Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
STAR Protoc ; 4(4): 102691, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37925637

ABSTRACT

G-protein-coupled receptors (GPCRs) are important therapeutic targets expressed on the cell surface. Here, we present a protocol for identifying physiologically relevant binding proteins of adhesion GPCR GPR110. We describe steps for in-cell chemical crosslinking, immunoprecipitation, and quantitative high-resolution mass spectrometry. Notably, we detail a label-free quantitation strategy that eliminates irrelevant interacting proteins using an inactive GPR110 mutant with impaired surface expression. Furthermore, we outline procedures for validating the identified partners. For complete details on the use and execution of this protocol, please refer to Huang et al. (2023).1.


Subject(s)
Carrier Proteins , Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/genetics , Cell Membrane , Immunoprecipitation , Mass Spectrometry
2.
iScience ; 26(4): 106550, 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37123219

ABSTRACT

Activation of adhesion receptor GPR110 by the endogenous ligand synaptamide promotes neurogenesis, neurite growth, and synaptogenesis in developing brains through cAMP signal transduction. However, interacting partners of GPR110 and their involvement in cellular function remain unclear. Here, we demonstrate using chemical crosslinking, affinity purification, and quantitative mass spectrometry that GPR110 interacts with the tight junction adhesion protein occludin. By removing non-specific partners by comparing the binding proteins of GPR110 WT and an inactive mutant exhibiting impaired surface expression, occludin was distinguished as a true binding partner which was further confirmed by reciprocal co-immunoprecipitation assay. Deletion of GPR110 in mice led to the disruption of blood-brain barrier (BBB) and reduced occludin phosphorylation at Y285 in the brain. The Y285 phosphorylation increased upon the ligand-induced activation of GPR110. These data suggest an important role of GPR110-occludin interaction in BBB function and association of previously unknown GPR110-dependent occludin phosphorylation at Y285 with BBB integrity.

3.
Int J Mol Sci ; 23(9)2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35563025

ABSTRACT

The neurodevelopmental and neuroprotective actions of docosahexaenoic acid (DHA) are mediated by mechanisms involving membrane- and metabolite-related signal transduction. A key characteristic in the membrane-mediated action of DHA results from the stimulated synthesis of neuronal phosphatidylserine (PS). The resulting DHA-PS-rich membrane domains facilitate the translocation and activation of kinases such as Raf-1, protein kinase C (PKC), and Akt. The activation of these signaling pathways promotes neuronal development and survival. DHA is also metabolized in neural tissues to bioactive mediators. Neuroprotectin D1, a docosatriene synthesized by the lipoxygenase activity, has an anti-inflammatory property, and elovanoids formed from DHA elongation products exhibit antioxidant effects in the retina. Synaptamide, an endocannabinoid-like lipid mediator synthesized from DHA in the brain, promotes neurogenesis and synaptogenesis and exerts anti-inflammatory effects. It binds to the GAIN domain of the GPR110 (ADGRF1) receptor, triggers the cAMP/protein kinase A (PKA) signaling pathway, and activates the cAMP-response element binding protein (CREB). The DHA status in the brain influences not only the PS-dependent signal transduction but also the metabolite formation and expression of pre- and post-synaptic proteins that are downstream of the CREB and affect neurotransmission. The combined actions of these processes contribute to the neurodevelopmental and neuroprotective effects of DHA.


Subject(s)
Docosahexaenoic Acids , Neuroprotection , Anti-Inflammatory Agents/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Endocannabinoids/metabolism , Signal Transduction
4.
Commun Biol ; 3(1): 109, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144388

ABSTRACT

Adhesion G protein-coupled receptors (aGPCR) are characterized by a large extracellular region containing a conserved GPCR-autoproteolysis-inducing (GAIN) domain. Despite their relevance to several disease conditions, we do not understand the molecular mechanism by which aGPCRs are physiologically activated. GPR110 (ADGRF1) was recently deorphanized as the functional receptor of N-docosahexaenoylethanolamine (synaptamide), a potent synaptogenic metabolite of docosahexaenoic acid. Thus far, synaptamide is the first and only small-molecule endogenous ligand of an aGPCR. Here, we demonstrate the molecular basis of synaptamide-induced activation of GPR110 in living cells. Using in-cell chemical cross-linking/mass spectrometry, computational modeling and mutagenesis-assisted functional assays, we discover that synaptamide specifically binds to the interface of GPR110 GAIN subdomains through interactions with residues Q511, N512 and Y513, causing an intracellular conformational change near TM6 that triggers downstream signaling. This ligand-induced GAIN-targeted activation mechanism provides a framework for understanding the physiological function of aGPCRs and therapeutic targeting in the GAIN domain.


Subject(s)
Ethanolamines/pharmacology , Oncogene Proteins/agonists , Receptors, G-Protein-Coupled/agonists , Binding Sites , Ethanolamines/metabolism , HEK293 Cells , Humans , Ligands , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutation , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Binding , Protein Domains , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
5.
Phytochem Anal ; 29(4): 375-386, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29785715

ABSTRACT

INTRODUCTION: Medicinal plants have played an important role in maintaining human health for thousands of years. However, the interactions between the active components in medicinal plants and some certain biological targets during a disease are still unclear in most cases. OBJECTIVE: To conduct the high-throughput screening for small active molecules that can interact with biological targets, which is of great theoretical significance and practical value. METHODOLOGY: The ultrafiltration mass spectrometry (UF-LC/MS) is a powerful bio-analytical method by combining affinity ultrafiltration and liquid chromatography-mass spectrometry (LC/MS), which could rapidly screen and identify small active molecules that bind to biological targets of interest at the same time. Compared with other analytical methods, affinity UF-LC/MS has the characteristics of fast, sensitive and high throughput, and is especially suitable for the complicated extracts of medicinal plants. RESULTS: In this review, the basic principle, characteristics and some most recent challenges in UF-LC/MS have been demonstrated. Meanwhile, the progress and applications of affinity UF-LC/MS in the discovery of the active components from natural medicinal plants and the interactions between small molecules and biological target proteins are also briefly summarised. In addition, the future directions for UF-LC/MS are also prospected. CONCLUSION: Affinity UF-LC/MS is a powerful tool in studies on the interactions between small active molecules and biological protein targets, especially in the high-throughput screening of active components from the natural medicinal plants.


Subject(s)
Chromatography, Affinity/methods , Mass Spectrometry/methods , Plants, Medicinal/chemistry , Ultrafiltration/methods , Drugs, Chinese Herbal/chemistry , HT29 Cells , Hep G2 Cells , Humans
6.
Sci Rep ; 7(1): 11673, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28916818

ABSTRACT

Akt plays a major role in tumorigenesis and the development of specific Akt inhibitors as effective cancer therapeutics has been challenging. Here, we report the identification of a highly specific allosteric inhibitor of Akt through a FRET-based high-throughput screening, and characterization of its inhibitory mechanism. Out of 373,868 compounds screened, 4-phenylquinolin-2(1H)-one specifically decreased Akt phosphorylation at both T308 and S473, and inhibited Akt kinase activity (IC50 = 6 µM) and downstream signaling. 4-Phenylquinolin-2(1H)-one did not alter the activity of upstream kinases including PI3K, PDK1, and mTORC2 as well as closely related kinases that affect cell proliferation and survival such as SGK1, PKA, PKC, or ERK1/2. This compound inhibited the proliferation of cancer cells but displayed less toxicity compared to inhibitors of PI3K or mTOR. Kinase profiling efforts revealed that 4-phenylquinolin-2(1H)-one does not bind to the kinase active site of over 380 human kinases including Akt. However, 4-phenylquinolin-2(1H)-one interacted with the PH domain of Akt, apparently inducing a conformation that hinders S473 and T308 phosphorylation by mTORC2 and PDK1. In conclusion, we demonstrate that 4-phenylquinolin-2(1H)-one is an exquisitely selective Akt inhibitor with a distinctive molecular mechanism, and a promising lead compound for further optimization toward the development of novel cancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Quinolones/pharmacology , Allosteric Regulation , Animals , Antineoplastic Agents/isolation & purification , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Enzyme Inhibitors/isolation & purification , Fluorescence Resonance Energy Transfer , High-Throughput Screening Assays , Mice , Neurons/drug effects , Neurons/physiology , Protein Binding , Quinolones/isolation & purification
7.
Nat Commun ; 7: 13123, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759003

ABSTRACT

Docosahexaenoic acid (DHA, 22:6n-3) is an omega-3 fatty acid essential for proper brain development. N-docosahexaenoylethanolamine (synaptamide), an endogenous metabolite of DHA, potently promotes neurogenesis, neuritogenesis and synaptogenesis; however, the underlying molecular mechanism is not known. Here, we demonstrate orphan G-protein coupled receptor 110 (GPR110, ADGRF1) as the synaptamide receptor, mediating synaptamide-induced bioactivity in a cAMP-dependent manner. Mass spectrometry-based proteomic characterization and cellular fluorescence tracing with chemical analogues of synaptamide reveal specific binding of GPR110 to synaptamide, which triggers cAMP production with low nM potency. Disruption of this binding or GPR110 gene knockout abolishes while GPR110 overexpression enhances synaptamide-induced bioactivity. GPR110 is highly expressed in fetal brains but rapidly decreases after birth. GPR110 knockout mice show significant deficits in object recognition and spatial memory. GPR110 deorphanized as a functional synaptamide receptor provides a novel target for neurodevelopmental control and new insight into mechanisms by which DHA promotes brain development and function.


Subject(s)
Cognition/physiology , Docosahexaenoic Acids/metabolism , Endocannabinoids/physiology , Neurogenesis/physiology , Oncogene Proteins/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Brain/cytology , Cell Line , Cyclic AMP/metabolism , Endocannabinoids/metabolism , Female , Gene Knockout Techniques , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Models, Animal , Neurons/physiology , Oncogene Proteins/metabolism , Primary Cell Culture , Proteomics/methods , Rats , Receptors, G-Protein-Coupled/metabolism , Recognition, Psychology/physiology , Signal Transduction/physiology , Spatial Memory/physiology
8.
Neurobiol Aging ; 41: 73-85, 2016 May.
Article in English | MEDLINE | ID: mdl-27103520

ABSTRACT

Aging has been related to diminished cognitive function, which could be a result of ineffective synaptic function. We have previously shown that synaptic plasma membrane proteins supporting synaptic integrity and neurotransmission were downregulated in docosahexaenoic acid (DHA)-deprived brains, suggesting an important role of DHA in synaptic function. In this study, we demonstrate aging-induced synaptic proteome changes and DHA-dependent mitigation of such changes using mass spectrometry-based protein quantitation combined with western blot or messenger RNA analysis. We found significant reduction of 15 synaptic plasma membrane proteins in aging brains including fodrin-α, synaptopodin, postsynaptic density protein 95, synaptic vesicle glycoprotein 2B, synaptosomal-associated protein 25, synaptosomal-associated protein-α, N-methyl-D-aspartate receptor subunit epsilon-2 precursor, AMPA2, AP2, VGluT1, munc18-1, dynamin-1, vesicle-associated membrane protein 2, rab3A, and EAAT1, most of which are involved in synaptic transmission. Notably, the first 9 proteins were further reduced when brain DHA was depleted by diet, indicating that DHA plays an important role in sustaining these synaptic proteins downregulated during aging. Reduction of 2 of these proteins was reversed by raising the brain DHA level by supplementing aged animals with an omega-3 fatty acid sufficient diet for 2 months. The recognition memory compromised in DHA-depleted animals was also improved. Our results suggest a potential role of DHA in alleviating aging-associated cognitive decline by offsetting the loss of neurotransmission-regulating synaptic proteins involved in synaptic function.


Subject(s)
Aging/genetics , Aging/metabolism , Brain/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteome , Synapses/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Aging/psychology , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Down-Regulation , Fatty Acids, Omega-3/administration & dosage , Female , Male , Membrane Proteins/physiology , Memory , Mice, Inbred C57BL , Pregnancy , Recognition, Psychology
9.
Int J Biochem Cell Biol ; 64: 195-201, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25912234

ABSTRACT

Akt is a key mediator of cell proliferation, survival and metabolism. After translocation to the membrane and phosphorylation at T308 and S473, the activated Akt dissociates from the plasma membrane to cytoplasm, which is an important step to phosphorylate its downstream targets. In addition to its central role in regulating the kinase activity, phosphorylation of T308 in the kinase loop has been reported to be necessary for this dissociation process. However, it is not clear whether the membrane detachment requires further mechanisms. In the present report, we demonstrate that membrane dissociation of Akt requires phosphoinositide-dependent protein kinase 1 (PDK1) which directly phosphorylates not only T308 but also T34 in the pleckstrin homology (PH) domain. Like T308, T34 was phosphorylated in a phosphatidylinositol 3,4,5-trisphosphate- and phosphatidylserine-dependent manner. Phosphorylation of T34 also occurred in cells following growth factor stimulation, concurrently with T308 phosphorylation. Moreover, when T34 was mutated to aspartic acid (T34D) to mimic its phosphorylation, Akt-membrane association assessed by surface plasmon resonance spectroscopy was significantly reduced. In cells, this mutation impaired the IGF-induced Akt membrane translocation and subsequent phosphorylation at T308 and S473. Taken together, our results demonstrate that T34 phosphorylation by PDK1 promotes the membrane dissociation of activated Akt for its downstream action through attenuating membrane binding affinity. This membrane dissociation mechanism offers a new insight for Akt activation process and provides a potential new target for controlling the Akt-dependent cellular processes.


Subject(s)
Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Tumor , Cell Membrane/enzymology , Mice , Phosphorylation , Protein Binding , Protein Transport , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Somatomedins/physiology , Threonine/metabolism
10.
Prog Lipid Res ; 56: 1-18, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24992464

ABSTRACT

Phosphatidylserine (PS) is the major anionic phospholipid class particularly enriched in the inner leaflet of the plasma membrane in neural tissues. PS is synthesized from phosphatidylcholine or phosphatidylethanolamine by exchanging the base head group with serine, and this reaction is catalyzed by phosphatidylserine synthase 1 and phosphatidylserine synthase 2 located in the endoplasmic reticulum. Activation of Akt, Raf-1 and protein kinase C signaling, which supports neuronal survival and differentiation, requires interaction of these proteins with PS localized in the cytoplasmic leaflet of the plasma membrane. Furthermore, neurotransmitter release by exocytosis and a number of synaptic receptors and proteins are modulated by PS present in the neuronal membranes. Brain is highly enriched with docosahexaenoic acid (DHA), and brain PS has a high DHA content. By promoting PS synthesis, DHA can uniquely expand the PS pool in neuronal membranes and thereby influence PS-dependent signaling and protein function. Ethanol decreases DHA-promoted PS synthesis and accumulation in neurons, which may contribute to the deleterious effects of ethanol intake. Improvement of some memory functions has been observed in cognitively impaired subjects as a result of PS supplementation, but the mechanism is unclear.


Subject(s)
Brain/metabolism , Docosahexaenoic Acids/metabolism , Neurons/metabolism , Phosphatidylserines/metabolism , Animals , Biosynthetic Pathways , Brain/cytology , Cell Differentiation , Cell Survival , Humans , Models, Biological , Neurons/cytology
11.
PLoS One ; 8(4): e61430, 2013.
Article in English | MEDLINE | ID: mdl-23613850

ABSTRACT

Akt is a critical protein for cell survival and known to interact with various proteins. However, Akt binding partners that modulate or regulate Akt activation have not been fully elucidated. Identification of Akt-interacting proteins has been customarily achieved by co-immunoprecipitation combined with western blot and/or MS analysis. An intrinsic problem of the method is loss of interacting proteins during procedures to remove non-specific proteins. Moreover, antibody contamination often interferes with the detection of less abundant proteins. Here, we developed a novel two-step chemical crosslinking strategy to overcome these problems which resulted in a dramatic improvement in identifying Akt interacting partners. Akt antibody was first immobilized on protein A/G beads using disuccinimidyl suberate and allowed to bind to cellular Akt along with its interacting proteins. Subsequently, dithiobis[succinimidylpropionate], a cleavable crosslinker, was introduced to produce stable complexes between Akt and binding partners prior to the SDS-PAGE and nanoLC-MS/MS analysis. This approach enabled identification of ten Akt partners from cell lysates containing as low as 1.5 mg proteins, including two new potential Akt interacting partners. None of these but one protein was detectable without crosslinking procedures. The present method provides a sensitive and effective tool to probe Akt-interacting proteins. This strategy should also prove useful for other protein interactions, particularly those involving less abundant or weakly associating partners.


Subject(s)
Cross-Linking Reagents/pharmacology , Immunoprecipitation/methods , Proto-Oncogene Proteins c-akt/metabolism , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Animals , Blotting, Western , Cell Line, Tumor , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/chemistry , Somatomedins/pharmacology
12.
Proteomics ; 13(3-4): 424-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23125184

ABSTRACT

Reversible phosphorylation, tightly controlled by protein kinases and phosphatases, plays a central role in mediating biological processes, such as protein-protein interactions, subcellular translocation, and activation of cellular enzymes. MS-based phosphoproteomics has now allowed the detection and quantification of tens of thousands of phosphorylation sites from a typical biological sample in a single experiment, which has posed new challenges in functional analysis of each and every phosphorylation site on specific signaling phosphoproteins of interest. In this article, we review recent advances in the functional analysis of targeted phosphorylation carried out by various chemical and biological approaches in combination with the MS-based phosphoproteomics. This review focuses on three types of strategies, including forward functional analysis, defined for the result-driven phosphoproteomics efforts in determining the substrates of a specific protein kinase; reverse functional analysis, defined for tracking the kinase(s) for specific phosphosite(s) derived from the discovery-driven phosphoproteomics efforts; and MS-based analysis on the structure-function relationship of phosphoproteins. It is expected that this review will provide a state-of-the-art overview of functional analysis of site-specific phosphorylation and explore new perspectives and outline future challenges.


Subject(s)
Phosphoproteins/metabolism , Protein Processing, Post-Translational , Proteome/metabolism , Amino Acid Motifs , Animals , Deuterium Exchange Measurement , Humans , Phosphoprotein Phosphatases/metabolism , Phosphoproteins/chemistry , Phosphorylation , Protein Interaction Mapping , Protein Kinases/metabolism , Proteome/chemistry , Proteomics , Signal Transduction
13.
J Biol Chem ; 287(4): 2579-90, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22128152

ABSTRACT

Bacterial elongation factor P (EF-P) is the ortholog of archaeal and eukaryotic initiation factor 5A (eIF5A). EF-P shares sequence homology and crystal structure with eIF5A, but unlike eIF5A, EF-P does not undergo hypusine modification. Recently, two bacterial genes, yjeA and yjeK, encoding truncated homologs of class II lysyl-tRNA synthetase and of lysine-2,3-aminomutase, respectively, have been implicated in the modification of EF-P to convert a specific lysine to a hypothetical ß-lysyl-lysine. Here we present biochemical evidence for ß-lysyl-lysine modification in Escherichia coli EF-P and for its role in EF-P activity by characterizing native and recombinant EF-P proteins for their modification status and activity in vitro. Mass spectrometric analyses confirmed the lysyl modification at lysine 34 in native and recombinant EF-P proteins. The ß-lysyl-lysine isopeptide was identified in the exhaustive Pronase digests of native EF-P and recombinant EF-P isolated from E. coli coexpressing EF-P, YjeA, and YjeK but not in the digests of proteins derived from the vectors encoding EF-P alone or EF-P together with YjeA, indicating that both enzymes, YjeA and YjeK, are required for ß-lysylation of EF-P. Endogenous EF-P as well as the recombinant EF-P preparation containing ß-lysyl-EF-P stimulated N-formyl-methionyl-puromycin synthesis ∼4-fold over the preparations containing unmodified EF-P and/or α-lysyl-EF-P. The mutant lacking the modification site lysine (K34A) was inactive. This is the first report of biochemical evidence for the ß-lysylation of EF-P in vivo and the requirement for this modification for the activity of EF-P.


Subject(s)
Deoxyribonucleases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Lysine/metabolism , Peptide Elongation Factors/metabolism , Protein Processing, Post-Translational/physiology , Deoxyribonucleases/chemistry , Deoxyribonucleases/genetics , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Lysine/chemistry , Lysine/genetics , Mass Spectrometry , Peptide Elongation Factors/chemistry , Peptide Elongation Factors/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
ACS Chem Biol ; 7(2): 387-94, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22129086

ABSTRACT

Although PI3K/Akt signaling that regulates neuronal survival has been implicated in the deleterious effects of ethanol on the central nervous system, underlying molecular mechanisms have not been fully elucidated. Akt-membrane interaction is a prerequisite step for Akt activation since it induces interdomain conformational changes to an open conformer that allows Akt phosphorylation by upstream kinases. In this study, we investigated the effect of ethanol on Akt activation by quantitatively probing Akt conformation using chemical cross-linking, (18)O labeling and mass spectrometry. We found that ethanol at pharmacologically relevant concentrations (20 or 170 mM) directly interacts with Akt and alters the local pleckstrin homology domain configuration near the PIP(3)-binding site. We also found that ethanol significantly impairs subsequent membrane-induced interdomain conformational changes needed for Akt activation. The observed alteration of Akt conformation caused by ethanol during the activation sequence provides a new molecular basis for the effects of ethanol on Akt signaling. The in vitro conformation-based approach employed in this study should also be useful in probing the molecular mechanisms for the action of ethanol or drugs on other signaling proteins, particularly for those undergoing dramatic conformational change during activation processes such as members of AGC kinase super family.


Subject(s)
Ethanol/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Membrane/metabolism , Cross-Linking Reagents/chemistry , Enzyme Activation , Humans , Mass Spectrometry , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/chemistry , Unilamellar Liposomes/metabolism
15.
J Proteome Res ; 10(12): 5472-80, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22003853

ABSTRACT

Docosahexenoic acid (DHA, 22:6n-3) plays an important role in development of proper brain function in mammals. We have previously reported that DHA promotes synaptogenesis and synaptic function in hippocampal neurons while DHA-depletion in the brain due to n-3 fatty acid deficiency produces opposite effects. To gain insight into underlying molecular mechanisms, we investigated whether the brain DHA status affects the synaptic plasma membrane (SPM) proteome by using nanoLC-ESI-MS/MS and (16)O/(18)O labeling. The DHA level in mouse brains was lowered by dietary depletion of n-3 fatty acids, and SPM was prepared by differential centrifugation followed by osmotic shock. SPM proteins from DHA-adequate and depleted brains were analyzed by nanoLC-ESI-MS/MS after SDS-PAGE, in-gel digestion, and differential O(18)/O(16) labeling. This strategy allowed comparative quantitation of more than 200 distinct membrane or membrane-associated proteins from DHA-adequate or depleted brains. We found that 18 pre- and postsynaptic proteins that are relevant to synaptic physiology were significantly down-regulated in DHA-depleted mouse brains. The protein network analysis suggests involvement of CREB and caspase-3 pathways in the DHA-dependent modulation of synaptic proteome. Reduction of specific synaptic proteins due to brain DHA-depletion may be an important mechanism for the suboptimal brain function associated with n-3 fatty acid deficiency.


Subject(s)
Cerebral Cortex/drug effects , Docosahexaenoic Acids/pharmacology , Isotope Labeling/methods , Proteome/analysis , Synaptic Membranes/drug effects , Animals , Blotting, Western , Centrifugation/methods , Cerebral Cortex/chemistry , Cyclic AMP Response Element-Binding Protein/chemistry , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Fatty Acids, Omega-3/chemistry , Female , Mass Spectrometry/methods , Membrane Proteins/analysis , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Osmotic Pressure , Oxygen Isotopes/chemistry , Pregnancy , Proteome/chemistry , Synapses/chemistry , Synaptic Membranes/chemistry
16.
J Cell Biol ; 192(6): 979-92, 2011 Mar 21.
Article in English | MEDLINE | ID: mdl-21402788

ABSTRACT

Akt activation relies on the binding of Akt to phosphatidylinositol-3,4,5-trisphosphate (PIP(3)) in the membrane. Here, we demonstrate that Akt activation requires not only PIP(3) but also membrane phosphatidylserine (PS). The extent of insulin-like growth factor-induced Akt activation and downstream signaling as well as cell survival under serum starvation conditions positively correlates with plasma membrane PS levels in living cells. PS promotes Akt-PIP(3) binding, participates in PIP(3)-induced Akt interdomain conformational changes for T308 phosphorylation, and causes an open conformation that allows for S473 phosphorylation by mTORC2. PS interacts with specific residues in the pleckstrin homology (PH) and regulatory (RD) domains of Akt. Disruption of PS-Akt interaction by mutation impairs Akt signaling and increases susceptibility to cell death. These data identify a critical function of PS for Akt activation and cell survival, particularly in conditions with limited PIP(3) availability. The novel molecular interaction mechanism for Akt activation suggests potential new targets for controlling Akt-dependent cell survival and proliferation.


Subject(s)
Inositol 1,4,5-Trisphosphate/metabolism , Phosphatidylserines/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/physiology , CHO Cells , Cell Membrane/chemistry , Cell Survival , Cricetinae , Cricetulus , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Mice , Models, Molecular , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/chemistry , Signal Transduction/physiology
17.
Biochem J ; 435(2): 327-36, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21281269

ABSTRACT

DHA (docosahexaenoic acid, C22:6,n-3) has been shown to promote neurite growth and synaptogenesis in embryonic hippocampal neurons, supporting the importance of DHA known for hippocampus-related learning and memory function. In the present study, we demonstrate that DHA metabolism to DEA (N-docosahexaenoylethanolamide) is a significant mechanism for hippocampal neuronal development, contributing to synaptic function. We found that a fatty acid amide hydrolase inhibitor URB597 potentiates DHA-induced neurite growth, synaptogenesis and synaptic protein expression. Active metabolism of DHA to DEA was observed in embryonic day 18 hippocampal neuronal cultures, which was increased further by URB597. Synthetic DEA promoted hippocampal neurite growth and synaptogenesis at substantially lower concentrations in comparison with DHA. DEA-treated neurons increased the expression of synapsins and glutamate receptor subunits and exhibited enhanced glutamatergic synaptic activity, as was the case for DHA. The DEA level in mouse fetal hippocampi was altered according to the maternal dietary supply of n-3 fatty acids, suggesting that DEA formation is a relevant in vivo process responding to the DHA status. In conclusion, DHA metabolism to DEA is a significant biochemical mechanism for neurite growth, synaptogenesis and synaptic protein expression, leading to enhanced glutamatergic synaptic function. The novel DEA-dependent mechanism offers a new molecular insight into hippocampal neurodevelopment and function.


Subject(s)
Docosahexaenoic Acids/analogs & derivatives , Ethanolamines/pharmacology , Hippocampus/drug effects , Hippocampus/embryology , Neurons/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Docosahexaenoic Acids/pharmacology , Drug Evaluation, Preclinical , Embryo, Mammalian , Endocannabinoids , Female , Maternal Nutritional Physiological Phenomena , Mice , Mice, Inbred C57BL , Neurites/drug effects , Neurites/physiology , Neurogenesis/drug effects , Neurons/physiology , Pregnancy
18.
Rapid Commun Mass Spectrom ; 23(13): 1885-91, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19462409

ABSTRACT

Amide hydrogen exchange coupled to nano-electrospray ionization mass spectrometry (nano-ESI-MS) has been used to identify and characterize localized conformational changes of Akt upon activation. Active or inactive Akt was incubated in D(2)O buffer, digested with pepsin, and analyzed by nano-ESI-MS to determine the deuterium incorporation. The hydrogen/deuterium (H/D) exchange profiles revealed that Akt undergoes considerable conformational changes in the core structures of all three individual domains after activation. In the PH domain, four beta-strand (beta1, beta2 beta5 and beta6) regions containing membrane-binding residues displayed higher solvent accessibility in the inactive state, suggesting that the PH domain is readily available for the binding to the plasma membrane for activation. In contrast, these beta-strands became less exposed or more folded in the active form, which is favored for the dissociation of Akt from the membrane. The beginning alpha-helix J region and the C-terminal locus (T450-470P) of the regulatory domain showed less folded structures that probably enable substrate entry. Our data also revealed detailed conformational changes of Akt in the kinase domain due to activation, some of which may be attributed to the interaction of the basic residues with phosphorylation sites. Our H/D exchange results indicating the conformational status of Akt at different activation states provided new insight for the regulation of this critical protein involved in cell survival.


Subject(s)
Deuterium/chemistry , Hydrogen/chemistry , Proto-Oncogene Proteins c-akt/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Amides/chemistry , Amino Acid Sequence , Enzyme Activation , Molecular Sequence Data , Peptide Mapping , Protein Conformation , Protein Structure, Tertiary
19.
J Am Soc Mass Spectrom ; 20(8): 1504-13, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19446470

ABSTRACT

The serine/threonine kinase Akt is a critical enzyme that regulates cell survival. As high Akt activity has been shown to contribute to the pathogenesis of various human malignancies, inhibition of Akt activation is a promising therapeutic strategy for cancers. We have previously demonstrated that changes in Akt interdomain arrangements from a closed to open conformation occur upon Akt-membrane interaction, which in turn allows Akt phosphorylation/activation. In the present study, we demonstrate a novel strategy to discern mechanisms for Akt inhibition based on Akt conformational changes using chemical cross-linking and (18)O labeling mass spectrometry. By quantitative comparison of two interdomain cross-linked peptides, which represent the proximity of the domains involved, we found that the binding of Akt to an inhibitor (PI analog) caused the open interdomain conformation where the PH and regulatory domains moved away from the kinase domain, even before interacting with membranes, subsequently preventing translocation of Akt to the plasma membrane. In contrast, the interdomain conformation remained unchanged after incubating with another type of inhibitor (peptide TCL1). Subsequent interaction with unilamellar vesicles suggested that TCL1 impaired particularly the opening of the PH domain for exposing T308 for phosphorylation at the plasma membrane. This novel approach based on the conformation-based molecular interaction mechanism should be potentially useful for drug discovery efforts for specific Akt inhibitors or anti-tumor agents.


Subject(s)
Mass Spectrometry/methods , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/ultrastructure , Binding Sites , Cross-Linking Reagents/chemistry , Protein Binding , Protein Interaction Mapping
20.
Neuropharmacology ; 54(1): 1-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17631919

ABSTRACT

Endocannabinoids, including anandamide (arachidonoyl ethanolamide) have been implicated in the regulation of a growing number of physiological and pathological processes. Anandamide can be generated from its membrane phospholipid precursor N-arachidonoyl phosphatidylethanolamine (NAPE) through hydrolysis by a phospholipase D (NAPE-PLD). Recent evidence indicates, however, the existence of two additional, parallel pathways. One involves the sequential deacylation of NAPE by alpha,beta-hydrolase 4 (Abhd4) and the subsequent cleavage of glycerophosphate to yield anandamide, and the other one proceeds through phospholipase C-mediated hydrolysis of NAPE to yield phosphoanandamide, which is then dephosphorylated by phosphatases, including the tyrosine phosphatase PTPN22 and the inositol 5' phosphatase SHIP1. Conversion of synthetic NAPE to AEA by brain homogenates from wild-type and NAPE-PLD(-/-) mice can proceed through both the PLC/phosphatase and Abdh4 pathways, with the former being dominant at shorter (<10 min) and the latter at longer (60 min) incubations. In macrophages, the endotoxin-induced synthesis of anandamide proceeds uniquely through the phospholipase C/phosphatase pathway.


Subject(s)
Arachidonic Acids/biosynthesis , Metabolic Networks and Pathways/physiology , Animals , Cell Line, Transformed , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Drug Interactions , Endocannabinoids , Glycerophosphates/metabolism , Hydrolases/metabolism , Hydrolysis/drug effects , Inositol Polyphosphate 5-Phosphatases , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Metabolic Networks and Pathways/drug effects , Mice , Mice, Knockout , Neomycin/pharmacology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phospholipase D/deficiency , Phosphoric Monoester Hydrolases/deficiency , Polyunsaturated Alkamides , Protein Synthesis Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/deficiency , RNA, Small Interfering/metabolism , Transfection/methods , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...