Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.774
Filtrar
1.
Sci Rep ; 14(1): 11444, 2024 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769383

RESUMEN

Neonatal sepsis is a major cause of childhood mortality. Limited diagnostic tools and mechanistic insights have hampered our abilities to develop prophylactic or therapeutic interventions. Biomarkers in human neonatal sepsis have been repeatedly identified as associated with dysregulation of angiopoietin signaling and altered arachidonic acid metabolism. We here provide the mechanistic evidence in support of the relevance for these observations. Angiopoetin-1 (Ang-1), which promotes vascular integrity, was decreased in blood plasma of human and murine septic newborns. In preclinical models, administration of Ang-1 provided prophylactic protection from septic death. Arachidonic acid metabolism appears to be functionally connected to Ang-1 via reactive oxygen species (ROS) with a direct role of nitric oxide (NO). Strengthening this intersection via oral administration of arachidonic acid and/or the NO donor L-arginine provided prophylactic as well as therapeutic protection from septic death while also increasing plasma Ang-1 levels among septic newborns. Our data highlight that targeting angiogenesis-associated pathways with interventions that increase Ang-1 activity directly or indirectly through ROS/eNOS provide promising avenues to prevent and/or treat severe neonatal sepsis.


Asunto(s)
Angiopoyetina 1 , Sepsis Neonatal , Óxido Nítrico , Especies Reactivas de Oxígeno , Humanos , Animales , Recién Nacido , Angiopoyetina 1/sangre , Angiopoyetina 1/metabolismo , Ratones , Especies Reactivas de Oxígeno/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico/sangre , Ácido Araquidónico/metabolismo , Ácido Araquidónico/sangre , Femenino , Masculino , Arginina/sangre , Arginina/metabolismo , Transducción de Señal , Óxido Nítrico Sintasa de Tipo III/metabolismo , Neovascularización Patológica/metabolismo , Biomarcadores/sangre , Modelos Animales de Enfermedad , Animales Recién Nacidos , Angiogénesis
2.
FASEB J ; 38(10): e23647, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38787599

RESUMEN

Arginine methylation is a protein posttranslational modification important for the development of skeletal muscle mass and function. Despite this, our understanding of the regulation of arginine methylation under settings of health and disease remains largely undefined. Here, we investigated the regulation of arginine methylation in skeletal muscles in response to exercise and hypertrophic growth, and in diseases involving metabolic dysfunction and atrophy. We report a limited regulation of arginine methylation under physiological settings that promote muscle health, such as during growth and acute exercise, nor in disease models of insulin resistance. In contrast, we saw a significant remodeling of asymmetric dimethylation in models of atrophy characterized by the loss of innervation, including in muscle biopsies from patients with myotrophic lateral sclerosis (ALS). Mass spectrometry-based quantification of the proteome and asymmetric arginine dimethylome of skeletal muscle from individuals with ALS revealed the largest compendium of protein changes with the identification of 793 regulated proteins, and novel site-specific changes in asymmetric dimethyl arginine (aDMA) of key sarcomeric and cytoskeletal proteins. Finally, we show that in vivo overexpression of PRMT1 and aDMA resulted in increased fatigue resistance and functional recovery in mice. Our study provides evidence for asymmetric dimethylation as a regulator of muscle pathophysiology and presents a valuable proteomics resource and rationale for numerous methylated and nonmethylated proteins, including PRMT1, to be pursued for therapeutic development in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Arginina , Músculo Esquelético , Proteína-Arginina N-Metiltransferasas , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Arginina/metabolismo , Arginina/análogos & derivados , Humanos , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Ratones , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Masculino , Metilación , Femenino , Procesamiento Proteico-Postraduccional , Ratones Endogámicos C57BL , Proteoma/metabolismo
3.
Mol Cell ; 84(10): 1819-1821, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38759621

RESUMEN

In this issue of Molecular Cell, Yang et al.1 find that arginine-to-cysteine substitutants are enriched in a subset of lung cancer proteomes, potentiated by arginine deprivation, and promote resistance to chemotherapy.


Asunto(s)
Arginina , Cisteína , Neoplasias Pulmonares , Proteoma , Humanos , Cisteína/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Arginina/metabolismo , Proteoma/metabolismo , Resistencia a Antineoplásicos/genética
4.
Food Res Int ; 187: 114311, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38763626

RESUMEN

The efficacy of amino acids as popular sports supplements has triggered debates, with their impact on athletic performance varying across sports disciplines due to diversity and heterogeneity in clinical trials. This review evaluates the ergogenic potential of amino acids, by critical appraisal of results of clinical trials of Branched chain amino acids (BCAAs), arginine, glutamine, citrulline, ß-alanine, and taurine, performed on elite sportsmen from various land and water sports. Clinical trials reviewed here confirm notable physiological benefits thereby supporting the claim that BCAA, citrulline and arginine in various doses can have positive effects on endurance and overall performance in sportsperson. Furthermore, results of clinical trials and metabolomic studies indicate that in future it would be more beneficial to design precise formulations to target the requirement of specific sports. For instance, some combinations of amino acids may be more suitable for long term endurance and some others may be suitable for short burst of excessive energy. The most important insights from this review are the identification of three key areas where research is urgently needed: a) Biomarkers that can identify the physiological end points and to distinguish the specific role of amino acid as anti-fatigue or reducing muscle soreness or enhancing energy b) In-depth sports-wise clinical trials on elite sportsperson to understand the ergogenic needs for the particular sports c) Design of precision formula for similar types of sports instead of common supplements.


Asunto(s)
Aminoácidos , Rendimiento Atlético , Suplementos Dietéticos , Fenómenos Fisiológicos en la Nutrición Deportiva , Humanos , Rendimiento Atlético/fisiología , Resistencia Física/efectos de los fármacos , Aminoácidos de Cadena Ramificada/metabolismo , beta-Alanina , Arginina/metabolismo
5.
Mol Cell ; 84(10): 1904-1916.e7, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38759626

RESUMEN

Many types of human cancers suppress the expression of argininosuccinate synthase 1 (ASS1), a rate-limiting enzyme for arginine production. Although dependency on exogenous arginine can be harnessed by arginine-deprivation therapies, the impact of ASS1 suppression on the quality of the tumor proteome is unknown. We therefore interrogated proteomes of cancer patients for arginine codon reassignments (substitutants) and surprisingly identified a strong enrichment for cysteine (R>C) in lung tumors specifically. Most R>C events did not coincide with genetically encoded R>C mutations but were likely products of tRNA misalignments. The expression of R>C substitutants was highly associated with oncogenic kelch-like epichlorohydrin (ECH)-associated protein 1 (KEAP1)-pathway mutations and suppressed by intact-KEAP1 in KEAP1-mutated cancer cells. Finally, functional interrogation indicated a key role for R>C substitutants in cell survival to cisplatin, suggesting that regulatory codon reassignments endow cancer cells with more resilience to stress. Thus, we present a mechanism for enriching lung cancer proteomes with cysteines that may affect therapeutic decisions.


Asunto(s)
Arginina , Cisteína , Proteína 1 Asociada A ECH Tipo Kelch , Neoplasias Pulmonares , Proteoma , Humanos , Cisteína/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteoma/metabolismo , Arginina/metabolismo , Mutación , Argininosuccinato Sintasa/metabolismo , Argininosuccinato Sintasa/genética , Cisplatino/farmacología , Línea Celular Tumoral , Proteómica/métodos , Regulación Neoplásica de la Expresión Génica , Supervivencia Celular/efectos de los fármacos , ARN de Transferencia/metabolismo , ARN de Transferencia/genética
6.
Proc Natl Acad Sci U S A ; 121(16): e2401313121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38602916

RESUMEN

All forms of life are presumed to synthesize arginine from citrulline via a two-step pathway consisting of argininosuccinate synthetase and argininosuccinate lyase using citrulline, adenosine 5'-triphosphate (ATP), and aspartate as substrates. Conversion of arginine to citrulline predominantly proceeds via hydrolysis. Here, from the hyperthermophilic archaeon Thermococcus kodakarensis, we identified an enzyme which we designate "arginine synthetase". In arginine synthesis, the enzyme converts citrulline, ATP, and free ammonia to arginine, adenosine 5'-diphosphate (ADP), and phosphate. In the reverse direction, arginine synthetase conserves the energy of arginine deimination and generates ATP from ADP and phosphate while releasing ammonia. The equilibrium constant of this reaction at pH 7.0 is [Cit][ATP][NH3]/[Arg][ADP][Pi] = 10.1 ± 0.7 at 80 °C, corresponding to a ΔG°' of -6.8 ± 0.2 kJ mol-1. Growth of the gene disruption strain was compared to the host strain in medium composed of amino acids. The results suggested that arginine synthetase is necessary in providing ornithine, the precursor for proline biosynthesis, as well as in generating ATP. Growth in medium supplemented with citrulline indicated that arginine synthetase can function in the direction of arginine synthesis. The enzyme is widespread in nature, including bacteria and eukaryotes, and catalyzes a long-overlooked energy-conserving reaction in microbial amino acid metabolism. Along with ornithine transcarbamoylase and carbamate kinase, the pathway identified here is designated the arginine synthetase pathway.


Asunto(s)
Arginina , Ligasas , Arginina/metabolismo , Citrulina/metabolismo , Amoníaco , Ornitina/genética , Adenosina Trifosfato/metabolismo , Fosfatos , Adenosina , Catálisis
7.
Biomolecules ; 14(4)2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38672418

RESUMEN

The post-translational modifications (PTMs) of proteins play a crucial role in increasing the functional diversity of proteins and are associated with the pathogenesis of various diseases. This review focuses on a less explored PTM called citrullination, which involves the conversion of arginine to citrulline. This process is catalyzed by peptidyl arginine deiminases (PADs). Different members of the PAD family have distinct tissue distribution patterns and functions. Citrullination is a post-translational modification of native proteins that can alter their structure and convert them into autoantigens; thus, it mediates the occurrence of autoimmune diseases. CD4+ T cells, including Th1, Th2, and Th17 cells, are important immune cells involved in mediating autoimmune diseases, allergic reactions, and tumor immunity. PADs can induce citrullination in CD4+ T cells, suggesting a role for citrullination in CD4+ T cell subset differentiation and function. Understanding the role of citrullination in CD4+ T cells may provide insights into immune-related diseases and inflammatory processes.


Asunto(s)
Linfocitos T CD4-Positivos , Citrulinación , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Animales , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/inmunología , Desiminasas de la Arginina Proteica/metabolismo , Procesamiento Proteico-Postraduccional , Citrulina/metabolismo , Arginina/metabolismo
8.
Front Immunol ; 15: 1363938, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38605962

RESUMEN

Arginine and tryptophan are pivotal in orchestrating cytokine-driven macrophage polarization and immune activation. Specifically, interferon-gamma (IFN-γ) stimulates inducible nitric oxide synthase (iNOS) expression), leading to the conversion of arginine into citrulline and nitric oxide (NO), while Interleukin-4 (IL4) promotes arginase activation, shifting arginine metabolism toward ornithine. Concomitantly, IFN-γ triggers indoleamine 2,3-dioxygenase 1 (IDO1) and Interleukin-4 induced 1 (IL4i1), resulting in the conversion of tryptophan into kynurenine and indole-3-pyruvic acid. These metabolic pathways are tightly regulated by NAD+-dependent sirtuin proteins, with Sirt2 and Sirt5 playing integral roles. In this review, we present novel insights that augment our understanding of the metabolic pathways of arginine and tryptophan following Mycobacterium tuberculosis infection, particularly their relevance in macrophage responses. Additionally, we discuss arginine methylation and demethylation and the role of Sirt2 and Sirt5 in regulating tryptophan metabolism and arginine metabolism, potentially driving macrophage polarization.


Asunto(s)
Arginina , Tuberculosis , Humanos , Arginina/metabolismo , Triptófano/metabolismo , Interleucina-4 , Sirtuina 2 , Activación de Macrófagos , Interferón gamma/farmacología
9.
J Anim Sci ; 1022024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38622951

RESUMEN

We determined apparent ileal digestibility (AID) and standardized ileal digestibility (SID) values of crude protein (CP) and amino acids (AA) in fermented soybean meal from five different sources (FSBM 1 to 5) in China when fed to mid and late-gestating sows. Twenty-four parity four sows (12 at 30 d in gestation and 12 at 80 d in gestation) were fitted with a T-cannula in the distal ileum and used in this experiment. Sows were randomly assigned to a replicated 6 × 3 Youden square design including six diets and three periods. Six diets were provided for sows in mid and late gestation, including a nitrogen-free diet and five test diets containing 26% FSBM from different sources. Results showed that there were differences in AID and SID of CP among the different FSBM samples, but no differences between sow physiological stages were observed. Specifically, when mid-gestating sows were fed FSBM 2, the AID of CP was the lowest, whereas FSBM 3 exhibited a greater AID of CP when compared to the other FSBM samples (P < 0.01). Furthermore, during late gestation, FSBM 3 consistently had greater SID of CP when compared to other FSBM samples (P < 0.01). The ileal digestibility of most AA varied with different FSBM samples. In both mid and late gestation, differences (P < 0.05) were observed for AID of lysine, tryptophan, histidine, and arginine across different FSBM samples. Similarly, the AID of dispensable AA (cysteine, glutamine, and serine) also exhibited differences (P < 0.05) across different FSBM samples in both mid and late-gestating sows. For mid-gestating sows, SID differences relating to lysine, phenylalanine, tryptophan, threonine, and arginine were observed among different diets (P < 0.05). In late-gestating sows, SID values for lysine, tryptophan, leucine, and arginine differed across diets (P < 0.05). Furthermore, the ileal digestibility of some dispensable AA was influenced by physiological stage, as evidenced by greater AID and SID values for glycine, glutamine, cysteine, and serine in late-gestating sows when compared to mid-gestating sows (P < 0.01). In summary, our study determined AA ileal digestibility of different FSBM fed to mid and late-gestating sows. We observed that the AA ileal digestibility differed among five FSBM samples, but the physiological stage of sows did not affect the ileal digestibility of CP and most AA. Additionally, when formulating diets for sows, it is crucial to consider the nutritional value differences of FSBM.


Fermented soybean meal (FSBM) is obtained from the microbial fermentation of soybean meal, which reduces anti-nutritional factor levels and enhances other nutrient content. Substituting soybean meal with FSBM in piglet and growing pig diets improves nutrient digestibility. However, its nutritional value for sows remains unclear. Therefore, five sources of FSBM were fed to sows in mid and late gestation to evaluate apparent ileal digestibility (AID) and standardized ileal digestibility (SID) values of amino acids (AA). We found that different FSBM samples impacted the SID value of AA when fed to gestating sows. Additionally, sow physiological stage influenced the SID of some dispensable AA. These findings provide valuable insights into the incorporation of FSBM into sow diets.


Asunto(s)
Aminoácidos , Alimentos Fermentados , Porcinos , Animales , Femenino , Embarazo , Aminoácidos/metabolismo , Digestión/fisiología , Glutamina/metabolismo , Triptófano/metabolismo , Cisteína/metabolismo , Lisina/metabolismo , Glycine max , Dieta/veterinaria , Arginina/metabolismo , Serina , Alimentación Animal/análisis , Íleon/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales
10.
Nat Commun ; 15(1): 2809, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38561334

RESUMEN

Protein arginine methyltransferase 9 (PRMT9) is a recently identified member of the PRMT family, yet its biological function remains largely unknown. Here, by characterizing an intellectual disability associated PRMT9 mutation (G189R) and establishing a Prmt9 conditional knockout (cKO) mouse model, we uncover an important function of PRMT9 in neuronal development. The G189R mutation abolishes PRMT9 methyltransferase activity and reduces its protein stability. Knockout of Prmt9 in hippocampal neurons causes alternative splicing of ~1900 genes, which likely accounts for the aberrant synapse development and impaired learning and memory in the Prmt9 cKO mice. Mechanistically, we discover a methylation-sensitive protein-RNA interaction between the arginine 508 (R508) of the splicing factor 3B subunit 2 (SF3B2), the site that is exclusively methylated by PRMT9, and the pre-mRNA anchoring site, a cis-regulatory element that is critical for RNA splicing. Additionally, using human and mouse cell lines, as well as an SF3B2 arginine methylation-deficient mouse model, we provide strong evidence that SF3B2 is the primary methylation substrate of PRMT9, thus highlighting the conserved function of the PRMT9/SF3B2 axis in regulating pre-mRNA splicing.


Asunto(s)
Empalme Alternativo , ARN , Animales , Humanos , Ratones , Arginina/metabolismo , Ratones Noqueados , Mutación , Proteína-Arginina N-Metiltransferasas/metabolismo , ARN/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN/genética
11.
Proc Natl Acad Sci U S A ; 121(17): e2320312121, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38625935

RESUMEN

In gram-positive bacteria, phosphorylated arginine functions as a protein degradation signal in a similar manner as ubiquitin in eukaryotes. The protein-arginine phosphorylation is mediated by the McsAB complex, where McsB possesses kinase activity and McsA modulates McsB activity. Although mcsA and mcsB are regulated within the same operon, the role of McsA in kinase activity has not yet been clarified. In this study, we determined the molecular mechanism by which McsA regulates kinase activity. The crystal structure of the McsAB complex shows that McsA binds to the McsB kinase domain through a second zinc-coordination domain and the subsequent loop region. This binding activates McsB kinase activity by rearranging the catalytic site, preventing McsB self-assembly, and enhancing stoichiometric substrate binding. The first zinc-coordination and coiled-coil domains of McsA further activate McsB by reassembling the McsAB oligomer. These results demonstrate that McsA is the regulatory subunit for the reconstitution of the protein-arginine kinase holoenzyme. This study provides structural insight into how protein-arginine kinase directs the cellular protein degradation system.


Asunto(s)
Arginina Quinasa , Proteínas Quinasas , Proteínas Quinasas/metabolismo , Arginina Quinasa/metabolismo , Arginina/metabolismo , Proteínas Bacterianas/metabolismo , Fosforilación , Zinc
12.
Biochim Biophys Acta Biomembr ; 1866(5): 184323, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38614236

RESUMEN

Protamine, an antimicrobial protein derived from salmon sperm with a molecular weight of approximately 5 kDa, is composed of 60-70 % arginine and is a highly charged protein. Here, we investigated the mechanism of antimicrobial action of protamine against Cutibacterium acnes (C. acnes) focusing on its rich arginine content and strong positive charge. Especially, we focused on the attribution of dual mechanisms of antimicrobial protein, including membrane disruption or interaction with intracellular components. We first determined the dose-dependent antibacterial activity of protamine against C. acnes. In order to explore the interaction between bacterial membrane and protamine, we analyzed cell morphology, zeta potential, membrane permeability, and the composition of membrane fatty acid. In addition, the localization of protamine in bacteria was observed using fluorescent-labeled protamine. For investigation of the intracellular targets of protamine, bacterial translation was examined using a cell-free translation system. Based on our results, the mechanism of the antimicrobial action of protamine against C. acnes is as follows: 1) electrostatic interactions with the bacterial cell membrane; 2) self-internalization into the bacterial cell by changing the composition of the bacterial membrane; and 3) inhibition of bacterial growth by blocking translation inside the bacteria. However, owing to its strong electric charge, protamine can also interact with DNA, RNA, and other proteins inside the bacteria, and may inhibit various bacterial life processes beyond the translation process.


Asunto(s)
Arginina , Membrana Celular , Protaminas , Protaminas/química , Protaminas/farmacología , Protaminas/metabolismo , Arginina/química , Arginina/farmacología , Arginina/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Animales , Antibacterianos/farmacología , Antibacterianos/química , Electricidad Estática , Permeabilidad de la Membrana Celular/efectos de los fármacos , Pruebas de Sensibilidad Microbiana
13.
ACS Synth Biol ; 13(5): 1549-1561, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38632869

RESUMEN

ATP is a universal energy currency that is essential for life. l-Arginine degradation via deamination is an elegant way to generate ATP in synthetic cells, which is currently limited by a slow l-arginine/l-ornithine exchange. We are now implementing a new antiporter with better kinetics to obtain faster ATP recycling. We use l-arginine-dependent ATP formation for the continuous synthesis and export of glycerol 3-phosphate by including glycerol kinase and the glycerol 3-phosphate/Pi antiporter. Exported glycerol 3-phosphate serves as a precursor for the biosynthesis of phospholipids in a second set of vesicles, which forms the basis for the expansion of the cell membrane. We have therefore developed an out-of-equilibrium metabolic network for ATP recycling, which has been coupled to lipid synthesis. This feeder-utilizer system serves as a proof-of-principle for the systematic buildup of synthetic cells, but the vesicles can also be used to study the individual reaction networks in confinement.


Asunto(s)
Adenosina Trifosfato , Arginina , Adenosina Trifosfato/metabolismo , Arginina/metabolismo , Células Artificiales/metabolismo , Glicerofosfatos/metabolismo , Glicerol Quinasa/metabolismo , Glicerol Quinasa/genética , Escherichia coli/metabolismo , Escherichia coli/genética , Lípidos/biosíntesis , Fosfolípidos/metabolismo , Redes y Vías Metabólicas
14.
Curr Biol ; 34(7): R267-R268, 2024 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-38593766

RESUMEN

In this Quick guide, Palmer and Berks introduce the twin-arginine translocation (Tat) systems. Tats are found in a variety of microbes and microbe-derived organelles, and are known to translocate folded substrate proteins across biological membranes.


Asunto(s)
Proteínas de Escherichia coli , Sistema de Translocación de Arginina Gemela , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Escherichia coli/metabolismo , Sistema de Translocación de Arginina Gemela/metabolismo , Membrana Celular/metabolismo , Arginina/metabolismo , Transporte de Proteínas , Señales de Clasificación de Proteína , Proteínas Bacterianas/metabolismo
15.
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
16.
Cell Signal ; 119: 111189, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38670475

RESUMEN

In patients on maintenance hemodialysis (MHD), vascular calcification (VC) is an independent predictor of cardiovascular disease (CVD), which is the primary cause of death in chronic kidney disease (CKD). The main component of VC in CKD is the vascular smooth muscle cells (VSMCs). VC is an ordered, dynamic activity. Under the stresses of oxidative stress and calcium-­phosphorus imbalance, VSMCs undergo osteogenic phenotypic transdifferentiation, which promotes the formation of VC. In addition to traditional epigenetics like RNA and DNA control, post-translational modifications have been discovered to be involved in the regulation of VC in recent years. It has been reported that the process of osteoblast differentiation is impacted by catalytic histone or non-histone arginine methylation. Its function in the osteogenic process is comparable to that of VC. Thus, we propose that arginine methylation regulates VC via many signaling pathways, including as NF-B, WNT, AKT/PI3K, TGF-/BMP/SMAD, and IL-6/STAT3. It might also regulate the VC-related calcification regulatory factors, oxidative stress, and endoplasmic reticulum stress. Consequently, we propose that arginine methylation regulates the calcification of the arteries and outline the regulatory mechanisms involved.


Asunto(s)
Arginina , Calcificación Vascular , Arginina/metabolismo , Humanos , Calcificación Vascular/metabolismo , Calcificación Vascular/patología , Metilación , Animales , Transducción de Señal , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Estrés Oxidativo
17.
FEBS Lett ; 598(8): 889-901, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38563123

RESUMEN

BeKm-1 is a peptide toxin from scorpion venom that blocks the pore of the potassium channel hERG (Kv11.1) in the human heart. Although individual protein structures have been resolved, the structure of the complex between hERG and BeKm-1 is unknown. Here, we used molecular dynamics and ensemble docking, guided by previous double-mutant cycle analysis data, to obtain an in silico model of the hERG-BeKm-1 complex. Adding to the previous mutagenesis study of BeKm-1, our model uncovers the key role of residue Arg20, which forms three interactions (a salt bridge and hydrogen bonds) with the channel vestibule simultaneously. Replacement of this residue even by lysine weakens the interactions significantly. In accordance, the recombinantly produced BeKm-1R20K mutant exhibited dramatically decreased activity on hERG. Our model may be useful for future drug design attempts.


Asunto(s)
Arginina , Canal de Potasio ERG1 , Simulación de Dinámica Molecular , Venenos de Escorpión , Animales , Humanos , Arginina/química , Arginina/metabolismo , Canal de Potasio ERG1/química , Canal de Potasio ERG1/metabolismo , Células HEK293 , Simulación del Acoplamiento Molecular , Mutación , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/metabolismo , Venenos de Escorpión/química , Venenos de Escorpión/genética , Venenos de Escorpión/metabolismo
18.
Arch Biochem Biophys ; 756: 110011, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38649133

RESUMEN

Structure-function relationships are key to understanding enzyme mechanisms, controlling enzyme activities, and designing biocatalysts. Here, we investigate the functions of arginine residues in the active sites of pyridoxal-5'-phosphate (PLP)-dependent non-canonical d-amino acid transaminases, focusing on the analysis of a transaminase from Haliscomenobacter hydrossis. Our results show that the tandem of arginine residues R28* and R90, which form the conserved R-[RK] motif in non-canonical d-amino acid transaminases, not only facilitates effective substrate binding but also regulates the catalytic properties of PLP. Non-covalent interactions between residues R28*, R90, and Y147 strengthen the hydrogen bond between Y147 and PLP, thereby maintaining the reactivity of the cofactor. Next, the R90 residue contributes to the stability of the holoenzyme. Finally, the R90I substitution induces structural changes that lead to substrate promiscuity, as evidenced by the effective binding of substrates with and without the α-carboxylate group. This study sheds light on the structural determinants of the activity of non-canonical d-amino acid transaminases. Understanding the structural basis of the active site plasticity in the non-canonical transaminase from H. hydrossis, which is characterized by effective conversion of d-amino acids and α-keto acids, may help to tailor it for industrial applications.


Asunto(s)
Arginina , Dominio Catalítico , Fosfato de Piridoxal , Transaminasas , Transaminasas/metabolismo , Transaminasas/química , Arginina/química , Arginina/metabolismo , Fosfato de Piridoxal/metabolismo , Fosfato de Piridoxal/química , Especificidad por Sustrato , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Modelos Moleculares
19.
BMC Vet Res ; 20(1): 167, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689278

RESUMEN

Arginine, which is metabolized into ornithine, proline, and nitric oxide, plays an important role in embryonic development. The present study was conducted to investigate the molecular mechanism of arginine in proliferation, differentiation, and physiological function of porcine trophoblast cells (pTr2) through metabolic pathways. The results showed that arginine significantly increased cell viability (P < 0.05). The addition of arginine had a quadratic tendency to increase the content of progesterone (P = 0.06) and protein synthesis rate (P = 0.03), in which the maximum protein synthesis rate was observed at 0.4 mM arginine. Arginine quadratically increased (P < 0.05) the intracellular contents of spermine, spermidine and putrescine, as well as linearly increased (P < 0.05) the intracellular content of NO in a dose-dependent manner. Arginine showed a quadratic tendency to increase the content of putrescine (P = 0.07) and a linear tendency to increase NO content (P = 0.09) in cell supernatant. Moreover, increasing arginine activated (P < 0.05) the mRNA expressions for ARG, ODC, iNOS and PCNA. Furthermore, inhibitors of arginine metabolism (L-NMMA and DFMO) both inhibited cell proliferation, while addition of its metabolites (NO and putrescine) promoted the cell proliferation and cell cycle, the mRNA expressions of PCNA, EGF and IGF-1, and increased (P < 0.05) cellular protein synthesis rate, as well as estradiol and hCG secretion (P < 0.05). In conclusion, our results suggested that arginine could promote cell proliferation and physiological function by regulating the metabolic pathway. Further studies showed that arginine and its metabolites modulate cell function mainly through ß-catenin and mTOR pathways.


Asunto(s)
Arginina , Diferenciación Celular , Proliferación Celular , Serina-Treonina Quinasas TOR , Trofoblastos , beta Catenina , Animales , Arginina/farmacología , Arginina/metabolismo , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Porcinos , Proliferación Celular/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Diferenciación Celular/efectos de los fármacos , beta Catenina/metabolismo , Supervivencia Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Óxido Nítrico/metabolismo , Línea Celular
20.
mSystems ; 9(5): e0024624, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38564708

RESUMEN

Dietary fiber deprivation is linked to probiotic extinction, mucus barrier dysbiosis, and the overgrowth of mucin-degrading bacteria. However, whether and how mucin could rescue fiber deprivation-induced intestinal barrier defects remains largely unexplored. Here, we sought to investigate the potential role and mechanism by which exogenous mucin maintains the gut barrier function. The results showed that dietary mucin alleviated fiber deprivation-induced disruption of colonic barrier integrity and reduced spermine production in vivo. Importantly, we highlighted that microbial-derived spermine production, but not host-produced spermine, increased significantly after mucin supplementation, with a positive association with upgraded colonic Lactobacillus abundance. After employing an in vitro model, the microbial-derived spermine was consistently dominated by both mucin and Lactobacillus spp. Furthermore, Limosilactobacillus mucosae was identified as an essential spermine-producing Lactobacillus spp., and this isolated strain was responsible for spermine accumulation, especially after adhering to mucin in vitro. Specifically, the mucin-supplemented bacterial supernatant of Limosilactobacillus mucosae was verified to promote intestinal barrier functions through the increased spermine production with a dependence on enhanced arginine metabolism. Overall, these findings collectively provide evidence that mucin-modulated microbial arginine metabolism bridged the interplay between microbes and gut barrier function, illustrating possible implications for host gut health. IMPORTANCE: Microbial metabolites like short-chain fatty acids produced by dietary fiber fermentation have been demonstrated to have beneficial effects on intestinal health. However, it is essential to acknowledge that certain amino acids entering the colon can be metabolized by microorganisms to produce polyamines. The polyamines can promote the renewal of intestinal epithelial cell and maintain host-microbe homeostasis. Our study highlighted the specific enrichment by mucin on promoting the arginine metabolism in Limosilactobacillus mucosae to produce spermine, suggesting that microbial-derived polyamines support a significant enhancement on the goblet cell proliferation and barrier function.


Asunto(s)
Arginina , Colon , Microbioma Gastrointestinal , Mucosa Intestinal , Mucinas , Espermina , Espermina/metabolismo , Mucinas/metabolismo , Arginina/metabolismo , Arginina/farmacología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Animales , Microbioma Gastrointestinal/fisiología , Colon/microbiología , Colon/metabolismo , Masculino , Ratones , Lactobacillus/metabolismo , Humanos , Fibras de la Dieta/metabolismo , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA