Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochemistry ; 62(11): 1589-1593, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37184546

RESUMO

Fragment antigen-binding domains of antibodies (Fabs) are powerful probes of structure-function relationships of assembly line polyketide synthases (PKSs). We report the discovery and characterization of Fabs interrogating the structure and function of the ketosynthase-acyltransferase (KS-AT) core of Module 2 of the 6-deoxyerythronolide B synthase (DEBS). Two Fabs (AC2 and BB1) were identified to potently inhibit the catalytic activity of Module 2. Both AC2 and BB1 were found to modulate ACP-mediated reactions catalyzed by this module, albeit by distinct mechanisms. AC2 primarily affects the rate (kcat), whereas BB1 increases the KM of an ACP-mediated reaction. A third Fab, AA5, binds to the KS-AT fragment of DEBS Module 2 without altering either parameter; it is phenotypically reminiscent of a previously characterized Fab, 1B2, shown to principally recognize the N-terminal helical docking domain of DEBS Module 3. Crystal structures of AA5 and 1B2 bound to the KS-AT fragment of Module 2 were solved to 2.70 and 2.65 Å resolution, respectively, and revealed entirely distinct recognition features of the two antibodies. The new tools and insights reported here pave the way toward advancing our understanding of the structure-function relationships of DEBS Module 2, arguably the most well-studied module of an assembly line PKS.


Assuntos
Eritromicina , Policetídeo Sintases , Policetídeo Sintases/química , Aciltransferases/química , Anticorpos
2.
PLoS One ; 16(5): e0250318, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33983947

RESUMO

Viral infection and pathogenesis is mediated by host protein-viral protein complexes that are important targets for therapeutic intervention as they are potentially less prone to development of drug resistance. We have identified human, recombinant antibodies (Fabs) from a phage display library that bind to three HIV-host complexes. We used these Fabs to 1) stabilize the complexes for structural studies; and 2) facilitate characterization of the function of these complexes. Specifically, we generated recombinant Fabs to Vif-CBF-ß-ELOB-ELOC (VCBC); ESCRT-I complex and AP2-complex. For each complex we measured binding affinities with KD values of Fabs ranging from 12-419 nM and performed negative stain electron microscopy (nsEM) to obtain low-resolution structures of the HIV-Fab complexes. Select Fabs were converted to scFvs to allow them to fold intracellularly and perturb HIV-host protein complex assembly without affecting other pathways. To identify these recombinant Fabs, we developed a rapid screening pipeline that uses quantitative ELISAs and nsEM to establish whether the Fabs have overlapping or independent epitopes. This pipeline approach is generally applicable to other particularly challenging antigens that are refractory to immunization strategies for antibody generation including multi-protein complexes providing specific, reproducible, and renewable antibody reagents for research and clinical applications. The curated antibodies described here are available to the scientific community for further structural and functional studies on these critical HIV host-factor proteins.


Assuntos
HIV-1/metabolismo , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Anticorpos/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Complexos Multiproteicos/metabolismo , Ligação Proteica , Replicação Viral/fisiologia
3.
Elife ; 92020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33236988

RESUMO

Membrane protein biogenesis in the endoplasmic reticulum (ER) is complex and failure-prone. The ER membrane protein complex (EMC), comprising eight conserved subunits, has emerged as a central player in this process. Yet, we have limited understanding of how EMC enables insertion and integrity of diverse clients, from tail-anchored to polytopic transmembrane proteins. Here, yeast and human EMC cryo-EM structures reveal conserved intricate assemblies and human-specific features associated with pathologies. Structure-based functional studies distinguish between two separable EMC activities, as an insertase regulating tail-anchored protein levels and a broader role in polytopic membrane protein biogenesis. These depend on mechanistically coupled yet spatially distinct regions including two lipid-accessible membrane cavities which confer client-specific regulation, and a non-insertase EMC function mediated by the EMC lumenal domain. Our studies illuminate the structural and mechanistic basis of EMC's multifunctionality and point to its role in differentially regulating the biogenesis of distinct client protein classes.


