Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Biomed Pharmacother ; 161: 114494, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36917886

RESUMO

Metastasis and chemoresistance in colorectal cancer are mediated by certain poorly differentiated cancer cells, known as cancer stem cells, that are maintained by Notch downstream signaling initiated upon Notch cleavage by the metalloprotease ADAM10. It has been shown that ADAM10 overexpression correlates with aberrant signaling from Notch, erbBs, and other receptors, as well as a more aggressive metastatic phenotype, in a range of cancers including colon, gastric, prostate, breast, ovarian, uterine, and leukemia. ADAM10 inhibition, therefore, stands out as an important and new approach to deter the progression of advanced CRC. For targeting the ADAM10 substrate-binding region, which is located outside of the catalytic domain of the protease, we generated a human anti-ADAM10 monoclonal antibody named 1H5. Structural and functional characterization of 1H5 reveals that it binds to the substrate-binding cysteine-rich domain and recognizes an activated ADAM10 conformation present on tumor cells. The mAb inhibits Notch cleavage and proliferation of colon cancer cell lines in vitro and in mouse models. Consistent with its binding to activated ADAM10, the mAb augments the catalytic activity of ADAM10 towards small peptide substrates in vitro. Most importantly, in a mouse model of colon cancer, when administered in combination with the therapeutic agent Irinotecan, 1H5 causes highly effective tumor growth inhibition without any discernible toxicity effects. Our singular approach to target the ADAM10 substrate-binding region with therapeutic antibodies could overcome the shortcomings of previous intervention strategies of targeting the protease active site with small molecule inhibitors that exhibit musculoskeletal toxicity.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Masculino , Camundongos , Animais , Humanos , Anticorpos Monoclonais/farmacologia , Proteínas de Membrana/metabolismo , Proteína ADAM10/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Secretases da Proteína Precursora do Amiloide/metabolismo
2.
Transl Oncol ; 15(1): 101265, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34768098

RESUMO

ADAM17 is upregulated in many cancers and in turn activates signaling pathways, including EGFR/ErbB, as well as those underlying resistance to targeted anti-EGFR therapies. Due to its central role in oncogenic pathways and drug resistance mechanisms, specific and efficacious monoclonal antibodies against ADAM17 could be useful for a broad patient population with solid tumors. Hence, we describe here an inhibitory anti-ADAM17 monoclonal antibody, named D8P1C1, that preferentially recognizes ADAM17 on cancer cells. D8P1C1 inhibits the catalytic activity of ADAM17 in a fluorescence-based peptide cleavage assay, as well as the proliferation of a range of cancer cell lines, including breast, ovarian, glioma, colon and the lung adenocarcinoma. In mouse models of triple-negative breast cancer and ovarian cancer, treatment with the mAb results in 78% and 45% tumor growth inhibition, respectively. Negative staining electron microscopy analysis of the ADAM17 ectodomain in complex with D8P1C1 reveals that the mAb binds the ADAM17 protease domain, consistent with its ability to inhibit the ADAM17 catalytic activity. Collectively, our results demonstrate the therapeutic potential of the D8P1C1 mAb to treat solid tumors.

3.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34638814

RESUMO

The Eph receptor tyrosine kinases and their ephrin ligands direct axon pathfinding and neuronal cell migration, as well as mediate many other cell-cell communication events. Their dysfunctional signaling has been shown to lead to various diseases, including cancer. The Ephs and ephrins both localize to the plasma membrane and, upon cell-cell contact, form extensive signaling assemblies at the contact sites. The Ephs and the ephrins are divided into A and B subclasses based on their sequence conservation and affinities for each other. The molecular details of Eph-ephrin recognition have been previously revealed and it has been documented that ephrin binding induces higher-order Eph assemblies, which are essential for full biological activity, via multiple, distinct Eph-Eph interfaces. One Eph-Eph interface type is characterized by a homotypic, head-to-tail interaction between the ligand-binding and the fibronectin domains of two adjacent Eph molecules. While the previous Eph ectodomain structural studies were focused on A class receptors, we now report the crystal structure of the full ectodomain of EphB2, revealing distinct and unique head-to-tail receptor-receptor interactions. The EphB2 structure and structure-based mutagenesis document that EphB2 uses the head-to-tail interactions as a novel autoinhibitory control mechanism for regulating downstream signaling and that these interactions can be modulated by posttranslational modifications.


Assuntos
Receptor EphB2/química , Receptor EphB2/metabolismo , Transdução de Sinais , Animais , Células HEK293 , Humanos , Camundongos , Domínios Proteicos , Receptor EphB2/genética , Relação Estrutura-Atividade
4.
PLoS One ; 16(3): e0247335, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33770085

RESUMO

Eph receptors are the largest group amongst the receptor tyrosine kinases and are divided into two subgroups, A and B, based on ligand binding specificities and sequence conservation. Through ligand-induced and ligand-independent activities, Ephs play central roles in diverse biological processes, including embryo development, regulation of neuronal signaling, immune responses, vasculogenesis, as well as tumor initiation, progression, and metastasis. The Eph extracellular regions (ECDs) are constituted of multiple domains, and previous structural studies of the A class receptors revealed how they interact with ephrin ligands and simultaneously mediate Eph-Eph clustering necessary for biological activity. Specifically, EphA structures highlighted a model, where clustering of ligand-bound receptors relies on two distinct receptor/receptor interfaces. Interestingly, most unliganded A class receptors also form an additional, third interface, between the ligand binding domain (LBD) and the fibronectin III domain (FN3) of neighboring molecules. Structures of B-class Eph ECDs, on the other hand, have never been reported. To further our understanding of Eph receptor function, we crystallized the EphB6-ECD and determined its three-dimensional structure using X-ray crystallography. EphB6 has important functions in both normal physiology and human malignancies and is especially interesting because this atypical receptor innately lacks kinase activity and our understanding of the mechanism of action is still incomplete. Our structural data reveals the overall EphB6-ECD architecture and shows EphB6-LBD/FN3 interactions similar to those observed for the unliganded A class receptors, suggesting that these unusual interactions are of general importance to the Eph group. We also observe unique structural features, which likely reflect the atypical signaling properties of EphB6, namely the need of co-receptor(s) for this kinase-inactive Eph. These findings provide new valuable information on the structural organization and mechanism of action of the B-class Ephs, and specifically EphB6, which in the future will assist in identifying clinically relevant targets for cancer therapy.


Assuntos
Receptor EphB6/ultraestrutura , Receptores da Família Eph/ultraestrutura , Linhagem Celular , Cristalografia por Raios X/métodos , Efrinas/metabolismo , Fibronectinas/metabolismo , Humanos , Ligantes , Fosforilação , Ligação Proteica/fisiologia , Domínios Proteicos/fisiologia , Receptor EphA1/metabolismo , Receptor EphA1/ultraestrutura , Receptor EphB6/metabolismo , Receptores da Família Eph/metabolismo , Transdução de Sinais
5.
Cancer Lett ; 467: 50-57, 2019 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-31593799

RESUMO

ADAM proteases are multi domain transmembrane metalloproteases that cleave a range of cell surface proteins and activate signaling pathways implicated in tumor progression, including those mediated by Notch, EFGR, and the Eph receptors. Consequently, they have emerged as key therapeutic targets in the efforts to inhibit tumor initiation and progression. To that end, two main approaches have been taken to develop ADAM antagonists: (i) small molecule inhibitors, and (ii) monoclonal antibodies. In this mini-review we describe the distinct features of ADAM proteases, particularly of ADAM10 and ADAM17, their domain organization, conformational rearrangements, regulation, as well as their emerging importance as therapeutic targets in cancer. Further, we highlight an anti-ADAM10 monoclonal antibody that we have recently developed, which has shown significant promise in inhibiting Notch signaling and deterring growth of solid tumors in pre-clinical settings.


Assuntos
Proteínas ADAM/química , Proteínas ADAM/metabolismo , Neoplasias/metabolismo , Proteínas ADAM/antagonistas & inibidores , Proteína ADAM10/antagonistas & inibidores , Proteína ADAM10/química , Proteína ADAM10/metabolismo , Proteína ADAM17/química , Proteína ADAM17/metabolismo , Animais , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Domínio Catalítico , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/tratamento farmacológico , Conformação Proteica , Domínios Proteicos
6.
Int J Biochem Cell Biol ; 105: 123-133, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30343150

RESUMO

The Eph-ephrin signaling pathway mediates developmental processes and the proper functioning of the adult human body. This distinctive bidirectional signaling pathway includes a canonical downstream signal cascade inside the Eph-bearing cells, as well as a reverse signaling in the ephrin-bearing cells. The signaling is terminated by ADAM metalloproteinase cleavage, internalization, and degradation of the Eph/ephrin complexes. Consequently, the Eph-ephrin-ADAM signaling cascade has emerged as a key target with immense therapeutic potential particularly in the context of cancer. An interesting twist was brought forth by the emergence of ephrins as the entry receptors for the pathological Henipaviruses, which has spurred new studies to target the viral entry. The availability of high-resolution structures of the multi-modular Eph receptors in complexes with ephrins and other binding partners, such as peptides, small molecule inhibitors and antibodies, offers a wealth of information for the structure-guided development of therapeutic intervention. Furthermore, genomic data mining of Eph mutants involved in cancer provides information for targeted drug development. In this review we summarize the distinct avenues for targeting the Eph-ephrin signaling pathway, including its termination by ADAM proteinases. We highlight the latest developments in Eph-related pharmacology in the context of Eph-ephrin-ADAM-based antibodies and small molecules. Finally, the future prospects of genomics- and proteomics-based medicine are discussed.


Assuntos
Efrinas/efeitos dos fármacos , Efrinas/metabolismo , Receptores da Família Eph/efeitos dos fármacos , Receptores da Família Eph/metabolismo , Proteínas ADAM/efeitos dos fármacos , Proteínas ADAM/metabolismo , Anticorpos/química , Anticorpos/farmacologia , Antineoplásicos/farmacologia , Sítios de Ligação , Desenvolvimento de Medicamentos , Efrinas/química , Humanos , Modelos Biológicos , Modelos Moleculares , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Receptores da Família Eph/genética , Transdução de Sinais/efeitos dos fármacos
7.
Genes Dev ; 29(8): 785-90, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25838500

RESUMO

The amyloid precursor protein (APP) has garnered considerable attention due to its genetic links to Alzheimer's disease. Death receptor 6 (DR6) was recently shown to bind APP via the protein extracellular regions, stimulate axonal pruning, and inhibit synapse formation. Here, we report the crystal structure of the DR6 ectodomain in complex with the E2 domain of APP and show that it supports a model for APP-induced dimerization and activation of cell surface DR6.


Assuntos
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Modelos Moleculares , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/metabolismo , Animais , Cristalização , Dimerização , Células HEK293 , Humanos , Camundongos , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Transdução de Sinais
8.
Science ; 344(6189): 1275-9, 2014 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-24876346

RESUMO

Netrins are secreted proteins that regulate axon guidance and neuronal migration. Deleted in colorectal cancer (DCC) is a well-established netrin-1 receptor mediating attractive responses. We provide evidence that its close relative neogenin is also a functional netrin-1 receptor that acts with DCC to mediate guidance in vivo. We determined the structures of a functional netrin-1 region, alone and in complexes with neogenin or DCC. Netrin-1 has a rigid elongated structure containing two receptor-binding sites at opposite ends through which it brings together receptor molecules. The ligand/receptor complexes reveal two distinct architectures: a 2:2 heterotetramer and a continuous ligand/receptor assembly. The differences result from different lengths of the linker connecting receptor domains fibronectin type III domain 4 (FN4) and FN5, which differs among DCC and neogenin splice variants, providing a basis for diverse signaling outcomes.


Assuntos
Axônios/fisiologia , Proteínas de Membrana/química , Fatores de Crescimento Neural/química , Receptores de Superfície Celular/química , Proteínas Supressoras de Tumor/química , Animais , Movimento Celular , Receptor DCC , Fibronectinas/química , Ligantes , Proteínas de Membrana/genética , Proteínas de Membrana/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/ultraestrutura , Receptores de Netrina , Netrina-1 , Neurônios/fisiologia , Multimerização Proteica , Estrutura Terciária de Proteína , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/ultraestrutura , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/ultraestrutura
9.
Proc Natl Acad Sci U S A ; 110(36): 14634-9, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-23959867

RESUMO

Eph receptor tyrosine kinases and their ephrin ligands mediate cell signaling during normal and oncogenic development. Eph signaling is initiated in a multistep process leading to the assembly of higher-order Eph/ephrin clusters that set off bidirectional signaling in interacting cells. Eph and ephrins are divided in two subclasses based on their abilities to bind and activate each other and on sequence conservation. EphA4 is an exception to the general rule because it can be activated by both A- and B-class ephrin ligands. Here we present high-resolution structures of the complete EphA4 ectodomain and its complexes with ephrin-A5. The structures reveal how ligand binding promotes conformational changes in the EphA4 ligand-binding domain allowing the formation of signaling clusters at the sites of cell-cell contact. In addition, the structural data, combined with structure-based mutagenesis, reveal a previously undescribed receptor-receptor interaction between the EphA4 ligand-binding and membrane-proximal fibronectin domains, which is functionally important for efficient receptor activation.


Assuntos
Efrina-A5/química , Estrutura Terciária de Proteína , Receptor EphA4/química , Transdução de Sinais , Sítios de Ligação/genética , Western Blotting , Cristalografia por Raios X , Ativação Enzimática , Efrina-A5/genética , Efrina-A5/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Receptor EphA4/genética , Receptor EphA4/metabolismo
10.
Structure ; 21(9): 1683-9, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-23891289

RESUMO

The leucine-rich repeat-containing G-protein-coupled receptors (Lgrs) are a large membrane protein family mediating signaling events during development and in the adult organism. Type 2 Lgrs, including Lgr4, Lgr5, and Lgr6, play crucial roles in embryonic development and in several cancers. They also regulate adult stem cell maintenance via direct association with proteins in the Wnt signaling pathways, including Lrp5/6 and frizzled receptors. The R-spondins (Rspo) were recently identified as functional ligands for type 2 Lgrs and were shown to synergize with both canonical and noncanonical Wnt signaling pathways. We determined and report the structure of the Lgr4 ectodomain alone and bound to Rspo1. The structures reveal an extended horseshoe leucine-rich repeat (LRR) receptor architecture that binds, with its concave side, the ligand furin-like repeats via an intimate interface. The molecular details of ligand/receptor recognition provide insight into receptor activation and could serve as template for stem-cell-based regenerative therapeutics development.


Assuntos
Receptores Acoplados a Proteínas G/química , Trombospondinas/química , Proteínas de Xenopus/química , Xenopus , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Células HEK293 , Humanos , Modelos Moleculares , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína
11.
Proc Natl Acad Sci U S A ; 110(18): 7205-10, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23592718

RESUMO

Angiogenesis is a complex cellular process involving multiple regulatory growth factors and growth factor receptors. Among them, the ligands for the endothelial-specific tunica intima endothelial receptor tyrosine kinase 2 (Tie2) receptor kinase, angiopoietin-1 (Ang1) and Ang2, play essential roles in balancing vessel stability and regression during both developmental and tumor-induced angiogenesis. Despite possessing a high degree of sequence identity, Ang1 and Ang2 have distinct functional roles and cell-signaling characteristics. Here, we present the crystal structures of Ang1 both unbound and in complex with the Tie2 ectodomain. Comparison of the Ang1-containing structures with their Ang2-containing counterparts provide insight into the mechanism of receptor activation and reveal molecular surfaces important for interactions with Tie2 coreceptors and associated signaling proteins. Using structure-based mutagenesis, we identify a loop within the angiopoietin P domain, adjacent to the receptor-binding interface, which confers the specific agonist/antagonist properties of the molecule. We demonstrate using cell-based assays that an Ang2 chimera containing the Ang1 loop sequence behaves functionally similarly to Ang1 as a constitutive Tie2 agonist, able to efficiently dissociate the inhibitory Tie1/Tie2 complex and elicit Tie2 clustering and downstream signaling.


Assuntos
Angiopoietina-1/química , Angiopoietina-1/metabolismo , Transdução de Sinais , Angiopoietina-2/química , Angiopoietina-2/metabolismo , Sequência Conservada , Cristalografia por Raios X , Células HEK293 , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína , Receptor de TIE-1/química , Receptor de TIE-1/metabolismo , Receptor TIE-2/química , Receptor TIE-2/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Eletricidade Estática , Relação Estrutura-Atividade
12.
Mol Cell ; 37(5): 643-55, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20227369

RESUMO

The Tie family of endothelial-specific receptor tyrosine kinases is essential for cell proliferation, migration, and survival during angiogenesis. Despite considerable similarity, experiments with Tie1- or Tie2-deficient mice highlight distinct functions for these receptors in vivo. The Tie2 receptor is further unique with respect to its structurally homologous ligands. Angiopoietin-2 and -3 can function as agonists or antagonists; angiopoietin-1 and -4 are constitutive agonists. To address the role of Tie1 in angiopoietin-mediated Tie2 signaling and determine the basis for the behavior of the individual angiopoietins, we used an in vivo FRET-based proximity assay to monitor Tie1 and -2 localization and association. We provide evidence for Tie1-Tie2 complex formation on the cell surface and identify molecular surface areas essential for receptor-receptor recognition. We further demonstrate that the Tie1-Tie2 interactions are dynamic, inhibitory, and differentially modulated by angiopoietin-1 and -2. Based on the available data, we propose a unified model for angiopoietin-induced Tie2 signaling.


Assuntos
Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Células Endoteliais/enzimologia , Receptor de TIE-1/metabolismo , Receptor TIE-2/metabolismo , Transdução de Sinais , Linhagem Celular , Membrana Celular/enzimologia , Transferência Ressonante de Energia de Fluorescência , Humanos , Ligantes , Modelos Moleculares , Mutação , Conformação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Interferência de RNA , Receptor Cross-Talk , Receptor de TIE-1/química , Receptor de TIE-1/genética , Receptor TIE-2/química , Receptor TIE-2/genética , Proteínas Recombinantes de Fusão/metabolismo , Relação Estrutura-Atividade , Fatores de Tempo , Transfecção
13.
Protein Sci ; 15(8): 2008-13, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16823037

RESUMO

The advent of the multiwavelength anomalous diffraction phasing method has significantly accelerated crystal structure determination and has become the norm in protein crystallography. This method allows researchers to take advantage of the anomalous signal from diverse atoms, but the dominant method for derivative preparation is selenomethionine substitution. Several generally applicable, high-efficiency labeling protocols have been developed for use in the bacterial, yeast, and baculovirus/insect cell expression systems but not for mammalian tissue culture. As a large number of proteins of biomedical importance can only be produced in yields sufficient for X-ray diffraction experiments in mammalian expression systems, it becomes all the more important to develop such protocols. We therefore evaluated several variables that play roles in determining incorporation levels and report here a simple protocol for selenomethionine modification of proteins in mammalian cells routinely yielding >90% labeling efficiency.


Assuntos
Substituição de Aminoácidos , Cristalografia por Raios X/métodos , Proteínas Recombinantes/biossíntese , Selenometionina/metabolismo , Coloração e Rotulagem/métodos , Técnicas de Cultura de Células/métodos , Linhagem Celular , Cristalização , Meios de Cultura , Humanos , Receptor TIE-2/biossíntese , Proteínas Recombinantes/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
14.
Nat Struct Mol Biol ; 13(6): 524-32, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16732286

RESUMO

The Tie receptor tyrosine kinases and their angiopoietin (Ang) ligands play central roles in developmental and tumor-induced angiogenesis. Here we present the crystal structures of the Tie2 ligand-binding region alone and in complex with Ang2. In contrast to prediction, Tie2 contains not two but three immunoglobulin (Ig) domains, which fold together with the three epidermal growth factor domains into a compact, arrowhead-shaped structure. Ang2 binds at the tip of the arrowhead utilizing a lock-and-key mode of ligand recognition-unique for a receptor kinase-where two complementary surfaces interact with each other with no domain rearrangements and little conformational change in either molecule. Ang2-Tie2 recognition is similar to antibody-protein antigen recognition, including the location of the ligand-binding site within the Ig fold. Analysis of the structures and structure-based mutagenesis provide insight into the mechanism of receptor activation and support the hypothesis that all angiopoietins interact with Tie2 in a structurally similar manner.


Assuntos
Angiopoietina-2/química , Receptor TIE-2/química , Sequência de Aminoácidos , Cálcio/química , Cálcio/metabolismo , Cromatografia em Gel , Cristalografia por Raios X , Fator de Crescimento Epidérmico/química , Fibrinogênio/química , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Receptor TIE-2/metabolismo , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA