Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biomolecules ; 14(5)2024 May 17.
Article in English | MEDLINE | ID: mdl-38786000

ABSTRACT

Cataract disease is strongly associated with progressively accumulating oxidative damage to the extremely long-lived crystallin proteins of the lens. Cysteine oxidation affects crystallin folding, interactions, and light-scattering aggregation especially strongly due to the formation of disulfide bridges. Minimizing crystallin aggregation is crucial for lifelong lens transparency, so one might expect the ubiquitous lens crystallin superfamilies (α and ßγ) to contain little cysteine. Yet, the Cys content of γ-crystallins is well above the average for human proteins. We review literature relevant to this longstanding puzzle and take advantage of expanding genomic databases and improved machine learning tools for protein structure prediction to investigate it further. We observe remarkably low Cys conservation in the ßγ-crystallin superfamily; however, in γ-crystallin, the spatial positioning of Cys residues is clearly fine-tuned by evolution. We propose that the requirements of long-term lens transparency and high lens optical power impose competing evolutionary pressures on lens ßγ-crystallins, leading to distinct adaptations: high Cys content in γ-crystallins but low in ßB-crystallins. Aquatic species need more powerful lenses than terrestrial ones, which explains the high methionine content of many fish γ- (and even ß-) crystallins. Finally, we discuss synergies between sulfur-containing and aromatic residues in crystallins and suggest future experimental directions.


Subject(s)
Cysteine , Lens, Crystalline , gamma-Crystallins , gamma-Crystallins/metabolism , gamma-Crystallins/chemistry , gamma-Crystallins/genetics , Cysteine/metabolism , Cysteine/chemistry , Humans , Lens, Crystalline/metabolism , Lens, Crystalline/chemistry , Animals , Cataract/metabolism
2.
Biophys J ; 122(16): 3238-3253, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37422697

ABSTRACT

Many secreted proteins, including viral proteins, contain multiple disulfide bonds. How disulfide formation is coupled to protein folding in the cell remains poorly understood at the molecular level. Here, we combine experiment and simulation to address this question as it pertains to the SARS-CoV-2 receptor binding domain (RBD). We show that the RBD can only refold reversibly if its native disulfides are present before folding. But in their absence, the RBD spontaneously misfolds into a nonnative, molten-globule-like state that is structurally incompatible with complete disulfide formation and that is highly prone to aggregation. Thus, the RBD native structure represents a metastable state on the protein's energy landscape with reduced disulfides, indicating that nonequilibrium mechanisms are needed to ensure native disulfides form before folding. Our atomistic simulations suggest that this may be achieved via co-translational folding during RBD secretion into the endoplasmic reticulum. Namely, at intermediate translation lengths, native disulfide pairs are predicted to come together with high probability, and thus, under suitable kinetic conditions, this process may lock the protein into its native state and circumvent highly aggregation-prone nonnative intermediates. This detailed molecular picture of the RBD folding landscape may shed light on SARS-CoV-2 pathology and molecular constraints governing SARS-CoV-2 evolution.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Disulfides/chemistry , Proteins/chemistry , Protein Folding
3.
Mol Cell ; 83(11): 1936-1952.e7, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37267908

ABSTRACT

Non-native conformations drive protein-misfolding diseases, complicate bioengineering efforts, and fuel molecular evolution. No current experimental technique is well suited for elucidating them and their phenotypic effects. Especially intractable are the transient conformations populated by intrinsically disordered proteins. We describe an approach to systematically discover, stabilize, and purify native and non-native conformations, generated in vitro or in vivo, and directly link conformations to molecular, organismal, or evolutionary phenotypes. This approach involves high-throughput disulfide scanning (HTDS) of the entire protein. To reveal which disulfides trap which chromatographically resolvable conformers, we devised a deep-sequencing method for double-Cys variant libraries of proteins that precisely and simultaneously locates both Cys residues within each polypeptide. HTDS of the abundant E. coli periplasmic chaperone HdeA revealed distinct classes of disordered hydrophobic conformers with variable cytotoxicity depending on where the backbone was cross-linked. HTDS can bridge conformational and phenotypic landscapes for many proteins that function in disulfide-permissive environments.


Subject(s)
Escherichia coli Proteins , Protein Folding , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Conformation , Disulfides/metabolism , High-Throughput Nucleotide Sequencing , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
4.
J Am Chem Soc ; 145(12): 6781-6797, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36918380

ABSTRACT

Cataracts are caused by high-molecular-weight aggregates of human eye lens proteins that scatter light, causing lens opacity. Metal ions have emerged as important potential players in the etiology of cataract disease, as human lens γ-crystallins are susceptible to metal-induced aggregation. Here, the interaction of Cu2+ ions with γD-, γC-, and γS-crystallins, the three most abundant γ-crystallins in the lens, has been evaluated. Cu2+ ions induced non-amyloid aggregation in all three proteins. Solution turbidimetry, sodium dodecyl sulfate poly(acrylamide) gel electrophoresis (SDS-PAGE), circular dichroism, and differential scanning calorimetry showed that the mechanism for Cu-induced aggregation involves: (i) loss of ß-sheet structure in the N-terminal domain; (ii) decreased thermal and kinetic stability; (iii) formation of metal-bridged species; and (iv) formation of disulfide-bridged dimers. Isothermal titration calorimetry (ITC) revealed distinct Cu2+ binding affinities in the γ-crystallins. Electron paramagnetic resonance (EPR) revealed two distinct Cu2+ binding sites in each protein. Spin quantitation demonstrated the reduction of γ-crystallin-bound Cu2+ ions to Cu+ under aerobic conditions, while X-ray absorption spectroscopy (XAS) confirmed the presence of linear or trigonal Cu+ binding sites in γ-crystallins. Our EPR and XAS studies revealed that γ-crystallins' Cu2+ reductase activity yields a protein-based free radical that is likely a Tyr-based species in human γD-crystallin. This unique free radical chemistry carried out by distinct redox-active Cu sites in human lens γ-crystallins likely contributes to the mechanism of copper-induced aggregation. In the context of an aging human lens, γ-crystallins could act not only as structural proteins but also as key players for metal and redox homeostasis.


Subject(s)
Cataract , Crystallins , gamma-Crystallins , Humans , gamma-Crystallins/chemistry , Copper/chemistry , Ions , Oxidoreductases
5.
ArXiv ; 2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36776823

ABSTRACT

Non-native conformations drive protein misfolding diseases, complicate bioengineering efforts, and fuel molecular evolution. No current experimental technique is well-suited for elucidating them and their phenotypic effects. Especially intractable are the transient conformations populated by intrinsically disordered proteins. We describe an approach to systematically discover, stabilize, and purify native and non-native conformations, generated in vitro or in vivo, and directly link conformations to molecular, organismal, or evolutionary phenotypes. This approach involves high-throughput disulfide scanning (HTDS) of the entire protein. To reveal which disulfides trap which chromatographically resolvable conformers, we devised a deep-sequencing method for double-Cys variant libraries of proteins that precisely and simultaneously locates both Cys residues within each polypeptide. HTDS of the abundant E. coli periplasmic chaperone HdeA revealed distinct classes of disordered hydrophobic conformers with variable cytotoxicity depending on where the backbone was cross-linked. HTDS can bridge conformational and phenotypic landscapes for many proteins that function in disulfide-permissive environments.

6.
bioRxiv ; 2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36380756

ABSTRACT

Many secreted proteins contain multiple disulfide bonds. How disulfide formation is coupled to protein folding in the cell remains poorly understood at the molecular level. Here, we combine experiment and simulation to address this question as it pertains to the SARS-CoV-2 receptor binding domain (RBD). We show that, whereas RBD can refold reversibly when its disulfides are intact, their disruption causes misfolding into a nonnative molten-globule state that is highly prone to aggregation and disulfide scrambling. Thus, non-equilibrium mechanisms are needed to ensure disulfides form prior to folding in vivo. Our simulations suggest that co-translational folding may accomplish this, as native disulfide pairs are predicted to form with high probability at intermediate lengths, ultimately committing the RBD to its metastable native state and circumventing nonnative intermediates. This detailed molecular picture of the RBD folding landscape may shed light on SARS-CoV-2 pathology and molecular constraints governing SARS-CoV-2 evolution.

7.
Elife ; 112022 06 20.
Article in English | MEDLINE | ID: mdl-35723573

ABSTRACT

Cataract is one of the most prevalent protein aggregation disorders and still the most common cause of vision loss worldwide. The metabolically quiescent core region of the human lens lacks cellular or protein turnover; it has therefore evolved remarkable mechanisms to resist light-scattering protein aggregation for a lifetime. We now report that one such mechanism involves an unusually abundant lens metabolite, myo-inositol, suppressing aggregation of lens crystallins. We quantified aggregation suppression using our previously well-characterized in vitro aggregation assays of oxidation-mimicking human γD-crystallin variants and investigated myo-inositol's molecular mechanism of action using solution NMR, negative-stain TEM, differential scanning fluorometry, thermal scanning Raman spectroscopy, turbidimetry in redox buffers, and free thiol quantitation. Unlike many known chemical chaperones, myo-inositol's primary target was not the native, unfolded, or final aggregated states of the protein; rather, we propose that it was the rate-limiting bimolecular step on the aggregation pathway. Given recent metabolomic evidence that it is severely depleted in human cataractous lenses compared to age-matched controls, we suggest that maintaining or restoring healthy levels of myo-inositol in the lens may be a simple, safe, and globally accessible strategy to prevent or delay lens opacification due to age-onset cataract.


Subject(s)
Cataract , Lens, Crystalline , Cataract/metabolism , Humans , Inositol/analysis , Inositol/metabolism , Lens, Crystalline/metabolism , Molecular Chaperones/metabolism , Protein Aggregates
8.
Exp Eye Res ; 211: 108707, 2021 10.
Article in English | MEDLINE | ID: mdl-34332989

ABSTRACT

The nuclear region of the lens is metabolically quiescent, but it is far from inert chemically. Without cellular renewal and with decades of environmental exposures, the lens proteome, lipidome, and metabolome change. The lens crystallins have evolved exquisite mechanisms for resisting, slowing, adapting to, and perhaps even harnessing the effects of these cumulative chemical modifications to minimize the amount of light-scattering aggregation in the lens over a lifetime. Redox chemistry is a major factor in these damages and mitigating adaptations, and as such, it is likely to be a key component of any successful therapeutic strategy for preserving or rescuing lens transparency, and perhaps flexibility, during aging. Protein redox chemistry is typically mediated by Cys residues. This review will therefore focus primarily on the Cys-rich γ-crystallins of the human lens, taking care to extend these findings to the ß- and α-crystallins where pertinent.


Subject(s)
Cysteine/metabolism , Lens, Crystalline/metabolism , gamma-Crystallins/metabolism , Aging/physiology , Disulfides/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Humans , Oxidation-Reduction , Sulfhydryl Compounds/metabolism
9.
Biophys J ; 117(2): 269-280, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31266635

ABSTRACT

The γ-crystallins of the eye lens nucleus are among the longest-lived proteins in the human body. Synthesized in utero, they must remain folded and soluble throughout adulthood to maintain lens transparency and avoid cataracts. γD- and γS-crystallin are two major monomeric crystallins of the human lens. γD-crystallin is concentrated in the oldest lens fiber cells, the lens nucleus, whereas γS-crystallin is concentrated in the younger cells of the lens cortex. The kinetic stability parameters of these two-domain proteins and their isolated domains were determined and compared. Kinetic unfolding experiments monitored by fluorescence spectroscopy in varying concentrations of guanidinium chloride were used to extrapolate unfolding rate constants and half-lives of the crystallins in the absence of the denaturant. Consistent with their long lifespans in the lens, extrapolated half-lives for the initial unfolding step were on the timescale of years. Both proteins' isolated N-terminal domains were less kinetically stable than their respective C-terminal domains at denaturant concentrations predicted to disrupt the domain interface, but at low denaturant concentrations, the relative kinetic stabilities were reversed. Cataract-associated aggregation has been shown to proceed from partially unfolded intermediates in these proteins; their extreme kinetic stability likely evolved to protect the lens from the initiation of aggregation reactions. Our findings indicate that the domain interface is the source of significant kinetic stability. The gene duplication and fusion event that produced the modern two-domain architecture of vertebrate lens crystallins may be the origin of their high kinetic as well as thermodynamic stability.


Subject(s)
Lens, Crystalline/metabolism , gamma-Crystallins/chemistry , gamma-Crystallins/metabolism , Humans , Kinetics , Models, Molecular , Protein Domains , Protein Folding , Protein Stability , Temperature , Time Factors
10.
J Biol Chem ; 293(46): 17997-18009, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30242128

ABSTRACT

Increased light scattering in the eye lens due to aggregation of the long-lived lens proteins, crystallins, is the cause of cataract disease. Several mutations in the gene encoding human γD-crystallin (HγD) cause misfolding and aggregation. Cataract-associated substitutions at Trp42 cause the protein to aggregate in vitro from a partially unfolded intermediate locked by an internal disulfide bridge, and proteomic evidence suggests a similar aggregation precursor is involved in age-onset cataract. Surprisingly, WT HγD can promote aggregation of the W42Q variant while itself remaining soluble. Here, a search for a biochemical mechanism for this interaction has revealed a previously unknown oxidoreductase activity in HγD. Using in vitro oxidation, mutational analysis, cysteine labeling, and MS, we have assigned this activity to a redox-active internal disulfide bond that is dynamically exchanged among HγD molecules. The W42Q variant acts as a disulfide sink, reducing oxidized WT and forming a distinct internal disulfide that kinetically traps the aggregation-prone intermediate. Our findings suggest a redox "hot potato" competition among WT and mutant or modified polypeptides wherein variants with the lowest kinetic stability are trapped in aggregation-prone intermediate states upon accepting disulfides from more stable variants. Such reactions may occur in other long-lived proteins that function in oxidizing environments. In these cases, aggregation may be forestalled by inhibiting disulfide flow toward mutant or damaged polypeptides.


Subject(s)
Disulfides/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , gamma-Crystallins/metabolism , Amino Acid Substitution , Cysteine/chemistry , Disulfides/chemistry , Escherichia coli , Humans , Mutagenesis, Site-Directed , Mutation , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Oxidoreductases Acting on Sulfur Group Donors/genetics , Protein Domains , Protein Multimerization , Protein Unfolding , Proteomics , gamma-Crystallins/chemistry , gamma-Crystallins/genetics
11.
Biochemistry ; 57(33): 4959-4962, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30064223

ABSTRACT

Cataract disease results from non-amyloid aggregation of eye lens proteins and is the leading cause of blindness in the world. Zinc concentrations in cataractous lenses are increased significantly relative to those in healthy lenses. It was recently reported that Zn(II) ions induce the aggregation of one of the more abundant proteins in the core of the lens, human γD-crystallin. Here, the mechanism of Zn-induced aggregation has been revealed through a comparative study of three homologous human lens γ-crystallins and a combination of spectroscopic, electron microscopy, and site-directed mutagenesis studies. This work reveals that a single His residue acts as a "switch" for the Zn-induced non-amyloid aggregation of human γ-crystallins. Aggregation can be reversed by a chelating agent, revealing a metal-bridging mechanism. This study sheds light on an aberrant Zn-crystallin interaction that promotes aggregation, a process that is relevant to cataract disease.


Subject(s)
Histidine/chemistry , Zinc/chemistry , gamma-Crystallins/metabolism , Cataract/etiology , Edetic Acid/chemistry , Humans , Mutation , Protein Binding , Protein Multimerization , gamma-Crystallins/chemistry , gamma-Crystallins/genetics
12.
FEBS Lett ; 590(23): 4308-4317, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27800613

ABSTRACT

Cooperativity in ligand binding is a key emergent property of protein oligomers. Positive cooperativity (higher affinity for subsequent binding events than for initial binding) is frequent. However, the symmetrically homodimeric ligand-binding domain (LBD) of metabotropic glutamate receptor type 1 exhibits negative cooperativity. To investigate its origin and functional significance, we measured the response to glutamate in vitro of wild-type and C140S LBD as a function of the extent of dimerization. Our results indicate that homodimerization enhances the affinity of the first, but not the second, binding site, relative to the monomer, giving the dimeric receptor both greater sensitivity and a broader dynamic range.


Subject(s)
Protein Multimerization , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/metabolism , Animals , Glutamic Acid/pharmacology , Ligands , Mice , Models, Molecular , Mutation , Protein Multimerization/drug effects , Protein Structure, Quaternary/drug effects , Receptors, Metabotropic Glutamate/genetics
13.
J Biol Chem ; 291(36): 19172-83, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27417136

ABSTRACT

Considerable mechanistic insight has been gained into amyloid aggregation; however, a large number of non-amyloid protein aggregates are considered "amorphous," and in most cases, little is known about their mechanisms. Amorphous aggregation of γ-crystallins in the eye lens causes cataract, a widespread disease of aging. We combined simulations and experiments to study the mechanism of aggregation of two γD-crystallin mutants, W42R and W42Q: the former a congenital cataract mutation, and the latter a mimic of age-related oxidative damage. We found that formation of an internal disulfide was necessary and sufficient for aggregation under physiological conditions. Two-chain all-atom simulations predicted that one non-native disulfide in particular, between Cys(32) and Cys(41), was likely to stabilize an unfolding intermediate prone to intermolecular interactions. Mass spectrometry and mutagenesis experiments confirmed the presence of this bond in the aggregates and its necessity for oxidative aggregation under physiological conditions in vitro Mining the simulation data linked formation of this disulfide to extrusion of the N-terminal ß-hairpin and rearrangement of the native ß-sheet topology. Specific binding between the extruded hairpin and a distal ß-sheet, in an intermolecular chain reaction similar to domain swapping, is the most probable mechanism of aggregate propagation.


Subject(s)
Cataract , Disulfides/chemistry , Mutation, Missense , Protein Aggregates , Protein Folding , gamma-Crystallins/chemistry , Amino Acid Substitution , Cysteine , Disulfides/metabolism , Humans , Protein Domains , Protein Structure, Secondary , gamma-Crystallins/genetics , gamma-Crystallins/metabolism
14.
Protein Sci ; 25(6): 1115-28, 2016 06.
Article in English | MEDLINE | ID: mdl-26991007

ABSTRACT

Numerous mutations and covalent modifications of the highly abundant, long-lived crystallins of the eye lens cause their aggregation leading to progressive opacification of the lens, cataract. The nature and biochemical mechanisms of the aggregation process are poorly understood, as neither amyloid nor native-state polymers are commonly found in opaque lenses. The ßγ-crystallin fold contains four highly conserved buried tryptophans, which can be oxidized to more hydrophilic products, such as kynurenine, upon UV-B irradiation. We mimicked this class of oxidative damage using Trp→Glu point mutants of human γD-crystallin. Such substitutions may represent a model of UV-induced photodamage-introduction of a charged group into the hydrophobic core generating "denaturation from within." The effects of Trp→Glu substitutions were highly position dependent. While each was destabilizing, only the two located in the bottom of the double Greek key fold-W42E and W130E-yielded robust aggregation of partially unfolded intermediates at 37°C and pH 7. The αB-crystallin chaperone suppressed aggregation of W130E, but not W42E, indicating distinct aggregation pathways from damage in the N-terminal vs C-terminal domain. The W130E aggregates had loosely fibrillar morphology, yet were nonamyloid, noncovalent, showed little surface hydrophobicity, and formed at least 20°C below the melting temperature of the native ß-sheets. These features are most consistent with domain-swapped polymerization. Aggregation of partially destabilized crystallins under physiological conditions, as occurs in this class of point mutants, could provide a simple in vitro model system for drug discovery and optimization.


Subject(s)
Cataract , Models, Biological , Point Mutation , Protein Aggregates , Protein Folding/radiation effects , Ultraviolet Rays , gamma-Crystallins , Amino Acid Substitution , Cataract/genetics , Cataract/metabolism , Humans , Hydrogen-Ion Concentration , Protein Aggregates/genetics , Protein Aggregates/radiation effects , Protein Structure, Secondary , gamma-Crystallins/chemistry , gamma-Crystallins/genetics , gamma-Crystallins/metabolism
15.
ACS Chem Biol ; 11(1): 263-72, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26579725

ABSTRACT

Cataract is the leading cause of blindness in the world. It results from aggregation of eye lens proteins into high-molecular-weight complexes, causing light scattering and lens opacity. Copper and zinc concentrations in cataractous lens are increased significantly relative to a healthy lens, and a variety of experimental and epidemiological studies implicate metals as potential etiological agents for cataract. The natively monomeric, ß-sheet rich human γD (HγD) crystallin is one of the more abundant proteins in the core of the lens. It is also one of the most thermodynamically stable proteins in the human body. Surprisingly, we found that both Cu(II) and Zn(II) ions induced rapid, nonamyloid aggregation of HγD, forming high-molecular-weight light-scattering aggregates. Unlike Zn(II), Cu(II) also substantially decreased the thermal stability of HγD and promoted the formation of disulfide-bridged dimers, suggesting distinct aggregation mechanisms. In both cases, however, metal-induced aggregation depended strongly on temperature and was suppressed by the human lens chaperone αB-crystallin (HαB), implicating partially folded intermediates in the aggregation process. Consistently, distinct site-specific interactions of Cu(II) and Zn(II) ions with the protein and conformational changes in specific hinge regions were identified by nuclear magnetic resonance. This study provides insights into the mechanisms of metal-induced aggregation of one of the more stable proteins in the human body, and it reveals a novel and unexplored bioinorganic facet of cataract disease.


Subject(s)
Copper/pharmacology , Crystallins/metabolism , Protein Aggregation, Pathological/chemically induced , Zinc/pharmacology , Copper/chemistry , Electrophoresis, Polyacrylamide Gel , Humans , Ions , Magnetic Resonance Spectroscopy , Microscopy, Electron, Transmission , Models, Molecular , Protein Folding , Protein Stability , Temperature , Zinc/chemistry
16.
J Biol Chem ; 290(18): 11491-503, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25787081

ABSTRACT

Non-native protein conformers generated by mutation or chemical damage template aggregation of wild-type, undamaged polypeptides in diseases ranging from amyotrophic lateral sclerosis to cancer. We tested for such interactions in the natively monomeric human eye lens protein γd-crystallin, whose aggregation leads to cataract disease. The oxidation-mimicking W42Q mutant of γd-crystallin formed non-native polymers starting from a native-like state under physiological conditions. Aggregation occurred in the temperature range 35-45 °C, in which the mutant protein began to lose the native conformation of its N-terminal domain. Surprisingly, wild-type γd-crystallin promoted W42Q polymerization in a catalytic manner, even at mutant concentrations too low for homogeneous nucleation to occur. The presence of wild-type protein also downshifted the temperature range of W42Q aggregation. W42Q aggregation required formation of a non-native intramolecular disulfide bond but not intermolecular cross-linking. Transient WT/W42Q binding may catalyze this oxidative misfolding event in the mutant. That a more stable variant in a mixture can specifically promote aggregation of a less stable one rationalizes how extensive aggregation of rare damaged polypeptides can occur during the course of aging.


Subject(s)
Mutation , Protein Folding , Protein Multimerization , gamma-Crystallins/chemistry , gamma-Crystallins/genetics , Amino Acid Sequence , Biocatalysis , Cold Temperature , Disulfides/chemistry , Humans , Kinetics , Models, Molecular , Oxidation-Reduction , Protein Stability , Protein Structure, Quaternary , Protein Structure, Tertiary , gamma-Crystallins/metabolism
17.
Prog Biophys Mol Biol ; 115(1): 32-41, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24835736

ABSTRACT

The ßγ-crystallins are among the most stable and long-lived proteins in the human body. With increasing age, however, they transform to high molecular weight light-scattering aggregates, resulting in cataracts. This occurs despite the presence in the lens of high concentrations of the a-crystallin chaperones. Aggregation of crystallins can be induced in vitro by a variety of stresses, including acidic pH, ultraviolet light, oxidative damage, heating or freezing, and specific amino acid substitutions. Accumulating evidence points to the existence of specific biochemical pathways of protein: protein interaction and polymerization. We review the methods used for studying crystallin stability and aggregation and discuss the sometimes counterintuitive relationships between factors that favor native state stability and those that favor non-native aggregation. We discuss the behavior of ßγ-crystallins in mixtures and their chaperone ability; the consequences of missense mutations and covalent damage to the side-chains; and the evolutionary strategies that have shaped these proteins. Efforts are ongoing to reveal the nature of cataractous crystallin aggregates and understand the mechanisms of aggregation in the context of key models of protein polymerization: amyloid, native-state, and domain-swapped. Such mechanistic understanding is likely to be of value for the development of therapeutic interventions and draw attention to unanswered questions about the relationship between a protein's native state stability and its transformation to an aggregated state.


Subject(s)
Protein Aggregates , beta-Crystallins/chemistry , gamma-Crystallins/chemistry , Animals , Cataract/metabolism , Humans , Molecular Chaperones/metabolism , Protein Stability , beta-Crystallins/metabolism , gamma-Crystallins/metabolism
18.
ACS Chem Biol ; 8(3): 617-25, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23237450

ABSTRACT

GPCRs mediate intracellular signaling upon external stimuli, making them ideal drug targets. However, little is known about their activation mechanisms due to the difficulty in purification. Here, we introduce a method to purify GPCRs in nanodiscs, which incorporates GPCRs into lipid bilayers immediately after membrane solubilization, followed by single-step purification. Using this approach, we purified a family B GPCR, parathyroid hormone 1 receptor (PTH1R), which regulates calcium and phosphate homeostasis and is a drug target for osteoporosis. We demonstrated that the purified PTH1R in nanodiscs can bind to PTH(1-34) and activate G protein. We also observed that Ca(2+) is a weak agonist of PTH1R, and Ca(2+) in millimolar concentration can switch PTH(1-34) from an inverse agonist to an agonist. Hence, our results show that nanodiscs are a viable vehicle for GPCR purification, enabling studies of GPCRs under precise experimental conditions without interference from other cellular or membrane components.


Subject(s)
Calcium/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Nanostructures/chemistry , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction , Binding Sites , Calcium/pharmacology , HEK293 Cells , Humans , Ligands , Models, Molecular , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/isolation & purification , Structure-Activity Relationship
19.
Biochim Biophys Acta ; 1818(2): 225-33, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21851807

ABSTRACT

Functional reconstitution of transmembrane proteins remains a significant barrier to their biochemical, biophysical, and structural characterization. Studies of seven-transmembrane G-protein coupled receptors (GPCRs) in vitro are particularly challenging because, ideally, they require access to the receptor on both sides of the membrane as well as within the plane of the membrane. However, understanding the structure and function of these receptors at the molecular level within a native-like environment will have a large impact both on basic knowledge of cell signaling and on pharmacological research. The goal of this article is to review the main classes of membrane mimics that have been, or could be, used for functional reconstitution of GPCRs. These include the use of micelles, bicelles, lipid vesicles, nanodiscs, lipidic cubic phases, and planar lipid membranes. Each of these approaches is evaluated with respect to its fundamental advantages and limitations and its applications in the field of GPCR research. This article is part of a Special Issue entitled: Membrane protein structure and function.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/chemistry , Receptors, G-Protein-Coupled/chemistry , Animals , Cell Membrane/chemistry , Cell Membrane/genetics , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Protein Conformation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...