Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 37(19)2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28694330

ABSTRACT

The activation of Raf kinases by the small GTPase Ras requires two major sets of phosphorylations. One set lies within the activation loop, and the other lies within the N-terminal acidic region (N region). In the most abundant isoform of Raf, C-Raf, N-region phosphorylations occur on serine 338 (S338) and tyrosine 341 (Y341) and are thought to provide allosteric activation of the Raf dimer. We show that the phosphorylations of these N-region sites does not require C-Raf dimerization, but rather, they precede dimerization. One of these phosphorylations (phospho-Y341) is required for C-Raf dimerization, and this action can be replicated by phosphomimetic mutants both in vivo and in vitro The role of the phosphorylation of Y341 in promoting Raf dimerization is distinct from its well-known function in facilitating S338 phosphorylation. In Ras mutant pancreatic cancer cell lines, the phosphorylation and dimerization of C-Raf are basally elevated. Dimerization is thought to contribute to their elevated growth rate through their activation of the mitogen-activated protein (MAP) kinase (extracellular signal-regulated kinase [ERK]) signaling cascade. Blocking the tyrosine phosphorylation of C-Raf with Src family inhibitors blocks growth, basal dimerization, and ERK activation in these cells. We suggest that the kinases mediating C-Raf Y341 phosphorylation are potential candidate drug targets in selected Ras-dependent cancers.

2.
Cell Rep ; 19(3): 617-629, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423323

ABSTRACT

Protein kinase A (PKA) has diverse functions in neurons. At rest, the subcellular localization of PKA is controlled by A-kinase anchoring proteins (AKAPs). However, the dynamics of PKA upon activation remain poorly understood. Here, we report that elevation of cyclic AMP (cAMP) in neuronal dendrites causes a significant percentage of the PKA catalytic subunit (PKA-C) molecules to be released from the regulatory subunit (PKA-R). Liberated PKA-C becomes associated with the membrane via N-terminal myristoylation. This membrane association does not require the interaction between PKA-R and AKAPs. It slows the mobility of PKA-C and enriches kinase activity on the membrane. Membrane-residing PKA substrates are preferentially phosphorylated compared to cytosolic substrates. Finally, the myristoylation of PKA-C is critical for normal synaptic function and plasticity. We propose that activation-dependent association of PKA-C renders the membrane a unique PKA-signaling compartment. Constrained mobility of PKA-C may synergize with AKAP anchoring to determine specific PKA function in neurons.


Subject(s)
Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Myristic Acid/metabolism , A Kinase Anchor Proteins/metabolism , Animals , Cytosol/metabolism , Enzyme Activation , HEK293 Cells , Humans , Neuronal Plasticity , Neurons/metabolism , Phosphorylation , Protein Binding , Rats , Substrate Specificity , Synapses/metabolism
3.
J Biol Chem ; 292(4): 1449-1461, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28003362

ABSTRACT

Cyclic adenosine monophosphate (cAMP) is an important mediator of hormonal stimulation of cell growth and differentiation through its activation of the extracellular signal-regulated kinase (ERK) cascade. Two small G proteins, Ras and Rap1 have been proposed to mediate this activation. Using HEK293 cells as a model system, we have recently shown that both Ras and Rap1 are required for cAMP signaling to ERKs. However, cAMP-dependent Ras signaling to ERKs is transient and rapidly terminated by PKA phosphorylation of the Raf isoforms C-Raf and B-Raf. In contrast, cAMP-dependent Rap1 signaling to ERKs and Rap1 is potentiated by PKA. We show that this is due to sustained binding of B-Raf to Rap1. One of the targets of PKA is Rap1 itself, directly phosphorylating Rap1a on serine 180 and Rap1b on serine 179. We show that these phosphorylations create potential binding sites for the adaptor protein 14-3-3 that links Rap1 to the scaffold protein KSR. These results suggest that Rap1 activation of ERKs requires PKA phosphorylation and KSR binding. Because KSR and B-Raf exist as heterodimers within the cell, this binding also brings B-Raf to Rap1, allowing Rap1 to couple to ERKs through B-Raf binding to Rap1 independently of its Ras-binding domain.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lysine-tRNA Ligase/metabolism , rap GTP-Binding Proteins/metabolism , Animals , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , HEK293 Cells , Humans , Lysine-tRNA Ligase/genetics , Mice , Mice, Knockout , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , rap GTP-Binding Proteins/genetics
4.
Plant J ; 89(5): 940-956, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27885735

ABSTRACT

2,4-Dichlorophenoxyacetic acid (2,4-D), a functional analogue of auxin, is used as an exogenous source of auxin as it evokes physiological responses like the endogenous auxin, indole-3-acetic acid (IAA). Previous molecular analyses of the auxin response pathway revealed that IAA and 2,4-D share a common mode of action to elicit downstream physiological responses. However, recent findings with 2,4-D-specific mutants suggested that 2,4-D and IAA might also use distinct pathways to modulate root growth in Arabidopsis. Using genetic and cellular approaches, we demonstrate that the distinct effects of 2,4-D and IAA on actin filament organization partly dictate the differential responses of roots to these two auxin analogues. 2,4-D but not IAA altered the actin structure in long-term and short-term assays. Analysis of the 2,4-D-specific mutant aar1-1 revealed that small acidic protein 1 (SMAP1) functions positively to facilitate the 2,4-D-induced depolymerization of actin. The ubiquitin proteasome mutants tir1-1 and axr1-12, which show enhanced resistance to 2,4-D compared with IAA for inhibition of root growth, were also found to have less disrupted actin filament networks after 2,4-D exposure. Consistently, a chemical inhibitor of the ubiquitin proteasome pathway mitigated the disrupting effects of 2,4-D on the organization of actin filaments. Roots of the double mutant aar1-1 tir1-1 also showed enhanced resistance to 2,4-D-induced inhibition of root growth and actin degradation compared with their respective parental lines. Collectively, these results suggest that the effects of 2,4-D on actin filament organization and root growth are mediated through synergistic interactions between SMAP1 and SCFTIR1 ubiquitin proteasome components.


Subject(s)
2,4-Dichlorophenoxyacetic Acid/metabolism , Actins/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Ubiquitin/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Plant Roots/genetics , Plant Roots/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
5.
J Biol Chem ; 291(41): 21584-21595, 2016 Oct 07.
Article in English | MEDLINE | ID: mdl-27531745

ABSTRACT

Cyclic adenosine monophosphate (cAMP) is an important mediator of hormonal stimulation of cell growth and differentiation through its activation of the extracellular signal-regulated kinase (ERK) cascade. Two small G proteins, Ras and Rap1, have been proposed to mediate this activation, with either Ras or Rap1 acting in distinct cell types. Using Hek293 cells, we show that both Ras and Rap1 are required for cAMP signaling to ERKs. The roles of Ras and Rap1 were distinguished by their mechanism of activation, dependence on the cAMP-dependent protein kinase (PKA), and the magnitude and kinetics of their effects on ERKs. Ras was required for the early portion of ERK activation by cAMP and was activated independently of PKA. Ras activation required the Ras/Rap guanine nucleotide exchange factor (GEF) PDZ-GEF1. Importantly, this action of PDZ-GEF1 was disrupted by mutation within its putative cyclic nucleotide-binding domain within PDZ-GEF1. Compared with Ras, Rap1 activation of ERKs was of longer duration. Rap1 activation was dependent on PKA and required Src family kinases and the Rap1 exchanger C3G. This is the first report of a mechanism for the cooperative actions of Ras and Rap1 in cAMP activation of ERKs. One physiological role for the sustained activation of ERKs is the transcription and stabilization of a range of transcription factors, including c-FOS. We show that the induction of c-FOS by cAMP required both the early and sustained phases of ERK activation, requiring Ras and Rap1, as well as for each of the Raf isoforms, B-Raf and C-Raf.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Telomere-Binding Proteins/metabolism , Animals , Cattle , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Shelterin Complex , Telomere-Binding Proteins/genetics
6.
Cell Signal ; 28(3): 177-189, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26739108

ABSTRACT

Extracellular signal-regulated kinases (ERKs) play important roles in proliferation, differentiation and gene expression. In our previous study, we demonstrated that both ERK5 and ERK1/2 were responsible for neurite outgrowth and tyrosine hydroxylase (TH) expression in rat pheochromocytoma cells (PC12) (J Biol Chem 284, 23,564-23,573, 2009). However, the functional differences between ERK5 and ERK1/2 signaling in neural differentiation remain unclear. In the present study, we show that ERK5, but not ERK1/2 regulates TH levels in rat sympathetic neurons. Furthermore, microarray analysis performed in PC12 cells using ERK5 and ERK1/2-specific inhibitors, identified ankyrin repeat domain 1 (ankrd1) as an ERK5-dependent and ERK1/2-independent gene. Here, we report a novel role of the ERK5/ankrd1 signaling in regulating TH levels and catecholamine biosynthesis. Ankrd1 mRNA was induced by nerve growth factor in time- and concentration-dependent manners. TH levels were reduced by ankrd1 knockdown with no changes in the mRNA levels, suggesting that ankrd1 was involved in stabilization of TH protein. Interestingly, ubiquitination of TH was enhanced and catecholamine biosynthesis was reduced by ankrd1 knockdown. Finally, we examined the relationship of ERK5 to TH levels in human adrenal pheochromocytomas. Whereas TH levels were correlated with ERK5 levels in normal adrenal medullas, ERK5 was down-regulated and TH was up-regulated in pheochromocytomas, indicating that TH levels are regulated by alternative mechanisms in tumors. Taken together, ERK5 signaling is required for catecholamine biosynthesis during neural differentiation, in part to induce ankrd1, and to maintain appropriate TH levels. This pathway is disrupted in pathological conditions.


Subject(s)
Catecholamines/biosynthesis , Mitogen-Activated Protein Kinase 7/metabolism , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Adolescent , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Catecholamines/analysis , Chromatography, High Pressure Liquid , Down-Regulation/drug effects , Female , Humans , Male , Microscopy, Fluorescence , Middle Aged , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Nerve Growth Factor/pharmacology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , PC12 Cells , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , RNA Interference , RNA, Small Interfering/metabolism , Rats , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction/drug effects , Tandem Mass Spectrometry , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation/drug effects
7.
Viruses ; 7(3): 1271-83, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25793940

ABSTRACT

The smaller tea tortrix, Adoxophyes honmai (Lepidoptera: Tortricidae), is an economically important pest of tea in Japan. Previous work showed that a fast-killing nucleopolyhedrovirus (NPV) isolated from A. orana (AdorNPV) and a slow-killing NPV isolated from A. honmai (AdhoNPV) are both infectious to A. honmai larvae. Field application of these different NPVs was conducted against an A. honmai larval population in tea plants, and the control efficacy and transmission rate of the two NPVs were compared. The slow-killing AdhoNPV showed lower field efficacy, in terms of preventing damage caused by A. honmai larvae against the tea plants, than the fast-killing AdorNPV. However, AdhoNPV had a significantly higher horizontal transmission rate than AdorNPV. These results show that AdorNPV is suitable as an inundative agent, while AdhoNPV is an appropriate inoculative agent.


Subject(s)
Lepidoptera/physiology , Lepidoptera/virology , Nucleopolyhedroviruses/growth & development , Nucleopolyhedroviruses/isolation & purification , Animals , Japan , Larva/physiology , Larva/virology , Pest Control, Biological/methods , Survival Analysis , Tea/parasitology
8.
J Biol Chem ; 288(39): 27712-23, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23946483

ABSTRACT

The small G protein Rap1 can mediate "inside-out signaling" by recruiting effectors to the plasma membrane that signal to pathways involved in cell adhesion and cell migration. This action relies on the membrane association of Rap1, which is dictated by post-translational prenylation as well as by a stretch of basic residues within its carboxyl terminus. One feature of this stretch of acidic residues is that it lies adjacent to a functional phosphorylation site for the cAMP-dependent protein kinase PKA. This phosphorylation has two effects on Rap1 action. One, it decreases the level of Rap1 activity as measured by GTP loading and the coupling of Rap1 to RapL, a Rap1 effector that couples Rap1 GTP loading to integrin activation. Two, it destabilizes the membrane localization of Rap1, promoting its translocation into the cytoplasm. These two actions, decreased GTP loading and decreased membrane localization, are related, as the translocation of Rap1-GTP into the cytoplasm is associated with its increased GTP hydrolysis and inactivation. The consequences of this phosphorylation in Rap1-dependent cell adhesion and cell migration were also examined. Active Rap1 mutants that lack this phosphorylation site had a minimal effect on cell adhesion but strongly reduced cell migration, when compared with an active Rap1 mutant that retained the phosphorylation site. This suggests that optimal cell migration is associated with cycles of Rap1 activation, membrane egress, and inactivation, and requires the regulated phosphorylation of Rap1 by PKA.


Subject(s)
Cell Movement , Cyclic AMP-Dependent Protein Kinases/chemistry , rap1 GTP-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Cattle , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Enzyme Activation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Microscopy, Confocal , Molecular Sequence Data , Mutation , Phosphorylation , Signal Transduction
9.
J Biol Chem ; 288(38): 27646-27657, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23893412

ABSTRACT

The small G protein Ras regulates proliferation through activation of the mitogen-activated protein (MAP) kinase (ERK) cascade. The first step of Ras-dependent activation of ERK signaling is Ras binding to members of the Raf family of MAP kinase kinase kinases, C-Raf and B-Raf. Recently, it has been reported that in melanoma cells harboring oncogenic Ras mutations, B-Raf does not bind to Ras and does not contribute to basal ERK activation. For other types of Ras-mutant tumors, the relative contributions of C-Raf and B-Raf are not known. We examined non-melanoma cancer cell lines containing oncogenic Ras mutations and express both C-Raf and B-Raf isoforms, including the lung cancer cell line H1299 cells. Both B-Raf and C-Raf were constitutively bound to oncogenic Ras and contributed to Ras-dependent ERK activation. Ras binding to B-Raf and C-Raf were both subject to inhibition by the cAMP-dependent protein kinase PKA. cAMP inhibited the growth of H1299 cells and Ras-dependent ERK activation via PKA. PKA inhibited the binding of Ras to both C-Raf and B-Raf through phosphorylations of C-Raf at Ser-259 and B-Raf at Ser-365, respectively. These studies demonstrate that in non-melanocytic Ras-mutant cancer cells, Ras signaling to B-Raf is a significant contributor to ERK activation and that the B-Raf pathway, like that of C-Raf, is a target for inhibition by PKA. We suggest that cAMP and hormones coupled to cAMP may prove useful in dampening the effects of oncogenic Ras in non-melanocytic cancer cells through PKA-dependent actions on B-Raf as well as C-Raf.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lung Neoplasms/metabolism , Oncogene Protein p21(ras)/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction , Cell Line, Tumor , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Oncogene Protein p21(ras)/genetics , Phosphorylation/genetics , Protein Binding/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism
10.
Int Immunol ; 25(4): 259-69, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23334952

ABSTRACT

The duration of signaling through the MAP kinase (or ERK pathway) cascade has been implicated in thymic development, particularly positive and negative selection. In T cells, two isoforms of the MAP kinase kinase kinase Raf function to transmit signals from the T-cell receptor to ERK: C-Raf and B-Raf. In this study, we conditionally ablated B-Raf expression within thymocytes to assess the effects on ERK activation and thymocyte development. The complete loss of B-Raf is accompanied by a dramatic loss of ERK activation in both the double positive (DP) and single positive (SP) thymocytes, as well as peripheral splenocytes. There was a significant decrease in the cellularity of KO thymi, largely due to a loss of pre-selected DP cells, a decrease in DP cells undergoing positive selection, and a defect in SP maturation. B-Raf plays significant roles in survival of DP thymocytes and function of SP cells in the periphery. Surprisingly, we saw no effect of B-Raf deficiency on negative selection of autoreactive SP thymocytes, despite the greatly reduced ERK activation in these cells.


Subject(s)
Proto-Oncogene Proteins B-raf/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation/genetics , Cell Survival/genetics , Cells, Cultured , Clonal Deletion/genetics , Enzyme Activation/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/immunology
11.
Proc Natl Acad Sci U S A ; 110(1): 312-7, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23248303

ABSTRACT

The ASPP2 (also known as 53BP2L) tumor suppressor is a proapoptotic member of a family of p53 binding proteins that functions in part by enhancing p53-dependent apoptosis via its C-terminal p53-binding domain. Mounting evidence also suggests that ASPP2 harbors important nonapoptotic p53-independent functions. Structural studies identify a small G protein Ras-association domain in the ASPP2 N terminus. Because Ras-induced senescence is a barrier to tumor formation in normal cells, we investigated whether ASPP2 could bind Ras and stimulate the protein kinase Raf/MEK/ERK signaling cascade. We now show that ASPP2 binds to Ras-GTP at the plasma membrane and stimulates Ras-induced signaling and pERK1/2 levels via promoting Ras-GTP loading, B-Raf/C-Raf dimerization, and C-Raf phosphorylation. These functions require the ASPP2 N terminus because BBP (also known as 53BP2S), an alternatively spliced ASPP2 isoform lacking the N terminus, was defective in binding Ras-GTP and stimulating Raf/MEK/ERK signaling. Decreased ASPP2 levels attenuated H-RasV12-induced senescence in normal human fibroblasts and neonatal human epidermal keratinocytes. Together, our results reveal a mechanism for ASPP2 tumor suppressor function via direct interaction with Ras-GTP to stimulate Ras-induced senescence in nontransformed human cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cellular Senescence/physiology , MAP Kinase Signaling System/physiology , ras Proteins/metabolism , Dimerization , Fibroblasts , HCT116 Cells , Humans , Keratinocytes , Microscopy, Fluorescence , Phosphorylation , Plasmids/genetics , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , RNA, Small Interfering/genetics
12.
Mol Cell Biol ; 30(16): 3956-69, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20547757

ABSTRACT

Epac1 (exchange protein directly activated by cyclic AMP [cAMP]) couples intracellular cAMP to the activation of Rap1, a Ras family GTPase that regulates cell adhesion, proliferation, and differentiation. Using mass spectrometry, we identified the small G protein Ran and Ran binding protein 2 (RanBP2) as potential binding partners of Epac1. Ran is a small G protein best known for its role in nuclear transport and can be found at the nuclear pore through its interaction with RanBP2. Here we demonstrate that Ran-GTP and Epac1 interact with each other in vivo and in vitro. This binding requires a previously uncharacterized Ras association (RA) domain in Epac1. Surprisingly, the interaction of Epac1 with Ran is necessary for the efficient activation of Rap1 by Epac1. We propose that Ran and RanBP2 anchor Epac1 to the nuclear pore, permitting cAMP signals to activate Rap1 at the nuclear envelope.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Nuclear Envelope/metabolism , ran GTP-Binding Protein/metabolism , rap1 GTP-Binding Proteins/metabolism , Base Sequence , Cyclic AMP/metabolism , Enzyme Activation , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , In Vitro Techniques , Models, Biological , Nuclear Pore/metabolism , Protein Interaction Domains and Motifs , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , ran GTP-Binding Protein/chemistry , ran GTP-Binding Protein/genetics , rap1 GTP-Binding Proteins/genetics
13.
Plant Cell ; 22(6): 1762-76, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20562236

ABSTRACT

In the root, the transport of auxin from the tip to the elongation zone, referred to here as shootward, governs gravitropic bending. Shootward polar auxin transport, and hence gravitropism, depends on the polar deployment of the PIN-FORMED auxin efflux carrier PIN2. In Arabidopsis thaliana, PIN2 has the expected shootward localization in epidermis and lateral root cap; however, this carrier is localized toward the root tip (rootward) in cortical cells of the meristem, a deployment whose function is enigmatic. We use pharmacological and genetic tools to cause a shootward relocation of PIN2 in meristematic cortical cells without detectably altering PIN2 polarization in other cell types or PIN1 polarization. This relocation of cortical PIN2 was negatively regulated by the membrane trafficking factor GNOM and by the regulatory A1 subunit of type 2-A protein phosphatase (PP2AA1) but did not require the PINOID protein kinase. When GNOM was inhibited, PINOID abundance increased and PP2AA1 was partially immobilized, indicating both proteins are subject to GNOM-dependent regulation. Shootward PIN2 specifically in the cortex was accompanied by enhanced shootward polar auxin transport and by diminished gravitropism. These results demonstrate that auxin flow in the root cortex is important for optimal gravitropic response.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/physiology , Gravitropism , Meristem/physiology , Plant Roots/physiology , Protein Phosphatase 2/metabolism , Biological Transport , Brefeldin A/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Indoleacetic Acids/metabolism
14.
J Biol Chem ; 284(35): 23564-73, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19581298

ABSTRACT

Extracellular signal-regulated kinases (ERKs) play important physiological roles in proliferation, differentiation, and gene expression. ERK5 is approximately twice the size of ERK1/2, and its amino-terminal half contains the kinase domain that shares homology with ERK1/2 and TEY activation motif, whereas the carboxyl-terminal half is unique. In this study, we examined a physiological role of ERK5 in rat pheochromocytoma cells (PC12), comparing it with ERK1/2. Nerve growth factor (NGF) induced phosphorylation of both ERK5 and ERK1/2, whereas the cAMP analog dibutyryl cAMP (Bt(2)cAMP) caused only ERK1/2 phosphorylation. U0126, at 30 mum, that blocks ERK1/2 signaling selectively attenuated neurite outgrowth induced by NGF and Bt(2)cAMP, but BIX02188 and BIX02189, at 30 mum, that block ERK5 signaling and an ERK5 dominant-negative mutant suppressed only NGF-induced neurite outgrowth. Next, we examined the expression of tyrosine hydroxylase, a rate-limiting enzyme of catecholamine biosynthesis. Both NGF and Bt(2)cAMP increased tyrosine hydroxylase gene promoter activity in an ERK1/2-dependent manner but was ERK5-independent. However, when both ERK5 and ERK1/2 signalings were inhibited, tyrosine hydroxylase protein up-regulation by NGF and Bt(2)cAMP was abolished, because of the loss of stabilization of tyrosine hydroxylase protein by ERK5. Taking these results together, ERK5 is involved in neurite outgrowth and stabilization of tyrosine hydroxylase in PC12 cells, and ERK5, along with ERK1/2, plays essential roles in the neural differentiation process.


Subject(s)
Mitogen-Activated Protein Kinase 7/metabolism , Nerve Growth Factor/metabolism , Neurites/enzymology , Neurogenesis , Tyrosine 3-Monooxygenase/metabolism , Animals , Mitogen-Activated Protein Kinase 7/genetics , Nerve Growth Factor/genetics , PC12 Cells , Phosphorylation , Rats , Signal Transduction , Tyrosine 3-Monooxygenase/genetics
15.
Mol Cell Biol ; 28(23): 7109-25, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18824540

ABSTRACT

Exchange proteins activated by cAMP (cyclic AMP) 2 (Epac2) is a guanine nucleotide exchange factor for Rap1, a small G protein involved in many cellular functions, including cell adhesion, differentiation, and exocytosis. Epac2 interacts with Ras-GTP via a Ras association (RA) domain. Previous studies have suggested that the RA domain was dispensable for Epac2 function. Here we show for the first time that Ras and cAMP regulate Epac2 function in a parallel fashion and the Ras-Epac2 interaction is required for the cAMP-dependent activation of endogenous Rap1 by Epac2. The mechanism for this requirement is not allosteric activation of Epac2 by Ras but the compartmentalization of Epac2 on the Ras-containing membranes. A computational modeling is consistent with this compartmentalization being a function of both the level of Ras activation and the affinity between Ras and Epac2. In PC12 cells, a well-established model for sympathetic neurons, the Epac2 signaling is coupled to activation of mitogen-activated protein kinases and contributes to neurite outgrowth. Taken together, the evidence shows that Epac2 is not only a cAMP sensor but also a bona fide Ras effector. Coincident detection of both cAMP and Ras signals is essential for Epac2 to activate Rap1 in a temporally and spatially controlled manner.


Subject(s)
Carrier Proteins/metabolism , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Cell Compartmentation , Cell Line , Humans , MAP Kinase Signaling System , Mice , Neurites , Neurons/ultrastructure , Protein Binding
16.
Int J Oncol ; 29(2): 413-21, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16820884

ABSTRACT

Familial adenomatous polyposis (FAP) is an autosomal dominant familial cancer syndrome caused by germline mutations of the tumor suppressor adenomatous polyposis coli (APC) gene. Heterozygous apc mutations have been identified in the majority of classical FAP patients who develop more than 100 colorectal adenomas. However, classical FAP patients often fail to display germline APC mutations detectable by routine mutation analysis. These apparently mutation-negative cases may be caused by heterozygous large genomic deletions. In the present study, FAP patients who showed no APC germline mutation detectable by the protein truncation assay and direct sequencing of protein coding exons were screened for APC gene deletion by a gene dose assay based on double competitive polymerase chain reaction. Gene dosage measurements within exon 15 of the APC gene identified two patients with gene deletion and one with possible gene duplication among 41 apparently mutation-negative patients. The deleted sequences in the two patients were determined by fine gene dose mapping around the APC gene and nucleotide sequencing of the deletion breakpoints. They were approximately 435-kilobase pair (kb) and 737-kb regions including the whole APC gene and flanked by a 4-base pair repeat and LINE-1 repetitive sequences, respectively. The chimeric LINE-1 element created at the breakpoint in the latter case also contained a short sequence derived from another LINE-1 element, suggesting a complex unequal homologous recombination event. These findings indicate that this gene dose assay is a useful technique to detect large gene deletions of the APC gene and to determine their genomic breakpoints.


Subject(s)
Adenomatous Polyposis Coli Protein/biosynthesis , Adenomatous Polyposis Coli/genetics , Genes, APC , Genetic Predisposition to Disease , Base Sequence , Exons , Gene Deletion , Heterozygote , Humans , Microsatellite Repeats , Models, Genetic , Molecular Sequence Data , Mutation , Recombination, Genetic
17.
J Biol Chem ; 280(32): 28927-35, 2005 Aug 12.
Article in English | MEDLINE | ID: mdl-15944154

ABSTRACT

Molecular diversity through alternative splicing is important for cellular function and development. However, little is known about the factors that regulate alternative splicing. Here we demonstrate that one isoform of coactivator-associated arginine methyltransferase 1 (named CARM1-v3) associates with the U1 small nuclear RNP-specific protein U1C and affects 5' splice site selection of the pre-mRNA splicing. CARM1-v3 was generated by the retention of introns 15 and 16 of the primary transcript of CARM1. Its deduced protein lacks the C-terminal domain of the major isoform of CARM1 and instead has v3-specific sequences at the C terminus. CARM1-v3, but not the other isoforms, strongly stimulates a shift to the distal 5' splice site of the pre-mRNA when the adenoviral E1A minigene is used as a reporter and enhances the exon skips in the CD44 reporter. A CARM1-v3 mutant lacking the v3-specific sequences completely lost the ability to regulate the alternative splicing patterns. In addition, CARM1-v3 shows tissue-specific expression patterns distinct from those of the other isoforms. These results suggest that the transcriptional coactivator can affect the splice site decision in an isoform-specific manner.


Subject(s)
Protein-Arginine N-Methyltransferases/physiology , Adenovirus E1A Proteins/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western , COS Cells , Cell Line , DNA Methylation , DNA, Complementary/metabolism , Gene Library , Genes, Reporter , Humans , Hyaluronan Receptors/metabolism , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Methyltransferases/metabolism , Models, Genetic , Molecular Sequence Data , Plasmids/metabolism , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/metabolism , RNA/metabolism , RNA Splicing , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoprotein, U1 Small Nuclear/chemistry , Sequence Homology, Amino Acid , Transcriptional Activation , Transfection , Two-Hybrid System Techniques , Ultraviolet Rays
18.
Mol Cell Biol ; 24(15): 6569-80, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15254225

ABSTRACT

MEN1 is a tumor suppressor gene that is responsible for multiple endocrine neoplasia type 1 (MEN1) and that encodes a 610-amino-acid protein, called menin. While the majority of germ line mutations identified in MEN1 patients are frameshift and nonsense mutations resulting in truncation of the menin protein, various missense mutations have been identified whose effects on menin activity are unclear. For this study, we analyzed a series of menin proteins with single amino acid alterations and found that all of the MEN1-causing missense mutations tested led to greatly diminished levels of the affected proteins in comparison with wild-type and benign polymorphic menin protein levels. We demonstrate here that the reduced levels of the mutant proteins are due to rapid degradation via the ubiquitin-proteasome pathway. Furthermore, the mutants, but not wild-type menin, interact both with the molecular chaperone Hsp70 and with the Hsp70-associated ubiquitin ligase CHIP, and the overexpression of CHIP promotes the ubiquitination of the menin mutants in vivo. These findings reveal that MEN1-causing missense mutations lead to a loss of function of menin due to enhanced proteolytic degradation, which may be a common mechanism for inactivating tumor suppressor gene products in familial cancer.


Subject(s)
Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Multiple Endocrine Neoplasia Type 1/genetics , Mutation, Missense , Proto-Oncogene Proteins/genetics , Ubiquitin/metabolism , Amino Acids/chemistry , Animals , Blotting, Northern , Blotting, Western , COS Cells , Cell Line , Chromatin/metabolism , Genes, Tumor Suppressor , HSP70 Heat-Shock Proteins/metabolism , Humans , Mass Spectrometry , Mice , Microscopy, Fluorescence , Mutation , Peptides/chemistry , Plasmids/metabolism , Polymorphism, Genetic , Precipitin Tests , Proteasome Endopeptidase Complex , RNA/metabolism , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...