Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Pharmacol Exp Ther ; 375(2): 296-307, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32958629

RESUMO

Infarct expansion can occur after myocardial infarction (MI), which leads to adverse left ventricular (LV) remodeling and failure. An imbalance between matrix metalloproteinase (MMP) induction and tissue inhibitors of MMPs (TIMPs) can accelerate this process. Past studies have shown different biologic effects of TIMP-3, which may depend upon specific domains within the TIMP-3 molecule. This study tested the hypothesis that differential effects of direct myocardial injections of either a full-length recombinant TIMP-3 (F-TIMP-3) or a truncated form encompassing the N-terminal region (N-TIMP-3) could be identified post-MI. MI was induced in pigs that were randomized for MI injections (30 mg) and received targeted injections within the MI region of F-TIMP-3 (n = 8), N-TIMP-3 (n = 9), or saline injection (MI-only, n = 11). At 14 days post-MI, LV ejection fraction fell post-MI but remained higher in both TIMP-3 groups. Tumor necrosis factor and interleukin-10 mRNA increased by over 10-fold in the MI-only and N-TIMP-3 groups but were reduced with F-TIMP-3 at this post-MI time point. Direct MI injection of either a full-length or truncated form of TIMP-3 is sufficient to favorably alter the course of post-MI remodeling. The functional and differential relevance of TIMP-3 domains has been established in vivo since the TIMP-3 constructs demonstrated different MMP/cytokine expression profiles. These translational studies identify a unique and more specific therapeutic strategy to alter the course of LV remodeling and dysfunction after MI. SIGNIFICANCE STATEMENT: Using different formulations of tissue inhibitor of matrix metalloproteinase-3 (TIMP-3), when injected into the myocardial infarction (MI) region, slowed the progression of indices of left ventricular (LV) failure, suggesting that the N terminus of TIMP-3 is sufficient to attenuate early adverse functional events post-MI. Injections of full-length recombinant TIMP-3, but not of the N-terminal region of TIMP-3, reduced relative indices of inflammation at the mRNA level, suggesting that the C-terminal region affects other biological pathways. These unique proof-of-concept studies demonstrate the feasibility of using recombinant small molecules to selectively interrupt adverse LV remodeling post-MI.


Assuntos
Infarto do Miocárdio/patologia , Fragmentos de Peptídeos/farmacologia , Inibidor Tecidual de Metaloproteinase-3/química , Remodelação Ventricular/efeitos dos fármacos , Sequência de Aminoácidos , Colágeno/genética , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Injeções , Metaloproteinases da Matriz/genética , Fragmentos de Peptídeos/química , Domínios Proteicos , RNA Mensageiro/genética , Inibidor Tecidual de Metaloproteinase-3/genética
2.
Pharmacol Res Perspect ; 6(6): e00442, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30459952

RESUMO

Tissue Inhibitor of Metalloproteinase 3 (TIMP3) is a secreted protein that has a great utility to inhibit elevated metalloproteinase (MMP) activity in injured tissues including infarcted cardiac tissue, inflamed vessels, and joint cartilages. An imbalance between TIMP3 and active MMP levels in the local tissue area may cause worsening of disease progression. To counter balance elevated MMP levels, exogenous administration of TIMP3 appeared to be beneficial in preclinical studies. However, the current form of WT-TIMP3 molecule has a limitation to be a therapeutic candidate due to low production yield, short plasma half-life, injection site retention, and difficulty in delivery, etc. We have engineered TIMP3 molecules by adding extra glycosylation sites or fusing with albumin, Fc, and antibody to improve pharmacokinetic properties. In general, the C-terminal fusion of TIMP3 improved expression and production in mammalian cells and extended half-lives dramatically 5-20 folds. Of note, a site-specific glycosylation at K22S/F34N resulted in a higher level of expression and better cardiac function compared to other fusion proteins in the context of left ventricle ejection fraction (LVEF) changes in a rat myocardial infarction model. It appeared that cardiac efficacy depends on a high ECM binding affinity, in which K22S/F34N and N-TIMP3 showed a higher binding to the ECM compared to other engineered molecules. In conclusion, we found that the ECM binding and sustained residence of injected TIMP3 molecules are important for cardiac tissue localization and inhibition of adverse remodeling activity.


Assuntos
Proteína ADAM17/antagonistas & inibidores , Metaloproteinases da Matriz/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Inibidor Tecidual de Metaloproteinase-3/farmacologia , Função Ventricular Esquerda/efeitos dos fármacos , Proteína ADAM17/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Progressão da Doença , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fibroblastos , Glicosilação , Humanos , Infusões Intravenosas , Injeções Intralesionais , Masculino , Mutação , Infarto do Miocárdio/etiologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Inibidor Tecidual de Metaloproteinase-3/química , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/uso terapêutico , Resultado do Tratamento
3.
Anal Biochem ; 511: 17-23, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27485270

RESUMO

Prostaglandin D2 synthase (PGDS) catalyzes the isomerization of prostaglandin H2 (PGH2) to prostaglandin D2 (PGD2). PGD2 produced by hematopoietic prostaglandin D2 synthase (H-PGDS) in mast cells and Th2 cells is proposed to be a mediator of allergic and inflammatory responses. Consequently, inhibitors of H-PGDS represent potential therapeutic agents for the treatment of inflammatory diseases such as asthma. Due to the instability of the PGDS substrate PGH2, an in-vitro enzymatic assay is not feasible for large-scale screening of H-PGDS inhibitors. Herein, we report the development of a competition binding assay amenable to high-throughput screening (HTS) in a scintillation proximity assay (SPA) format. This assay was used to screen an in-house compound library of approximately 280,000 compounds for novel H-PGDS inhibitors. The hit rate of the H-PGDS primary screen was found to be 4%. This high hit rate suggests that the active site of H-PGDS can accommodate a large diversity of chemical scaffolds. For hit prioritization, these initial hits were rescreened at a lower concentration in SPA and tested in the LAD2 cell assay. 116 compounds were active in both assays with IC50s ranging from 6 to 807 nM in SPA and 82 nM to 10 µM in the LAD2 cell assay.


Assuntos
Inibidores Enzimáticos/química , Oxirredutases Intramoleculares/antagonistas & inibidores , Oxirredutases Intramoleculares/química , Lipocalinas/antagonistas & inibidores , Lipocalinas/química , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Lipocalinas/genética , Lipocalinas/metabolismo , Prostaglandina D2/biossíntese , Prostaglandina D2/sangue , Prostaglandina H2/química , Prostaglandina H2/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
Mol Pharm ; 13(7): 2387-96, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27248573

RESUMO

Antibody-drug conjugates (ADC) rely on the target-binding specificity of an antibody to selectively deliver potent drugs to cancer cells. IgG antibody half-life is regulated by neonatal Fc receptor (FcRn) binding. Histidine 435 of human IgG was mutated to alanine (H435A) to explore the effect of FcRn binding on the pharmacokinetics, efficacy, and tolerability of two separate maytansine-based ADC pairs with noncleavable linkers, (c-DM1 and c-H435A-DM1) and (7v-Cys-may and 7v-H435A-Cys-may). The in vitro cell-killing potency of each pair of ADCs was similar, demonstrating that H435A showed no measurable impact on ADC bioactivity. The H435A mutant antibodies showed no detectable binding to human or mouse FcRn in vitro, whereas their counterpart wild-type IgG ADCs were found to bind to FcRn at pH = 6.0. In xenograft bearing SCID mice expressing mouse FcRn, the AUC of 7v-Cys-may was 1.6-fold higher than that of 7v-H435A-may, yet the observed efficacy was similar. More severe thrombocytopenia was observed with 7v-H435A-Cys-may as compared to 7v-Cys-may at multiple dose levels. The AUC of c-DM1 was approximately 3-fold higher than that of c-H435A-DM1 in 786-0 xenograft bearing SCID mice, which led to a 3-fold difference in efficacy by dose. Murine FcRn knockout, human FcRn transgenic line 32 SCID animals bearing 786-0 xenografts showed an amplified exposure difference between c-DM1 and c-H435A-DM1 as compared to murine FcRn expressing SCID mice, leading to a 10-fold higher dose required for efficacy despite a 6-fold higher AUC of the c-H435A-DM1. The accelerated clearance observed for the noncleavable maytansine ADCs with the H435A FcRn mutation led to reduced efficacy at equivalent doses and exacerbation of clinical pathology parameters (decreased tolerability) at equivalent doses. The results show that reduced ADC clearance mediated by FcRn modulation can improve therapeutic index.


Assuntos
Anticorpos/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoconjugados/farmacologia , Imunoglobulina G/metabolismo , Receptores Fc/metabolismo , Animais , Anticorpos/genética , Ligante CD27/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Imunoconjugados/química , Maitansina/metabolismo , Camundongos , Camundongos SCID , Receptores Fc/genética
5.
Cancer Res ; 75(24): 5329-40, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26631267

RESUMO

Antibody-drug conjugates (ADC) target cytotoxic drugs to antigen-positive cells for treating cancer. After internalization, ADCs with noncleavable linkers are catabolized to amino acid-linker-warheads within the lysosome, which then enter the cytoplasm by an unknown mechanism. We hypothesized that a lysosomal transporter was responsible for delivering noncleavable ADC catabolites into the cytoplasm. To identify candidate transporters, we performed a phenotypic shRNA screen with an anti-CD70 maytansine-based ADC. This screen revealed the lysosomal membrane protein SLC46A3, the genetic attenuation of which inhibited the potency of multiple noncleavable antibody-maytansine ADCs, including ado-trastuzumab emtansine. In contrast, the potencies of noncleavable ADCs carrying the structurally distinct monomethyl auristatin F were unaffected by SLC46A3 attenuation. Structure-activity experiments suggested that maytansine is a substrate for SLC46A3. Notably, SLC46A3 silencing led to relative increases in catabolite concentrations in the lysosome. Taken together, our results establish SLC46A3 as a direct transporter of maytansine-based catabolites from the lysosome to the cytoplasm, prompting further investigation of SLC46A3 as a predictive response marker in breast cancer specimens.


Assuntos
Antineoplásicos Fitogênicos/metabolismo , Imunoconjugados/metabolismo , Maitansina/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Antineoplásicos Fitogênicos/administração & dosagem , Linhagem Celular Tumoral , Citoplasma/metabolismo , Sistemas de Liberação de Medicamentos , Humanos , Imunoconjugados/administração & dosagem , Lisossomos/metabolismo , Maitansina/administração & dosagem
6.
Cancer Cell ; 26(2): 177-89, 2014 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-25043603

RESUMO

Death receptor agonist therapies have exhibited limited clinical benefit to date. Investigations into why Apo2L/TRAIL and AMG 655 preclinical data were not predictive of clinical response revealed that coadministration of Apo2L/TRAIL with AMG 655 leads to increased antitumor activity in vitro and in vivo. The combination of Apo2L/TRAIL and AMG 655 results in enhanced signaling and can sensitize Apo2L/TRAIL-resistant cells. Structure determination of the Apo2L/TRAIL-DR5-AMG 655 ternary complex illustrates how higher order clustering of DR5 is achieved when both agents are combined. Enhanced agonism generated by combining Apo2L/TRAIL and AMG 655 provides insight into the limited efficacy observed in previous clinical trials and suggests testable hypotheses to reconsider death receptor agonism as a therapeutic strategy.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Anticorpos Monoclonais/química , Antineoplásicos/química , Linhagem Celular Tumoral , Sobrevivência Celular , Cristalografia por Raios X , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Humanos , Camundongos , Modelos Moleculares , Multimerização Proteica , Estrutura Quaternária de Proteína , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/química , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/química , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Sci Transl Med ; 6(223): 223ra21, 2014 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-24523321

RESUMO

An imbalance between matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs) contributes to the left ventricle (LV) remodeling that occurs after myocardial infarction (MI). However, translation of these observations into a clinically relevant, therapeutic strategy remains to be established. The present study investigated targeted TIMP augmentation through regional injection of a degradable hyaluronic acid hydrogel containing recombinant TIMP-3 (rTIMP-3) in a large animal model. MI was induced in pigs by coronary ligation. Animals were then randomized to receive targeted hydrogel/rTIMP-3, hydrogel alone, or saline injection and followed for 14 days. Instrumented pigs with no MI induction served as referent controls. Multimodal imaging (fluoroscopy/echocardiography/magnetic resonance imaging) revealed that LV ejection fraction was improved, LV dilation was reduced, and MI expansion was attenuated in the animals treated with rTIMP-3 compared to all other controls. A marked reduction in proinflammatory cytokines and increased smooth muscle actin content indicative of myofibroblast proliferation occurred in the MI region with hydrogel/rTIMP-3 injections. These results provide the first proof of concept that regional sustained delivery of an MMP inhibitor can effectively interrupt adverse post-MI remodeling.


Assuntos
Hidrogel de Polietilenoglicol-Dimetacrilato/química , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Inibidor Tecidual de Metaloproteinase-3/administração & dosagem , Inibidor Tecidual de Metaloproteinase-3/uso terapêutico , Remodelação Ventricular/fisiologia , Animais , Modelos Animais de Doenças , Hidrogel de Polietilenoglicol-Dimetacrilato/administração & dosagem , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Remodelação Ventricular/efeitos dos fármacos
8.
Curr Pharm Biotechnol ; 11(3): 241-5, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20210749

RESUMO

The expression of proteins which do not express well on their own can be enhanced by linking them to human serum albumin (HSA) or antibody crystallizable fragment (Fc). The constructs shown here are designed to secrete the proteins after transient transfection of mammalian cell lines. The fusion partners are appended to the N-terminus of the proteins and contain a linker designed to be proteolytically cleaved. Transient transfection and purification protocols are provided as well as experimental results with five interleukins.


Assuntos
Células Eucarióticas/fisiologia , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Animais , Humanos , Mamíferos
9.
Protein Sci ; 19(2): 357-62, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20014434

RESUMO

The expression levels of five secreted target interleukins (IL-11, 15, 17B, 32, and IL23 p19 subunit) were tested with three different fusion partners in 2936E cells. When fused to the N-terminus, human serum albumin (HSA) was found to enhance the expression of both IL-17B and IL-15, cytokines which did not express at measurable levels on their own. Although the crystallizable fragment of an antibody (Fc) was also an effective fusion partner for IL-17B, Fc did not increase expression of IL-15. Fc was superior to HSA for the expression of the p19 subunit of IL-23, but no partner led to measurable levels of IL-32gamma secretion. Glutathione S-transferase (GST) did not enhance the expression of any target and suppressed the production of IL-11, a cytokine which expressed robustly both on its own and when fused to HSA or Fc. Cleavage of the fusion partner was not always possible. The use of HSA or Fc as N-terminal fusions can be an effective technique to express difficult proteins, especially for applications in which the fusion partner need not be removed.


Assuntos
Fragmentos Fc das Imunoglobulinas/metabolismo , Interleucinas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Albumina Sérica/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Humanos , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/genética , Interleucinas/química , Interleucinas/genética , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Albumina Sérica/genética , Transfecção
10.
J Mol Biol ; 356(2): 367-81, 2006 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-16368107

RESUMO

Dimeric interactions among anti- and pro-apoptotic members of the BCL-2 protein family are dynamically regulated and intimately involved in survival and death functions. We report the structure of a BCL-X(L) homodimers a 3D-domain swapped dimer (3DDS). The X-ray crystal structure demonstrates the mutual exchange of carboxy-terminal regions including BH2 (Bcl-2 homology 2) between monomer subunits, with the hinge region occurring at the hairpin turn between the fifth and sixth alpha helices. Both BH3 peptide-binding hydrophobic grooves are unoccupied in the 3DDS dimer and available for BH3 peptide binding, as confirmed by sedimentation velocity analysis. BCL-X(L) 3DDS dimers have increased pore-forming activity compared to monomers, suggesting that 3DDS dimers may act as intermediates in membrane pore formation. Chemical crosslinking studies of Cys-substituted BCL-X(L) proteins demonstrate that 3DDS dimers form in synthetic lipid vesicles.


Assuntos
Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteína bcl-X/química , Reagentes de Ligações Cruzadas/química , Cristalografia por Raios X , Dimerização , Humanos , Modelos Moleculares , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
11.
Oncogene ; 23(28): 4818-27, 2004 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-15122313

RESUMO

Enforced expression of the antiapoptotic Bcl-2 family protein Mcl-1 promotes lymphomagenesis in the mouse; however, the functional role of Mcl-1 in human B-cell lymphoma remains unclear. We demonstrate that Mcl-1 is widely expressed in malignant B-cells, and high-level expression of Mcl-1 is required for B-lymphoma cell survival, since transfection of Mcl-1-specific antisense oligodeoxynucleotides was sufficient to promote apoptosis in Akata6 lymphoma cells. Mcl-1 was efficiently cleaved by caspases at evolutionarily conserved aspartic acid residues in vitro, and during cisplatin-induced apoptosis in B-lymphoma cell lines and spontaneous apoptosis of primary malignant B-cells. Overexpression of the Mcl-1 cleavage product that accumulated during apoptosis was sufficient to kill cells. Therefore, Mcl-1 is an essential survival molecule for B-lymphoma cells and is cleaved by caspases to a death-promoting molecule during apoptosis. In contrast to Mcl-1, Bcl-2 and Bcl-XL were relatively resistant to caspase cleavage in vitro and in intact cells. Interfering with Mcl-1 function appears to be an effective means of inducing apoptosis in Mcl-1-positive B-cell lymphoma, and the unique sensitivity of Mcl-1 to caspase-mediated cleavage suggests an attractive strategy for converting it to a proapoptotic molecule.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular/fisiologia , Linfoma de Células B/patologia , Proteínas Oncogênicas/metabolismo , Biópsia , Proteínas de Ciclo Celular/genética , Morte Celular , Linhagem Celular Tumoral , Humanos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Proteínas Oncogênicas/genética , Fases de Leitura Aberta , Plasmídeos , Tionucleotídeos/farmacologia
12.
J Biol Chem ; 279(3): 2159-65, 2004 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-14534311

RESUMO

Cells expressing high levels of the BCL-X(L) anti-apoptotic protein are preferentially killed by the mitochondrial inhibitor antimycin A (AA). Computational modeling predicts a binding site for AA in the extended hydrophobic groove on BCL-X(L), previously identified as an interface for dimerization to BAX and related proapoptotic proteins. Here, we identify BCL-X(L) hydrophobic groove mutants with normal cellular anti-apoptotic function but suppressed sensitivity to AA. The LD(50) of AA for cells expressing BCL-X(L) mutants directly correlates with the measured in vitro dissociation constants for AA binding. These results indicate that BCL-X(L) is a principal target mediating AA cytotoxicity.


Assuntos
Antimicina A/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/química , Sequência de Aminoácidos , Sítios de Ligação , Sobrevivência Celular/efeitos dos fármacos , Interações Hidrofóbicas e Hidrofílicas , Dados de Sequência Molecular , Mutação Puntual , Dobramento de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Proteína bcl-X
13.
Curr Med Chem ; 10(16): 1553-62, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12871126

RESUMO

The Bcl-2 family of proteins provide the most unambiguous link between mitochondrial functions and apoptosis, as their only (or principal) functions appear to be as regulators of this cell death pathway. Rational drug design to manipulate the functions of these proteins has been hampered by the lack of a clear understanding of a biochemical or molecular function, with disruption of intra-family protein-protein interactions as the only known, but daunting, objective. There has been substantial progress in this task using molecular modeling and drug leads. The prospects are also good for development of chemical tools for functional analysis of the Bcl-2 proteins.


Assuntos
Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Antimicina A/metabolismo , Antimicina A/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Benzamidas/metabolismo , Benzamidas/farmacologia , Benzopiranos/metabolismo , Benzopiranos/farmacologia , Citocromos c/metabolismo , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Canais Iônicos/metabolismo , Mitocôndrias/fisiologia , Modelos Moleculares , Nitrilas/metabolismo , Nitrilas/farmacologia , Peptídeos/metabolismo , Peptídeos/farmacologia , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/química , Proteínas Proto-Oncogênicas c-bcl-2/genética , Tiazóis/metabolismo , Tiazóis/farmacologia
15.
J Mol Biol ; 315(3): 471-7, 2002 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-11786026

RESUMO

The rational design of loops and turns is a key step towards creating proteins with new functions. We used a computational design procedure to create new backbone conformations in the second turn of protein L. The Protein Data Bank was searched for alternative turn conformations, and sequences optimal for these turns in the context of protein L were identified using a Monte Carlo search procedure and an energy function that favors close packing. Two variants containing 12 and 14 mutations were found to be as stable as wild-type protein L. The crystal structure of one of the variants has been solved at a resolution of 1.9 A, and the backbone conformation in the second turn is remarkably close to that of the in silico model (1.1 A RMSD) while it differs significantly from that of wild-type protein L (the turn residues are displaced by an average of 7.2 A). The folding rates of the redesigned proteins are greater than that of the wild-type protein and in contrast to wild-type protein L the second beta-turn appears to be formed at the rate limiting step in folding.


Assuntos
Proteínas de Bactérias/química , Simulação por Computador , Engenharia de Proteínas , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biologia Computacional , Cristalografia por Raios X , Bases de Dados de Proteínas , Cinética , Modelos Moleculares , Método de Monte Carlo , Mutação/genética , Desnaturação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...