Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ecotoxicol Environ Saf ; 272: 116084, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38350217

RESUMO

Polyhexamethylene guanidine (PHMG) is manufactured and applied extensively due to its superior disinfectant capabilities. However, the inhalatory exposure to PHMG aerosols is increasingly recognized as a potential instigator of pulmonary fibrosis, prompting an urgent call for elucidation of the underlying pathophysiological mechanisms. Within this context, alveolar macrophages play a pivotal role in the primary immune defense in the respiratory tract. Dysregulated lipid metabolism within alveolar macrophages leads to the accumulation of foam cells, a process that is intimately linked with the pathogenesis of pulmonary fibrosis. Therefore, this study examines PHMG's effects on alveolar macrophage foaminess and its underlying mechanisms. We conducted a 3-week inhalation exposure followed by a 3-week recovery period in C57BL/6 J mice using a whole-body exposure system equipped with a disinfection aerosol generator (WESDAG). The presence of lipid-laden alveolar macrophages and downregulation of pulmonary tissue lipid transport proteins ABCA1 and ABCG1 were observed in mice. In cell culture models involving lipid-loaded macrophages, we demonstrated that PHMG promotes foam cell formation by inhibiting lipid efflux in mouse alveolar macrophages. Furthermore, PHMG-induced foam cells were found to promote an increase in the release of TGF-ß1, fibronectin deposition, and collagen remodeling. In vivo interventions were subsequently implemented on mice exposed to PHMG aerosols, aiming to restore macrophage lipid efflux function. Remarkably, this intervention demonstrated the potential to retard the progression of pulmonary fibrosis. In conclusion, this study underscores the pivotal role of macrophage foaming in the pathogenesis of PHMG disinfectants-induced pulmonary fibrosis. Moreover, it provides compelling evidence to suggest that the regulation of macrophage efflux function holds promise for mitigating the progression of pulmonary fibrosis, thereby offering novel insights into the mechanisms underlying inhaled PHMG disinfectants-induced pulmonary fibrosis.


Assuntos
Desinfetantes , Fibrose Pulmonar , Camundongos , Animais , Fibrose Pulmonar/metabolismo , Guanidina/toxicidade , Guanidina/metabolismo , Camundongos Endogâmicos C57BL , Aerossóis e Gotículas Respiratórios , Pulmão , Guanidinas/metabolismo , Macrófagos , Desinfetantes/farmacologia , Lipídeos
2.
Microbiol Spectr ; 11(1): e0346922, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36475769

RESUMO

The increasing emergence of antibiotic resistance is an urgent threat to global health care; thus, there is a need for new therapeutics. Guanidine is the preferred functional group for antimicrobial design and development. Herein, the potential antibacterial activity of the guanidine derivative isopropoxy benzene guanidine (IBG) against multidrug-resistant (MDR) bacteria was discovered. The synergistic antibacterial activity of IBG and colistin was determined by checkerboard assay, time-killing curve, and mouse experiments. The antibacterial mechanism of IBG was verified in fluorescent probe experiments, intracellular oxidative phosphorylation assays, and transcriptome analysis. The results showed that IBG displays efficient antibacterial activity against Gram-positive pathogens and Gram-negative pathogens with permeabilized outer membranes. Further mechanistic studies showed that IBG triggers cytoplasmic membrane damage by binding to phosphatidylglycerol and cardiolipin, leading to the dissipation of proton motive force and accumulation of intracellular ATP. IBG combined with low levels of colistin enhances bacterial outer membrane permeability and increases the accumulation of reactive oxygen species, as further evidenced by transcriptome analysis. Furthermore, the efficacy of IBG with colistin against MDR Escherichia coli in three infection models was demonstrated. Together, these results suggest that IBG is a promising adjuvant of colistin, providing an alternative approach to address the prevalent infections caused by MDR Gram-negative pathogens. IMPORTANCE As antibiotic discovery stagnates, the world is facing a growing menace from the emergence of bacteria that are resistant to almost all available antibiotics. The key to winning this race is to explore distinctive mechanisms of antibiotics. Thus, novel efficient antibacterial agents and alternative strategies are urgently required to fill the void in antibiotic development. Compared with the large amount of money and time required to develop new agents, the antibiotic adjuvant strategy is a promising approach to inhibit bacterial resistance and increase killing of bacteria. In this study, we found that the guanidine derivatives IBG not only displayed efficient antibacterial activities against Gram-positive bacteria but also restored colistin susceptibility of Gram-negative pathogens as an antibiotic adjuvant. More in-depth study showed that IBG is a potential lead to overcome antibiotic resistance, providing new insight into future antibiotic discovery and development.


Assuntos
Benzeno , Colistina , Animais , Camundongos , Colistina/farmacologia , Benzeno/metabolismo , Benzeno/farmacologia , Bactérias Gram-Negativas/metabolismo , Guanidina/metabolismo , Guanidina/farmacologia , Antibacterianos/química , Bactérias/metabolismo , Escherichia coli/metabolismo , Farmacorresistência Bacteriana Múltipla , Guanidinas/farmacologia , Testes de Sensibilidade Microbiana
3.
Bioorg Med Chem Lett ; 76: 129017, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36209968

RESUMO

Despite the enormous potential of siRNAs to transcriptionally downregulate disease causing proteins in many genetic diseases, efficient delivery and endosomal escape are the two bottlenecks that have resulted in only a handful of FDA approved drugs. In this report, we have successfully delivered siRNA against Nanog with the help of pentafluorobenzyl modified Internal Oligo-guanidinium transporter (IGT) that has previously shown promising results in peptide and antisense morpholino delivery. Nanog downregulation in prostate cancer cell line DU145 in serum containing media led to suppression of associated proteins such as KLF4, FAK and cMyc and also enhanced the chemosensitivity of Epirubicin, an anthracycline based drug, in DU145 cells by associated MDR-1 downregulation in vitro. These results show that IGT is a promising candidate for siRNA delivery and its conjugation with stable siRNAs could enhance the chemotherapeutic efficiency of siRNAs alone and in combination with small molecule-based drugs.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Epirubicina , Proteína Homeobox Nanog , Proteínas de Transporte de Cátions Orgânicos , Neoplasias da Próstata , RNA Interferente Pequeno , Humanos , Masculino , Linhagem Celular Tumoral , Epirubicina/farmacologia , Guanidina/metabolismo , Morfolinos , Proteína Homeobox Nanog/genética , Peptídeos/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , RNA Interferente Pequeno/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética
4.
FEBS Open Bio ; 12(10): 1700-1716, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35979612

RESUMO

Recent biochemical experiments have indicated that in Candida albicans, a commensal fungal pathogen, the Ras signaling pathway plays a significant role in the yeast-to-hyphal transition; specifically, two enzymes in this pathway, Adenyl Cyclase Cyr1 and GTPase activating protein Ira2, facilitate this transition, in the presence of energy sensor ATP. However, the precise mechanism by which protein interactions between Ira2 and Cyr1 and the energy sensor ATP result in the yeast-to-hyphal transition and create a switch-like process are unknown. We propose a new set of biochemical reaction steps that captures all the essential interactions between Ira2, Cyr1, and ATP in the Ras pathway. With the help of chemical reaction network theory, we demonstrate that this set of biochemical reaction steps results in bistability. Further, bifurcation analysis of the differential equations based on this set of reaction steps supports the existence of a bistable switch, and this switch may act as a checkpoint mechanism for the promotion of growth-to-hyphal transition in C. albicans.


Assuntos
Adenilil Ciclases , Candida albicans , Trifosfato de Adenosina/metabolismo , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Candida albicans/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Guanidina/metabolismo , Modelos Teóricos
5.
J Cell Mol Med ; 26(14): 3816-3827, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35678269

RESUMO

Radix puerariae, a traditional Chinese herbal medication, has been used to treat patients with diabetic kidney disease (DKD). Our previous studies demonstrated that puerarin, the active compound of radix puerariae, improves podocyte injury in type 1 DKD mice. However, the direct molecular target of puerarin and its underlying mechanisms in DKD remain unknown. In this study, we confirmed that puerarin also improved DKD in type 2 diabetic db/db mice. Through RNA-sequencing odf isolated glomeruli, we found that differentially expressed genes (DEGs) that were altered in the glomeruli of these diabetic mice but reversed by puerarin treatment were involved mostly in oxidative stress, inflammatory and fibrosis. Further analysis of these reversed DEGs revealed protein kinase A (PKA) was among the top pathways. By utilizing the drug affinity responsive target stability method combined with mass spectrometry analysis, we identified guanine nucleotide-binding protein Gi alpha-1 (Gnai1) as the direct binding partner of puerarin. Gnai1 is an inhibitor of cAMP production which is known to have protection against podocyte injury. In vitro, we showed that puerarin not only interacted with Gnai1 but also increased cAMP production in human podocytes and mouse diabetic kidney in vivo. Puerarin also enhanced CREB phosphorylation, a downstream transcription factor of cAMP/PKA. Overexpression of CREB reduced high glucose-induced podocyte apoptosis. Inhibition of PKA by Rp-cAMP also diminished the effects of puerarin on high glucose-induced podocyte apoptosis. We conclude that the renal protective effects of puerarin are likely through inhibiting Gnai1 to activate cAMP/PKA/CREB pathway in podocytes.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Podócitos , Animais , Apoptose , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/farmacologia , Glucose/metabolismo , Guanidina/metabolismo , Guanidina/farmacologia , Guanidina/uso terapêutico , Humanos , Isoflavonas , Camundongos , Nucleotídeos/metabolismo , Podócitos/metabolismo
6.
Nat Commun ; 12(1): 5150, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34446715

RESUMO

Recent studies have revealed the prevalence and biological significance of guanidine metabolism in nature. However, the metabolic pathways used by microbes to degrade guanidine or mitigate its toxicity have not been widely studied. Here, via comparative proteomics and subsequent experimental validation, we demonstrate that Sll1077, previously annotated as an agmatinase enzyme in the model cyanobacterium Synechocystis sp. PCC 6803, is more likely a guanidinase as it can break down guanidine rather than agmatine into urea and ammonium. The model cyanobacterium Synechococcus elongatus PCC 7942 strain engineered to express the bacterial ethylene-forming enzyme (EFE) exhibits unstable ethylene production due to toxicity and genomic instability induced by accumulation of the EFE-byproduct guanidine. Co-expression of EFE and Sll1077 significantly enhances genomic stability and enables the resulting strain to achieve sustained high-level ethylene production. These findings expand our knowledge of natural guanidine degradation pathways and demonstrate their biotechnological application to support ethylene bioproduction.


Assuntos
Proteínas de Bactérias/metabolismo , Etilenos/biossíntese , Instabilidade Genômica , Guanidina/metabolismo , Synechococcus/genética , Synechococcus/metabolismo , Synechocystis/enzimologia , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Synechocystis/genética
7.
J Nutr Biochem ; 81: 108381, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32422424

RESUMO

One-carbon metabolism is a collection of metabolic cycles that supports methylation and provides one-carbon bound folates for the de novo synthesis of purine and thymidine nucleotides. The methylation of phosphatidylethanolamine to form choline has been extensively studied in the context of fatty liver disease. However, the role of one-carbon metabolism in supporting nucleotide synthesis during liver damage has not been addressed. The objective of this study is to determine how the disruption of one-carbon metabolism influences nucleotide metabolism in the liver after dietary methionine and choline restriction. Mice (n=8) were fed a methionine-choline-deficient or control diet for 3 weeks. We treated mice with the compound alloxazine (0.5 mg/kg), a known adenosine receptor antagonist, every second day during the final week of feeding to probe the function of adenosine signaling during liver damage. We found that concentrations of several hepatic nucleotides were significantly lower in methionine- and choline-deficient mice vs. controls (adenine: 13.9±0.7 vs. 10.1±0.6, guanine: 1.8±0.1 vs. 1.4±0.1, thymidine: 0.0122±0.0027 vs. 0.0059±0.0027 nmol/mg dry tissue). Treatment of alloxazine caused a specific decrease in thymidine nucleotides, decrease in mitochondrial content in the liver and exacerbation of steatohepatitis as shown by the increased hepatic lipid content and altered macrophage morphology. This study demonstrates a role for one-carbon metabolism in supporting de novo nucleotide synthesis and mitochondrial function during liver damage.


Assuntos
Carbono/metabolismo , Fígado Gorduroso/metabolismo , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Nucleotídeos/metabolismo , Adenosina/metabolismo , Animais , Colina/farmacologia , Deficiência de Colina/metabolismo , Dieta , Modelos Animais de Doenças , Flavinas/farmacologia , Guanidina/metabolismo , Inflamação/metabolismo , Fígado/metabolismo , Fígado/patologia , Masculino , Metionina/deficiência , Metionina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Antagonistas de Receptores Purinérgicos P1/farmacologia , Timidina/metabolismo
8.
ACS Appl Mater Interfaces ; 12(16): 18363-18374, 2020 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-32242658

RESUMO

The development of novel antimicrobials is a top priority to address the growing epidemic of multidrug-resistant pathogens. Since cationic nonamphiphilic star-shaped antimicrobials are promising molecular scaffolds that provide a high charge density in binding anionic bacterial bilayers, this research aimed to further increase their membrane perturbation capability by introducing guanidinium groups to the antimicrobials via enhancing membrane insertion. In particular, computational simulation and experimental investigations revealed that our designed guanidinium-rich alternating copolypeptide, four-armed poly(arginine-alt-glycine), can interact with both the headgroups and unsaturated tails of phospholipids in bacterial membranes through multiple interactions, including electrostatic, cation-π, and T-shaped π-π interactions, allowing it to penetrate deeper inside the biologically inaccessible high-energy barrier of the hydrophobic lipid bilayer interior to cause membrane permeabilization and precipitation of the bacterial cytoplasm. Furthermore, glycine was observed to have a unique effect in enhancing the performance of arginine-based copolypeptide. Four-armed poly(arginine-alt-glycine) exhibited broad-spectrum antimicrobial activity, high bactericidal efficiency, and negligible hemolysis. The in vivo antibacterial performance of the copolypeptide was superior to that of doxycycline in a mouse model of Pseudomonas aeruginosa skin infection, accompanied by negligible local and systemic toxicity. Our results demonstrate that this guanidinium-rich, nonamphiphilic, star-shaped structure may promote the development of next-generation antimicrobials.


Assuntos
Antibacterianos , Bactérias/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Guanidina , Peptídeos , Animais , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Bactérias/citologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Guanidina/química , Guanidina/metabolismo , Guanidina/farmacologia , Hemólise/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Nanoestruturas/química , Peptídeos/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Ratos , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus/efeitos dos fármacos
9.
Chem Commun (Camb) ; 55(95): 14387-14390, 2019 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-31723950

RESUMO

We designed a tandem stimuli-responsive assembly based on a guanidinium-modified calix[5]arene (GC5A-6C) and eosin Y modified hyaluronic acid (EY-HA), which showed hyaluronidase-triggered disassembly and ATP-activated release of EY. Both hyaluronidase and ATP are tumor biomarkers, and therefore, the present system shows potential in precision delivery with respect to tumor phototheranostics.


Assuntos
Trifosfato de Adenosina/metabolismo , Calixarenos/metabolismo , Amarelo de Eosina-(YS)/metabolismo , Guanidina/metabolismo , Ácido Hialurônico/metabolismo , Hialuronoglucosaminidase/metabolismo , Trifosfato de Adenosina/química , Biomarcadores Tumorais/química , Biomarcadores Tumorais/metabolismo , Calixarenos/química , Amarelo de Eosina-(YS)/química , Guanidina/química , Humanos , Ácido Hialurônico/química , Hialuronoglucosaminidase/química , Substâncias Macromoleculares/química , Substâncias Macromoleculares/metabolismo , Polímeros/química , Polímeros/metabolismo , Nanomedicina Teranóstica , Microambiente Tumoral
10.
J Phys Chem B ; 123(44): 9302-9311, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31597039

RESUMO

Guanidinium cation (Gdm+) interacts strongly with amino acids of different polarities modulating protein structure and function. Using density functional theory calculations and molecular dynamics simulations, we studied the interaction of Gdm+ with carboxylate ions mimicking its interaction with acidic amino acids and explored its effect in enzymatic folding and activity. We show that, in low concentrations, Gdm+ stabilizes carboxylate ion dimers by acting as a bridge between them, thereby reducing the electrostatic repulsion. We further show that this carboxylate-Gdm+-carboxylate interaction can have an effect on the structure-activity relationship in enzymes with active sites containing two acidic residues. Using five enzymes (hen egg white lysozyme, T4 lysozyme, HIV-1 protease, pepsin, and creatine kinase), which have two acidic amino acids in their active sites, we show that, in low concentrations (<0.5 M), Gdm+ strongly binds to the enzyme active site, thereby potentially inhibiting its activity without unfolding it. This can lead to misleading conclusions in experiments, which infer the extent of enzyme unfolding from activity measurements. However, the carboxylate-Gdm+-carboxylate specific interaction can be exploited in drug discovery as drugs based on guanidinium derivatives are already being used to treat various maladies related to muscle weakness, cancer, diabetes etc. Guanidinium derivatives can be designed as potential drug molecules to inhibit activity or functioning of enzymes, which have binding pockets with two acidic residues in close vicinity.


Assuntos
Ácidos Carboxílicos/química , Enzimas/química , Enzimas/metabolismo , Guanidina/química , Domínio Catalítico , Creatina Quinase/química , Creatina Quinase/metabolismo , Teoria da Densidade Funcional , Desenho de Fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Guanidina/metabolismo , Guanidina/farmacologia , Protease de HIV/química , Protease de HIV/metabolismo , Íons , Simulação de Dinâmica Molecular , Muramidase/antagonistas & inibidores , Muramidase/química , Muramidase/metabolismo , Pepsina A/química , Pepsina A/metabolismo , Conformação Proteica , Eletricidade Estática
11.
Inorg Chem ; 58(5): 2954-2964, 2019 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-30735039

RESUMO

Emulating enzymatic reactivity using small molecules has been a long-time challenging pursuit of the scientific community. Peroxidases, ubiquitous heme enzymes that are involved in hormone synthesis and the immune system, have been a prime target of such efforts due to their tremendous potential in the chemical industry as well as in wastewater treatment. Here it is demonstrated that inclusion of a second sphere guanidine moiety in an iron porphyrin not only makes this small molecule a veritable peroxidase catalyst but also offers an auxiliary binding site for organic substrates, facilitating their rapid oxidation with a green oxidant like H2O2. This small molecule analogue exhibits a "ping-pong" mechanism and Michaelis-Menten type kinetics, which is generally typical of metallo-enzymes and follows a mechanism of the natural enzyme in its entirety, including the formation of compound I as the primary oxidant.


Assuntos
Ferro/metabolismo , Metaloporfirinas/metabolismo , Peroxidases/metabolismo , Sítios de Ligação , Biocatálise , Cristalografia por Raios X , Guanidina/química , Guanidina/metabolismo , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Ferro/química , Cinética , Metaloporfirinas/síntese química , Metaloporfirinas/química , Modelos Moleculares , Estrutura Molecular , Oxirredução , Peroxidases/química
12.
Nature ; 566(7742): 94-99, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30728519

RESUMO

Small molecules containing the N-nitroso group, such as the bacterial natural product streptozotocin, are prominent carcinogens1,2 and important cancer chemotherapeutics3,4. Despite the considerable importance of this functional group to human health, enzymes dedicated to the assembly of the N-nitroso unit have not been identified. Here we show that SznF, a metalloenzyme from the biosynthesis of streptozotocin, catalyses an oxidative rearrangement of the guanidine group of Nω-methyl-L-arginine to generate an N-nitrosourea product. Structural characterization and mutagenesis of SznF reveal two separate active sites that promote distinct steps in this transformation using different iron-containing metallocofactors. This biosynthetic reaction, which has little precedent in enzymology or organic synthesis, expands the catalytic capabilities of non-haem-iron-dependent enzymes to include N-N bond formation. We find that biosynthetic gene clusters that encode SznF homologues are widely distributed among bacteria-including environmental organisms, plant symbionts and human pathogens-which suggests an unexpectedly diverse and uncharacterized microbial reservoir of bioactive N-nitroso metabolites.


Assuntos
Metaloproteínas/metabolismo , Estreptozocina/biossíntese , Estreptozocina/química , Arginina/análogos & derivados , Domínio Catalítico/genética , Coenzimas/metabolismo , Cristalografia por Raios X , Guanidina/metabolismo , Ferro/metabolismo , Metaloproteínas/química , Metaloproteínas/genética , Modelos Moleculares , Família Multigênica , Compostos de Nitrosoureia/metabolismo , Streptomyces/enzimologia , Streptomyces/genética
13.
J Med Chem ; 61(7): 2910-2923, 2018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29522685

RESUMO

Peptide-binding G protein-coupled receptors (GPCRs) are key effectors in numerous pathological and physiological pathways. The assessment of the receptor-bound conformation of a peptidic ligand within a membrane receptor such as a GPCR is of great impact for a rational drug design of more potent analogues. In this work, we applied multiple ligand-based nuclear magnetic resonance (NMR) methods to study the interaction of peptide heptamers, derived from the C-X-C Motif Chemokine 12 (CXCL12), and the C-X-C Chemokine Receptor Type 4 (CXCR4) on membranes of human T-Leukemia cells (CCRF-CEM cells). This study represents the first structural investigation reporting the receptor-bound conformation of a peptide to a GPCR directly on a living cell. The results obtained in the field of CXCL12/CXCR4 are proofs of concept, although important information for researchers dealing with the CXCR4 field arises. General application of the presented NMR methodologies is possible and surely may help to boost the development of new therapeutic agents targeting GPCRs.


Assuntos
Neoplasias/metabolismo , Receptores CXCR4/antagonistas & inibidores , Animais , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Quimiocina CXCL12/antagonistas & inibidores , Quimiocina CXCL12/metabolismo , Cricetinae , Cricetulus , Mapeamento de Epitopos , Guanidina/metabolismo , Humanos , Leucemia de Células T/metabolismo , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Conformação Molecular , Simulação de Dinâmica Molecular , Receptores CXCR4/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
14.
J Am Chem Soc ; 140(7): 2687-2692, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29381064

RESUMO

We developed dendritic caged molecular glues (CagedGlue-R) as tags for nucleus-targeted drug delivery, whose multiple guanidinium ion (Gu+) pendants are protected by an anionic photocleavable unit (butyrate-substituted nitroveratryloxycarbonyl; BANVOC). Negatively charged CagedGlue-R hardly binds to anionic biomolecules because of their electrostatic repulsion. However, upon exposure of CagedGlue-R to UV light or near-infrared (NIR) light, the BANVOC groups of CagedGlue-R are rapidly detached to yield an uncaged molecular glue (UncagedGlue-R) that carries multiple Gu+ pendants. Because Gu+ forms a salt bridge with PO4-, UncagedGlue-R tightly adheres to anionic biomolecules such as DNA and phospholipids in cell membranes by a multivalent salt-bridge formation. When tagged with CagedGlue-R, guests can be taken up into living cells via endocytosis and hide in endosomes. However, when the CagedGlue-R tag is photochemically uncaged to form UncagedGlue-R, the guests escape from the endosome and migrate into the cytoplasm followed by the cell nucleus. We demonstrated that quantum dots (QDs) tagged with CagedGlue-R can be delivered efficiently to cell nuclei eventually by irradiation with light.


Assuntos
Adesivos/metabolismo , Núcleo Celular/metabolismo , Guanidina/metabolismo , Luz , Adesivos/química , Linhagem Celular Tumoral , Núcleo Celular/química , Endocitose , Endossomos/química , Endossomos/metabolismo , Guanidina/química , Humanos , Estrutura Molecular , Processos Fotoquímicos , Pontos Quânticos/química , Pontos Quânticos/metabolismo
15.
Biophys J ; 111(11): 2349-2357, 2016 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-27926836

RESUMO

Unfolded protein, a disordered structure found before folding of newly synthesized protein or after protein denaturation, is a substrate for binding by many cellular factors such as heat-stable proteins, chaperones, and many small molecules. However, it is challenging to directly probe such interactions in physiological solution conditions because proteins are largely in their folded state. In this work we probed small molecule binding to mechanically unfolded polyprotein using sodium dodecyl sulfate (SDS) as an example. The effect of binding is quantified based on changes in the elasticity and refolding of the unfolded polyprotein in the presence of SDS. We show that this single-molecule mechanical detection of binding to unfolded polyprotein can serve, to our knowledge, as a novel label-free assay with a great potential to study many factors that interact with unfolded protein domains, which underlie many important biological processes.


Assuntos
Elasticidade , Desnaturação Proteica , Redobramento de Proteína , Proteínas/química , Proteínas/metabolismo , Guanidina/metabolismo , Guanidina/farmacologia , Modelos Moleculares , Desnaturação Proteica/efeitos dos fármacos , Redobramento de Proteína/efeitos dos fármacos , Dodecilsulfato de Sódio/metabolismo , Dodecilsulfato de Sódio/farmacologia , Ureia/metabolismo , Ureia/farmacologia
16.
Biochemistry ; 54(38): 5920-36, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26352925

RESUMO

Vis toxin was identified by a bioinformatics strategy as a putative virulence factor produced by Vibrio splendidus with mono-ADP-ribosyltransferase activity. Vis was purified to homogeneity as a 28 kDa single-domain enzyme and was shown to possess NAD(+)-glycohydrolase [KM(NAD(+)) = 276 ± 12 µM] activity and with an R-S-E-X-E motif; it targets arginine-related compounds [KM(agmatine) = 272 ± 18 mM]. Mass spectrometry analysis revealed that Vis labels l-arginine with ADP-ribose from the NAD(+) substrate at the amino nitrogen of the guanidinium side chain. Vis is toxic to yeast when expressed in the cytoplasm under control of the CUP1 promotor, and catalytic variants lost the ability to kill the yeast host, indicating that the toxin exerts its lethality through its enzyme activity. Several small molecule inhibitors were identified from a virtual screen, and the most potent compounds were found to inhibit the transferase activity of the enzyme with Ki values ranging from 25 to 134 µM. Inhibitor compound M6 bears the necessary attributes of a solid candidate as a lead compound for therapeutic development. Vis toxin was crystallized, and the structures of the apoenzyme (1.4 Å) and the enzyme bound with NAD(+) (1.8 Å) and with the M6 inhibitor (1.5 Å) were determined. The structures revealed that Vis represents a new subgroup within the mono-ADP-ribosyltransferase toxin family.


Assuntos
ADP Ribose Transferases/química , Toxinas Bacterianas/química , Vibrio/enzimologia , ADP Ribose Transferases/metabolismo , Adenosina Difosfato Ribose/metabolismo , Sequência de Aminoácidos , Toxinas Bacterianas/metabolismo , Cristalografia por Raios X , Guanidina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , NAD+ Nucleosidase/química , NAD+ Nucleosidase/metabolismo , Conformação Proteica , Alinhamento de Sequência , Vibrio/química , Vibrio/metabolismo
17.
Amino Acids ; 47(9): 1751-62, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26123990

RESUMO

Hyperargininemia is caused by deficiency of arginase 1, which catalyzes the hydrolysis of L-arginine to urea as the final enzyme in the urea cycle. In contrast to other urea cycle defects, arginase 1 deficiency usually does not cause catastrophic neonatal hyperammonemia but rather presents with progressive neurological symptoms including seizures and spastic paraplegia in the first years of life and hepatic pathology, such as neonatal cholestasis, acute liver failure, or liver fibrosis. Some patients have developed hepatocellular carcinoma. A usually mild or moderate hyperammonemia may occur at any age. The pathogenesis of arginase I deficiency is yet not fully understood. However, the accumulation of L-arginine and the resulting abnormalities in the metabolism of guanidine compounds and nitric oxide have been proposed to play a major pathophysiological role. This article provides an update on the first patients ever described, gives an overview of the distinct clinical characteristics, biochemical as well as genetical background and discusses treatment options.


Assuntos
Arginase , Arginina/metabolismo , Hiperargininemia , Arginina/genética , Pré-Escolar , Feminino , Guanidina/metabolismo , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Hiperamonemia/fisiopatologia , Hiperargininemia/genética , Hiperargininemia/metabolismo , Hiperargininemia/patologia , Hiperargininemia/fisiopatologia , Lactente , Hepatopatias/genética , Hepatopatias/metabolismo , Hepatopatias/patologia , Hepatopatias/fisiopatologia , Paraplegia/genética , Paraplegia/metabolismo , Paraplegia/patologia , Paraplegia/fisiopatologia , Convulsões/genética , Convulsões/metabolismo , Convulsões/patologia , Convulsões/fisiopatologia
18.
Acta Physiol (Oxf) ; 215(1): 13-23, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25939450

RESUMO

AIM: As post-translational modifications of proteins may have an impact on the pathogenesis of diseases such as atherosclerosis, diabetes mellitus and chronic kidney disease (CKD), post-translational modifications are currently gaining increasing interest. In this study, a comprehensive method for analysis of these post-translational modifications is established for the clinical diagnostic routine. METHODS: Here, we analysed albumin - the most abundant plasma protein in human - isolated from patients with CKD and healthy controls by chromatographic steps and identified by MALDI mass spectrometry. Post-translational modifications of albumin were identified after digestion by analysing mass signal shifts of albumin peptides using pertinent mass databases. RESULTS: Albumin isolated from plasma of patients with CKD but not from healthy control subjects was specifically post-translationally modified by guanidinylation of lysines at positions 249, 468, 548, 565 and 588. After identification of guanidinylations as post-translational modifications of albumin isolated from patients with CKD, these modifications were quantified by mass spectrometry demonstrating a significant increase in the corresponding mass signal intensities in patients with CKD compared to healthy controls. The relative amount of guanidinylation of lysine at position 468 in patients with CKD was determined as 63 ± 32% (N = 3). Subsequently, we characterized the pathophysiological impact of the post-translational guanidinylation on the binding capacity of albumin for representative hydrophobic metabolic waste products. In vitro guanidinylation of albumin from healthy control subjects caused a decreased binding capacity of albumin in a time-dependent manner. Binding of indoxyl sulphate (protein-bound fraction) decreased from 82 ± 1% of not post-translationally modified albumin to 56 ± 1% after in vitro guanidinylation (P < 0.01), whereas the binding of tryptophan decreased from 20 to 4%. These results are in accordance with the binding of indoxyl sulphate to albumin from healthy control subjects and patients with CKD (88 ± 3 vs. 74 ± 10, P < 0.01). Thus, in vitro post-translational guanidinylation of albumin had a direct effect on the binding capacity of hydrophobic metabolites such as indoxyl sulphate and tryptophan. CONCLUSION: We used a mass spectrometry-based method for the characterization of post-translational modification and demonstrated the pathophysiological impact of a representative post-translational modification of plasma albumin. The data described in this study may help to elucidate the pathophysiological role of protein modifications.


Assuntos
Processamento de Proteína Pós-Traducional/fisiologia , Insuficiência Renal Crônica/sangue , Albumina Sérica/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Guanidina/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lisina/metabolismo , Masculino , Espectrometria de Massas/métodos , Pessoa de Meia-Idade , Peso Molecular , Peptídeos/metabolismo
19.
J Am Chem Soc ; 137(1): 362-8, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25495130

RESUMO

The rational design of theranostic nanoparticles exhibiting synergistic turn-on of therapeutic potency and enhanced diagnostic imaging in response to tumor milieu is critical for efficient personalized cancer chemotherapy. We herein fabricate self-reporting theranostic drug nanocarriers based on hyperbranched polyprodrug amphiphiles (hPAs) consisting of hyperbranched cores conjugated with reduction-activatable camptothecin prodrugs and magnetic resonance (MR) imaging contrast agent (Gd complex), and hydrophilic coronas functionalized with guanidine residues. Upon cellular internalization, reductive milieu-actuated release of anticancer drug in the active form, activation of therapeutic efficacy (>70-fold enhancement in cytotoxicity), and turn-on of MR imaging (∼9.6-fold increase in T1 relaxivity) were simultaneously achieved in the simulated cytosol milieu. In addition, guanidine-decorated hPAs exhibited extended blood circulation with a half-life up to ∼9.8 h and excellent tumor cell penetration potency. The hyperbranched chain topology thus provides a novel theranostic polyprodrug platform for synergistic imaging/chemotherapy and enhanced tumor uptake.


Assuntos
Antineoplásicos/metabolismo , Permeabilidade da Membrana Celular , Liberação Controlada de Fármacos , Imageamento por Ressonância Magnética , Polímeros/metabolismo , Pró-Fármacos/metabolismo , Tensoativos/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Liberação Controlada de Fármacos/efeitos dos fármacos , Guanidina/química , Guanidina/metabolismo , Guanidina/farmacologia , Células Hep G2 , Humanos , Modelos Moleculares , Estrutura Molecular , Nanopartículas/química , Nanopartículas/metabolismo , Oxirredução/efeitos dos fármacos , Polímeros/química , Polímeros/farmacologia , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Relação Estrutura-Atividade , Tensoativos/química , Tensoativos/farmacologia
20.
Inorg Chem ; 53(12): 6113-26, 2014 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-24884156

RESUMO

With the aim of assessing whether ruthenium(II) compounds with meridional geometry might be utilized as potential antitumor agents, a series of new, water-soluble, monofunctional ruthenium(II) complexes of the general formula mer-[Ru(L3)(N-N)X][Y]n (where L3 = 2,2':6',2″-terpyridine (tpy) or 4'-chloro-2,2':6',2″-terpyridine (Cl-tpy), N-N = 1,2-diaminoethane (en), 1,2-diaminocyclohexane (dach), or 2,2'-bipyridine (bpy); X = Cl or dmso-S; Y = Cl, PF6, or CF3SO3; n = 1 or 2, depending on the nature of X) were synthesized. All complexes were fully characterized by elemental analysis and spectroscopic techniques (IR, UV/visible, and 1D and 2D NMR), and for three of them, i.e., [Ru(Cl-tpy)(bpy)Cl][Cl] (3Cl), [Ru(Cl-tpy)(en)(dmso-S)][Y]2 [Y = PF6 (6PF6), CF3SO3 (6OTf)] and [Ru(Cl-tpy)(bpy)(dmso-S)][CF3SO3]2 (8OTf), the X-ray structure was also determined. The new terpyridine complexes, with the exception of 8, are well soluble in water (>25 mg/mL). (1)H and (31)P NMR spectroscopy studies performed on the three selected complexes [Ru(Cl-tpy)(N-N)Cl](+) [N-N = en (1), dach (2), and bpy (3)] demonstrated that, after hydrolysis of the Cl ligand, they are capable of interacting with guanine derivatives [i.e., 9-methylguanine (9MeG) or guanosine-5'-monophosphate (5'-GMP)] through N7, forming monofunctional adducts with rates and extents that depend strongly on the nature of N-N: 1 ≈ 2 ≫ 3. In addition, compound 1 shows high selectivity toward 5'-GMP compared to adenosine-5'-monophosphate (5'-AMP), in a competition experiment. Quantitative kinetic investigations on 1 and 2 were performed by means of UV/visible spectroscopy. Overall, the complexes with bidentate aliphatic diamines proved to be superior to those with bpy in terms of solubility and reactivity (i.e., release of Cl(-) and capability to bind guanine derivatives). Contrary to the chlorido compounds, the corresponding dmso derivatives proved to be inert (viz., they do not release the monodentate ligand) in aqueous media.


Assuntos
2,2'-Dipiridil/análogos & derivados , Antineoplásicos/química , Guanidina/análogos & derivados , Compostos Organometálicos/química , Rutênio/química , 2,2'-Dipiridil/química , 2,2'-Dipiridil/farmacologia , Monofosfato de Adenosina/metabolismo , Antineoplásicos/farmacologia , Cristalografia por Raios X , Guanidina/metabolismo , Guanosina Monofosfato/metabolismo , Cinética , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Compostos Organometálicos/farmacologia , Rutênio/farmacologia , Solubilidade , Água/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA