Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 76(21): 6331-6339, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27543601

RESUMO

The fibroblast growth factor receptor FGFR2 is overexpressed in a variety of solid tumors, including breast, gastric, and ovarian tumors, where it offers a potential therapeutic target. In this study, we present evidence of the preclinical efficacy of BAY 1187982, a novel antibody-drug conjugate (ADC). It consists of a fully human FGFR2 monoclonal antibody (mAb BAY 1179470), which binds to the FGFR2 isoforms FGFR2-IIIb and FGFR2-IIIc, conjugated through a noncleavable linker to a novel derivative of the microtubule-disrupting cytotoxic drug auristatin (FGFR2-ADC). In FGFR2-expressing cancer cell lines, this FGFR2-ADC exhibited potency in the low nanomolar to subnanomolar range and was more than 100-fold selective against FGFR2-negative cell lines. High expression levels of FGFR2 in cells correlated with efficient internalization, efficacy, and cytotoxic effects in vitro Pharmacokinetic analyses in mice bearing FGFR2-positive NCI-H716 tumors indicated that the toxophore metabolite of FGFR2-ADC was enriched more than 30-fold in tumors compared with healthy tissues. Efficacy studies demonstrated that FGFR2-ADC treatment leads to a significant tumor growth inhibition or tumor regression of cell line-based or patient-derived xenograft models of human gastric or breast cancer. Furthermore, FGFR2 amplification or mRNA overexpression predicted high efficacy in both of these types of in vivo model systems. Taken together, our results strongly support the clinical evaluation of BAY 1187982 in cancer patients and a phase I study (NCT02368951) has been initiated. Cancer Res; 76(21); 6331-9. ©2016 AACR.


Assuntos
Aminobenzoatos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Imunoconjugados/uso terapêutico , Neoplasias/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/análise , Animais , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
ChemMedChem ; 11(2): 199-206, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26333652

RESUMO

Human neutrophil elastase (HNE) is a key driver of inflammation in many cardiopulmonary and systemic inflammatory and autoimmune conditions. Overshooting high HNE activity is the consequence of a disrupted protease-antiprotease balance. Accordingly, there has been an intensive search for potent and selective HNE inhibitors with suitable pharmacokinetics that would allowing oral administration in patients. Based on the chemical probe BAY-678 and the clinical candidate BAY 85-8501 we explored further ring topologies along the equator of the parent pyrimidinone lead series. Novel ring systems were annulated in the east, yielding imidazolo-, triazolo-, and tetrazolopyrimidines in order to ensure additional inhibitor-HNE contacts beyond the S1 and the S2 pocket of HNE. The western annulation of pyridazines led to the polar pyrimidopyridazine BAY-8040, which combines excellent potency and selectivity with a promising pharmacokinetic profile. In vivo efficacy with regard to decreasing cardiac remodeling and amelioration of cardiac function was shown in a monocrotaline-induced rat model for pulmonary arterial hypertension. This demonstrated in vivo proof of concept in animals.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Elastase de Leucócito/antagonistas & inibidores , Proteínas Secretadas Inibidoras de Proteinases/química , Proteínas Secretadas Inibidoras de Proteinases/farmacologia , Piridazinas/química , Piridazinas/farmacologia , Pirimidinas/química , Pirimidinas/farmacologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Hipertensão Pulmonar/metabolismo , Elastase de Leucócito/metabolismo , Modelos Moleculares , Estrutura Molecular , Proteínas Secretadas Inibidoras de Proteinases/síntese química , Piridazinas/síntese química , Pirimidinas/síntese química , Ratos , Relação Estrutura-Atividade
3.
Endocrinology ; 156(11): 4365-73, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26284426

