Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 3(18)2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30232287

RESUMO

During the last half-century, numerous antiinflammatory agents were tested in dozens of clinical trials and have proven ineffective for treating septic shock. The observation in multiple studies that cell-free hemoglobin (CFH) levels are elevated during clinical sepsis and that the degree of increase correlates with higher mortality suggests an alternative approach. Human haptoglobin binds CFH with high affinity and, therefore, can potentially reduce iron availability and oxidative activity. CFH levels are elevated over approximately 24-48 hours in our antibiotic-treated canine model of S. aureus pneumonia that simulates the cardiovascular abnormalities of human septic shock. In this 96-hour model, resuscitative treatments, mechanical ventilation, sedation, and continuous care are translatable to management in human intensive care units. We found, in this S. aureus pneumonia model inducing septic shock, that commercial human haptoglobin concentrate infusions over 48-hours bind canine CFH, increase CFH clearance, and lower circulating iron. Over the 96-hour study, this treatment was associated with an improved metabolic profile (pH, lactate), less lung injury, reversal of shock, and increased survival. Haptoglobin binding compartmentalized CFH to the intravascular space. This observation, in combination with increasing CFHs clearance, reduced available iron as a potential source of bacterial nutrition while decreasing the ability for CFH and iron to cause extravascular oxidative tissue injury. In contrast, haptoglobin therapy had no measurable antiinflammatory effect on elevations in proinflammatory C-reactive protein and cytokine levels. Haptoglobin therapy enhances normal host defense mechanisms in contrast to previously studied antiinflammatory sepsis therapies, making it a biologically plausible novel approach to treat septic shock.


Assuntos
Haptoglobinas/farmacologia , Lesão Pulmonar/tratamento farmacológico , Pneumonia/tratamento farmacológico , Choque Séptico/tratamento farmacológico , Animais , Antibacterianos , Anti-Inflamatórios/farmacologia , Gasometria , Anormalidades Cardiovasculares , Citocinas , Modelos Animais de Doenças , Cães , Haptoglobinas/uso terapêutico , Hematócrito , Humanos , Imunidade Inata , Ferro , Estimativa de Kaplan-Meier , Pneumonia/microbiologia , Pneumonia/mortalidade , Artéria Pulmonar , Staphylococcus aureus
2.
Mol Pharmacol ; 91(2): 75-86, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27895162

RESUMO

Cannabinoid receptor interacting protein 1a (CRIP1a) is a CB1 receptor (CB1R) distal C-terminal-associated protein that alters CB1R interactions with G-proteins. We tested the hypothesis that CRIP1a is capable of also altering CB1R interactions with ß-arrestin proteins that interact with the CB1R at the C-terminus. Coimmunoprecipitation studies indicated that CB1R associates in complexes with either CRIP1a or ß-arrestin, but CRIP1a and ß-arrestin fail to coimmunoprecipitate with each other. This suggests a competition for CRIP1a and ß-arrestin binding to the CB1R, which we hypothesized could attenuate the action of ß-arrestin to mediate CB1R internalization. We determined that agonist-mediated reduction of the density of cell surface endogenously expressed CB1Rs was clathrin and dynamin dependent and could be modeled as agonist-induced aggregation of transiently expressed GFP-CB1R. CRIP1a overexpression attenuated CP55940-mediated GFP-CB1R as well as endogenous ß-arrestin redistribution to punctae, and conversely, CRIP1a knockdown augmented ß-arrestin redistribution to punctae. Peptides mimicking the CB1R C-terminus could bind to both CRIP1a in cell extracts as well as purified recombinant CRIP1a. Affinity pull-down studies revealed that phosphorylation at threonine-468 of a CB1R distal C-terminus 14-mer peptide reduced CB1R-CRIP1a association. Coimmunoprecipitation of CB1R protein complexes demonstrated that central or distal C-terminal peptides competed for the CB1R association with CRIP1a, but that a phosphorylated central C-terminal peptide competed for association with ß-arrestin 1, and phosphorylated central or distal C-terminal peptides competed for association with ß-arrestin 2. Thus, CRIP1a can compete with ß-arrestins for interaction with C-terminal CB1R domains that could affect agonist-driven, ß-arrestin-mediated internalization of the CB1R.


Assuntos
Proteínas de Transporte/metabolismo , Receptor CB1 de Canabinoide/metabolismo , beta-Arrestinas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas de Membrana , Peptídeos/química , Fosforilação , Ligação Proteica , Ratos
3.
Theranostics ; 6(5): 698-709, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27022417

RESUMO

The theranostic potential of (225)Ac-based radiopharmaceuticals continues to increase as researchers seek innovative ways to harness the nuclear decay of this radioisotope for therapeutic and imaging applications. This communication describes the evaluation of (225)Ac-DOTA-c(RGDyK) in both biodistribution and Cerenkov luminescence imaging (CLI) studies. Initially, La-DOTA-c(RGDyK) was prepared as a non-radioactive surrogate to evaluate methodologies that would contribute to an optimized radiochemical synthetic strategy and estimate the radioactive conjugate's affinity for αvß3, using surface plasmon resonance spectroscopy. Surface plasmon resonance spectroscopy studies revealed the IC50 and Ki of La-DOTA-c(RGDyK) to be 33 ± 13 nM and 26 ± 11 nM, respectively, and suggest that the complexation of the La(3+) ion to the conjugate did not significantly alter integrin binding. Furthermore, use of this surrogate allowed optimization of radiochemical synthesis strategies to prepare (225)Ac-DOTA-c(RGDyK) with high radiochemical purity and specific activity similar to other (225)Ac-based radiopharmaceuticals. This radiopharmaceutical was highly stable in vitro. In vivo biodistribution studies confirmed the radiotracer's ability to target αvß3 integrin with specificity; specificity was detected in tumor-bearing animals using Cerenkov luminescence imaging. Furthermore, tumor growth control was achieved using non-toxic doses of the radiopharmaceutical in U87mg tumor-bearing nude mice. To our knowledge, this is the first report to describe the CLI of αvß3 (+) tumors in live animals using the daughter products derived from (225)Ac decay in situ. This concept holds promise to further enhance development of targeted alpha particle therapy.


Assuntos
Complexos de Coordenação/farmacocinética , Complexos de Coordenação/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/radioterapia , Imagem Óptica/métodos , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/uso terapêutico , Animais , Integrina alfaVbeta3/metabolismo , Camundongos Nus , Resultado do Tratamento
4.
PLoS One ; 10(7): e0132668, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26172836

RESUMO

Ends of human chromosomes consist of the six nucleotide repeat d[pTTAGGG]n known as telomeric DNA, which protects chromosomes. We have previously shown that the DHX36 gene product, G4 Resolvase 1 (G4R1), binds parallel G-quadruplex (G4) DNA with an unusually tight apparent Kd. Recent work associates G4R1 with the telomerase holoenzyme, which may allow it to access telomeric G4-DNA. Here we show that G4R1 can tightly bind telomeric G4-DNA, and in the context of the telomeric sequence, we determine length, sequence, and structural requirements sufficient for tight G4R1 telomeric binding. Specifically, G4R1 binds telomeric DNA in the K+-induced "3+1" G4-topology with an apparent Kd = 10 ± 1.9 pM, a value similar as previously found for binding to unimolecular parallel G4-DNA. G4R1 binds to the Na+-induced "2+2" basket G4-structure formed by the same DNA sequence with an apparent Kd = 71 ± 2.2 pM. While the minimal G4-structure is not sufficient for G4R1 binding, a 5' G4-structure with a 3' unstructured tail containing a guanine flanked by adenine(s) is sufficient for maximal binding. Mutations directed to disrupt G4-structure similarly disrupt G4R1 binding; secondary mutations that restore G4-structure also restore G4R1 binding. We present a model showing that a replication fork disrupting a T-loop could create a 5' quadruplex with an opened 3'tail structure that is recognized by G4R1.


Assuntos
RNA Helicases DEAD-box/metabolismo , DNA/química , DNA/metabolismo , Quadruplex G , Sequência de Bases , Sítios de Ligação/genética , Dicroísmo Circular , DNA/genética , Humanos , Cinética , Modelos Moleculares , Mutação , Potássio/metabolismo , Sequências Repetitivas de Ácido Nucleico , Sódio/metabolismo , Telômero/química , Telômero/genética , Telômero/metabolismo
5.
Atherosclerosis ; 238(2): 231-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25528432

RESUMO

Oleic acid consumption is considered cardio-protective according to studies conducted examining effects of the Mediterranean diet. However, animal models have shown that oleic acid consumption increases LDL particle cholesteryl oleate content which is associated with increased LDL-proteoglycan binding and atherosclerosis. The objective was to examine effects of varying oleic, linoleic and docosahexaenoic acid consumption on human LDL-proteoglycan binding in a non-random subset of the Canola Oil Multi-center Intervention Trial (COMIT) participants. COMIT employed a randomized, double-blind, five-period, cross-over trial design. Three of the treatment oil diets: 1) a blend of corn/safflower oil (25:75); 2) high oleic canola oil; and 3) DHA-enriched high oleic canola oil were selected for analysis of LDL-proteoglycan binding in 50 participants exhibiting good compliance. LDL particles were isolated from frozen plasma by gel filtration chromatography and LDL cholesteryl esters quantified by mass-spectrometry. LDL-proteoglycan binding was assessed using surface plasmon resonance. LDL particle cholesterol ester fatty acid composition was sensitive to the treatment fatty acid compositions, with the main fatty acids in the treatments increasing in the LDL cholesterol esters. The corn/safflower oil and high-oleic canola oil diets lowered LDL-proteoglycan binding relative to their baseline values (p = 0.0005 and p = 0.0012, respectively). At endpoint, high-oleic canola oil feeding resulted in lower LDL-proteoglycan binding than corn/safflower oil (p = 0.0243) and DHA-enriched high oleic canola oil (p = 0.0249), although high-oleic canola oil had the lowest binding at baseline (p = 0.0344). Our findings suggest that high-oleic canola oil consumption in humans increases cholesteryl oleate percentage in LDL, but in a manner not associated with a rise in LDL-proteoglycan binding.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Ésteres do Colesterol/sangue , LDL-Colesterol/sangue , Dieta Mediterrânea , Gorduras na Dieta/administração & dosagem , Ácidos Graxos Monoinsaturados/administração & dosagem , Ácido Oleico/administração & dosagem , Proteoglicanas/sangue , Adulto , Canadá , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/diagnóstico , Óleo de Milho/administração & dosagem , Estudos Cross-Over , Ácidos Docosa-Hexaenoicos/administração & dosagem , Método Duplo-Cego , Feminino , Humanos , Ácido Linoleico/administração & dosagem , Masculino , Pessoa de Meia-Idade , Óleo de Brassica napus , Fatores de Risco , Comportamento de Redução do Risco , Óleo de Cártamo/administração & dosagem , Fatores de Tempo , Estados Unidos
6.
Nanomedicine ; 10(2): 451-61, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23988714

RESUMO

DNA and porphyrin based therapeutics are important for anti-cancer treatment. The present studies demonstrate single-stranded DNA (ssDNA) assembles with meso-tetra-4-pyridyl porphine (MTP) forming porphyrin:DNA nano-complexes (PDN) that are stable in aqueous solution under physiologically relevant conditions and undergo dissociation with DNA release in hydrophobic environments, including cell membranes. PDN formation is DNA-dependent with the ratio of porphyrin:DNA being approximately two DNA nucleobases per porphyrin. PDN produce reactive oxygen species (ROS) in a light-dependent manner under conditions that favor nano-complex dissociation in the presence of hydrophobic solvents. PDN induce light-dependent cytotoxicity in vitro and anti-tumor activity towards bladder cancer xenografts in vivo. Light-dependent, PDN-mediated cell death results from ROS-mediated localized membrane damage due to lipid peroxidation with mass spectrometry indicating the generation of the lipid peroxidation products 9- and 13-hydroxy octadecanoic acid. Our results demonstrate that PDN have properties useful for therapeutic applications, including cancer treatment. FROM THE CLINICAL EDITOR: In this study, porphyrin-DNA nanocomplexes were investigated as anti-cancer therapeutics inducing ROS production in a light-dependent manner. Efficacy is demonstrated in vitro as well as a in a bladder cancer xenograft model.


Assuntos
Antineoplásicos/química , DNA de Cadeia Simples/química , Neoplasias/tratamento farmacológico , Porfirinas/química , Animais , Morte Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Endossomos , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Peroxidação de Lipídeos , Camundongos , Camundongos Nus , Nanomedicina , Transplante de Neoplasias , Fármacos Fotossensibilizantes/química , Espécies Reativas de Oxigênio , Ácidos Esteáricos/química , Neoplasias da Bexiga Urinária/terapia
7.
Nano Life ; 3(1): 13400051-134000511, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098311

RESUMO

The interaction between platelet integrin αIIbß3 and fibrin(ogen) plays a key role in blood clot formation and stability. Integrin antagonists, a class of pharmaceuticals used to prevent and treat cardiovascular disease, are designed to competitively interfere with this process. However, the energetics of the integrin-drug binding are not fully understood, potentially hampering further development of this class of pharmaceuticals. We integrated dynamic force spectroscopy (DFS) and surface plasmon resonance (SPR) to probe the energetics of complex formation between αIIbß3 and cHarGD, a cyclic peptide integrin antagonist. Analysis of αIIbß3:cHarGD DFS rupture force data at pulling rates of 14 000 pN/s, 42 000 pN/s and 70 000 pN/s yielded koff = 0.02-0.09 s-1, a dissociation energy barrier [Formula: see text] = 22-29 kJ/mol, and a potential well width x-1 = 0.5-0.8 nm. SPR kinetic data yielded an association rate constant kon = 7 × 103 L/mol-s and a dissociation rate constant koff = 10-2 s-1, followed by a slower stabilization step (τ ~ 400 s). Both DFS and SPR detected minimal interactions between αIIbß3 and cHarGA demonstrating a key role for electrostatic interactions between the ligand aspartate and the integrin metal ion-dependent adhesion site (MIDAS). Our work provides new insights into the energy landscape of αIIbß3's interactions with pharmacological and physiological ligands.

8.
J Lipid Res ; 54(9): 2495-503, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23804810

RESUMO

Several studies in humans and animals suggest that LDL particle core enrichment in cholesteryl oleate (CO) is associated with increased atherosclerosis. Diet enrichment with MUFAs enhances LDL CO content. Steroyl O-acyltransferase 2 (SOAT2) is the enzyme that catalyzes the synthesis of much of the CO found in LDL, and gene deletion of SOAT2 minimizes CO in LDL and protects against atherosclerosis. The purpose of this study was to test the hypothesis that the increased atherosclerosis associated with LDL core enrichment in CO results from an increased affinity of the LDL particle for arterial proteoglycans. ApoB-100-only Ldlr(-/-) mice with and without Soat2 gene deletions were fed diets enriched in either cis-MUFA or n-3 PUFA, and LDL particles were isolated. LDL:proteogylcan binding was measured using surface plasmon resonance. Particles with higher CO content consistently bound with higher affinity to human biglycan and the amount of binding was shown to be proportional to the extent of atherosclerosis of the LDL donor mice. The data strongly support the thesis that atherosclerosis was induced through enhanced proteoglycan binding of LDL resulting from LDL core CO enrichment.


Assuntos
Aterosclerose/metabolismo , Ésteres do Colesterol/metabolismo , LDL-Colesterol/metabolismo , Proteoglicanas/metabolismo , Ressonância de Plasmônio de Superfície/métodos , Animais , Artérias/metabolismo , Biglicano/metabolismo , Humanos , Camundongos
9.
J Biol Chem ; 288(25): 18448-57, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23661698

RESUMO

The EphA2 receptor tyrosine kinase is overexpressed in a number of malignancies and is activated by ephrin ligands, most commonly by ephrin-A1. The crystal structure of the ligand-receptor complex revealed a glycosylation on the Asn-26 of ephrin-A1. Here we report for the first time the significance of the glycosylation in the biology of EphA2 and ephrin-A1. Ephrin-A1 was enzymatically deglycosylated, and its activity was evaluated in several assays using glioblastoma (GBM) cells and recombinant EphA2. We found that deglycosylated ephrin-A1 does not efficiently induce EphA2 receptor internalization and degradation, and does not activate the downstream signaling pathways involved in cell migration and proliferation. Data obtained by surface plasmon resonance confirms that deglycosylated ephrin-A1 does not bind EphA2 with high affinity. Mutations in the glycosylation site on ephrin-A1 result in protein aggregation and mislocalization. Analysis of Eph/ephrin crystal structures reveals an interaction between the ligand's carbohydrates and two residues of EphA2: Asp-78 and Lys-136. These findings suggest that the glycosylation on ephrin-A1 plays a critical role in the binding and activation of the EphA2 receptor.


Assuntos
Efrina-A1/metabolismo , Receptor EphA2/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Sítios de Ligação/genética , Ligação Competitiva , Western Blotting , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cristalografia por Raios X , Efrina-A1/química , Efrina-A1/genética , Efrina-A2/genética , Efrina-A2/metabolismo , Glicosilação , Humanos , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Receptor EphA2/química , Receptor EphA2/genética , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície
10.
Biomaterials ; 34(10): 2492-500, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23332318

RESUMO

Uncontrolled bleeding continues to be one of the leading causes of death in individuals following traumatic injury. Prognosis is worsened with the onset of acute coagulopathy characterized by metabolic acidosis, hypothermia and hemodilution, which consequently perpetuates blood loss and increases mortality. While there are several limitations to biomaterials employed as hemostatic agents, keratin biomaterials have demonstrated efficacy in mitigating blood loss in an animal model of hemorrhage in prior studies. Here we investigate the hypothesis that keratins actively participate in coagulation and that a potential mechanism of action is independent of temperature and dilution of clotting factors. Data from this study show that keratins appear to contribute to hemostasis by significantly decreasing plasma clotting lag times and are able to maintain activity under simulated conditions of coagulopathy. Moreover, a system of isolated fibrin polymerization provided evidence of increased fibril lateral assembly in the presence of keratin. The data provided here provides a platform for further development of keratin biomaterials as hemostatic agents.


Assuntos
Materiais Biocompatíveis/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Hemostasia/efeitos dos fármacos , Queratinas/farmacologia , Materiais Biocompatíveis/química , Células Cultivadas , Humanos , Queratinas/química , Microscopia Eletrônica de Varredura
11.
Biomaterials ; 34(11): 2632-40, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23340195

RESUMO

Driven by new discoveries in stem-cell biology and regenerative medicine, there is broad interest in biomaterials that go beyond basic interactions with cells and tissues to actively direct and sustain cellular behavior. Keratin biomaterials have the potential to achieve these goals but have been inadequately described in terms of composition, structure, and cell-instructive characteristics. In this manuscript we describe and characterize a keratin-based biomaterial, demonstrate self-assembly of cross-linked hydrogels, investigate a cell-specific interaction that is dependent on the hydrogel structure and mediated by specific biomaterial-receptor interactions, and show one potential medical application that relies on receptor binding - the ability to achieve hemostasis in a lethal liver injury model. Keratin biomaterials represent a significant advance in biotechnology as they combine the compatibility of natural materials with the chemical flexibility of synthetic materials. These characteristics allow for a system that can be formulated into several varieties of cell-instructive biomaterials with potential uses in tissue engineering, regenerative medicine, drug and cell delivery, and trauma.


Assuntos
Materiais Biocompatíveis/química , Cabelo/química , Hemostáticos/metabolismo , Hidrogéis/química , Queratinas/química , Animais , Materiais Biocompatíveis/análise , Plaquetas/citologia , Plaquetas/metabolismo , Western Blotting , Adesão Celular , Colorimetria , Eletroforese , Hemostasia , Humanos , Hidrogéis/análise , Queratinas/análise , Espectrometria de Massas , Microscopia Confocal , Microscopia Eletrônica de Varredura , Medicina Regenerativa/métodos , Reologia , Suínos , Engenharia Tecidual/métodos
12.
Neuro Oncol ; 14(10): 1239-53, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22952195

RESUMO

Our objective was to exploit a novel ligand-based delivery system for targeting diagnostic and therapeutic agents to cancers that express interleukin 13 receptor alpha 2 (IL13Rα2), a tumor-restricted plasma membrane receptor overexpressed in glioblastoma multiforme (GBM), meningiomas, peripheral nerve sheath tumors, and other peripheral tumors. On the basis of our prior work, we designed a novel IL13Rα2-targeted quadruple mutant of IL13 (TQM13) to selectively bind the tumor-restricted IL13Rα2 with high affinity but not significantly interact with the physiologically abundant IL13Rα1/IL4Rα heterodimer that is also expressed in normal brain. We then assessed the in vitro binding profile of TQM13 and its potential to deliver diagnostic and therapeutic radioactivity in vivo. Surface plasmon resonance (SPR; Biacore) binding experiments demonstrated that TQM13 bound strongly to recombinant IL13Rα2 (Kd∼5 nM). In addition, radiolabeled TQM13 specifically bound IL13Rα2-expressing GBM cells and specimens but not normal brain. Of importance, TQM13 did not functionally activate IL13Rα1/IL4Rα in cells or bind to it in SPR binding assays, in contrast to wtIL13. Furthermore, in vivo targeting of systemically delivered radiolabeled TQM13 to IL13Rα2-expressing subcutaneous tumors was demonstrated and confirmed non-invasively for the first time with 124I-TQM13 positron emission tomography imaging. In addition, 131I-TQM13 demonstrated in vivo efficacy against subcutaneous IL13Rα2-expressing GBM tumors and in an orthotopic synergeic IL13Rα2-positive murine glioma model, as evidenced by statistically significant survival advantage. Our results demonstrate that we have successfully generated an optimized biomarker-targeted scaffolding that exhibited specific binding activity toward the tumor-associated IL13Rα2 in vitro and potential to deliver diagnostic and therapeutic payloads in vivo.


Assuntos
Neoplasias Encefálicas/metabolismo , Proliferação de Células , Glioblastoma/metabolismo , Subunidade alfa1 de Receptor de Interleucina-13/metabolismo , Subunidade alfa2 de Receptor de Interleucina-13/metabolismo , Interleucina-13/genética , Animais , Sítios de Ligação , Western Blotting , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Cromatografia em Camada Fina , Dicroísmo Circular , Ensaio de Desvio de Mobilidade Eletroforética , Glioblastoma/patologia , Humanos , Processamento de Imagem Assistida por Computador , Técnicas Imunoenzimáticas , Interleucina-13/metabolismo , Ligantes , Masculino , Camundongos , Camundongos Nus , Mutagênese Sítio-Dirigida , Mutação/genética , Tomografia por Emissão de Pósitrons , Ligação Proteica , Proteínas Recombinantes , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Biochemistry ; 51(12): 2526-38, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22397628

RESUMO

Our previous studies revealed that the fibrinogen αC-domains undergo conformational changes and adopt a physiologically active conformation upon their self-association into αC polymers in fibrin. In the present study, we analyzed the mechanism of αC polymer formation and tested our hypothesis that self-association of the αC-domains occurs through the interaction between their N-terminal subdomains and may include ß-hairpin swapping. Our binding experiments performed by size-exclusion chromatography and optical trap-based force spectroscopy revealed that the αC-domains self-associate exclusively through their N-terminal subdomains, while their C-terminal subdomains were found to interact with the αC-connectors that tether the αC-domains to the bulk of the molecule. This interaction should reinforce the structure of αC polymers and provide the proper orientation of their reactive residues for efficient cross-linking by factor XIIIa. Molecular modeling of self-association of the N-terminal subdomains confirmed that the hypothesized ß-hairpin swapping does not impose any steric hindrance. To "freeze" the conformation of the N-terminal subdomain and prevent the hypothesized ß-hairpin swapping, we introduced by site-directed mutagenesis an extra disulfide bond between two ß-hairpins of the bovine Aα406-483 fragment corresponding to this subdomain. The experiments performed by circular dichroism revealed that Aα406-483 mutant containing Lys429Cys/Thr463Cys mutations preserved its ß-sheet structure. However, in contrast to wild-type Aα406-483, this mutant had lower tendency for oligomerization, and its structure was not stabilized upon oligomerization, in agreement with the above hypothesis. On the basis of the results obtained and our previous findings, we propose a model of fibrin αC polymer structure and molecular mechanism of assembly.


Assuntos
Fibrina/química , Fibrinogênio/química , Fragmentos de Peptídeos/química , Multimerização Proteica , Animais , Bovinos , Dissulfetos/química , Fibrinogênio/genética , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Fragmentos de Peptídeos/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
14.
J Biol Chem ; 287(17): 14012-22, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22362770

RESUMO

The EphA2 receptor is overexpressed in glioblastoma multiforme and has been to shown to contribute to cell transformation, tumor initiation, progression, and maintenance. EphrinA1 (eA1) is a preferred ligand for the receptor. Treatment with monomeric eA1, the form of eA1 found in the extracellular environment, causes receptor phosphorylation, internalization, and down-regulation with subsequent anti-tumor effects. Here, we investigated the structure-function relationship of a monomeric eA1 focusing on its G-H loop ((108)FQRFTPFTLGKEFKE(123)G), a highly conserved region among eAs that mediates binding to their receptors. Alanine substitution mutants of the G-H loop amino acids were transfected into U-251 MG glioblastoma multiforme cells, and functional activity of each mutant in conditioned media was assessed by EphA2 down-regulation, ERK and AKT activation and cellular response assays. Alanine substitutions at positions Pro-113 Thr-115, Gly-117, Glu-122, and also Gln-109 enhanced the EphA2 receptor down-regulation and decreased p-ERK and p-AKT. Substitution mutants of eA1 at positions Phe-108, Arg-110, Phe-111, Thr-112, Phe-114, Leu-116, Lys-118, Glu-119, and Phe-120 had a deleterious effect on EphA2 down-regulation when compared with eA1-WT. Mutants at positions Phe-108, Lys-18, Lys-121, Gly-123 retained similar properties to eA1-WT. Recombinant eA1-R110A, -T115A, -G117A, and -F120A have been found to exhibit the same characteristics as the ligands contained in the conditioned media mainly due to the differences in their binding to the receptor. Thus, we have identified variants of eA1 that possess either superagonistic or antagonistic properties. These new findings will be important in the understanding of the receptor/ligand interactions and in further design of anti-cancer therapies targeting the eA/EphA system.


Assuntos
Efrina-A1/química , Regulação da Expressão Gênica , Receptor EphA2/química , Alanina/química , Antineoplásicos/farmacologia , Sítios de Ligação , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Humanos , Ligantes , Dados de Sequência Molecular , Mutagênese , Ligação Proteica , Conformação Proteica , Isoformas de Proteínas , Estrutura Secundária de Proteína , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície
15.
PLoS One ; 6(10): e26021, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21998747

RESUMO

BACKGROUND: 4-hydroxy-2-oxoglutarate (HOG) aldolase is a unique enzyme in the hydroxyproline degradation pathway catalyzing the cleavage of HOG to pyruvate and glyoxylate. Mutations in this enzyme are believed to be associated with the excessive production of oxalate in primary hyperoxaluria type 3 (PH3), although no experimental data is available to support this hypothesis. Moreover, the identity, oligomeric state, enzymatic activity, and crystal structure of human HOGA have not been experimentally determined. METHODOLOGY/PRINCIPAL FINDINGS: In this study human HOGA (hHOGA) was identified by mass spectrometry of the mitochondrial enzyme purified from bovine kidney. hHOGA performs a retro-aldol cleavage reaction reminiscent of the trimeric 2-keto-3-deoxy-6-phosphogluconate aldolases. Sequence comparisons, however, show that HOGA is related to the tetrameric, bacterial dihydrodipicolinate synthases, but the reaction direction is reversed. The 1.97 Å resolution crystal structure of hHOGA bound to pyruvate was determined and enabled the modeling of the HOG-Schiff base intermediate and the identification of active site residues. Kinetic analyses of site-directed mutants support the importance of Lys196 as the nucleophile, Tyr168 and Ser77 as components of a proton relay, and Asn78 and Ser198 as unique residues that facilitate substrate binding. CONCLUSIONS/SIGNIFICANCE: The biochemical and structural data presented support that hHOGA utilizes a type I aldolase reaction mechanism, but employs novel residue interactions for substrate binding. A mapping of the PH3 mutations identifies potential rearrangements in either the active site or the tetrameric assembly that would likely cause a loss in activity. Altogether, these data establish a foundation to assess mutant forms of hHOGA and how their activity could be pharmacologically restored.


Assuntos
Hidroxiprolina/metabolismo , Hiperoxalúria Primária/metabolismo , Oxo-Ácido-Liases/química , Oxo-Ácido-Liases/metabolismo , Sequência de Aminoácidos , Animais , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/isolamento & purificação , Apoenzimas/metabolismo , Bactérias/enzimologia , Domínio Catalítico , Bovinos , Cristalografia por Raios X , Humanos , Hiperoxalúria Primária/genética , Hiperoxalúria Primária/patologia , Rim/patologia , Espectrometria de Massas , Mitocôndrias/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Oxo-Ácido-Liases/genética , Oxo-Ácido-Liases/isolamento & purificação , Multimerização Proteica , Estrutura Quaternária de Proteína , Ácido Pirúvico/metabolismo , Bases de Schiff/metabolismo , Soluções , Especificidade por Substrato
16.
Nucleic Acids Res ; 39(16): 7161-78, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21586581

RESUMO

It has been previously shown that the DHX36 gene product, G4R1/RHAU, tightly binds tetramolecular G4-DNA with high affinity and resolves these structures into single strands. Here, we test the ability of G4R1/RHAU to bind and unwind unimolecular G4-DNA. Gel mobility shift assays were used to measure the binding affinity of G4R1/RHAU for unimolecular G4-DNA-formed sequences from the Zic1 gene and the c-Myc promoter. Extremely tight binding produced apparent K(d)'s of 6, 3 and 4 pM for two Zic1 G4-DNAs and a c-Myc G4-DNA, respectively. The low enzyme concentrations required for measuring these K(d)'s limit the precision of their determination to upper boundary estimates. Similar tight binding was not observed in control non-G4 forming DNA sequences or in single-stranded DNA having guanine-rich runs capable of forming tetramolecular G4-DNA. Using a peptide nucleic acid (PNA) trap assay, we show that G4R1/RHAU catalyzes unwinding of unimolecular Zic1 G4-DNA into an unstructured state capable of hybridizing to a complementary PNA. Binding was independent of adenosine triphosphate (ATP), but the PNA trap assay showed that unwinding of G4-DNA was ATP dependent. Competition studies indicated that unimolecular Zic1 and c-Myc G4-DNA structures inhibit G4R1/RHAU-catalyzed resolution of tetramolecular G4-DNA. This report provides evidence that G4R1/RHAU tightly binds and unwinds unimolecular G4-DNA structures.


Assuntos
RNA Helicases DEAD-box/metabolismo , DNA/química , Quadruplex G , Dicroísmo Circular , DNA/metabolismo , Genes myc , Humanos , Hibridização de Ácido Nucleico , Oligodesoxirribonucleotídeos/química , Ácidos Nucleicos Peptídicos/química , Recombinases/metabolismo
17.
Biochemistry ; 49(43): 9217-25, 2010 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-20828133

RESUMO

This study demonstrates that two orthogonal events regulate integrin αIIbß3's interactions with fibrinogen, its primary physiological ligand: (1) conformational changes at the αIIb-ß3 interface and (2) flexibility in the carboxy terminus of fibrinogen's γ-module. The first postulate was tested by capturing αIIbß3 on a biosensor and measuring binding by surface plasmon resonance. Binding of fibrinogen to eptifibatide-primed αIIbß3 was characterized by a k(on) of ~2 × 10(4) L mol(-1) s(-1) and a k(off) of ~8 × 10(-5) s(-1) at 37 °C. In contrast, even at 150 nM fibrinogen, no binding was detected with resting αIIbß3. Eptifibatide competitively inhibited fibrinogen's interactions with primed αIIbß3 (K(i) ~0.4 nM), while a synthetic γ-module peptide (HHLGGAKQAGDV) was only weakly inhibitory (K(i) > 10 µM). The second postulate was tested by measuring αIIbß3's interactions with recombinant fibrinogen, both normal (rFgn) and a deletion mutant lacking the γ-chain AGDV sites (rFgn γΔ408-411). Normal rFgn bound rapidly, tightly, and specifically to primed αIIbß3; no interaction was detected with rFgn γΔ408-411. Equilibrium and transition-state thermodynamic data indicated that binding of fibrinogen to primed αIIbß3, while enthalpy-favorable, must overcome an entropy-dominated activation energy barrier. The hypothesis that fibrinogen binding is enthalpy-driven fits with structural data showing that its γ-C peptide and eptifibatide exhibit comparable electrostatic contacts with αIIbß3's ectodomain. The concept that fibrinogen's αIIbß3 targeting sequence is intrinsically disordered may explain the entropy penalty that limits its binding rate. In the hemostatic milieu, platelet-platelet interactions may be localized to vascular injury sites because integrins must be activated before they can bind their most abundant ligand.


Assuntos
Fibrinogênio/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Entropia , Eptifibatida , Fibrinogênio/metabolismo , Hemostáticos , Humanos , Peptídeos/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ligação Proteica , Conformação Proteica , Ressonância de Plasmônio de Superfície , Termodinâmica
18.
Biochemistry ; 48(51): 12191-201, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-19928926

RESUMO

Our recent study established the NMR structure of the recombinant bAalpha406-483 fragment corresponding to the NH(2)-terminal half of the bovine fibrinogen alphaC-domain and revealed that at increasing concentrations this fragment forms oligomers (self-associates). The major goals of the study presented here were to determine the structure and self-association of the full-length human fibrinogen alphaC-domains. To accomplish these goals, we prepared a recombinant human fragment, hAalpha425-503, homologous to bovine bAalpha406-483, and demonstrated using NMR, CD, and size-exclusion chromatography that its overall fold and ability to form oligomers are similar to those of bAalpha406-483. We also prepared recombinant hAalpha392-610 and bAalpha374-568 fragments corresponding to the full-length human and bovine alphaC-domains, respectively, and tested their structure, stability, and ability to self-associate. Size-exclusion chromatography revealed that both fragments form reversible oligomers in a concentration-dependent manner. Their oligomerization was confirmed in sedimentation equilibrium experiments, which also established the self-association affinities of these fragments and revealed that the addition of each monomer to assembling alphaC-oligomers substantially increases the stabilizing free energy. In agreement, unfolding experiments monitored by CD established that self-association of both fragments results in a significant increase in their thermal stability. Analysis of CD spectra of both fragments revealed that alphaC self-association results in an increase in the level of regular structure, implying that the COOH-terminal half of the alphaC-domain adopts an ordered conformation in alphaC-oligomers and that this domain contains two independently folded subdomains. Altogether, these data further clarify the structure of the human and bovine alphaC-domains and the molecular mechanism of their self-association into alphaC-polymers in fibrin.


Assuntos
Fibrinogênio/química , Animais , Bovinos , Dicroísmo Circular , Fibrinogênio/genética , Fibrinogênio/isolamento & purificação , Fibrinogênio/metabolismo , Humanos , Cinética , Ligação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína
19.
Biochemistry ; 48(35): 8355-65, 2009 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-19640007

RESUMO

This investigation addressed the paradox that disintegrins and small RGD-ligands readily bind to the resting alphaIIbbeta3 integrin, while macromolecules with similar integrin recognition motifs require an activated, or primed, receptor. Three structurally similar pharmaceutical integrin antagonists (eptifibatide, tirofiban, and roxifiban) were each incubated with resting alphaIIbbeta3; after drug wash-out, the receptor's ability to recognize PAC-1, an activation-dependent IgM with an RYD integrin-targeting site was measured. Their promotion of PAC-1:alphaIIbbeta3 binding (solid phase assay), eptifibatide > tirofiban > roxifiban, correlated with their ability to shift the receptor to an open conformer, as measured by analytical ultracentrifugation. Surface plasmon resonance (SPR) demonstrated that PAC-1 bound rapidly (k(on) approximately 5 x 10(5) l/mol-s, 25 degrees C) and tightly (Kd approximately 1 nM) to eptifibatide-primed integrins, captured on a biosensor using an IgG specific for alphaIIb's cytoplasmic domain. Varying the interval between integrin capture and antagonist dissociation indicated that transiently primed alphaIIbbeta3 retains the ability to rapidly bind PAC-1 from 2-90 min, although the dissociation rate increased at later times, indicative of a weakening of the complex. Fluorescence anisotropy (fluorophore-tagged analogue exchange assay) demonstrated that eptifibatide dissociates rapidly from alphaIIbbeta3 (half-time <2 min), consistent with the priming window determined by SPR. van't Hoff analysis of alphaIIbbeta3:PAC-1's temperature-dependent Kd indicated entropy/enthalpy compensation, similar to (resting) integrin binding to the disintegrin echistatin. Eyring analysis of k(on) yielded DeltaG degrees approximately 10 kcal/mol for PAC-1 binding to primed alphaIIbbeta3, 3 kcal/mol lower than that of echistatin. These observations suggest that priming lowers the transition-state energy barrier, enabling rapid macromolecular ligand binding to activated integrins. Recognizing the limitations in extrapolating from laboratory to pathophysiological conditions, we propose that similar priming mechanisms may contribute to the unexpected platelet-activating effects of pharmaceutical integrin antagonists.


Assuntos
Fosfatase 2 de Especificidade Dupla/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Amidinas/química , Amidinas/farmacologia , Eptifibatida , Integrinas/química , Integrinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Isoxazóis/química , Isoxazóis/farmacologia , Modelos Moleculares , Peptídeos/química , Peptídeos/farmacologia , Inibidores da Agregação Plaquetária/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Ligação Proteica , Termodinâmica , Tirofibana , Tirosina/análogos & derivados , Tirosina/química , Tirosina/farmacologia
20.
Clin Vaccine Immunol ; 16(1): 21-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18987167

RESUMO

A number of studies have clearly demonstrated that flagellin is a potent adjuvant that promotes robust immune responses when it is given with a protein antigen. In view of the potential biological and practical benefits of a recombinant protein vaccine composed of a single fusion protein containing flagellin and antigen, we have evaluated the efficacy of a fusion protein composed of flagellin and two protective antigens of Yersinia pestis (F1 and V) in eliciting protection against respiratory challenge with Y. pestis. Flagellin-F1-V was produced and purified in high yield under good manufacturing practices conditions. The fusion protein retains full Toll-like receptor 5-stimulating activity in vitro. Using a prime-boost immunization protocol, we found that flagellin-F1-V elicits robust antigen-specific humoral immunity in mice and two species of nonhuman primates. Immune mice were fully protected against intranasal challenge with 150 mean tolerated doses of Y. pestis CO92. In immune mice, the bacteria were completely cleared within 3 days after challenge. Flagellin-F1-V exhibited full stability for at least 297 days at 4 degrees C and at least 168 days at 25 degrees C. At between 29 and 84 days at 37 degrees C, the protein exhibited a loss of biological activity that appeared to be associated with a substantial change in protein diameter, possibly due to oligomerization. On the basis of our results, we believe that flagellin-F1-V is an outstanding candidate for evaluation in studies with humans.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Flagelina/imunologia , Vacina contra a Peste/imunologia , Proteínas Citotóxicas Formadoras de Poros/imunologia , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Chlorocebus aethiops , Estabilidade de Medicamentos , Armazenamento de Medicamentos , Feminino , Flagelina/genética , Imunização Secundária , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Vacina contra a Peste/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Análise de Sobrevida , Temperatura , Fatores de Tempo , Receptor 5 Toll-Like/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Yersinia pestis/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...