Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.023
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(7): 1465-1477.e18, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-37001505

RESUMEN

Receptor activity-modifying proteins (RAMPs) modulate the activity of many Family B GPCRs. We show that RAMP2 directly interacts with the glucagon receptor (GCGR), a Family B GPCR responsible for blood sugar homeostasis, and broadly inhibits receptor-induced downstream signaling. HDX-MS experiments demonstrate that RAMP2 enhances local flexibility in select locations in and near the receptor extracellular domain (ECD) and in the 6th transmembrane helix, whereas smFRET experiments show that this ECD disorder results in the inhibition of active and intermediate states of the intracellular surface. We determined the cryo-EM structure of the GCGR-Gs complex at 2.9 Å resolution in the presence of RAMP2. RAMP2 apparently does not interact with GCGR in an ordered manner; however, the receptor ECD is indeed largely disordered along with rearrangements of several intracellular hallmarks of activation. Our studies suggest that RAMP2 acts as a negative allosteric modulator of GCGR by enhancing conformational sampling of the ECD.


Asunto(s)
Glucagón , Receptores de Glucagón , Membrana Celular/metabolismo , Glucagón/metabolismo , Receptores de Glucagón/metabolismo , Proteína 2 Modificadora de la Actividad de Receptores/metabolismo
2.
Cell ; 172(3): 605-617.e11, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29336887

RESUMEN

The bacterial chaperonin GroEL and its cofactor, GroES, form a nano-cage for a single molecule of substrate protein (SP) to fold in isolation. GroEL and GroES undergo an ATP-regulated interaction cycle to close and open the folding cage. GroEL consists of two heptameric rings stacked back to back. Here, we show that GroEL undergoes transient ring separation, resulting in ring exchange between complexes. Ring separation occurs upon ATP-binding to the trans ring of the asymmetric GroEL:7ADP:GroES complex in the presence or absence of SP and is a consequence of inter-ring negative allostery. We find that a GroEL mutant unable to perform ring separation is folding active but populates symmetric GroEL:GroES2 complexes, where both GroEL rings function simultaneously rather than sequentially. As a consequence, SP binding and release from the folding chamber is inefficient, and E. coli growth is impaired. We suggest that transient ring separation is an integral part of the chaperonin mechanism.


Asunto(s)
Chaperonina 60/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Chaperonina 10/metabolismo , Chaperonina 60/química , Chaperonina 60/genética , Mutación , Unión Proteica
3.
Cell ; 171(7): 1625-1637.e13, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29198525

RESUMEN

When unfolded proteins accumulate in the endoplasmic reticulum (ER), the unfolded protein response (UPR) increases ER-protein-folding capacity to restore protein-folding homeostasis. Unfolded proteins activate UPR signaling across the ER membrane to the nucleus by promoting oligomerization of IRE1, a conserved transmembrane ER stress receptor. However, the coupling of ER stress to IRE1 oligomerization and activation has remained obscure. Here, we report that the ER luminal co-chaperone ERdj4/DNAJB9 is a selective IRE1 repressor that promotes a complex between the luminal Hsp70 BiP and the luminal stress-sensing domain of IRE1α (IRE1LD). In vitro, ERdj4 is required for complex formation between BiP and IRE1LD. ERdj4 associates with IRE1LD and recruits BiP through the stimulation of ATP hydrolysis, forcibly disrupting IRE1 dimers. Unfolded proteins compete for BiP and restore IRE1LD to its default, dimeric, and active state. These observations establish BiP and its J domain co-chaperones as key regulators of the UPR.


Asunto(s)
Endorribonucleasas/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Respuesta de Proteína Desplegada , Animales , Cricetinae , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Humanos , Pliegue de Proteína
4.
Mol Cell ; 82(2): 304-314, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35063098

RESUMEN

Owing to their unique abilities to manipulate, label, and image individual molecules in vitro and in cellulo, single-molecule techniques provide previously unattainable access to elementary biological processes. In imaging, single-molecule fluorescence resonance energy transfer (smFRET) and protein-induced fluorescence enhancement in vitro can report on conformational changes and molecular interactions, single-molecule pull-down (SiMPull) can capture and analyze the composition and function of native protein complexes, and single-molecule tracking (SMT) in live cells reveals cellular structures and dynamics. In labeling, the abilities to specifically label genomic loci, mRNA, and nascent polypeptides in cells have uncovered chromosome organization and dynamics, transcription and translation dynamics, and gene expression regulation. In manipulation, optical tweezers, integration of single-molecule fluorescence with force measurements, and single-molecule force probes in live cells have transformed our mechanistic understanding of diverse biological processes, ranging from protein folding, nucleic acids-protein interactions to cell surface receptor function.


Asunto(s)
Genómica/tendencias , Imagen Molecular/tendencias , Imagen Óptica/tendencias , Imagen Individual de Molécula/tendencias , Animales , Difusión de Innovaciones , Transferencia Resonante de Energía de Fluorescencia/tendencias , Humanos , Microscopía Fluorescente/tendencias , Proteómica/tendencias
5.
Mol Cell ; 70(1): 60-71.e15, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29606590

RESUMEN

Fidaxomicin is an antibacterial drug in clinical use for treatment of Clostridium difficile diarrhea. The active ingredient of fidaxomicin, lipiarmycin A3 (Lpm), functions by inhibiting bacterial RNA polymerase (RNAP). Here we report a cryo-EM structure of Mycobacterium tuberculosis RNAP holoenzyme in complex with Lpm at 3.5-Å resolution. The structure shows that Lpm binds at the base of the RNAP "clamp." The structure exhibits an open conformation of the RNAP clamp, suggesting that Lpm traps an open-clamp state. Single-molecule fluorescence resonance energy transfer experiments confirm that Lpm traps an open-clamp state and define effects of Lpm on clamp dynamics. We suggest that Lpm inhibits transcription by trapping an open-clamp state, preventing simultaneous interaction with promoter -10 and -35 elements. The results account for the absence of cross-resistance between Lpm and other RNAP inhibitors, account for structure-activity relationships of Lpm derivatives, and enable structure-based design of improved Lpm derivatives.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Escherichia coli/efectos de los fármacos , Fidaxomicina/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Antibacterianos/química , Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Sitios de Unión , Microscopía por Crioelectrón , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/ultraestructura , Diseño de Fármacos , Farmacorresistencia Bacteriana/genética , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/ultraestructura , Fidaxomicina/química , Fidaxomicina/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Modelos Moleculares , Mutación , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/ultraestructura , Unión Proteica , Conformación Proteica , Imagen Individual de Molécula , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/enzimología , Staphylococcus aureus/genética , Relación Estructura-Actividad
6.
J Biol Chem ; 300(2): 105636, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38199572

RESUMEN

The sequence-specific endoribonuclease MazF is widely conserved among prokaryotes. Approximately 20 different MazF cleavage sequences have been discovered, varying from three to seven nucleotides in length. Although MazFs from various prokaryotes were found, the cleavage sequences of most MazFs are unknown. Here, we characterized the conserved MazF of Salmonella enterica subsp. arizonae (MazF-SEA). Using massive parallel sequencing and fluorometric assays, we revealed that MazF-SEA preferentially cleaves the sequences U∧ACG and U∧ACU (∧ represents cleavage sites). In addition, we predicted the 3D structure of MazF-SEA using AlphaFold2 and aligned it with the crystal structure of RNA-bound Bacillus subtilis MazF to evaluate RNA interactions. We found Arg-73 of MazF-SEA interacts with RNAs containing G and U at the third position from the cleavage sites (U∧ACG and U∧ACU). We then obtained the mutated MazF-SEA R73L protein to evaluate the significance of Arg-73 interaction with RNAs containing G and U at this position. We also used fluorometric and kinetic assays and showed the enzymatic activity of MazF-SEA R73L for the sequence UACG and UACU was significantly decreased. These results suggest Arg-73 is essential for recognizing G and U at the third position from the cleavage sites. This is the first study to our knowledge to identify a single residue responsible for RNA recognition by MazF. Owing to its high specificity and ribosome-independence, MazF is useful for RNA cleavage in vitro. These results will likely contribute to increasing the diversity of MazF specificity and to furthering the application of MazF in RNA engineering.


Asunto(s)
Salmonella enterica , Endonucleasas , Endorribonucleasas/metabolismo , Guanina , ARN Bacteriano/metabolismo , ARN Mensajero/genética , Salmonella enterica/enzimología , Salmonella enterica/genética , Uracilo
7.
J Biol Chem ; 300(10): 107778, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39270821

RESUMEN

Propofol is a widely used anesthetic and sedative that acts as a positive allosteric modulator of gamma-aminobutyric acid type A (GABAA) receptors. Several potential propofol binding sites that may mediate this effect have been identified using propofol-analogue photoaffinity labeling. Ortho-propofol diazirine (o-PD) labels ß-H267, a pore-lining residue, whereas AziPm labels residues ß-M286, ß-M227, and α-I239 in the two membrane-facing interfaces [ß(+)/α(-) and α(+)/ß(-)] between α and ß subunits. This study used photoaffinity labeling of α1ß3 GABAA receptors to reconcile the apparently conflicting results obtained with AziPm and o-PD labeling, focusing on whether ß3-H267 identifies specific propofol binding site(s). The results show that propofol, but not AziPm protects ß3-H267 from labeling by o-PD, whereas both propofol and o-PD protect against AziPm labeling of ß3-M286, ß3-M227, and α1I239. These data indicate that there are three distinct classes of propofol binding sites, with AziPm binding to two of the classes and o-PD to all three. Analysis of binding stoichiometry using native mass spectrometry in ß3 homomeric receptors, demonstrated a minimum of five AziPm labeled residues and three o-PD labeled residues per pentamer, suggesting that there are two distinct propofol binding sites per ß-subunit. The native mass spectrometry data, coupled with photolabeling performed in the presence of zinc, indicate that the binding site(s) identified by o-PD are adjacent to, but not within the channel pore, since the pore at the 17' H267 residue can accommodate only one propofol molecule. These data validate the existence of three classes of specific propofol binding sites on α1ß3 GABAA receptors.

8.
J Biol Chem ; 300(5): 107267, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38583863

RESUMEN

Phospholamban (PLB) is a transmembrane micropeptide that regulates the sarcoplasmic reticulum Ca2+-ATPase (SERCA) in cardiac muscle, but the physical mechanism of this regulation remains poorly understood. PLB reduces the Ca2+ sensitivity of active SERCA, increasing the Ca2+ concentration required for pump cycling. However, PLB does not decrease Ca2+ binding to SERCA when ATP is absent, suggesting PLB does not inhibit SERCA Ca2+ affinity. The prevailing explanation for these seemingly conflicting results is that PLB slows transitions in the SERCA enzymatic cycle associated with Ca2+ binding, altering transport Ca2+ dependence without actually affecting the equilibrium binding affinity of the Ca2+-coordinating sites. Here, we consider another hypothesis, that measurements of Ca2+ binding in the absence of ATP overlook important allosteric effects of nucleotide binding that increase SERCA Ca2+ binding affinity. We speculated that PLB inhibits SERCA by reversing this allostery. To test this, we used a fluorescent SERCA biosensor to quantify the Ca2+ affinity of non-cycling SERCA in the presence and absence of a non-hydrolyzable ATP-analog, AMPPCP. Nucleotide activation increased SERCA Ca2+ affinity, and this effect was reversed by co-expression of PLB. Interestingly, PLB had no effect on Ca2+ affinity in the absence of nucleotide. These results reconcile the previous conflicting observations from ATPase assays versus Ca2+ binding assays. Moreover, structural analysis of SERCA revealed a novel allosteric pathway connecting the ATP- and Ca2+-binding sites. We propose this pathway is disrupted by PLB binding. Thus, PLB reduces the equilibrium Ca2+ affinity of SERCA by interrupting allosteric activation of the pump by ATP.


Asunto(s)
Proteínas de Unión al Calcio , Calcio , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Animales , Humanos , Adenosina Trifosfato/metabolismo , Regulación Alostérica , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/química , Miocardio/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/química , Perros , Células HEK293 , Modelos Moleculares , Estructura Terciaria de Proteína
9.
J Biol Chem ; 300(8): 107591, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39032647

RESUMEN

Neuronal exocytosis requires the assembly of three SNARE proteins, syntaxin and SNAP25 on the plasma membrane and synaptobrevin on the vesicle membrane. However, the precise steps in this process and the points at which assembly and fusion are controlled by regulatory proteins are unclear. In the present work, we examine the kinetics and intermediate states during SNARE assembly in vitro using a combination of time resolved fluorescence and EPR spectroscopy. We show that syntaxin rapidly forms a dimer prior to forming the kinetically stable 2:1 syntaxin:SNAP25 complex and that the 2:1 complex is not diminished by the presence of excess SNAP25. Moreover, the 2:1 complex is temperature-dependent with a reduced concentration at 37 °C. The two segments of SNAP25 behave differently. The N-terminal SN1 segment of SNAP25 exhibits a pronounced increase in backbone ordering from the N- to the C-terminus that is not seen in the C-terminal SNAP25 segment SN2. Both the SN1 and SN2 segments of SNAP25 will assemble with syntaxin; however, while the association of the SN1 segment with syntaxin produces a stable 2:2 (SN1:syntaxin) complex, the complex formed between SN2 and syntaxin is largely disordered. Synaptobrevin fails to bind syntaxin alone but will associate with syntaxin in the presence of either the SN1 or SN2 segments; however, the synaptobrevin:syntaxin:SN2 complex remains disordered. Taken together, these data suggest that synaptobrevin and syntaxin do not assemble in the absence of SNAP25 and that the SN2 segment of SNAP25 is the last to enter the SNARE complex.


Asunto(s)
Neuronas , Proteínas Qa-SNARE , Proteína 25 Asociada a Sinaptosomas , Proteína 25 Asociada a Sinaptosomas/metabolismo , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/química , Neuronas/metabolismo , Animales , Proteínas Qa-SNARE/metabolismo , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/química , Cinética , Proteínas SNARE/metabolismo , Proteínas SNARE/genética , Ratas , Multimerización de Proteína
10.
J Biol Chem ; 300(6): 107410, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38796062

RESUMEN

Over the past decade, the connection between APOBEC3 cytosine deaminases and cancer mutagenesis has become increasingly apparent. This growing awareness has created a need for biochemical tools that can be used to identify and characterize potential inhibitors of this enzyme family. In response to this challenge, we have developed a Real-time APOBEC3-mediated DNA Deamination assay. This assay offers a single-step set-up and real-time fluorescent read-out, and it is capable of providing insights into enzyme kinetics. The assay also offers a high-sensitivity and easily scalable method for identifying APOBEC3 inhibitors. This assay serves as a crucial addition to the existing APOBEC3 biochemical and cellular toolkit and possesses the versatility to be readily adapted into a high-throughput format for inhibitor discovery.


Asunto(s)
Citidina Desaminasa , ADN , Humanos , Desaminación , Citidina Desaminasa/metabolismo , ADN/metabolismo , ADN/química , Cinética , Desaminasas APOBEC/metabolismo , Inhibidores Enzimáticos/farmacología
11.
RNA ; 29(9): 1411-1422, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37311599

RESUMEN

k-Junctions are elaborated forms of kink turns with an additional helix on the nonbulged strand, thus forming a three-way helical junction. Two were originally identified in the structures of Arabidopsis and Escherichia coli thiamine pyrophosphate (TPP) riboswitches, and another called DUF-3268 was tentatively identified from sequence information. In this work we show that the Arabidopsis and E. coli riboswitch k-junctions fold in response to the addition of magnesium or sodium ions, and that atomic mutations that should disrupt key hydrogen bonding interactions greatly impair folding. Using X-ray crystallography, we have determined the structure of the DUF-3268 RNA and thus confirmed that it is a k-junction. It also folds upon the addition of metal ions, though requiring a 40-fold lower concentration of either divalent or monovalent ions. The key difference between the DUF-3268 and riboswitch k-junctions is the lack of nucleotides inserted between G1b and A2b in the former. We show that this insertion is primarily responsible for the difference in folding properties. Finally, we show that the DUF-3268 can functionally substitute for the k-junction in the E. coli TPP riboswitch such that the chimera can bind the TPP ligand, although less avidly.


Asunto(s)
Arabidopsis , Riboswitch , Riboswitch/genética , Escherichia coli/metabolismo , Arabidopsis/genética , Pliegue del ARN , Tiamina Pirofosfato/genética , Tiamina Pirofosfato/metabolismo , Iones , Conformación de Ácido Nucleico
12.
Exp Cell Res ; 441(2): 114185, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39069150

RESUMEN

Dysfunction of the tumor suppressor p53 occurs in most human cancers, Hdm2 and HdmX play critical roles in p53 inactivation and degradation. Under unstressed conditions, HdmX binds to p53 like Hdm2, but HdmX cannot directly induce p53 degradation. Moreover, HdmX has been reported to stimulate Hdm2-mediated ubiquitination and degradation of p53. Here we reported that HdmX promoted the nuclear export of p53 independent of Hdm2 in living cells using FRET technology. Whereas, Hdm2 impeded HdmX-mediated nuclear export of p53 by sequestering it in nucleus. Interestingly, the C-terminal RING domain mutant Hdm2C464A formed heterooligomers with p53 in nucleus, which was inhibited by HdmX. The heterooligomers were located near PML-NBs. This study indicate that the nuclear Hdm2-HdmX interaction aborts the HdmX-mediated nuclear export of p53.


Asunto(s)
Transporte Activo de Núcleo Celular , Proteínas de Ciclo Celular , Núcleo Celular , Proteínas Proto-Oncogénicas c-mdm2 , Proteínas Proto-Oncogénicas , Proteína p53 Supresora de Tumor , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Humanos , Núcleo Celular/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Ubiquitinación , Unión Proteica
13.
J Mol Cell Cardiol ; 186: 125-137, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38008210

RESUMEN

N-terminal cardiac myosin-binding protein C (cMyBP-C) domains (C0-C2) bind to thick (myosin) and thin (actin) filaments to coordinate contraction and relaxation of the heart. These interactions are regulated by phosphorylation of the M-domain situated between domains C1 and C2. In cardiomyopathies and heart failure, phosphorylation of cMyBP-C is significantly altered. We aimed to investigate how cMyBP-C interacts with myosin and actin. We developed complementary, high-throughput, C0-C2 FRET-based binding assays for myosin and actin to characterize the effects due to 5 HCM-linked variants or functional mutations in unphosphorylated and phosphorylated C0-C2. The assays indicated that phosphorylation decreases binding to both myosin and actin, whereas the HCM mutations in M-domain generally increase binding. The effects of mutations were greatest in phosphorylated C0-C2, and some mutations had a larger effect on actin than myosin binding. Phosphorylation also altered the spatial relationship of the probes on C0-C2 and actin. The magnitude of these structural changes was dependent on C0-C2 probe location (C0, C1, or M-domain). We conclude that binding can differ between myosin and actin due to phosphorylation or mutations. Additionally, these variables can change the mode of binding, affecting which of the interactions in cMyBP-C N-terminal domains with myosin or actin take place. The opposite effects of phosphorylation and M-domain mutations is consistent with the idea that cMyBP-C phosphorylation is critical for normal cardiac function. The precision of these assays is indicative of their usefulness in high-throughput screening of drug libraries for targeting cMyBP-C as therapy.


Asunto(s)
Citoesqueleto de Actina , Actinas , Proteínas Portadoras , Actinas/metabolismo , Fosforilación , Citoesqueleto de Actina/metabolismo , Miosinas/genética , Miosinas/metabolismo , Mutación
14.
J Biol Chem ; 299(5): 104634, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36963492

RESUMEN

Upon activation by RAS, RAF family kinases initiate signaling through the MAP kinase cascade to control cell growth, proliferation, and differentiation. Among RAF isoforms (ARAF, BRAF, and CRAF), oncogenic mutations are by far most frequent in BRAF. The BRAFV600E mutation drives more than half of all malignant melanoma and is also found in many other cancers. Selective inhibitors of BRAFV600E (vemurafenib, dabrafenib, encorafenib) are used clinically for these indications, but they are not effective inhibitors in the context of oncogenic RAS, which drives dimerization and activation of RAF, nor for malignancies driven by aberrantly dimerized truncation/fusion variants of BRAF. By contrast, a number of "type II" RAF inhibitors have been developed as potent inhibitors of RAF dimers. Here, we compare potency of type II inhibitors tovorafenib (TAK-580) and naporafenib (LHX254) in biochemical assays against the three RAF isoforms and describe crystal structures of both compounds in complex with BRAF. We find that tovorafenib and naporafenib are most potent against CRAF but markedly less potent against ARAF. Crystal structures of both compounds with BRAFV600E or WT BRAF reveal the details of their molecular interactions, including the expected type II-binding mode, with full occupancy of both subunits of the BRAF dimer. Our findings have important clinical ramifications. Type II RAF inhibitors are generally regarded as pan-RAF inhibitors, but our studies of these two agents, together with recent work with type II inhibitors belvarafenib and naporafenib, indicate that relative sparing of ARAF may be a property of multiple drugs of this class.


Asunto(s)
Modelos Moleculares , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas B-raf , Humanos , Línea Celular Tumoral , Cristalografía por Rayos X , Sistema de Señalización de MAP Quinasas , Melanoma/tratamiento farmacológico , Estructura Molecular , Mutación , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/química , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo
15.
J Biol Chem ; 299(12): 105382, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37866628

RESUMEN

Proteomic studies have identified moesin (MSN), a protein containing a four-point-one, ezrin, radixin, moesin (FERM) domain, and the receptor CD44 as hub proteins found within a coexpression module strongly linked to Alzheimer's disease (AD) traits and microglia. These proteins are more abundant in Alzheimer's patient brains, and their levels are positively correlated with cognitive decline, amyloid plaque deposition, and neurofibrillary tangle burden. The MSN FERM domain interacts with the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) and the cytoplasmic tail of CD44. Inhibiting the MSN-CD44 interaction may help limit AD-associated neuronal damage. Here, we investigated the feasibility of developing inhibitors that target this protein-protein interaction. We have employed structural, mutational, and phage-display studies to examine how CD44 binds to the FERM domain of MSN. Interestingly, we have identified an allosteric site located close to the PIP2 binding pocket that influences CD44 binding. These findings suggest a mechanism in which PIP2 binding to the FERM domain stimulates CD44 binding through an allosteric effect, leading to the formation of a neighboring pocket capable of accommodating a receptor tail. Furthermore, high-throughput screening of a chemical library identified two compounds that disrupt the MSN-CD44 interaction. One compound series was further optimized for biochemical activity, specificity, and solubility. Our results suggest that the FERM domain holds potential as a drug development target. Small molecule preliminary leads generated from this study could serve as a foundation for additional medicinal chemistry efforts with the goal of controlling microglial activity in AD by modifying the MSN-CD44 interaction.


Asunto(s)
Enfermedad de Alzheimer , Unión Proteica , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Dominios FERM , Receptores de Hialuranos/metabolismo , Unión Proteica/efectos de los fármacos , Proteómica
16.
J Biol Chem ; 299(12): 105369, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37865311

RESUMEN

Cardiac MyBP-C (cMyBP-C) interacts with actin and myosin to fine-tune cardiac muscle contractility. Phosphorylation of cMyBP-C, which reduces the binding of cMyBP-C to actin and myosin, is often decreased in patients with heart failure (HF) and is cardioprotective in model systems of HF. Therefore, cMyBP-C is a potential target for HF drugs that mimic its phosphorylation and/or perturb its interactions with actin or myosin. We labeled actin with fluorescein-5-maleimide (FMAL) and the C0-C2 fragment of cMyBP-C (cC0-C2) with tetramethylrhodamine (TMR). We performed two complementary high-throughput screens (HTS) on an FDA-approved drug library, to discover small molecules that specifically bind to cMyBP-C and affect its interactions with actin or myosin, using fluorescence lifetime (FLT) detection. We first excited FMAL and detected its FLT, to measure changes in fluorescence resonance energy transfer (FRET) from FMAL (donor) to TMR (acceptor), indicating binding. Using the same samples, we then excited TMR directly, using a longer wavelength laser, to detect the effects of compounds on the environmentally sensitive FLT of TMR, to identify compounds that bind directly to cC0-C2. Secondary assays, performed on selected modulators with the most promising effects in the primary HTS assays, characterized the specificity of these compounds for phosphorylated versus unphosphorylated cC0-C2 and for cC0-C2 versus C1-C2 of fast skeletal muscle (fC1-C2). A subset of identified compounds modulated ATPase activity in cardiac and/or skeletal myofibrils. These assays establish the feasibility of the discovery of small-molecule modulators of the cMyBP-C-actin/myosin interaction, with the ultimate goal of developing therapies for HF.


Asunto(s)
Proteínas Portadoras , Descubrimiento de Drogas , Insuficiencia Cardíaca , Miofibrillas , Bibliotecas de Moléculas Pequeñas , Humanos , Actinas/metabolismo , Descubrimiento de Drogas/métodos , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Miosinas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Evaluación Preclínica de Medicamentos , Miofibrillas/efectos de los fármacos , Proteínas Portadoras/metabolismo , Técnicas Biosensibles , Adenosina Trifosfatasas/metabolismo , Músculo Esquelético/metabolismo , Proteínas Recombinantes/metabolismo , Activación Enzimática/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia
17.
Arch Microbiol ; 206(5): 237, 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38678508

RESUMEN

Invasive fungal infections (IFIs) are common and life-threatening complications in post-hematopoietic stem cell transplantation (post-HSCT) recipients, Severe IFIs can lead to systemic infection and organ damage, which results in high mortality in HSCT recipients. With the development of the field of fungal infection diagnosis, more and more advanced non-culture diagnostic tools have been developed, such as glip biosensors, metagenomic next-generation sequencing, Magnetic Nanoparticles and Identified Using SERS via AgNPs+ , and artificial intelligence-assisted diagnosis. The advanced diagnostic approaches contribute to the success of HSCT and improve the overall survival of post-HSCT leukemia patients by supporting therapeutical decisions. This review provides an overview of the characteristics of two high-incidence IFIs in post-HSCT recipients and discusses some of the recently developed IFI detection technologies. Additionally, it explores the potential application of cationic conjugated polymer fluorescence resonance energy transfer (CCP-FRET) technology for IFI detection. The aim is to offer insights into selecting appropriate IFI detection methods and gaining an understanding of novel fungal diagnostic approaches in laboratory settings.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Infecciones Fúngicas Invasoras , Humanos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Infecciones Fúngicas Invasoras/diagnóstico , Transferencia Resonante de Energía de Fluorescencia , Secuenciación de Nucleótidos de Alto Rendimiento , Técnicas Biosensibles/métodos
18.
J Fluoresc ; 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38647960

RESUMEN

A selective and sensitive fluorometric assay was developed for specific determination of curcumin (Cur) based on fluorescence resonance energy transfer (FRET) between molybdenum disulfide quantum dots (MoS2 QDs) and Cur. The MoS2 QDs were prepared via a one-step hydrothermal protocol using sodium molybdate dihydrate, L-cysteine (Cys) as precursors, and sodium cholate (SC) as a modification agent. The as-prepared MoS2 QDs possessed maximum fluorescence emission at 460 nm with a 20% of fluorescence quantum yield (FQY). It was found that the fluorescence of MoS2 QDs could be quantitatively quenched by Cur through FRET mechanism. Therefore, Cur could be detected in the range of 0.1-20 µg mL- 1 with a detection limit of 5 ng mL- 1. Additionally, the developed MoS2 QDs based fluorescent assay has been successfully applied for real food sample analysis with satisfactory results.

19.
Anal Bioanal Chem ; 416(4): 883-893, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38052994

RESUMEN

The developed method for simultaneous detection of aflatoxin B1 (AFB1) and aflD genes can effectively monitor from the source and reduce the safety problems and economic losses caused by the production of aflatoxin, which can be of great significance for food safety regulations. In this paper, we constructed a sensitive and convenient fluorescent biosensor to detect AFB1 and aflD genes simultaneously based on fluorescence resonance energy transfer (FRET) between quantum dots (QDs) and a black hole quenching agent. A stable "Y" shaped aptasensor was employed as the detection platform and a double quantum dot labeled DNA fragment was utilized to be the sensing element in this work. When the targets of AFB1 and aflD genes were presented in the solution, the aptamer in the "Y" shaped probe is specifically recognized by the target. At this time, both Si-carbon quantum dots (Si-CDs) and CdTe QDs are far away from the BHQ1 and BHQ3 to recover the fluorescence. The linear range of the prepared fluorescence simultaneous detection method was as wide as 0.5-500 ng·mL-1 with detection lines of 0.64 ng·mL-1 for AFB1 and 0.5-500 nM with detection lines of 0.75 nM for aflD genes (3σ/k). This fabricated fluorescent biosensor was further validated in real rice flour and corn flour samples, which also achieved good results. The recoveries were calculated by comparing the known and found amounts of AFB1 which ranged from 88.4 to approximately 115.32% in the rice flour samples and 90.7 ~ 102.58% in the corn flour samples. The recoveries of aflD genes ranged from 84.32 to approximately 109.3% in the rice flour samples and 89.48 ~ 100.99% in the corn flour samples. Therefore, the proposed biosensor can significantly improve food safety and quality control through a simple, fast, and sensitive agricultural product monitoring and detection system.


Asunto(s)
Aptámeros de Nucleótidos , Técnicas Biosensibles , Compuestos de Cadmio , Puntos Cuánticos , Aflatoxina B1/análisis , Aptámeros de Nucleótidos/genética , Telurio , Colorantes Fluorescentes , Técnicas Biosensibles/métodos , Límite de Detección
20.
Luminescence ; 39(3): e4716, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38497410

RESUMEN

A fluorescence resonance energy transfer (FRET) method was developed for double-stranded deoxyribonucleic acid (dsDNA) detection in living cells using the RecA-GFP (green fluorescent protein) fusion protein filament. In brief, the thiol-modified single-stranded DNA (ssDNA) was attached to gold nanoparticles (AuNPs); on the contrary, the prepared RecA-GFP fusion protein interacted with ssDNA. Due to the FRET between AuNPs and RecA-GFP, fluorescence of RecA-GFP fusion protein was quenched. In the presence of homologous dsDNA, homologous recombination occurred to release RecA-GFP fusion protein. Thus, the fluorescence of RecA-GFP was recovered. The dsDNA concentration was detected using fluorescence intensity of RecA-GFP. Under optimal conditions, this method could detect dsDNA activity as low as 0.015 optical density (OD) Escherichia coli cells, with a wide linear range from 0.05 to 0.9 OD cells, and the regression equation was ΔF = 342.7c + 78.9, with a linear relationship coefficient of 0.9920. Therefore, it provided a promising approach for the selective detection of dsDNA in living cells for early clinical diagnosis of genetic diseases.


Asunto(s)
ADN de Cadena Simple , Nanopartículas del Metal , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/genética , Oro/metabolismo , ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA