Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Nanotechnol ; 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38491184

ABSTRACT

Multivalent presentation of ligands often enhances receptor activation and downstream signalling. DNA origami offers a precise nanoscale spacing of ligands, a potentially useful feature for therapeutic nanoparticles. Here we use a square-block DNA origami platform to explore the importance of the spacing of CpG oligonucleotides. CpG engages Toll-like receptors and therefore acts to activate dendritic cells. Through in vitro cell culture studies and in vivo tumour treatment models, we demonstrate that square blocks induce Th1 immune polarization when CpG is spaced at 3.5 nm. We observe that this DNA origami vaccine enhances DC activation, antigen cross-presentation, CD8 T-cell activation, Th1-polarized CD4 activation and natural-killer-cell activation. The vaccine also effectively synergizes with anti-PD-L1 for improved cancer immunotherapy in melanoma and lymphoma models and induces long-term T-cell memory. Our results suggest that DNA origami may serve as a platform for controlling adjuvant spacing and co-delivering antigens in vaccines.

2.
Nat Nanotechnol ; 16(12): 1362-1370, 2021 12.
Article in English | MEDLINE | ID: mdl-34675411

ABSTRACT

Decoding the identity of biomolecules from trace samples is a longstanding goal in the field of biotechnology. Advances in DNA analysis have substantially affected clinical practice and basic research, but corresponding developments for proteins face challenges due to their relative complexity and our inability to amplify them. Despite progress in methods such as mass spectrometry and mass cytometry, single-molecule protein identification remains a highly challenging objective. Towards this end, we combine DNA nanotechnology with single-molecule force spectroscopy to create a mechanically reconfigurable DNA nanoswitch caliper capable of measuring multiple coordinates on single biomolecules with atomic resolution. Using optical tweezers, we demonstrate absolute distance measurements with ångström-level precision for both DNA and peptides, and using multiplexed magnetic tweezers, we demonstrate quantification of relative abundance in mixed samples. Measuring distances between DNA-labelled residues, we perform single-molecule fingerprinting of synthetic and natural peptides, and show discrimination, within a heterogeneous population, between different posttranslational modifications. DNA nanoswitch calipers are a powerful and accessible tool for characterizing distances within nanoscale complexes that will enable new applications in fields such as single-molecule proteomics.


Subject(s)
DNA/chemistry , Nanotechnology , Single Molecule Imaging , Amino Acid Sequence , Calibration , Peptides/chemistry , Protein Processing, Post-Translational , Reproducibility of Results , Spectrum Analysis
3.
J Biol Chem ; 296: 100108, 2021.
Article in English | MEDLINE | ID: mdl-33219128

ABSTRACT

The retinoblastoma tumor suppressor protein (RB) plays an important role in biological processes such as cell cycle control, DNA damage repair, epigenetic regulation, and genome stability. The canonical model of RB regulation is that cyclin-CDKs phosphorylate and render RB inactive in late G1/S, promoting entry into S phase. Recently, monophosphorylated RB species were described to have distinct cell-cycle-independent functions, suggesting that a phosphorylation code dictates diversity of RB function. However, a biologically relevant, functional role of RB phosphorylation at non-CDK sites has remained elusive. Here, we investigated S838/T841 dual phosphorylation, its upstream stimulus, and downstream functional output. We found that mimicking T-cell receptor activation in Jurkat leukemia cells induced sequential activation of downstream kinases including p38 MAPK and RB S838/T841 phosphorylation. This signaling pathway disrupts RB and condensin II interaction with chromatin. Using cells expressing a WT or S838A/T841A mutant RB fragment, we present evidence that deficiency for this phosphorylation event prevents condensin II release from chromatin.


Subject(s)
Chromatin/metabolism , Retinoblastoma Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic/genetics , Humans , Multiprotein Complexes/metabolism , Mutation/genetics , Phosphorylation/genetics , Phosphorylation/physiology , Receptors, Antigen, T-Cell/metabolism
4.
J Am Chem Soc ; 139(10): 3767-3773, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28207247

ABSTRACT

The synthesis of complex protein-based bioconjugates has been facilitated greatly by recent developments in chemoselective methods for biomolecular modification. The oxidative coupling of o-aminophenols or catechols with aniline functional groups is chemoselective, mild, and rapid; however, the oxidatively sensitive nature of the electron-rich aromatics and the paucity of commercial sources pose some obstacles to the general use of these reactive strategies. Herein, we identify o-methoxyphenols as air-stable, commercially available derivatives that undergo efficient oxidative couplings with anilines in the presence of periodate as oxidant. Mechanistic considerations informed the development of a preoxidation protocol that can greatly reduce the amount of periodate necessary for effective coupling. The stability and versatility of these reagents was demonstrated through the synthesis of complex protein-protein bioconjugates using a site-selective heterobifunctional cross-linker comprising both o-methoxyphenol and 2-pyridinecarboxaldehyde moieties. This compound was used to link epidermal growth factor to genome-free MS2 viral capsids, affording nanoscale delivery vectors that can target a variety of cancer cell types.


Subject(s)
Aminophenols/chemistry , Biomimetic Materials/chemistry , Cross-Linking Reagents/chemistry , Aniline Compounds/chemistry , Cross-Linking Reagents/chemical synthesis , Humans , MCF-7 Cells , Molecular Structure , Oxidation-Reduction
5.
Mol Cell Biol ; 36(20): 2596-611, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27503856

ABSTRACT

Macropinocytosis is a normal cellular process by which cells internalize extracellular fluids and nutrients from their environment and is one strategy that Ras-transformed pancreatic cancer cells use to increase uptake of amino acids to meet the needs of rapid growth. Paradoxically, in non-Ras transformed medulloblastoma brain tumors, we have shown that expression and activation of the receptor tyrosine kinase TrkA overactivates macropinocytosis, resulting in the catastrophic disintegration of the cell membrane and in tumor cell death. The molecular basis of this uncontrolled form of macropinocytosis has not been previously understood. Here, we demonstrate that the overactivation of macropinocytosis is caused by the simultaneous activation of two TrkA-mediated pathways: (i) inhibition of RhoB via phosphorylation at Ser(185) by casein kinase 1, which relieves actin stress fibers, and (ii) FRS2-scaffolded Src and H-Ras activation of RhoA, which stimulate actin reorganization and the formation of lamellipodia. Since catastrophic macropinocytosis results in brain tumor cell death, improved understanding of the mechanisms involved will facilitate future efforts to reprogram tumors, even those resistant to apoptosis, to die.


Subject(s)
Casein Kinase I/metabolism , Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Pinocytosis , Receptor, trkA/metabolism , rhoB GTP-Binding Protein/metabolism , Actins/metabolism , Cell Death , Cell Line, Tumor , Humans , Phosphorylation , Proto-Oncogene Proteins p21(ras)/metabolism , Serine/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism
6.
Biomaterials ; 102: 268-76, 2016 09.
Article in English | MEDLINE | ID: mdl-27348850

ABSTRACT

Polyacrylamide hydrogels have been used extensively to study cell responses to the mechanical and biochemical properties of extracellular matrix substrates. A key step in fabricating these substrates is the conjugation of cell adhesion proteins to the polyacrylamide surfaces, which typically involves nonspecifically anchoring these proteins via side-chain functional groups. This can result in a loss of presentation control and altered bioactivity. Here, we describe a new functionalization strategy in which we anchor full-length extracellular matrix proteins to polyacrylamide substrates using 2-pyridinecarboxaldehyde, which can be co-polymerized into polyacrylamide gels and used to immobilize proteins by their N-termini. This one-step reaction proceeds under mild aqueous conditions and does not require additional reagents. We demonstrate that these substrates can readily conjugate to various extracellular matrix proteins, as well as promote cell adhesion and spreading. Notably, this chemistry supports the assembly and cellular remodeling of large collagen fibers, which is not observed using conventional side-chain amine-conjugation chemistry.


Subject(s)
Acrylic Resins/chemistry , Biocompatible Materials/chemistry , Extracellular Matrix Proteins/chemistry , Pyridines/chemistry , Animals , Cattle , Cell Adhesion , Cell Line , Collagen/chemistry , Hardness , Humans , Mechanical Phenomena , Mice
7.
Angew Chem Int Ed Engl ; 55(30): 8585-9, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27239057

ABSTRACT

Enzyme-mediated protein modification often requires large amounts of biocatalyst, adding significant costs to the process and limiting industrial applications. Herein, we demonstrate a scalable and straightforward strategy for the efficient capture and recycling of enzymes using a small-molecule affinity tag. A proline variant of an evolved sortase A (SrtA 7M) was N-terminally labeled with lithocholic acid (LA)-an inexpensive bile acid that exhibits strong binding to ß-cyclodextrin (ßCD). Capture and recycling of the LA-Pro-SrtA 7M conjugate was achieved using ßCD-modified sepharose resin. The LA-Pro-SrtA 7M conjugate retained full enzymatic activity, even after multiple rounds of recycling.

8.
Nat Chem Biol ; 11(5): 326-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25822913

ABSTRACT

The chemical modification of proteins is an enabling technology for many scientific fields, including chemical biology, biophysics, bioengineering and materials science. These methods allow the attachment of strategically selected detection probes, polymers, drug molecules and analysis platforms. However, organic reactions that can proceed under conditions mild enough to maintain biomolecular function are limited. Even more rare are chemical strategies that can target a single site, leading to products with uniform properties and optimal function. We present a versatile method for the selective modification of protein N termini that does not require any genetic engineering of the protein target. This reaction is demonstrated for 12 different proteins, including the soluble domain of the human estrogen receptor. The function of this protein was confirmed through the binding of a fluorescent estrogen mimic, and the modified protein was explored as a prototype for the detection of endocrine-disrupting chemicals in water.


Subject(s)
Proteins/chemistry , Pyridines/chemistry , Aldehydes/chemistry , Biotinylation , Endocrine Disruptors/chemistry , Endocrine Disruptors/pharmacology , Humans , Indicators and Reagents , Peptides/chemistry , Protein Conformation , Receptors, Estrogen/chemistry , Receptors, Estrogen/drug effects , Ribonuclease, Pancreatic/chemistry , Small Molecule Libraries
9.
Mol Cell Biol ; 34(12): 2221-34, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24710275

ABSTRACT

Mammalian DREAM is a conserved protein complex that functions in cellular quiescence. DREAM contains an E2F, a retinoblastoma (RB)-family protein, and the MuvB core (LIN9, LIN37, LIN52, LIN54, and RBBP4). In mammals, MuvB can alternatively bind to BMYB to form a complex that promotes mitotic gene expression. Because BMYB-MuvB is essential for proliferation, loss-of-function approaches to study MuvB have generated limited insight into DREAM function. Here, we report a gene-targeted mouse model that is uniquely deficient for DREAM complex assembly. We have targeted p107 (Rbl1) to prevent MuvB binding and combined it with deficiency for p130 (Rbl2). Our data demonstrate that cells from these mice preferentially assemble BMYB-MuvB complexes and fail to repress transcription. DREAM-deficient mice show defects in endochondral bone formation and die shortly after birth. Micro-computed tomography and histology demonstrate that in the absence of DREAM, chondrocytes fail to arrest proliferation. Since DREAM requires DYRK1A (dual-specificity tyrosine phosphorylation-regulated protein kinase 1A) phosphorylation of LIN52 for assembly, we utilized an embryonic bone culture system and pharmacologic inhibition of (DYRK) kinase to demonstrate a similar defect in endochondral bone growth. This reveals that assembly of mammalian DREAM is required to induce cell cycle exit in chondrocytes.


Subject(s)
Chondrocytes/cytology , Chondrocytes/metabolism , Mammals/metabolism , Multiprotein Complexes/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cell Cycle/genetics , Cell Proliferation/drug effects , Chondrocytes/drug effects , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Harmine/pharmacology , Humans , Mice , Mice, Mutant Strains , Models, Animal , Molecular Sequence Data , Multiprotein Complexes/chemistry , Mutation/genetics , Osteogenesis/drug effects , Protein Binding/drug effects , Retinoblastoma Protein/metabolism , Tibia/drug effects , Tibia/metabolism , Tibia/pathology
10.
Mol Cell Biol ; 34(11): 2029-45, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24662053

ABSTRACT

The retinoblastoma protein (pRB) is best known for regulating cell proliferation through E2F transcription factors. In this report, we investigate the properties of a targeted mutation that disrupts pRB interactions with the transactivation domain of E2Fs. Mice that carry this mutation endogenously (Rb1(ΔG)) are defective for pRB-dependent repression of E2F target genes. Except for an accelerated entry into S phase in response to serum stimulation, cell cycle regulation in Rb1(ΔG/ΔG) mouse embryonic fibroblasts (MEFs) strongly resembles that of the wild type. In a serum deprivation-induced cell cycle exit, Rb1(ΔG/ΔG) MEFs display a magnitude of E2F target gene derepression similar to that of Rb1(-/-) cells, even though Rb1(ΔG/ΔG) cells exit the cell cycle normally. Interestingly, cell cycle arrest in Rb1(ΔG/ΔG) MEFs is responsive to p16 expression and gamma irradiation, indicating that alternate mechanisms can be activated in G1 to arrest proliferation. Some Rb1(ΔG/ΔG) mice die neonatally with a muscle degeneration phenotype, while the others live a normal life span with no evidence of spontaneous tumor formation. Most tissues appear histologically normal while being accompanied by derepression of pRB-regulated E2F targets. This suggests that non-E2F-, pRB-dependent pathways may have a more relevant role in proliferative control than previously identified.


Subject(s)
E2F Transcription Factors/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , S Phase Cell Cycle Checkpoints/genetics , Adenocarcinoma/genetics , Alleles , Animals , Binding Sites , Cell Line , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Fibroblasts/cytology , Gene Targeting , Mice , Mice, Knockout , Mutation , Pituitary Neoplasms/genetics , S Phase/genetics
11.
J Mol Neurosci ; 49(1): 38-51, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22744634

ABSTRACT

We previously demonstrated that the guanine nucleotide exchange factor, RasGrf1, binds nerve growth factor (NGF)-activated TrkA and facilitates neurotrophin-induced neurite outgrowth in PC12 cells. RasGrf1 can activate both Ras and Rac, via intrinsic Cdc25 and DH domains, respectively, suggesting that the activation of both could contribute to this process. Previous studies have assayed constitutive neurite outgrowth following RasGrf1 over-expression in PC12 cells, in either the absence or presence of ectopic H-Ras, and have suggested an essential role for either Ras or Rac depending on the presence of H-Ras over-expression. In contrast, in this study, we have addressed the mechanism of how RasGrf1 facilitates neurite outgrowth in response to the neurotrophins, NGF and BDNF. Using Ras/Rac activation assays and site-directed RasGrf1 mutants, we find that both Ras and Rac are essential to neurotrophin-induced neurite outgrowth. Moreover, we find that H-Ras over-expression rescues the loss of neurite outgrowth observed with a Rac minus mutant and that RasGrf1 differentially stimulates NGF-dependent activation of Rac or Ras, depending on cell type. Collectively, these studies clarify the mechanism of how RasGrf1 expression facilitates neurotrophin-induced neurite outgrowth. Moreover, they suggest that H-Ras over-expression should be used with caution to measure phenotypic responses.


Subject(s)
Neurites/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Nerve Growth Factor/metabolism , rac GTP-Binding Proteins/metabolism , ras-GRF1/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , HEK293 Cells , Humans , Mice , Mutagenesis, Site-Directed , Mutation , Nerve Growth Factor/metabolism , Neurites/metabolism , PC12 Cells , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Up-Regulation , ras-GRF1/genetics
12.
J Neurochem ; 121(6): 861-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22404429

ABSTRACT

Vesicular transport in neurons plays a vital role in neuronal function and survival. Nesca is a novel protein that we previously identified and herein describe its pattern of expression, subcellular localization and protein-protein interactions both in vitro and in vivo. Specifically, a large proportion of Nesca is in tight association with both actin and microtubule cytoskeletal proteins. Nesca binds to F-actin, microtubules, ßIII and acetylated α-tubulin, but not neurofilaments or the actin-binding protein drebrin, in in vitro-binding assays. Nesca co-immunoprecipitates with kinesin heavy chain (KIF5B) and kinesin light-chain motors as well as with the synaptic membrane precursor protein, syntaxin-1, and is a constituent of the post-synaptic density. Moreover, in vitro-binding assays indicate that Nesca directly binds KIF5B, kinesin light-chain and syntaxin-1. In contrast, Nesca does not co-immunoprecipitate with the kinesin motors KIF1B, KIF3A nor does it bind syntaxin-4 or the synaptosome-associated protein 25 kDa (SNAP-25) in vitro. Nesca expression in neurons is highly punctuate, co-stains with syntaxin-1, and is found in fractions containing markers of early endosomes and Golgi suggesting that it is involved in vesicular transport. Collectively, these data suggest that Nesca functions as an adapter involved in neuronal vesicular transport including vesicles containing soluble N-ethylmaleimide sensitive factor attachment protein receptors that are essential to exocytosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Kinesins/metabolism , Neurons/metabolism , Syntaxin 1/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Blotting, Western , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Hippocampus/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Neurogenesis/physiology , Post-Synaptic Density/metabolism , Protein Transport/physiology , Synaptic Membranes/metabolism , Transfection
13.
Genes Cancer ; 3(11-12): 619-33, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23634251

ABSTRACT

The retinoblastoma tumor suppressor protein (pRB) plays an integral role in G1-S checkpoint control and consequently is a frequent target for inactivation in cancer. The RB protein can function as an adaptor, nucleating components such as E2Fs and chromatin regulating enzymes into the same complex. For this reason, pRB's regulation by posttranslational modifications is thought to be critical. pRB is phosphorylated by a number of different kinases such as cyclin dependent kinases (Cdks), p38 MAP kinase, Chk1/2, Abl, and Aurora b. Although phosphorylation of pRB by Cdks has been extensively studied, activities regulated through phosphorylation by other kinases are just starting to be understood. As well as being phosphorylated, pRB is acetylated, methylated, ubiquitylated, and SUMOylated. Acetylation, methylation, and SUMOylation play roles in pRB mediated gene silencing. Ubiquitinylation of pRB promotes its degradation and may be used to regulate apoptosis. Recent proteomic data have revealed that pRB is posttranslationally modified to a much greater extent than previously thought. This new information suggests that many unknown pathways affect pRB regulation. This review focuses on posttranslational modifications of pRB and how they influence its function. The final part of the review summarizes new phosphorylation sites from accumulated proteomic data and discusses the possibilities that might arise from this data.

14.
Bioorg Med Chem Lett ; 20(5): 1569-72, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20149653

ABSTRACT

Modifications to the sugar portion of C-aryl glycoside sodium glucose transporter 2 (SGLT2) inhibitors were explored, including systematic deletion and modification of each of the glycoside hydroxyl groups. Based on results showing activity to be quite tolerant of structural change at the C-5 position, a series of novel C-5 spiro analogues was prepared. Some of these analogues exhibit low nanomolar potency versus SGLT2 and promote urinary glucose excretion (UGE) in rats. However, due to sub-optimal pharmacokinetic parameters (in particular half-life), predicted human doses did not meet criteria for further advancement.


Subject(s)
Glycosides/chemistry , Hypoglycemic Agents/chemistry , Sodium-Glucose Transporter 2 Inhibitors , Spiro Compounds/chemistry , Animals , Cyclization , Glycosides/chemical synthesis , Glycosides/pharmacokinetics , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacokinetics , Male , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 2/metabolism
15.
J Neurochem ; 112(4): 882-99, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19943845

ABSTRACT

Ectopic expression of the TrkA receptor tyrosine kinase in tumors of the nervous system can mediate nerve growth factor (NGF)-dependent cell death by apoptosis and /or autophagy. Herein, we demonstrate that TrkA can also induce cell death in medulloblastoma Daoy cells by a caspase-independent mechanism that involves the hyperstimulation of macropinocytosis. Specifically, NGF-stimulates the uptake of AlexaFluor546-dextran into lysosome-associated membrane protein-1 positive vacuoles which fuse with microtubule associated protein light chain 3 (LC3) positive autophagosomes, to form large intracellular vacuoles (> 1 mum), which then fuse with lysotracker positive lysosomes. While LC3 cleavage and the appearance of LC3 positive vacuoles suggest the induction of autophagy, siRNA reduced expression of four proteins essential to autophagy (beclin-1, Atg5, LC3 and Atg9) neither blocks NGF-induced vacuole formation nor cell death. TrkA activated cell death does not require p38, JNK or Erk1/2 kinases but does require activation of class III PI-3 kinase and is blocked by the casein kinase 1 (CK1) inhibitor, D4476. This inhibitor does not interfere with TrkA activation but does block NGF-dependent AlexaFluor546-dextran uptake and CK1 dependent phosphorylation of beta-catenin. Collectively, these data demonstrate that TrkA stimulates cell death by a novel mechanism involving CK1-dependent hyperstimulation of macropinocytosis.


Subject(s)
Autophagy/drug effects , Nerve Growth Factor/pharmacology , Pinocytosis/drug effects , Pinocytosis/physiology , Receptor, trkA/metabolism , Analysis of Variance , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Cell Line, Tumor , Cytochromes c/metabolism , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation/methods , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Lysosomes/drug effects , Lysosomes/ultrastructure , Medulloblastoma/pathology , Medulloblastoma/ultrastructure , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Electron, Transmission/methods , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Time Factors , Transfection/methods
16.
J Neurochem ; 112(4): 924-39, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19943849

ABSTRACT

Fibroblast growth factor (FGF) mediated signaling is essential to many aspects of neural development. Activated FGF receptors signal primarily through the FGF receptor substrate (Frs) adapters, which include Frs2/Frs2alpha and Frs3/Frs2beta. While some studies suggest that Frs3 can compensate for the loss of Frs2 in transfected cells, the lack of an effective Frs3 specific antibody has prevented efforts to determine the role(s) of the endogenous protein. To this end, we have generated a Frs3 specific antibody and have characterized the pattern of Frs3 expression in the developing nervous system, its subcellular localization as well as its biochemical properties. We demonstrate that Frs3 is expressed at low levels in the ventricular zone of developing cortex, between E12 and E15, and it co-localizes with nestin and acetylated alpha-tubulin in radial processes in the ventricular/subventricular zones as well as with betaIII tubulin in differentiated cortical neurons. Subcellular fractionation studies demonstrate that endogenous Frs3 is both soluble and plasma membrane associated while Frs3 expressed in 293T cells associates exclusively with lipid rafts. Lastly, we demonstrate that neuronal Frs3 binds microtubules comparable to the microtubule-associated protein, MAP2, while Frs2 does not. Collectively, these data suggest that neuronal Frs3 functions as a novel microtubule binding protein and they provide the first biochemical evidence that neuronal Frs3 is functionally distinct from Frs2/Frs2alpha.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation/physiology , Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Microtubule-Associated Proteins/metabolism , Neurons/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/physiology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cyclic AMP/pharmacology , Embryo, Mammalian , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hippocampus , Humans , In Vitro Techniques , Ki-67 Antigen/metabolism , Lysosomal-Associated Membrane Protein 1/metabolism , Membrane Microdomains/metabolism , Mice , Microtubule-Associated Proteins/genetics , Neurons/drug effects , Protein Binding , T-Box Domain Proteins/metabolism , Tubulin/metabolism
17.
Biochim Biophys Acta ; 1763(4): 366-80, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16697063

ABSTRACT

We have investigated the signaling properties of the fibroblast growth factor (FGF) receptor substrate 3 (FRS3), also known as SNT-2 or FRS2beta, in neurotrophin-dependent differentiation in comparison with the related adapter FRS2 (SNT1 or FRS2alpha). We demonstrate that FRS3 binds all neurotrophin Trk receptor tyrosine kinases and becomes tyrosine phosphorylated in response to NGF, BDNF, NT-3 and FGF stimulation in transfected cells and/or primary cortical neurons. Second, the signaling molecules Grb2 and Shp2 bind FRS3 at consensus sites that are highly conserved among FRS family members and that Shp2, in turn, becomes tyrosine phosphorylated. While FRS3 over-expression in PC12 cells neither increases NGF-induced neuritogenesis nor activation of Map kinase/AKT, comparable to previous reports on FRS2, over-expression of a chimeric adapter containing the PH/PTB domains of the insulin receptor substrate (IRS) 2, in place of the PTB domain of FRS3 (IRS2-FRS3) supports insulin-dependent Map kinase activation and neurite outgrowth in PC12 cells. Collectively, these data demonstrate that FRS3 supports ligand-induced Map kinase activation and that the chimeric IRS2-FRS3 adapter is stimulating sufficient levels of activated MapK to support neurite outgrowth in PC12 cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fibroblast Growth Factors/physiology , Nerve Growth Factors/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , PC12 Cells , Rats , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Receptor, trkC/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Substrate Specificity/physiology
18.
Analyst ; 130(12): 1608-16, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16284659

ABSTRACT

Long molecules such as fibrous proteins are particularly difficult to characterise structurally. We have recently designed a microvolume Couette flow linear dichroism (LD) cell whose sample volume is only 20-40 microL in contrast to previous cells where the volume of sample required has typically been of the order of 1000-2000 microL. This brings the sample requirements of LD to a level where it can be used for biological samples. Since LD is the difference in absorption of light polarised parallel to an orientation direction and perpendicular to that direction, it is the ideal technique for determining relative orientations of subunits of e.g. fibrous proteins, DNA-drug systems, etc. For solution phase samples, Couette flow orientation, whereby the sample is sandwiched between two cylinders, one of which rotates, has proved to be the optimal technique for LD experiments in many laboratories. Our capillary microvolume LD cell has been designed using extruded quartz rods and capillaries and focusing and collecting lenses. We have developed applications with PCR products, fibrous proteins, liposome-bound membrane proteins, as well as DNA-dye systems. Despite this range of applications, to date there is nothing reported in the literature to enable one to validate the performance of Couette flow LD cells. In this paper we establish validation criteria and show that the data from the microvolume cells are reproducible, vary by less than 1% with sample reloading, follow the Beer-Lambert law, and have signals linear in voltage over a wide voltage range. The microvolume cell data are consistent with those from the large-volume cells for DNA samples. Surprisingly, upon extending the wavelength range by adding the intercalator ethidium bromide, the spectra in the microvolume and large-volume cells differ by a wavelength dependent orientation parameter. This wavelength variation was concluded to be the result of Taylor-vortices in the large-volume cells which have inner rotating cylinders in our laboratory. Thus the microvolume LD cells can be concluded to provide better data than our large-volume LD cells, though the latter are still to be preferred for titration series as it is extremely difficult to add sample to the capillary cells without introducing artefacts.


Subject(s)
DNA/analysis , Microchemistry/instrumentation , Microchemistry/methods , Spectrum Analysis/instrumentation , Spectrum Analysis/methods
19.
J Biol Chem ; 280(20): 19461-71, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15753086

ABSTRACT

The human tumorous imaginal disc 1 (TID1) proteins including TID1(L) and TID1(S), members of the DnaJ domain protein family, are involved in multiple intracellular signaling pathways such as apoptosis induction, cell proliferation, and survival. Here we report that TID1 associates with the Trk receptor tyrosine kinases and regulates nerve growth factor (NGF)-induced neurite outgrowth in PC12-derived nnr5 cells. Binding assays and transfection studies showed that the carboxyl-terminal end of TID1 (residues 224-429) bound to Trk at the activation loop (Tyr(P)(683)-Tyr(684)(P)(684) in rat TrkA) and that TID1 was tyrosine phosphorylated by Trk both in yeast and in transfected cells. Moreover endogenous TID1 was also tyrosine phosphorylated by and co-immunoprecipitated with Trk in neurotrophin-stimulated primary rat hippocampal neurons. Overexpression studies showed that both TID1(L) and TID1(S) significantly facilitated NGF-induced neurite outgrowth in TrkA-expressing nnr5 cells possibly through a mechanism involving increased activation of mitogen-activated protein kinase. Consistently knockdown of endogenous TID1, mediated with specific short hairpin RNA, significantly reduced NGF-induced neurite growth in nnr5-TrkA cells. These data provide the first evidence that TID1 is a novel intracellular adaptor that interacts with the Trk receptor tyrosine kinases in an activity-dependent manner to facilitate Trk-dependent intracellular signaling.


Subject(s)
Heat-Shock Proteins/metabolism , Receptor, trkA/metabolism , Animals , Base Sequence , Binding Sites , DNA/genetics , HSP40 Heat-Shock Proteins , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Humans , Kinetics , Neurites/metabolism , Neurites/ultrastructure , PC12 Cells , Phosphorylation , RNA Interference , Rats , Receptor, trkA/chemistry , Receptor, trkA/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Two-Hybrid System Techniques , Tyrosine/chemistry
20.
J Biol Chem ; 280(1): 225-35, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15513915

ABSTRACT

Ras guanine-releasing factor 1 (RasGrf1), a guanine nucleotide exchange factor for members of the Ras and Rho family of GTPases, is highly expressed in the brain. It is regulated by two separate mechanisms, calcium regulation through interaction with its calcium/calmodulin-binding IQ domain and serine and tyrosine phosphorylation. RasGrf1 is activated downstream of G-protein-coupled receptors and the non-receptor tyrosine kinases, Src and Ack1. Previously, we demonstrated a novel interaction between the intracellular domain of the nerve growth factor-regulated TrkA receptor tyrosine kinase and an N-terminal fragment of RasGrf1. We now show that RasGrf1 is phosphorylated and interacts with TrkA, -B, and -C in co-transfection studies. This interaction and phosphorylation of RasGrf1 is dependent on the HIKE domain of TrkA (a region shown to interact with pleckstrin homology domains) but not on any of the phosphotyrosine residues that act as docking sites for intracellular signaling molecules such as Shc and FRS-2. The PH1 domain alone of RasGrf1 is sufficient for phosphorylation by the TrkA receptor. A potential role for Trk activation of RasGrf1 is suggested through transfection studies in PC12 cells in which RasGrf1 significantly increases neurite outgrowth at low doses of neurotrophin stimulation. Notably, this neurite outgrowth is dependent on an intact HIKE domain, as nnr5-S10 cells expressing a TrkA HIKE domain mutant do not exhibit potentiated neurite outgrowth in the presence of RasGrf1. These studies identify RasGrf1 as a novel target of neurotrophin activation and suggest an additional pathway whereby neurotrophin-stimulated neurite outgrowth may be regulated.


Subject(s)
Nerve Growth Factors/metabolism , Neurites/physiology , Receptor, trkA/metabolism , ras-GRF1/metabolism , Animals , Binding Sites , Cell Enlargement , Humans , Mice , Neurites/ultrastructure , PC12 Cells , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Rats , Receptor, trkB/metabolism , Signal Transduction , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...