Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Mol Pharm ; 16(3): 1350-1357, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30742442

RESUMO

Integrin αvß3 has been considered as a promising biomarker for vulnerable atherosclerotic plaques, and it is highly expressed by those instability-associated factors, such as macrophages, vessel endothelial cells, and smooth muscle cells. Our previous study successfully showed that the 64Cu-labeled divalent (containing two RGD motifs) cystine knot peptide, 64Cu-NOTA-3-4A, had high binding affinity and specificity in targeting vulnerable carotid atherosclerotic plaques with increased αvß3 levels. Therefore, considering that 68Ga has excellent nuclear physical properties for positron emission tomography (PET), this study aimed to investigate the feasibility of using 68Ga-NOTA-3-4A for PET study of vulnerable atherosclerotic plaques. The vulnerable carotid atherosclerotic plaques were induced and maintained in ApoE-/- mice through carotid artery ligation and a high-fat diet. Divalent knottin peptide 3-4A was synthesized through solid-phase peptide synthesis chemistry and radiolabeled with 68Ga after being conjugated with 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA). The probe stability was analyzed in phosphate buffered saline (PBS) buffer and mouse serum. ApoE-/- mice with atherosclerotic plaques ( n = 4) were imaged by PET/CT at 1 and 2 h after the tail vein injection of 68Ga-NOTA-3-4A. The targeting specificity was determined by coinjection of 68Ga-NOTA-3-4A and nonradioactive c(RGDyK) peptide. The carotid artery tissues were removed, and immunofluorescent staining was performed to evaluate αvß3 integrin expression. It was found that 68Ga-NOTA-3-4A displayed high stability in both PBS buffer and mouse serum. Small animal PET/CT images and quantification analysis indicated the quick and high plaque uptake of 68Ga-NOTA-3-4A (6.67 ± 1.44 and 2.97 ± 0.46%ID/g at 1 and 2 h, respectively). The plaque-to-normal artery ratio was 15.88 and 9.90 at 1 and 2 h, respectively. Furthermore, the plaque accumulation of 68Ga-NOTA-3-4A was significantly inhibited via coinjection of c(RGDyK). Finally, immunostaining identified integrin αvß3 expressed by macrophages, vessel endothelial cells, and smooth muscle cells. In summary, 68Ga-NOTA-3-4A has high potential to be a promising PET tracer for imaging vulnerable atherosclerotic plaques.


Assuntos
Miniproteínas Nó de Cistina/metabolismo , Cistina/química , Radioisótopos de Gálio/metabolismo , Placa Aterosclerótica/diagnóstico por imagem , Traçadores Radioativos , Animais , Artérias Carótidas/cirurgia , Miniproteínas Nó de Cistina/sangue , Miniproteínas Nó de Cistina/síntese química , Dieta Hiperlipídica/efeitos adversos , Estabilidade de Medicamentos , Radioisótopos de Gálio/sangue , Integrina alfaVbeta3/metabolismo , Marcação por Isótopo , Ligadura/efeitos adversos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout para ApoE , Placa Aterosclerótica/etiologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos/metabolismo
2.
Adv Exp Med Biol ; 917: 121-44, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27236555

RESUMO

Peptides with the cystine-knot architecture, often termed knottins, are promising scaffolds for biomolecular engineering. These unique molecules combine diverse bioactivities with excellent structural, thermal, and proteolytical stability. Being different in the composition and structure of their amino acid backbone, knottins share the same core element, namely cystine knot, which is built by six cysteine residues forming three disulfides upon oxidative folding. This motif ensures a notably rigid framework that highly tolerates both rational and combinatorial changes in the primary structure. Being accessible through recombinant production and total chemical synthesis, cystine-knot miniproteins can be endowed with novel bioactivities by variation of surface-exposed loops and incorporation of non-natural elements within their non-conserved regions towards the generation of tailor-made peptidic compounds. In this chapter the topology of cystine-knot peptides, their synthesis and applications for diagnostics and therapy is discussed.


Assuntos
Miniproteínas Nó de Cistina/uso terapêutico , Desenho de Fármacos , Neoplasias/tratamento farmacológico , Fragmentos de Peptídeos/uso terapêutico , Engenharia de Proteínas/métodos , Animais , Miniproteínas Nó de Cistina/síntese química , Humanos , Neoplasias/diagnóstico
3.
J Am Chem Soc ; 137(1): 6-9, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25486381

RESUMO

Molecules that target and inhibit αvß3, αvß5, and α5ß1 integrins have generated great interest because of the role of these receptors in mediating angiogenesis and metastasis. Attempts to increase the binding affinity and hence the efficacy of integrin inhibitors by dimerization have been marginally effective. In the present work, we achieved this goal by using oxime-based chemical conjugation to synthesize dimers of integrin-binding cystine knot (knottin) miniproteins with low-picomolar binding affinity to tumor cells. A non-natural amino acid containing an aminooxy side chain was introduced at different locations within a knottin monomer and reacted with dialdehyde-containing cross-linkers of different lengths to create knottin dimers with varying molecular topologies. Dimers cross-linked through an aminooxy functional group located near the middle of the protein exhibited higher apparent binding affinity to integrin-expressing tumor cells compared with dimers cross-linked through an aminooxy group near the C-terminus. In contrast, the cross-linker length had no effect on the integrin binding affinity. A chemical-based dimerization strategy was critical, as knottin dimers created through genetic fusion to a bivalent antibody domain exhibited only modest improvement (less than 5-fold) in tumor cell binding relative to the knottin monomer. The best oxime-conjugated knottin dimer achieved an unprecedented 150-fold increase in apparent binding affinity over the knottin monomer. Also, this dimer bound 3650-fold stronger and inhibited tumor cell migration and proliferation compared with cilengitide, an integrin-targeting peptidomimetic that performed poorly in recent clinical trials, suggesting promise for further therapeutic development.


Assuntos
Movimento Celular/efeitos dos fármacos , Reagentes de Ligações Cruzadas/química , Miniproteínas Nó de Cistina/química , Miniproteínas Nó de Cistina/farmacologia , Integrinas/antagonistas & inibidores , Integrinas/química , Neoplasias/patologia , Multimerização Proteica , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Miniproteínas Nó de Cistina/síntese química , Relação Dose-Resposta a Droga , Humanos , Integrinas/metabolismo , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
4.
Molecules ; 17(11): 12533-52, 2012 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-23095896

RESUMO

Cystine-knot peptides display exceptional structural, thermal, and biological stability. Their eponymous motif consists of six cysteine residues that form three disulfide bonds, resulting in a notably rigid structural core. Since they highly tolerate either rational or combinatorial changes in their primary structure, cystine knots are considered to be promising frameworks for the development of peptide-based pharmaceuticals. Despite their relatively small size (two to three dozens amino acid residues), the chemical synthesis route is challenging since it involves critical steps such as head-to-tail cyclization and oxidative folding towards the respective bioactive isomer. Herein we describe the topology of cystine-knot peptides, their synthetic availability and briefly discuss potential applications of engineered variants in diagnostics and therapy.


Assuntos
Miniproteínas Nó de Cistina/síntese química , Sequência de Aminoácidos , Ciclização , Motivos Nó de Cisteína , Miniproteínas Nó de Cistina/química , Desenho de Fármacos , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Oxirredução , Dobramento de Proteína , Estabilidade Proteica
5.
Methods Enzymol ; 503: 223-51, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22230571

RESUMO

Cystine-knot miniproteins, also known as knottins, contain a conserved core of three tightly woven disulfide bonds which impart extraordinary thermal and proteolytic stability. Interspersed between their conserved cysteine residues are constrained loops that possess high levels of sequence diversity among knottin family members. Together these attributes make knottins promising molecular scaffolds for protein engineering and translational applications. While naturally occurring knottins have shown potential as both diagnostic agents and therapeutics, protein engineering is playing an important and increasing role in creating designer molecules that bind to a myriad of biomedical targets. Toward this goal, rational and combinatorial approaches have been used to engineer knottins with novel molecular recognition properties. Here, methods are described for creating and screening knottin libraries using yeast surface display and fluorescence-activated cell sorting. Protocols are also provided for producing knottins by synthetic and recombinant methods, and for measuring the binding affinity of knottins to target proteins expressed on the cell surface.


Assuntos
Proteínas de Transporte/química , Miniproteínas Nó de Cistina/química , Biblioteca de Peptídeos , Engenharia de Proteínas/métodos , Marcadores de Afinidade/química , Animais , Proteínas de Transporte/síntese química , Proteínas de Transporte/genética , Proteínas de Transporte/isolamento & purificação , Cisteína/química , Miniproteínas Nó de Cistina/síntese química , Miniproteínas Nó de Cistina/genética , Miniproteínas Nó de Cistina/isolamento & purificação , DNA/química , DNA/genética , Citometria de Fluxo , Corantes Fluorescentes/química , Humanos , Oligonucleotídeos/química , Oligonucleotídeos/genética , Fases de Leitura Aberta , Pichia/química , Plasmídeos/química , Ligação Proteica , Dobramento de Proteína , Receptores de Superfície Celular/química , Proteínas Recombinantes/síntese química , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Técnicas de Síntese em Fase Sólida , Especificidade por Substrato , Leveduras/química
6.
Clin Cancer Res ; 18(3): 839-49, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22173551

RESUMO

PURPOSE: Detection of pancreatic cancer remains a high priority and effective diagnostic tools are needed for clinical applications. Many cancer cells overexpress integrin α(v)ß(6), a cell surface receptor being evaluated as a novel clinical biomarker. EXPERIMENTAL DESIGN: To validate this molecular target, several highly stable cystine knot peptides were engineered by directed evolution to bind specifically and with high affinity (3-6 nmol/L) to integrin α(v)ß(6). The binders do not cross-react with related integrin α(v)ß(5), integrin α(5)ß(1), or tumor-angiogenesis-associated integrin, α(v)ß(3). RESULTS: Positron emission tomography showed that these disulfide-stabilized peptides rapidly accumulate at tumors expressing integrin α(v)ß(6). Clinically relevant tumor-to-muscle ratios of 7.7 ± 2.4 to 11.3 ± 3.0 were achieved within 1 hour after radiotracer injection. Minimization of off-target dosing was achieved by reformatting α(v)ß(6)-binding activities across various natural and pharmacokinetically stabilized cystine knot scaffolds with different amino acid content. We show that the primary sequence of a peptide scaffold directs its pharmacokinetics. Scaffolds with high arginine or glutamic acid content suffered high renal retention of more than 75% injected dose per gram (%ID/g). Substitution of these amino acids with renally cleared amino acids, notably serine, led to significant decreases in renal accumulation of less than 20%ID/g 1 hour postinjection (P < 0.05, n = 3). CONCLUSIONS: We have engineered highly stable cystine knot peptides with potent and specific integrin α(v)ß(6)-binding activities for cancer detection. Pharmacokinetic engineering of scaffold primary sequence led to significant decreases in off-target radiotracer accumulation. Optimization of binding affinity, specificity, stability, and pharmacokinetics will facilitate translation of cystine knots for cancer molecular imaging.


Assuntos
Antígenos de Neoplasias/metabolismo , Miniproteínas Nó de Cistina/farmacocinética , Integrinas/metabolismo , Neoplasias Pancreáticas/diagnóstico , Compostos Radiofarmacêuticos/farmacocinética , Animais , Bioengenharia , Biomarcadores Tumorais/análise , Miniproteínas Nó de Cistina/síntese química , Miniproteínas Nó de Cistina/química , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Ligação Proteica , Compostos Radiofarmacêuticos/síntese química , Distribuição Tecidual
7.
Curr Pharm Des ; 17(38): 4329-36, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22204431

RESUMO

Cystine knot miniproteins define a class of peptides in the size range of approximately 28-35 amino acid residues that often combine high chemical and biological stability with high potency and selectivity. They share a common structural motif that is defined by three intramolecular disulfide bonds that gives rise to a very stable scaffold. Members of this family cover a broad spectrum of natural bioactivities ranging from antimicrobial and antiviral activities to selective blockage or activation of ion channels, cell surface receptors and extracelluar proteases. In recent years, the spectrum of natural bioactivities of this class of miniproteins was further expanded by application of protein design and directed evolution technologies. Miniproteins have been developed that inhibit platelet aggregation, block asthma-related proteases, act as growth factor mimics or address human tumor targets. Recent reports on miniproteins binding to cancer specific targets indicate that these biomolecules due to their particularly high in vivo stability, high target affinity, good tissue distribution, and fast body clearance are very promising agents that can be endowed with important beneficial features for imaging and therapeutic applications. With the first cystine-knot miniprotein already marketed as an analgesic, more candidates can be expected to find their way into the clinic for diagnostic and therapeutic applications over next years.


Assuntos
Miniproteínas Nó de Cistina , Descoberta de Drogas , Engenharia de Proteínas , Administração Oral , Sequência de Aminoácidos , Animais , Fenômenos Biofísicos , Miniproteínas Nó de Cistina/síntese química , Miniproteínas Nó de Cistina/genética , Miniproteínas Nó de Cistina/uso terapêutico , Estabilidade de Medicamentos , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Inibidores da Agregação Plaquetária/síntese química , Inibidores da Agregação Plaquetária/química , Inibidores da Agregação Plaquetária/farmacologia , Inibidores de Proteases/síntese química , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Conformação Proteica , Estabilidade Proteica , Técnicas de Síntese em Fase Sólida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA