Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Hum Mol Genet ; 31(20): 3439-3457, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-35642742

RESUMEN

The correct expression of folded, functional rhodopsin (Rho) is critical for visual perception. However, this seven-transmembrane helical G protein-coupled receptor is prone to mutations with pathological consequences of retinal degeneration in retinitis pigmentosa (RP) due to Rho misfolding. Pharmacological chaperones that stabilize the inherited Rho variants by assisting their folding and membrane targeting could slow the progression of RP. In this study, we employed virtual screening of synthetic compounds with a natural product scaffold in conjunction with in vitro and in vivo evaluations to discover a novel chromenone-containing small molecule with favorable pharmacological properties that stabilize rod opsin. This compound reversibly binds to unliganded bovine rod opsin with an EC50 value comparable to the 9-cis-retinal chromophore analog and partially rescued membrane trafficking of multiple RP-related rod opsin variants in vitro. Importantly, this novel ligand of rod opsin was effective in vivo in murine models, protecting photoreceptors from deterioration caused by either bright light or genetic insult. Together, our current study suggests potential broad therapeutic implications of the new chromenone-containing non-retinoid small molecule against retinal diseases associated with photoreceptor degeneration.


Asunto(s)
Productos Biológicos , Degeneración Retiniana , Retinitis Pigmentosa , Animales , Productos Biológicos/uso terapéutico , Bovinos , Ligandos , Ratones , Receptores Acoplados a Proteínas G , Degeneración Retiniana/tratamiento farmacológico , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Retinitis Pigmentosa/tratamiento farmacológico , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Opsinas de Bastones/genética
2.
J Biol Chem ; 298(8): 102266, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35850308

RESUMEN

Over 100 mutations in the rhodopsin gene have been linked to a spectrum of retinopathies that include retinitis pigmentosa and congenital stationary night blindness. Though most of these variants exhibit a loss of function, the molecular defects caused by these underlying mutations vary considerably. In this work, we utilize deep mutational scanning to quantitatively compare the plasma membrane expression of 123 known pathogenic rhodopsin variants in the presence and absence of the stabilizing cofactor 9-cis-retinal. We identify 69 retinopathy variants, including 20 previously uncharacterized variants, that exhibit diminished plasma membrane expression in HEK293T cells. Of these apparent class II variants, 67 exhibit a measurable increase in expression in the presence of 9-cis-retinal. However, the magnitude of the response to this molecule varies considerably across this spectrum of mutations. Evaluation of the observed shifts relative to thermodynamic estimates for the coupling between binding and folding suggests underlying differences in stability constrains the magnitude of their response to retinal. Nevertheless, estimates from computational modeling suggest that many of the least sensitive variants also directly compromise binding. Finally, we evaluate the functional properties of three previous uncharacterized, retinal-sensitive variants (ΔN73, S131P, and R135G) and show that two of these retain residual function in vitro. Together, our results provide a comprehensive experimental characterization of the proteostatic properties of retinopathy variants and their response to retinal.


Asunto(s)
Enfermedades Hereditarias del Ojo , Rodopsina , Diterpenos/farmacología , Resistencia a Medicamentos/genética , Enfermedades Hereditarias del Ojo/genética , Células HEK293 , Humanos , Mutación , Retinaldehído/farmacología , Rodopsina/efectos de los fármacos , Rodopsina/genética , Rodopsina/metabolismo
3.
Nucleic Acids Res ; 49(22): 12943-12954, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34871407

RESUMEN

Programmed ribosomal frameshifting (PRF) is a translational recoding mechanism that enables the synthesis of multiple polypeptides from a single transcript. During translation of the alphavirus structural polyprotein, the efficiency of -1PRF is coordinated by a 'slippery' sequence in the transcript, an adjacent RNA stem-loop, and a conformational transition in the nascent polypeptide chain. To characterize each of these effectors, we measured the effects of 4530 mutations on -1PRF by deep mutational scanning. While most mutations within the slip-site and stem-loop reduce the efficiency of -1PRF, the effects of mutations upstream of the slip-site are far more variable. We identify several regions where modifications of the amino acid sequence of the nascent polypeptide impact the efficiency of -1PRF. Molecular dynamics simulations of polyprotein biogenesis suggest the effects of these mutations primarily arise from their impacts on the mechanical forces that are generated by the translocon-mediated cotranslational folding of the nascent polypeptide chain. Finally, we provide evidence suggesting that the coupling between cotranslational folding and -1PRF depends on the translation kinetics upstream of the slip-site. These findings demonstrate how -1PRF is coordinated by features within both the transcript and nascent chain.


Asunto(s)
Sistema de Lectura Ribosómico/genética , Simulación de Dinámica Molecular , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , Ribosomas/genética , Alphavirus/genética , Alphavirus/metabolismo , Células HEK293 , Humanos , Cinética , Mutación , Conformación de Ácido Nucleico , Poliproteínas/genética , Poliproteínas/metabolismo , ARN Mensajero/química , ARN Mensajero/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , ARN Viral/química , ARN Viral/genética , ARN Viral/metabolismo , Ribosomas/metabolismo
4.
Biophys J ; 121(14): 2712-2720, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35715957

RESUMEN

Missense mutations that compromise the plasma membrane expression (PME) of integral membrane proteins are the root cause of numerous genetic diseases. Differentiation of this class of mutations from those that specifically modify the activity of the folded protein has proven useful for the development and targeting of precision therapeutics. Nevertheless, it remains challenging to predict the effects of mutations on the stability and/ or expression of membrane proteins. In this work, we utilize deep mutational scanning data to train a series of artificial neural networks to predict the PME of transmembrane domain variants of G protein-coupled receptors from structural and/ or evolutionary features. We show that our best-performing network, which we term the PME predictor, can recapitulate mutagenic trends within rhodopsin and can differentiate pathogenic transmembrane domain variants that cause it to misfold from those that compromise its signaling. This network also generates statistically significant predictions for the relative PME of transmembrane domain variants for another class A G protein-coupled receptor (ß2 adrenergic receptor) but not for an unrelated voltage-gated potassium channel (KCNQ1). Notably, our analyses of these networks suggest structural features alone are generally sufficient to recapitulate the observed mutagenic trends. Moreover, our findings imply that networks trained in this manner may be generalizable to proteins that share a common fold. Implications of our findings for the design of mechanistically specific genetic predictors are discussed.


Asunto(s)
Canal de Potasio KCNQ1 , Canales de Potasio con Entrada de Voltaje , Canal de Potasio KCNQ1/metabolismo , Mutagénesis , Mutación , Canales de Potasio con Entrada de Voltaje/metabolismo , Rodopsina/química
5.
J Biol Chem ; 297(6): 101359, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34756884

RESUMEN

Membrane protein variants with diminished conformational stability often exhibit enhanced cellular expression at reduced growth temperatures. The expression of "temperature-sensitive" variants is also typically sensitive to corrector molecules that bind and stabilize the native conformation. There are many examples of temperature-sensitive rhodopsin variants, the misfolding of which is associated with the molecular basis of retinitis pigmentosa. In this work, we employ deep mutational scanning to compare the effects of reduced growth temperature and 9-cis-retinal, an investigational corrector, on the plasma membrane expression of 700 rhodopsin variants in HEK293T cells. We find that the change in expression at reduced growth temperatures correlates with the response to 9-cis-retinal among variants bearing mutations within a hydrophobic transmembrane domain (TM2). The most sensitive variants appear to disrupt a native helical kink within this transmembrane domain. By comparison, mutants that alter the structure of a polar transmembrane domain (TM7) exhibit weaker responses to temperature and retinal that are poorly correlated. Statistical analyses suggest that this observed insensitivity cannot be attributed to a single variable, but likely arises from the composite effects of mutations on the energetics of membrane integration, the stability of the native conformation, and the integrity of the retinal-binding pocket. Finally, we show that the characteristics of purified temperature- and retinal-sensitive variants suggest that the proteostatic effects of retinal may be manifested during translation and cotranslational folding. Together, our findings highlight several biophysical constraints that appear to influence the sensitivity of genetic variants to temperature and small-molecule correctors.


Asunto(s)
Mutación , Retinaldehído/metabolismo , Rodopsina/metabolismo , Células HEK293 , Humanos , Rodopsina/genética , Temperatura
6.
J Biol Chem ; 296: 100719, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33933451

RESUMEN

Peripheral myelin protein 22 (PMP22) folds and trafficks inefficiently, with only 20% of newly expressed protein trafficking to the cell surface. This behavior is exacerbated in many of the mutants associated with Charcot-Marie-Tooth disease, motivating further study. Here we characterized the role of N-glycosylation in limiting PMP22 trafficking. We first eliminated N-glycosylation using an N41Q mutation, which resulted in an almost 3-fold increase in trafficking efficiency of wildtype (WT) PMP22 and a 10-fold increase for the severely unstable L16P disease mutant in HEK293 cells, with similar results in Schwann cells. Total cellular levels were also much higher for the WT/N41Q mutant, although not for the L16P/N41Q form. Depletion of oligosaccharyltransferase OST-A and OST-B subunits revealed that WT PMP22 is N-glycosylated posttranslationally by OST-B, whereas L16P is cotranslationally glycosylated by OST-A. Quantitative proteomic screens revealed similarities and differences in the interactome for WT, glycosylation-deficient, and unstable mutant forms of PMP22 and also suggested that L16P is sequestered at earlier stages of endoplasmic reticulum quality control. CRISPR knockout studies revealed a role for retention in endoplasmic reticulum sorting receptor 1 (RER1) in limiting the trafficking of all three forms, for UDP-glucose glycoprotein glucosyltransferase 1 (UGGT1) in limiting the trafficking of WT and L16P but not N41Q, and calnexin (CNX) in limiting the trafficking of WT and N41Q but not L16P. This work shows that N-glycosylation is a limiting factor to forward trafficking PMP22 and sheds light on the proteins involved in its quality control.


Asunto(s)
Proteínas de la Mielina/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Glicosilación , Células HEK293 , Humanos , Modelos Moleculares , Mutación , Proteínas de la Mielina/química , Proteínas de la Mielina/genética , Conformación Proteica , Transporte de Proteínas
7.
J Biol Chem ; 295(33): 11418-11419, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32817126

RESUMEN

Proteins must acquire and maintain a specific fold to execute their biochemical function(s). In solution, unfolded proteins typically find this native structure through a biased sampling of preferred intermediate conformations. However, the initial search for these structures begins during protein synthesis, and it is unclear how much interactions between the ribosome and nascent polypeptide skew folding pathways. In this issue, Jensen and colleagues use a ribosomal force-profiling assay to show that RNase H forms a similar folding intermediate on and off the ribosome. In conjunction with measurements of the rate of RNase H unfolding on and off the ribosome, their results show that ribosomal interactions have little impact on the folding pathway of RNase H. These findings suggest that the ribosome itself does not necessarily rewire protein folding reactions.


Asunto(s)
Ribonucleasa H , Ribosomas , Biosíntesis de Proteínas , Pliegue de Proteína , Proteínas/metabolismo , Ribonucleasa H/genética , Ribonucleasa H/metabolismo , Ribosomas/metabolismo
8.
J Biol Chem ; 295(20): 6798-6808, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32169904

RESUMEN

Viruses maximize their genetic coding capacity through a variety of biochemical mechanisms, including programmed ribosomal frameshifting (PRF), which facilitates the production of multiple proteins from a single mRNA transcript. PRF is typically stimulated by structural elements within the mRNA that generate mechanical tension between the transcript and ribosome. However, in this work, we show that the forces generated by the cotranslational folding of the nascent polypeptide chain can also enhance PRF. Using an array of biochemical, cellular, and computational techniques, we first demonstrate that the Sindbis virus structural polyprotein forms two competing topological isomers during its biosynthesis at the ribosome-translocon complex. We then show that the formation of one of these topological isomers is linked to PRF. Coarse-grained molecular dynamics simulations reveal that the translocon-mediated membrane integration of a transmembrane domain upstream from the ribosomal slip site generates a force on the nascent polypeptide chain that scales with observed frameshifting. Together, our results indicate that cotranslational folding of this viral protein generates a tension that stimulates PRF. To our knowledge, this constitutes the first example in which the conformational state of the nascent polypeptide chain has been linked to PRF. These findings raise the possibility that, in addition to RNA-mediated translational recoding, a variety of cotranslational folding or binding events may also stimulate PRF.


Asunto(s)
Alphavirus/clasificación , Sistema de Lectura Ribosómico , Poliproteínas/biosíntesis , Biosíntesis de Proteínas , Pliegue de Proteína , Virus Sindbis/metabolismo , Proteínas Virales/biosíntesis , Alphavirus/química , Células HEK293 , Humanos , Virus Sindbis/genética
9.
Chem Rev ; 119(9): 5537-5606, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-30608666

RESUMEN

Advances over the past 25 years have revealed much about how the structural properties of membranes and associated proteins are linked to the thermodynamics and kinetics of membrane protein (MP) folding. At the same time biochemical progress has outlined how cellular proteostasis networks mediate MP folding and manage misfolding in the cell. When combined with results from genomic sequencing, these studies have established paradigms for how MP folding and misfolding are linked to the molecular etiologies of a variety of diseases. This emerging framework has paved the way for the development of a new class of small molecule "pharmacological chaperones" that bind to and stabilize misfolded MP variants, some of which are now in clinical use. In this review, we comprehensively outline current perspectives on the folding and misfolding of integral MPs as well as the mechanisms of cellular MP quality control. Based on these perspectives, we highlight new opportunities for innovations that bridge our molecular understanding of the energetics of MP folding with the nuanced complexity of biological systems. Given the many linkages between MP misfolding and human disease, we also examine some of the exciting opportunities to leverage these advances to address emerging challenges in the development of therapeutics and precision medicine.


Asunto(s)
Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Deficiencias en la Proteostasis/metabolismo , Humanos , Cinética , Modelos Moleculares , Conformación Proteica , Pliegue de Proteína , Estabilidad Proteica , Proteostasis , Deficiencias en la Proteostasis/patología , Termodinámica
10.
Biochemistry ; 59(13): 1367-1377, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32207963

RESUMEN

More than 80 loss-of-function (LOF) mutations in the SLC6A8 creatine transporter (hCRT1) are responsible for cerebral creatine deficiency syndrome (CCDS), which gives rise to a spectrum of neurological defects, including intellectual disability, epilepsy, and autism spectrum disorder. To gain insight into the nature of the molecular defects caused by these mutations, we quantitatively profiled the cellular processing, trafficking, expression, and function of eight pathogenic CCDS variants in relation to the wild type (WT) and one neutral isoform. All eight CCDS variants exhibit measurable proteostatic deficiencies that likely contribute to the observed LOF. However, the magnitudes of their specific effects on the expression and trafficking of hCRT1 vary considerably, and we find that the LOF associated with two of these variants primarily arises from the disruption of the substrate-binding pocket. In conjunction with an analysis of structural models of the transporter, we use these data to suggest mechanistic classifications for these variants. To evaluate potential avenues for therapeutic intervention, we assessed the sensitivity of these variants to temperature and measured their response to the proteostasis regulator 4-phenylbutyrate (4-PBA). Only one of the tested variants (G132V) is sensitive to temperature, though its response to 4-PBA is negligible. Nevertheless, 4-PBA significantly enhances the activity of WT hCRT1 in HEK293T cells, which suggests it may be worth evaluating as a therapeutic for female intellectual disability patients carrying a single CCDS mutation. Together, these findings reveal that pathogenic SLC6A8 mutations cause a spectrum of molecular defects that should be taken into consideration in future efforts to develop CCDS therapeutics.


Asunto(s)
Encefalopatías Metabólicas Innatas/metabolismo , Creatina/deficiencia , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/deficiencia , Encefalopatías Metabólicas Innatas/genética , Creatina/genética , Creatina/metabolismo , Células HEK293 , Humanos , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/genética , Mutación Missense , Proteínas del Tejido Nervioso/química , Fenilbutiratos/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/química , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo
11.
J Am Chem Soc ; 141(1): 204-215, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30537820

RESUMEN

Membrane proteins are prone to misfolding and degradation within the cell, yet the nature of the conformational defects involved in this process remain poorly understood. The earliest stages of membrane protein folding are mediated by the Sec61 translocon, a molecular machine that facilitates the lateral partitioning of the polypeptide into the membrane. Proper membrane integration is an essential prerequisite for folding of the nascent chain. However, the marginal energetic drivers of this reaction suggest the translocon may operate with modest fidelity. In this work, we employed biophysical modeling in conjunction with quantitative biochemical measurements in order to evaluate the extent to which cotranslational folding defects influence membrane protein homeostasis. Protein engineering was employed to selectively perturb the topological energetics of human rhodopsin, and the expression and cellular trafficking of engineered variants were quantitatively compared. Our results reveal clear relationships between topological energetics and the efficiency of rhodopsin biogenesis, which appears to be limited by the propensity of a polar transmembrane domain to achieve its correct topological orientation. Though the polarity of this segment is functionally constrained, we find that its topology can be stabilized in a manner that enhances biogenesis without compromising the functional properties of rhodopsin. Furthermore, sequence alignments reveal this topological instability has been conserved throughout the course of evolution. These results suggest that topological defects significantly contribute to the inefficiency of membrane protein folding in the cell. Additionally, our findings suggest that the marginal stability of rhodopsin may represent an evolved trait.


Asunto(s)
Pliegue de Proteína , Proteostasis , Rodopsina/biosíntesis , Rodopsina/química , Humanos , Modelos Moleculares , Conformación Proteica , Ingeniería de Proteínas , Rodopsina/genética , Rodopsina/metabolismo , Termodinámica
12.
Biochemistry ; 57(35): 5188-5201, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30085663

RESUMEN

G protein-coupled receptors can exist as dimers and higher-order oligomers in biological membranes. The specific oligomeric assembly of these receptors is believed to play a major role in their function, and the disruption of native oligomers has been implicated in specific human pathologies. Computational predictions and biochemical analyses suggest that two molecules of rhodopsin (Rho) associate through the interactions involving its fifth transmembrane helix (TM5). Interestingly, there are several pathogenic loss-of-function mutations within TM5 that face the lipid bilayer in a manner that could potentially influence the dimerization of Rho. Though several of these mutations are known to induce misfolding, the pathogenic defects associated with V209M and F220C Rho remain unclear. In this work, we utilized a variety of biochemical and biophysical approaches to elucidate the effects of these mutations on the dimerization, folding, trafficking, and function of Rho in relation to other pathogenic TM5 variants. Chemical cross-linking, bioluminescence energy transfer, and pulsed-interleaved excitation fluorescence cross-correlation spectroscopy experiments revealed that each of these mutants exhibits a wild type-like propensity to self-associate within the plasma membrane. However, V209M and F220C each exhibit subtle defects in cellular trafficking. Together, our results suggest that the RP pathology associated with the expression of the V209M and F220C mutants could arise from defects in folding and cellular trafficking rather than the disruption of dimerization, as has been previously proposed.


Asunto(s)
Mutación , Multimerización de Proteína , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Rodopsina/genética , Rodopsina/metabolismo , Secuencia de Aminoácidos , Membrana Celular/metabolismo , Células HEK293 , Humanos , Conformación Proteica , Transporte de Proteínas , Rodopsina/química , Homología de Secuencia
13.
Q Rev Biophys ; 48(1): 1-34, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25420508

RESUMEN

Most biological processes require the production and degradation of proteins, a task that weighs heavily on the cell. Mutations that compromise the conformational stability of proteins place both specific and general burdens on cellular protein homeostasis (proteostasis) in ways that contribute to numerous diseases. Efforts to elucidate the chain of molecular events responsible for diseases of protein folding address one of the foremost challenges in biomedical science. However, relatively little is known about the processes by which mutations prompt the misfolding of α-helical membrane proteins, which rely on an intricate network of cellular machinery to acquire and maintain their functional structures within cellular membranes. In this review, we summarize the current understanding of the physical principles that guide membrane protein biogenesis and folding in the context of mammalian cells. Additionally, we explore how pathogenic mutations that influence biogenesis may differ from those that disrupt folding and assembly, as well as how this may relate to disease mechanisms and therapeutic intervention. These perspectives indicate an imperative for the use of information from structural, cellular, and biochemical studies of membrane proteins in the design of novel therapeutics and in personalized medicine.


Asunto(s)
Células/metabolismo , Proteínas de la Membrana/química , Pliegue de Proteína , Secuencia de Aminoácidos , Animales , Homeostasis , Humanos , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Transporte de Proteínas
14.
Biophys J ; 110(11): 2475-2485, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27276265

RESUMEN

Caveolins mediate the formation of caveolae, which are small omega-shaped membrane invaginations involved in a variety of cellular processes. There are three caveolin isoforms, the third of which (Cav3) is expressed in smooth and skeletal muscles. Mutations in Cav3 cause a variety of human muscular diseases. In this work, we characterize the secondary structure, dynamics, and topology of the monomeric form of the full-length lipidated protein. Cav3 consists of a series of membrane-embedded or surface-associated helical elements connected by extramembrane connecting loops or disordered domains. Our results also reveal that the N-terminal domain undergoes a large scale pH-mediated topological rearrangement between soluble and membrane-anchored forms. Considering that roughly one-third of pathogenic mutations in Cav3 influence charged residues located in this domain, we hypothesize that this transition is likely to be relevant to the molecular basis of Cav3-linked diseases. These results provide insight into the structure of Cav3 and set the stage for mechanistic investigations of the effects of pathogenic mutations.


Asunto(s)
Caveolina 3/metabolismo , Concentración de Iones de Hidrógeno , Secuencia de Aminoácidos , Caveolina 3/genética , Dicroismo Circular , Humanos , Membranas Artificiales , Micelas , Modelos Moleculares , Mutación , Resonancia Magnética Nuclear Biomolecular , Fosfatidilgliceroles/química , Estructura Secundaria de Proteína , Solubilidad , Soluciones
15.
Biochemistry ; 55(7): 985-8, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26859249

RESUMEN

The integration of membrane proteins into "lipid raft" membrane domains influences many biochemical processes. The intrinsic structural properties of membrane proteins are thought to mediate their partitioning between membrane domains. However, whether membrane topology influences the targeting of proteins to rafts remains unclear. To address this question, we examined the domain preference of three putative raft-associated membrane proteins with widely different topologies: human caveolin-3, C99 (the 99 residue C-terminal domain of the amyloid precursor protein), and peripheral myelin protein 22. We find that each of these proteins are excluded from the ordered domains of giant unilamellar vesicles containing coexisting liquid-ordered and liquid-disordered phases. Thus, the intrinsic structural properties of these three topologically distinct disease-linked proteins are insufficient to confer affinity for synthetic raft-like domains.


Asunto(s)
Precursor de Proteína beta-Amiloide/química , Caveolina 3/química , Microdominios de Membrana/química , Modelos Moleculares , Proteínas de la Mielina/química , Fragmentos de Péptidos/química , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Caveolina 3/genética , Caveolina 3/metabolismo , Colesterol/química , Colesterol/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Microdominios de Membrana/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/química , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Rodaminas/química , Esfingomielinas/química , Esfingomielinas/metabolismo , Liposomas Unilamelares
16.
J Am Chem Soc ; 137(27): 8758-68, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26102530

RESUMEN

Despite broad biochemical relevance, our understanding of the physiochemical reactions that limit the assembly and cellular trafficking of integral membrane proteins remains superficial. In this work, we report the first experimental assessment of the relationship between the conformational stability of a eukaryotic membrane protein and the degree to which it is retained by cellular quality control in the secretory pathway. We quantitatively assessed both the conformational equilibrium and cellular trafficking of 12 variants of the α-helical membrane protein peripheral myelin protein 22 (PMP22), the intracellular misfolding of which is known to cause peripheral neuropathies associated with Charcot-Marie-Tooth disease (CMT). We show that the extent to which these mutations influence the energetics of Zn(II)-mediated PMP22 folding is proportional to the observed reduction in cellular trafficking efficiency. Strikingly, quantitative analyses also reveal that the reduction of motor nerve conduction velocities in affected patients is proportional to the extent of the mutagenic destabilization. This finding provides compelling evidence that the effects of these mutations on the energetics of PMP22 folding lie at the heart of the molecular basis of CMT. These findings highlight conformational stability as a key factor governing membrane protein biogenesis and suggest novel therapeutic strategies for CMT.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Mutación Missense , Proteínas de la Mielina/química , Proteínas de la Mielina/genética , Pliegue de Proteína , Secuencia de Aminoácidos , Animales , Enfermedad de Charcot-Marie-Tooth/metabolismo , Perros , Humanos , Células de Riñón Canino Madin Darby , Metales/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas de la Mielina/metabolismo , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/metabolismo , Conformación Proteica , Estabilidad Proteica , Transporte de Proteínas , Termodinámica
17.
J Membr Biol ; 248(3): 371-81, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25192979

RESUMEN

Aberrant protein folding and assembly contribute to a number of diseases, and efforts to rationalize how pathogenic mutations cause this phenomenon represent an important imperative in biochemical research. However, for α-helical membrane proteins, this task is complicated by the fact that membrane proteins require intricate machinery to achieve structural and functional maturity under cellular conditions. In this work, we utilized the ΔG predictor algorithm ( www.dgpred.cbr.su.se ) to survey 470 known pathogenic mutations occurring in five misfolding-prone α-helical membrane proteins for their predicted effects on the translocon-mediated membrane integration of transmembrane helices, a critical step in biosynthesis and folding of nascent membrane proteins. The results suggest that about 10 % of these mutations are likely to have adverse effects on the topogenesis of nascent membrane proteins. These results suggest that the misfolding of a modest but nonetheless significant subset of pathogenic variants may begin at the translocon. Potential implications for therapeutic design and personalized medicine are discussed.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Canal de Potasio KCNQ1/química , Membrana Dobles de Lípidos/química , Proteínas de la Mielina/química , Receptores de Vasopresinas/química , Rodopsina/química , Secuencia de Aminoácidos , Animales , Bovinos , Secuencia Conservada , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Canal de Potasio KCNQ1/genética , Mutación , Mutación Missense , Proteínas de la Mielina/genética , Estructura Secundaria de Proteína , Transporte de Proteínas , Receptores de Vasopresinas/genética , Rodopsina/genética , Termodinámica
18.
Biophys J ; 117(5): 793-794, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31400915
19.
Biochemistry ; 53(27): 4320-2, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24960539

RESUMEN

Caveolin-3 (Cav3) is an unconventional membrane protein that serves as a critical scaffolding hub in caveolae and is genetically linked to various muscle disorders. In this work, we report the expression, purification, and characterization of full-length human Cav3. To mimic the palmitoylation of endogenous Cav3, we developed a generally applicable approach to covalently attached thioalkyl chains at natively modified cysteine residues. Nuclear magnetic resonance measurements indicate that lipidation exerts only a modest and local effect on the Cav3 structure, with little impact on the structures of the N-terminal domain, the scaffolding domain, and the extreme C-terminus.


Asunto(s)
Caveolina 3/química , Caveolina 3/genética , Humanos , Lipoilación , Mutación , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
20.
J Am Chem Soc ; 136(47): 16574-81, 2014 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-25369295

RESUMEN

The folding mechanisms of helical membrane proteins remain largely uncharted. Here we characterize the kinetics of bacteriorhodopsin folding and employ φ-value analysis to explore the folding transition state. First, we developed and confirmed a kinetic model that allowed us to assess the rate of folding from SDS-denatured bacteriorhodopsin (bRU) and provides accurate thermodynamic information even under influence of retinal hydrolysis. Next, we obtained reliable φ-values for 16 mutants of bacteriorhodopsin with good coverage across the protein. Every φ-value was less than 0.4, indicating the transition state is not uniquely structured. We suggest that the transition state is a loosely organized ensemble of conformations.


Asunto(s)
Bacteriorodopsinas/química , Bacteriorodopsinas/genética , Hidrólisis , Cinética , Modelos Moleculares , Mutación , Desnaturalización Proteica , Pliegue de Proteína , Dodecil Sulfato de Sodio/química , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA