiently inhibited IQGAP1-mediated anoikis resistance and metastasis. CONCLUSIONS: Our study indicated an important mechanism by which upregulated IQGAP1 by HBV promoted anoikis resistance, migration and invasion of HCC cells through Rac1-dependent ROS accumulation and activation of Src/FAK signalling, suggesting IQGAP1 as a prognostic indicator and a novel therapeutic target in HCC patients with HBV infection.


Subject(s)
Carcinoma, Hepatocellular/pathology , Focal Adhesion Kinase 1/physiology , Liver Neoplasms/pathology , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/physiology , ras GTPase-Activating Proteins/physiology , src-Family Kinases/physiology , Animals , Anoikis , Cell Line, Tumor , Female , Hepatitis B/complications , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Signal Transduction/physiology
6.
Mol Cell ; 45(2): 196-209, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22206868

ABSTRACT

Growth factors activate Ras, PI3K, and other signaling pathways. It is not well understood how these signals are translated by individual cells into a decision to proliferate or differentiate. Here, using single-cell image analysis of nerve growth factor (NGF)-stimulated PC12 cells, we identified a two-dimensional phospho-ERK (pERK)-phospho-AKT (pAKT) response map with a curved boundary that separates differentiating from proliferating cells. The boundary position remained invariant when different stimuli were used or upstream signaling components perturbed. We further identified Rasa2 as a negative feedback regulator that links PI3K to Ras, placing the stochastically distributed pERK-pAKT signals close to the decision boundary. This allows for uniform NGF stimuli to create a subpopulation of cells that differentiates with each cycle of proliferation. Thus, by linking a complex signaling system to a simpler intermediate response map, cells gain unique integration and control capabilities to balance cell number expansion with differentiation.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Nerve Growth Factor/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cyclin D/genetics , Cyclin D/metabolism , Cyclin D/physiology , Gene Expression Regulation , Gene Knockdown Techniques , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Protein Stability , Proto-Oncogene Proteins c-akt/physiology , Rats , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/physiology
7.
Dev Biol ; 418(2): 258-67, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27565025

ABSTRACT

Plexins (Plexs) comprise a large family of cell surface receptors for semaphorins (Semas) that function as evolutionarily conserved guidance molecules. GTPase activating protein (GAP) activity for Ras family small GTPases has been implicated in plexin signaling cascades through its RasGAP domain. However, little is known about how Ras family GTPases are controlled in vivo by plexin signaling. Here, we found that Drosophila Rap1, a member of the Ras family of GTPases, plays an important role controlling intersegmental nerve b motor axon guidance during neural development. Gain-of-function studies using dominant-negative and constitutively active forms of Rap1 indicate that Rap1 contributes to axonal growth and guidance. Genetic interaction analyses demonstrate that the Sema-1a/PlexA-mediated repulsive guidance function is regulated positively by Rap1. Furthermore, neuronal expression of mutant PlexA robustly restored defasciculation defects in PlexA null mutants when the catalytic arginine fingers of the PlexA RasGAP domain critical for GAP activity were disrupted. However, deleting the RasGAP domain abolished the ability of PlexA to rescue the PlexA guidance phenotypes. These findings suggest that PlexA-mediated motor axon guidance is dependent on the presence of the PlexA RasGAP domain, but not on its GAP activity toward Ras family small GTPases.


Subject(s)
Axon Guidance/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Drosophila melanogaster/physiology , Monomeric GTP-Binding Proteins/physiology , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/physiology , Telomere-Binding Proteins/physiology , ras GTPase-Activating Proteins/physiology , Animals , Animals, Genetically Modified , Axon Guidance/genetics , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Genes, Insect , Monomeric GTP-Binding Proteins/deficiency , Monomeric GTP-Binding Proteins/genetics , Motor Neurons/physiology , Mutagenesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Shelterin Complex , Telomere-Binding Proteins/deficiency , Telomere-Binding Proteins/genetics , Up-Regulation , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
8.
EMBO J ; 32(19): 2617-30, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-23982733

ABSTRACT

Phosphatidylinositol 4,5 bisphosphate (PIP2) is a key lipid messenger for regulation of cell migration. PIP2 modulates many effectors, but the specificity of PIP2 signalling can be defined by interactions of PIP2-generating enzymes with PIP2 effectors. Here, we show that type Iγ phosphatidylinositol 4-phosphate 5-kinase (PIPKIγ) interacts with the cytoskeleton regulator, IQGAP1, and modulates IQGAP1 function in migration. We reveal that PIPKIγ is required for IQGAP1 recruitment to the leading edge membrane in response to integrin or growth factor receptor activation. Moreover, IQGAP1 is a PIP2 effector that directly binds PIP2 through a polybasic motif and PIP2 binding activates IQGAP1, facilitating actin polymerization. IQGAP1 mutants that lack PIPKIγ or PIP2 binding lose the ability to control directional cell migration. Collectively, these data reveal a synergy between PIPKIγ and IQGAP1 in the control of cell migration.


Subject(s)
Cell Movement/physiology , Phosphatidylinositol 4,5-Diphosphate/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , ras GTPase-Activating Proteins/physiology , Cell Line, Tumor , Humans
9.
J Am Soc Nephrol ; 27(9): 2597-608, 2016 09.
Article in English | MEDLINE | ID: mdl-26839366

ABSTRACT

Diabetic nephropathy (DN) is a frequent and severe complication of diabetes that is structurally characterized by glomerular basement membrane thickening, extracellular matrix accumulation, and destabilization of podocyte foot processes. MicroRNAs (miRNAs) are dysregulated in DN, but identification of the specific miRs involved remains incomplete. Here, we confirm that the peripheral blood from patients with diabetes and the kidneys of animals with type 1 or 2 diabetes have low levels of miR-23b compared with those of their nondiabetic counterparts. Furthermore, exposure to high glucose downregulated miR-23b in cultured kidney cells. In contrast, renal expression of Ras GTPase-activating protein SH3 domain-binding protein 2 (G3BP2), a putative miR-23b target, increased in DN. In vitro, overexpression of miR-23b decreased, and inhibition of miR-23b increased, G3BP2 expression levels. Bioinformatics analysis also revealed p53 binding sites in the miR-23b promoter; in vitro inhibition of p53 or the upstream p38 mitogen-activated protein kinase (p38MAPK) upregulated miR-23b expression in high-glucose conditions. In turn, inhibition of G3BP2 or overexpression of miR-23b downregulated p53 and p38MAPK expression in high-glucose conditions. In vivo, overexpression of miR-23b or inhibition of p53 in db/db mice reversed hyperalbuminuria and kidney fibrosis, whereas miR-23b antagomir treatment promoted renal fibrosis and increased albuminuria in wild-type mice. These data suggest that hyperglycemia regulates pathogenic processes in DN through an miR-23b/G3BP2 feedback circuit involving p38MAPK and p53. In conclusion, these results reveal a role for miR-23b in DN and indicate a novel potential therapeutic target.


Subject(s)
Albuminuria/enzymology , Diabetic Nephropathies/enzymology , Kidney/pathology , MicroRNAs/physiology , ras GTPase-Activating Proteins/physiology , Adaptor Proteins, Signal Transducing , Albuminuria/complications , Animals , Diabetic Nephropathies/complications , Fibrosis/complications , Fibrosis/enzymology , Male , Mice , Mice, Inbred C57BL , RNA-Binding Proteins
10.
J Proteome Res ; 15(12): 4675-4685, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27934296

ABSTRACT

Intracellular actin-based motility of the melioidosis pathogen Burkholderia pseudomallei requires the bacterial factor BimA. Located at one pole of the bacterium, BimA recruits and polymerizes cellular actin to promote bacterial motility within and between cells. Here, we describe an affinity approach coupled with mass spectrometry to identify cellular proteins recruited to BimA-expressing bacteria under conditions that promote actin polymerization. We identified a group of cellular proteins that are recruited to the B. pseudomallei surface in a BimA-dependent manner, a subset of which were independently validated with specific antisera including the ubiquitous scaffold protein Ras GTPase-activating-like protein (IQGAP1). IQGAP1 integrates several key cellular signaling pathways including those involved in actin dynamics and has been shown to be involved in the adhesion of attaching and effacing Escherichia coli to infected cells and invasion of host cells by Salmonella enterica serovar Typhimurium. Although a direct interaction between BimA and IQGAP1 could not be detected using either conventional pulldown or yeast two hybrid techniques, confocal microscopy revealed that IQGAP1 is recruited to B. pseudomallei actin tails in infected cells, and siRNA-mediated knockdown highlighted a role for this protein in controlling the length and actin density of B. pseudomallei actin tails.


Subject(s)
Actins/metabolism , Burkholderia pseudomallei/chemistry , Cell Movement , Bacterial Proteins/analysis , Bacterial Proteins/physiology , Burkholderia pseudomallei/cytology , Cell Polarity , Humans , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Polymerization , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/physiology
11.
Dev Neurosci ; 37(2): 131-41, 2015.
Article in English | MEDLINE | ID: mdl-25721469

ABSTRACT

Dab2IP (DOC-2/DAB2 interacting protein) is a GTPase-activating protein which is involved in various aspects of brain development in addition to its roles in tumor formation and apoptosis in other systems. In this study, we carefully examined the expression profile of Dab2IP and investigated its physiological role during brain development using a Dab2IP-knockdown (KD) mouse model created by retroviral insertion of a LacZ-encoding gene-trapping cassette. LacZ staining revealed that Dab2IP is expressed in the ventricular zone as well as the cortical plate and the intermediate zone. Immunohistochemical analysis showed that Dab2IP protein is localized in the leading process and proximal cytoplasmic regions of migrating neurons in the intermediate zone. Bromodeoxyuridine birth dating experiments in combination with immunohistochemical analysis using layer-specific markers showed that Dab2IP is important for proper positioning of a subset of layer II-IV neurons in the developing cortex. Notably, neuronal migration was not completely disrupted in the cerebral cortex of Dab2IP-KD mice and disruption of migration was not strictly layer specific. Previously, we found that Dab2IP regulates multipolar transition in cortical neurons. Others have shown that Rap1 regulates the transition from multipolar to bipolar morphology in migrating postmitotic neurons through N-cadherin signaling and somal translocation in the superficial layer of the cortical plate through integrin signaling. Therefore, we examined whether Rap1 and integrin signaling were affected in Dab2IP-KD brains. We found that Dab2IP-KD resulted in higher levels of activated Rap1 and integrin in the developing cortex. Taken together, our results suggest that Dab2IP plays an important role in the migration and positioning of a subpopulation of later-born (layers II-IV) neurons, likely through the regulation of Rap1 and integrin signaling.


Subject(s)
Cell Movement/physiology , Cerebral Cortex , Integrins/metabolism , Neurons/cytology , rap1 GTP-Binding Proteins/metabolism , ras GTPase-Activating Proteins/physiology , Animals , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , ras GTPase-Activating Proteins/metabolism
12.
Arterioscler Thromb Vasc Biol ; 34(3): 603-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24407031

ABSTRACT

OBJECTIVE: To investigate the novel function of ASK1-interacting protein-1 (AIP1) in vascular endothelial cell growth factor receptor (VEGFR)-3 signaling, and VEGFR-3-dependent angiogenesis and lymphangiogenesis. APPROACH AND RESULTS: AIP1, a signaling scaffold protein, is highly expressed in the vascular endothelium. We have previously reported that AIP1 functions as an endogenous inhibitor in pathological angiogenesis by blocking VEGFR-2 activity. Surprisingly, here we observe that mice with a global deletion of AIP1-knockout mice (AIP1-KO) exhibit reduced retinal angiogenesis with less sprouting and fewer branches. Vascular endothelial cell (but not neuronal)-specific deletion of AIP1 causes similar defects in retinal angiogenesis. The reduced retinal angiogenesis correlates with reduced expression in VEGFR-3 despite increased VEGFR-2 levels in AIP1-KO retinas. Consistent with the reduced expression of VEGFR-3, AIP1-KO show delayed developmental lymphangiogenesis in neonatal skin and mesentery, and mount weaker VEGF-C-induced cornea lymphangiogenesis. In vitro, human lymphatic endothelial cells with AIP1 small interfering RNA knockdown, retinal endothelial cells, and lymphatic endothelial cells isolated from AIP1-KO all show attenuated VEGF-C-induced VEGFR-3 signaling. Mechanistically, we demonstrate that AIP1 via vegfr-3-specific miR-1236 increases VEGFR-3 protein expression and that, by directly binding to VEGFR-3, it enhances VEGFR-3 endocytosis and stability. CONCLUSION: Our in vivo and in vitro results provide the first insight into the mechanism by which AIP1 mediates VEGFR-3-dependent angiogenic and lymphangiogenic signaling.


Subject(s)
Carrier Proteins/physiology , Lymphangiogenesis/physiology , Retinal Neovascularization/physiopathology , Vascular Endothelial Growth Factor Receptor-3/physiology , ras GTPase-Activating Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cells, Cultured , Cornea , Endocytosis , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Eye Proteins/physiology , Guanylate Kinases , Humans , Mice , Mice, Knockout , MicroRNAs/physiology , Neurons/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Receptors, Notch/physiology , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/physiology , Vascular Endothelial Growth Factor Receptor-3/biosynthesis , Vascular Endothelial Growth Factor Receptor-3/genetics , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
13.
Cell Mol Life Sci ; 71(14): 2759-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24352566

ABSTRACT

p-21 activated 6 (PAK6), first identified as interacting with the androgen receptor (AR), is over-expressed in multiple cancer tissues and has been linked to the progression of prostate cancer, however little is known about PAK6 function in the absence of AR signaling. We report here that PAK6 is specifically required for carcinoma cell-cell dissociation downstream of hepatocyte growth factor (HGF) for both DU145 prostate cancer and HT29 colon cancer cells. Moreover, PAK6 overexpression can drive cells to escape from adhesive colonies in the absence of stimulation. We have localized PAK6 to cell-cell junctions and have detected a direct interaction between the kinase domain of PAK6 and the junctional protein IQGAP1. Co-expression of IQGAP1 and PAK6 increases cell colony escape and leads to elevated PAK6 activation. Further studies have identified a PAK6/E-cadherin/IQGAP1 complex downstream of HGF. Moreover, we find that ß-catenin is also localized with PAK6 in cell-cell junctions and is a novel PAK6 substrate. We propose a unique role for PAK6, independent of AR signaling, where PAK6 drives junction disassembly during HGF-driven cell-cell dissociation via an IQGAP1/E-cadherin complex that leads to the phosphorylation of ß-catenin and the disruption of cell-cell adhesions.


Subject(s)
Intercellular Junctions/metabolism , p21-Activated Kinases/physiology , ras GTPase-Activating Proteins/physiology , Cell Adhesion/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition , HEK293 Cells , HT29 Cells , Humans , Phosphorylation , Protein Interaction Mapping , beta Catenin/metabolism , p21-Activated Kinases/metabolism , ras GTPase-Activating Proteins/metabolism
14.
J Neurosci ; 33(25): 10447-52, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785156

ABSTRACT

Critical periods of developmental plasticity contribute to the refinement of neural connections that broadly shape brain development. These windows of plasticity are thought to be important for the maturation of perception, language, and cognition. Synaptic properties in cortical regions that underlie critical periods influence the onset and duration of windows, although it remains unclear how mechanisms that shape synapse development alter critical-period properties. In this study, we demonstrate that inactivation of a single copy of syngap1, which causes a surprisingly common form of sporadic, non-syndromic intellectual disability with autism in humans, induced widespread early functional maturation of excitatory connections in the mouse neocortex. This accelerated functional maturation was observed across distinct areas and layers of neocortex and directly influenced the duration of a critical-period synaptic plasticity associated with experience-dependent refinement of cortical maps. These studies support the idea that genetic control over synapse maturation influences the duration of critical-period plasticity windows. These data also suggest that critical-period duration links synapse maturation rates to the development of intellectual ability.


Subject(s)
Critical Period, Psychological , Neuronal Plasticity/physiology , Synapses/physiology , ras GTPase-Activating Proteins/physiology , Animals , Brain/growth & development , Brain/physiology , Brain Mapping , Cognition/physiology , Data Interpretation, Statistical , Electric Stimulation , Female , In Vitro Techniques , Long-Term Potentiation/physiology , Male , Mice , Neocortex/growth & development , Neocortex/physiology , Patch-Clamp Techniques , Social Behavior , Thalamus/growth & development , Thalamus/physiology , ras GTPase-Activating Proteins/genetics
15.
J Biol Chem ; 288(31): 22378-86, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23788642

ABSTRACT

Nrf2 plays a critical role in the regulation of cellular oxidative stress. MEK-ERK activation has been shown to be one of the major pathways resulting in the activation of Nrf2 and induction of Nrf2 downstream targets, including phase II detoxifying/antioxidant genes in response to oxidative stress and xenobiotics. In this study, IQGAP1 (IQ motif-containing GTPase-activating protein 1), a new Nrf2 interaction partner that we have published previously, was found to modulate MEK-ERK-mediated Nrf2 activation and induction of phase II detoxifying/antioxidant genes. Nrf2 binds directly to the IQ domain (amino acids 699-905) of IQGAP1. Knockdown of IQGAP1 significantly attenuated phenethyl isothiocyanate- or MEK-mediated activation of the MEK-ERK-Nrf2 pathway. Knockdown of IQGAP1 also attenuated MEK-mediated increased stability of Nrf2, which in turn was associated with a decrease in the nuclear translocation of Nrf2 and a decrease in the expression of phase II detoxifying/antioxidant genes. In the aggregate, these results suggest that IQGAP1 may play an important role in the MEK-ERK-Nrf2 signaling pathway.


Subject(s)
MAP Kinase Signaling System , NF-E2-Related Factor 2/metabolism , ras GTPase-Activating Proteins/physiology , Base Sequence , Cell Nucleus/metabolism , DNA Primers , Enzyme Activation , HEK293 Cells , Humans , Polymerase Chain Reaction
16.
J Immunol ; 188(5): 2057-63, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22345702

ABSTRACT

Activating and inhibiting receptors of lymphocytes collect valuable information about their mikròs kósmos. This information is essential to initiate or to turn off complex signaling pathways. Irrespective of these advances, our knowledge on how these intracellular activation cascades are coordinated in a spatiotemporal manner is far from complete. Among multiple explanations, the scaffolding proteins have emerged as a critical piece of this evolutionary tangram. Among many, IQGAP1 is one of the essential scaffolding proteins that coordinate multiple signaling pathways. IQGAP1 possesses multiple protein interaction motifs to achieve its scaffolding functions. Using these domains, IQGAP1 has been shown to regulate a number of essential cellular events. This includes actin polymerization, tubulin multimerization, microtubule organizing center formation, calcium/calmodulin signaling, Pak/Raf/Mek1/2-mediated Erk1/2 activation, formation of maestrosome, E-cadherin, and CD44-mediated signaling and glycogen synthase kinase-3/adenomatous polyposis coli-mediated ß-catenin activation. In this review, we summarize the recent developments and exciting new findings of cellular functions of IQGAP1.


Subject(s)
Cell Communication/immunology , Intracellular Space/immunology , Intracellular Space/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Protein Multimerization/immunology , ras GTPase-Activating Proteins/physiology , Animals , Cells, Cultured , Intracellular Space/chemistry , Lymphocyte Subsets/chemistry , Mice , Mice, Knockout , Protein Interaction Mapping/methods , Signal Transduction/immunology , ras GTPase-Activating Proteins/chemistry , ras GTPase-Activating Proteins/deficiency
17.
J Neurosci ; 32(47): 16971-81, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23175848

ABSTRACT

Neurofibromatosis type I (NF1), caused by the mutation in the NF1 gene, is characterized by multiple pathological symptoms. Importantly, ~50% of NF1 patients also suffer learning difficulty. Although downstream pathways are well studied, regulation of the NF1-encoded neurofibromin protein is less clear. Here, we focused on the pathophysiology of Drosophila NF1 mutants in synaptic growth at neuromuscular junctions. Our analysis suggests that the Drosophila neurofibromin protein NF1 is required to constrain synaptic growth and transmission. NF1 functions downstream of the Drosophila focal adhesion kinase (FAK) Fak56 and physically interacts with Fak56. The N-terminal region of NF1 mediates the interaction with Fak56 and is required for the signaling activity and presynaptic localization of NF1. In presynapses, NF1 acts via the cAMP pathway, but independent of its GAP activity, to restrain synaptic growth. Thus, presynaptic FAK signaling may be disrupted, causing abnormal synaptic growth and transmission in the NF1 genetic disorder.


Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , Focal Adhesion Kinase 1/physiology , Nerve Tissue Proteins/physiology , Neuromuscular Junction/physiology , Synapses/physiology , ras GTPase-Activating Proteins/physiology , Adenylyl Cyclases/physiology , Animals , Cyclic AMP/physiology , Electrophysiological Phenomena/physiology , Female , Image Processing, Computer-Assisted , Immunohistochemistry , Larva , Male , Microscopy, Electron , Mutation/physiology , Receptors, Presynaptic/physiology , Signal Transduction/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology
18.
Am J Nephrol ; 38(5): 430-44, 2013.
Article in English | MEDLINE | ID: mdl-24247724

ABSTRACT

BACKGROUND/AIMS: The mechanism underlying angiotensin II (AngII)-promoted podocyte apoptosis has not been established. IQ domain GTPase-activating protein 1 (IQGAP1) is a scaffolding protein of the mitogen-activated protein kinases (MAPK) signaling pathway, and plays a significant role in apoptosis. The present study evaluates the role of IQGAP1 in AngII-induced podocyte apoptosis. METHODS: We randomly assigned 36 male Wistar rats to a normal saline-infused group, an AngII-infused group, or a normal control group, and measured podocyte apoptosis by the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay and transmission electron microscopic analysis. In addition, we exposed differentiated mouse podocytes to AngII and then assessed apoptosis by flow cytometry and Hoechst-33258 staining. Expression of IQGAP1 was measured by Western blotting, real-time PCR and immunofluorescence assay in vivo and in vitro. IQGAP1 siRNA and MAPK pathway inhibitors were further introduced to investigate the role of IQGAP1 and MAPK signaling in the process. Coimmunoprecipitation was used to evaluate the interaction between ERK1/2 and IQGAP1. RESULTS: AngII promoted podocyte apoptosis in vivo and in vitro. IQGAP1 had a linear distribution along the capillary loops of glomeruli in vivo, and was in the cellular membrane and cytoplasm of cultured podocytes. AngII stimulated IQGAP1 expression and increased phosphorylation of P38, JNK, and ERK1/2. Knockdown of IQGAP1 with siRNA prevented AngII-induced apoptosis of podocytes and reduced AngII-induced phosphorylation of ERK1/2, but not that of P38, JNK. This was accompanied by a reduced interaction between ERK1/2 and IQGAP1. CONCLUSION: IQGAP1 contributes to AngII-induced apoptosis of podocytes by interacting with the ERK1/2 signaling protein.


Subject(s)
Angiotensin II/metabolism , Apoptosis , Gene Expression Regulation , MAP Kinase Signaling System , Podocytes/cytology , ras GTPase-Activating Proteins/physiology , Animals , Disease Progression , Kidney Glomerulus/drug effects , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Rats , Rats, Wistar
19.
J Immunol ; 187(1): 151-63, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21646295

ABSTRACT

Activation of the Ras small GTP-binding protein is necessary for normal T cell development and function. However, it is unknown which Ras GTPase-activating proteins (RasGAPs) inactivate Ras in T cells. We used a T cell-specific RASA1-deficient mouse model to investigate the role of the p120 RasGAP (RASA1) in T cells. Death of CD4(+)CD8(+) double-positive thymocytes was increased in RASA1-deficient mice. Despite this finding, on an MHC class II-restricted TCR transgenic background, evidence was obtained for increased positive selection of thymocytes associated with augmented activation of the Ras-MAPK pathway. In the periphery, RASA1 was found to be dispensable as a regulator of Ras-MAPK activation and T cell functional responses induced by full agonist peptides. However, numbers of naive T cells were substantially reduced in RASA1-deficient mice. Loss of naive T cells in the absence of RASA1 could be attributed in part to impaired responsiveness to the IL-7 prosurvival cytokine. These findings reveal an important role for RASA1 as a regulator of double-positive survival and positive selection in the thymus as well as naive T cell survival in the periphery.


Subject(s)
Resting Phase, Cell Cycle/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/immunology , p120 GTPase Activating Protein/physiology , ras GTPase-Activating Proteins/physiology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Resting Phase, Cell Cycle/genetics , T-Lymphocyte Subsets/metabolism , Thymus Gland/metabolism , p120 GTPase Activating Protein/deficiency , p120 GTPase Activating Protein/genetics , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
20.
Proc Natl Acad Sci U S A ; 107(6): 2485-90, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20080667

ABSTRACT

A single nucleotide polymorphism in the DAB2IP gene is associated with risk of aggressive prostate cancer (PCa), and loss of DAB2IP expression is frequently detected in metastatic PCa. However, the functional role of DAB2IP in PCa remains unknown. Here, we show that the loss of DAB2IP expression initiates epithelial-to-mesenchymal transition (EMT), which is visualized by repression of E-cadherin and up-regulation of vimentin in both human normal prostate epithelial and prostate carcinoma cells as well as in clinical prostate-cancer specimens. Conversely, restoring DAB2IP in metastatic PCa cells reversed EMT. In DAB2IP knockout mice, prostate epithelial cells exhibited elevated mesenchymal markers, which is characteristic of EMT. Using a human prostate xenograft-mouse model, we observed that knocking down endogenous DAB2IP in human carcinoma cells led to the development of multiple lymph node and distant organ metastases. Moreover, we showed that DAB2IP functions as a scaffold protein in regulating EMT by modulating nuclear beta-catenin/T-cell factor activity. These results show the mechanism of DAB2IP in EMT and suggest that assessment of DAB2IP may provide a prognostic biomarker and potential therapeutic target for PCa metastasis.


Subject(s)
Epithelial Cells/pathology , Mesoderm/pathology , Prostatic Neoplasms/pathology , ras GTPase-Activating Proteins/physiology , Animals , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Cell Line , Cell Line, Tumor , Cell Movement , Epithelial Cells/metabolism , Gene Expression , Humans , Immunohistochemistry , Male , Mesoderm/metabolism , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , TCF Transcription Factors/metabolism , Transfection , Transplantation, Heterologous , Vimentin/genetics , Vimentin/metabolism , beta Catenin/metabolism , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL