Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 246: 125710, 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37414319

ABSTRACT

p75 neurotrophin receptor (p75NTR) contains a C-terminal globular protein module known as the death domain (DD), which plays a central role in apoptotic and inflammatory signaling through the formation of oligomeric protein complexes. A monomeric state of the p75NTR-DD also exists depending on its chemical environment in vitro. However, studies on the oligomeric states of the p75NTR-DD have produced conflicting findings and sparked great controversy. Here we present new evidence from biophysical and biochemical studies to demonstrate the coexistence of symmetric and asymmetric dimers of the p75NTR-DD, which may equilibrate with the monomeric form in solution and in the absence of any other protein. The reversible close-open solution behavior may be important for the p75NTR-DD to serve as an intracellular signaling hub. This result supports an intrinsic ability of the p75NTR-DD to self-associate, in congruence with the oligomerization properties of all members of the DD superfamily.


Subject(s)
Death Domain Superfamily , Receptor, Nerve Growth Factor , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/metabolism , Death Domain , Signal Transduction
2.
Life Sci Alliance ; 4(4)2021 04.
Article in English | MEDLINE | ID: mdl-33536237

ABSTRACT

γ-secretase inhibitors (GSI) were developed to reduce the generation of Aß peptide to find new Alzheimer's disease treatments. Clinical trials on Alzheimer's disease patients, however, showed several side effects that worsened the cognitive symptoms of the treated patients. The observed side effects were partially attributed to Notch signaling. However, the effect on other γ-secretase substrates, such as the p75 neurotrophin receptor (p75NTR) has not been studied in detail. p75NTR is highly expressed in the basal forebrain cholinergic neurons (BFCNs) during all life. Here, we show that GSI treatment induces the oligomerization of p75CTF leading to the cell death of BFCNs, and that this event is dependent on TrkA activity. The oligomerization of p75CTF requires an intact cholesterol recognition sequence (CRAC) and the constitutive binding of TRAF6, which activates the JNK and p38 pathways. Remarkably, TrkA rescues from cell death by a mechanism involving the endocytosis of p75CTF. These results suggest that the inhibition of γ-secretase activity in aged patients, where the expression of TrkA in the BFCNs is already reduced, could accelerate cholinergic dysfunction and promote neurodegeneration.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Endocytosis , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Motifs , Amyloid Precursor Protein Secretases/metabolism , Cell Death/drug effects , Cycloheximide/pharmacology , Humans , Ligands , MAP Kinase Signaling System , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization/drug effects , Proteolysis , Receptor, Nerve Growth Factor/chemistry
3.
Biol Rev Camb Philos Soc ; 94(4): 1282-1293, 2019 08.
Article in English | MEDLINE | ID: mdl-30762293

ABSTRACT

The death domain (DD) is a globular protein motif with a signature feature of an all-helical Greek-key motif. It is a primary mediator of a variety of biological activities, including apoptosis, cell survival and cytoskeletal changes, which are related to many neurodegenerative diseases, neurotrauma, and cancers. DDs exist in a wide range of signalling proteins including p75 neurotrophin receptor (p75NTR ), a member of the tumour necrosis factor receptor superfamily. The specific signalling mediated by p75NTR in a given cell depends on the type of ligand engaging the extracellular domain and the recruitment of cytosolic interactors to the intracellular domain, especially the DD, of the receptor. In solution, the p75NTR -DDs mainly form a symmetric non-covalent homodimer. In response to extracellular signals, conformational changes in the p75NTR extracellular domain (ECD) propagate to the p75NTR -DD through the disulfide-bonded transmembrane domain (TMD) and destabilize the p75NTR -DD homodimer, leading to protomer separation and exposure of binding sites on the DD surface. In this review, we focus on recent advances in the study of the structural mechanism of p75NTR -DD signalling through recruitment of diverse intracellular interactors for the regulation and control of diverse functional outputs.


Subject(s)
Death Domain , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/metabolism , Amino Acid Sequence , Animals , Gene Expression Regulation , Models, Molecular , Protein Conformation , Receptor, Nerve Growth Factor/genetics
4.
J Alzheimers Dis ; 63(1): 139-147, 2018.
Article in English | MEDLINE | ID: mdl-29578485

ABSTRACT

The p75 neurotrophin receptor (p75NTR) is an amyloid-ß (Aß) receptor that both mediates Aß neurotoxicity and regulates Aß production and deposition, thus playing an important role in the pathogenesis of Alzheimer's disease (AD). The extracellular domain of p75NTR (p75ECD), consisting of four cysteine-rich repeat domains (CRDs), was recently reported to be an endogenous anti-Aß scavenger to block p75NTR-mediated neuronal death and neurite degeneration signaling of Aß and pro-neurotrophins. Identification of the specific Aß binding domains of p75NTR is crucial for illuminating their interactions and the etiology of AD. CRDs of p75ECD were obtained by expression of recombinant plasmids or direct synthesis. Aß aggregation inhibiting test and immunoprecipitation assay were applied to locate the specific binding domains of Aß to p75ECD. The Aß neurotoxicity antagonistic effects of different CRDs were examined by cytotoxicity experiments including neurite outgrowth assay, propidium iodide (PI) staining, and MTT assay. In the Aß aggregation inhibiting test, the fluorescence intensity in the CRD2 and CRD4 treatment groups was significantly lower than that in the CRD1 and CRD3 treatment groups. Immunoprecipitation assay and western blot confirmed that Aß could bind to CRD2 and CRD4. Besides, CRD2 and CRD4 antagonized Aß neurotoxicity suggested by longer neurite length, less PI labelled cells, and higher cell viability than the control group. Our results indicate that CRD2 and CRD4 are Aß binding domains of p75NTR and capable of antagonizing Aß neurotoxicity, and therefore are potential therapeutic targets to block the interaction of Aß and p75NTR in the pathogenesis of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Neurons/drug effects , Protein Structure, Tertiary/physiology , Receptor, Nerve Growth Factor/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Differentiation/drug effects , Cell Line, Transformed , Cysteine/metabolism , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Neuroblastoma/pathology , Neuronal Outgrowth/drug effects , Neuronal Outgrowth/genetics , Neurons/metabolism , Protein Binding/physiology , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/genetics , Transfection
5.
Curr Top Med Chem ; 18(32): 2800-2815, 2018.
Article in English | MEDLINE | ID: mdl-30747071

ABSTRACT

The failure of the Integrase Strand Transfer Inhibitors (INSTIs) due to the mutations occurring at the catalytic site of HIV integrase (IN) has led to the design of allosteric integrase inhibitors (ALLINIs). Lens epithelium derived growth factor (LEDGF/p75) is the host cellular cofactor which helps chaining IN to the chromatin. The protein-protein interactions (PPIs) were observed at the allosteric site (LEDGF/p75 binding domain) between LEDGF/p75 of the host cell and IN of virus. In recent years, many small molecules such as CX04328, CHIBA-3053 and CHI-104 have been reported as LEDGF/p75-IN interaction inhibitors (LEDGINs). LEDGINs have emerged as promising therapeutics to halt the PPIs by binding at the interface of both the proteins. In the present work, we correlated the docking scores for the reported LEDGINs containing quinoline scaffold with the in vitro biological data. The hierarchal clustering method was used to divide the compounds into test and training set. The robustness of the generated model was validated by q2 and r2 for the predicted set of compounds. The generated model between the docking score and biological data was assessed to predict the activity of the hits (quinoline scaffold) obtained from virtual screening of LEDGINs providing their structureactivity relationships to aim for the generation of potent agents.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Computer Simulation , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , Quinolines/pharmacology , Receptor, Nerve Growth Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Transcription Factors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/chemistry , Allosteric Site/drug effects , Drug Evaluation, Preclinical , HIV Integrase/chemistry , HIV Integrase Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Protein Binding/drug effects , Quinolines/chemistry , Receptor, Nerve Growth Factor/chemistry , Small Molecule Libraries/chemistry , Transcription Factors/chemistry
6.
Vitam Horm ; 104: 57-87, 2017.
Article in English | MEDLINE | ID: mdl-28215307

ABSTRACT

Although p75 neurotrophin receptor (p75NTR) was the founding member of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF), it is an atypical TNFRSF protein. p75NTR like TNF-R1 and Fas-R contain an extracellular domain with four cysteine-rich domains (CRD) and a death domain (DD) in the intracellular region. While TNFRSF proteins are activated by trimeric TNFSF ligands, p75NTR forms dimers activated by dimeric neurotrophins that are structurally unrelated to TNFSF proteins. In addition, although p75NTR shares with other members the interaction with the TNF receptor-associated factors to activate the NF-κB and cell death pathways, p75NTR does not interact with the DD-containing proteins FADD, TRADD, or MyD88. By contrast, the DD of p75NTR is able to recruit several protein interactors via a full catalog of DD interactions not described before in the TNFRSF. p75-DD forms homotypic symmetrical DD-DD complexes with itself and with the related p45-DD; forms heterotypic DD-CARD interactions with the RIP2-CARD domain, and forms a new interaction between a DD and RhoGDI. All these features, in addition to its promiscuous interactions with several ligands and coreceptors, its processing by α- and γ-secretases, the dimeric nature of its transmembrane domain and its "special" juxtamembrane region, make p75NTR a truly stranger in the TNFR superfamily. In this chapter, I will summarize the known structural aspects of p75NTR and I will analyze from a structural point of view, the similitudes and differences between p75NTR and the other members of the TNFRSF.


Subject(s)
Models, Molecular , Receptor, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Binding Sites , Dimerization , Humans , Ligands , Nerve Growth Factors/chemistry , Nerve Growth Factors/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Precursors/chemistry , Protein Precursors/metabolism , Receptor, Nerve Growth Factor/agonists , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor, Type II/agonists , Receptors, Tumor Necrosis Factor, Type II/chemistry , Receptors, Tumor Necrosis Factor, Type II/genetics
7.
Cell Tissue Res ; 368(1): 13-27, 2017 04.
Article in English | MEDLINE | ID: mdl-27726026

ABSTRACT

Recently, NIX, a pro-apoptotic BH3-only protein, was found to be a novel p75 neurotrophin receptor (p75NTR) binding protein by screening a human fetal brain two-hybrid library in our laboratory. We further study the interaction of these two proteins and the possible roles of p75NTR and NIX in intracerebral hemorrhage (ICH)-induced neuronal death. Using the split-ubiquitin yeast two-hybrid system, we found that the "Copper" domain in p75NTR and the TM region in NIX were sufficient for the interaction of these two proteins. Co-immunoprecipitation and in vitro binding assays demonstrated the direct interaction between p75NTR and NIX. NIX protein was stabilized by p75NTR at post-translational levels. Moreover, p75NTR was able to work together with NIX to promote apoptosis and affected the NIX-induced JNK-p53-Bax pathway in neuronal PC12 cells. Previous work has indicated that p75NTR and NIX are induced in neurons in human ICH and the rat ICH model, respectively. We confirm that both p75NTR and NIX levels were up-regulated in glutamate-treated primary cortical neurons (a cellular in vitro model for ICH) and in the rat ICH model. Glutamate exposure increased the association between p75NTR and NIX and elevated the activation of the JNK-p53-Bax pathway and neuronal apoptosis; all of these observations were similar in the rat ICH model. Importantly, p75NTR and NIX appeared to be involved in cortical neuronal apoptosis, because knockdown of p75NTR or NIX not only inhibited the JNK pathway but also impaired neuronal apoptosis. Thus, p75NTR and NIX may play critical roles in ICH-induced neuronal apoptosis in vitro and in vivo.


Subject(s)
Apoptosis , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Membrane Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins/metabolism , Receptor, Nerve Growth Factor/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Cerebral Cortex/pathology , Cerebral Hemorrhage/enzymology , Disease Models, Animal , Enzyme Activation/drug effects , Glutamic Acid/pharmacology , HEK293 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Membrane Proteins/chemistry , Protein Binding/drug effects , Protein Interaction Domains and Motifs , Protein Stability/drug effects , Proto-Oncogene Proteins/chemistry , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/chemistry , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/chemistry , Two-Hybrid System Techniques , Ubiquitin/metabolism , Up-Regulation/drug effects , bcl-2-Associated X Protein/metabolism
8.
J Biol Chem ; 291(23): 12346-57, 2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27056327

ABSTRACT

Dimerization of single span transmembrane receptors underlies their mechanism of activation. p75 neurotrophin receptor plays an important role in the nervous system, but the understanding of p75 activation mechanism is still incomplete. The transmembrane (TM) domain of p75 stabilizes the receptor dimers through a disulfide bond, essential for the NGF signaling. Here we solved by NMR the three-dimensional structure of the p75-TM-WT and the functionally inactive p75-TM-C257A dimers. Upon reconstitution in lipid micelles, p75-TM-WT forms the disulfide-linked dimers spontaneously. Under reducing conditions, p75-TM-WT is in a monomer-dimer equilibrium with the Cys(257) residue located on the dimer interface. In contrast, p75-TM-C257A forms dimers through the AXXXG motif on the opposite face of the α-helix. Biochemical and cross-linking experiments indicate that AXXXG motif is not on the dimer interface of p75-TM-WT, suggesting that the conformation of p75-TM-C257A may be not functionally relevant. However, rather than mediating p75 homodimerization, mutagenesis of the AXXXG motif reveals its functional role in the regulated intramembrane proteolysis of p75 catalyzed by the γ-secretase complex. Our structural data provide an insight into the key role of the Cys(257) in stabilization of the weak transmembrane dimer in a conformation required for the NGF signaling.


Subject(s)
Membrane Proteins/chemistry , Protein Domains , Protein Multimerization , Protein Structure, Secondary , Receptor, Nerve Growth Factor/chemistry , Amino Acid Motifs/genetics , Amino Acid Sequence , Blotting, Western , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , HeLa Cells , Humans , Lipids/chemistry , Magnetic Resonance Spectroscopy , Membrane Proteins/genetics , Membrane Proteins/metabolism , Micelles , Models, Molecular , Mutation , Oxidation-Reduction , Proteolysis , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism
9.
J Neurochem ; 138(1): 163-73, 2016 07.
Article in English | MEDLINE | ID: mdl-26991827

ABSTRACT

The neurotrophin receptor p75 (p75NTR) is a receptor for amyloid-beta (Aß) and mediates Aß-induced neurodegenerative signals. The ectodomain of p75NTR (p75ECD) is a physiological protective factor against Aß in Alzheimer's disease (AD). We have previously demonstrated that the shedding of p75ECD from the cell surface is down-regulated in AD brains and restoration of the p75ECD level in the brain, through intracranial administration of p75ECD by adeno-associated virus vectors, attenuates AD-like pathologies in an AD mouse model. In this study, we further investigated the feasibility and efficacy of peripheral administration of AAV-p75ECD on brain amyloid burden and associated pathogenesis. We found that intramuscular delivery of AAV-p75ECD increased the level of p75ECD in the blood, significantly improved the behavioral phenotype of amyloid precursor protein/PS1 transgenic mice, and reduced brain amyloid burden, attenuated Tau hyperphosphorylation, and neuroinflammation. Furthermore, intramuscular delivery of AAV-p75ECD was well tolerated. Our results indicate that peripheral delivery of p75ECD represents a safe and effective therapeutic strategy for AD. The ectodomain of p75NTR (p75ECD) is a physiological protective factor against amyloid-beta (Aß) in Alzheimer's disease (AD). Intramuscular delivery of AAV-p75ECD increased the p75ECD levels in the blood, reduced brain amyloid burden through a 'peripheral sink' mechanism and alleviates AD-type pathologies. Peripheral delivery of p75ECD represents a promising therapeutic strategy for AD.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/therapy , Cognition Disorders/therapy , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cognition Disorders/genetics , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Injections, Intramuscular , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics , Receptor, Nerve Growth Factor/genetics , Transduction, Genetic
10.
PLoS Biol ; 12(8): e1001918, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25093680

ABSTRACT

The p75 neurotrophin receptor, a member of the tumor necrosis factor receptor superfamily, is required as a co-receptor for the Nogo receptor (NgR) to mediate the activity of myelin-associated inhibitors such as Nogo, MAG, and OMgp. p45/NRH2/PLAIDD is a p75 homologue and contains a death domain (DD). Here we report that p45 markedly interferes with the function of p75 as a co-receptor for NgR. P45 forms heterodimers with p75 and thereby blocks RhoA activation and inhibition of neurite outgrowth induced by myelin-associated inhibitors. p45 binds p75 through both its transmembrane (TM) domain and DD. To understand the underlying mechanisms, we have determined the three-dimensional NMR solution structure of the intracellular domain of p45 and characterized its interaction with p75. We have identified the residues involved in such interaction by NMR and co-immunoprecipitation. The DD of p45 binds the DD of p75 by electrostatic interactions. In addition, previous reports suggested that Cys257 in the p75 TM domain is required for signaling. We found that the interaction of the cysteine 58 of p45 with the cysteine 257 of p75 within the TM domain is necessary for p45-p75 heterodimerization. These results suggest a mechanism involving both the TM domain and the DD of p45 to regulate p75-mediated signaling.


Subject(s)
Protein Multimerization , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Cysteine/metabolism , HEK293 Cells , Humans , Magnetic Resonance Spectroscopy , Mice , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Interaction Mapping , Protein Stability , Receptors, Cell Surface/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Solutions , Structure-Activity Relationship , Up-Regulation
12.
J Chem Inf Model ; 54(7): 2051-67, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-24941229

ABSTRACT

Crystallographic structures of NGF/p75NTR and proNGF/p75NTR were previously obtained in 2:1 and 2:2 stoichiometries, respectively. However, evidence shows that both stoichiometries can occur for mature neurotrophins and pro-neurotrophins. We used Molecular Dynamics (MD) simulations to examine the energetic and structural characteristics of these two complete systems as well as the uncomplexed forms of NGF and understand how these could translate in a new view of different biological outcomes. Here, we show that one chain at the 2:2 proNGF complex seems to be preferentially lost creating a 2:1 structure able to interact with sortilin. We also demonstrated that the structure of the neurotrophin dimers is not pre-established and suffers large structural modifications upon p75NTR binding. Moreover, our data suggests an elegant explanation for the dual role of NGF in neuronal cell death and survival, where different stoichiometries induce conformational changes that might be the basis for the different biological outcomes observed with the mature and proforms of neurotrophins.


Subject(s)
Molecular Dynamics Simulation , Nerve Growth Factor/chemistry , Nerve Growth Factor/metabolism , Protein Multimerization , Protein Precursors/chemistry , Protein Precursors/metabolism , Receptor, Nerve Growth Factor/metabolism , Protein Binding , Protein Structure, Quaternary , Receptor, Nerve Growth Factor/chemistry , Thermodynamics
13.
Handb Exp Pharmacol ; 220: 121-64, 2014.
Article in English | MEDLINE | ID: mdl-24668472

ABSTRACT

The p75 neurotrophin receptor (p75(NTR)) regulates a wide range of cellular functions, including programmed cell death, axonal growth and degeneration, cell proliferation, myelination, and synaptic plasticity. The multiplicity of cellular functions governed by the receptor arises from the variety of ligands and co-receptors which associate with p75(NTR) and regulate its signaling. P75(NTR) promotes survival through interactions with Trk receptors, inhibits axonal regeneration via partnerships with Nogo receptor (Nogo-R) and Lingo-1, and promotes apoptosis through association with Sortilin. Signals downstream of these interactions are further modulated through regulated intramembrane proteolysis (RIP) of p75(NTR) and by interactions with numerous cytosolic partners. In this chapter, we discuss the intricate signaling mechanisms of p75(NTR), emphasizing how these signals are differentially regulated to mediate these diverse cellular functions.


Subject(s)
Receptor, Nerve Growth Factor/physiology , Signal Transduction/physiology , Adaptor Proteins, Vesicular Transport/physiology , Animals , Apoptosis , Cell Cycle , Cell Survival , Humans , JNK Mitogen-Activated Protein Kinases/physiology , Myelin Sheath/physiology , NF-kappa B/physiology , Neuronal Plasticity , Protein Precursors/physiology , Receptor, Nerve Growth Factor/chemistry , Receptor, trkA/physiology
14.
Bioorg Khim ; 40(4): 451-7, 2014.
Article in Russian | MEDLINE | ID: mdl-25898755

ABSTRACT

Neurotoxic beta-amyloid peptide plays an important role in the pathology of Alzheimer's disease. In aggregated form it binds to several proteins on the surface of the brain cells leading to their death. p75 receptor in- volved in supporting of cell balance is one of the targets for toxic beta-amyloid. We proposed that induction of antibodies against potential binding sites of p75 with beta-amyloid can be a promising approach towards new drug development for Alzheimer's disease therapy. Four potentially immunoactive fragments of p75 were chosen and chemically synthesized. Investigation of immunoprotective effect of the peptide fragments carried out in mice with experimentally induced form of Alzheimer's disease helped to reveal two fragments effectively preserving murine memory from impairment. Results obtained by ELISA biochemical analysis showed that only immunization with fragment p75 155-164 led to significant decrease in beta-amyloid level in the brain of the experimental mice. Thus, immunization with both fragments of p75 receptor is believed to be an effective tool for the development of new drugs against Alzheimer's disease.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Antibodies/administration & dosage , Peptide Fragments/administration & dosage , Receptor, Nerve Growth Factor/immunology , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Animals , Antibodies/chemistry , Antibodies/immunology , Binding Sites/immunology , Hippocampus/immunology , Hippocampus/pathology , Humans , Immunization , Memory Disorders/drug therapy , Memory Disorders/immunology , Mice , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Protein Binding/immunology , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/therapeutic use
15.
Proc Natl Acad Sci U S A ; 110(51): 20461-6, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24282309

ABSTRACT

Drosophila Toll functions in embryonic development and innate immunity and is activated by an endogenous ligand, Spätzle (Spz). The related Toll-like receptors in vertebrates also function in immunity but are activated directly by pathogen-associated molecules such as bacterial endotoxin. Here, we present the crystal structure at 2.35-Å resolution of dimeric Spz bound to a Toll ectodomain encompassing the first 13 leucine-rich repeats. The cystine knot of Spz binds the concave face of the Toll leucine-rich repeat solenoid in an area delineated by N-linked glycans and induces a conformational change. Mutagenesis studies confirm that the interface observed in the crystal structure is relevant for signaling. The asymmetric binding mode of Spz to Toll is similar to that of nerve growth factor (NGF) in complex with the p75 neurotrophin receptor but is distinct from that of microbial ligands bound to the Toll-like receptors. Overall, this study indicates an allosteric signaling mechanism for Toll in which ligand binding to the N terminus induces a conformational change that couples to homodimerization of juxtamembrane structures in the Toll ectodomain C terminus.


Subject(s)
Drosophila Proteins/chemistry , Protein Multimerization/physiology , Toll-Like Receptors/chemistry , Animals , Crystallography, X-Ray , Drosophila Proteins/immunology , Drosophila Proteins/metabolism , Drosophila melanogaster , Endotoxins/chemistry , Endotoxins/immunology , Endotoxins/metabolism , Immunity, Innate/physiology , Nerve Growth Factor/chemistry , Nerve Growth Factor/immunology , Nerve Growth Factor/metabolism , Protein Binding , Protein Structure, Quaternary , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/immunology , Receptor, Nerve Growth Factor/metabolism , Repetitive Sequences, Amino Acid , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
16.
Mol Biol Cell ; 24(12): 1996-2007, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23637462

ABSTRACT

The sorting signals that direct proteins to the apical surface of polarized epithelial cells are complex and can include posttranslational modifications, such as N- and O-linked glycosylation. Efficient apical sorting of the neurotrophin receptor p75 is dependent on its O-glycosylated membrane proximal stalk, but how this domain mediates targeting is unknown. Protein oligomerization or clustering has been suggested as a common step in the segregation of all apical proteins. Like many apical proteins, p75 forms dimers, and we hypothesized that formation of higher-order clusters mediated by p75 dimerization and interactions of the stalk facilitate its apical sorting. Using fluorescence fluctuation techniques (photon-counting histogram and number and brightness analyses) to study p75 oligomerization status in vivo, we found that wild-type p75-green fluorescent protein forms clusters in the trans-Golgi network (TGN) but not at the plasma membrane. Disruption of either the dimerization motif or the stalk domain impaired both clustering and polarized delivery. Manipulation of O-glycan processing or depletion of multiple galectins expressed in Madin-Darby canine kidney cells had no effect on p75 sorting, suggesting that the stalk domain functions as a structural prop to position other determinants in the lumenal domain of p75 for oligomerization. Additionally, a p75 mutant with intact dimerization and stalk motifs but with a dominant basolateral sorting determinant (Δ250 mutant) did not form oligomers, consistent with a requirement for clustering in apical sorting. Artificially enhancing dimerization restored clustering to the Δ250 mutant but was insufficient to reroute this mutant to the apical surface. Together these studies demonstrate that clustering in the TGN is required for normal biosynthetic apical sorting of p75 but is not by itself sufficient to reroute a protein to the apical surface in the presence of a strong basolateral sorting determinant. Our studies shed new light on the hierarchy of polarized sorting signals and on the mechanisms by which newly synthesized proteins are segregated in the TGN for eventual apical delivery.


Subject(s)
Protein Multimerization , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/metabolism , trans-Golgi Network/metabolism , Animals , Binding Sites/genetics , Cell Line , Dogs , Galectins/genetics , Galectins/metabolism , Glycosylation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Confocal , Models, Biological , Mutation , Protein Transport , RNA Interference , Receptor, Nerve Growth Factor/genetics
17.
FEBS Lett ; 586(20): 3658-64, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22968039

ABSTRACT

Grb2 and ShcA are two phosphotyrosine-binding proteins that link receptor protein-tyrosine kinases to activation of the Ras-Erk pathway. While some receptors bind Grb2 directly, others bind ShcA, which provides a binding site for Grb2. In order to compare signal transduction through a Grb2-binding site with signal transduction through a ShcA-binding site, we replaced the ShcA-binding site in the NGF receptor with a Grb2-binding site. Our results show that the Grb2- and ShcA-binding sites have similar abilities to activate the Ras-Erk and PI 3-kinase-Akt pathways. In contrast, they displayed dramatic differences in their ability to activate DNA synthesis.


Subject(s)
GRB2 Adaptor Protein/metabolism , Protein Engineering/methods , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism , Shc Signaling Adaptor Proteins/metabolism , Signal Transduction , Amino Acid Motifs , Animals , Binding Sites , DNA/biosynthesis , Humans , Mice , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Nerve Growth Factor/chemistry , ras Proteins/metabolism
18.
J Neurosci Res ; 90(12): 2259-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22903500

ABSTRACT

Nerve growth factor (NGF) is a homodimer that binds to two distinct receptor types, TrkA and p75, to support survival and differentiation of neurons. The high-affinity binding on the cell surface is believed to involve a heteroreceptor complex, but its exact nature is unclear. We developed a heterodimer (heteromutein) of two NGF muteins that can bind p75 and TrkA on opposite sides of the heterodimer, but not two TrkA receptors. Previously described muteins are Δ9/13 that is TrkA negative and 7-84-103 that is signal selective through TrkA. The heteromutein (Htm1) was used to study the heteroreceptor complex formation and function, in the putative absence of NGF-induced TrkA dimerization. Cellular binding assays indicated that Htm1 does not bind TrkA as efficiently as wild-type (wt) NGF but has better affinity than either homodimeric mutein. Htm1, 7-84-103, and Δ9/13 were each able to compete for cold-temperature, cold-chase stable binding on PC12 cells, indicating that binding to p75 was required for a portion of this high-affinity binding. Survival, neurite outgrowth, and MAPK signaling in PC12 cells also showed a reduced response for Htm1, compared with wtNGF, but was better than the parent muteins in the order wtNGF > Htm1 > 7-84-103 >> Δ9/13. Htm1 and 7-84-103 demonstrated similar levels of survival on cells expressing only TrkA. In the longstanding debate on the NGF receptor binding mechanism, our data support the ligand passing of NGF from p75 to TrkA involving a transient heteroreceptor complex of p75-NGF-TrkA.


Subject(s)
Nerve Growth Factor/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Animals , Binding Sites , Binding, Competitive , Cell Differentiation , Cell Line , Cell Survival , Dimerization , Fibroblasts , Ligands , Mice , Models, Molecular , Mutation , Nerve Growth Factor/chemistry , Nerve Growth Factor/genetics , Neurites/ultrastructure , PC12 Cells , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/metabolism , Rats , Receptor, Nerve Growth Factor/chemistry , Receptor, trkA/chemistry
19.
Postepy Hig Med Dosw (Online) ; 66: 304-10, 2012 May 30.
Article in Polish | MEDLINE | ID: mdl-22706116

ABSTRACT

 The neurotrophins play an important role in the development of the nervous system. These trophic factors affect the cells through the neurotrophin receptors Trk and p75NTR. Trk (tyrosine kinase receptor) mediated signaling promotes survival and growth, while p75NTR-mediated signaling promotes cell death. The structure of p75NTR and its role in the regulation of survival, growth and induction of apoptosis are discussed. p75NTR can interact with the aggregated form of Aß peptides and by influencing protein tau hyperphosphorylation plays an important role in etiopathogenesis of Alzheimer's disease.


Subject(s)
Apoptosis/physiology , Cell Cycle/physiology , Cell Proliferation , Receptor, Nerve Growth Factor/metabolism , Amino Acid Sequence , Animals , Heredodegenerative Disorders, Nervous System/metabolism , Heredodegenerative Disorders, Nervous System/pathology , Humans , Nerve Growth Factors/metabolism , Protein Isoforms/chemistry , Receptor, Nerve Growth Factor/chemistry , Signal Transduction/physiology
20.
Microb Cell Fact ; 11: 45, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22510322

ABSTRACT

BACKGROUND: Receptors with a single transmembrane (TM) domain are essential for the signal transduction across the cell membrane. NMR spectroscopy is a powerful tool to study structure of the single TM domain. The expression and purification of a TM domain in Escherichia coli (E.coli) is challenging due to its small molecular weight. Although ketosteroid isomerase (KSI) is a commonly used affinity tag for expression and purification of short peptides, KSI tag needs to be removed with the toxic reagent cyanogen bromide (CNBr). RESULT: The purification of the TM domain of p75 neurotrophin receptor using a KSI tag with the introduction of a thrombin cleavage site is described herein. The recombinant fusion protein was refolded into micelles and was cleaved with thrombin. Studies showed that purified protein could be used for structural study using NMR spectroscopy. CONCLUSIONS: These results provide another strategy for obtaining a single TM domain for structural studies without using toxic chemical digestion or acid to remove the fusion tag. The purified TM domain of p75 neurotrophin receptor will be useful for structural studies.


Subject(s)
Escherichia coli/genetics , Gene Expression , Isomerases/genetics , Ketosteroids/metabolism , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/isolation & purification , Escherichia coli/metabolism , Humans , Isomerases/chemistry , Isomerases/isolation & purification , Isomerases/metabolism , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Protein Structure, Tertiary , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...