RESUMO

The prolactin receptor (PRLR) has been implicated in a variety of physiological processes (lactation, reproduction) and diseases (breast cancer, autoimmune diseases). Prolactin synthesis in the pituitary and extrapituitary sites is regulated by different promoters. Dopamine receptor agonists such as bromocriptine can only interfere with pituitary prolactin synthesis and thus do not induce a complete blockade of PRLR signaling. Here we describe the identification of a human monoclonal antibody 005-C04 that blocks PRLR-mediated signaling at nanomolar concentrations in vitro. In contrast to a negative control antibody, the neutralizing PRLR antibody 005-C04 inhibits signal transducer and activator of transcription 5 phosphorylation in T47D cells and proliferation of BaF3 cells stably expressing murine or human PRLRs in a dose-dependent manner. In vivo application of this new function-blocking PRLR antibody reflects the phenotype of PRLR-deficient mice. After antibody administration female mice become infertile in a reversible manner. In lactating dams, the antibody induces mammary gland involution and negatively interferes with lactation capacity as evidenced by reduced milk protein expression in mammary glands and impaired litter weight gain. Antibody-mediated blockade of the PRLR in vivo stimulates hair regrowth in female mice. Compared with peptide-derived PRLR antagonists, the PRLR antibody 005-C04 exhibits several advantages such as higher potency, noncompetitive inhibition of PRLR signaling, and a longer half-life, which allows its use as a tool compound also in long-term in vivo studies. Therefore, we suggest that this antibody will help to further our understanding of the role of auto- and paracrine PRLR signaling in health and disease.


Assuntos
Anticorpos Neutralizantes/administração & dosagem , Fenótipo , Receptores da Prolactina/imunologia , Animais , Relação Dose-Resposta a Droga , Feminino , Camundongos , Camundongos Knockout , Receptores da Prolactina/genética , Receptores da Prolactina/metabolismo
4.
Mol Cancer Ther ; 13(6): 1537-48, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24714131

RESUMO

Mesothelin is a tumor differentiation antigen frequently overexpressed in tumors such as mesothelioma, ovarian, pancreatic, and lung adenocarcinomas while showing limited expression in nonmalignant tissues. Mesothelin is therefore an attractive target for cancer therapy using antibody-drug conjugates (ADC). This study describes the detailed characterization of anetumab ravtansine, here referred to as BAY 94-9343, a novel ADC consisting of a human anti-mesothelin antibody conjugated to the maytansinoid tubulin inhibitor DM4 via a disulfide-containing linker. Binding properties of the anti-mesothelin antibody were analyzed using surface plasmon resonance, immunohistochemistry, flow cytometry, and fluorescence microscopy. Effects of BAY 94-9343 on cell proliferation were first studied in vitro and subsequently in vivo using subcutaneous, orthotopic, and patient-derived xenograft tumor models. The antibody binds to human mesothelin with high affinity and selectivity, thereby inducing efficient antigen internalization. In vitro, BAY 94-9343 demonstrated potent and selective cytotoxicity of mesothelin-expressing cells with an IC(50) of 0.72 nmol/L, without affecting mesothelin-negative or nonproliferating cells. In vivo, BAY 94-9343 localized specifically to mesothelin-positive tumors and inhibited tumor growth in both subcutaneous and orthotopic xenograft models. In addition, BAY 94-9343 was able to induce a bystander effect on neighboring mesothelin-negative tumor cells. Antitumor efficacy of BAY 94-9343 correlated with the amount of mesothelin expressed and was generally superior to that of standard-of-care regimen resulting in complete tumor eradication in most of the models. BAY 94-9343 is a selective and highly potent ADC, and our data support its development for the treatment of patients with mesothelin-expressing tumors.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Proteínas Ligadas por GPI/imunologia , Imunoconjugados/administração & dosagem , Maitansina/análogos & derivados , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Efeito Espectador , Linhagem Celular Tumoral , Proteínas Ligadas por GPI/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Maitansina/administração & dosagem , Mesotelina , Neoplasias/imunologia , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
MAbs ; 6(2): 367-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24492302

RESUMO

Human antibody-ribonuclease (RNase) fusion proteins, referred to as immunoRNases, have been proposed as an alternative to heterologous immunotoxins, without their immunogenicity and unspecific toxicity issues. In this study, we investigated if human pancreatic RNase will be suitable as effector component in a therapeutic antibody development platform. We generated several fusion proteins consisting of tumor-specific human immunoglobulins (IgGs) and human pancreatic RNase. Transient mammalian cell production was efficient and IgG-RNases were purified to homogeneity. Antigen binding was comparable to the parental antibodies and RNase catalytic activity was retained even in the presence of 50-fold molar excess of human cytosolic RNase inhibitor (RI). Serum stability, cell binding and internalization of IgG-RNases were comparable to the parental IgGs. Despite these promising properties, none of the IgG-RNases revealed significant inhibition of tumor cell growth in vitro even when targeting different antigens putatively employing different endocytotic pathways. The introduction of different linkers containing endosomal protease cleavage sites into the IgG-RNase did not enhance cytotoxicity. Similarly, RI evasive human pancreatic RNase variants mediated only small inhibiting effects on tumor cell growth at high concentrations, potentially reflecting inefficient cytosolic translocation. Taken together, human pancreatic RNase and variants did not prove to be generally suitable as effector component for a therapeutic antibody drug development platform.


Assuntos
Adenocarcinoma/tratamento farmacológico , Anticorpos Catalíticos/metabolismo , Neoplasias do Colo/tratamento farmacológico , Imunoglobulina G/metabolismo , Imunoterapia/métodos , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Recombinantes de Fusão/metabolismo , Ribonucleases/metabolismo , Adenocarcinoma/imunologia , Anticorpos Catalíticos/genética , Antígenos de Neoplasias/imunologia , Processos de Crescimento Celular/efeitos dos fármacos , Neoplasias do Colo/imunologia , Citotoxicidade Imunológica , Endocitose , Células HEK293 , Células HT29 , Humanos , Imunoglobulina G/genética , Imunoterapia/tendências , Neoplasias Pulmonares/imunologia , Terapia de Alvo Molecular , Pâncreas/enzimologia , Proteínas Recombinantes de Fusão/genética , Ribonucleases/genética
6.
Antiviral Res ; 95(2): 182-91, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22580131

RESUMO

The RNA-dependent RNA polymerase NS5B of the hepatitis C virus (HCV) has emerged as one of the key targets for antiviral drug discovery. Here we describe a novel non-nucleoside inhibitor (NNI) chemotype identified by screening: The substituted N-phenylbenzenesulphonamides (SPBS) which showed reversible inhibition of NS5B from HCV genotype 1b with IC(50) values up to 40 nM. Based on the decreased inhibitory activity against a recombinant NS5B protein carrying the mutation L419M or M423T we assumed that the SPBS inhibitors bind to the thumb site II which has already been described as the allosteric binding site for the NNI carboxy thiophene. The postulated binding site was consequently confirmed by solving two co-crystal structures of NS5B in complex with SPBS analogues at 2.3 and 2.2Å resolutions. The inhibitors are hydrogen-bonded to the main chain Ser476 and Tyr477 and to the side chain of Arg501. In addition, the inhibitors displayed van der Waals interactions with several residues of the hydrophobic binding pocket Leu419, Ile482, Leu497, Met423 and Trp528. Notably, the two SPBS analogues reported here revealed significant differences in addressing the NH-group of the main chain Tyr477 by hydrogen-bonds, water-mediated or directly, which provoked a shift of the carboxyphenyl group of the inhibitors towards the His475 position for the water-mediated binding mode. Interestingly, the differences observed in the binding mode led to a different cross resistance profile at positions M423 and I482. Using a panel of 38 individual NS5B proteins derived from different HCV genotypes, we could demonstrate inhibitory activity of the SPBS against polymerases from HCV genotypes 1a and 1b whereas the inhibitor class failed to inhibit any of the non-genotype 1 polymerases efficiently. Furthermore we demonstrated initial antiviral activity for SPBS against the subgenomic replicons of HCV genotypes 1a and 1b, respectively, and no considerable cytotoxic potential against a panel of ten different cell types.


Assuntos
Antivirais/farmacologia , Inibidores Enzimáticos/farmacologia , Hepacivirus/efeitos dos fármacos , Hepacivirus/enzimologia , Sulfonamidas/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Sítios de Ligação , Linhagem Celular , Cristalografia por Raios X , Hepatócitos/virologia , Humanos , Concentração Inibidora 50 , Testes de Sensibilidade Microbiana , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo
7.
BMC Biotechnol ; 11: 76, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21777442

RESUMO

BACKGROUND: A promising new approach in cancer therapy is the use of tumor specific antibodies coupled to cytotoxic agents. Currently these immunoconjugates are prepared by rather unspecific coupling chemistries, resulting in heterogeneous products. As the drug load is a key parameter for the antitumor activity, site-specific strategies are desired. Expressed protein ligation (EPL) and protein trans-splicing (PTS) are methods for the specific C-terminal modification of a target protein. Both include the expression as an intein fusion protein, followed by the exchange of the intein for a functionalized moiety. RESULTS: A full-length IgG specific for fibronectin ED-B was expressed as fusion protein with an intein (Mxe GyrA or Npu DnaE) attached to each heavy chain. In vitro protocols were established to site-specifically modify the antibodies in high yields by EPL or PTS, respectively. Although reducing conditions had to be employed during the process, the integrity or affinity of the antibody was not affected. The protocols were used to prepare immunoconjugates containing two biotin molecules per antibody, attached to the C-termini of the heavy chains. CONCLUSION: Full-length antibodies can be efficiently and site-specifically modified at the C-termini of their heavy chains by intein-fusion technologies. The described protocols can be used to prepare immunoconjugates of high homogeneity and with a defined drug load of two. The attachment to the C-termini is expected to retain the affinity and effector functions of the antibodies.


Assuntos
Fibronectinas/imunologia , Imunoconjugados/química , Imunoglobulina G/química , Inteínas/genética , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/química , Sequência de Aminoácidos , Eletroforese em Gel de Poliacrilamida , Humanos , Imunoconjugados/imunologia , Imunoconjugados/metabolismo , Imunoglobulina G/biossíntese , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Trans-Splicing
8.
Chembiochem ; 12(11): 1774-80, 2011 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-21656631

RESUMO

Sortase A from Staphylococcus aureus attracts growing interest for its use in biotechnological protein modification. This enzyme binds to a short signal sequence at the C terminus of a target protein, cleaves it by formation of an acyl-enzyme intermediate, and subsequently attaches an oligoglycine with a peptide bond. In this work, we explored its usability for the modification of the L19 Fab fragment (specific for fibronectin ED-B), a promising candidate for antibody-based cancer therapy. The Fab fragment was expressed with a sortase signal sequence attached to its light chain, and was successfully modified with a fluorescent oligoglycine probe in good yield. Our interest focused on performance under conditions of limited oligoglycine concentrations. Two unproductive side reactions of sortase were observed. The first was hydrolysis of the acyl-enzyme intermediate; in the second, sortase accepted the ε-amino group of lysine as substrate, thereby resulting in polypeptide crosslinking. In case of the L19 Fab fragment, it led to the covalent connection of the heavy and light chains. Both side reactions were effectively suppressed by sufficient concentrations of the oligoglycine probe.


Assuntos
Aminoaciltransferases/química , Proteínas de Bactérias/química , Cisteína Endopeptidases/química , Fragmentos Fab das Imunoglobulinas/química , Lisina/química , Água/química , Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Humanos , Hidrólise , Fragmentos Fab das Imunoglobulinas/metabolismo , Lisina/metabolismo , Modelos Moleculares , Peptídeos/metabolismo , Especificidade por Substrato , Água/metabolismo
9.
J Mol Biol ; 409(5): 681-91, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21549129

RESUMO

Human neutrophil elastase (HNE), a trypsin-type serine protease, is of pivotal importance in the onset and progression of chronic obstructive pulmonary disease (COPD). COPD encompasses a group of slowly progressive respiratory disorders and is a major medical problem and the fifth leading cause of death worldwide. HNE is a major target for the development of compounds that inhibit the progression of long-term lung function decline in COPD patients. Here, we present the three-dimensional structure of a potent dihydropyrimidone inhibitor (DHPI) non-covalently bound to HNE at a resolution of 2.0 Å. The inhibitor binds to the active site in a unique orientation addressing S1 and S2 subsites of the protease. To facilitate further analysis of this binding mode, we determined the structure of the uncomplexed enzyme at a resolution of 1.86 Å. Detailed comparisons of the HNE:DHPI complex with the uncomplexed HNE structure and published structures of other elastase:inhibitor complexes revealed that binding of DHPI leads to large conformational changes in residues located in the S2 subsite. The rearrangement of residues Asp95-Leu99B creates a deep, well-defined cavity, which is filled by the P2 moiety of the inhibitor molecule to almost perfect shape complementarity. The shape of the S2 subsite in complex with DHPI clearly differs from all other observed HNE structures. The observed structural flexibility of the S2 subsite is a key feature for the understanding of the binding mode of DHPIs in general and the development of new HNE selective inhibitors.


Assuntos
Inibidores Enzimáticos/química , Elastase de Leucócito/antagonistas & inibidores , Elastase de Leucócito/química , Pirimidinas/química , Sítios de Ligação , Domínio Catalítico , Humanos , Modelos Moleculares , Conformação Proteica , Doença Pulmonar Obstrutiva Crônica/enzimologia
10.
J Mol Biol ; 358(5): 1328-40, 2006 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-16600296

RESUMO

DNA topoisomerases are a family of enzymes altering the topology of DNA by concerted breakage and rejoining of the phosphodiester backbone of DNA. Bacterial and archeal type IA topoisomerases, including topoisomerase I, topoisomerase III, and reverse gyrase, are crucial in regulation of DNA supercoiling and maintenance of genetic stability. The crystal structure of full length topoisomerase I from Thermotoga maritima was determined at 1.7A resolution and represents an intact and fully active bacterial topoisomerase I. It reveals the torus-like structure of the conserved transesterification core domain comprising domains I-IV and a tightly associated C-terminal zinc ribbon domain (domain V) packing against domain IV of the core domain. The previously established zinc-independence of the functional activity of T.maritima topoisomerase I is further supported by its crystal structure as no zinc ion is bound to domain V. However, the structural integrity is preserved by the formation of two disulfide bridges between the four Zn-binding cysteine residues. A functional role of domain V in DNA binding and recognition is suggested and discussed in the light of the structure and previous biochemical findings. In addition, implications for bacterial topoisomerases I are provided.


Assuntos
DNA Topoisomerases Tipo I/química , Thermotoga maritima/enzimologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Sequência de Bases , Domínio Catalítico , Sequência Conservada , Cristalografia por Raios X , DNA Topoisomerases Tipo I/genética , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Escherichia coli/enzimologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Thermotoga maritima/genética
11.
Structure ; 12(11): 2081-93, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15530372

RESUMO

The biological activity of chemokines requires interactions with cell surface proteoglycans. We have determined the structure of the chemokine RANTES (regulated on activation normal T cell expressed) in the presence of heparin-derived disaccharide analogs by X-ray crystallography. These structures confirm the essential role of the BBXB motif in the interaction between the chemokine and the disaccharide. Unexpected interactions were observed in the 30s loop and at the amino terminus. Mutant RANTES molecules were designed to abrogate these interactions and their biological activity examined in vivo. The K45E mutant within the BBXB motif lost the capacity to bind heparin and the ability to elicit cellular recruitment. The Y3A mutant maintained its capacity to bind heparin but was unable to elicit cellular recruitment. Finally, a tetrasaccharide is the smallest oligosaccharide which effectively abolishes the ability of RANTES to recruit cells in vivo. These crystallographic structures provide a description of the molecular interaction of a chemokine with glycosaminoglycans.


Assuntos
Quimiocina CCL5/química , Quimiocinas/antagonistas & inibidores , Sequência de Carboidratos , Quimiocina CCL5/genética , Cristalografia por Raios X , Heparina/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica
12.
J Biol Chem ; 279(1): 311-8, 2004 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-14578355

RESUMO

Protein tyrosine phosphatases (PTPs) play important, highly dynamic roles in signaling. Currently about 90 different PTP genes have been described. The enzymes are highly regulated at all levels of expression, and it is becoming increasingly clear that substrate specificity of the PTP catalytic domains proper contributes considerably to PTP functionality. To investigate PTP substrate selectivity, we have set up a procedure to generate phage libraries that presents randomized, phosphotyrosine-containing peptides. Phages that expressed suitable substrates were selected by immobilized, substrate-trapping GST-PTP fusion proteins. After multiple rounds of selection, positive clones were confirmed by SPOT analysis, dephosphorylation by wild-type enzyme, and Km determinations. We have identified distinct consensus substrate motifs for PTP1B, Sap-1, PTP-beta, SHP1, and SHP2. Our results confirm substrate specificity for individual PTPs at the peptide level. Such consensus sequences may be useful both for identifying potential PTP substrates and for the development of peptidomimetic inhibitors.


Assuntos
Bacteriófagos/genética , Fosfotirosina/metabolismo , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/metabolismo , Sequência de Aminoácidos , Capsídeo/química , Domínio Catalítico , Primers do DNA , Biblioteca Gênica , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Mapeamento por Restrição , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
13.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 4): 677-87, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12657787

RESUMO

The antibody Fv fragment 7E2 has previously been employed in the induced crystallization of the integral membrane protein cytochrome c oxidase from Paracoccus denitrificans. The 1.3 A X-ray structure of the uncomplexed antibody fragment reveals conserved water networks on the surfaces of the framework regions. A novel consensus motif for water coordination, XX(S/T), is found along the edges of the beta-sandwich, where a water molecule forms hydrogen bonds to the carbonyl O atom of a residue at position N and the OG hydroxyl groups of conserved serines or threonines at position N + 2. Multiple conformations were found in the hydrophobic core for residues IleL21, LeuL33 and the disulfide bridges. An internal water molecule that is compatible with only one of the three packing states of the V(L) core suggests local 'breathing' of the variable domain. TrpH47, a conserved key residue of the V(H)/V(L) interface, is crucially involved in the formation of the antigen-binding site by adopting a novel conformation that specifically stabilizes the non-canonical CDR-L3 loop. Finally, a comparison with 7E2-cytochrome c oxidase complexes demonstrates that binding of this membrane-bound antigen proceeds without major conformational changes of the 7E2 antibody fragment.


Assuntos
Região Variável de Imunoglobulina/química , Proteínas de Membrana/química , Anticorpos/química , Sítios de Ligação , Fenômenos Químicos , Físico-Química , Cristalização , Cristalografia por Raios X , Bicamadas Lipídicas , Modelos Moleculares , Conformação Proteica , Receptores de Antígenos/química , Solubilidade
14.
J Biol Chem ; 278(17): 15162-7, 2003 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-12551909

RESUMO

Protein-protein recognition usually involves multiple interactions among different motifs that are scattered over protein surfaces. To identify such weak interactions, we have developed a novel double peptide synthesis (DS) method. This method allows us to map protein-protein interactions that involve two linear dis- continuous components from a polypeptide by the use of spatially addressable synergistic pairs of synthetic peptides. The DS procedure is based on the "SPOT" membrane-bound peptide synthesis technique, but to synthesize a mixture of two peptides, it uses both Fmoc (N-(9-fluorenyl)methoxycarbonyl))-alanine and Alloc-alanine at the first cycle. This allows their selective deprotection by either piperidine or tributyltin/palladium treatment, respectively. Using SPOT DS, we confirmed as a proof of principle that Elk-1 Ser(383) phosphorylation by ERK-2 kinase is stimulated by the presence of the Elk-1-docking domain. SPOT DS can also be used to dissect protein-protein motifs that define phosphatase substrate affinity. Using this technique, we identified three new regions in the insulin receptor that stimulate the dephosphorylation of the receptor by protein-tyrosine phosphatase (PTP) 1B and presumably increase the selectivity of PTP for this substrate. These data demonstrate that the SPOT DS technique allows the identification of non-linear weakly interacting protein motifs, which are an important determinant of protein kinase and phosphatase substrate specificity and of protein-protein interactions in general.


Assuntos
Proteínas de Ligação a DNA , Oligopeptídeos/metabolismo , Mapeamento de Peptídeos/métodos , Fatores de Transcrição , Técnicas de Química Combinatória , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Mutação , Oligopeptídeos/síntese química , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor de Insulina/metabolismo , Serina/metabolismo , Proteínas Elk-1 do Domínio ets
15.
Expert Opin Ther Targets ; 6(6): 637-47, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12472377

RESUMO

Protein tyrosine phosphatases (PTPs) control signal transduction pathways and have recently emerged as potential drug targets. Inhibition of individual PTPs can result in the activation of therapeutically relevant kinase cascades. This is particularly useful in cases where disease is associated with hormonal resistance, such as insensitivity to insulin or leptin. Currently, PTP1B is being investigated by a number of companies as a promising target for leptin/insulin mimetics and in the treatment of diabetes and obesity. Since all 90-100 PTPs have been identified in the human genome, the challenge now is to identify the function of these enzymes and the therapeutic indications that may exist for specific PTP inhibitors.


Assuntos
Proteínas Tirosina Fosfatases/antagonistas & inibidores , Permeabilidade Capilar/fisiologia , Quinases Ciclina-Dependentes/metabolismo , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/enzimologia , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Genes Supressores de Tumor , Humanos , Sistema Imunitário/enzimologia , Infecções/tratamento farmacológico , Infecções/enzimologia , Insulina/fisiologia , Leptina/fisiologia , Família Multigênica , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/fisiologia , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Obesidade/tratamento farmacológico , Obesidade/enzimologia , Osteoporose/tratamento farmacológico , Osteoporose/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/classificação , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/fisiologia , Receptores de Antígenos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA