Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 296: 100447, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33617878

RESUMO

The fibronectin type III (FN3) monobody domain is a promising non-antibody scaffold, which features a less complex architecture than an antibody while maintaining analogous binding loops. We previously developed FN3Con, a hyperstable monobody derivative with diagnostic and therapeutic potential. Prestabilization of the scaffold mitigates the stability-function trade-off commonly associated with evolving a protein domain toward biological activity. Here, we aimed to examine if the FN3Con monobody could take on antibody-like binding to therapeutic targets, while retaining its extreme stability. We targeted the first of the Adnectin derivative of monobodies to reach clinical trials, which was engineered by directed evolution for binding to the therapeutic target VEGFR2; however, this function was gained at the expense of large losses in thermostability and increased oligomerization. In order to mitigate these losses, we grafted the binding loops from Adnectin-anti-VEGFR2 (CT-322) onto the prestabilized FN3Con scaffold to produce a domain that successfully bound with high affinity to the therapeutic target VEGFR2. This FN3Con-anti-VEGFR2 construct also maintains high thermostability, including remarkable long-term stability, retaining binding activity after 2 years of storage at 36 °C. Further investigations into buffer excipients doubled the presence of monomeric monobody in accelerated stability trials. These data suggest that loop grafting onto a prestabilized scaffold is a viable strategy for the development of monobody domains with desirable biophysical characteristics and that FN3Con is therefore well-suited to applications such as the evolution of multiple paratopes or shelf-stable diagnostics and therapeutics.


Assuntos
Anticorpos/metabolismo , Domínio de Fibronectina Tipo III/genética , Anticorpos/imunologia , Domínio de Fibronectina Tipo III/imunologia , Fibronectinas/genética , Fibronectinas/imunologia , Fibronectinas/metabolismo , Engenharia Genética/métodos , Humanos , Regiões de Interação com a Matriz , Mutação , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
2.
Elife ; 92020 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-32838837

RESUMO

Endothelial Tie2 signaling plays a pivotal role in vascular barrier maintenance at baseline and after injury. We previously demonstrated that a sharp drop in Tie2 expression observed across various murine models of critical illnesses is associated with increased vascular permeability and mortality. Matrix metalloprotease (MMP)-14-mediated Tie2 ectodomain shedding has recently been recognized as a possible mechanism for Tie2 downregulation in sepsis. Here, we identified the exact MMP14-mediated Tie2 ectodomain cleavage sites and could show that pharmacological MMP14 blockade in experimental murine sepsis exerts barrier protective and anti-inflammatory effects predominantly through the attenuation of Tie2 cleavage to improve survival both in a pre-treatment and rescue approach. Overall, we show that protecting Tie2 shedding might offer a new therapeutic opportunity for the treatment of septic vascular leakage.


Assuntos
Receptor TIE-2 , Sepse , Animais , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Modelos Animais de Doenças , Domínio de Fibronectina Tipo III/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Metaloproteinase 14 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor TIE-2/química , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Sepse/metabolismo , Sepse/fisiopatologia , Transdução de Sinais/efeitos dos fármacos
3.
Cells ; 9(7)2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32640697

RESUMO

Apoptotic cells expressing phosphatidylserine (PS) on their cell surface are directly or indirectly recognized by phagocytes through PS-binding proteins. The PS-binding protein Tim-4 secures apoptotic cells to phagocytes to facilitate the engulfment of apoptotic cells. However, the molecular mechanism by which Tim-4 transduces signals to phagocytes during Tim-4-mediated efferocytosis is incompletely understood. Here, we report that Tim-4 collaborates with Mertk during efferocytosis through a biochemical interaction with Mertk. Proximal localization between the two proteins in phagocytes was observed by immunofluorescence and proximal ligation assays. Physical association between Tim-4 and Mertk, which was mediated by an interaction between the IgV domain of Tim-4 and the fibronectin type-III domain of Mertk, was also detected with immunoprecipitation. Furthermore, the effect of Mertk on Tim-4-mediated efferocytosis was abolished by GST-MertkFnIII, a soluble form of the fibronectin type-III domain of Mertk that disrupts the interaction between Tim-4 and Mertk. Taken together, the results from our study suggest that a physical interaction between Tim-4 and Mertk is necessary for Mertk to enhance efferocytosis mediated by Tim-4.


Assuntos
Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , c-Mer Tirosina Quinase/química , c-Mer Tirosina Quinase/metabolismo , Animais , Domínio de Fibronectina Tipo III/genética , Domínio de Fibronectina Tipo III/fisiologia , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Proteínas de Membrana/genética , Camundongos , Camundongos Mutantes , Fagocitose/genética , Fagocitose/fisiologia , Ligação Proteica , c-Mer Tirosina Quinase/genética
4.
Parasit Vectors ; 13(1): 202, 2020 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-32307003

RESUMO

BACKGROUND: Malaria vector mosquitoes acquire midgut microbiota primarily from their habitat. The homeostasis of these microbial communities plays an essential role in the mosquito longevity, the most essential factor in the mosquito vectorial capacity. Our recent study revealed that silencing genes involved in regulation of the midgut homeostasis including FN3D1, FN3D3 and GPRGr9 reduced the survival of female adult Anopheles arabiensis mosquitoes. In the present study, we investigate the stability of the gene silencing efficiency of mosquitoes reared in three different breeding conditions representing distinct larval habitat types: town brick pits in Jimma, flood pools in the rural land of Asendabo and roadside pools in Wolkite. METHODS: First-instar larvae of An. arabiensis mosquitoes were reared separately using water collected from the three breeding sites. The resulting adult females were micro-injected with dsRNA targeting the FN3D1 gene (AARA003032) and their survival was monitored. Control mosquitoes were injected with dsRNA Lacz. In addition, the load of midgut microbiota of these mosquitoes was determined using flow cytometry. RESULTS: Survival of naïve adult female mosquitoes differed between the three sites. Mosquitoes reared using water collected from brick pits and flood pools survived longer than mosquitoes reared using water collected from roadside. However, the FN3D1 gene silencing effect on survival did not differ between the three sites. CONCLUSIONS: The present study revealed that the efficacy of FN3D1 gene silencing is not affected by variation in the larval habitat. Thus, silencing this gene has potential for application throughout sub-Saharan Africa.


Assuntos
Anopheles/genética , Domínio de Fibronectina Tipo III/genética , Interferência de RNA/fisiologia , Animais , Anopheles/fisiologia , Cruzamento , Ecossistema , Larva/genética , Larva/fisiologia , Controle de Mosquitos/métodos , Mosquitos Vetores/genética , Mosquitos Vetores/fisiologia
5.
Tissue Cell ; 63: 101323, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32223958

RESUMO

Previous reports showed that fibronectin (FN) was effective in stimulating the recovery of damaged dermis. However, native FN has multifunctional domains transmitting beneficial as well as unbeneficial signals to dermal tissue cells through the mediation of integrin heterodimers. The use of a functional domain [FN type III9-10 fragments (FNIII9-10)] providing beneficial effects on the physiology of dermal tissue cells would enhance an in vitro culture system for dermal fibroblasts (DFs). We therefore investigated the FNIII9-10-derived extracellular signaling effect on the physiology of DFs during in vitro culture. Recombinant FNIII9-10 proteins were constructed and their functionality was determined by observing the adhesion of adult human DFs (aHDFs) to recombinant FNIII9-10 and of low adhesion integrin α5ß1- and αvß3-blocked aHDFs to recombinant FNIII9-10. Cellular proliferation, morphology, and senescence were measured and compared in the aHDFs cultured on native FN and recombinant FNIII9-10 for short or long periods. The results show that recombinant FNIII9-10-derived extracellular signaling stimulated increased proliferation of aHDF (both in short- and long-term cultures) and inhibited the generation of morphological abnormalities (in short- and long-term cultures) and cellular senescence (long-term culture) when compared with native FN-derived extracellular signaling. Our results suggest that, instead of native FN, recombinant FNIII9-10 better enhanced the in vitro culture of aHDFs while diminishing the adverse effects associated with the use of human-derived materials.


Assuntos
Senescência Celular/genética , Derme/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibronectinas/genética , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Proliferação de Células/genética , Células Cultivadas , Derme/metabolismo , Fibroblastos/metabolismo , Domínio de Fibronectina Tipo III/genética , Fibronectinas/farmacologia , Humanos , Integrina alfa5beta1/genética , Transdução de Sinais/genética
6.
Biotechnol Lett ; 41(11): 1265-1274, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31541332

RESUMO

OBJECTIVE: To improve the production yield of N-glycosylated anti-VEGFR2 (vascular endothelial growth factor receptor 2) monobody (FN3VEGFR2-Gly) in lpp knockout Escherichia coli cells harboring Campylobacter jejuni N-glycosylation pathway. RESULTS: The leaky CLM37-Δlpp strain efficiently secreted FN3VEGFR2-Gly into culture medium. The extracellular levels of glycosylated FN3VEGFR2-Gly in CLM37-Δlpp culture medium were approximately 11 and 15 times higher compared to those in CLM37 cells via IPTG and auto-induction, respectively. In addition, the highest level of total glycosylated FN3VEGFR2-Gly (70 ± 3.4 mg/L) was found in culture medium via auto-induction. Furthermore, glycosylated FN3VEGFR2-Gly was more stable than unglycosylated FN3VEGFR2-Gly in this expression system, but their bioactivities were relatively similar. CONCLUSIONS: Lpp knockout leaky E. coli strain combined with auto-induction method can enhance the extracellular production of homogenous N-glycosylated FN3VEGFR2-Gly, and facilitate the downstream protein purification. The findings of this study may provide practical implications for the large-scale production and cost-effective harvesting of N-glycosylation proteins.


Assuntos
Anticorpos , Escherichia coli/genética , Espaço Extracelular/metabolismo , Domínio de Fibronectina Tipo III/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Anticorpos/química , Anticorpos/genética , Anticorpos/imunologia , Anticorpos/metabolismo , Glicosilação , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
7.
Nano Lett ; 19(9): 6124-6132, 2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-31389705

RESUMO

We describe a genetically encoded micelle for targeted delivery consisting of a diblock polypeptide with segments derived from repetitive protein motifs inspired by Drosophila melanogaster Rec-1 resilin and human tropoelastin with a C-terminal fusion of an integrin-targeting fibronectin type III domain. By systematically varying the weight fraction of the hydrophilic elastin-like polypeptide (ELP) block and molecular weight of the diblock polypeptide, we designed micelles of different morphologies that modulate the binding avidity of the human wild-type 10th fibronectin domain (Fn3) as a function of shape. We show that wormlike micelles that present the Fn3 domain have a 1000-fold greater avidity for the αvß3 receptor compared to the monomer ligand and an avidity that is greater than a clinically relevant antibody that is driven by their multivalency. The amplified avidity of these micelles leads to significantly increased cellular internalization, a feature that may have utility for the intracellular delivery of drugs that are loaded into the core of these micelles.


Assuntos
Proteínas de Drosophila/química , Sistemas de Liberação de Medicamentos , Fibronectinas/química , Nanopartículas/química , Tropoelastina/química , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/química , Drosophila melanogaster/genética , Elastina/química , Elastina/genética , Domínio de Fibronectina Tipo III/genética , Fibronectinas/genética , Humanos , Ligantes , Micelas , Peptídeos/química , Peptídeos/farmacologia , Temperatura , Tropoelastina/genética
8.
Methods Mol Biol ; 2033: 301-313, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31332762

RESUMO

Bioconjugation of biologically useful proteins is in great demand (e.g., conjugation to biotins, metal chelators, and drug carriers to target specific tissues for both in vitro and in vivo use). These conjugates provide widespread opportunities for various biological and biomedical applications. Evolving state-of-the-art protein conjugation strategies have led to the development of many affinity ligands, including for cancer imaging and diagnosis. However, to achieve the desirable protein conjugates, there are many challenges that remain to be addressed in order to obtain a reproducible procedure for all proteins and ligands. These include a control over the protein modification and the efficiency of the conjugation while retaining the original biological protein affinity postmodification. Here we present detailed conjugation methods for the human fibronectin tenth type III domain (FN3) protein scaffold for use in preclinical PET imaging. More specifically, this chapter provides detailed methods to produce a FN3 and a FN3-chelator-conjugate, its labeling with the radionuclide 64-Cu, and its use for noninvasive PET imaging in mice.


Assuntos
Portadores de Fármacos/química , Domínio de Fibronectina Tipo III/genética , Imagem Molecular/métodos , Neoplasias/diagnóstico , Animais , Biotina/química , Linhagem Celular Tumoral , Quelantes/química , Radioisótopos de Cobre/química , Humanos , Ligantes , Camundongos , Neoplasias/genética
9.
Proc Natl Acad Sci U S A ; 116(28): 13937-13942, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31239342

RESUMO

Despite being the subject of intense effort and scrutiny, kinases have proven to be consistently challenging targets in inhibitor drug design. A key obstacle has been promiscuity and consequent adverse effects of drugs targeting the ATP binding site. Here we introduce an approach to controlling kinase activity by using monobodies that bind to the highly specific regulatory allosteric pocket of the oncoprotein Aurora A (AurA) kinase, thereby offering the potential for more specific kinase modulators. Strikingly, we identify a series of highly specific monobodies acting either as strong kinase inhibitors or activators via differential recognition of structural motifs in the allosteric pocket. X-ray crystal structures comparing AurA bound to activating vs inhibiting monobodies reveal the atomistic mechanism underlying allosteric modulation. The results reveal 3 major advantages of targeting allosteric vs orthosteric sites: extreme selectivity, ability to inhibit as well as activate, and avoidance of competing with ATP that is present at high concentrations in the cells. We envision that exploiting allosteric networks for inhibition or activation will provide a general, powerful pathway toward rational drug design.


Assuntos
Aurora Quinase A/química , Aurora Quinase B/química , Inibidores de Proteínas Quinases/química , Proteínas Quinases/química , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Regulação Alostérica/genética , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/genética , Aurora Quinase B/antagonistas & inibidores , Aurora Quinase B/genética , Sítios de Ligação/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Cristalografia por Raios X , Desenho de Fármacos , Domínio de Fibronectina Tipo III/genética , Humanos , Conformação Proteica , Proteínas Quinases/genética
10.
PLoS One ; 14(1): e0210193, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30629639

RESUMO

The human natural killer-1 (HNK-1) carbohydrate epitope, composed of a unique sulfated trisaccharide (HSO3-3GlcAß1-3Galß1-4GlcNAc-R), is highly expressed during brain development and regulates higher brain function. However, it remains unclear which glycoprotein carries the HNK-1 epitope in the embryonic brain and the functional role it plays. Here, we showed that one of the major HNK-1 carrier proteins in the embryonic brain is tenascin-C (TNC), an extracellular matrix protein that regulates neurite outgrowth by interacting with the GPI-anchored protein contactin-1 (CNTN). Because the alternatively spliced fibronectin type-III (FNIII) repeats in TNC give rise to many isoforms and affect neuronal function, we evaluated neurite outgrowth of primary hippocampal neurons on purified recombinant FNIII repeats with or without the HNK-1 epitope as a substrate. We found that the presence of the HNK-1 epitope on the C domain of TNC promoted neurite outgrowth, and that this signal was mediated by CNTN, which is an HNK-1-expressing neuronal receptor. The neurite-promoting activity of the HNK-1 epitope on TNC required neuronal HNK-1 expression, which was defective in neurons lacking the glucuronyltransferases GlcAT-P and GlcAT-S. These results suggest that the HNK-1 epitope is a key modifier of TNC and CNTN in the regulation of embryonic brain development.


Assuntos
Antígenos CD57/imunologia , Contactina 1/fisiologia , Hipocampo/crescimento & desenvolvimento , Crescimento Neuronal/imunologia , Tenascina/imunologia , Processamento Alternativo/imunologia , Animais , Embrião de Mamíferos , Epitopos/imunologia , Domínio de Fibronectina Tipo III/genética , Domínio de Fibronectina Tipo III/imunologia , Glucuronosiltransferase/genética , Células HEK293 , Hipocampo/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuritos/fisiologia , Crescimento Neuronal/genética , Cultura Primária de Células , Tenascina/genética
11.
Clin Cancer Res ; 25(6): 1774-1785, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30373750

RESUMO

PURPOSE: To design and evaluate a small engineered protein binder targeting human programmed death-1 ligand (hPD-L1) in vivo for PET imaging in four mouse tumor models, and in situ in human cancer specimens.Experimental Design: The hPD-L1 protein binder, FN3hPD-L1, was engineered using a 12-kDa human fibronectin type-3 domain (FN3) scaffold. The binder's affinity was assayed in CT26 mouse colon carcinoma cells stably expressing hPD-L1 (CT26/hPD-L1). 64Cu-FN3hPD-L1 was assayed for purity, specific activity, and immunoreactivity. Four groups of NSG mice (n = 3-5/group) were imaged with 64Cu-FN3hPD-L1 PET imaging (1-24 hours postinjection of 3.7 MBq/7 µg of Do-FN3 in 200 µL PBS): Nod SCID Gamma (NSG) mice bearing (i) syngeneic CT26/hPD-L1tumors, (ii) CT26/hPD-L1 tumors blocked (blk) by preinjected nonradioactive FN3hPD-L1 binder, (iii) hPD-L1-negative Raji xenografts, and (iv) MDA-MB-231 xenografts. The FN3hPD-L1 binder staining was evaluated against validated hPD-L1 antibodies by immunostaining in human cancer specimens. RESULTS: FN3hPD-L1 bound hPD-L1 with 1.4 ± 0.3 nmol/L affinity in CT26/hPD-L1 cells. 64Cu-FN3hPD-L1 radiotracer showed >70% yield and >95% purity. 64Cu-FN3hPD-L1 PET imaging of mice bearing CT26/hPD-L1 tumors showed tumor-to-muscle ratios of 5.6 ± 0.9 and 13.1 ± 2.3 at 1 and 4 hours postinjection, respectively. The FN3hPD-L1 binder detected hPD-L1 expression in human tissues with known hPD-L1 expression status based on two validated antibodies. CONCLUSIONS: The 64Cu-FN3hPD-L1 radiotracer represents a novel, small, and high-affinity binder for imaging hPD-L1 in tumors. Our data support further exploration and clinical translation of this binder for noninvasive identification of cancer patients who may respond to immune checkpoint blockade therapies.


Assuntos
Antígeno B7-H1/metabolismo , Imagem Molecular/métodos , Sondas Moleculares/administração & dosagem , Neoplasias/diagnóstico por imagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Adulto , Idoso , Animais , Antígeno B7-H1/imunologia , Linhagem Celular Tumoral/transplante , Radioisótopos de Cobre/administração & dosagem , Radioisótopos de Cobre/química , Modelos Animais de Doenças , Feminino , Domínio de Fibronectina Tipo III/genética , Humanos , Masculino , Camundongos , Sondas Moleculares/química , Sondas Moleculares/genética , Neoplasias/patologia , Ligação Proteica , Engenharia de Proteínas , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/química , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Distribuição Tecidual
12.
Dev Comp Immunol ; 93: 78-88, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30590066

RESUMO

Interleukin-35 (IL-35) is a member of the IL-12 cytokine family and a heterodimeric protein formed by Epstein-Barr virus-induced gene 3 (EBI3) and IL-12p35. Emerging evidence showed that IL-35 is a key player in the regulation of cellular communication, differentiation, and inflammation. To date, no studies on fish IL-35 have been documented. In this work, we first identify two splicing isoforms of EBI3, EBI3a and EBI3b, from grass carp (Ctenopharyngodon idella). EBI3a is composed of 299 amino acid residues and possesses an immunoglobulin-like (Ig-like) domain and a fibronectin type 3 (FN3) domain that is a conservative domain in vertebrate EBI3. However, the EBI3b is composed of 177 amino acid residues and only contains an Ig-like domain. The result of Co-immunoprecipitation suggests that only EBI3a can associate with IL-12p35 to form IL-35 in grass carp. Like the function of IL-35 in human and mouse, recombinant grass carp IL-35 protein could induce the expression of genes EBI3a, IL-12p35, and CD25-like and downregulate the expression of genes CD4-1, CD4-2, IL-17A/F1, and RORγ2. Taken together, these results indicate for the first time that a teleost IL-35 may also have the ability to induce regulatory T (Treg) cells, inhibit effector T (Teff) cell proliferation and restrict the differentiation and function of T helper 17 (Th17) cells in teleost.


Assuntos
Carpas/imunologia , Interleucinas/genética , Interleucinas/imunologia , Isoformas de Proteínas/genética , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Proliferação de Células , Domínio de Fibronectina Tipo III/genética , Regulação da Expressão Gênica/imunologia , Imunoglobulinas/genética , Subunidade p35 da Interleucina-12/metabolismo , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia
13.
PLoS One ; 13(5): e0197029, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29738555

RESUMO

Mesothelin is a cell surface protein that is overexpressed in numerous cancers, including breast, ovarian, lung, liver, and pancreatic tumors. Aberrant expression of mesothelin has been shown to promote tumor progression and metastasis through interaction with established tumor biomarker CA125. Therefore, molecules that specifically bind to mesothelin have potential therapeutic and diagnostic applications. However, no mesothelin-targeting molecules are currently approved for routine clinical use. While antibodies that target mesothelin are in development, some clinical applications may require a targeting molecule with an alternative protein fold. For example, non-antibody proteins are more suitable for molecular imaging and may facilitate diverse chemical conjugation strategies to create drug delivery complexes. In this work, we engineered variants of the fibronectin type III domain (Fn3) non-antibody protein scaffold to bind to mesothelin with high affinity, using directed evolution and yeast surface display. Lead engineered Fn3 variants were solubly produced and purified from bacterial culture at high yield. Upon specific binding to mesothelin on human cancer cell lines, the engineered Fn3 proteins internalized and co-localized to early endosomes. To our knowledge, this is the first report of non-antibody proteins engineered to bind mesothelin. The results validate that non-antibody proteins can be engineered to bind to tumor biomarker mesothelin, and encourage the continued development of engineered variants for applications such as targeted diagnostics and therapeutics.


Assuntos
Domínio de Fibronectina Tipo III/genética , Proteínas Ligadas por GPI/genética , Neoplasias/genética , Engenharia de Proteínas/métodos , Anticorpos Monoclonais/farmacologia , Biomarcadores Tumorais/genética , Antígeno Ca-125/genética , Linhagem Celular Tumoral , Proteínas Ligadas por GPI/uso terapêutico , Humanos , Mesotelina , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Proteínas Associadas à Matriz Nuclear/genética , Ligação Proteica
14.
Cell Death Differ ; 25(8): 1503-1516, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29396549

RESUMO

In response to stroke, astrocytes become reactive astrogliosis and are a major component of a glial scar. This results in the formation of both a physical and chemical (production of chondroitin sulfate proteoglycans) barrier, which prevent neurite regeneration that, in turn, interferes with functional recovery. However, the mechanisms of reactive astrogliosis and glial scar formation are poorly understood. In this work, we hypothesized that repulsive guidance molecule a (RGMa) regulate reactive astrogliosis and glial scar formation. We first found that RGMa was strongly expressed by reactive astrocytes in the glial scar in a rat model of middle cerebral artery occlusion/reperfusion. Genetic or pharmacologic inhibition of RGMa in vivo resulted in a strong reduction of reactive astrogliosis and glial scarring as well as in a pronounced improvement in functional recovery. Furthermore, we showed that transforming growth factor ß1 (TGFß1) stimulated RGMa expression through TGFß1 receptor activin-like kinase 5 (ALK5) in primary cultured astrocytes. Knockdown of RGMa abrogated key steps of reactive astrogliosis and glial scar formation induced by TGFß1, including cellular hypertrophy, glial fibrillary acidic protein upregulation, cell migration, and CSPGs secretion. Finally, we demonstrated that RGMa co-immunoprecipitated with ALK5 and Smad2/3. TGFß1-induced ALK5-Smad2/3 interaction and subsequent phosphorylation of Smad2/3 were impaired by RGMa knockdown. Taken together, we identified that after stroke, RGMa promotes reactive astrogliosis and glial scar formation by forming a complex with ALK5 and Smad2/3 to promote ALK5-Smad2/3 interaction to facilitate TGFß1/Smad2/3 signaling, thereby inhibiting neurological functional recovery. RGMa may be a new therapeutic target for stroke.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/patologia , Transdução de Sinais/efeitos dos fármacos , Acidente Vascular Cerebral/patologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Células Cultivadas , Cicatriz/patologia , Modelos Animais de Doenças , Domínio de Fibronectina Tipo III/genética , Proteínas Ligadas por GPI , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Acidente Vascular Cerebral/metabolismo
15.
Mol Microbiol ; 105(6): 839-859, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28657670

RESUMO

Colonization of mucosal respiratory surfaces is a prerequisite for the human pathobiont Streptococcus pneumoniae (the pneumococcus) to cause severe invasive infections. The arsenal of pneumococcal adhesins interacts with a multitude of extracellular matrix proteins. A paradigm for pneumococci is their interaction with the adhesive glycoprotein fibronectin, which facilitates bacterial adherence to host cells. Here, we deciphered the molecular interaction between fibronectin and pneumococcal fibronectin-binding proteins (FnBPs) PavA and PavB respectively. We show in adherence and binding studies that the pneumococcal interaction with fibronectin is a non-human specific trait. PavA and PavB target at least 13 out of 15 type III fibronectin domains as demonstrated in ligand overlay assays, surface plasmon resonance studies and SPOT peptide arrays. Strikingly, both pneumococcal FnBPs recognize similar peptides in targeted type III repeats. Structural comparisons revealed that the targeted type III repeat epitopes cluster on the inner strands of both ß-sheets forming the fibronectin domains. Importantly, synthetic peptides of FnIII1 , FnIII5 or FnIII15 bind directly to FnBPs PavA and PavB respectively. In conclusion, our study suggests a common pattern of molecular interactions between pneumococcal FnBPs and fibronectin. The specific epitopes recognized in this study can potentially be tested as antimicrobial targets in further scientific endeavours.


Assuntos
Proteínas de Bactérias/metabolismo , Domínio de Fibronectina Tipo III/fisiologia , Fibronectinas/metabolismo , Adesinas Bacterianas/metabolismo , Aderência Bacteriana/genética , Aderência Bacteriana/fisiologia , Proteínas de Bactérias/genética , Proteínas de Transporte/metabolismo , Domínio de Fibronectina Tipo III/genética , Interações Hospedeiro-Patógeno , Humanos , Ligação Proteica/genética , Ligação Proteica/fisiologia , Domínios Proteicos , Streptococcus pneumoniae/metabolismo , Fatores de Virulência/metabolismo
16.
Biochemistry ; 56(10): 1504-1517, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28233978

RESUMO

Polysialic acid (polySia) is a unique post-translational modification found on a small set of mammalian glycoproteins. Composed of long chains of α2,8-linked sialic acid, this large, negatively charged polymer attenuates protein and cell adhesion and modulates signaling mediated by its carriers and proteins that interact with these carriers. PolySia is crucial for the proper development of the nervous system and is upregulated during tissue regeneration and in highly invasive cancers. Our laboratory has previously shown that the neural cell adhesion molecule, NCAM, has an acidic surface patch in its first fibronectin type III repeat (FN1) that is critical for the polysialylation of N-glycans on the adjacent immunoglobulin domain (Ig5). We have also identified a polysialyltransferase (polyST) polybasic region (PBR) that may mediate substrate recognition. However, a direct interaction between the NCAM FN1 acidic patch and the polyST PBR has yet to be demonstrated. Here, we have probed this interaction using isothermal titration calorimetry and nuclear magnetic resonance (NMR) spectroscopy. We observe direct and specific binding between FN1 and the PBR peptide that is dependent upon acidic residues in FN1 and basic residues of the PBR. NMR titration experiments verified the role of the FN1 acidic patch in the recognition of the PBR and suggest a conformational change of the Ig5-FN1 linker region following binding of the PBR to the acidic patch. Finally, mutation of residues identified by NMR titration experiments impacts NCAM polysialylation, supporting their mechanistic role in protein-specific polysialylation.


Assuntos
Domínio de Fibronectina Tipo III/genética , Moléculas de Adesão de Célula Nervosa/química , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/química , Ácidos Siálicos/química , Sialiltransferases/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Chlorocebus aethiops , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Histidina/genética , Histidina/metabolismo , Humanos , Modelos Moleculares , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Alinhamento de Sequência , Ácidos Siálicos/metabolismo , Sialiltransferases/genética , Sialiltransferases/metabolismo
17.
Cancer Res ; 77(7): 1730-1740, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28108514

RESUMO

The contribution of somatic mutations to metastasis of colorectal cancers is currently unknown. To find mutations involved in the colorectal cancer metastatic process, we performed deep mutational analysis of 676 genes in 107 stages II to IV primary colorectal cancer, of which half had metastasized. The mutation prevalence in the ephrin (EPH) family of tyrosine kinase receptors was 10-fold higher in primary tumors of metastatic colorectal than in nonmetastatic cases and preferentially occurred in stage III and IV tumors. Mutational analyses in situ confirmed expression of mutant EPH receptors. To enable functional studies of EPHB1 mutations, we demonstrated that DLD-1 colorectal cancer cells expressing EPHB1 form aggregates upon coculture with ephrin B1 expressing cells. When mutations in the fibronectin type III and kinase domains of EPHB1 were compared with wild-type EPHB1 in DLD-1 colorectal cancer cells, they decreased ephrin B1-induced compartmentalization. These observations provide a mechanistic link between EPHB receptor mutations and metastasis in colorectal cancer. Cancer Res; 77(7); 1730-40. ©2017 AACR.


Assuntos
Neoplasias Colorretais/patologia , Mutação , Metástase Neoplásica , Receptor EphB1/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Domínio de Fibronectina Tipo III/genética , Humanos , Estadiamento de Neoplasias , Proteínas Tirosina Quinases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...