Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
1.
Chembiochem ; 25(1): e202300551, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-37856284

RESUMO

Stress-activated signaling pathways orchestrate cellular behaviors and fates. Studying the precise role(s) of stress-activated protein kinases is challenging, because stress conditions induce adaptation and impose selection pressure. To meet this challenge, we have applied an optogenetic system with a single plasmid to express light-activated p38α or its upstream activator, MKK6, in conjunction with live-cell fluorescence microscopy. In starved cells, decaging of constitutively active p38α or MKK6 by brief exposure to UV light elicits rapid p38-mediated signaling, release of cytochrome c from mitochondria, and apoptosis with different kinetics. In parallel, light activation of p38α also suppresses autophagosome formation, similarly to stimulation with growth factors that activate PI3K/Akt/mTORC1 signaling. Active MKK6 negatively regulates serum-induced ERK activity, which is p38-independent as previously reported. Here, we reproduce that result with the one plasmid system and show that although decaging active p38α does not reduce basal ERK activity in our cells, it can block growth factor-stimulated ERK signaling in serum-starved cells. These results clarify the roles of MKK6 and p38α in dynamic signaling programs, which act in concert to actuate apoptotic death while suppressing cell survival mechanisms.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina , Proteínas Quinases Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Fosfatidilinositol 3-Quinases , Proteínas Quinases p38 Ativadas por Mitógeno , MAP Quinase Quinase 6/genética
2.
Org Biomol Chem ; 22(2): 302-308, 2024 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-38054844

RESUMO

The STING pathway is critical to innate immunity and is being investigated as a potential therapeutic target. Existing agents targeting STING suffer from several undesirable effects, particularly the possibility of systematic activation, which increases the risk of autoimmune disorders. In this proof-of-concept study, we report the development of a light-activated STING agonist, based on the potent compound SR-717. We first screened the activity of the non-caged agonist toward 5 human STING variants to identify the most viable target. A photocaged agonist was designed and synthesized in order to block an essential interaction between the carboxy acid group of the ligand with the R238 residue of the STING protein. We then investigated the selective activation of STING with the photocaged agonist, demonstrating an irradiation-dependent response. The development and characterization of this selective agonist expands the growing toolbox of conditionally controlled STING agonists to avoid systematic immune activation.


Assuntos
Imunidade Inata , Proteínas de Membrana , Humanos , Proteínas de Membrana/agonistas
3.
Angew Chem Int Ed Engl ; 63(17): e202318773, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38411401

RESUMO

Conditionally controlled antisense oligonucleotides provide precise interrogation of gene function at different developmental stages in animal models. Only one example of small molecule-induced activation of antisense function exist. This has been restricted to cyclic caged morpholinos that, based on sequence, can have significant background activity in the absence of the trigger. Here, we provide a new approach using azido-caged nucleobases that are site-specifically introduced into antisense morpholinos. The caging group design is a simple azidomethylene (Azm) group that, despite its very small size, efficiently blocks Watson-Crick base pairing in a programmable fashion. Furthermore, it undergoes facile decaging via Staudinger reduction when exposed to a small molecule phosphine, generating the native antisense oligonucleotide under conditions compatible with biological environments. We demonstrated small molecule-induced gene knockdown in mammalian cells, zebrafish embryos, and frog embryos. We validated the general applicability of this approach by targeting three different genes.


Assuntos
Oligonucleotídeos , Peixe-Zebra , Animais , Morfolinos/genética , Morfolinos/farmacologia , Oligonucleotídeos Antissenso , Fenótipo , Mamíferos
4.
J Am Chem Soc ; 145(4): 2395-2403, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36662675

RESUMO

Precise temporally regulated protein function directs the highly complex processes that make up embryo development. The zebrafish embryo is an excellent model organism to study development, and conditional control over enzymatic activity is desirable to target chemical intervention to specific developmental events and to investigate biological mechanisms. Surprisingly few, generally applicable small molecule switches of protein function exist in zebrafish. Genetic code expansion allows for site-specific incorporation of unnatural amino acids into proteins that contain caging groups that are removed through addition of small molecule triggers such as phosphines or tetrazines. This broadly applicable control of protein function was applied to activate several enzymes, including a GTPase and a protease, with temporal precision in zebrafish embryos. Simple addition of the small molecule to the media produces robust and tunable protein activation, which was used to gain insight into the development of a congenital heart defect from a RASopathy mutant of NRAS and to control DNA and protein cleavage events catalyzed by a viral recombinase and a viral protease, respectively.


