Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Mol Biol ; 436(6): 168495, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38360090

ABSTRACT

Under prebiotic conditions, peptides are capable of self-replication through a structure-based template-assisted mechanism when they form amyloids. Furthermore, peptide amyloids can spontaneously form inside fatty acid vesicles creating membrane enclosed complex structures of variable morphologies. This is possible because fatty acid vesicle membranes act as filters allowing passage of activated amino acids while some amino acids derived from the activated species become non-permeable and trapped in the vesicles. Similarly, nascent peptides derived from the condensation of the activated amino acids are also trapped in the vesicles. It is hypothesized that such preselected peptide amyloids become a sequence pool for the emergence of proteins in life and that after billions of years of cellular evolution, the sequences in the current proteome have diverged significantly from these original seed peptides. If this hypothesis is correct, it could be possible to detect the traces of these seed sequences in current proteomes. Here, we show for all possible 3, 6, 7, 8 or 9 residue sequence motifs that those motifs that are most amyloidogenic/aggregation prone are over-represented in extant proteomes compared to a sequence-randomized proteome. Furthermore, we find that there is a greater proportion of amyloidogenic sequence motifs in archaea proteomes than in the larger primate proteomes. This suggests that the evolution towards larger proteomes leads to smaller proportion of amyloidogenic sequences.


Subject(s)
Amyloid , Peptides , Proteome , Animals , Amino Acids/chemistry , Amyloid/chemistry , Fatty Acids , Peptides/chemistry , Evolution, Molecular
2.
Int J Mol Sci ; 24(18)2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37762631

ABSTRACT

Protein-fragment complex structures are particularly sought after in medicinal chemistry to rationally design lead molecules. These structures are usually derived using X-ray crystallography, but the failure rate is non-neglectable. NMR is a possible alternative for the calculation of weakly interacting complexes. Nevertheless, the time-consuming protein signal assignment step remains a barrier to its routine application. NMR Molecular Replacement (NMR2) is a versatile and rapid method that enables the elucidation of a protein-ligand complex structure. It has been successfully applied to peptides, drug-like molecules, and more recently to fragments. Due to the small size of the fragments, ca < 300 Da, solving the structures of the protein-fragment complexes is particularly challenging. Here, we present the expected performances of NMR2 when applied to protein-fragment complexes. The NMR2 approach has been benchmarked with the SERAPhic fragment library to identify the technical challenges in protein-fragment NMR structure calculation. A straightforward strategy is proposed to increase the method's success rate further. The presented work confirms that NMR2 is an alternative method to X-ray crystallography for solving protein-fragment complex structures.


Subject(s)
Benchmarking , Magnetic Resonance Imaging , Chemistry, Pharmaceutical , Crystallography, X-Ray , Gene Library
3.
Angew Chem Int Ed Engl ; 60(10): 5561-5568, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33325627

ABSTRACT

Cellular life requires a high degree of molecular complexity and self-organization, some of which must have originated in a prebiotic context. Here, we demonstrate how both of these features can emerge in a plausibly prebiotic system. We found that chemical gradients in simple mixtures of activated amino acids and fatty acids can lead to the formation of amyloid-like peptide fibrils that are localized inside of a proto-cellular compartment. In this process, the fatty acid or lipid vesicles act both as a filter, allowing the selective passage of activated amino acids, and as a barrier, blocking the diffusion of the amyloidogenic peptides that form spontaneously inside the vesicles. This synergy between two distinct building blocks of life induces a significant increase in molecular complexity and spatial order thereby providing a route for the early molecular evolution that could give rise to a living cell.


Subject(s)
Amino Acids/chemistry , Amyloidogenic Proteins/chemistry , Liposomes/chemistry , Origin of Life , Peptides/chemistry , Amino Acids/metabolism , Amyloidogenic Proteins/metabolism , Decanoic Acids/chemistry , Decanoic Acids/metabolism , Liposomes/metabolism , Oleic Acid/chemistry , Oleic Acid/metabolism , Peptides/metabolism , Permeability , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Protein Multimerization
4.
Orig Life Evol Biosph ; 49(4): 213-224, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31845164

ABSTRACT

Prebiotic chemical replication is a commonly assumed precursor to and prerequisite for life and as such is the one of the goals of our research. We have previously reported on the role that short peptide amyloids could have played in a template-based chemical elongation. Here we take a step closer to the goal by reproducing amyloid-templated peptide elongation with carbonyl sulfide (COS) in place of the less-prebiotically relevant carbonyldiimidazole (CDI) used in the earlier study. Our investigation shows that the sequence-selectivity and stereoselectivity of the amyloid-templated reaction is similar for both activation chemistries. Notably, the amyloid protects the peptides from some of the side-reactions that take place with the COS-activation.


Subject(s)
Evolution, Chemical , Origin of Life , Peptides/chemistry , Sulfur Oxides/chemistry
5.
Biochemistry ; 58(39): 4017-4027, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31365236

ABSTRACT

The human voltage-gated proton channel [Hv1(1) or VSDO(2)] plays an important role in the human innate immune system. Its structure differs considerably from those of other cation channels. It is built solely of a voltage-sensing domain and thus lacks the central pore domain, which is essential for other cation channels. Here, we determined the solution structure of an N- and C-terminally truncated human Hv1 (Δ-Hv1) in the resting state by nuclear magnetic resonance (NMR) spectroscopy. Δ-Hv1 comprises the typical voltage-sensing antiparallel four-helix bundle (S1-S4) preceded by an amphipathic helix (S0). The solution structure corresponds to an intermediate state between resting and activated forms of voltage-sensing domains. Furthermore, Zn2+-induced closing of proton channel Δ-Hv1 was studied with two-dimensional NMR spectroscopy, which showed that characteristic large scale dynamics of open Δ-Hv1 are absent in the closed state of the channel. Additionally, pH titration studies demonstrated that a higher H+ concentration is required for the protonation of side chains in the Zn2+-induced closed state than in the open state. These observations demonstrate both structural and dynamical changes involved in the process of voltage gating of the Hv1 channel and, in the future, may help to explain the unique properties of unidirectional conductance and the exceptional ion selectivity of the channel.


Subject(s)
Ion Channel Gating , Ion Channels/chemistry , Magnetic Resonance Spectroscopy/methods , Basic-Leucine Zipper Transcription Factors/chemistry , Crystallization , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Hydrogen Bonding , Hydrogen-Ion Concentration , Ion Channels/genetics , Kinetics , Models, Molecular , Phosphoric Monoester Hydrolases/chemistry , Protein Binding , Protein Structure, Secondary , Protons , Saccharomyces cerevisiae Proteins/chemistry , Zinc/chemistry
6.
Biophys J ; 115(12): 2336-2347, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30503535

ABSTRACT

Interactions between biological membranes and disease-associated amyloids are well documented, and their prevalence suggests that an inherent affinity exists between these molecular assemblies. Our interest in the molecular origins of life have led us to investigate the nature of such interactions in the context of their molecular predecessors (i.e., vesicle-forming amphiphiles and small peptides). Under certain conditions, amyloidogenic peptides or fatty acids are each able to form ordered structures on their own; however, we report here on their cooperative assembly into novel, to our knowledge, highly ordered structures. We first examined an amyloidogenic eight-residue peptide, which forms amyloids at pH 11, yet because of its positive electrostatic character remains soluble at a neutral pH. In mixtures with simple fatty acids, this peptide is also able to form novel, to our knowledge, coaggregates at a neutral pH whose structures are sensitive to both the fatty acid concentration and identity. Below the critical vesicle concentration, the mixtures of fatty acid and peptide yield a flocculent precipitate with an underlying ß-structure. Above the critical vesicle concentration, the mixtures yield a translucent precipitate that consists of tube-like structures. Small-angle x-ray scattering and fiber diffraction data were used to model their structures as hollow-core two-shell cylinders in which the inner shell is a bilayer of fatty acid and the outer shell alternates between amyloid and bilayers of fatty acid. The further analysis of decanoic acid with a panel of 13 other basic amyloidogenic peptides confirmed the general nature of the observed interactions. The cooperativity within this heterogeneous system is attributed to the structurally repetitive natures of the fatty acid bilayer and the cross-ß-sheet motif, providing compatible scaffolds for attractive electrostatic interactions. We show these interactions to be mutually beneficial, expanding the phase space of both peptides and fatty acids while providing a simple yet robust physical connection between two distinct entities relevant for life.


Subject(s)
Fatty Acids/metabolism , Oligopeptides/metabolism , Amino Acid Sequence , Amyloid/chemistry , Hydrogen-Ion Concentration , Oligopeptides/chemistry , Protein Binding
7.
J Mol Biol ; 430(20): 3735-3750, 2018 10 12.
Article in English | MEDLINE | ID: mdl-29890117

ABSTRACT

How life can emerge from non-living matter is one of the fundamental mysteries of the universe. A bottom-up approach to this problem focuses on the potential chemical precursors of life, in particular the nature of the first replicative molecules. Such thinking has led to the currently most popular idea: that an RNA-like molecule played a central role as the first replicative and catalytic molecule. Here, we review an alternative hypothesis that has recently gained experimental support, focusing on the role of amyloidogenic peptides rather than nucleic acids, in what has been by some termed "the amyloid-world" hypothesis. Amyloids are well-ordered peptide aggregates that have a fibrillar morphology due to their underlying structure of a one-dimensional crystal-like array of peptides in a ß-strand conformation. While they are notorious for their implication in several neurodegenerative diseases including Alzheimer's disease, amyloids also have many biological functions. In this review, we will elaborate on the following properties of amyloids in relation to their fitness as a prebiotic entity: they can be formed by very short peptides with simple amino acids sequences; as aggregates they are more chemically stable than their isolated component peptides; they can possess diverse catalytic activities; they can form spontaneously during the prebiotic condensation of amino acids; they can act as templates in their own chemical replication; they have a structurally repetitive nature that enables them to interact with other structurally repetitive biopolymers like RNA/DNA and polysaccharides, as well as with structurally repetitive surfaces like amphiphilic membranes and minerals.


Subject(s)
Amyloid/chemistry , Amyloid/metabolism , Origin of Life , Peptides/chemistry , Peptides/metabolism , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/metabolism , Biophysical Phenomena , Catalysis , Cell Membrane/chemistry , Cell Membrane/metabolism , Models, Molecular , Prebiotics , Protein Conformation , Protein Multimerization , RNA/genetics , RNA/metabolism , Repetitive Sequences, Nucleic Acid , Signal Transduction , Structure-Activity Relationship
8.
J Mol Biol ; 428(19): 3737-51, 2016 09 25.
Article in English | MEDLINE | ID: mdl-27473602

ABSTRACT

S-Nitrosylation is well established as an important post-translational regulator in protein function and signaling. However, relatively little is known about its structural and dynamical consequences. We have investigated the effects of S-nitrosylation on the rhodanese domain of the Escherichia coli integral membrane protein YgaP by NMR, X-ray crystallography, and mass spectrometry. The results show that the active cysteine in the rhodanese domain of YgaP is subjected to two competing modifications: S-nitrosylation and S-sulfhydration, which are naturally occurring in vivo. It has been observed that in addition to inhibition of the sulfur transfer activity, S-nitrosylation of the active site residue Cys63 causes an increase in slow motion and a displacement of helix 5 due to a weakening of the interaction between the active site and the helix dipole. These findings provide an example of how nitrosative stress can exert action at the atomic level.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Protein Processing, Post-Translational , Thiosulfate Sulfurtransferase/chemistry , Thiosulfate Sulfurtransferase/metabolism , Catalytic Domain , Crystallography, X-Ray , Cysteine/metabolism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Protein Conformation
9.
Trends Pharmacol Sci ; 35(12): 648-57, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25458539

ABSTRACT

TGF-ß superfamily ligands govern normal tissue development and homeostasis, and their dysfunction is a hallmark of many diseases. These ligands are also well defined both structurally and functionally. This review focuses on TGF-ß superfamily ligand engineering for therapeutic purposes, in particular for regenerative medicine and musculoskeletal disorders. We describe the key discovery that structure-guided mutation of receptor-binding epitopes, especially swapping of these epitopes between ligands, results in new ligands with unique functional properties that can be harnessed clinically. Given the promising results with prototypical engineered TGF-ß superfamily ligands, and the vast number of such molecules that remain to be produced and tested, this strategy is likely to hold great promise for the development of new biologics.


Subject(s)
Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/metabolism , Animals , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Protein Engineering/methods , Protein Structure, Secondary , Structure-Activity Relationship
10.
J Endocrinol ; 223(1): 35-45, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25100748

ABSTRACT

Activins and bone morphogenetic proteins (BMPs) share activin type 2 signaling receptors but utilize different type 1 receptors and Smads. We designed AB215, a potent BMP2-like Activin A/BMP2 chimera incorporating the high-affinity type 2 receptor-binding epitope of Activin A. In this study, we compare the signaling properties of AB215 and BMP2 in HEK293T cells and gonadotroph LßT2 cells in which Activin A and BMP2 synergistically induce FSHß. In HEK293T cells, AB215 is more potent than BMP2 and competitively blocks Activin A signaling, while BMP2 has a partial blocking activity. Activin A signaling is insensitive to BMP pathway antagonism in HEK293T cells but is strongly inhibited by constitutively active (CA) BMP type 1 receptors. By contrast, the potencies of AB215 and BMP2 are indistinguishable in LßT2 cells and although AB215 blocks Activin A signaling, BMP2 has no inhibitory effect. Unlike HEK293T, Activin A signaling is strongly inhibited by BMP pathway antagonism in LßT2 cells but is largely unaffected by CA BMP type 1 receptors. BMP2 increases phospho-Smad3 levels in LßT2 cells, in both the absence and the presence of Activin A treatment, and augments Activin A-induced FSHß. AB215 has the opposite effect and sharply decreases basal phospho-Smad3 levels and blocks Smad2 phosphorylation and FSHß induction resulting from Activin A treatment. These findings together demonstrate that while AB215 activates the BMP pathway, it has opposing effects to those of BMP2 on FSHß induction in LßT2 cells apparently due to its ability to block Activin A signaling.


Subject(s)
Activins/pharmacology , Bone Morphogenetic Protein 2/pharmacology , Follicle Stimulating Hormone, beta Subunit/genetics , Gonadotrophs/drug effects , Recombinant Fusion Proteins/pharmacology , Activins/genetics , Activins/metabolism , Animals , Blotting, Western , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cell Line , Gonadotrophs/cytology , Gonadotrophs/metabolism , HEK293 Cells , Humans , Mice , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcriptional Activation/drug effects
11.
BMC Cancer ; 14: 549, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25070479

ABSTRACT

BACKGROUND: One in eight women will be affected by breast cancer in her lifetime. Approximately 75% of breast cancers express estrogen receptor alpha (ERα) and/or progesterone receptor and these receptors are markers for tumor dependence on estrogen. Anti-estrogenic drugs such as tamoxifen are commonly used to block estrogen-mediated signaling in breast cancer. However, many patients either do not respond to these therapies (de novo resistance) or develop resistance to them following prolonged treatment (acquired resistance). Therefore, it is imperative to continue efforts aimed at developing new efficient and safe methods of targeting ER activity in breast cancer. METHODS: AB215 is a chimeric ligand assembled from sections of Activin A and BMP2. BMP2's and AB215's inhibition of breast cancer cells growth was investigated. In vitro luciferase and MTT proliferation assays together with western blot, RT_PCR, and mRNA knockdown methods were used to determine the mechanism of inhibition of estrogen positive breast cancer cells growth by BMP2 and AB215. Additionally in vivo xenograft tumor model was used to investigate anticancer properties of AB215. RESULTS: Here we report that AB215, a chimeric ligand assembled from sections of Activin A and BMP2 with BMP2-like signaling, possesses stronger anti-proliferative effects on ERα positive breast cancer cells than BMP2. We further show that AB215 inhibits estrogen signaling by inducing expression of inhibitor of DNA binding proteins (IDs). Specifically, we demonstrate that knockdown of ID proteins attenuates the anti-estrogen effects of AB215. Remarkably, we find that AB215 is more effective than tamoxifen in suppressing tumor growth in a xenograft model. CONCLUSION: This study shows that IDs have profound role to inhibit estrogen signaling in ERα positive breast cancer cells, and that engineered TGF-beta ligands may have high therapeutic value.


Subject(s)
Activins/pharmacology , Bone Morphogenetic Protein 2/pharmacology , Breast Neoplasms/pathology , Inhibitor of Differentiation Proteins/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Signal Transduction/drug effects , Activins/therapeutic use , Bone Morphogenetic Protein 2/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Female , Humans , Inhibitor of Differentiation Proteins/genetics , MCF-7 Cells , Neoplasm Transplantation , Recombinant Fusion Proteins/therapeutic use , Tamoxifen/therapeutic use , Xenograft Model Antitumor Assays
12.
J Bone Miner Res ; 29(9): 1950-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24692083

ABSTRACT

Recombinant bone morphogenetic protein 2 (rhBMP2) has been used clinically to treat bone fractures in human patients. However, the high doses of rhBMP2 required for a therapeutic response can cause undesirable side effects. Here, we demonstrate that a novel Activin A/BMP2 (AB2) chimera, AB204, promotes osteogenesis and bone healing much more potently and effectively than rhBMP2. Remarkably, 1 month of AB204 treatment completely heals tibial and calvarial defects of critical size in mice at a concentration 10-fold lower than a dose of rhBMP2 that only partially heals the defect. We determine the structure of AB204 to 2.3 Å that reveals a distinct BMP2-like fold in which the Activin A sequence segments confer insensitivity to the BMP2 antagonist Noggin and an affinity for the Activin/BMP type II receptor ActRII that is 100-fold greater than that of BMP2. The structure also led to our identification of a single Activin A-derived amino acid residue, which, when mutated to the corresponding BMP2 residue, resulted in a significant increase in the affinity of AB204 for its type I receptor BMPRIa and a further enhancement in AB204's osteogenic potency. Together, these findings demonstrate that rationally designed AB2 chimeras can provide BMP2 substitutes with enhanced potency for treating non-union bone fractures.


Subject(s)
Activins/pharmacology , Bone Morphogenetic Protein 2/pharmacology , Recombinant Fusion Proteins/pharmacology , Transforming Growth Factor beta/pharmacology , Wound Healing/drug effects , Activin Receptors, Type I/metabolism , Activins/chemistry , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 2/chemistry , Calcification, Physiologic/drug effects , Cell Line , Humans , Mice , Osteogenesis/drug effects , Point Mutation/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Skull/drug effects , Skull/pathology , Tibia/drug effects , Tibia/pathology , Transforming Growth Factor beta/chemistry
13.
J Biol Chem ; 289(3): 1788-97, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24311780

ABSTRACT

Nodal, a member of the TGF-ß superfamily, plays an important role in vertebrate and invertebrate early development. The biochemical study of Nodal and its signaling pathway has been a challenge, mainly because of difficulties in producing the protein in sufficient quantities. We have developed a library of stable, chemically refoldable Nodal/BMP2 chimeric ligands (NB2 library). Three chimeras, named NB250, NB260, and NB264, show Nodal-like signaling properties including dependence on the co-receptor Cripto and activation of the Smad2 pathway. NB250, like Nodal, alters heart looping during the establishment of embryonic left-right asymmetry, and both NB250 and NB260, as well as Nodal, induce chondrogenic differentiation of human adipose-derived stem cells. This Nodal-induced differentiation is shown to be more efficient than BPM2-induced differentiation. Interestingly, the crystal structure of NB250 shows a backbone scaffold similar to that of BMP2. Our results show that these chimeric ligands may have therapeutic implications in cartilage injuries.


Subject(s)
Adipose Tissue/metabolism , Bone Morphogenetic Protein 2 , Chondrogenesis/drug effects , Nodal Protein , Recombinant Fusion Proteins , Signal Transduction/drug effects , Stem Cells/metabolism , Adipose Tissue/pathology , Adult , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/pharmacology , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Cell Line , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nodal Protein/chemistry , Nodal Protein/genetics , Nodal Protein/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Signal Transduction/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Stem Cells/pathology
14.
Stem Cell Res ; 10(3): 464-76, 2013 May.
Article in English | MEDLINE | ID: mdl-23500646

ABSTRACT

Human adipose derived stem cells (hASCs) can be easily isolated and their plasticity has been well characterized. Several TGF-ß superfamily ligands can direct hASCs towards chondrocytes. However, these ligands are difficult to purify and expensive. We have developed a library of Activin/BMP2 chimeric ligands (AB2 ligands) by systematically mixing their sequence segments and have tested their chondrogenic potential in hASCs. Cells cultured in monolayer or in a pellet culture system were incubated with a chemically defined medium supplemented with the chimeric ligands for 4 or 6 weeks and showed higher expression levels of type II collagen, aggrecan, and Sox9 mRNAs when compared with control and non-treated cells. Moreover, toluidine blue, alcian blue, and Masson's trichrome staining was markedly increased in treated cells, both in cell pellet and monolayer assays. In addition, immunohistochemical staining for detection of type I collagen, type II collagen, and Sox 9 demonstrated the acquisition of a chondrogenic phenotype in both culture systems. We present here an inexpensive and robust protocol for differentiation of hASCs towards chondrocytes in a reproducible and highly efficient manner. The AB2 ligands employed are easily produced and have properties that may become useful in cell therapy.


Subject(s)
Adipose Tissue/cytology , Chondrocytes/cytology , Mesenchymal Stem Cells/cytology , Activins/genetics , Activins/metabolism , Adult , Aggrecans/genetics , Aggrecans/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chondrocytes/metabolism , Chondrogenesis/drug effects , Collagen Type II/genetics , Collagen Type II/metabolism , Female , Humans , Ligands , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
15.
Cell ; 151(2): 304-19, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063122

ABSTRACT

Evolution of minimal DNA tumor virus' genomes has selected for small viral oncoproteins that hijack critical cellular protein interaction networks. The structural basis for the multiple and dominant functions of adenovirus oncoproteins has remained elusive. E4-ORF3 forms a nuclear polymer and simultaneously inactivates p53, PML, TRIM24, and MRE11/RAD50/NBS1 (MRN) tumor suppressors. We identify oligomerization mutants and solve the crystal structure of E4-ORF3. E4-ORF3 forms a dimer with a central ß core, and its structure is unrelated to known polymers or oncogenes. E4-ORF3 dimer units coassemble through reciprocal and nonreciprocal exchanges of their C-terminal tails. This results in linear and branched oligomer chains that further assemble in variable arrangements to form a polymer network that partitions the nuclear volume. E4-ORF3 assembly creates avidity-driven interactions with PML and an emergent MRN binding interface. This reveals an elegant structural solution whereby a small protein forms a multivalent matrix that traps disparate tumor suppressors.


Subject(s)
Adenovirus E4 Proteins/chemistry , Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/metabolism , Tumor Suppressor Proteins/metabolism , Adenovirus Infections, Human/virology , Cell Line , Cells, Cultured , Crystallography, X-Ray , Humans , Plant Cells/virology , Protein Folding , Nicotiana/virology
16.
Nat Methods ; 9(8): 834-9, 2012 May 20.
Article in English | MEDLINE | ID: mdl-22609626

ABSTRACT

Although nearly half of today's major pharmaceutical drugs target human integral membrane proteins (hIMPs), only 30 hIMP structures are currently available in the Protein Data Bank, largely owing to inefficiencies in protein production. Here we describe a strategy for the rapid structure determination of hIMPs, using solution NMR spectroscopy with systematically labeled proteins produced via cell-free expression. We report new backbone structures of six hIMPs, solved in only 18 months from 15 initial targets. Application of our protocols to an additional 135 hIMPs with molecular weight <30 kDa yielded 38 hIMPs suitable for structural characterization by solution NMR spectroscopy without additional optimization.


Subject(s)
Membrane Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Databases, Protein , Humans , Models, Molecular , Molecular Weight , Protein Conformation
17.
Protein Sci ; 20(6): 1030-41, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21465615

ABSTRACT

G-protein coupled receptors (GPCRs) constitute the largest family of intercellular signaling molecules and are estimated to be the target of more than 50% of all modern drugs. As with most integral membrane proteins (IMPs), a major bottleneck in the structural and biochemical analysis of GPCRs is their expression by conventional expression systems. Cell-free (CF) expression provides a relatively new and powerful tool for obtaining preparative amounts of IMPs. However, in the case of GPCRs, insufficient homogeneity of the targeted protein is a problem as the in vitro expression is mainly done with detergents, in which aggregation and solubilization difficulties, as well as problems with proper folding of hydrophilic domains, are common. Here, we report that using CF expression with the help of a fructose-based polymer, NV10 polymer (NVoy), we obtained preparative amounts of homogeneous GPCRs from the three GPCR families. We demonstrate that two GPCR B family members, corticotrophin-releasing factor receptors 1 and 2ß are not only solubilized in NVoy but also have functional ligand-binding characteristics with different agonists and antagonists in a detergent-free environment as well. Our findings open new possibilities for functional and structural studies of GPCRs and IMPs in general.


Subject(s)
Cell-Free System/metabolism , Fructans/metabolism , Gene Expression , Polymers/metabolism , Receptors, G-Protein-Coupled/genetics , Detergents , Fructans/chemistry , Humans , Ligands , Nuclear Magnetic Resonance, Biomolecular , Polymers/chemistry , Protein Binding , Receptors, G-Protein-Coupled/metabolism
18.
J Neurosci ; 31(10): 3565-79, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21389213

ABSTRACT

The precise knowledge of the subunit assembly process of NMDA receptors (NMDA-Rs) is essential to understand the receptor architecture and underlying mechanism of channel function. Because NMDA-Rs are obligatory heterotetramers requiring the GluN1 subunit, it is critical to investigate how GluN1 and GluN2 type subunits coassemble into tetramers. By combining approaches in cell biology, biochemistry, single particle electron microscopy, and x-ray crystallography, we report the mechanisms and phenotypes of mutant GluN1 subunits that are defective in receptor maturation. The T110A mutation in the N-terminal domain (NTD) of the GluN1 promotes heterodimerization between the NTDs of GluN1 and GluN2, whereas the Y109C mutation in the adjacent residue stabilizes the homodimer of the NTD of GluN1. The crystal structure of the NTD of GluN1 revealed the mechanism underlying the biochemical properties of these mutants. Effects of these mutations on the maturation of heteromeric NMDA-Rs were investigated using a receptor trafficking assay. Our results suggest that the NTDs of the GluN1 subunit initially form homodimers and the subsequent dimer dissociation is critical for forming heterotetrameric NMDA-Rs containing GluN2 subunits, defining a molecular determinant for receptor assembly. The domain arrangement of the dimeric NTD of GluN1 is unique among the ionotropic glutamate receptors and predicts that the structure and mechanism around the NTDs of NMDA-Rs are different from those of the homologous AMPA and kainate receptors.


Subject(s)
Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Cell Line , Cells, Cultured , Chromatography, Gel , Crystallography, X-Ray , Humans , Protein Conformation , Protein Subunits/chemistry , Receptors, N-Methyl-D-Aspartate/chemistry
19.
Proc Natl Acad Sci U S A ; 108(14): 5831-6, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21422294

ABSTRACT

G protein-gated inwardly rectifying potassium (GIRK) channels are important gatekeepers of neuronal excitability. The surface expression of neuronal GIRK channels is regulated by the psychostimulant-sensitive sorting nexin 27 (SNX27) protein through a class I (-X-Ser/Thr-X-Φ, where X is any residue and Φ is a hydrophobic amino acid) PDZ-binding interaction. The G protein-insensitive inward rectifier channel (IRK1) contains the same class I PDZ-binding motif but associates with a different synaptic PDZ protein, postsynaptic density protein 95 (PSD95). The mechanism by which SNX27 and PSD95 discriminate these channels was previously unclear. Using high-resolution structures coupled with biochemical and functional analyses, we identified key amino acids upstream of the channel's canonical PDZ-binding motif that associate electrostatically with a unique structural pocket in the SNX27-PDZ domain. Changing specific charged residues in the channel's carboxyl terminus or in the PDZ domain converts the selective association and functional regulation by SNX27. Elucidation of this unique interaction site between ion channels and PDZ-containing proteins could provide a therapeutic target for treating brain diseases.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Models, Molecular , Neurons/physiology , Sorting Nexins/metabolism , Amino Acid Motifs/physiology , Animals , COS Cells , Chlorocebus aethiops , Crystallization , Disks Large Homolog 4 Protein , Electrophysiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Hippocampus/cytology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microscopy, Confocal , Mutagenesis, Site-Directed , Neurons/metabolism , Protein Structure, Tertiary , Rats
20.
Biochemistry ; 49(42): 9089-95, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20849081

ABSTRACT

Mistic is a small Bacillus subtilis protein which is of current interest to the field of structural biology and biochemistry because of its unique ability to increase integral membrane protein yields in Escherichia coli expression. Using the osmosensing histidine kinase receptor, EnvZ, an E. coli two-component system, and its cytoplasmic cognate response regulator, OmpR, we provide the first evidence that a Mistic-fused integral membrane protein maintains functionality both in vitro and in vivo. When the purified and detergent-solubilized receptor EnvZ is fused to Mistic, it maintains the ability to autophosphorylate on residue His(243) and phosphotransfers to residue Asp(55) located on OmpR. Functionality was also observed in vivo by means of a ß-galactosidase assay in which RU1012 [Φ(ompC-lacZ)10-15, ΔenvZ::Km(r)] cells transformed with Mistic-fused EnvZ led to an increase in downstream signal transduction events detected by the activation of ompC gene expression. These findings illustrate that Mistic preserves the functionality of the Mistic-fused cargo protein and thus provides a beneficial alternate approach to study integral membrane proteins not only by improving expression levels but also for direct use in functional characterization.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Models, Biological , Models, Molecular , Multienzyme Complexes/genetics , Porins/chemistry , Porins/genetics , Porins/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...