Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomaterials ; 303: 122401, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38006645

RESUMEN

Peptide amphiphile (PA) nanofibers have been shown to target and deliver drugs when administered via an intravenous (IV) injection. Subcutaneous administration can broaden the applicability of PA nanofibers in the medical field. The ability of PA nanofibers to be absorbed into systemic circulation after subcutaneous administration was investigated. Four PA molecules with different amino acid sequences were designed to understand the effect of nanofiber cohesion and charge on uptake. Solution small-angle X-ray scattering confirmed nanostructure morphology and provided characteristic lengths for co-assemblies. Circular dichroism and solution wide-angle X-ray scattering confirmed PA secondary structure and molecular order. PAs were co-assembled in a 95 %:5 % molar ratio of unlabeled PA to fluorescently labeled PA. Male and female Sprague Dawley rats were injected in the nape of the neck with PA co-assemblies. In vivo normalized abdominal fluorescence was measured 1-72 h after injection. PA nanofibers with a negative charge and low internal order showed the highest amount of systemic absorption at 1, 6, and 24 h. At 24 h after injection, white blood cell count decreased and glucose was elevated. Glucose began to decrease at 48 h. These data indicate that PA nanofibers can be absorbed into the systemic circulation after subcutaneous injection.


Asunto(s)
Nanofibras , Ratas , Animales , Masculino , Femenino , Nanofibras/química , Ratas Sprague-Dawley , Péptidos/química , Inyecciones Subcutáneas , Glucosa
2.
ACS Appl Mater Interfaces ; 15(22): 26340-26348, 2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37235485

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection relies on its spike protein binding to angiotensin-converting enzyme 2 (ACE2) on host cells to initiate cellular entry. Blocking the interactions between the spike protein and ACE2 offers promising therapeutic opportunities to prevent infection. We report here on peptide amphiphile supramolecular nanofibers that display a sequence from ACE2 in order to promote interactions with the SARS-CoV-2 spike receptor binding domain. We demonstrate that displaying this sequence on the surface of supramolecular assemblies preserves its α-helical conformation and blocks the entry of a pseudovirus and its two variants into human host cells. We also found that the chemical stability of the bioactive structures was enhanced in the supramolecular environment relative to the unassembled peptide molecules. These findings reveal unique advantages of supramolecular peptide therapies to prevent viral infections and more broadly for other targets as well.


Asunto(s)
COVID-19 , Nanofibras , Humanos , SARS-CoV-2/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Unión Proteica , Péptidos/farmacología , Péptidos/metabolismo
3.
ACS Nano ; 16(5): 7309-7322, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35504018

RESUMEN

An abdominal aortic aneurysm (AAA) is a localized dilation of the aorta located in the abdomen that poses a severe risk of death when ruptured. The cause of AAA is not fully understood, but degradation of medial elastin due to elastolytic matrix metalloproteinases is a key step leading to aortic dilation. Current therapeutic interventions are limited to surgical repair to prevent catastrophic rupture. Here, we report the development of injectable supramolecular nanofibers using peptide amphiphile molecules designed to localize to AAA by targeting fragmented elastin, matrix metalloproteinase 2 (MMP-2), and membrane type 1 matrix metalloproteinase. We designed four targeting peptide sequences from X-ray crystallographic data and incorporated them into PA molecules via solid phase peptide synthesis. After coassembling targeted and diluent PAs at different molar ratios, we assessed their ability to form nanofibers using transmission electron microscopy and to localize to AAA in male and female Sprague-Dawley rats using light sheet fluorescence microscopy. We found that three formulations of the PA nanofibers were able to localize to AAA tissue, but the MMP-2 targeting PA substantially outperformed the other nanofibers. Additionally, we demonstrated that the MMP-2 targeting PA nanofibers had an optimal dose of 5 mg (∼12 mg/kg). Our results show that there was not a significant difference in targeting between male and female Sprague-Dawley rats. Given the ability of the MMP-2 targeting PA nanofiber to localize to AAA tissue, future studies will investigate potential diagnostic and targeted drug delivery applications for AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal , Nanofibras , Ratas , Animales , Masculino , Femenino , Metaloproteinasa 2 de la Matriz/metabolismo , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Elastina , Nanofibras/química , Ratas Sprague-Dawley , Péptidos/metabolismo , Aorta Abdominal/metabolismo
4.
Adv Ther (Weinh) ; 4(9)2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34926792

RESUMEN

Atherosclerotic plaque remains the leading contributor to cardiovascular disease and requires invasive surgical procedures for its removal. Nanomedicine offers a minimally invasive approach to alleviate plaque burden by targeted therapeutic delivery. However, nanocarriers are limited without the ability to sense and respond to the diseased microenvironment. In this study, targeted self-assembled peptide amphiphile (PA) nanofibers were developed that cleave in response to biochemical cues expressed in atherosclerotic lesions-reactive oxygen species (ROS) and intracellular glutathione-to deliver a liver X receptor agonist (LXR) to enhance macrophage cholesterol efflux. The PAs released LXR in response to physiological levels of ROS and reducing agents and could be co-assembled with plaque-targeting PAs to form nanofibers. The resulting LXR PA nanofibers promoted cholesterol efflux from macrophages in vitro as well as LXR alone and with lower cytotoxicity. Further, the ApoA1-LXR PA nanofibers targeted plaque within an atherosclerotic mouse model in vivo and activated ATP-binding cassette A1 (ABCA1) expression as well as LXR alone with reduced liver toxicity. Taken together, these results demonstrate the potential of self-assembled PA nanofibers for controlled therapeutic delivery to the atherosclerotic niche.

5.
Adv Healthc Mater ; 10(13): e2100302, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34061473

RESUMEN

Pulmonary hypertension is a highly morbid disease with no cure. Available treatments are limited by systemic adverse effects due to non-specific biodistribution. Self-assembled peptide amphiphile (PA) nanofibers are biocompatible nanomaterials that can be modified to recognize specific biological markers to provide targeted drug delivery and reduce off-target toxicity. Here, PA nanofibers that target the angiotensin I-converting enzyme and the receptor for advanced glycation end-products (RAGE) are developed, as both proteins are overexpressed in the lung with pulmonary hypertension. It is demonstrated that intravenous delivery of RAGE-targeted nanofibers containing the targeting epitope LVFFAED (LVFF) significantly accumulated within the lung in a chronic hypoxia-induced pulmonary hypertension mouse model. Using 3D light sheet fluorescence microscopy, it is shown that LVFF nanofiber localization is specific to the diseased pulmonary tissue with immunofluorescence analysis demonstrating colocalization of the targeted nanofiber to RAGE in the hypoxic lung. Furthermore, biodistribution studies show that significantly more LVFF nanofibers localized to the lung compared to major off-target organs. Targeted nanofibers are retained within the pulmonary tissue for 24 h after injection. Collectively, these data demonstrate the potential of a RAGE-targeted nanomaterial as a drug delivery platform to treat pulmonary hypertension.


Asunto(s)
Hipertensión Pulmonar , Nanofibras , Animales , Hipertensión Pulmonar/tratamiento farmacológico , Pulmón , Ratones , Receptor para Productos Finales de Glicación Avanzada , Distribución Tisular
6.
Biomaterials ; 274: 120862, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33975274

RESUMEN

Smoke inhalation injury is associated with significant mortality and current therapies remain supportive. The purpose of our study was to identify proteins upregulated in the lung after smoke inhalation injury and develop peptide amphiphile nanofibers that target these proteins. We hypothesize that nanofibers targeted to angiotensin-converting enzyme or receptor for advanced glycation end products will localize to smoke-injured lungs. METHODS: Five targeting sequences were incorporated into peptide amphiphile monomers methodically to optimize nanofiber formation. Nanofiber formation was assessed by conventional transmission electron microscopy. Rats received 8 min of wood smoke. Levels of angiotensin-converting enzyme and receptor for advanced glycation end products were evaluated by immunofluorescence. Rats received the targeted nanofiber 23 h after injury via tail vein injection. Nanofiber localization was determined by fluorescence quantification. RESULTS: Peptide amphiphile purity (>95%) and nanofiber formation were confirmed. Target proteins were increased in smoke inhalation versus sham (p < 0.001). After smoke inhalation and injection of targeted nanofibers, we found a 10-fold increase in angiotensin-converting enzyme-targeted nanofiber localization to lung (p < 0.001) versus sham with minimal localization of non-targeted nanofiber (p < 0.001). CONCLUSIONS: We synthesized, characterized, and evaluated systemically delivered targeted nanofibers that localized to the site of smoke inhalation injury in vivo. Angiotensin-converting enzyme-targeted nanofibers serve as the foundation for developing a novel nanotherapeutic that treats smoke inhalation lung injury.


Asunto(s)
Nanofibras , Lesión por Inhalación de Humo , Animales , Pulmón , Péptidos , Ratas , Humo
7.
ACS Nano ; 14(6): 6649-6662, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32469498

RESUMEN

Noncompressible torso hemorrhage accounts for a significant portion of preventable trauma deaths. We report here on the development of injectable, targeted supramolecular nanotherapeutics based on peptide amphiphile (PA) molecules that are designed to target tissue factor (TF) and, therefore, selectively localize to sites of injury to slow hemorrhage. Eight TF-targeting sequences were identified, synthesized into PA molecules, coassembled with nontargeted backbone PA at various weight percentages, and characterized via circular dichroism spectroscopy, transmission electron microscopy, and X-ray scattering. Following intravenous injection in a rat liver hemorrhage model, two of these PA nanofiber coassemblies exhibited the most specific localization to the site of injury compared to controls (p < 0.05), as quantified using immunofluorescence imaging of injured liver and uninjured organs. To determine if the nanofibers were targeting TF in vivo, a mouse saphenous vein laser injury model was performed and showed that TF-targeted nanofibers colocalized with fibrin, demonstrating increased levels of nanofiber at TF-rich sites. Thromboelastograms obtained using samples of heparinized rat whole blood containing TF demonstrated that no clots were formed in the absence of TF-targeted nanofibers. Lastly, both PA nanofiber coassemblies decreased blood loss in comparison to sham and backbone nanofiber controls by 35-59% (p < 0.05). These data demonstrate an optimal TF-targeted nanofiber that localizes selectively to sites of injury and TF exposure, and, interestingly, reduces blood loss. This research represents a promising initial phase in the development of a TF-targeted injectable therapeutic to reduce preventable deaths from hemorrhage.


Asunto(s)
Nanofibras , Animales , Hemorragia/tratamiento farmacológico , Ratones , Péptidos , Ratas , Tromboplastina , Torso
8.
Nanomaterials (Basel) ; 10(3)2020 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-32121105

RESUMEN

Atherosclerosis is the leading cause of death and disability around the world, with current treatments limited by neointimal hyperplasia. Our goal was to synthesize, characterize, and evaluate an injectable, targeted nanomaterial that will specifically bind to the site of arterial injury. Our target protein is fractalkine, a chemokine involved in both neointimal hyperplasia and atherosclerosis. We showed increased fractalkine staining in rat carotid arteries 24 h following arterial injury and in the aorta of low-density lipoprotein receptor knockout (LDLR-/-) mice fed a high-fat diet for 16 weeks. Three peptide amphiphiles (PAs) were synthesized: fractalkine-targeted, scrambled, and a backbone PA. PAs were ≥90% pure on liquid chromatography/mass spectrometry (LCMS) and showed nanofiber formation on transmission electron microscopy (TEM). Rats systemically injected with fractalkine-targeted nanofibers 24 h after carotid artery balloon injury exhibited a 4.2-fold increase in fluorescence in the injured artery compared to the scrambled nanofiber (p < 0.001). No localization was observed in the non-injured artery or with the backbone nanofiber. Fluorescence of the fractalkine-targeted nanofiber increased in a dose dependent manner and was observed for up to 48 h. These data demonstrate the presence of fractalkine after arterial injury and the localization of our fractalkine-targeted nanofiber to the site of injury and serve as the foundation to develop this technology further.

9.
Macromol Biosci ; 19(6): e1900066, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31066494

RESUMEN

The rising prevalence of cardiovascular disease worldwide necessitates novel therapeutic approaches to manage atherosclerosis. Intravenously administered nanostructures are a promising noninvasive approach to deliver therapeutics that reduce plaque burden. The drug liver X receptor agonist GW3965 (LXR) can reduce atherosclerosis by promoting cholesterol efflux from plaque but causes liver toxicity when administered systemically at effective doses, thus preventing its clinical use. The ability of peptide amphiphile nanofibers containing apolipoprotein A1-derived targeting peptide 4F to serve as nanocarriers for LXR delivery (ApoA1-LXR PA) in vivo is investigated here. These nanostructures are found to successfully target atherosclerotic lesions in a mouse model within 24 h of injection. After 8 weeks of intravenous administration, the nanostructures significantly reduce plaque burden in both male and female mice to a similar extent as LXR alone in comparison to saline-treated controls. Furthermore, they do not cause increased liver toxicity in comparison to LXR treatments, which may be related to more controlled release by the nanostructure. These findings demonstrate the potential of supramolecular nanostructures as safe, effective drug nanocarriers to manage atherosclerosis.


Asunto(s)
Apolipoproteína A-I/farmacología , Aterosclerosis/tratamiento farmacológico , Receptores X del Hígado/química , Péptidos/farmacología , Animales , Apolipoproteína A-I/química , Aterosclerosis/genética , Benzoatos/efectos adversos , Benzoatos/química , Bencilaminas/efectos adversos , Bencilaminas/química , Modelos Animales de Enfermedad , Humanos , Receptores X del Hígado/genética , Receptores X del Hígado/uso terapéutico , Ratones , Terapia Molecular Dirigida , Nanofibras/química , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Péptidos/química , Tensoactivos/química , Tensoactivos/farmacología
10.
Adv Healthc Mater ; 8(3): e1801545, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30620448

RESUMEN

Nanomedicine is a promising, noninvasive approach to reduce atherosclerotic plaque burden. However, drug delivery is limited without the ability of nanocarriers to sense and respond to the diseased microenvironment. In this study, nanomaterials are developed from peptide amphiphiles (PAs) that respond to the increased levels of matrix metalloproteinases 2 and 9 (MMP2/9) or reactive oxygen species (ROS) found within the atherosclerotic niche. A pro-resolving therapeutic, Ac2-26, derived from annexin-A1 protein, is tethered to PAs using peptide linkages that cleave in response to MMP2/9 or ROS. By adjusting the molar ratios and processing conditions, the Ac2-26 PA can be co-assembled with a PA containing an apolipoprotein A1-mimetic peptide to create a targeted, therapeutic nanofiber (ApoA1-Ac226 PA). The ApoA1-Ac2-26 PAs demonstrate release of Ac2-26 within 24 h after treatment with MMP2 or ROS. The niche-responsive ApoA1-Ac2-26 PAs are cytocompatible and reduce macrophage activation from interferon gamma and lipopolysaccharide treatment, evidenced by decreased nitric oxide production. Interestingly, the linkage chemistry of ApoA1-Ac2-26 PAs significantly affects macrophage uptake and retention. Taken together, these findings demonstrate the potential of PAs to serve as an atheroma niche-responsive nanocarrier system to modulate the inflammatory microenvironment, with implications for atherosclerosis treatment.


Asunto(s)
Anexina A1 , Apolipoproteína A-I , Aterosclerosis , Portadores de Fármacos , Inmunoterapia , Nanofibras , Péptidos , Placa Aterosclerótica , Animales , Anexina A1/química , Anexina A1/farmacología , Apolipoproteína A-I/química , Apolipoproteína A-I/farmacología , Aterosclerosis/inmunología , Aterosclerosis/patología , Aterosclerosis/terapia , Línea Celular , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 2 de la Matriz/farmacología , Metaloproteinasa 9 de la Matriz/química , Metaloproteinasa 9 de la Matriz/farmacología , Ratones , Nanofibras/química , Nanofibras/uso terapéutico , Péptidos/química , Péptidos/farmacología , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/patología , Placa Aterosclerótica/terapia
11.
Cancer Immunol Res ; 6(9): 1025-1038, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29915023

RESUMEN

Antitumor T-cell responses have the potential to be curative in cancer patients, but the induction of potent T-cell immunity through vaccination remains a largely unmet goal of immunotherapy. We previously reported that the immunogenicity of peptide vaccines could be increased by maximizing delivery to lymph nodes (LNs), where T-cell responses are generated. This was achieved by conjugating the peptide to 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-PEG (DSPE-PEG) to promote albumin binding, which resulted in enhanced lymphatic drainage and improved T-cell responses. Here, we expanded upon these findings and mechanistically dissected the properties that contribute to the potency of this amphiphile-vaccine (amph-vaccine). We found that multiple linkage chemistries could be used to link peptides with DSPE-PEG, and further, that multiple albumin-binding moieties conjugated to peptide antigens enhanced LN accumulation and subsequent T-cell priming. In addition to enhancing lymphatic trafficking, DSPE-PEG conjugation increased the stability of peptides in serum. DSPE-PEG peptides trafficked beyond immediate draining LNs to reach distal nodes, with antigen presented for at least a week in vivo, whereas soluble peptide presentation quickly decayed. Responses to amph-vaccines were not altered in mice deficient in the albumin-binding neonatal Fc receptor (FcRn), but required Batf3-dependent dendritic cells (DCs). Amph-peptides were processed by human DCs equivalently to unmodified peptides. These data define design criteria for enhancing the immunogenicity of molecular vaccines to guide the design of next-generation peptide vaccines. Cancer Immunol Res; 6(9); 1025-38. ©2018 AACR.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inmunogenicidad Vacunal , Péptidos/inmunología , Linfocitos T/inmunología , Vacunas de Subunidad/inmunología , Animales , Células Dendríticas/inmunología , Femenino , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunoterapia , Ganglios Linfáticos/inmunología , Ratones Endogámicos C57BL , Fosfatidiletanolaminas/metabolismo , Polietilenglicoles/metabolismo , Receptores Fc/genética , Receptores Fc/inmunología , Albúmina Sérica/metabolismo , Vacunas de Subunidad/química
14.
Bioconjug Chem ; 22(11): 2263-70, 2011 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21950520

RESUMEN

1,2,4,5-Tetrazines have been established as effective dienes for inverse electron demand [4 + 2] Diels-Alder cycloaddition reactions with strained alkenes for over 50 years. Recently, this reaction pair combination has been applied to bioorthogonal labeling and cell detection applications; however, to date, there has been no detailed examination and optimization of tetrazines for use in biological experiments. Here, we report the synthesis and characterization of 12 conjugatable tetrazines. The tetrazines were all synthesized in a similar fashion and were screened in parallel to identify candidates most ideally suited for biological studies. In depth follow-up studies revealed compounds with varying degrees of stability and reactivity that could each be useful in different bioorthogonal applications. One promising, highly stable, and water-soluble derivative was used in pretargeted cancer cell labeling studies, confirming its utility as a bioorthogonal moiety.


Asunto(s)
Química Clic/métodos , Compuestos Orgánicos/química , Compuestos Orgánicos/síntesis química , Alquenos/química , Línea Celular Tumoral/citología , Línea Celular Tumoral/metabolismo , Humanos , Estructura Molecular , Norbornanos/química , Coloración y Etiquetado/métodos
15.
Mol Oncol ; 4(6): 511-28, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20851695

RESUMEN

Nanotechnology has enabled a renaissance in the diagnosis of cancers. This is due, in part to the ability to develop agents bearing multiple functionalities, including those utilized for targeting, imaging, and therapy, allowing for the tailoring of the properties of the nanomaterials. Whereas many nanomaterials exhibit localization to diseased tissues via intrinsic targeting, the addition of targeting ligands, such as antibodies, peptides, aptamers, and small molecules, facilitates far more sensitive cancer detection. As such, this review focuses upon some of the most poignant examples of the utility of affinity ligand targeted nanoagents in the detection of cancer.


Asunto(s)
Nanoestructuras , Neoplasias/diagnóstico , Animales , Aptámeros de Péptidos , Sistemas de Liberación de Medicamentos/métodos , Colorantes Fluorescentes/metabolismo , Humanos , Imagenología Tridimensional/métodos , Ligandos , Estructura Molecular , Neoplasias/patología , Péptidos
16.
J Inorg Biochem ; 104(3): 268-73, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20083307

RESUMEN

Selective protein tyrosine phosphatase (PTP) inhibition is often difficult to achieve owing to the high degree of similarity of the catalytic domains of this family of enzymes. Selective inhibitors of the lymphoid specific tyrosine phosphatase, LYP, are of great interest due to the involvement of LYP in several autoimmune disorders. This manuscript describes a study into the mechanistic details of selective LYP inhibition by a Au(I)-phosphine complex. The complex, [Au((CH(2)CH(2)CN)(2)PPh)Cl], selectively inhibits LYP activity both in vitro and in cells, but does not inhibit other T-cell derived PTPs including the highly homologous PTP-PEST. The mode of inhibition was probed by investigating inhibition of LYP, the LYP mutant C129/231S, and PTP-PEST. Inhibition of LYP and PTP-PEST was competitive, while the LYP double mutant appeared mixed. Wild-type LYP was inhibited more potently than LYP C129/231S, indicating an important role for at least one of these residues in Au(I) binding. Coordination of Au(I) by both the active site cysteine residue as well as either Cys129 or 231 is suggested as a potential mechanism for LYP selective inhibition.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Compuestos Orgánicos de Oro/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Cisteína/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Células Jurkat , Ligandos , Datos de Secuencia Molecular , Estructura Molecular , Compuestos Orgánicos de Oro/química , Compuestos Orgánicos de Oro/metabolismo , Conformación Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 12/química , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 22/química , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 22/metabolismo , Alineación de Secuencia
17.
J Med Chem ; 52(21): 6912-8, 2009 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-19888762

RESUMEN

Therapeutic inhibition of protein tyrosine phosphatase activity is a compelling yet challenging approach to the treatment of human disease. Toward this end, a library of 40 gold complexes with the general formula R(3)P-Au-Cl was screened to identify novel inhibitors of PTP activity. The most promising inhibitor obtained for the lymphoid tyrosine phosphatase LYP, (2-pyridine)(Ph(2))P-Au-Cl, is one of the most potent and selective LYP inhibitors identified to date with an IC(50) of 1.5 +/- 0.3 microM, 10-fold selectivity for LYP over PTP-PEST, HePTP, and CD45 in vitro, and activity in cellular studies as well.


Asunto(s)
Complejos de Coordinación/síntesis química , Oro , Compuestos Orgánicos de Oro/síntesis química , Fosfinas/síntesis química , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Animales , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Bases de Datos Factuales , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Compuestos Orgánicos de Oro/química , Compuestos Orgánicos de Oro/farmacología , Fosfinas/química , Fosfinas/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 12/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 2/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 22/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 22/metabolismo , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos , Linfocitos T/enzimología
18.
Anal Biochem ; 382(1): 63-5, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18692018

RESUMEN

A significant challenge in the field of medicinal inorganic chemistry is the identification of biological targets of metal-based drugs and the characterization of the metal-biomolecule adducts. A classic example is Au(I), which has long been used to treat rheumatoid arthritis despite a poor understanding of its biological targets due to the lability, reactivity, and "spectroscopic silence" that are characteristic of Au(I). Here, we report two qualitative methods for characterizing Au(I)-protein adducts: a thiol-reactive probe that facilitates the identification of biological cysteine-Au(I) adducts and a photoreactive Au(I) complex that produces a covalent bond between the Au(I) complex and the biomolecule.


Asunto(s)
Oro/metabolismo , Oro/uso terapéutico , Animales , Auranofina/metabolismo , Auranofina/uso terapéutico , Sitios de Unión , Bovinos , Humanos , Papaína/metabolismo , Unión Proteica , Estilbenos/metabolismo
19.
J Med Chem ; 51(15): 4790-5, 2008 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-18605719

RESUMEN

Gold(I) complexes containing N-heterocyclic carbene ligands were synthesized, characterized, and along with the antiarthritic drug, auranofin, tested as inhibitors of the cysteine-dependent protein tyrosine phosphatases, which are implicated in several disease states. These compounds exhibit potencies in the low micromolar range against the enzymes in vitro. At therapeutically relevant concentrations, all compounds inhibit PTP activity in Jurkat T leukemia cells with some selectivity. In addition, the gold-carbene compounds inhibit phosphatase activity in primary mouse thymocytes.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Compuestos de Oro/síntesis química , Compuestos de Oro/farmacología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Línea Celular , Inhibidores Enzimáticos/química , Compuestos de Oro/química , Humanos , Concentración de Iones de Hidrógeno , Estructura Molecular , Proteínas Tirosina Fosfatasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...