Assuntos
Proteínas , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Proteínas/metabolismo , Aminoácidos/metabolismo , Código Genético , Embrião não Mamífero , Engenharia de Proteínas
5.
J Am Chem Soc ; 145(30): 16458-16463, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37473438

RESUMO

Covalent aptamers are novel biochemical tools for fast and selective transfer of labels to target proteins. Equipped with cleavable electrophiles, these nucleic acid probes enable the installation of functional handles onto native proteins. The high affinity and specificity with which aptamers bind their selected targets allows for quick, covalent labeling that can compete with nuclease-mediated degradation. Here, we introduce the first application of covalent aptamers to modify a specific cell surface protein through proximity-driven label transfer. We targeted protein tyrosine kinase 7 (PTK7), a prominent cancer marker, and demonstrated aptamer-mediated biotin transfer to specific lysine residues on the extracellular domain of the protein. This allowed for tracking of PTK7 expression, localization, and cellular internalization. These studies validate the programmability of covalent aptamers and highlight their applicability in a cellular context, including protein and small molecule delivery.


Assuntos
Aptâmeros de Nucleotídeos , Aptâmeros de Nucleotídeos/química , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Lisina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Técnica de Seleção de Aptâmeros
6.
J Am Chem Soc ; 145(4): 2414-2420, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36669466

RESUMO

Genetic code expansion has pushed protein chemistry past the canonical 22 amino acids. The key enzymes that make this possible are engineered aminoacyl tRNA synthetases. However, as the number of genetically encoded amino acids has increased over the years, obvious limits in the type and size of novel side chains that can be accommodated by the synthetase enzyme become apparent. Here, we show that chemically acylating tRNAs allow for robust, site-specific incorporation of unnatural amino acids into proteins in zebrafish embryos, an important model organism for human health and development. We apply this approach to incorporate a unique photocaged histidine analogue for which synthetase engineering efforts have failed. Additionally, we demonstrate optical control over different enzymes in live embryos by installing photocaged histidine into their active sites.


Assuntos
Aminoacil-tRNA Sintetases , RNA de Transferência , Peixe-Zebra , Animais , Aminoácidos/química , Aminoacil-tRNA Sintetases/genética , Aminoacil-tRNA Sintetases/metabolismo , Código Genético , Histidina/genética , RNA de Transferência/genética , RNA de Transferência/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
7.
J Am Chem Soc ; 145(45): 24459-24465, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-38104267

RESUMO

Light is well-established for control of bond breakage but not for control of specific bond formation in complex environments. We previously engineered the diffusion-limited reactivity of the SpyTag003 peptide with its protein partner SpyCatcher003 through spontaneous isopeptide bond formation. This system enables precise and irreversible assembly of biological building blocks with applications from biomaterials to vaccines. Here we establish a system for the rapid control of this amide bond formation with visible light. We have generated a caged SpyCatcher003, which allows light triggering of covalent bond formation to SpyTag003 in mammalian cells. Photocaging is achieved through site-specific incorporation of an unnatural coumarin-lysine at the reactive site of SpyCatcher003. We showed a uniform specific reaction in cell lysate upon light activation. We then used the spatiotemporal precision of a 405 nm confocal laser for uncaging in seconds, probing the earliest events in mechanotransduction by talin, the key force sensor between the cytoskeleton and the extracellular matrix. Reconstituting talin induced rapid biphasic extension of lamellipodia, revealing the kinetics of talin-regulated cell spreading and polarization. Thereafter we determined the hierarchy of the recruitment of key components for cell adhesion. Precise control over site-specific protein reaction with visible light creates diverse opportunities for cell biology and nanoassembly.


Assuntos
Mecanotransdução Celular , Talina , Animais , Adesão Celular , Talina/metabolismo , Mecanotransdução Celular/fisiologia , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Mamíferos/metabolismo
8.
Chembiochem ; 24(7): e202200721, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36642698

RESUMO

The use of light to control protein function is a critical tool in chemical biology. Here we describe the addition of a photocaged histidine to the genetic code. This unnatural amino acid becomes histidine upon exposure to light and allows for the optical control of enzymes that utilize active-site histidine residues. We demonstrate light-induced activation of a blue fluorescent protein and a chloramphenicol transferase. Further, we genetically encoded photocaged histidine in mammalian cells. We then used this approach in live cells for optical control of firefly luciferase and, Renilla luciferase. This tool should have utility in manipulating and controlling a wide range of biological processes.


Assuntos
Aminoácidos , Histidina , Animais , Histidina/genética , Aminoácidos/química , Proteínas/metabolismo , Luciferases de Renilla/genética , Código Genético , Mamíferos/genética , Mamíferos/metabolismo
9.
Nat Chem Biol ; 17(9): 998-1007, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34341589

RESUMO

Subcellular compartmentalization of macromolecules increases flux and prevents inhibitory interactions to control biochemical reactions. Inspired by this functionality, we sought to build designer compartments that function as hubs to regulate the flow of information through cellular control systems. We report a synthetic membraneless organelle platform to control endogenous cellular activities through sequestration and insulation of native proteins. We engineer and express a disordered protein scaffold to assemble micron-size condensates and recruit endogenous clients via genomic tagging with high-affinity dimerization motifs. By relocalizing up to 90% of targeted enzymes to synthetic condensates, we efficiently control cellular behaviors, including proliferation, division and cytoskeletal organization. Further, we demonstrate multiple strategies for controlled cargo release from condensates to switch cells between functional states. These synthetic organelles offer a powerful and generalizable approach to modularly control cell decision-making in a variety of model systems with broad applications for cellular engineering.


Assuntos
Engenharia Celular , Organelas/metabolismo , Divisão Celular , Linhagem Celular Tumoral , Proliferação de Células , Citoesqueleto/metabolismo , Humanos , Organelas/química
10.
Biochemistry ; 61(23): 2687-2697, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36346979

RESUMO

Signal transduction pathways are responsible for maintaining cellular functions, including proliferation, differentiation, apoptosis, and cell cycle progression. These pathways are maintained through the propagation of phosphorylation signals by protein kinases, as well as the removal of phosphorylation signals by protein phosphatases. Depending on the context, post-translational modification could have either a positive or negative effect on a signaling pathway. Intricate networks of positive and negative regulators offer a challenging target for the dissection of cell signaling mechanisms, particularly regarding the more subtle dampening of signal transduction through phosphatases. We report the development of two complimentary methods for the optical control of a complex phosphatase: SH2 domain-containing protein tyrosine phosphatase-2 (SHP2). We investigated controlling the catalytic function of SHP2 through (1) site-specific incorporation of a caged tyrosine for light activation of catalytic activity for the control of an essential substrate binding residue and (2) site-specific incorporation of a caged lysine at a conserved residue within an allosteric pocket for the control of SHP2 binding partner docking sites. These methods are generalizable to proteins bearing either a protein tyrosine phosphatase (PTP) catalytic domain or an SH2 domain, including SHP1, PTP family phosphatases, and a diverse range of SH2 domain-containing proteins.


Assuntos
Proteína Tirosina Fosfatase não Receptora Tipo 11 , Domínios de Homologia de src , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Tirosina Fosfatases Contendo o Domínio SH2 , Transdução de Sinais , Fosforilação
11.
Biochemistry ; 61(22): 2470-2481, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-35918061

RESUMO

Many proteins harboring low complexity or intrinsically disordered sequences (IDRs) are capable of undergoing liquid-liquid phase separation to form mesoscale condensates that function as biochemical niches with the ability to concentrate or sequester macromolecules and regulate cellular activity. Engineered disordered proteins have been used to generate programmable synthetic membraneless organelles in cells. Phase separation is governed by the strength of interactions among polypeptides with multivalency enhancing phase separation at lower concentrations. Previously, we and others demonstrated enzymatic control of IDR valency from multivalent precursors to dissolve condensed phases. Here, we develop noncovalent strategies to multimerize an individual IDR, the RGG domain of LAF-1, using protein interaction domains to regulate condensate formation in vitro and in living cells. First, we characterize modular dimerization of RGG domains at either terminus using cognate high-affinity coiled-coil pairs to form stable condensates in vitro. Second, we demonstrate temporal control over phase separation of RGG domains fused to FRB and FKBP in the presence of dimerizer. Further, using a photocaged dimerizer, we achieve optically induced condensation both in cell-sized emulsions and within live cells. Collectively, these modular tools allow multiple strategies to promote phase separation of a common core IDR for tunable control of condensate assembly.


Assuntos
Fenômenos Bioquímicos , Proteínas Intrinsicamente Desordenadas , Proteínas Intrinsicamente Desordenadas/química , Transição de Fase , Domínios Proteicos , Biossíntese de Proteínas
12.
Dev Biol ; 475: 21-29, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33684434

RESUMO

Echinoderms are important experimental models for analyzing embryonic development, but a lack of spatial and temporal control over gene perturbations has hindered developmental studies using these animals. Morpholino antisense oligonucleotides (MOs) have been used successfully by the echinoderm research community for almost two decades, and MOs remain the most widely used tool for acute gene knockdowns in these organisms. Echinoderm embryos develop externally and are optically transparent, making them ideally-suited to many light-based approaches for analyzing and manipulating development. Studies using zebrafish embryos have demonstrated the effectiveness of photoactivatable (caged) MOs for conditional gene knockdowns. Here we show that caged MOs, synthesized using nucleobase-caged monomers, provide light-regulated control over gene expression in sea urchin embryos. Our work provides the first robust approach for conditional gene silencing in this prominent model system.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Morfolinos/farmacologia , Ouriços-do-Mar/genética , Animais , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/genética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Inativação Gênica/fisiologia , Morfolinos/química , Oligonucleotídeos Antissenso/genética
13.
J Biol Chem ; 296: 100094, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33485192

RESUMO

Controlling unmodified serotonin levels in brain synapses is a primary objective when treating major depressive disorder-a disease that afflicts ∼20% of the world's population. Roughly 60% of patients respond poorly to first-line treatments and thus new therapeutic strategies are sought. To this end, we have constructed isoform-specific inhibitors of the human cytosolic sulfotransferase 1A3 (SULT1A3)-the isoform responsible for sulfonating ∼80% of the serotonin in the extracellular brain fluid. The inhibitor design includes a core ring structure, which anchors the inhibitor into a SULT1A3-specific binding pocket located outside the active site, and a side chain crafted to act as a latch to inhibit turnover by fastening down the SULT1A3 active-site cap. The inhibitors are allosteric, they bind with nanomolar affinity and are highly specific for the 1A3 isoform. The cap-stabilizing effects of the latch can be accurately calculated and are predicted to extend throughout the cap and into the surrounding protein. A free-energy correlation demonstrates that the percent inhibition at saturating inhibitor varies linearly with cap stabilization - the correlation is linear because the rate-limiting step of the catalytic cycle, nucleotide release, scales linearly with the fraction of enzyme in the cap-open form. Inhibitor efficacy in cultured cells was studied using a human mammary epithelial cell line that expresses SULT1A3 at levels comparable with those found in neurons. The inhibitors perform similarly in ex vivo and in vitro studies; consequently, SULT1A3 turnover can now be potently suppressed in an isoform-specific manner in human cells.


Assuntos
Células Epiteliais/metabolismo , Neurotransmissores/metabolismo , Sítio Alostérico , Arilsulfotransferase/metabolismo , Catecolaminas/metabolismo , Transtorno Depressivo Maior/metabolismo , Humanos , Cinética , Simulação de Dinâmica Molecular , Estrutura Molecular , Serotonina/metabolismo , Relação Estrutura-Atividade , Sulfotransferases/metabolismo
14.
J Am Chem Soc ; 144(37): 16819-16826, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-36073798

RESUMO

MicroRNAs play crucial and dynamic roles in vertebrate development and diseases. Some, like miR-430, are highly expressed during early embryo development and regulate hundreds of transcripts, which can make it difficult to study their role in the timing and location of specific developmental processes using conventional morpholino oligonucleotide (MO) knockdown or genetic deletion approaches. We demonstrate that light-activated circular morpholino oligonucleotides (cMOs) can be applied to the conditional control of microRNA function. We targeted miR-430 in zebrafish embryos to study its role in the development of the embryo body and the heart. Using 405 nm irradiation, precise spatial and temporal control over miR-430 function was demonstrated, offering insight into the cell populations and developmental timepoints involved in each process.


Assuntos
MicroRNAs , Peixe-Zebra , Animais , Embrião não Mamífero , MicroRNAs/genética , Morfolinos/farmacologia , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Proteínas de Peixe-Zebra/genética
15.
Chembiochem ; 23(23): e202200297, 2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36196665

RESUMO

A wide array of optogenetic tools are available that allow for precise spatiotemporal control over cellular processes. These tools are particularly important to zebrafish researchers who take advantage of the embryo's transparency. However, photocleavable optogenetic proteins have not been utilized in zebrafish. We demonstrate successful optical control of protein cleavage in embryos using PhoCl, a photocleavable fluorescent protein. This optogenetic tool offers temporal and spatial control over protein cleavage events, which we demonstrate in light-triggered protein translocation and light-triggered apoptosis.


Assuntos
Optogenética , Peixe-Zebra , Animais , Peixe-Zebra/genética , Transporte Proteico , Proteólise , Apoptose
16.
Chembiochem ; 23(21): e202200374, 2022 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-36068175

RESUMO

Caged morpholino oligonucleotides (cMOs) are synthetic tools that allow light-inducible gene silencing in live organisms. Previously reported cMOs have utilized hairpin, duplex, and cyclic structures, as well as caged nucleobases. While these antisense technologies enable efficient optical control of RNA splicing and translation, they can have limited dynamic range. A new caging strategy was developed where the two MO termini are conjugated to an internal position through a self-immolative trifunctional linker, thereby generating a bicyclic cMO that is conformationally resistant to RNA binding. The efficacy of this alternative cMO design has been demonstrated in zebrafish embryos and compared to linear MOs and monocyclic constructs.


Assuntos
Inativação Gênica , Peixe-Zebra , Animais , Morfolinos/química , Peixe-Zebra/genética
17.
Bioconjug Chem ; 33(12): 2361-2369, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36459098

RESUMO

Despite a range of covalent protein modifications, few techniques exist for quantification of protein bioconjugation in cells. Here, we describe a novel method for quantifying in cellulo protein bioconjugation through covalent bond formation with HaloTag. This approach utilizes unnatural amino acid (UAA) mutagenesis to selectively install a small and bioorthogonally reactive handle onto the surface of a protein. We utilized the fast kinetics and high selectivity of inverse electron-demand Diels-Alder cycloadditions to evaluate reactions of tetrazine phenylalanine (TetF) with strained trans-cyclooctene-chloroalkane (sTCO-CA) and trans-cyclooctene lysine (TCOK) with tetrazine-chloroalkane (Tet-CA). Following bioconjugation, the chloroalkane ligand is exposed for labeling by the HaloTag enzyme, allowing for straightforward quantification of bioconjugation via simple western blot analysis. We demonstrate the versatility of this tool for quickly and accurately determining the bioconjugation efficiency of different UAA/chloroalkane pairs and for different sites on different proteins of interest, including EGFP and the estrogen-related receptor ERRα.


Assuntos
Compostos Heterocíclicos , Proteínas , Animais , Proteínas/química , Aminoácidos/química , Fenilalanina , Ciclo-Octanos/química , Reação de Cicloadição , Mamíferos/metabolismo
18.
Chem Soc Rev ; 50(23): 13253-13267, 2021 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-34739027

RESUMO

Translation of mRNA into protein is one of the most fundamental processes within biological systems. Gene expression is tightly regulated both in space and time, often involving complex signaling or gene regulatory networks, as most prominently observed in embryo development. Thus, studies of gene function require tools with a matching level of external control. Light is an excellent conditional trigger as it is minimally invasive, can be easily tuned in wavelength and amplitude, and can be applied with excellent spatial and temporal resolution. To this end, modification of established oligonucleotide-based technologies with optical control elements, in the form of photocaging groups and photoswitches, has rendered these tools capable of navigating the dynamic regulatory pathways of mRNA translation in cellular and in vivo models. In this review, we discuss the different optochemical approaches used to generate photoresponsive nucleic acids that activate and deactivate gene expression and function at the translational level.


Assuntos
Ácidos Nucleicos , Expressão Gênica , Luz , Oligonucleotídeos , Proteínas/genética
19.
J Biol Chem ; 295(25): 8494-8504, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32371393

RESUMO

The selective pressure imposed by extrinsic death signals and stressors adds to the challenge of isolating and interpreting the roles of proteins in stress-activated signaling networks. By expressing a kinase with activating mutations and a caged lysine blocking the active site, we can rapidly switch on catalytic activity with light and monitor the ensuing dynamics. Applying this approach to MAP kinase 6 (MKK6), which activates the p38 subfamily of MAPKs, we found that decaging active MKK6 in fibroblasts is sufficient to trigger apoptosis in a p38-dependent manner. Both in fibroblasts and in a murine melanoma cell line expressing mutant B-Raf, MKK6 activation rapidly and potently inhibited the pro-proliferative extracellular signal-regulated kinase (ERK) pathway; to our surprise, this negative cross-regulation was equally robust when all p38 isoforms were inhibited. These results position MKK6 as a new pleiotropic signal transducer that promotes both pro-apoptotic and anti-proliferative signaling, and they highlight the utility of caged, light-activated kinases for dissecting stress-activated signaling networks.


Assuntos
MAP Quinase Quinase 6/metabolismo , Transdução de Sinais/efeitos da radiação , Raios Ultravioleta , Animais , Apoptose/efeitos da radiação , Linhagem Celular , Proliferação de Células/efeitos da radiação , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , MAP Quinase Quinase 6/genética , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mutagênese , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
J Am Chem Soc ; 143(24): 9222-9229, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34121391

RESUMO

Development of methodologies for optically triggered protein degradation enables the study of dynamic protein functions, such as those involved in cell signaling, that are difficult to be probed with traditional genetic techniques. Here, we describe the design and implementation of a novel light-controlled peptide degron conferring N-end pathway degradation to its protein target. The degron comprises a photocaged N-terminal amino acid and a lysine-rich, 13-residue linker. By caging the N-terminal residue, we were able to optically control N-degron recognition by an E3 ligase, consequently controlling ubiquitination and proteasomal degradation of the target protein. We demonstrate broad applicability by applying this approach to a diverse set of target proteins, including EGFP, firefly luciferase, the kinase MEK1, and the phosphatase DUSP6 (also known as MKP3). The caged degron can be used with minimal protein engineering and provides virtually complete, light-triggered protein degradation on a second to minute time scale.


Assuntos
Fosfatase 6 de Especificidade Dupla/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Luciferases de Vaga-Lume/metabolismo , MAP Quinase Quinase 1/metabolismo , Peptídeos/metabolismo , Animais , Fosfatase 6 de Especificidade Dupla/química , Vaga-Lumes , Proteínas de Fluorescência Verde/química , Humanos , Luciferases de Vaga-Lume/química , MAP Quinase Quinase 1/química , Peptídeos/química , Conformação Proteica , Engenharia de Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA