Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 695: 149467, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38211531

RESUMEN

Staphylococcus aureus (S. aureus), a Gram-positive bacterium, causes a wide range of infections, and diagnosis at an early stage is challenging. Targeting the maltodextrin transporter has emerged as a promising strategy for imaging bacteria and has been able to image a wide range of bacteria including S. aureus. However, little is known about the maltodextrin transporter in S. aureus, and this prevents new S. aureus specific ligands for the maltodextrin transporter from being developed. In Gram-positive bacteria, including S. aureus, the first step of maltodextrin transport is the binding of the maltodextrin-binding protein malE to maltodextrins. Thus, understanding the binding affinity and characteristics of malE from S. aureus is important to developing efficient maltodextrin-based imaging probes. We evaluated the affinity of malE of S. aureus to maltodextrins of various lengths. MalE of S. aureus (SAmalE) was expressed in E. coli BL21(DE3) and purified by Ni-NTA resin. The affinities of SAmalE to maltodextrins were evaluated with isothermal titration calorimetry. SAmalE has low affinity to maltose but binds to maltotriose and longer maltodextrins up to maltoheptaose with affinities up to Ka = 9.02 ± 0.49 × 105 M-1. SAmalE binding to maltotriose-maltoheptaose was exothermic and fit a single-binding site model. The van't Hoff enthalpy in the binding reaction of SAmalE with maltotriose was 9.9 ± 1.3 kcal/mol, and the highest affinity of SAmalE was observed with maltotetraose with Ka = 9.02 ± 0.49 × 105 M-1. In the plot of ΔH-T*ΔS, the of Enthalpy-Entropy Compensation effect was observed in binding reaction of SAmalE to maltodextrins. Acarbose and maltotetraiol bind with SAmalE indicating that SAmalE is tolerant of modifications on both the reducing and non-reducing ends of maltodextrins. Our results show that unlike ECmalE and similar to the maltodextrin binding protein of Streptococci, SAmalE primarily binds to maltodextrins via hydrogen bonds. This is distinct from the maltodextrin binding protein of Streptococci, SAmalE that binds to maltotetraiol with high affinity. Understanding the binding characteristics and tolerance to maltodextrins modifications by maltodextrin binding proteins will hopefully provide the basis for developing bacterial species-specific maltodextrin-based imaging probes.


Asunto(s)
Proteínas Portadoras , Staphylococcus aureus , Proteínas Portadoras/metabolismo , Staphylococcus aureus/metabolismo , Escherichia coli/metabolismo , Oligosacáridos/metabolismo , Proteínas Bacterianas/metabolismo , Polisacáridos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Calorimetría , Unión Proteica
2.
Biochemistry ; 62(24): 3533-3547, 2023 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-37729550

RESUMEN

Neurological disorders are often debilitating conditions with no cure. The majority of current therapies are palliative rather than disease-modifying; therefore, new strategies for treating neurological disorders are greatly needed. mRNA-based therapeutics have great potential for treating such neurological disorders; however, challenges with delivery have limited their clinical potential. Lipid nanoparticles (LNPs) are a promising delivery vector for the brain, given their safer toxicity profile and higher efficacy. Despite this, very little is known about LNP-mediated delivery of mRNA into the brain. Here, we employ MC3-based LNPs and successfully deliver Cre mRNA and Cas9 mRNA/Ai9 sgRNA to the adult Ai9 mouse brain; greater than half of the entire striatum and hippocampus was found to be penetrated along the rostro-caudal axis by direct intracerebral injections of MC3 LNP mRNAs. MC3 LNP Cre mRNA successfully transfected cells in the striatum (∼52% efficiency) and hippocampus (∼49% efficiency). In addition, we demonstrate that MC3 LNP Cas9 mRNA/Ai9 sgRNA edited cells in the striatum (∼7% efficiency) and hippocampus (∼3% efficiency). Further analysis demonstrates that MC3 LNPs mediate mRNA delivery to multiple cell types including neurons, astrocytes, and microglia in the brain. Overall, LNP-based mRNA delivery is effective in brain tissue and shows great promise for treating complex neurological disorders.


Asunto(s)
Nanopartículas , Enfermedades del Sistema Nervioso , Animales , Ratones , ARN Guía de Sistemas CRISPR-Cas , Encéfalo , ARN Mensajero/genética , ARN Interferente Pequeño
3.
Mol Syst Biol ; 18(1): e10495, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35005851

RESUMEN

Understanding mechanisms of antibiotic failure is foundational to combating the growing threat of multidrug-resistant bacteria. Prodrugs-which are converted into a pharmacologically active compound after administration-represent a growing class of therapeutics for treating bacterial infections but are understudied in the context of antibiotic failure. We hypothesize that strategies that rely on pathogen-specific pathways for prodrug conversion are susceptible to competing rates of prodrug activation and bacterial replication, which could lead to treatment escape and failure. Here, we construct a mathematical model of prodrug kinetics to predict rate-dependent conditions under which bacteria escape prodrug treatment. From this model, we derive a dimensionless parameter we call the Bacterial Advantage Heuristic (BAH) that predicts the transition between prodrug escape and successful treatment across a range of time scales (1-104 h), bacterial carrying capacities (5 × 104 -105 CFU/µl), and Michaelis constants (KM = 0.747-7.47 mM). To verify these predictions in vitro, we use two models of bacteria-prodrug competition: (i) an antimicrobial peptide hairpin that is enzymatically activated by bacterial surface proteases and (ii) a thiomaltose-conjugated trimethoprim that is internalized by bacterial maltodextrin transporters and hydrolyzed by free thiols. We observe that prodrug failure occurs at BAH values above the same critical threshold predicted by the model. Furthermore, we demonstrate two examples of how failing prodrugs can be rescued by decreasing the BAH below the critical threshold via (i) substrate design and (ii) nutrient control. We envision such dimensionless parameters serving as supportive pharmacokinetic quantities that guide the design and administration of prodrug therapeutics.


Asunto(s)
Infecciones Bacterianas , Profármacos , Antibacterianos/farmacología , Péptidos Antimicrobianos , Bacterias , Humanos
4.
Nat Immunol ; 11(7): 608-17, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20495560

RESUMEN

The mechanisms that initiate T helper type 2 (T(H)2) responses are poorly understood. Here we demonstrate that cysteine protease-induced T(H)2 responses occur via 'cooperation' between migratory dermal dendritic cells (DCs) and basophils positive for interleukin 4 (IL-4). Subcutaneous immunization with papain plus antigen induced reactive oxygen species (ROS) in lymph node DCs and in dermal DCs and epithelial cells of the skin. ROS orchestrated T(H)2 responses by inducing oxidized lipids that triggered the induction of thymic stromal lymphopoietin (TSLP) by epithelial cells mediated by Toll-like receptor 4 (TLR4) and the adaptor protein TRIF; by suppressing production of the T(H)1-inducing molecules IL-12 and CD70 in lymph node DCs; and by inducing the DC-derived chemokine CCL7, which mediated recruitment of IL-4(+) basophils to the lymph node. Thus, the T(H)2 response to cysteine proteases requires DC-basophil cooperation via ROS-mediated signaling.


Asunto(s)
Basófilos/metabolismo , Citocinas/biosíntesis , Células de Langerhans/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células Th2/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antígenos/inmunología , Basófilos/inmunología , Basófilos/patología , Comunicación Celular , Citocinas/genética , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Interleucina-4/biosíntesis , Células de Langerhans/inmunología , Células de Langerhans/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Papaína/inmunología , Especies Reactivas de Oxígeno/inmunología , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Linfopoyetina del Estroma Tímico
5.
Mol Pharm ; 19(1): 67-79, 2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34931518

RESUMEN

The development of endosomal disruptive agents is a major challenge in the field of drug delivery and pharmaceutical chemistry. Current endosomal disruptive agents are composed of polymers, peptides, and nanoparticles and have had limited clinical impact. Alternatives to traditional endosomal disruptive agents are therefore greatly needed. In this report, we introduce a new class of low molecular weight endosomal disruptive agents, termed caged surfactants, that selectively disrupt endosomes via reversible PEGylation under acidic endosomal conditions. The caged surfactants have the potential to address several of the limitations hindering the development of current endosomal disruptive agents, such as high toxicity and low excretion, and are amenable to traditional medicinal chemistry approaches for optimization. In this report, we synthesized three generations of caged surfactants and demonstrated that they can enhance the ability of cationic lipids to deliver mRNA into primary cells. We also show that caged surfactants can deliver siRNA into cells when modified with the RNA-binding dye thiazole orange. We anticipate that the caged surfactants will have numerous applications in pharmaceutical chemistry and drug delivery given their versatility.


Asunto(s)
Sistemas de Liberación de Medicamentos , Ácidos Nucleicos/administración & dosificación , Tensoactivos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Endosomas/efectos de los fármacos , Hemólisis/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , ARN Mensajero/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Relación Estructura-Actividad , Tensoactivos/administración & dosificación , Tensoactivos/química
6.
Nat Immunol ; 9(10): 1157-64, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18758466

RESUMEN

Robust production of type I interferon (IFN-alpha/beta) in plasmacytoid dendritic cells (pDCs) is crucial for antiviral immunity. Here we show involvement of the mammalian target of rapamycin (mTOR) pathway in regulating interferon production by pDCs. Inhibition of mTOR or its 'downstream' mediators, the p70 ribosomal S6 protein kinases p70S6K1 and p70S6K2, during pDC activation by Toll-like receptor 9 (TLR9) blocked the interaction of TLR9 with the adaptor MyD88 and subsequent activation of the interferon-regulatory factor IRF7, which resulted in impaired IFN-alpha/beta production. Microarray analysis confirmed that inhibition of mTOR by the immunosuppressive drug rapamycin suppressed antiviral and anti-inflammatory gene expression. Consistent with this, targeting rapamycin-encapsulated microparticles to antigen-presenting cells in vivo resulted in less IFN-alpha/beta production in response to CpG DNA or the yellow fever vaccine virus strain 17D. Thus, mTOR signaling is crucial in TLR-mediated IFN-alpha/beta responses by pDCs.


Asunto(s)
Células Dendríticas/metabolismo , Interferón Tipo I/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 9/metabolismo , Animales , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Regulación de la Expresión Génica/inmunología , Humanos , Immunoblotting , Inmunosupresores/farmacología , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas/inmunología , Proteínas Quinasas S6 Ribosómicas 70-kDa/inmunología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Receptor Toll-Like 9/inmunología , Transfección
7.
Chembiochem ; 19(20): 2173-2177, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30079487

RESUMEN

Biochemical assays that can identify ß-lactamase activity directly from patient samples have the potential to significantly improve the treatment of bacterial infections. However, current ß-lactamase probes do not have the sensitivity needed to measure ß-lactam resistance directly from patient samples. Here, we report the development of an instrument-free signal amplification technology, DETECT, that connects the activity of two enzymes in series to effectively amplify the activity of ß-lactamase 40 000-fold, compared to the standard ß-lactamase probe nitrocefin.


Asunto(s)
Infecciones Bacterianas/diagnóstico , Infecciones Bacterianas/microbiología , beta-Lactamasas/orina , Cefalosporinas/química , Humanos , Límite de Detección , Resistencia betalactámica
8.
Bioconjug Chem ; 29(5): 1729-1735, 2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29660287

RESUMEN

Trimethoprim is one of the most widely used antibiotics in the world. However, its efficacy is frequently limited by its poor water solubility and dose limiting toxicity. Prodrug strategies based on conjugation of oligosaccharides to trimethoprim have great potential for increasing the solubility of trimethoprim and lowering its toxicity, but they have been challenging to develop due to the sensitivity of trimethoprim to chemical modifications, and the rapid degradation of oligosaccharides in serum. In this report, we present a trimethoprim conjugate of maltodextrin termed TM-TMP, which increased the water solubility of trimethoprim by over 100 times, was stable to serum enzymes, and was active against urinary tract infections in mice. TM-TMP is composed of thiomaltose conjugated to trimethoprim, via a self-immolative disulfide linkage, and releases 4'-OH-trimethoprim (TMP-OH) after disulfide cleavage, which is a known metabolic product of trimethoprim and is as potent as trimethoprim. TM-TMP also contains a new maltodextrin targeting ligand composed of thiomaltose, which is stable to hydrolysis by serum amylases and therefore has the metabolic stability needed for in vivo use. TM-TMP has the potential to significantly improve the treatment of a wide number of infections given its high water solubility and the widespread use of trimethoprim.


Asunto(s)
Antibacterianos/química , Antibacterianos/uso terapéutico , Polisacáridos/química , Polisacáridos/uso terapéutico , Trimetoprim/análogos & derivados , Trimetoprim/uso terapéutico , Infecciones Urinarias/tratamiento farmacológico , Animales , Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Femenino , Maltosa/análogos & derivados , Maltosa/farmacología , Maltosa/uso terapéutico , Ratones , Polisacáridos/farmacología , Trimetoprim/farmacología
9.
Nature ; 470(7335): 543-7, 2011 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-21350488

RESUMEN

Many successful vaccines induce persistent antibody responses that can last a lifetime. The mechanisms by which they do so remain unclear, but emerging evidence indicates that they activate dendritic cells via Toll-like receptors (TLRs). For example, the yellow fever vaccine YF-17D, one of the most successful empiric vaccines ever developed, activates dendritic cells via multiple TLRs to stimulate proinflammatory cytokines. Triggering specific combinations of TLRs in dendritic cells can induce synergistic production of cytokines, which results in enhanced T-cell responses, but its impact on antibody responses remain unknown. Learning the critical parameters of innate immunity that program such antibody responses remains a major challenge in vaccinology. Here we demonstrate that immunization of mice with synthetic nanoparticles containing antigens plus ligands that signal through TLR4 and TLR7 induces synergistic increases in antigen-specific, neutralizing antibodies compared to immunization with nanoparticles containing antigens plus a single TLR ligand. Consistent with this there was enhanced persistence of germinal centres and of plasma-cell responses, which persisted in the lymph nodes for >1.5 years. Surprisingly, there was no enhancement of the early short-lived plasma-cell response relative to that observed with single TLR ligands. Molecular profiling of activated B cells, isolated 7 days after immunization, indicated that there was early programming towards B-cell memory. Antibody responses were dependent on direct triggering of both TLRs on B cells and dendritic cells, as well as on T-cell help. Immunization protected completely against lethal avian and swine influenza virus strains in mice, and induced robust immunity against pandemic H1N1 influenza in rhesus macaques.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Vacunas contra la Influenza/inmunología , Animales , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Células Dendríticas/citología , Células Dendríticas/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Ácido Láctico , Ligandos , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Macaca mulatta/inmunología , Macaca mulatta/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Linfocitos T/inmunología , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
10.
Nat Mater ; 14(7): 701-6, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25915034

RESUMEN

Therapeutics based on transcription factors have the potential to revolutionize medicine but have had limited clinical success as a consequence of delivery problems. The delivery of transcription factors is challenging because it requires the development of a delivery vehicle that can complex transcription factors, target cells and stimulate endosomal disruption, with minimal toxicity. Here, we present a multifunctional oligonucleotide, termed DARTs (DNA assembled recombinant transcription factors), which can deliver transcription factors with high efficiency in vivo. DARTs are composed of an oligonucleotide that contains a transcription-factor-binding sequence and hydrophobic membrane-disruptive chains that are masked by acid-cleavable galactose residues. DARTs have a unique molecular architecture, which allows them to bind transcription factors, trigger endocytosis in hepatocytes, and stimulate endosomal disruption. The DARTs have enhanced uptake in hepatocytes as a result of their galactose residues and can disrupt endosomes efficiently with minimal toxicity, because unmasking of their hydrophobic domains selectively occurs in the acidic environment of the endosome. We show that DARTs can deliver the transcription factor nuclear erythroid 2-related factor 2 (Nrf2) to the liver, catalyse the transcription of Nrf2 downstream genes, and rescue mice from acetaminophen-induced liver injury.


Asunto(s)
ADN/química , Oligonucleótidos/química , Factores de Transcripción/metabolismo , Alanina Transaminasa/metabolismo , Animales , Sistemas de Liberación de Medicamentos , Endosomas/metabolismo , Células Hep G2 , Hepatocitos/citología , Humanos , Concentración de Iones de Hidrógeno , Hidrólisis , Hígado/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Espectrometría de Fluorescencia , Distribución Tisular
11.
Anal Chem ; 87(3): 1941-9, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25540985

RESUMEN

Flow cytometry holds promise to accelerate antibiotic susceptibility determinations; however, without robust multidimensional statistical analysis, general discrimination criteria have remained elusive. In this study, a new statistical method, probability binning signature quadratic form (PB-sQF), was developed and applied to analyze flow cytometric data of bacterial responses to antibiotic exposure. Both sensitive lab strains (Escherichia coli and Pseudomonas aeruginosa) and a multidrug resistant, clinically isolated strain (E. coli) were incubated with the bacteria-targeted dye, maltohexaose-conjugated IR786, and each of many bactericidal or bacteriostatic antibiotics to identify changes induced around corresponding minimum inhibition concentrations (MIC). The antibiotic-induced damages were monitored by flow cytometry after 1-h incubation through forward scatter, side scatter, and fluorescence channels. The 3-dimensional differences between the flow cytometric data of the no-antibiotic treated bacteria and the antibiotic-treated bacteria were characterized by PB-sQF into a 1-dimensional linear distance. A 99% confidence level was established by statistical bootstrapping for each antibiotic-bacteria pair. For the susceptible E. coli strain, statistically significant increments from this 99% confidence level were observed from 1/16x MIC to 1x MIC for all the antibiotics. The same increments were recorded for P. aeruginosa, which has been reported to cause difficulty in flow-based viability tests. For the multidrug resistant E. coli, significant distances from control samples were observed only when an effective antibiotic treatment was utilized. Our results suggest that a rapid and robust antimicrobial susceptibility test (AST) can be constructed by statistically characterizing the differences between sample and control flow cytometric populations, even in a label-free scheme with scattered light alone. These distances vs paired controls coupled with rigorous statistical confidence limits offer a new path toward investigating initial biological responses, screening for drugs, and shortening time to result in antimicrobial sensitivity testing.


Asunto(s)
Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Citometría de Flujo/métodos , Pruebas de Sensibilidad Microbiana/métodos , Pseudomonas aeruginosa/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Citometría de Flujo/economía , Humanos , Pruebas de Sensibilidad Microbiana/economía , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Factores de Tiempo
12.
Proc Natl Acad Sci U S A ; 108(21): 8803-8, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21555563

RESUMEN

The mechanisms by which enteric commensal microbiota influence maturation and repair of the epithelial barrier are relatively unknown. Epithelial restitution requires active cell migration, a process dependent on dynamic turnover of focal cell-matrix adhesions (FAs). Here, we demonstrate that natural, commensal bacteria stimulate generation of reactive oxygen species (ROS) in intestinal epithelia. Bacteria-mediated ROS generation induces oxidation of target cysteines in the redox-sensitive tyrosine phosphatases, LMW-PTP and SHP-2, which in turn results in increased phosphorylation of focal adhesion kinase (FAK), a key protein regulating the turnover of FAs. Accordingly, phosphorylation of FAK substrate proteins, focal adhesion formation, and cell migration are all significantly enhanced by bacterial contact in both in vitro and in vivo models of wound closure. These results suggest that commensal bacteria regulate cell migration via induced generation of ROS in epithelial cells.


Asunto(s)
Enterobacteriaceae/metabolismo , Células Epiteliales/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Movimiento Celular , Adhesiones Focales/metabolismo , Ratones , Ratones Endogámicos C57BL , Monoéster Fosfórico Hidrolasas , Cicatrización de Heridas
13.
BMC Complement Altern Med ; 14: 97, 2014 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-24617993

RESUMEN

BACKGROUND: Snail transcription factor can induce epithelial-mesenchymal transition (EMT), associated with decreased cell adhesion-associated molecules like E-cadherin, increased mesenchymal markers like vimentin, leading to increased motility, invasion and metastasis. Muscadine grape skin extract (MSKE) has been shown to inhibit prostate cancer cell growth and induce apoptosis without affecting normal prostate epithelial cells. We investigated novel molecular mechanisms by which Snail promotes EMT in prostate cancer cells via Reactive Oxygen Species (ROS) and whether it can be antagonized by MSKE. METHODS: ARCaP and LNCaP cells overexpressing Snail were utilized to examine levels of reactive oxygen species (ROS), specifically, superoxide, in vitro using Dihydroethidium (DHE) or HydroCy3 dyes. Mitosox staining was performed to determine whether the source of ROS was mitochondrial in origin. We also investigated the effect of Muscadine grape skin extract (MSKE) on EMT marker expression by western blot analysis. Migration and cell viability using MTS proliferation assay was performed following MSKE treatments. RESULTS: Snail overexpression in ARCaP and LNCaP cells was associated with increased concentration of mitochondrial superoxide, in vitro. Interestingly, MSKE decreased superoxide levels in ARCaP and LNCaP cells. Additionally, MSKE and Superoxide Dismutase (SOD) reverted EMT as evidenced by decreased vimentin levels and re-induction of E-cadherin expression in ARCaP-Snail cells after 3 days, concomitant with reduced cell migration. MSKE also decreased Stat-3 activity in ARCaP-Snail cells. CONCLUSIONS: This study shows that superoxide species may play a role in Snail transcription factor-mediated EMT. Therefore, therapeutic targeting of Snail with various antioxidants such as MSKE may prove beneficial in abrogating EMT and ROS-mediated tumor progression in human prostate cancer.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Extractos Vegetales/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Superóxidos/metabolismo , Factores de Transcripción/metabolismo , Vitis/química , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Factores de Transcripción de la Familia Snail
14.
Angew Chem Int Ed Engl ; 53(51): 14096-14101, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25330976

RESUMEN

A positron emission tomography (PET) tracer composed of (18)F-labeled maltohexaose (MH(18)F) can image bacteria in vivo with a sensitivity and specificity that are orders of magnitude higher than those of fluorodeoxyglucose ((18)FDG). MH(18)F can detect early-stage infections composed of as few as 10(5) E. coli colony-forming units (CFUs), and can identify drug resistance in bacteria in vivo. MH(18)F has the potential to improve the diagnosis of bacterial infections given its unique combination of high specificity and sensitivity for bacteria.


Asunto(s)
Infecciones por Escherichia coli/diagnóstico , Radioisótopos de Flúor , Oligosacáridos , Tomografía de Emisión de Positrones , Animales , Infecciones por Escherichia coli/tratamiento farmacológico , Radioisótopos de Flúor/química , Estructura Molecular , Oligosacáridos/síntesis química , Oligosacáridos/química , Ratas
15.
Bioconjug Chem ; 24(1): 4-8, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23237198

RESUMEN

In this report, we present a new strategy for targeting chemotherapeutics to tumors, based on targeting extracellular DNA. A gemcitabine prodrug was synthesized, termed H-gemcitabine, which is composed of Hoechst conjugated to gemcitabine. H-gemcitabine has low toxicity because it is membrane-impermeable; however, it still has high tumor efficacy because of its ability to target gemcitabine to E-DNA in tumors. We demonstrate here that H-gemcitabine has a wider therapeutic window than free gemcitabine.


Asunto(s)
Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/uso terapéutico , Desoxicitidina/análogos & derivados , Neoplasias/tratamiento farmacológico , Profármacos/química , Profármacos/uso terapéutico , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/farmacocinética , Sitios de Unión , Línea Celular Tumoral , ADN/metabolismo , Desoxicitidina/administración & dosificación , Desoxicitidina/química , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Ratones , Ratones Desnudos , Neoplasias/metabolismo , Profármacos/administración & dosificación , Profármacos/farmacocinética , Gemcitabina
17.
Arthritis Rheum ; 64(6): 1899-908, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22231023

RESUMEN

OBJECTIVE: To assess temporal changes in cartilage and bone morphology, reactive oxygen species (ROS), and vascularization in rats with monosodium iodoacetate (MIA)-induced osteoarthritis (OA), using advanced imaging methodologies. METHODS: Right knees of 8-week-old male Wistar rats were injected with 1 mg MIA in 50 µl saline and left knees were injected with 50 µl saline as controls. After 1, 2, and 3 weeks (n = 5 at each time point), changes in cartilage morphology and composition were quantified using equilibrium partitioning of an ionic contrast agent microfocal computed tomography (µCT), and changes in subchondral and trabecular bone were assessed by standard µCT. ROS were characterized by in vivo fluorescence imaging at 1, 11, and 21 days (n = 5 at each time point). Three weeks following fluorescence imaging, alterations in knee joint vascularity were quantified with µCT after perfusion of a vascular contrast agent. RESULTS: Femoral cartilage volume, thickness, and proteoglycan content were significantly decreased in MIA-injected knees compared with control knees, accompanied by loss of trabecular bone and erosion of subchondral bone surface. ROS quantities were significantly increased 1 day after MIA injection and subsequently decreased gradually, having returned to normal by 21 days. Vascularity in whole knees and distal femora was significantly increased at 21 days after MIA injection. CONCLUSION: Contrast-enhanced µCT and fluorescence imaging were combined to characterize articular cartilage, subchondral bone, vascularization, and ROS, providing unprecedented 3-dimensional joint imaging and quantification in multiple tissues during OA progression. These advanced imaging techniques have the potential to become standardized methods for comprehensive evaluation of articular joint degeneration and evaluation of therapeutic efficacy.


Asunto(s)
Artritis Experimental/diagnóstico por imagen , Huesos/diagnóstico por imagen , Cartílago Articular/diagnóstico por imagen , Neovascularización Patológica/diagnóstico por imagen , Osteoartritis de la Rodilla/diagnóstico por imagen , Especies Reactivas de Oxígeno/metabolismo , Animales , Artritis Experimental/metabolismo , Huesos/metabolismo , Cartílago Articular/metabolismo , Articulación de la Rodilla/diagnóstico por imagen , Articulación de la Rodilla/metabolismo , Masculino , Neovascularización Patológica/metabolismo , Osteoartritis de la Rodilla/inducido químicamente , Osteoartritis de la Rodilla/metabolismo , Radiografía , Ratas , Ratas Wistar
18.
J Am Soc Nephrol ; 23(5): 793-800, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22282594

RESUMEN

Antioxidant therapy can protect against ischemic injury, but the inability to selectively target the kidney would require extremely high doses to achieve effective local concentrations of drug. Here, we developed a directed therapeutic that specifically targets an antioxidant to renal proximal tubule cells via the folate receptor. Because a local increase in superoxide contributes to renal ischemic injury, we created the folate-antioxidant conjugate 4-hydroxy-Tempo (tempol)-folate to target folate receptors, which are highly expressed in the proximal tubule. Dihydroethidium high-performance liquid chromatography demonstrated that conjugated tempol retained its efficacy to scavenge superoxide in proximal tubule cells. In a mouse model of renal ischemia-reperfusion injury, tempol-folate reduced renal superoxide levels more effectively than tempol alone. Furthermore, electron spin resonance revealed the successful targeting of the tempol-folate conjugate to the kidney and other tissues expressing folate receptors. Administration of tempol-folate protected the renal function of mice after ischemia-reperfusion injury and inhibited infiltration of macrophages. In conclusion, kidney-specific targeting of an antioxidant has therapeutic potential to prevent renal ischemic injury. Conjugation of other pharmaceuticals to folate may also facilitate the development of treatments for other kidney diseases.


Asunto(s)
Antioxidantes/uso terapéutico , Transportadores de Ácido Fólico/fisiología , Riñón/irrigación sanguínea , Daño por Reperfusión/tratamiento farmacológico , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Células Cultivadas , Óxidos N-Cíclicos/farmacocinética , Óxidos N-Cíclicos/farmacología , Humanos , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/metabolismo , Marcadores de Spin , Superóxidos/metabolismo
19.
J Control Release ; 354: 188-195, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36596342

RESUMEN

Gene therapy approaches that utilize Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) ribonucleases have tremendous potential to treat human disease. However, CRISPR therapies delivered by integrating viral vectors are limited by potential off-target genome editing caused by constitutive activation of ribonuclease functions. Thus, biomaterial formulations are being used for the delivery of purified CRISPR components to increase the efficiency and safety of genome editing approaches. We previously demonstrated that a novel peptide identified by phage display, TAxI-peptide, mediates delivery of recombinant proteins into neurons. In this report we utilized NeutrAvidin protein to formulate neuron-targeted genome-editing nanoparticles. Cas12a ribonucleases was loaded with biotinylated guide RNA and biotinylated TAxI-peptide onto NeutrAvidin protein to coordinate the formation a targeted ribonuclease protein (RNP) complex. TAxI-RNP complexes are polydisperse with a 14.3 nm radius. The nanoparticles are stable after formulation and show good stability in the presence of normal mouse serum. TAxI-RNP nanoparticles increased neuronal delivery of Cas12a in reporter mice, resulting in induced tdTomato expression after direct injection into the dentate gyrus of the hippocampus. TAxI-RNP nanoparticles also increased genome editing efficacy in hippocampal neurons versus glia. These studies demonstrate the ability to assemble RNP nanoformulations with NeutrAvidin by binding biotinylated peptides and gRNA-loaded Cas12a ribonucleases into protein nanoparticles that target CRISPR delivery to specific cell-types in vivo. The potential to deliver CRISPR nanoparticles to specific cell-types and control off-target delivery to further reduce deleterious genome editing is essential for the creation of viable therapies to treat nervous system disease.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Ratones , Animales , Humanos , Edición Génica/métodos , Ribonucleasas , Péptidos , Neuronas
20.
Adv Drug Deliv Rev ; 200: 115026, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37516409

RESUMEN

The field of gene editing has received much attention in recent years due to its immense therapeutic potential. In particular, gene editing therapeutics, such as the CRISPR-Cas systems, base editors, and other emerging gene editors, offer the opportunity to address previously untreatable disorders. This review aims to summarize the therapeutic applications of gene editing based on mRNA delivery. We introduce gene editing therapeutics using mRNA and focus on engineering and improvement of gene editing technology. We subsequently examine ex vivo and in vivo gene editing techniques and conclude with an exploration of the next generation of CRISPR and base editing systems.


Asunto(s)
Edición Génica , Técnicas de Transferencia de Gen , Humanos , Edición Génica/métodos , ARN Mensajero/genética , Sistemas CRISPR-Cas , Terapia Genética/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA