Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Anal Chem ; 96(12): 4764-4773, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38484023

RESUMO

Ancient peptides are remnants of early biochemistry that continue to play pivotal roles in current proteins. They are simple molecules yet complex enough to exhibit independent functions, being products of an evolved biochemistry at the interface of life and nonlife. Their adsorption to minerals may contribute to their stabilization and preservation over time. To investigate the feasibility of conserved peptide sequences and structures as target biomarkers for the search for life on Mars or other planetary bodies, we conducted a bioinformatics selection of well-conserved ancient peptides and produced polyclonal antibodies for their detection using fluorescence microarray immunoassays. Additionally, we explored how adsorbing peptides to Mars-representative minerals to form organomineral complexes could affect their immunological detection. The results demonstrated that the selected peptides exhibited autonomous folding, with some of them regaining their structure, even after denaturation. Furthermore, their cognate antibodies detected their conformational features regardless of amino acid sequences, thereby broadening the spectrum of target peptide sequences. While certain antibodies displayed unspecific binding to bare minerals, we validated that peptide-mineral complexes can be detected using sandwich immunoassays, as confirmed through desorption and competitive assays. Consequently, we conclude that the diversity of peptide sequences and structures suitable for use as target biomarkers in astrobiology can be constrained to a few well conserved sets, and they can be detected even if they are adsorbed in organomineral complexes.


Assuntos
Exobiologia , Marte , Exobiologia/métodos , Minerais , Sequência de Aminoácidos , Peptídeos , Anticorpos , Biomarcadores
2.
Nucleic Acids Res ; 50(22): 12938-12950, 2022 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-36511856

RESUMO

Genetic interventions on microbiomes, for clinical or biotechnological purposes, remain challenging. Conjugation-based delivery of genetic cargo is still unspecific and limited by low conjugation rates. Here we report an approach to overcome these problems, based on a synthetic bacterial adhesion system. Mating assemblers consist on a synthetic adhesion formed by the expression on the surface of donor and target cells of specific nanobodies (Nb) and their cognate antigen (Ag). The Nb-Ag bridge increased 1-3 logs transfer of a variety of plasmids, especially in liquid media, confirming that cell-cell docking is a main determinant limiting mating efficiency. Synthetic cell-to-cell adhesion allows efficient conjugation to targeted recipients, enhancing delivery of desired genes to a predefined subset of prey species, or even specific pathogenic strains such as enterohemorrhagic Escherichia coli (EHEC), within a bacterial community. The synthetic conjugation enhancer presented here optimizes plasmid delivery by selecting the target hosts with high selectivity.


Assuntos
Conjugação Genética , Técnicas Genéticas , Microbiota , Adesão Celular , Conjugação Genética/fisiologia , Escherichia coli/genética , Escherichia coli/metabolismo , Plasmídeos/genética , Biotecnologia/métodos , Microbiota/genética
3.
Trends Microbiol ; 30(6): 524-533, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34840074

RESUMO

Type III secretion system (T3SS) effectors are key virulence factors that underpin the infection strategy of many clinically important Gram-negative pathogens, including Salmonella enterica, Shigella spp., enteropathogenic and enterohemorrhagic Escherichia coli and their murine equivalent, Citrobacter rodentium. The cellular processes or proteins targeted by the effectors can be common to multiple pathogens or pathogen-specific. The main approach to understanding T3SS-mediated pathogenesis has been to determine the contribution of one effector at a time, with the aim of piecing together individual functions and unveiling infection mechanisms. However, in contrast to this prevailing approach, simultaneous deletion of multiple effectors revealed that they function as an interconnected network in vivo, uncovering effector codependency and context-dependent effector essentiality. This paradigm shift in T3SS biology is at the heart of this opinion article.


Assuntos
Escherichia coli Êntero-Hemorrágica , Proteínas de Escherichia coli , Salmonella enterica , Citrobacter rodentium/genética , Escherichia coli Êntero-Hemorrágica/genética , Proteínas de Escherichia coli/metabolismo , Salmonella enterica/metabolismo , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
4.
Microb Biotechnol ; 14(3): 1228-1236, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33929101

RESUMO

The surge of SARS-CoV-2 has challenged health systems worldwide and efficient tests to detect viral particles, as well as antibodies generated against them, are needed. Specificity, sensitivity, promptness or scalability are the main parameters to estimate the final performance, but rarely all of them match in a single test. We have developed SCOVAM, a protein microarray with several viral antigens (spike, nucleocapsid, main protease Nsp5) as capturing probes in a fluorescence immunoassay for COVID-19 serological testing. SCOVAM depicts IgG and IgM antibody responses against each of these proteins of 22 individuals in a single microscope slide. It detects specific IgM (0.094 µg ml-1 ) and IgG (~0.017 µg ml-1 ) and is scalable and cost-effective. We validated SCOVAM by comparing with a widely used chemiluminescent commercial serological test (n = 742). SCOVAM showed twice the sensitivity and allowed following seroconversion in a single assay. By analysing the prevalence 4 months later in a subset of 76 positive sera, we still detected 93.42% of positives, almost doubling the detection of the commercial assay. The higher sensitivity of SCOVAM is especially relevant to screen sera for convalescent plasma-based treatments, high-throughput antibody response monitoring after vaccination or evaluation of vaccine efficiency.


Assuntos
COVID-19 , SARS-CoV-2 , Anticorpos Antivirais , COVID-19/terapia , Teste Sorológico para COVID-19 , Humanos , Imunização Passiva , Imunoglobulina G , Imunoglobulina M , Sensibilidade e Especificidade , Estudos Soroepidemiológicos , Glicoproteína da Espícula de Coronavírus , Soroterapia para COVID-19
5.
Science ; 371(6534)2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33707240

RESUMO

Infections with many Gram-negative pathogens, including Escherichia coli, Salmonella, Shigella, and Yersinia, rely on type III secretion system (T3SS) effectors. We hypothesized that while hijacking processes within mammalian cells, the effectors operate as a robust network that can tolerate substantial contractions. This was tested in vivo using the mouse pathogen Citrobacter rodentium (encoding 31 effectors). Sequential gene deletions showed that effector essentiality for infection was context dependent and that the network could tolerate 60% contraction while maintaining pathogenicity. Despite inducing very different colonic cytokine profiles (e.g., interleukin-22, interleukin-17, interferon-γ, or granulocyte-macrophage colony-stimulating factor), different networks induced protective immunity. Using data from >100 distinct mutant combinations, we built and trained a machine learning model able to predict colonization outcomes, which were confirmed experimentally. Furthermore, reproducing the human-restricted enteropathogenic E. coli effector repertoire in C. rodentium was not sufficient for efficient colonization, which implicates effector networks in host adaptation. These results unveil the extreme robustness of both T3SS effector networks and host responses.


Assuntos
Proteínas de Bactérias/metabolismo , Citrobacter rodentium/patogenicidade , Infecções por Enterobacteriaceae/microbiologia , Redes e Vias Metabólicas , Sistemas de Secreção Tipo III/metabolismo , Animais , Proteínas de Bactérias/genética , Citrobacter rodentium/genética , Infecções por Enterobacteriaceae/imunologia , Feminino , Deleção de Genes , Imunidade , Camundongos , Camundongos Endogâmicos C57BL , Proteólise , Sistemas de Secreção Tipo III/genética , Virulência
6.
Methods Mol Biol ; 2291: 253-272, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33704757

RESUMO

Therapeutic antibodies (Abs) inhibiting bacterial adhesion to host epithelia are an attractive option to reduce the load of Shiga toxin-producing E. coli (STEC) in the intestine of the patient and also in the bovine reservoir, thereby minimizing the risk of STEC contamination in the food chain. Of particular interest are recombinant single-domain Ab fragments called nanobodies (Nbs) derived from the variable domain of camelid heavy chain-only antibodies (VHH). The outer membrane adhesin intimin and the translocated intimin receptor (Tir) are essential for the attachment of STEC to host epithelia. In addition, EspA filaments of the bacterial type III protein secretion system are needed for Tir translocation into the host cell. Given their importance for bacterial adhesion and colonization, we developed Nbs against intimin, Tir and EspA proteins of STEC serotype O157:H7. Here, we report the screening methods used to isolate inhibitory Nbs blocking intimin-Tir protein-protein interaction, actin-pedestal formation, and intimate adhesion of STEC to epithelial cells in vitro. First, we describe how VHH gene repertoires can be produced as Nbs secreted by E. coli using the α-hemolysin (HlyA) protein secretion system. Next, we report the methods for identification of inhibitors of intimin-Tir protein-protein interaction and of STEC intimate adhesion to HeLa cells in culture. These methods can be adapted for the screening of Nbs against different adhesin-receptor complexes to block the adhesion of other pathogens to host cells.


Assuntos
Adesinas Bacterianas/imunologia , Anticorpos Antibacterianos/imunologia , Aderência Bacteriana/imunologia , Células Epiteliais , Escherichia coli O157/imunologia , Proteínas de Escherichia coli/imunologia , Receptores de Superfície Celular/imunologia , Anticorpos de Domínio Único/imunologia , Animais , Bovinos , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Escherichia coli O157/patogenicidade , Humanos
7.
Mucosal Immunol ; 13(2): 322-333, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31772324

RESUMO

Given the global burden of diarrheal diseases on healthcare it is surprising how little is known about the drivers of disease severity. Colitis caused by infection and inflammatory bowel disease (IBD) is characterised by neutrophil infiltration into the intestinal mucosa and yet our understanding of neutrophil responses during colitis is incomplete. Using infectious (Citrobacter rodentium) and chemical (dextran sulphate sodium; DSS) murine colitis models, as well as human IBD samples, we find that faecal neutrophil elastase (NE) activity reflects disease severity. During C. rodentium infection intestinal epithelial cells secrete the serine protease inhibitor SerpinA3N to inhibit and mitigate tissue damage caused by extracellular NE. Mice suffering from severe infection produce insufficient SerpinA3N to control excessive NE activity. This activity contributes to colitis severity as infection of these mice with a recombinant C. rodentium strain producing and secreting SerpinA3N reduces tissue damage. Thus, uncontrolled luminal NE activity is involved in severe colitis. Taken together, our findings suggest that NE activity could be a useful faecal biomarker for assessing disease severity as well as therapeutic target for both infectious and chronic inflammatory colitis.


Assuntos
Biomarcadores/metabolismo , Citrobacter rodentium/fisiologia , Colite/metabolismo , Infecções por Enterobacteriaceae/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Elastase de Leucócito/metabolismo , Neutrófilos/imunologia , Proteínas de Fase Aguda/metabolismo , Animais , Sulfato de Dextrana , Modelos Animais de Doenças , Progressão da Doença , Fezes/química , Humanos , Camundongos , Inibidores de Proteases/metabolismo , Serpinas/metabolismo , Índice de Gravidade de Doença
8.
PLoS Pathog ; 15(8): e1008031, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31465434

RESUMO

Enterohemorrhagic E. coli (EHEC) is a human intestinal pathogen that causes hemorrhagic colitis and hemolytic uremic syndrome. No vaccines or specific therapies are currently available to prevent or treat these infections. EHEC tightly attaches to the intestinal epithelium by injecting the intimin receptor Tir into the host cell via a type III secretion system (T3SS). In this project, we identified a camelid single domain antibody (nanobody), named TD4, that recognizes a conserved Tir epitope overlapping the binding site of its natural ligand intimin with high affinity and stability. We show that TD4 inhibits attachment of EHEC to cultured human HeLa cells by preventing Tir clustering by intimin, activation of downstream actin polymerization and pedestal formation. Furthermore, we demonstrate that TD4 significantly reduces EHEC adherence to human colonic mucosa in in vitro organ cultures. Altogether, these results suggest that nanobody-based therapies hold potential in the development of much needed treatment and prevention strategies against EHEC infection.


Assuntos
Aderência Bacteriana/fisiologia , Colo/metabolismo , Escherichia coli Êntero-Hemorrágica/fisiologia , Infecções por Escherichia coli/metabolismo , Proteínas de Escherichia coli/antagonistas & inibidores , Receptores de Superfície Celular/antagonistas & inibidores , Anticorpos de Domínio Único/farmacologia , Sequência de Aminoácidos , Animais , Aderência Bacteriana/efeitos dos fármacos , Sítios de Ligação , Camelus , Colo/microbiologia , Colo/patologia , Escherichia coli Êntero-Hemorrágica/efeitos dos fármacos , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Proteínas de Escherichia coli/imunologia , Proteínas de Escherichia coli/metabolismo , Células HeLa , Humanos , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Homologia de Sequência , Anticorpos de Domínio Único/imunologia
9.
Cell Rep ; 27(4): 1008-1017.e6, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31018119

RESUMO

Microbial infections can stimulate the assembly of inflammasomes, which activate caspase-1. The gastrointestinal pathogen enteropathogenic Escherichia coli (EPEC) causes localized actin polymerization in host cells. Actin polymerization requires the binding of the bacterial adhesin intimin to Tir, which is delivered to host cells via a type 3 secretion system (T3SS). We show that EPEC induces T3SS-dependent rapid non-canonical NLRP3 inflammasome activation in human macrophages. Notably, caspase-4 activation by EPEC triggers pyroptosis and cytokine processing through the NLRP3-caspase-1 inflammasome. Mechanistically, caspase-4 activation requires the detection of LPS and EPEC-induced actin polymerization, either via Tir tyrosine phosphorylation and the phosphotyrosine-binding adaptor NCK or Tir and the NCK-mimicking effector TccP. An engineered E. coli K12 could reconstitute Tir-intimin signaling, which is necessary and sufficient for inflammasome activation, ruling out the involvement of other virulence factors. Our studies reveal a crosstalk between caspase-4 and caspase-1 that is cooperatively stimulated by LPS and effector-driven actin polymerization.


Assuntos
Caspases Iniciadoras/fisiologia , Escherichia coli Enteropatogênica/patogenicidade , Macrófagos/microbiologia , Actinas/metabolismo , Caspase 1/genética , Caspase 1/metabolismo , Caspase 1/fisiologia , Caspases Iniciadoras/genética , Caspases Iniciadoras/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Inflamassomos/fisiologia , Modelos Biológicos , Polimerização
10.
Microb Cell Fact ; 18(1): 47, 2019 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-30857538

RESUMO

BACKGROUND: The hemolysin (Hly) secretion system of E. coli allows the one-step translocation of hemolysin A (HlyA) from the bacterial cytoplasm to the extracellular medium, without a periplasmic intermediate. In this work, we investigate whether the Hly secretion system of E. coli is competent to secrete a repertoire of functional single-domain VHH antibodies (nanobodies, Nbs), facilitating direct screening of VHH libraries and the purification of selected Nb from the extracellular medium. RESULTS: We employed a phagemid library of VHHs obtained by immunization of a dromedary with three protein antigens from enterohemorrhagic E. coli (EHEC), namely, the extracellular secreted protein A (EspA), the extracellular C-terminal region of Intimin (Int280), and the translocated intimin receptor middle domain (TirM). VHH clones binding each antigen were enriched and amplified by biopanning, and subsequently fused to the C-terminal secretion signal of HlyA to be expressed and secreted in a E. coli strain carrying the Hly export machinery (HlyB, HlyD and TolC). Individual E. coli clones were grown and induced in 96-well microtiter plates, and the supernatants of the producing cultures directly used in ELISA for detection of Nbs binding EspA, Int280 and TirM. A set of Nb sequences specifically binding each of these antigens were identified, indicating that the Hly system is able to secrete a diversity of functional Nbs. We performed thiol alkylation assays demonstrating that Nbs are correctly oxidized upon secretion, forming disulphide bonds between cysteine pairs despite the absence of a periplasmic intermediate. In addition, we show that the secreted Nb-HlyA fusions can be directly purified from the supernatant of E. coli cultures, avoiding cell lysis and in a single affinity chromatography step. CONCLUSIONS: Our data demonstrate the Hly secretion system of E. coli can be used as an expression platform for screening and purification of Nb binders from VHH repertories.


Assuntos
Meios de Cultura/análise , Escherichia coli/fisiologia , Proteínas Hemolisinas/metabolismo , Anticorpos de Domínio Único/isolamento & purificação , Transporte Biológico , Escherichia coli/imunologia
11.
Nat Commun ; 9(1): 5148, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30498235

RESUMO

The original version of this Article contained an error in the spelling of the author David Ruano-Gallego, which was incorrectly given as David R. Gallego. This has now been corrected in both the PDF and HTML versions of the Article.

12.
Nat Commun ; 9(1): 4187, 2018 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-30305622

RESUMO

Niche-adaptation of a bacterial pathogen hinges on the ability to recognize the complexity of signals from the environment and integrate that information with the regulation of genes critical for infection. Here we report the transcriptome of the attaching and effacing pathogen Citrobacter rodentium during infection of its natural murine host. Pathogen gene expression in vivo was heavily biased towards the virulence factor repertoire and was found to be co-ordinated uniquely in response to the host. Concordantly, we identified the host-specific induction of a metabolic pathway that overlapped with the regulation of virulence. The essential type 3 secretion system and an associated suite of distinct effectors were found to be modulated co-ordinately through a unique mechanism involving metabolism of microbiota-derived 1,2-propanediol, which dictated the ability to colonize the host effectively. This study provides novel insights into how host-specific metabolic adaptation acts as a cue to fine-tune virulence.


Assuntos
Sistemas de Secreção Bacterianos , Citrobacter rodentium/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Interações Hospedeiro-Patógeno , Animais , Aderência Bacteriana , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidade , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/patologia , Regulação Bacteriana da Expressão Gênica , Células HeLa , Interações Hospedeiro-Patógeno/genética , Humanos , Metabolômica , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Propilenoglicol/metabolismo , Análise de Sequência de RNA , Transcriptoma/genética , Virulência/genética , Fatores de Virulência/metabolismo
13.
Curr Top Microbiol Immunol ; 416: 51-72, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30088147

RESUMO

Infection with enteropathogenic and enterohaemorrhagic Escherichia coli (EPEC and EHEC), enteroinvasive E. coli (EIEC) and Shigella relies on the elaboration of a type III secretion system (T3SS). Few strains also encode a second T3SS, named ETT2. Through the integration of coordinated intracellular and extracellular cues, the modular T3SS is assembled within the bacterial cell wall, as well as the plasma membrane of the host cell. As such, the T3SS serves as a conduit, allowing the chaperone-regulated translocation of effector proteins directly into the host cytosol to subvert eukaryotic cell processes. Recent technological advances revealed high structural resolution of the T3SS apparatus and how it could be exploited to treat enteric disease. This chapter summarises the current knowledge of the structure and function of the E. coli T3SSs.


Assuntos
Escherichia coli Êntero-Hemorrágica/metabolismo , Escherichia coli Êntero-Hemorrágica/patogenicidade , Escherichia coli Enteropatogênica/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Sistemas de Secreção Tipo III/metabolismo , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Humanos
14.
Cell Rep ; 21(12): 3381-3389, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29262319

RESUMO

We investigated the role of commensals at the peak of infection with the colonic mouse pathogen Citrobacter rodentium. Bioluminescent and kanamycin (Kan)-resistant C. rodentium persisted avirulently in the cecal lumen of mice continuously treated with Kan. A single Kan treatment was sufficient to displace C. rodentium from the colonic mucosa, a phenomenon not observed following treatment with vancomycin (Van) or metronidazole (Met). Kan, Van, and Met induce distinct dysbiosis, suggesting C. rodentium relies on specific commensals for colonic colonization. Expression of the master virulence regulator ler is induced in germ-free mice, yet C. rodentium is only seen in the cecal lumen. Moreover, in conventional mice, a single Kan treatment was sufficient to displace C. rodentium constitutively expressing Ler from the colonic mucosa. These results show that expression of virulence genes is not sufficient for colonization of the colonic mucosa and that commensals are essential for a physiological infection course.


Assuntos
Antibacterianos/farmacologia , Citrobacter rodentium/patogenicidade , Colo/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Animais , Proteínas de Bactérias/genética , Citrobacter rodentium/efeitos dos fármacos , Canamicina/farmacologia , Metronidazol/farmacologia , Camundongos , Vancomicina/farmacologia , Virulência/genética
15.
Curr Opin Biotechnol ; 35: 94-102, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26070111

RESUMO

Although bacteria are generally regarded as the causative agents of infectious diseases, most bacteria inhabiting the human body are non-pathogenic and some of them can be turned, after proper engineering, into 'smart' living therapeutics of defined properties for the treatment of different illnesses. This review focuses on recent developments to engineer bacteria for the treatment of diverse human pathologies, including inflammatory bowel diseases, autoimmune disorders, cancer, metabolic diseases and obesity, as well as to combat bacterial and viral infections. We discuss significant advances provided by synthetic biology to fully reprogram bacteria as human therapeutics, including novel measures for strict biocontainment.


Assuntos
Bactérias/metabolismo , Engenharia Genética/métodos , Animais , Bactérias/genética , Desenho de Fármacos , Genoma Bacteriano , Humanos , Inflamação/tratamento farmacológico , Células-Tronco
16.
ACS Synth Biol ; 4(9): 1030-41, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26017572

RESUMO

Bacterial pathogens containing type III protein secretion systems (T3SS) assemble large needle-like protein complexes in the bacterial envelope, called injectisomes, for translocation of protein effectors into host cells. The application of these "molecular syringes" for the injection of proteins into mammalian cells is hindered by their structural and genomic complexity, requiring multiple polypeptides encoded along with effectors in various transcriptional units (TUs) with intricate regulation. In this work, we have rationally designed the controlled expression of the filamentous injectisomes found in enteropathogenic Escherichia coli (EPEC) in the nonpathogenic strain E. coli K-12. All structural components of EPEC injectisomes, encoded in a genomic island called the locus of enterocyte effacement (LEE), were engineered in five TUs (eLEEs) excluding effectors, promoters and transcriptional regulators. These eLEEs were placed under the control of the IPTG-inducible promoter Ptac and integrated into specific chromosomal sites of E. coli K-12 using a marker-less strategy. The resulting strain, named synthetic injector E. coli (SIEC), assembles filamentous injectisomes similar to those in EPEC. SIEC injectisomes form pores in the host plasma membrane and are able to translocate T3-substrate proteins (e.g., translocated intimin receptor, Tir) into the cytoplasm of HeLa cells reproducing the phenotypes of intimate attachment and polymerization of actin-pedestals elicited by EPEC bacteria. Hence, SIEC strain allows the controlled expression of functional filamentous injectisomes for efficient translocation of proteins with T3S-signals into mammalian cells.


Assuntos
Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/metabolismo , Engenharia Genética , Receptores de Superfície Celular/metabolismo , Primers do DNA , DNA Bacteriano/genética , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/genética , Genes Bacterianos , Células HeLa , Humanos , Regiões Promotoras Genéticas , Transporte Proteico , Receptores de Superfície Celular/genética
17.
Breast Cancer Res ; 15(6): R116, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24330716

RESUMO

INTRODUCTION: Aberrant turnover of the actin cytoskeleton is intimately associated with cancer cell migration and invasion. Frequently however, evidence is circumstantial, and a reliable assessment of the therapeutic significance of a gene product is offset by lack of inhibitors that target biologic properties of a protein, as most conventional drugs do, instead of the corresponding gene. Proteomic studies have demonstrated overexpression of CapG, a constituent of the actin cytoskeleton, in breast cancer. Indirect evidence suggests that CapG is involved in tumor cell dissemination and metastasis. In this study, we used llama-derived CapG single-domain antibodies or nanobodies in a breast cancer metastasis model to address whether inhibition of CapG activity holds therapeutic merit. METHODS: We raised single-domain antibodies (nanobodies) against human CapG and used these as intrabodies (immunomodulation) after lentiviral transduction of breast cancer cells. Functional characterization of nanobodies was performed to identify which biochemical properties of CapG are perturbed. Orthotopic and tail vein in vivo models of metastasis in nude mice were used to assess cancer cell spreading. RESULTS: With G-actin and F-actin binding assays, we identified a CapG nanobody that binds with nanomolar affinity to the first CapG domain. Consequently, CapG interaction with actin monomers or actin filaments is blocked. Intracellular delocalization experiments demonstrated that the nanobody interacts with CapG in the cytoplasmic environment. Expression of the nanobody in breast cancer cells restrained cell migration and Matrigel invasion. Notably, the nanobody prevented formation of lung metastatic lesions in orthotopic xenograft and tail-vein models of metastasis in immunodeficient mice. We showed that CapG nanobodies can be delivered into cancer cells by using bacteria harboring a type III protein secretion system (T3SS). CONCLUSIONS: CapG inhibition strongly reduces breast cancer metastasis. A nanobody-based approach offers a fast track for gauging the therapeutic merit of drug targets. Mapping of the nanobody-CapG interface may provide a platform for rational design of pharmacologic compounds.


Assuntos
Actinas/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proteínas dos Microfilamentos/imunologia , Terapia de Alvo Molecular/métodos , Proteínas Nucleares/imunologia , Anticorpos de Domínio Único/farmacologia , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Escherichia coli/genética , Feminino , Humanos , Camundongos SCID , Proteínas dos Microfilamentos/genética , Proteínas Nucleares/genética , Estrutura Terciária de Proteína
18.
PLoS One ; 8(9): e75126, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086454

RESUMO

Screening of antibody (Ab) libraries by direct display on the surface of E. coli cells is hampered by the presence of the outer membrane (OM). In this work we demonstrate that the native ß-domains of EhaA autotransporter and intimin, two proteins from enterohemorrhagic E. coli O157:H7 (EHEC) with opposite topologies in the OM, are effective systems for the display of immune libraries of single domain Abs (sdAbs) from camelids (nanobodies or VHH) on the surface of E. coli K-12 cells and for the selection of high affinity sdAbs using magnetic cell sorting (MACS). We analyzed the capacity of EhaA and intimin ß-domains to display individual sdAbs and sdAb libraries obtained after immunization with the extracellular domain of the translocated intimin receptor from EHEC (TirM(EHEC)). We demonstrated that both systems displayed functional sdAbs on the surface of E. coli cells with little proteolysis and cellular toxicity, although E. coli cells displaying sdAbs with the ß-domain of intimin showed higher antigen-binding capacity. Both E. coli display libraries were screened for TirM(EHEC) binding clones by MACS. High affinity binders were selected by both display systems, although more efficiently with the intimin ß-domain. The specificity of the selected clones against TirM(EHEC) was demonstrated by flow cytometry of E. coli cells, along with ELISA and surface plasmon resonance with purified sdAbs. Finally, we employed the E. coli cell display systems to provide an estimation of the affinity of the selected sdAb by flow cytometry analysis under equilibrium conditions.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Camelus/imunologia , Escherichia coli O157/metabolismo , Biblioteca de Peptídeos , Anticorpos de Domínio Único/metabolismo , Adesinas Bacterianas/metabolismo , Adesinas de Escherichia coli/metabolismo , Animais , Especificidade de Anticorpos , Western Blotting , Ensaio de Imunoadsorção Enzimática , Proteínas de Escherichia coli/metabolismo , Citometria de Fluxo , Oligonucleotídeos/genética , Plasmídeos/genética , Anticorpos de Domínio Único/química , Ressonância de Plasmônio de Superfície
19.
Cytoskeleton (Hoboken) ; 70(10): 604-22, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23818458

RESUMO

Nanobodies or VHHs are single domain antigen binding fragments derived from heavy-chain antibodies naturally occurring in species of the Camelidae. Due to their ease of cloning, high solubility and intrinsic stability, they can be produced at low cost. Their small size, combined with high affinity and antigen specificity, enables recognition of a broad range of structural (undruggable) proteins and enzymes alike. Focusing on two actin binding proteins, gelsolin and CapG, we summarize a general protocol for the generation, cloning and production of nanobodies. Furthermore, we describe multiple ways to characterize antigen-nanobody binding in more detail and we shed light on some applications with recombinant nanobodies. The use of nanobodies as intrabodies is clarified through several case studies revealing new cytoskeletal protein properties and testifying to the utility of nanobodies as intracellular bona fide protein inhibitors. Moreover, as nanobodies can traverse the plasma membrane of eukaryotic cells by means of the enteropathogenic E. coli type III protein secretion system, we show that in this promising way of nanobody delivery, actin pedestal formation can be affected following nanobody injection.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Mapeamento de Epitopos , Anticorpos de Domínio Único/metabolismo , Actinas/metabolismo , Calorimetria , Linhagem Celular , Membrana Celular/metabolismo , Movimento Celular , Núcleo Celular/metabolismo , Centrossomo/metabolismo , Escherichia coli Enteropatogênica/metabolismo , Gelsolina/metabolismo , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Fagocitose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA