Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 200
Filter
1.
J Biol Chem ; 297(2): 100916, 2021 08.
Article in English | MEDLINE | ID: mdl-34175311

ABSTRACT

The p75 neurotrophin receptor (p75NTR) is a critical mediator of neuronal death and tissue remodeling and has been implicated in various neurodegenerative diseases and cancers. The death domain (DD) of p75NTR is an intracellular signaling hub and has been shown to interact with diverse adaptor proteins. In breast cancer cells, binding of the adaptor protein TRADD to p75NTR depends on nerve growth factor and promotes cell survival. However, the structural mechanism and functional significance of TRADD recruitment in neuronal p75NTR signaling remain poorly understood. Here we report an NMR structure of the p75NTR-DD and TRADD-DD complex and reveal the mechanism of specific recognition of the TRADD-DD by the p75NTR-DD mainly through electrostatic interactions. Furthermore, we identified spatiotemporal overlap of p75NTR and TRADD expression in developing cerebellar granule neurons (CGNs) at early postnatal stages and discover the physiological relevance of the interaction between TRADD and p75NTR in the regulation of canonical NF-κB signaling and cell survival in CGNs. Our results provide a new structural framework for understanding how the recruitment of TRADD to p75NTR through DD interactions creates a membrane-proximal platform, which can be efficiently regulated by various neurotrophic factors through extracellular domains of p75NTR, to propagate downstream signaling in developing neurons.


Subject(s)
NF-kappa B/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/metabolism , TNF Receptor-Associated Death Domain Protein/metabolism , Animals , Death Domain , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Conformation , Protein Interaction Domains and Motifs , Receptor, Nerve Growth Factor/metabolism , Signal Transduction , TNF Receptor-Associated Death Domain Protein/chemistry
2.
Sci Rep ; 10(1): 13686, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792564

ABSTRACT

The neurotrophin receptor p75NTR plays crucial roles in neuron development and regulates important neuronal processes like degeneration, apoptosis and cell survival. At the same time the detailed mechanism of signal transduction is unclear. One of the main hypotheses known as the snail-tong mechanism assumes that in the inactive state, the death domains interact with each other and in response to ligand binding there is a conformational change leading to their exposure. Here, we show that neither rat nor human p75NTR death domains homodimerize in solution. Moreover, there is no interaction between the death domains in a more native context: the dimerization of transmembrane domains in liposomes and the presence of activating mutation in extracellular juxtamembrane region do not lead to intracellular domain interaction. These findings suggest that the activation mechanism of p75NTR should be revised. Thus, we propose a novel model of p75NTR functioning based on interaction with "helper" protein.


Subject(s)
Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Receptors, Growth Factor/chemistry , Receptors, Growth Factor/metabolism , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/metabolism , Animals , Humans , Ligands , Liposomes/metabolism , Models, Molecular , Nerve Tissue Proteins/genetics , Protein Binding , Protein Conformation , Protein Domains , Protein Multimerization , Rats , Receptors, Growth Factor/genetics , Receptors, Nerve Growth Factor/genetics
3.
J Mol Biol ; 432(13): 3749-3760, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32302608

ABSTRACT

Optically controlled receptor tyrosine kinases (opto-RTKs) allow regulation of RTK signaling using light. Until recently, the majority of opto-RTKs were activated with blue-green light. Fusing a photosensory core module of Deinococcus radiodurans bacterial phytochrome (DrBphP-PCM) to the kinase domains of neurotrophin receptors resulted in opto-RTKs controlled with light above 650 nm. To expand this engineering approach to RTKs of other families, here we combined the DrBpP-PCM with the cytoplasmic domains of EGFR and FGFR1. The resultant Dr-EGFR and Dr-FGFR1 opto-RTKs are rapidly activated with near-infrared and inactivated with far-red light. The opto-RTKs efficiently trigger ERK1/2, PI3K/Akt, and PLCγ signaling. Absence of spectral crosstalk between the opto-RTKs and green fluorescent protein-based biosensors enables simultaneous Dr-FGFR1 activation and detection of calcium transients. Action mechanism of the DrBphP-PCM-based opto-RTKs is considered using the available RTK structures. DrBphP-PCM represents a versatile scaffold for engineering of opto-RTKs that are reversibly regulated with far-red and near-infrared light.


Subject(s)
Phytochrome/ultrastructure , Receptor Protein-Tyrosine Kinases/ultrastructure , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biosensing Techniques , Deinococcus/chemistry , Deinococcus/genetics , Green Fluorescent Proteins/chemistry , HeLa Cells , Humans , Light , MAP Kinase Signaling System/radiation effects , Nerve Growth Factors/chemistry , Nerve Growth Factors/genetics , Phosphatidylinositol 3-Kinases/genetics , Phytochrome/chemistry , Phytochrome/genetics , Protein Conformation/radiation effects , Proto-Oncogene Proteins c-akt/genetics , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/genetics , Signal Transduction/radiation effects
4.
Cancer Res ; 78(9): 2262-2276, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29437707

ABSTRACT

Resistance to anoikis allows cancer cells to survive during systemic circulation; however, the mechanism underlying anoikis resistance remains unclear. Here we show that A disintegrin and metalloprotease 10 (ADAM10)-mediated cleavage of p75 neurotrophin receptor (p75NTR) and subsequent generation of the p75NTR intracellular domain (ICD) endow cancer cells with resistance to anoikis. p75NTR ICD promoted expression of TNF receptor-associated factor 6 (TRAF6), a critical intermediary in p75NTR ICD-mediated signal transduction, at the translational level. Cell detachment-induced activation of EGFR triggered autoubiquitination of TRAF6 by facilitating its dimerization, subsequently activated NFκB, and eventually led to anoikis resistance. ADAM10 and p75NTR ICD also promoted tumor metastasis formation in vivo Together, our findings uncover a previously unknown function for the ADAM10-p75NTR ICD-TRAF6-NFκB axis in preventing anoikis and suggest ADAM10 and p75NTR ICD as potential cancer therapeutic targets.Significance: These findings identify the ADAM10-p75NTR ICD-TRAF6-NFκB signaling axis as a potential candidate for cancer therapy. Cancer Res; 78(9); 2262-76. ©2018 AACR.


Subject(s)
ADAM10 Protein/metabolism , Anoikis , Nerve Tissue Proteins/metabolism , Protein Interaction Domains and Motifs , Receptors, Nerve Growth Factor/metabolism , Biomarkers , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Intracellular Signaling Peptides and Proteins , Neoplasm Metastasis , Neoplasm Staging , Nerve Tissue Proteins/chemistry , Protein Binding , Protein Multimerization , Proteolysis , RNA Interference , Receptors, Nerve Growth Factor/chemistry , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Ubiquitination
5.
Biochem Biophys Res Commun ; 495(1): 700-705, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29108999

ABSTRACT

Nerve growth factor (NGF) is the prototypic member of the neurotrophin family and binds two receptors, TrkA and the 75 kDa neurotrophin receptor (p75NTR), through which diverse and sometimes opposing effects are mediated. Using the FoldX protein design algorithm, we generated eight NGF variants with different point mutations predicted to have altered binding to TrkA or p75NTR. Of these, the I31R NGF variant exhibited specific binding to p75NTR. The generation of this NGF variant with selective affinity for p75NTR can be used to enhance understanding of neurotrophin receptor imbalance in diseases and identifies a key targetable residue for the development of small molecules to disrupt binding of NGF to TrkA with potential uses in chronic pain.


Subject(s)
Drug Design , Mutagenesis, Site-Directed/methods , Nerve Growth Factor/biosynthesis , Nerve Growth Factor/chemistry , Protein Engineering/methods , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/metabolism , Animals , Binding Sites , Nerve Growth Factor/genetics , PC12 Cells , Protein Binding , Rats , Structure-Activity Relationship
6.
Methods Mol Biol ; 1727: 295-314, 2018.
Article in English | MEDLINE | ID: mdl-29222790

ABSTRACT

We describe here a versatile methodological platform to achieve site-directed and stoichiometry-controlled labeling of neurotrophins and their receptors with various probes, ranging from biotin to small organic dyes. This labeling method works in vitro on purified neurotrophins as well as in a living cell context, where it achieves selective labeling of surface-exposed neurotrophin receptors. Here, we list all experimental details of our labeling protocols, along with examples of the wide range of applications in which these can be used.


Subject(s)
Fluorescent Dyes/chemistry , Nerve Growth Factors/chemistry , Receptors, Nerve Growth Factor/chemistry , Animals , Biotin/chemistry , Cell Line , HEK293 Cells , Humans , Mice , Staining and Labeling
7.
Sci Rep ; 7(1): 5127, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28698670

ABSTRACT

To comprehensively assess whether p75ECD in urine could be a candidate biomarker for ALS evaluation. Urine samples were collected from 101 ALS patients, 108 patients with other neurological disease (OND) and 97 healthy controls. 61 ALS patients were followed up with clinical data including ALSFRS-r every 6 to 12 months, 23 ALS patients died and 17 ALS patients lost touch during follow up period. Enzyme-linked immunoassay was employed to determine urine p75ECD concentration. The ALSFRS-r was employed to assess the severity of ALS. The concentration of p75ECD in ALS was significantly higher than that of OND and CTRL (p < 0.001). Additionally, urine p75ECD concentrations in ALS-definite grade patients were significantly higher than that in ALS-probable grade and ALS-possible grade patients (p < 0.001). Higher urine p75ECD concentrations were correlated with increased clinical stage (p = 0.0309); urine p75ECD concentrations and ALSFRS-r were negatively correlated (p = 0.022); and urine p75ECD concentration in the fast-progressing ALS group was significantly higher than that in slow-progression (p = 0.0026). Our finding indicates that urine p75ECD concentration provides additional evidence for patients with clinically suspected ALS, and can be employed to evaluate ALS-severity.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Biomarkers/urine , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/urine , Receptors, Nerve Growth Factor/chemistry , Adult , Aged , Amyotrophic Lateral Sclerosis/mortality , Amyotrophic Lateral Sclerosis/urine , Asian People , Biomarkers/chemistry , Cohort Studies , Disease Progression , Female , Humans , Male , Middle Aged , Protein Domains , Severity of Illness Index
8.
eNeuro ; 4(3)2017.
Article in English | MEDLINE | ID: mdl-28680965

ABSTRACT

Axonal damage and demyelination are major determinants of disability in patients with peripheral demyelinating neuropathies. The neurotrophin family of growth factors are essential for the normal development and myelination of the peripheral nervous system (PNS), and as such are potential therapeutic candidates for ameliorating axonal and myelin damage. In particular, BDNF promotes peripheral nerve myelination via p75 neurotrophin receptor (p75NTR) receptors. Here, we investigated the therapeutic efficacy of a small structural mimetic of the region of BDNF that binds to p75NTR (cyclo-dPAKKR) in experimental autoimmune neuritis (EAN), an established animal model of peripheral demyelinating neuropathy. Examination of rodents induced with EAN revealed that p75NTR is abundantly expressed in affected peripheral nerves. We found that systemic administration of cyclo-dPAKKR ameliorates EAN disease severity and accelerates recovery. Animals treated with cyclo-dPAKKR displayed significantly better motor performance compared to control animals. Histological assessment revealed that cyclo-dPAKKR administration limits the extent of inflammatory demyelination and axonal damage, and protects against the disruption of nodal architecture in affected peripheral nerves. In contrast, a structural control peptide of cyclo-dPAKKR exerted no influence. Moreover, all the beneficial effects of cyclo-dPAKKR in EAN are abrogated in p75NTR heterozygous mice, strongly suggesting a p75NTR-dependent effect. Taken together, our data demonstrate that cyclo-dPAKKR ameliorates functional and pathological defects of EAN in a p75NTR-dependant manner, suggesting that p75NTR is a therapeutic target to consider for future treatment of peripheral demyelinating diseases and targeting of p75NTR is a strategy worthy of further investigation.


Subject(s)
Axons/drug effects , Demyelinating Diseases/drug therapy , Demyelinating Diseases/etiology , Neuritis, Autoimmune, Experimental , Oligopeptides/therapeutic use , Receptors, Nerve Growth Factor/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Axons/pathology , Axons/ultrastructure , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Transmission , Myelin Basic Protein/metabolism , Neuritis, Autoimmune, Experimental/complications , Neuritis, Autoimmune, Experimental/genetics , Neuritis, Autoimmune, Experimental/pathology , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/therapeutic use , Statistics, Nonparametric
9.
Int J Mol Sci ; 18(4)2017 Mar 24.
Article in English | MEDLINE | ID: mdl-28338624

ABSTRACT

Membrane microdomains or "lipid rafts" have emerged as essential functional modules of the cell, critical for the regulation of growth factor receptor-mediated responses. Herein we describe the dichotomy between caveolin-1 and caveolin-2, structural and regulatory components of microdomains, in modulating proliferation and differentiation. Caveolin-2 potentiates while caveolin-1 inhibits nerve growth factor (NGF) signaling and subsequent cell differentiation. Caveolin-2 does not appear to impair NGF receptor trafficking but elicits prolonged and stronger activation of MAPK (mitogen-activated protein kinase), Rsk2 (ribosomal protein S6 kinase 2), and CREB (cAMP response element binding protein). In contrast, caveolin-1 does not alter initiation of the NGF signaling pathway activation; rather, it acts, at least in part, by sequestering the cognate receptors, TrkA and p75NTR, at the plasma membrane, together with the phosphorylated form of the downstream effector Rsk2, which ultimately prevents CREB phosphorylation. The non-phosphorylatable caveolin-1 serine 80 mutant (S80V), no longer inhibits TrkA trafficking or subsequent CREB phosphorylation. MC192, a monoclonal antibody towards p75NTR that does not block NGF binding, prevents exit of both NGF receptors (TrkA and p75NTR) from lipid rafts. The results presented herein underline the role of caveolin and receptor signaling complex interplay in the context of neuronal development and tumorigenesis.


Subject(s)
Caveolin 1/metabolism , Cell Nucleus/metabolism , Membrane Microdomains/metabolism , Nerve Growth Factor/pharmacology , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal/immunology , CREB-Binding Protein/metabolism , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Caveolin 2/antagonists & inhibitors , Caveolin 2/genetics , Caveolin 2/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Mice , Nerve Tissue Proteins , PC12 Cells , Phosphorylation/drug effects , Protein Binding , Protein Transport/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Rats , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/chemistry , Receptor, trkA/immunology , Receptor, trkA/metabolism , Receptors, Growth Factor , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/immunology , Receptors, Nerve Growth Factor/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
10.
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
11.
J Phys Chem B ; 121(4): 815-824, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28045258

ABSTRACT

Early research has shown that many neurodegenerative diseases are associated with the absence of a short and natural tripeptide sequence, Lys-Phe-Gly (KFG). Herein we report results of both experiments and extensive MD simulations of this tripeptide to understand the self-assembly and morphology as a function of its concentration. Morphologies of the aggregates formed by the tripeptide at low concentration (vesicles), and at high concentration (nanotubes) are studied by several independent 3 µs long Martini coarse-graining MD simulation runs. Further, prediction from MD at still higher concentrations about the formation of rectangular blocks, reported for the first time, has been verified through laboratory experiments. Thus, the computational studies performed are in agreement with the experimental findings observed in our laboratory and a complete control over the formation of various nanostructures is achieved simply by changing the concentration of a short and naturally conserved tripeptide.


Subject(s)
Molecular Dynamics Simulation , Nerve Tissue Proteins/chemistry , Oligopeptides/chemistry , Receptors, Nerve Growth Factor/chemistry , Humans
12.
Biomed Res Int ; 2016: 1674580, 2016.
Article in English | MEDLINE | ID: mdl-27294109

ABSTRACT

The RABVG ectodomain is a homotrimer, and trimers are often called spikes. They are responsible for the attachment of the virus through the interaction with nicotinic acetylcholine receptors, neural cell adhesion molecule (NCAM), and the p75 neurotrophin receptor (p75NTR). This makes them relevant in viral pathogenesis. The antigenic structure differs significantly between the trimers and monomers. Surfaces rich in hydrophobic amino acids are important for trimer stabilization in which the C-terminal of the ectodomain plays an important role; to understand these interactions between the G proteins, a mechanistic study of their functions was performed with a molecular model of G protein in its trimeric form. This verified its 3D conformation. The molecular modeling of G protein was performed by a I-TASSER server and was evaluated via a Rachamandran plot and ERRAT program obtained 84.64% and 89.9% of the residues in the favorable regions and overall quality factor, respectively. The molecular dynamics simulations were carried out on RABVG trimer at 310 K. From these theoretical studies, we retrieved the RMSD values from Cα atoms to assess stability. Preliminary model of G protein of rabies virus stable at 12 ns with molecular dynamics was obtained.


Subject(s)
Antigens, Viral/chemistry , Glycoproteins/chemistry , Imaging, Three-Dimensional/methods , Rabies virus , Viral Envelope Proteins/chemistry , Algorithms , CD56 Antigen/chemistry , Immune System , Molecular Dynamics Simulation , Nerve Tissue Proteins/chemistry , Protein Domains , Protein Multimerization , Protein Structure, Secondary , Receptors, Nerve Growth Factor/chemistry
13.
J Neurosci ; 36(20): 5587-95, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27194337

ABSTRACT

UNLABELLED: The p75 neurotrophin receptor (p75(NTR)) mediates neuronal death in response to neural insults by activating a caspase apoptotic pathway. The oligomeric state and activation mechanism that enable p75(NTR) to mediate these effects have recently been called into question. Here, we have investigated mutant mice lacking the p75(NTR) death domain (DD) or a highly conserved transmembrane (TM) cysteine residue (Cys(259)) implicated in receptor dimerization and activation. Neuronal death induced by proneurotrophins or epileptic seizures was assessed and compared with responses in p75(NTR) knock-out mice and wild-type animals. Proneurotrophins induced apoptosis of cultured hippocampal and cortical neurons from wild-type mice, but mutant neurons lacking p75(NTR), only the p75(NTR) DD, or just Cys(259) were all equally resistant to proneurotrophin-induced neuronal death. Homo-FRET anisotropy experiments demonstrated that both NGF and proNGF induce conformational changes in p75(NTR) that are dependent on the TM cysteine. In vivo, neuronal death induced by pilocarpine-mediated seizures was significantly reduced in the hippocampus and somatosensory, piriform, and entorhinal cortices of all three strains of p75(NTR) mutant mice. Interestingly, the levels of protection observed in mice lacking the DD or only Cys(259) were identical to those of p75(NTR) knock-out mice even though the Cys(259) mutant differed from the wild-type receptor in only one amino acid residue. We conclude that, both in vitro and in vivo, neuronal death induced by p75(NTR) requires the DD and TM Cys(259), supporting the physiological relevance of DD signaling by disulfide-linked dimers of p75(NTR) in the CNS. SIGNIFICANCE STATEMENT: A detailed understanding of the physiological significance of distinct structural determinants in the p75 neurotrophin receptor (p75(NTR)) is crucial for the identification of suitable drug targets in this receptor. We have tested the relevance of the p75(NTR) death domain (DD) and the highly conserved transmembrane residue Cys(259) for the ability of p75(NTR) to induce apoptosis in neurons of the CNS using gene-targeted mutant mice. The physiological importance of these determinants had been contested in some recent in vitro studies. Our results indicate a requirement for DD signaling by disulfide-linked dimers of p75(NTR) for neuronal death induced by proneurotrophins and epileptic seizures. These new mouse models will be useful for clarifying different aspects of p75(NTR) physiology.


Subject(s)
Apoptosis , Cerebral Cortex/metabolism , Mutation , Protein Multimerization , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Animals , COS Cells , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Chlorocebus aethiops , Cysteine/genetics , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Pilocarpine/toxicity , Protein Domains , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/genetics , Seizures/etiology , Seizures/genetics
15.
Elife ; 4: e11692, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26646181

ABSTRACT

Death domains (DDs) mediate assembly of oligomeric complexes for activation of downstream signaling pathways through incompletely understood mechanisms. Here we report structures of complexes formed by the DD of p75 neurotrophin receptor (p75(NTR)) with RhoGDI, for activation of the RhoA pathway, with caspase recruitment domain (CARD) of RIP2 kinase, for activation of the NF-kB pathway, and with itself, revealing how DD dimerization controls access of intracellular effectors to the receptor. RIP2 CARD and RhoGDI bind to p75(NTR) DD at partially overlapping epitopes with over 100-fold difference in affinity, revealing the mechanism by which RIP2 recruitment displaces RhoGDI upon ligand binding. The p75(NTR) DD forms non-covalent, low-affinity symmetric dimers in solution. The dimer interface overlaps with RIP2 CARD but not RhoGDI binding sites, supporting a model of receptor activation triggered by separation of DDs. These structures reveal how competitive protein-protein interactions orchestrate the hierarchical activation of downstream pathways in non-catalytic receptors.


Subject(s)
Nerve Tissue Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , NF-kappa B/metabolism , Nerve Tissue Proteins/chemistry , Protein Binding , Protein Conformation , Protein Multimerization , Receptor-Interacting Protein Serine-Threonine Kinase 2/chemistry , Receptors, Nerve Growth Factor/chemistry , rho-Specific Guanine Nucleotide Dissociation Inhibitors/chemistry
16.
J Neurosci ; 35(34): 11911-20, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311773

ABSTRACT

The p75 neurotrophin receptor (p75(NTR)) is a multifunctional receptor that participates in many critical processes in the nervous system, ranging from apoptosis to synaptic plasticity and morphological events. It is a member of the tumor necrosis factor receptor (TNFR) superfamily, whose members undergo trimeric oligomerization. Interestingly, p75(NTR) interacts with dimeric ligands (i.e., proneurotrophins or mature neurotrophins), but several of the intracellular adaptors that mediate p75(NTR) signaling are trimeric (i.e., TNFR-associated factor 6 or TRAF6). Consequently, the active receptor signaling unit remains uncertain. To identify the functional receptor complex, we evaluated its oligomerization in vitro and in mice brain tissues using a combination of biochemical techniques. We found that the most abundant homotypic arrangement for p75(NTR) is a trimer and that monomers and trimers coexist at the cell surface. Interestingly, trimers are not required for ligand-independent or ligand-dependent p75(NTR) activation in a growth cone retraction functional assay. However, monomers are capable of inducing acute morphological effects in neurons. We propose that p75(NTR) activation is regulated by its oligomerization status and its levels of expression. These results indicate that the oligomeric state of p75(NTR) confers differential responses and offers an explanation for the diverse and contradictory actions of this receptor in the nervous system. SIGNIFICANCE STATEMENT: The p75 neurotrophin receptor (p75(NTR)) regulates a wide range of cellular functions, including apoptosis, neuronal processes remodeling, and synaptic plasticity. The goal of our work was to inquire whether oligomers of the receptor are required for function. Here we report that p75(NTR) predominantly assembles as a trimer, similar to other tumor necrosis factor receptors. Interestingly, monomers and trimers coexist at the cell surface, but trimers are not required for p75(NTR) activation in a functional assay. However, monomers are capable of inducing acute morphological effects in neurons. Identification of the oligomerization state of p75(NTR) begins to provide insights to the mechanisms of signal initiation of this noncatalytic receptor, as well as to develop therapeutic interventions to diminish its activity.


Subject(s)
Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/chemistry , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/chemistry , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Female , HEK293 Cells , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , PC12 Cells , Rats , Receptors, Nerve Growth Factor/genetics , Stereoisomerism
17.
Biophys J ; 109(4): 772-82, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26287629

ABSTRACT

P75NTR is a type I integral membrane protein that plays a key role in neurotrophin signaling. However, structural data for the receptor in various functional states are sparse and controversial. In this work, we studied the spatial structure and mobility of the transmembrane and intracellular parts of p75NTR, incorporated into lipid-protein nanodiscs of various sizes and compositions, by solution NMR spectroscopy. Our data reveal a high level of flexibility and disorder in the juxtamembrane chopper domain of p75NTR, which results in the motions of the receptor death domain being uncoupled from the motions of the transmembrane helix. Moreover, none of the intracellular domains of p75NTR demonstrated a propensity to interact with the membrane or to self-associate under the experimental conditions. The obtained data are discussed in the context of the receptor activation mechanism.


Subject(s)
Membrane Lipids/chemistry , Nanostructures/chemistry , Receptors, Nerve Growth Factor/chemistry , Amino Acid Sequence , Animals , Dimyristoylphosphatidylcholine/chemistry , Escherichia coli , Magnetic Resonance Spectroscopy , Micelles , Molecular Sequence Data , Nerve Tissue Proteins , Phosphatidylglycerols/chemistry , Rats , Receptors, Growth Factor , Receptors, Nerve Growth Factor/genetics
18.
Small ; 11(36): 4626-31, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26097092

ABSTRACT

Multifunctional SiO2 microtubes for targeted drug delivery are produced with precise control over shape and size by combining lithography and electrochemical etching. The hollow core is loaded with a lipophilic anticancer drug generating nanopills and an antibody is conjugated to the external surface for cancer cell targeting. Results demonstrate selective killing of neuroblastoma cells that express the cognate receptor.


Subject(s)
Antineoplastic Agents/chemistry , Camptothecin/administration & dosage , Drug Delivery Systems , Nanomedicine/methods , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Electrochemistry , Humans , Microscopy, Confocal , Microscopy, Electron, Scanning , Microtubules/chemistry , Neoplasms/metabolism , Nerve Tissue Proteins/chemistry , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Receptors, Nerve Growth Factor/chemistry , Surface Properties
19.
Mol Psychiatry ; 20(11): 1301-10, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25917367

ABSTRACT

In Alzheimer's disease (AD), neurodegenerative signals such as amyloid-beta (Aß) and the precursors of neurotrophins, outbalance neurotrophic signals, causing synaptic dysfunction and neurodegeneration. The neurotrophin receptor p75 (p75NTR) is a receptor of Aß and mediates Aß-induced neurodegenerative signals. The shedding of its ectodomain from the cell surface is physiologically regulated; however, the function of the diffusible p75NTR ectodomain (p75ECD) after shedding remains largely not known. Here, we show that p75ECD levels in cerebrospinal fluid and in the brains of Alzheimer's patients and amyloid-beta precursor protein (APP)/PS1 transgenic mice were significantly reduced, due to inhibition of the sheddase-tumor necrosis factor-alpha-converting enzyme by Aß. Restoration of p75ECD to the normal level by brain delivery of the gene encoding human p75ECD before or after Aß deposition in the brain of APP/PS1 mice reversed the behavioral deficits and AD-type pathologies, such as Aß deposit, apoptotic events, neuroinflammation, Tau phosphorylation and loss of dendritic spine, neuronal structures and synaptic proteins. Furthermore, p75ECD can also reduce amyloidogenesis by suppressing ß-secretase expression and activities. Our data demonstrate that p75ECD is a physiologically neuroprotective molecule against Aß toxicity and would be a novel therapeutic target and biomarker for AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Brain/pathology , Nerve Tissue Proteins/chemistry , Protein Structure, Tertiary/physiology , Receptors, Nerve Growth Factor/chemistry , ADAM Proteins/metabolism , ADAM17 Protein , Age Factors , Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Protein Precursor/genetics , Animals , Apoptosis/physiology , Brain/drug effects , Brain/metabolism , Case-Control Studies , Cognition Disorders/etiology , Cognition Disorders/therapy , Disease Models, Animal , Down-Regulation/genetics , Humans , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Presenilin-1/genetics , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Recombinant Proteins/therapeutic use , Transduction, Genetic
20.
Cell Signal ; 27(6): 1225-36, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25748048

ABSTRACT

Direct interaction of α9ß1 integrin with nerve growth factor (NGF) has been previously reported to induce pro-proliferative and pro-survival activities of non-neuronal cells. We investigated participation of p75(NTR) in α9ß1 integrin-dependent cellular response to NGF stimulation. Using selective transfection of glioma cell lines with these receptors, we showed a strong, cation-independent association of α9 integrin subunit with p75(NTR) on the cellular membrane by selective immunoprecipitation experiments. The presence of the α9/p75(NTR) complex increases NGF-dependent cell adhesion, proliferation and migration. Other integrin subunits including ß1 were not found in complex with p75(NTR). FRET analysis indicated that p75(NTR) and α9 integrin subunit are not closely associated through their cytoplasmic domains, most probably because of the molecular interference with other cytoplasmic proteins such as paxillin. Interaction of α9ß1 integrin with another ligand, VCAM-1 was not modulated by the p75(NTR). α9/p75(NTR) complex elevated NGF-dependent activation of MAPK Erk1/2 arty for integrin that may create active complexes with other types of receptors belonging to the TNF superfamily.


Subject(s)
Cell Proliferation/drug effects , Integrins/metabolism , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Immunohistochemistry , Integrins/chemistry , Integrins/genetics , Mice , Microscopy, Confocal , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factor/isolation & purification , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Paxillin/metabolism , Protein Binding , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/genetics , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...