Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 159: 105517, 2021 11.
Article in English | MEDLINE | ID: mdl-34563643

ABSTRACT

Huntington's disease (HD) is a genetically inherited neurodegenerative disorder caused by expansion of a polyglutamine (polyQ) repeat in the exon-1 of huntingtin protein (HTT). The expanded polyQ enhances the amyloidogenic propensity of HTT exon 1 (HTTex1), which forms a heterogeneous mixture of assemblies with a broad neurotoxicity spectrum. While predominantly intracellular, monomeric and aggregated mutant HTT species are also present in the cerebrospinal fluids of HD patients, however, their biological properties are not well understood. To explore the role of extracellular mutant HTT in aggregation and toxicity, we investigated the uptake and amplification of recombinant HTTex1 assemblies in cell culture models. We find that small HTTex1 fibrils preferentially enter human neurons and trigger the amplification of neurotoxic assemblies; astrocytes or epithelial cells are not permissive. The amplification of HTTex1 in neurons depletes endogenous HTT protein with non-pathogenic polyQ repeat, activates apoptotic caspase-3 pathway and induces nuclear fragmentation. Using a panel of novel monoclonal antibodies and genetic mutation, we identified epitopes within the N-terminal 17 amino acids and proline-rich domain of HTTex1 to be critical in neural uptake and amplification. Synaptosome preparations from the brain homogenates of HD mice also contain mutant HTT species, which enter neurons and behave similar to small recombinant HTTex1 fibrils. These studies suggest that amyloidogenic extracellular mutant HTTex1 assemblies may preferentially enter neurons, propagate and promote neurodegeneration.


Subject(s)
Astrocytes/metabolism , Epithelial Cells/metabolism , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Neurons/metabolism , Protein Aggregation, Pathological/metabolism , Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/metabolism , Animals , Apoptosis , Caspase 3 , Exons , Gene Knock-In Techniques , Humans , Huntingtin Protein/genetics , Mice , Mice, Transgenic , Mutation , Peptides/genetics , Protein Aggregation, Pathological/genetics , Synaptosomes
2.
Nat Commun ; 12(1): 4272, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34257293

ABSTRACT

The first exon of the huntingtin protein (HTTex1) important in Huntington's disease (HD) can form cross-ß fibrils of varying toxicity. We find that the difference between these fibrils is the degree of entanglement and dynamics of the C-terminal proline-rich domain (PRD) in a mechanism analogous to polyproline film formation. In contrast to fibril strains found for other cross-ß fibrils, these HTTex1 fibril types can be interconverted. This is because the structure of their polyQ fibril core remains unchanged. Further, we find that more toxic fibrils of low entanglement have higher affinities for protein interactors and are more effective seeds for recombinant HTTex1 and HTTex1 in cells. Together these data show how the structure of a framing sequence at the surface of a fibril can modulate seeding, protein-protein interactions, and thereby toxicity in neurodegenerative disease.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/metabolism , Neurodegenerative Diseases/metabolism , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Neurodegenerative Diseases/genetics , Peptides/chemistry , Peptides/metabolism , Protein Interaction Maps
3.
J Biol Chem ; 297(1): 100804, 2021 07.
Article in English | MEDLINE | ID: mdl-34044018

ABSTRACT

The functional amyloid Orb2 belongs to the cytoplasmic polyadenylation element binding (CPEB) protein family and plays an important role in long-term memory formation in Drosophila. The Orb2 domain structure combines RNA recognition motifs with low-complexity sequences similar to many RNA-binding proteins shown to form protein droplets via liquid-liquid phase separation (LLPS) in vivo and in vitro. This similarity suggests that Orb2 might also undergo LLPS. However, cellular Orb2 puncta have very little internal protein mobility, and Orb2 forms fibrils in Drosophila brains that are functionally active indicating that LLPS might not play a role for Orb2. In the present work, we reconcile these two views on Orb2 droplet formation. Using fluorescence microscopy, we show that soluble Orb2 can indeed phase separate into protein droplets. However, fluorescence recovery after photobleaching (FRAP) data shows that these droplets have either no or only an extremely short-lived liquid phase and appear maturated right after formation. Orb2 fragments that lack the C-terminal RNA-binding domain (RBD) form fibrils out of these droplets. Solid-state NMR shows that these fibrils have well-ordered static domains in addition to the Gln/His-rich fibril core. Further, we find that full-length Orb2B, which is by far the major component of Orb2 fibrils in vivo, does not transition into fibrils but remains in the droplet phase. Together, our data suggest that phase separation might play a role in initiating the formation of functional Orb2 fibrils.


Subject(s)
Amyloid/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Amino Acid Sequence , Amyloid/ultrastructure , Animals , Benzothiazoles/metabolism , Carbon Isotopes , Drosophila Proteins/chemistry , Drosophila melanogaster/ultrastructure , Fluorescence , Osmolar Concentration , Protein Domains , Transcription Factors/chemistry , mRNA Cleavage and Polyadenylation Factors/chemistry
4.
Biochemistry ; 57(28): 4206-4213, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29928798

ABSTRACT

Structure-based "rational" drug design strategies fail for diseases associated with intrinsically disordered proteins (IDPs). However, structural disorder allows large-amplitude spontaneous intramolecular dynamics in a protein. We demonstrate a method that exploits this dynamics to provide quantitative information about the degree of interaction of an IDP with other molecules. A candidate ligand molecule may not bind strongly, but even momentary interactions can be expected to perturb the fluctuations. We measure the amplitude and frequency of the equilibrium fluctuations of fluorescently labeled small oligomers of hIAPP (an IDP associated with type II diabetes) in a physiological solution, using nanosecond fluorescence cross-correlation spectroscopy. We show that the interterminal distance fluctuates at a characteristic time scale of 134 ± 10 ns, and 6.4 ± 0.2% of the population is in the "closed" (quenched) state at equilibrium. These fluctuations are affected in a dose-dependent manner by a series of small molecules known to reduce the toxicity of various amyloid peptides. The degree of interaction increases in the following order: resveratrol < epicatechin ∼ quercetin < Congo red < epigallocatechin 3-gallate. Such ordering can provide a direction for exploring the chemical space for finding stronger-binding ligands. We test the biological relevance of these measurements by measuring the effect of these molecules on the affinity of hIAPP for lipid vesicles and cell membranes. We find that the ability of a molecule to modulate intramolecular fluctuations correlates well with its ability to lower membrane affinity. We conclude that structural disorder may provide new avenues for rational drug design for IDPs.


Subject(s)
Drug Design , Drug Discovery , Intrinsically Disordered Proteins/chemistry , Islet Amyloid Polypeptide/chemistry , Small Molecule Libraries/pharmacology , Drug Discovery/methods , Humans , Intrinsically Disordered Proteins/metabolism , Islet Amyloid Polypeptide/metabolism , Ligands , Liposomes/metabolism , Molecular Dynamics Simulation , Protein Binding , Protein Multimerization/drug effects , Small Molecule Libraries/chemistry
5.
Biochim Biophys Acta Biomembr ; 1860(9): 1863-1875, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29702073

ABSTRACT

Abnormal protein aggregation is a hallmark of various human diseases. α-Synuclein, a protein implicated in Parkinson's disease, is found in aggregated form within Lewy bodies that are characteristically observed in the brains of PD patients. Similarly, deposits of aggregated human islet amyloid polypeptide (IAPP) are found in the pancreatic islets in individuals with type 2 diabetes mellitus. Significant number of studies have focused on how monomeric, disaggregated proteins transition into various amyloid structures leading to identification of a vast number of aggregation promoting molecules and processes over the years. Inasmuch as these factors likely enhance the formation of toxic, misfolded species, they might act as risk factors in disease. Cellular membranes, and particularly certain lipids, are considered to be among the major players for aggregation of α-synuclein and IAPP, and membranes might also be the target of toxicity. Past studies have utilized an array of biophysical tools, both in vitro and in vivo, to expound the membrane-mediated aggregation. Here, we focus on membrane interaction of α-synuclein and IAPP, and how various kinds of membranes catalyze or modulate the aggregation of these proteins and how, in turn, these proteins disrupt membrane integrity, both in vitro and in vivo. The membrane interaction and subsequent aggregation has been briefly contrasted to aggregation of α-synuclein and IAPP in solution. This article is part of a Special Issue entitled: Protein Aggregation and Misfolding at the Cell Membrane Interface edited by Ayyalusamy Ramamoorthy.

6.
Biochim Biophys Acta Biomembr ; 1860(9): 1734-1740, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29626442

ABSTRACT

Islet amyloid polypeptide (IAPP) is a 37 residue intrinsically disordered protein whose aggregation is associated with Type II diabetes. Like most amyloids, it appears that the intermediate aggregates ("oligomers") of IAPP are more toxic than the mature fibrils, and interaction with the cell membrane is likely to be an integral component of the toxicity. Here we probe the membrane affinity and the conformation of the peptide as a function of its aggregation state. We find that the affinity of the peptide for artificial lipid bilayers is more than 15 times higher in the small oligomeric state (hydrodynamic radius ~ 1.6 nm) compared to the monomeric state (hydrodynamic radius ~ 0.7 nm). Binding with RIN-m5F cell membranes also shows qualitatively similar behavior. The monomeric state, as determined by Forster Resonance Energy Transfer, has a much larger end to end distance than the oligomeric state, suggesting conformational change between the monomers and the oligomers. Raman and Infrared spectroscopic measurements show the presence of considerable alpha helical content in the oligomers, whereas the larger aggregates have largely beta sheet character. Therefore, the conformation of the small oligomers is distinct from both the smaller monomers and the larger oligomers, and this is associated with an enhanced membrane affinity. This provides a possible structural basis for the enhanced toxicity of amyloid oligomers. Such change is also reminiscent of amyloid beta, another aggregation prone amyloidogenic peptide, though the nature of the conformational change is quite different in the two cases. We infer that conformational change underlying oligomer formation is a key factor in determining the enhanced membrane affinity of disease causing oligomers, but the toxic "oligomer fold" may not be universal.

7.
J Biol Chem ; 293(7): 2597-2605, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29282287

ABSTRACT

Aggregation of huntingtin protein arising from expanded polyglutamine (polyQ) sequences in the exon-1 region of mutant huntingtin plays a central role in the pathogenesis of Huntington's disease. The huntingtin aggregation pathways are of therapeutic and diagnostic interest, but obtaining critical information from the physiologically relevant htt exon-1 (Httex1) protein has been challenging. Using biophysical techniques and an expression and purification protocol that generates clean, monomeric Httex1, we identified and mapped three distinct aggregation pathways: 1) unseeded in solution; 2) seeded in solution; and 3) membrane-mediated. In solution, aggregation proceeded in a highly stepwise manner, in which the individual domains (N terminus containing 17 amino acids (N17), polyQ, and proline-rich domain (PRD)) become ordered at very different rates. The aggregation was initiated by an early oligomer requiring a pathogenic, expanded Gln length and N17 α-helix formation. In the second phase, ß-sheet forms in the polyQ. The slowest step is the final structural maturation of the PRD. This stepwise mechanism could be bypassed by seeding, which potently accelerated aggregation and was a prerequisite for prion-like spreading in vivo Remarkably, membranes could catalyze aggregation even more potently than seeds, in a process that caused significant membrane damage. The N17 governed membrane-mediated aggregation by anchoring Httex1 to the membrane, enhancing local concentration and promoting collision via two-dimensional diffusion. Considering its central roles in solution and in membrane-mediated aggregation, the N17 represents an attractive target for inhibiting multiple pathways. Our approach should help evaluate such inhibitors and identify diagnostic markers for the misfolded forms identified here.


Subject(s)
Cell Membrane/metabolism , Huntingtin Protein/chemistry , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Amino Acid Motifs , Cell Membrane/chemistry , Cell Membrane/genetics , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Kinetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Aggregates , Protein Conformation, alpha-Helical , Protein Domains
8.
ACS Chem Neurosci ; 9(3): 469-474, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29226666

ABSTRACT

Monoamine neurotransmission is key to neuromodulation, but imaging monoamines in live neurons has remained a challenge. Here we show that externally added ortho-phthalaldehyde (OPA) can permeate live cells and form bright fluorogenic adducts with intracellular monoamines (e.g., serotonin, dopamine, and norepinephrine) and with L-DOPA, which can be imaged sensitively using conventional single-photon excitation in a fluorescence microscope. The peak excitation and emission wavelengths (λex = 401 nm and λem = 490 nm for serotonin; λex = 446 nm and λem = 557 nm for dopamine; and λex = 446 nm and λem = 544 nm for norepinephrine, respectively) are accessible to most modern confocal imaging instruments. The identity of monoamine containing structures (possibly neurotransmitter vesicles) in serotonergic RN46A cells is established by quasi-simultaneous imaging of serotonin using three-photon excitation microscopy. Mass spectrometry of cell extracts and of in vitro solutions helps us identify the chemical nature of the adducts and establishes the reaction mechanisms. Our method has low toxicity, high selectivity, and the ability to directly report the location and concentration of monoamines in live cells.


Subject(s)
Dopamine/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Serotonin/metabolism , Humans , Mass Spectrometry/methods , Neuroimaging/methods , Norepinephrine/metabolism
9.
J Phys Chem B ; 121(8): 1835-1842, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28140589

ABSTRACT

Shape complementarity between close-packed residues plays a critical role in the amyloid aggregation process. Here, we probe such "steric zipper" interactions in amyloid-ß (Aß40), whose aggregation is linked to Alzheimer's disease, by replacing natural residues by their stereoisomers. Such mutations are expected to specifically destabilize the shape sensitive "packing" interactions, which may potentially increase their solubility and change other properties. We study the stereomutants DF19 and DL34 and also the DA2/DF4/DH6/DS8 mutant of Aß40. F19-L34 is a critical contact in a tightly packed region of Aß, while residues 1-9 are known to be disordered. While both DF19 and DL34 slow down the kinetics of aggregation and form amyloid fibrils efficiently, only DL34 increases the final solubility. DF19 gives rise to additional off-pathway aggregation which results in large, kinetically stable aggregates, and has lower net solubility. DA2/DF4/DH6/DS8 does not have an effect on the kinetics or the solubility. Notably, both DF19 and DL34 oligomers have a significantly lower level of interactions with lipid vesicles and live cells. We conclude that stereoisomers can cause complex site dependent changes in amyloid properties, and provide an effective tool to determine the role of individual residues in shaping the packed interiors of amyloid aggregates.


Subject(s)
Amyloid beta-Peptides/chemistry , Peptide Fragments/chemistry , Protein Aggregates , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/ultrastructure , Animals , Cell Line , Cell Membrane/metabolism , Humans , Mutation , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/ultrastructure , Rats , Solubility , Stereoisomerism
10.
J Biol Inorg Chem ; 22(1): 47-59, 2017 01.
Article in English | MEDLINE | ID: mdl-27822620

ABSTRACT

Amyloid-ß peptides and their metal-associated aggregated states have been implicated in the pathogenesis of Alzheimer's disease. The present paper epitomises the design and synthesis of a small, neutral, lipophilic benzothiazole Schiff base (E)-2-((6-chlorobenzo[d]thiazol-2-ylimino)methyl)-5-diethylamino)phenol (CBMDP), and explores its multifunctionalty as a potential metal chelator/fluorophore using UV-visible absorption, steady-state fluorescence, single molecule fluorescence correlation spectroscopic (FCS) techniques which is further corroborated by in silico studies. Some pharmaceutically relevant properties of the synthesized compound have also been calculated theoretically. Steady-state fluorescence and single molecule FCS reveal that the synthesized CBMDP not only recognizes oligomeric Aß40, but could also be used as an amyloid-specific extrinsic fluorophore as it shows tremendous increase in its emission intensity in the presence of Aß40. Molecular docking exercise and MD simulation reveal that CBMDP localizes itself in the crucial amyloidogenic and copper-binding region of Aß40 and undergoes a strong binding interaction via H-bonding and π-π stacking. It stabilizes the solitary α-helical Aß40 monomer by retaining the initial conformation of the Aß central helix and mostly interacts with the hydrophilic N-terminus and the α-helical region spanning from Ala-2 to Val-24. CBMDP exhibits strong copper as well as zinc chelation ability and retards the rapid copper-induced aggregation of amyloid peptide. In addition, CBMDP shows radical scavenging activity which enriches its functionality. Overall, the consolidated in vitro and in silico results obtained for the synthesized molecule could provide a rational template for developing new multifunctional agents.


Subject(s)
Chelating Agents/chemistry , Chelating Agents/pharmacology , Drug Discovery , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Spectrum Analysis , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Caco-2 Cells , Chelating Agents/metabolism , Heterocyclic Compounds/metabolism , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Protein Stability , Protein Structure, Secondary , Reactive Oxygen Species/metabolism , Schiff Bases/chemistry
11.
ACS Chem Neurosci ; 6(8): 1290-5, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-25951510

ABSTRACT

Small hydrophobic oligomers of aggregation-prone proteins are thought to be generically toxic. Here we examine this view by perturbing an early folding contact between Phe19 and Leu34 formed during the aggregation of Alzheimer's amyloid-ß (Aß40) peptide. We find that even conservative single mutations altering this interaction can abolish Aß40 toxicity. Significantly, the mutants are not distinguishable either by the oligomers size or by the end-state fibrillar structure from the wild type Aß40. We trace the change in their toxicity to a drastic lowering of membrane affinity. Therefore, nonlocal folding contacts play a key role in steering the oligomeric intermediates through specific conformations with very different properties and toxicity levels. Our results suggest that engineering the folding energy landscape may provide an alternative route to Alzheimer therapeutics.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Amyloid beta-Peptides/toxicity , Animals , Cell Survival , Cells, Cultured , Cerebral Cortex/physiology , Membranes, Artificial , Mutation , Neurons/physiology , Peptide Fragments/toxicity , Phosphatidylcholines/chemistry , Phosphatidylglycerols/chemistry , Protein Folding , Rats, Wistar , Unilamellar Liposomes/chemistry
12.
Curr Top Med Chem ; 14(6): 740-6, 2014.
Article in English | MEDLINE | ID: mdl-24444154

ABSTRACT

The process of self-assembly is universal and lies at the heart of biological structures and function. Peptide aggregation, while considered a nuisance in peptide chemistry, soon gained interest with the discovery of pore-forming peptide toxins and had been an area of intense research during last century and even to date. This has also resulted in the increasing use of the more respectable term peptide self-assembly. The discovery of amyloid forming peptides has rekindled the interest in peptide self-assembly since such aggregates are directly implicated in many debilitating diseases in human and animals. Amyloid aggregates have posed many fundamental questions to researchers. In addition, self-assembly of peptides has emerged as a bottom-up strategy for the fabrication of nanostructures owing to highly ordered nature of the process and considerable degree of flexibility and diversity provided by peptides as starting materials. This review provides a brief account of the progress in the field of peptide self-assembly from pore-forming toxins to amyloid forming peptides and those forming nanostructures.


Subject(s)
Amyloid/chemistry , Nanotubes/chemistry , Peptides/chemical synthesis , Toxins, Biological/chemistry , Peptides/chemistry
13.
J Pept Sci ; 19(12): 770-83, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24243599

ABSTRACT

Aggregation of a polypeptide chain into highly ordered amyloid aggregates is a complex process. Various factors, both extrinsic and intrinsic to the polypeptide chain, have been shown to perturb this process, leading to a drastic change in the amyloidogenic behavior, which is reflected in the polymorphism of amyloid aggregates at various levels of self-assembly. In this paper, we have investigated the ability of covalently linked long-chain fatty acids in modulating the self-assembly of an aromatic amino acid-rich highly amyloidogenic sequence derived from the amino acid region 59-71 of human ß2-microglobulin by thioflavin T (ThT) fluorescence microscopy, circular dichroism, and fluorescence spectroscopy. Our results indicate that under identical conditions of dissolution and concentration, each peptide enhances the fluorescence of ThT. However, the aggregates are morphologically distinct. For the same peptide, the aggregate morphologies are dependent on peptide concentration. Further, an optimum concentration, which varies with solution ionic strength, is required for the formation of fibrillar aggregates. We show that covalent modification of this amyloidogenic sequence, with long-chain fatty acids, affects the way the higher order amyloid structures assemble from the cross-ß units, in fatty acyl chain-dependent and position-dependent manner. Our data indicate that noncovalent interactions leading to amyloid fibril formation can be modulated by the hydrophobicity of covalently attached long-chain fatty acids resulting in self-assembly of the peptide chain to form nonfibrillar aggregates.


Subject(s)
Amyloid/chemistry , Myristic Acid/chemistry , Palmitic Acid/chemistry , Peptide Fragments/chemistry , beta 2-Microglobulin/chemistry , Acylation , Amino Acid Sequence , Amyloid/ultrastructure , Benzothiazoles , Circular Dichroism , Fluorescent Dyes/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Microscopy, Fluorescence , Protein Multimerization , Protein Stability , Protein Structure, Quaternary , Protein Structure, Secondary , Thiazoles/chemistry
14.
J Biosci ; 38(1): 63-71, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23385814

ABSTRACT

The SH4 domain of Src family of nonreceptor protein tyrosine kinases represents the extreme N-terminal 1-16 amino acid region which mediates membrane association of these proteins and facilitates their functions. The SH4 domains among Src members lack well-defined sequence consensus and vary in the net charge. However, they readily anchor to the cytoplasmic face of the plasma membrane upon fatty acid acylation. Here, we report the membrane association of differentially acylated SH4 domain of Lck kinase, which has net negative charge at physiological pH. Our results suggest that despite the net negative charge, the SH4 domain of Lck associates with membranes upon fatty acid acylation. While myristoylation at the N-terminus is sufficient for providing membrane anchorage, multiple acylation determines orientation of the peptide chain with respect to the lipid bilayer. Hence, fatty acylation serves more than just a lipid anchor. It has an important role in regulating the spatial orientation of the peptide domain with respect to the lipid bilayer, which could be important for the interaction of the other domains of these kinases with their partners.


Subject(s)
Fatty Acids/chemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Membrane Lipids/chemistry , Peptides/chemistry , Unilamellar Liposomes/chemistry , Acylation , Amino Acid Sequence , Binding Sites , Fluorescence Resonance Energy Transfer , Molecular Sequence Data , Myristic Acid/chemistry , Peptides/chemical synthesis , Protein Binding , Protein Structure, Tertiary , Solid-Phase Synthesis Techniques , Static Electricity
15.
Biochim Biophys Acta ; 1798(10): 1854-63, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20599685

ABSTRACT

The SH4 domain of Fyn, a member of the Src family of tyrosine kinases, though rich in polar amino acid residues, anchors to the cytosolic face of membranes upon fatty acylation. In order to probe the requirement of specific fatty acylation at the N-terminus and at the side-chain of this domain for membrane-association, we have studied the interaction of peptides corresponding to the polar segment of the SH4 domain of Fyn and its mono- and dually fatty acylated analogs with model membranes. While the polar segment without covalently linked fatty acids (KDKEATKLTEW-amide) does not interact with lipid vesicles, peptides with one covalently linked fatty acid at the N-terminus or in the side-chain, associate with zwitterionic and anionic lipids to varying degrees. The interaction of dually acylated peptides (Myr-GK(epsilon-myr)KDKEATKLTEW-amide and Myr-GC(S-pal)KDKEATKLTEW-amide) with lipids depends on the linkage between fatty acyl side-chain and peptide backbone. The peptide chain associates with membranes only when the side-chain acylation is via an amide bond and not via a thioester bond. Our investigations indicate that acylation is essential for membrane targeting and unacylated polar stretch of the SH4 domain does not have a role in membrane-anchoring. Side-chain acylation via a thioester bond not only provides membrane anchorage but also directs the peptide chain away from the bilayer which might be important to enable the full length protein to interact with other signaling partners.


Subject(s)
Lipid Bilayers/chemistry , Membrane Lipids/chemistry , Peptides/chemistry , Proto-Oncogene Proteins c-fyn/chemistry , Amino Acid Sequence , Binding Sites , Fatty Acids/chemistry , Fatty Acids/metabolism , Lipid Bilayers/metabolism , Membrane Lipids/metabolism , Models, Molecular , Myristic Acid/chemistry , Myristic Acid/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Protein Binding , Spectrometry, Fluorescence , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...