Cells are surrounded and contained by a plasma membrane consisting of a double layer of fats and proteins. These proteins monitor and facilitate the movement of food, oxygen and messages in and out of the cell, and help neighboring cells communicate. Membrane proteins are manufactured in a cell compartment called the endoplasmic reticulum. Cellular machines called ribosomes visit this compartment's membrane to manufacture proteins that need to be secreted or embedded into the cell's membranes. As these proteins are made, they are pulled into the endoplasmic reticulum so they can be folded correctly and inserted in the membrane. A cellular machine in this compartment's membrane that aids this process is the endoplasmic reticulum membrane protein complex (EMC). Many steps can go wrong during protein assembly, so to control protein quality, the EMC has to accommodate the variety of complex physical features that proteins can have. To explore the activity of the EMC, Miller-Vedam, Bräuning, Popova et al. studied the normal structure of the EMC in both yeast and human cells grown in the lab. These snapshots of the complex in different species had a lot in common, including how the complex was arranged within and around the membrane. Next, Miller-Vedam, Bräuning, Popova et al. generated 50 mutant versions of the EMC in human cells to determine how changing different parts of the complex affected the production of three proteins that rely on the EMC to fold correctly. These proteins were an enzyme called squalene synthase, a signaling protein called the beta adrenergic receptor and sigma intracellular receptor 2, a protein involved in the regulation of cholesterol levels. Mutations in the section of the EMC outside of the endoplasmic reticulum, within the main cellular compartment, negatively impacted the stability of squalene synthase. This section of the EMC provides a platform where proteins can associate before entering the membrane. The part of EMC that spans the membrane contains both a fat-filled cavity and a cavity with a 'door' that is either open or closed. Mutations in this section disrupted the insertion of both squalene synthase and the beta adrenergic receptor into the membrane, a role performed by the cavity with the door. The specific role of the fat-filled cavity is still not fully understood, but a mutation affecting this cavity disrupts the correct production of all three proteins studied. The largest section of the complex, which sits inside the endoplasmic reticulum, protected proteins as they folded, ensuring they were not destroyed for being folded incorrectly before they were fully formed. Mutations in this part of the EMC negatively impacted the stability of sigma intracellular receptor 2 without negatively affecting the other proteins. This molecular dissection of the activity of the EMC provides insights into how membrane proteins are manufactured, stabilized, coordinated, and monitored for quality. These findings could contribute towards the development of new treatments for certain congenital diseases. For example, cystic fibrosis, retinitis pigmentosa, and Charcot-Marie-Tooth disease are all thought to be caused by mutations within membrane proteins that require the EMC during their production.


Assuntos
Retículo Endoplasmático/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de Membrana/metabolismo , Western Blotting , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Estrutura Terciária de Proteína , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/biossíntese , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência
4.
J Am Chem Soc ; 142(35): 14933-14939, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32786753

RESUMO

The 6-deoxyerythronolide B synthase (DEBS) is a prototypical assembly line polyketide synthase (PKS) that synthesizes the macrocyclic core of the antibiotic erythromycin. Each of its six multidomain modules presumably sample distinct conformations, as biosynthetic intermediates tethered to their acyl carrier proteins interact with multiple active sites during the courses of their catalytic cycles. The spatiotemporal details underlying these protein dynamics remain elusive. Here, we investigate one aspect of this conformational flexibility using two domain-specific monoclonal antibody fragments (Fabs) isolated from a very large naïve human antibody library. Both Fabs, designated 1D10 and 2G10, were bound specifically and with high affinity to the ketoreductase domain of DEBS module 1 (KR1). Comparative kinetic analysis of stand-alone KR1 as well as a truncated bimodular derivative of DEBS revealed that 1D10 inhibited KR1 activity whereas 2G10 did not. Co-crystal structures of each KR1-Fab complex provided a mechanistic rationale for this difference. A hybrid PKS module harboring KR1 was engineered, whose individual catalytic domains have been crystallographically characterized at high resolution. Size exclusion chromatography coupled to small-angle X-ray scattering (SEC-SAXS) of this hybrid module bound to 1D10 provided further support for the catalytic relevance of the "extended" model of a PKS module. Our findings reinforce the power of monoclonal antibodies as tools to interrogate structure-function relationships of assembly line PKSs.


Assuntos
Aldo-Ceto Redutases/metabolismo , Anticorpos Monoclonais/metabolismo , Sondas Moleculares/metabolismo , Policetídeo Sintases/metabolismo , Aldo-Ceto Redutases/química , Anticorpos Monoclonais/química , Humanos , Lactonas/química , Lactonas/metabolismo , Conformação Molecular , Sondas Moleculares/química , Oxirredução , Policetídeo Sintases/química
5.
Biochemistry ; 57(43): 6201-6208, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30289692

RESUMO

Assembly line polyketide synthases (PKSs) are large multimodular enzymes responsible for the biosynthesis of diverse antibiotics in bacteria. Structural and mechanistic analysis of these megasynthases can benefit from the discovery of reagents that recognize individual domains or linkers in a site-specific manner. Monoclonal antibodies not only have proven themselves as premier tools in analogous applications but also have the added benefit of constraining the conformational flexibility of their targets in unpredictable but often useful ways. Here we have exploited a library based on the naïve human antibody repertoire to discover a Fab (3A6) that recognizes the terminal thioesterase (TE) domain of the 6-deoxyerythronolide B synthase with high specificity. Biochemical assays were used to verify that 3A6 binding does not inhibit enzyme turnover. The co-crystal structure of the TE-3A6 complex was determined at 2.45 Å resolution, resulting in atomic characterization of this protein-protein recognition mechanism. Fab binding had minimal effects on the structural integrity of the TE. In turn, these insights were used to interrogate via small-angle X-ray scattering the solution-phase conformation of 3A6 complexed to a catalytically competent PKS module and bimodule. Altogether, we have developed a high-affinity monoclonal antibody tool that recognizes the TE domain of the 6-deoxyerythronolide B synthase while maintaining its native function.


Assuntos
Anticorpos Monoclonais/metabolismo , Policetídeo Sintases/química , Policetídeo Sintases/metabolismo , Anticorpos Monoclonais/imunologia , Catálise , Cristalografia por Raios X , Eritromicina/análogos & derivados , Eritromicina/metabolismo , Humanos , Modelos Moleculares , Estrutura Molecular , Policetídeo Sintases/imunologia , Conformação Proteica , Especificidade por Substrato
6.
Bioconjug Chem ; 29(10): 3476-3482, 2018 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-30227708

RESUMO

The swell of experimental imaging technologies to noninvasively measure immune checkpoint protein expression presents the opportunity for rigorous comparative studies toward identifying a gold standard. 89Zr-atezolizumab is currently in man, and early data show tumor targeting but also abundant uptake in several normal tissues. Therefore, we conducted a reverse translational study both to understand if tumor to normal tissue ratios for 89Zr-atezolizumab could be improved and to make direct comparisons to 89Zr-C4, a radiotracer that we showed can detect a large dynamic range of tumor-associated PD-L1 expression. PET/CT and biodistribution studies in tumor bearing immunocompetent and nu/nu mice revealed that high specific activity 89Zr-atezolizumab (∼2 µCi/µg) binds to PD-L1 on tumors but also results in very high uptake in many normal mouse tissues, as expected. Unexpectedly, 89Zr-atezolizumab uptake was generally higher in normal mouse tissues compared to 89Zr-C4 and lower in H1975, a tumor model with modest PD-L1 expression. Also unexpectedly, reducing the specific activity at least 15-fold suppressed 89Zr-atezo uptake in normal mouse tissues but increased tumor uptake to levels observed with high specific activity 89Zr-C4. In summary, these data reveal that low specific activity 89Zr-atezo may be necessary for accurately measuring PD-L1 in the tumor microenvironment, assuming a threshold can be identified that preferentially suppresses binding in normal tissues without reducing binding to tumors with abundant expression. Alternatively, high specific activity approaches like 89Zr-C4 PET may be simpler to implement clinically to measure the broad dynamic range of PD-L1 expression known to manifest among tumors.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Compostos Radiofarmacêuticos/química , Zircônio/química , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Antineoplásicos/química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Portadores de Fármacos , Composição de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
7.
J Am Chem Soc ; 140(21): 6518-6521, 2018 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-29762030

RESUMO

Catalytic modules of assembly-line polyketide synthases (PKSs) have previously been observed in two very different conformations-an "extended" architecture and an "arch-shaped" architecture-although the catalytic relevance of neither has been directly established. By the use of a fully human naïve antigen-binding fragment (Fab) library, a high-affinity antibody was identified that bound to the extended conformation of a PKS module, as verified by X-ray crystallography and tandem size-exclusion chromatography-small-angle X-ray scattering (SEC-SAXS). Kinetic analysis proved that this antibody-stabilized module conformation was fully competent for catalysis of intermodular polyketide chain translocation as well as intramodular polyketide chain elongation and functional group modification of a growing polyketide chain. Thus, the extended conformation of a PKS module is fully competent for all of its essential catalytic functions.


Assuntos
Policetídeo Sintases/química , Biocatálise , Cristalografia por Raios X , Humanos , Cinética , Modelos Moleculares , Policetídeo Sintases/metabolismo , Conformação Proteica , Espalhamento a Baixo Ângulo , Difração de Raios X
8.
PLoS Pathog ; 14(1): e1006830, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29304101

RESUMO

The lentiviral protein Viral Infectivity Factor (Vif) counteracts the antiviral effects of host APOBEC3 (A3) proteins and contributes to persistent HIV infection. Vif targets A3 restriction factors for ubiquitination and proteasomal degradation by recruiting them to a multi-protein ubiquitin E3 ligase complex. Here, we describe a degradation-independent mechanism of Vif-mediated antagonism that was revealed through detailed structure-function studies of antibody antigen-binding fragments (Fabs) to the Vif complex. Two Fabs were found to inhibit Vif-mediated A3 neutralization through distinct mechanisms: shielding A3 from ubiquitin transfer and blocking Vif E3 assembly. Combined biochemical, cell biological and structural studies reveal that disruption of Vif E3 assembly inhibited A3 ubiquitination but was not sufficient to restore its packaging into viral particles and antiviral activity. These observations establish that Vif can neutralize A3 family members in a degradation-independent manner. Additionally, this work highlights the potential of Fabs as functional probes, and illuminates how Vif uses a multi-pronged approach involving both degradation dependent and independent mechanisms to suppress A3 innate immunity.


Assuntos
Antivirais/farmacologia , Citosina Desaminase/metabolismo , Fragmentos Fab das Imunoglobulinas/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/imunologia , Desaminases APOBEC , Antivirais/química , Proteínas Culina/química , Proteínas Culina/metabolismo , Citidina Desaminase , Células HEK293 , Infecções por HIV/imunologia , Infecções por HIV/terapia , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/metabolismo , Humanos , Ubiquitina/metabolismo , Ubiquitinação , Montagem de Vírus , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química
9.
Bioconjug Chem ; 29(1): 96-103, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29125731

RESUMO

High sensitivity imaging tools could provide a more holistic view of target antigen expression to improve the identification of patients who might benefit from cancer immunotherapy. We developed for immunoPET a novel recombinant human IgG1 (termed C4) that potently binds an extracellular epitope on human and mouse PD-L1 and radiolabeled the antibody with zirconium-89. Small animal PET/CT studies showed that 89Zr-C4 detected antigen levels on a patient derived xenograft (PDX) established from a non-small-cell lung cancer (NSCLC) patient before an 8-month response to anti-PD-1 and anti-CTLA4 therapy. Importantly, the concentration of antigen is beneath the detection limit of previously developed anti-PD-L1 radiotracers, including radiolabeled atezolizumab. We also show that 89Zr-C4 can specifically detect antigen in human NSCLC and prostate cancer models endogenously expressing a broad range of PD-L1. 89Zr-C4 detects mouse PD-L1 expression changes in immunocompetent mice, suggesting that endogenous PD-1/2 will not confound human imaging. Lastly, we found that 89Zr-C4 could detect acute changes in tumor expression of PD-L1 due to standard of care chemotherapies. In summary, we present evidence that low levels of PD-L1 in clinically relevant cancer models can be imaged with immunoPET using a novel recombinant human antibody.


Assuntos
Antígeno B7-H1/análise , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Imunoconjugados/química , Imunoglobulina G/química , Neoplasias Pulmonares/diagnóstico por imagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Radioisótopos/química , Zircônio/química , Animais , Linhagem Celular Tumoral , Células HEK293 , Humanos , Pulmão/diagnóstico por imagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/química
10.
Nutr Metab (Lond) ; 13: 32, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27141227

RESUMO

BACKGROUND: Apple polyphenols could represent a novel nutritional approach in the management and control of blood glucose, especially in type 2 diabetics. The aim of this study was to test the therapeutic potential of an apple polyphenol extract (APE) in an insulin-resistant rat model and to determine the molecular basis of insulin sensitivity action in skeletal muscle cells. METHODS: Acute effect of APE on the postprandial hyperglycemic response was assayed in 15 week old obese Zucker rats (OZR), by using a meal tolerance test (MTT). The ability of APE to improve whole peripheral insulin sensitivity was also assayed in a chronic study by using the euglycemic-hyperinsulinemic clamp technique. To elucidate the molecular mechanisms, rat L6 myotubes were used. Glucose uptake was measured by using 2-[3H]-Deoxy-Glucose (2-DG) and specific inhibitors, as well as phosphorylation status of key kinases, were used to determine the implicated signaling pathway. RESULTS: In vivo study showed that nutritional intervention with APE induced an increase of insulin sensitivity with an increase of glucose infusion rate (GIR) of 45 %. Additionally, in vitro results showed a synergistic effect between APE and insulin as well as increased glucose uptake through GLUT4 translocation in muscle cells. This translocation was mediated by phosphatydil inositol 3-kinase (PI3K) and peroxisome proliferator-activated receptor-gamma (PPARγ) signaling pathways. CONCLUSIONS: As a whole, this study describes the mechanisms involved in the insulin sensitizing effect of APE, which could be considered a promising ingredient for inclusion in nutritional products focused on the management of chronic diseases such as diabetes.

11.
J Cachexia Sarcopenia Muscle ; 7(1): 68-78, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27065075

RESUMO

BACKGROUND: L-Leu and its metabolite ß-hydroxy-ß-methylbutyrate (HMB) stimulate muscle protein synthesis enhancing the phosphorylation of proteins that regulate anabolic signalling pathways. Alterations in these pathways are observed in many catabolic diseases, and HMB and L-Leu have proven their anabolic effects in in vivo and in vitro models. The aim of this study was to compare the anabolic effects of L-Leu and HMB in myotubes grown in the absence of any catabolic stimuli. METHODS: Studies were conducted in vitro using rat L6 myotubes under normal growth conditions (non-involving L-Leu-deprived conditions). Protein synthesis and mechanistic target of rapamycin signalling pathway were determined. RESULTS: Only HMB was able to increase protein synthesis through a mechanism that involves the phosphorylation of the mechanistic target of rapamycin as well as its downstream elements, pS6 kinase, 4E binding protein-1, and eIF4E. HMB was significantly more effective than L-Leu in promoting these effects through an activation of protein kinase B/Akt. Because the conversion of L-Leu to HMB is limited in muscle, L6 cells were transfected with a plasmid that codes for α-keto isocaproate dioxygenase, the key enzyme involved in the catabolic conversion of α-keto isocaproate into HMB. In these transfected cells, L-Leu was able to promote protein synthesis and mechanistic target of rapamycin regulated pathway activation equally to HMB. Additionally, these effects of leucine were reverted to a normal state by mesotrione, a specific inhibitor of α-keto isocaproate dioxygenase. CONCLUSION: Our results suggest that HMB is an active L-Leu metabolite able to maximize protein synthesis in skeletal muscle under conditions, in which no amino acid deprivation occurred. It may be proposed that supplementation with HMB may be very useful to stimulate protein synthesis in wasting conditions associated with chronic diseases, such as cancer or chronic heart failure.

12.
ACS Chem Biol ; 11(6): 1587-94, 2016 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-27008570

RESUMO

New methodologies for site-specifically radiolabeling proteins with (18)F are required to generate high quality radiotracers for preclinical and clinical applications with positron emission tomography. Herein, we report an approach by which we use lipoic acid ligase (LplA) to conjugate [(18)F]-fluorooctanoic acid to an antibody fragment bearing the peptide substrate of LplA. The mild conditions of the reaction preserve antibody immunoreactivity, and the efficiency of LplA allows for >90% yield even with very small amounts of peptidic precursor (1-10 nmol). These features are advantageous compared to the current gold standard in the field. Moreover, the methodology introduces a new application for an important tool in chemical biology.


Assuntos
Caprilatos/química , Proteínas de Escherichia coli/química , Fragmentos Fab das Imunoglobulinas/química , Ligases/química , Caprilatos/síntese química , Radioisótopos de Flúor , Células HEK293 , Halogenação , Humanos , Peptídeos/química
13.
Cancer Res ; 75(7): 1225-35, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25672980

RESUMO

The increased proteolytic activity of membrane-bound and secreted proteases on the surface of cancer cells and in the transformed stroma is a common characteristic of aggressive metastatic prostate cancer. We describe here the development of an active site-specific probe for detecting a secreted peritumoral protease expressed by cancer cells and the surrounding tumor microenvironment. Using a human fragment antigen-binding phage display library, we identified a human antibody termed U33 that selectively inhibited the active form of the protease urokinase plasminogen activator (uPA, PLAU). In the full-length immunoglobulin form, U33 IgG labeled with near-infrared fluorophores or radionuclides allowed us to noninvasively detect active uPA in prostate cancer xenograft models using optical and single-photon emission computed tomography imaging modalities. U33 IgG labeled with (111)In had a remarkable tumor uptake of 43.2% injected dose per gram (%ID/g) 72 hours after tail vein injection of the radiolabeled probe in subcutaneous xenografts. In addition, U33 was able to image active uPA in small soft-tissue and osseous metastatic lesions using a cardiac dissemination prostate cancer model that recapitulated metastatic human cancer. The favorable imaging properties were the direct result of U33 IgG internalization through an uPA receptor-mediated mechanism in which U33 mimicked the function of the endogenous inhibitor of uPA to gain entry into the cancer cell. Overall, our imaging probe targets a prostate cancer-associated protease, through a unique mechanism, allowing for the noninvasive preclinical imaging of prostate cancer lesions.


Assuntos
Neoplasias da Próstata/enzimologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Linhagem Celular Tumoral , Imunofluorescência , Expressão Gênica , Humanos , Radioisótopos de Índio , Masculino , Camundongos Nus , Transplante de Neoplasias , Especificidade de Órgãos , Neoplasias da Próstata/diagnóstico por imagem , Compostos Radiofarmacêuticos , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Óptica , Ativador de Plasminogênio Tipo Uroquinase/genética
14.
Theranostics ; 4(3): 267-79, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24505235

RESUMO

Subtype-targeted therapies can have a dramatic impact on improving the quality and quantity of life for women suffering from breast cancer. Despite an initial therapeutic response, cancer recurrence and acquired drug-resistance are commonplace. Non-invasive imaging probes that identify drug-resistant lesions are urgently needed to aid in the development of novel drugs and the effective utilization of established therapies for breast cancer. The protease receptor urokinase plasminogen activator receptor (uPAR) is a target that can be exploited for non-invasive imaging. The expression of uPAR has been associated with phenotypically aggressive breast cancer and acquired drug-resistance. Acquired drug-resistance was modeled in cell lines from two different breast cancer subtypes, the uPAR negative luminal A subtype and the uPAR positive triple negative subtype cell line MDA-MB-231. MCF-7 cells, cultured to be resistant to tamoxifen (MCF-7 TamR), were found to significantly over-express uPAR compared to the parental cell line. uPAR expression was maintained when resistance was modeled in triple-negative breast cancer by generating doxorubicin and paclitaxel resistant MDA-MB-231 cells (MDA-MB-231 DoxR and MDA-MB-231 TaxR). Using the antagonistic uPAR antibody 2G10, uPAR was imaged in vivo by near-infrared (NIR) optical imaging and (111)In-single photon emission computed tomography (SPECT). Tumor uptake of the (111)In-SPECT probe was high in the three drug-resistant xenografts (> 46 %ID/g) and minimal in uPAR negative xenografts at 72 hours post-injection. This preclinical study demonstrates that uPAR can be targeted for imaging breast cancer models of acquired resistance leading to potential clinical applications.


Assuntos
Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Anticorpos/análise , Antineoplásicos/farmacologia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/farmacologia , Feminino , Humanos , Imunoglobulina G/análise , Radioisótopos de Índio , Células MCF-7 , Camundongos , Imagem Multimodal , Imagem Óptica , Paclitaxel/farmacologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/imunologia , Tamoxifeno/farmacologia , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
15.
Org Biomol Chem ; 7(13): 2681-4, 2009 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-19532982

RESUMO

Gene delivery systems based on the beta-cyclodextrin scaffold have been synthesized by combining the copper(I)-catalyzed azide-alkyne coupling ("click chemistry") and an efficient acylation method of the secondary hydroxyls; molecular flexibility, charge density and hydrophobic-hydrophilic balance are critical parameters that can be fine-tuned by the click approach.


Assuntos
Técnicas de Transferência de Genes , Substâncias Macromoleculares/química , beta-Ciclodextrinas/química , Alcinos/química , Animais , Azidas/química , Células CHO , Catálise , Cobre/química , Cricetinae , Cricetulus , DNA/química , DNA/metabolismo , Substâncias Macromoleculares/síntese química , Substâncias Macromoleculares/metabolismo , Estrutura Molecular , Nanopartículas/química , Estereoisomerismo , beta-Ciclodextrinas/síntese química , beta-Ciclodextrinas/metabolismo
16.
Clin Nutr ; 28(5): 565-74, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19477051

RESUMO

BACKGROUND AND AIMS: To evaluate if the antidiabetic properties of Salacia oblonga extract are mediated not only by inhibiting intestinal alpha-glycosidases but also by enhancing glucose transport in muscle and adipose cells. METHODS: S. oblonga extract effects on 2-deoxy-D-glucose uptake were assayed in muscle L6-myotubes and 3T3-adipocytes. In L6-myotubes, the amount and translocation of glucose transporters were assayed. A fractionation of the extract was carried out to identify the active compounds. Furthermore, we analyzed the phosphorylation status of key components of signaling pathways that are involved in the molecular mechanisms regulating glucose uptake. RESULTS: S. oblonga extract increased 2-deoxy-D-glucose uptake by 50% in L6-myotubes and 3T3-adipocytes. In L6-myotubes, the extract increased up to a 100% the GLUT4 content, activating GLUT4 promoter transcription and its translocation to the plasma membrane. Mangiferin was identified as the bioactive compound. Furthermore, mangiferin effects were concomitant with the phosphorylation of 5'-AMP-activated protein kinase without the activation of PKB/Akt. The effect of mangiferin on 2-deoxy-D-glucose uptake was blocked by GW9662, an irreversible PPAR-gamma antagonist. CONCLUSIONS: S. oblonga extract and mangiferin may exert their antidiabetic effect by increasing GLUT4 expression and translocation in muscle cells. These effects are probably mediated through two independent pathways that are related to 5'-AMP-activated protein kinase and PPAR-gamma.


Assuntos
Transportador de Glucose Tipo 4/metabolismo , Hipoglicemiantes/farmacologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Extratos Vegetais/farmacologia , Salacia/química , Xantonas/farmacologia , Células 3T3 , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Membrana Celular/metabolismo , Desoxiglucose/metabolismo , Relação Dose-Resposta a Droga , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 4/genética , Insulina/farmacologia , Camundongos , Fibras Musculares Esqueléticas/metabolismo , PPAR gama/antagonistas & inibidores , PPAR gama/metabolismo , Extratos Vegetais/química , Raízes de Plantas/química , Ratos , Rosiglitazona , Tiazolidinedionas/farmacologia , Xantonas/análise
17.
J Biochem ; 145(1): 21-30, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18849572

RESUMO

To dissect the rat receptor for advanced glycation end products (RAGE) subcellular distribution and trafficking in eukaryotic cells, an expression system coding for a fusion protein between the RAGE and an enhanced green fluorescent protein (EGFP) has been used. The RAGE-EGFP protein is expressed at the plasma membrane of CHO-k1 and Neuro-2a (N2a) cells and retains the capacity to bind Texas Red-labelled advanced glycation end products (AGEs). AGEs addition to the cell cultures induced a change in the subcellular distribution of the fluorescent RAGE-EGFP protein compatible with an internalization of the AGEs-RAGE complex. Furthermore, while N2a cells expressing the RAGE-EGFP showed an increase in ERK1/2 phosphorylation and NF-kappaB DNA binding in response to AGEs, pre-incubation with dansyl-cadaverine or phenylarsine oxide, inhibitors of receptors internalization, blocked the activation of ERKs and other intracellular responses mediated by AGEs. These results suggest that internalization plays a key role in the signal transduction mediated by RAGE.


Assuntos
Receptores Imunológicos/análise , Receptores Imunológicos/metabolismo , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Proteínas de Fluorescência Verde/metabolismo , Microscopia Confocal , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Ratos , Receptor para Produtos Finais de Glicação Avançada
18.
Biochem Biophys Res Commun ; 319(1): 247-55, 2004 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-15158469

RESUMO

Several putative polyadenylation sequences and an adenylate plus timidylate rich element (ARE) are present at the 3' end of the rat advanced glycation end products receptor (RAGE) gene. Two transcripts are generated by the use of alternative polyadenylation sequences, one containing the ARE sequence in its 3'-untranslated region (3'-UTR). Transfections of CHO-k1 or NRK cells with constructs expressing the 3'-UTRs of the transcripts fused to a green fluorescence protein mRNA show that the ARE sequence has a negative effect on protein expression correlating with a decrease in the amount of mRNA, as shown in CHO-k1 transfected cells. When transfected cells were incubated in the presence of Actinomycin D the amount of fluorescence decreased in cells transfected with the ARE sequence, indicating that this sequence induces lower mRNA stability. Thus, alternative polyadenylation signals and an ARE sequence provide a novel mechanism for the regulation of the rat RAGE gene expression.


Assuntos
Regiões 3' não Traduzidas , Regulação da Expressão Gênica , RNA Mensageiro/metabolismo , Receptores Imunológicos/genética , Animais , Sequência de Bases , Células CHO , Clonagem Molecular , Cricetinae , Dactinomicina/metabolismo , Biblioteca Gênica , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Rim/metabolismo , Proteínas Luminescentes/metabolismo , Modelos Genéticos , Dados de Sequência Molecular , Processamento Pós-Transcricional do RNA , Ratos , Receptor para Produtos Finais de Glicação Avançada , Espectrometria de Fluorescência , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...