Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.041
Filtrar
1.
Inorg Chem ; 63(29): 13602-13612, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-38973094

RESUMEN

Metal nanozymes have offered attractive opportunities for biocatalysis and biomedicine. However, fabricating nanozymes simultaneously possessing highly catalytic selectivity and activity remains a great challenge due to the lack of three-dimensional (3D) architecture of the catalytic pocket in natural enzymes. Here, we integrate rhodium nanocluster (RhNC), reduced graphene oxide (rGO), and protamine (PRTM, a typical arginine-rich peptide) into a composite facilely based on the single peptide. Remarkably, the PRTM-RhNC@rGO composite displays outstanding selectivity, activity, and stability for the catalytic degradation of uric acid. The reaction rate constant of the uric acid oxidation catalyzed by the PRTM-RhNC@rGO composite is about 1.88 × 10-3 s-1 (4 µg/mL), which is 37.6 times higher than that of reported RhNP (k = 5 × 10-5 s-1, 20 µg/mL). Enzyme kinetic studies reveal that the PRTM-RhNC@rGO composite exhibits a similar affinity for uric acid as natural uricase. Furthermore, the uricase-like activity of PRTM-RhNC@rGO nanozymes remains in the presence of sulfur substances and halide ions, displaying incredibly well antipoisoning abilities. The analysis of the structure-function relationship indicates the PRTM-RhNC@rGO composite features the substrate binding site near the catalytic site in a confined space contributed by 2D rGO and PRTM, resulting in the high-performance of the composite nanozyme. Based on the outstanding uricase-like activity and the interaction of PRTM and uric acid, the PRTM-RhNC@rGO composite can retard the urate crystallization significantly. The present work provides new insights into the design of metal nanozymes with suitable binding sites near catalytic sites by mimicking pocket-like structures in natural enzymes based on simple peptides, conducing to broadening the practical application of high-performance nanozymes in biomedical fields.


Asunto(s)
Grafito , Rodio , Ácido Úrico , Grafito/química , Ácido Úrico/química , Ácido Úrico/metabolismo , Rodio/química , Urato Oxidasa/química , Urato Oxidasa/metabolismo , Péptidos/química , Péptidos/farmacología , Oxidación-Reducción , Arginina/química , Nanopartículas del Metal/química
2.
Mikrochim Acta ; 191(7): 432, 2024 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951266

RESUMEN

Cystinuria is a genetic disorder, and in severe cases, it might lead to kidney failure. As an important biomarker for cystinuria, the level of arginine (Arg) in urine is a vital indicator for cystinuria screening. Therefore, it is urgently needed to detect Arg with high selectivity and sensitivity. In this work, a boric acid functionalized Zr-based metal-organic framework UiO-PhbA is prepared by grafting phenylboronic acid on UiO-66-NH2 through a Schiff base reaction using a covalent post-synthesis modification (CPSM) strategy. The prepared UiO-PhbA exhibits a sensitive and specific fluorescence "turn-on" response to Arg and can be exploited to detect Arg in human serum and urine samples with a broad linear range of 0.6-350 µM and low limit of detection (LOD) of 18.45 nM. This study provides a new and reliable rapid screening protocol for sulfite oxidase deficiency-related diseases.


Asunto(s)
Arginina , Biomarcadores , Ácidos Borónicos , Cistinuria , Colorantes Fluorescentes , Límite de Detección , Estructuras Metalorgánicas , Humanos , Cistinuria/diagnóstico , Cistinuria/orina , Estructuras Metalorgánicas/química , Colorantes Fluorescentes/química , Arginina/química , Arginina/sangre , Biomarcadores/orina , Biomarcadores/sangre , Ácidos Borónicos/química , Espectrometría de Fluorescencia/métodos , Circonio/química
3.
ACS Appl Mater Interfaces ; 16(30): 38942-38955, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39039973

RESUMEN

Exogenous electrical stimulation has attracted considerable attention due to the advantages of microelectric induction and subsequent biological effects such as actin reorganization and reactive oxygen species (ROS) generation. Herein, an injectable hydrogel of BPR-ARG@Gel (BAG) with pyroelectric BPR nanoparticle loading and l-arginine (ARG) introduction was fabricated for advanced cancer therapy in vivo. Due to the photothermal effect, the holes and electrons in BPR nanoparticles were separated to produce an open-circuit voltage and consequently catalyze water H2O to generate toxic superoxide (•O2-) and hydroxyl radicals (•OH). These ROS substances further oxidize ARG to produce NO for synergistic tumor treatments. The mice experiments indicated that the employment of BAG hydrogel incorporation with a near-infrared laser downregulated the heat shock protein and recruited immune cells with 5-fold-enhanced expression of proinflammatory cytokines of interferon-γ. It was also noteworthy that the injectable hydrogel of BAG substantially induced the generation of reactive oxygen/nitrogen species (ROS/RNS) with reliable biosafety and strong tumor inhibition. Overall, these findings have provided potentially new inspirations and a feasible strategy to translate this multifunctional hydrogel toward tumor therapy in a pyroelectric stimulation manner.


Asunto(s)
Hidrogeles , Especies Reactivas de Oxígeno , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Hidrogeles/química , Hidrogeles/farmacología , Catálisis , Rayos Infrarrojos , Humanos , Arginina/química , Especies de Nitrógeno Reactivo/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias/patología , Línea Celular Tumoral , Nanopartículas/química , Ratones Endogámicos BALB C , Antineoplásicos/química , Antineoplásicos/farmacología
4.
Cell Rep ; 43(7): 114459, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38985674

RESUMEN

Glycine- and arginine-rich (GAR) motifs, commonly found in RNA-binding and -processing proteins, can be symmetrically (SDMA) or asymmetrically (ADMA) dimethylated at the arginine residue by protein arginine methyltransferases. Arginine-methylated protein motifs are usually read by Tudor domain-containing proteins. Here, using a GFP-Trap, we identify a non-Tudor domain protein, squamous cell carcinoma antigen recognized by T cells 3 (SART3), as a reader for SDMA-marked GAR motifs. Structural analysis and mutagenesis of SART3 show that aromatic residues lining a groove between two adjacent aromatic-rich half-a-tetratricopeptide (HAT) repeat domains are essential for SART3 to recognize and bind to SDMA-marked GAR motif peptides, as well as for the interaction between SART3 and the GAR-motif-containing proteins fibrillarin and coilin. Further, we show that the loss of this reader ability affects RNA splicing. Overall, our findings broaden the range of potential SDMA readers to include HAT domains.


Asunto(s)
Secuencias de Aminoácidos , Arginina , Glicina , Arginina/metabolismo , Arginina/química , Humanos , Glicina/metabolismo , Glicina/química , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Unión Proteica , Empalme del ARN , Células HEK293 , Metilación , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/química , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/química
5.
Anal Chem ; 96(24): 9885-9893, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38848670

RESUMEN

Glutathione (GSH) redox control and arginine metabolism are critical in regulating the physiological response to injury and oxidative stress. Quantification assessment of the GSH/arginine redox metabolism supports monitoring metabolic pathway shifts during pathological processes and their linkages to redox regulation. However, assessing the redox status of organisms with complex matrices is challenging, and single redox molecule analysis may not be accurate for interrogating the redox status in cells and in vivo. Herein, guided by a paired derivatization strategy, we present a new ultraperformance liquid chromatography tandem mass spectrometry (UPLC-MS/MS)-based approach for the functional assessment of biological redox status. Two structurally analogous probes, 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate (AQC) and newly synthesized 2-methyl-6-aminoquinolyl-N-hydroxysuccinimidyl carbamate (MeAQC), were set for paired derivatization. The developed approach was successfully applied to LPS-stimulated RAW 264.7 cells and HDM-induced asthma mice to obtain quantitative information on GSH/arginine redox metabolism. The results suggest that the redox status was remarkably altered upon LPS and HDM stimulation. We expect that this approach will be of good use in a clinical biomarker assay and potential drug screening associated with redox metabolism, oxidative damage, and redox signaling.


Asunto(s)
Arginina , Glutatión , Oxidación-Reducción , Espectrometría de Masas en Tándem , Animales , Arginina/metabolismo , Arginina/análisis , Arginina/química , Glutatión/metabolismo , Glutatión/análisis , Ratones , Espectrometría de Masas en Tándem/métodos , Células RAW 264.7 , Carbamatos/metabolismo , Carbamatos/química , Cromatografía Líquida de Alta Presión , Lipopolisacáridos/farmacología , Aminoquinolinas/química
6.
Biomacromolecules ; 25(7): 4168-4176, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38902961

RESUMEN

We have successfully created self-assembled membranes by combining positively charged (Pro-X-(Phe-X)5-Pro) PFX peptides with negatively charged alginate. These PFX/alginate membranes were formed by three different peptides that contain either X = Arginine (R), Histidine (H), or Ornithine (O) as their charged amino acid. The assemblies were compared to membranes that were previously reported by us composed of X = lysine (K). This study enabled us to elucidate the impact of amino acids' specific interactions on membrane formation. SEM, SAXS, and cryo-TEM measurements show that although K, R, H, and O may have a similar net charge, the specific traits of the charged amino acid is an essential factor in determining the hierarchical structure of alginate/PFX self-assembled membranes.


Asunto(s)
Alginatos , Alginatos/química , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Péptidos/química , Cationes/química , Membranas Artificiales , Arginina/química
7.
Anal Chem ; 96(27): 10943-10952, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38918973

RESUMEN

Both controllable regulation of the conformational structure of a polypeptide and specific recognition of an amino acid are still arduous challenges. Here, a novel dual-mode (electrochemical and colorimetric) biosensor was built for arginine (Arg) recognition based on a conformation switch, utilizing controllable and synergistic self-assembly of a ferrocene-grafted hexadecapeptide (P16Fc) with gold nanoparticles (AuNPs). Benefiting from the flexibility and unique topological structure of P16Fc formed nanospheres, the assembly and disassembly can undergo a conformation transition induced by Arg through controlling the distance and number of Fc detached from the gold surface, producing on-off electrical signals. Also, they can induce aggregation and dispersion of AuNPs in solution, causing a color change. The mechanism of Arg recognition with polypeptide conformation regulation was well explored by combining microstructure characterizations with molecular mechanics calculations. The electrochemical and colorimetric assays for Arg were successfully established in sensitive and selective manner, not only obtaining a very low detection limit, but also effectively eliminating the interference from other amino acids and overcoming the limitation of AuNP aggregation. Notably, the conformational change-based assay with the peptide regulated by the target will make a powerful tool for the amino acid biosensing and health diagnosis.


Asunto(s)
Arginina , Técnicas Electroquímicas , Compuestos Ferrosos , Oro , Nanopartículas del Metal , Metalocenos , Péptidos , Arginina/química , Compuestos Ferrosos/química , Metalocenos/química , Oro/química , Nanopartículas del Metal/química , Péptidos/química , Técnicas Biosensibles/métodos , Colorimetría/métodos , Conformación Proteica , Límite de Detección
8.
Methods ; 229: 94-107, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38834165

RESUMEN

In this report, non-isomerisable analogs of arginine tRNA (Arg-triazole-tRNA) have been synthesized as tools to study tRNA-dependent aminoacyl-transferases. The synthesis involves the incorporation of 1,4 substituted-1,2,3 triazole ring to mimic the ester bond that connects the amino acid to the terminal adenosine in the natural substrate. The synthetic procedure includes (i) a coupling between 2'- or 3'-azido-adenosine derivatives and a cytidine phosphoramidite to access dinucleotide molecules, (ii) Cu-catalyzed cycloaddition reactions between 2'- or 3'-azido dinucleotide in the presence of an alkyne molecule mimicking the arginine, providing the corresponding Arg-triazole-dinucleotides, (iii) enzymatic phosphorylation of the 5'-end extremity of the Arg-triazole-dinucleotides with a polynucleotide kinase, and (iv) enzymatic ligation of the 5'-phosphorylated dinucleotides with a 23-nt RNA micro helix that mimics the acceptor arm of arg-tRNA or with a full tRNAarg. Characterization of nucleoside and nucleotide compounds involved MS spectrometry, 1H, 13C and 31P NMR analysis. This strategy allows to obtain the pair of the two stable regioisomers of arg-tRNA analogs (2' and 3') which are instrumental to explore the regiospecificity of arginyl transferases enzyme. In our study, a first binding assay of the arg-tRNA micro helix with the Arginyl-tRNA-protein transferase 1 (ATE1) was performed by gel shift assays.


Asunto(s)
Cobre , Reacción de Cicloadición , Catálisis , Cobre/química , Reacción de Cicloadición/métodos , Arginina/química , Arginina/análogos & derivados , ARN de Transferencia de Arginina/química , ARN de Transferencia de Arginina/genética , ARN de Transferencia de Arginina/metabolismo , Fosforilación , Triazoles/química , Triazoles/síntesis química , Estereoisomerismo , Adenosina/análogos & derivados , Adenosina/química , Aminoaciltransferasas/metabolismo , Aminoaciltransferasas/química , Aminoaciltransferasas/genética
9.
Arch Biochem Biophys ; 757: 110040, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38750922

RESUMEN

Purine salvage enzymes have been of significant interest in anti-Leishmanial drug development due to the parasite's critical dependence on this pathway for the supply of nucleotides in the absence of a de novo purine synthesis pathway. Adenylosuccinate lyase (ADSL) one of the key enzymes in this pathway is a homo-tetramer, where the active site is formed by residues from three distinct subunits. Analysis of the subunit interfaces of LdADSL, revealed a conserved Arg40 forming critical inter-subunit interactions and also involved in substrate binding. We hypothesized that mutating this residue can affect both the structural stability and activity of the enzyme. In our study, we used biochemical, biophysical, and computational simulation approaches to understand the structural and functional role of Arg40 in LdADSL. We have replaced Arg40 with an Ala and Glu using site directed mutagenesis. The mutant enzymes were similar to wild-type enzyme in secondary structure and subunit association. Thermal shift assays indicated that the mutations affected the protein stability. Both mutants showed decreased specific activities in both forward and reverse directions with significantly weakened affinities towards succinyl-adenosine monophosphate (SAMP). The mutations resulted in changes in C3 loop conformation and D3 domain rotation. Consequently, the orientation of the active site amino acid residues changed resulting in compromised activity and stability. Studies so far have majorly focused on the ADSL active site for designing drugs against it. Our work indicates that an alternative inhibitory mechanism for the enzyme can be designed by targeting the inter-subunit interface.


Asunto(s)
Adenilosuccinato Liasa , Arginina , Estabilidad de Enzimas , Leishmania donovani , Adenilosuccinato Liasa/genética , Adenilosuccinato Liasa/química , Adenilosuccinato Liasa/metabolismo , Leishmania donovani/enzimología , Leishmania donovani/genética , Arginina/metabolismo , Arginina/química , Purinas/metabolismo , Purinas/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Mutagénesis Sitio-Dirigida , Dominio Catalítico , Simulación de Dinámica Molecular
10.
Acta Biomater ; 182: 245-259, 2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38729545

RESUMEN

Diabetic wound healing is a great clinical challenge due to the microenvironment of hyperglycemia and high pH value, bacterial infection and persistent inflammation. Here, we develop a cascade nanoreactor hydrogel (Arg@Zn-MOF-GOx Gel, AZG-Gel) with arginine (Arg) loaded Zinc metal organic framework (Zn-MOF) and glucose oxidase (GOx) based on chondroitin sulfate (CS) and Pluronic (F127) to accelerate diabetic infected wound healing. GOx in AZG-Gel was triggered by hyperglycemic environment to reduce local glucose and pH, and simultaneously produced hydrogen peroxide (H2O2) to enable Arg-to release nitric oxide (NO) for inflammation regulation, providing a suitable microenvironment for wound healing. Zinc ions (Zn2+) released from acid-responsive Zn-MOF significantly inhibited the proliferation and biofilm formation of S.aureus and E.coli. AZG-Gel significantly accelerated diabetic infected wound healing by down-regulating pro-inflammatory tumor necrosis factor (TNF)-α and interleukin (IL)-6, up-regulating anti-inflammatory factor IL-4, promoting angiogenesis and collagen deposition in vivo. Collectively, our nanoreactor cascade strategy combining "endogenous improvement (reducing glucose and pH)" with "exogenous resistance (anti-bacterial and anti-inflammatory)" provides a new idea for promoting diabetic infected wound healing by addressing both symptoms and root causes. STATEMENT OF SIGNIFICANCE: A cascade nanoreactor (AZG-Gel) is constructed to solve three key problems in diabetic wound healing, namely, hyperglycemia and high pH microenvironment, bacterial infection and persistent inflammation. Local glucose and pH levels are reduced by GOx to provide a suitable microenvironment for wound healing. The release of Zn2+ significantly inhibits bacterial proliferation and biofilm formation, and NO reduces wound inflammation and promotes angiogenesis. The pH change when AZG-Gel is applied to wounds is expected to enable the visualization of wound healing to guide the treatment of diabetic wound. Our strategy of "endogenous improvement (reducing glucose and pH)" combined with "exogenous resistance (anti-bacterial and anti-inflammatory)" provides a new way for promoting diabetic wound healing.


Asunto(s)
Glucosa Oxidasa , Estructuras Metalorgánicas , Óxido Nítrico , Cicatrización de Heridas , Zinc , Cicatrización de Heridas/efectos de los fármacos , Animales , Zinc/química , Zinc/farmacología , Óxido Nítrico/metabolismo , Estructuras Metalorgánicas/farmacología , Estructuras Metalorgánicas/química , Glucosa Oxidasa/farmacología , Glucosa Oxidasa/metabolismo , Diabetes Mellitus Experimental/patología , Microambiente Celular/efectos de los fármacos , Ratones , Hidrogeles/química , Hidrogeles/farmacología , Masculino , Staphylococcus aureus/efectos de los fármacos , Biopelículas/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/química , Arginina/farmacología , Arginina/química
11.
ACS Biomater Sci Eng ; 10(6): 3825-3832, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38722049

RESUMEN

In recent years, a novel treatment method for cancer has emerged, which is based on the starvation of tumors of amino acids like arginine. The deprivation of arginine in serum is based on enzymatic degradation and can be realized by arginine deaminases like the l-amino acid oxidase found in the ink toxin of the sea hare Aplysia punctata. Previously isolated from the ink, the l-amino acid oxidase was described to oxidate the essential amino acids l-lysine and l-arginine to their corresponding deaminated alpha-keto acids. Here, we present the recombinant production and functionalization of the amino acid oxidase Aplysia punctata ink toxin (APIT). PEGylated APIT (APIT-PEG) increased the blood circulation time. APIT-PEG treatment of patient-derived xenografted mice shows a significant dose-dependent reduction of tumor growth over time mediated by amino acid starvation of the tumor. Treatment of mice with APIT-PEG, which led to deprivation of arginine, was well tolerated.


Asunto(s)
Aplysia , Arginina , Lisina , Polietilenglicoles , Animales , Arginina/farmacología , Arginina/química , Lisina/farmacología , Lisina/química , Polietilenglicoles/química , Polietilenglicoles/farmacología , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto , Toxinas Marinas/farmacología , Toxinas Marinas/uso terapéutico , Toxinas Marinas/química , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , L-Aminoácido Oxidasa/farmacología , L-Aminoácido Oxidasa/metabolismo , L-Aminoácido Oxidasa/química , Femenino , Línea Celular Tumoral
12.
Sci Rep ; 14(1): 11715, 2024 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778164

RESUMEN

Recent studies have revealed that arginine is the most favorable target of amino acid alteration in most cancer types and it has been suggested that the high preference for arginine mutations reflects the critical roles of this amino acid in the function of proteins. High rates of mutations of arginine residues in cancer, however, might also be due to increased mutability of arginine codons of the CGN family as the CpG dinucleotides of these codons may be methylated. In the present work we have analyzed spectra of single base substitutions of cancer genes (oncogenes, tumor suppressor genes) and passenger genes in cancer tissues to assess the contributions of CpG hypermutability and selection to arginine mutations. Our studies have shown that arginines encoded by the CGN codon family display higher rates of mutation in both cancer genes and passenger genes than arginine codons AGA and AGG that are devoid of CpG dinucleotide, suggesting that the predominance of arginine mutations in cancer is primarily due to CpG hypermutability, rather than selection for arginine replacement. Nevertheless, our results also suggest that CGN codons for arginines may serve as Achilles' heels of cancer genes. CpG hypermutability of key arginines of proto-oncogenes, leading to high rates of recurrence of driver mutations, contributes significantly to carcinogenesis. Similarly, our results indicate that hypermutability of the CpG dinucleotide of CGA codons (converting them to TGA stop codons) contributes significantly to recurrent truncation and inactivation of tumor suppressor genes.


Asunto(s)
Arginina , Codón , Islas de CpG , Neoplasias , Arginina/genética , Arginina/química , Humanos , Codón/genética , Neoplasias/genética , Islas de CpG/genética , Mutación , Oncogenes/genética , Genes Supresores de Tumor
13.
FEBS Lett ; 598(9): 1061-1079, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38649155

RESUMEN

The molecular mechanisms of selective RNA loading into exosomes and other extracellular vesicles are not yet completely understood. In order to show that a pool of RNA sequences binds both the amino acid arginine and lipid membranes, we constructed a bifunctional RNA 10Arg aptamer specific for arginine and lipid vesicles. The preference of RNA 10Arg for lipid rafts was visualized and confirmed using FRET microscopy in neuroblastoma cells. The selection-amplification (SELEX) method using a doped (with the other three nucleotides) pool of RNA 10Arg sequences yielded several RNA 10Arg(D) sequences, and the affinities of these RNAs both to arginine and liposomes are improved in comparison to pre-doped RNA. Generation of these bispecific aptamers supports the hypothesis that an RNA molecule can bind both to RNA-binding proteins (RBPs) through arginine within the RBP-binding site and to membrane lipid rafts, thus facilitating RNA loading into exosomes and other extracellular vesicles.


Asunto(s)
Arginina , Liposomas , Arginina/química , Arginina/metabolismo , Humanos , Liposomas/química , Liposomas/metabolismo , Microdominios de Membrana/metabolismo , Microdominios de Membrana/química , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/metabolismo , Aptámeros de Nucleótidos/genética , Línea Celular Tumoral , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Secuencia de Bases , ARN/metabolismo , ARN/química , ARN/genética , Exosomas/metabolismo , Exosomas/genética , Exosomas/química , Transferencia Resonante de Energía de Fluorescencia
14.
Food Chem ; 450: 139392, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-38640546

RESUMEN

The combinational effects of kojic acid and lauroyl arginine ethyl ester hydrochloride (ELAH) on fresh-cut potatoes were investigated. Kojic acid of 0.6% (w/w) effectively inhibited the browning of fresh-cut potatoes and displayed antimicrobial capacity. The color difference value of samples was decreased from 175 to 26 by kojic acid. In contrast, ELAH could not effectively bind with the active sites of tyrosinase and catechol oxidase at molecular level. Although 0.5% (w/w) of ELAH prominently inhibited the microbial growth, it promoted the browning of samples. However, combining kojic acid and ELAH effectively inhibited the browning of samples and microbial growth during the storage and the color difference value of samples was decreased to 52. This amount of kojic acid inhibited enzyme activities toward phenolic compounds. The results indicated that combination of kojic acid and ELAH could provide a potential strategy to extend the shelf life of fresh-cut products.


Asunto(s)
Arginina , Monofenol Monooxigenasa , Pironas , Solanum tuberosum , Pironas/farmacología , Pironas/química , Arginina/química , Arginina/análogos & derivados , Arginina/farmacología , Solanum tuberosum/química , Solanum tuberosum/crecimiento & desarrollo , Monofenol Monooxigenasa/metabolismo , Conservación de Alimentos/métodos , Catecol Oxidasa/metabolismo , Conservantes de Alimentos/farmacología , Conservantes de Alimentos/química , Bacterias/efectos de los fármacos , Bacterias/genética
15.
J Clin Lab Anal ; 38(7): e25030, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38525916

RESUMEN

BACKGROUND: The motor protein dynein is integral to retrograde transport along microtubules and interacts with numerous cargoes through the recruitment of cargo-specific adaptor proteins. This interaction is mediated by dynein light intermediate chain subunits LIC1 (DYNC1LI1) and LIC2 (DYNC1LI2), which govern the adaptor binding and are present in distinct dynein complexes with overlapping and unique functions. METHODS: Using bioinformatics, we analyzed the C-terminal domains (CTDs) of LIC1 and LIC2, revealing similar structural features but diverse post-translational modifications (PTMs). The methylation status of LIC2 and the proteins involved in this modification were examined through immunoprecipitation and immunoblotting analyses. The specific methylation sites on LIC2 were identified through a site-directed mutagenesis analysis, contributing to a deeper understanding of the regulatory mechanisms of the dynein complex. RESULTS: We found that LIC2 is specifically methylated at the arginine 397 residue, a reaction that is catalyzed by protein arginine methyltransferase 1 (PRMT1). CONCLUSIONS: The distinct PTMs of the LIC subunits offer a versatile mechanism for dynein to transport diverse cargoes efficiently. Understanding how these PTMs influence the functions of LIC2, and how they differ from LIC1, is crucial for elucidating the role of dynein-related transport pathways in a range of diseases. The discovery of the arginine 397 methylation site on LIC2 enhances our insight into the regulatory PTMs of dynein functions.


Asunto(s)
Arginina , Dineínas Citoplasmáticas , Proteína-Arginina N-Metiltransferasas , Proteínas Represoras , Metilación , Arginina/metabolismo , Arginina/química , Humanos , Dineínas Citoplasmáticas/metabolismo , Dineínas Citoplasmáticas/genética , Dineínas Citoplasmáticas/química , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Procesamiento Proteico-Postraduccional , Dineínas/metabolismo , Dineínas/genética , Dineínas/química , Secuencia de Aminoácidos
16.
J Phys Chem B ; 128(10): 2347-2359, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38416758

RESUMEN

Liquid-liquid phase separation mediated by proteins and/or nucleic acids is believed to underlie the formation of many distinct condensed phases, or membraneless organelles, within living cells. These condensates have been proposed to orchestrate a variety of important processes. Despite recent advances, the interactions that regulate the dynamics of molecules within a condensate remain poorly understood. We performed accumulated 564.7 µs all-atom molecular dynamics (MD) simulations (system size ∼200k atoms) of model condensates formed by a scaffold RNA oligomer and a scaffold peptide rich in arginine (Arg). These model condensates contained one of three possible guest peptides: the scaffold peptide itself or a variant in which six Arg residues were replaced by lysine (Lys) or asymmetric dimethyl arginine (ADMA). We found that the Arg-rich peptide can form the largest number of hydrogen bonds and bind the strongest to the scaffold RNA in the condensate, relative to the Lys- and ADMA-rich peptides. Our MD simulations also showed that the Arg-rich peptide diffused more slowly in the condensate relative to the other two guest peptides, which is consistent with a recent fluorescence microscopy study. There was no significant increase in the number of cation-π interactions between the Arg-rich peptide and the scaffold RNA compared to the Lys-rich and ADMA-rich peptides. Our results indicate that hydrogen bonds between the peptides and the RNA backbone, rather than cation-π interactions, play a major role in regulating peptide diffusion in the condensate.


Asunto(s)
Simulación de Dinámica Molecular , ARN , Enlace de Hidrógeno , Péptidos/química , Proteínas , Arginina/química , Lisina/química , Cationes
17.
Adv Sci (Weinh) ; 11(16): e2308493, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38380492

RESUMEN

Supramolecular chirality-mediated selective interaction among native assemblies is essential for precise disease diagnosis and treatment. Herein, to fully understand the supramolecular chiral binding affinity-achieved therapeutic efficiency, supramolecular chiral nanoparticles (WP5⊃D/L-Arg+DOX+ICG) with the chirality transfer from chiral arginine (D/L-Arg) to water-soluble pillar[5]arene (WP5) are developed through non-covalent interactions, in which an anticancer drug (DOX, doxorubicin hydrochloride) and a photothermal agent (ICG, indocyanine green) are successfully loaded. Interestingly, the WP5⊃D-Arg nanoparticles show 107 folds stronger binding capability toward phospholipid-composed liposomes compared with WP5⊃L-Arg. The enantioselective interaction further triggers the supramolecular chirality-specific drug accumulation in cancer cells. As a consequence, WP5⊃D-Arg+DOX+ICG exhibits extremely enhanced chemo-photothermal synergistic therapeutic efficacy (tumor inhibition rate of 99.4%) than that of WP5⊃L-Arg+DOX+ICG (tumor inhibition rate of 56.4%) under the same condition. This work reveals the breakthrough that supramolecular chiral assemblies can induce surprisingly large difference in cancer therapy, providing strong support for the significance of supramolecular chirality in bio-application.


Asunto(s)
Antineoplásicos , Doxorrubicina , Verde de Indocianina , Nanopartículas , Doxorrubicina/farmacología , Doxorrubicina/química , Animales , Ratones , Antineoplásicos/farmacología , Antineoplásicos/química , Verde de Indocianina/química , Nanopartículas/química , Humanos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Arginina/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/terapia , Compuestos de Amonio Cuaternario/química , Calixarenos/química , Estereoisomerismo
18.
Food Chem ; 446: 138809, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38402768

RESUMEN

This study investigated the individual and combined effects of l-arginine, l-lysine, and NaCl on the ultrastructure of porcine myofibrils to uncover the mechanism underlying meat tenderization. Arg or Lys alone shortened A-bands and damaged M-lines, while NaCl alone destroyed M- and Z-lines. Overall, Arg and Lys cooperated with NaCl to destroy the myofibrillar ultrastructure. Moreover, these two amino acids conjoined with NaCl to increase myosin solubility, actin band intensity, and the protein concentration of the actomyosin supernatant. However, they decreased the turbidity and particle size of both myosin and actomyosin solutions, and the remaining activities of Ca2+- and Mg2+-ATPase. The current results revealed that Arg/Lys combined with NaCl to extract myosin and dissociate actomyosin, thereby aggravating the destruction of the myofibrillar ultrastructure. The present results provide a good explanation for the previous phenomenon that Arg and Lys cooperated with NaCl to improve meat tenderness.


Asunto(s)
Actomiosina , Lisina , Animales , Porcinos , Actomiosina/química , Lisina/química , Cloruro de Sodio/química , Miosinas/química , Carne/análisis , Actinas/metabolismo , Arginina/química , Suplementos Dietéticos
19.
ACS Appl Mater Interfaces ; 16(9): 11159-11171, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38385360

RESUMEN

For the improved delivery of cancer therapeutics and imaging agents, the conjugation of cell-penetrating peptides (CPPs) increases the cellular uptake and water solubility of agents. Among the various CPPs, arginine-rich peptides have been the most widely used. Combining CPPs with enzyme-responsive peptides presents an innovative strategy to target specific intracellular enzymes in cancer cells and when combined with the appropriate click chemistry can enhance theranostic drug delivery through the formation of intracellular self-assembled nanostructures. However, one drawback of CPPs is their high positive charge which can cause nonspecific binding, leading to off-target accumulation and potential toxicity. Hence, balancing cell-specific penetration, toxicity, and biocompatibility is essential for future clinical efficacy. We synthesized six cancer-specific, legumain-responsive RnAANCK peptides containing one to six arginine residues, with legumain being an asparaginyl endopeptidase that is overexpressed in aggressive prostate tumors. When conjugated to Alexa Fluor 488, R1-R6AANCK peptides exhibited a concentration- and time-dependent cell penetration in prostate cancer cells, which was higher for peptides with higher R values, reaching a plateau after approximately 120 min. Highly aggressive DU145 prostate tumor cells, but not less aggressive LNCaP cells, self-assembled nanoparticles in the cytosol after the cleavage of the legumain-specific peptide. The in vivo biocompatibility was assessed in mice after the intravenous injection of R1-R6AANCK peptides, with concentrations ranging from 0.0125 to 0.4 mmol/kg. The higher arginine content in R4-6 peptides showed blood and urine indicators for the impairment of bone marrow, liver, and kidney function in a dose-dependent manner, with instant hemolysis and morbidity in extreme cases. These findings underscore the importance of designing peptides with the optimal arginine residue length for a proper balance of cell-specific penetration, toxicity, and in vivo biocompatibility.


Asunto(s)
Péptidos de Penetración Celular , Neoplasias , Animales , Ratones , Arginina/química , Péptidos de Penetración Celular/química , Neoplasias/tratamiento farmacológico
20.
J Photochem Photobiol B ; 252: 112867, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38368636

RESUMEN

Arginine methylation (ArgMet), as a post-translational modification, plays crucial roles in RNA processing, transcriptional regulation, signal transduction, DNA repair, apoptosis and liquid-liquid phase separation (LLPS). Since arginine methylation is associated with cancer pathogenesis and progression, protein arginine methyltransferases have gained interest as targets for anti-cancer therapy. Despite considerable process made to elucidate (patho)physiological mechanisms regulated by arginine methylation, there remains a lack of tools to visualize arginine methylation with high spatiotemporal resolution in live cells. To address this unmet need, we generated an ArgMet-sensitive genetically encoded, Förster resonance energy transfer-(FRET) based biosensor, called GEMS, capable of quantitative real-time monitoring of ArgMet dynamics. We optimized these biosensors by using different ArgMet-binding domains, arginine-glycine-rich regions and adjusting the linkers within the biosensors to improve their performance. Using a set of mammalian cell lines and modulators, we demonstrated the applicability of GEMS for monitoring changes in arginine methylation with single-cell and temporal resolution. The GEMS can facilitate the in vitro screening to find potential protein arginine methyltransferase inhibitors and will contribute to a better understanding of the regulation of ArgMet related to differentiation, development and disease.


Asunto(s)
Arginina , Transferencia Resonante de Energía de Fluorescencia , Animales , Arginina/química , Metilación , Regulación de la Expresión Génica , Colorantes , Procesamiento Proteico-Postraduccional , Mamíferos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA