Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Microb Biotechnol ; 17(1): e14328, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37608576

RESUMO

Biosafety of engineered bacteria as living therapeutics requires a tight regulation to control the specific delivery of protein effectors, maintaining minimum leakiness in the uninduced (OFF) state and efficient expression in the induced (ON) state. Here, we report a three repressors (3R) genetic circuit that tightly regulates the expression of multiple tac promoters (Ptac) integrated in the chromosome of E. coli and drives the expression of a complex type III secretion system injectisome for therapeutic protein delivery. The 3R genetic switch is based on the tetracycline repressor (TetR), the non-inducible lambda repressor cI (ind-) and a mutant lac repressor (LacIW220F ) with higher activity. The 3R switch was optimized with different protein translation and degradation signals that control the levels of LacIW220F . We demonstrate the ability of an optimized switch to fully repress the strong leakiness of the Ptac promoters in the OFF state while triggering their efficient activation in the ON state with anhydrotetracycline (aTc), an inducer suitable for in vivo use. The implementation of the optimized 3R switch in the engineered synthetic injector E. coli (SIEC) strain boosts expression of injectisomes upon aTc induction, while maintaining a silent OFF state that preserves normal growth in the absence of the inducer. Since Ptac is a commonly used promoter, the 3R switch may have multiple applications for tight control of protein expression in E. coli. In addition, the modularity of the 3R switch may enable its tuning for the control of Ptac promoters with different inducers.


Assuntos
Compostos Bicíclicos com Pontes , Escherichia coli , Tiadiazóis , Escherichia coli/genética , Escherichia coli/metabolismo , Regiões Promotoras Genéticas , Compostos Bicíclicos com Pontes/metabolismo , Repressores Lac/genética , Repressores Lac/metabolismo
2.
Microb Biotechnol ; 17(1): e14367, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37971317

RESUMO

Large gene libraries are frequently created in Escherichia coli plasmids, which can induce cell toxicity and expression instability due to the high gene dosage. To address these limitations, gene libraries can be integrated in a single copy into the bacterial chromosome. Here, we describe an efficient system for the massive integration (MAIN) of large gene libraries in the E. coli chromosome that generates in-frame gene fusions that are expressed stably. MAIN uses a thermosensitive integrative plasmid that is linearized in vivo to promote extensive integration of the gene library via homologous recombination. Positive and negative selections efficiently remove bacteria lacking gene integration in the target site. We tested MAIN with a library of 107 VHH genes that encode nanobodies (Nbs). The integration of VHH genes into a custom target locus of the E. coli chromosome enabled stable expression and surface display of the Nbs. Next-generation DNA sequencing confirmed that MAIN preserved the diversity of the gene library after integration. Finally, we screened the integrated library to select Nbs that bind a specific antigen using magnetic and fluorescence-activated cell sorting. This allowed us to identify Nbs binding the epidermal growth factor receptor that were not previously isolated in a similar screening of a multicopy plasmid library. Our results demonstrate that MAIN enables large gene library integration into the E. coli chromosome, creating stably expressed in-frame fusions for functional screening.


Assuntos
DNA , Escherichia coli , Escherichia coli/genética , Plasmídeos , Biblioteca Gênica , Cromossomos Bacterianos
3.
Environ Res ; 216(Pt 2): 114611, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36283441

RESUMO

While mercury (Hg) is a major concern in all aquatic environments because of its methylation and biomagnification pathways, very few studies consider Hg cycling in remote alpine lakes which are sensitive ecosystems. Nineteen high-altitude pristine lakes from Western/Central Pyrenees were investigated on both northern (France) and southern (Spain) slopes (1620-2600 m asl.). Subsurface water samples were collected in June 2017/2018/2019 and October 2017/2018 for Hg speciation analysis of inorganic mercury (iHg(II)), monomethylmercury (MMHg), and dissolved gaseous mercury (DGM) to investigate spatial and seasonal variations. In June 2018/2019 and October 2018, more comprehensive studies were performed in four lakes by taking water column depth profiles. Besides, in-situ incubation experiments using isotopically enriched Hg species (199iHg(II), 201MMHg) were conducted to investigate Hg transformation mechanisms in the water column. While iHg(II) (0.08-1.10 ng L-1 in filtered samples; 0.11-1.19 ng L-1 in unfiltered samples) did not show significant seasonal variations in the subsurface water samples, MMHg (<0.03-0.035 ng L-1 in filtered samples; <0.03-0.062 ng L-1 in unfiltered samples) was significantly higher in October 2018, mainly because of in-situ methylation. DGM (0.02-0.68 ng L-1) varies strongly and can exhibit higher levels in comparison with other pristine areas. Depth profiles and incubation experiments highlighted the importance of in-situ biotic methylation triggered by anoxic conditions in bottom waters. In-situ incubations confirm that significant methylation, demethylation and photoreduction extents are taking place in the water columns. Overall, drastic environmental changes occurring daily and seasonally in alpine lakes are providing conditions that can both promote Hg methylation (stratified anoxic waters) and MMHg photodemethylation (intense UV light). In addition, light induced photoreduction is a major pathway controlling significant gaseous Hg evasion. Global warming and potential eutrophication may thus have direct implications on Hg turnover and MMHg burden in those remote ecosystems.


Assuntos
Mercúrio , Compostos de Metilmercúrio , Poluentes Químicos da Água , Mercúrio/análise , Lagos , Ecossistema , Monitoramento Ambiental , Altitude , Poluentes Químicos da Água/análise , Gases/análise , Água/análise , Compostos de Metilmercúrio/análise
4.
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
5.
Front Med (Lausanne) ; 9: 1058455, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36507540

RESUMO

Triple-negative breast cancer (TNBC) is characterized by aggressiveness and high rates of metastasis. The identification of relevant biomarkers is crucial to improve outcomes for TNBC patients. Membrane type 1-matrix metalloproteinase (MT1-MMP) could be a good candidate because its expression has been reported to correlate with tumor malignancy, progression and metastasis. Moreover, single-domain variable regions (VHHs or Nanobodies) derived from camelid heavy-chain-only antibodies have demonstrated improvements in tissue penetration and blood clearance, important characteristics for cancer imaging. Here, we have developed a nanobody-based PET imaging strategy for TNBC detection that targets MT1-MMP. A llama-derived library was screened against the catalytic domain of MT1-MMP and a panel of specific nanobodies were identified. After a deep characterization, two nanobodies were selected to be labeled with gallium-68 (68Ga). ImmunoPET imaging with both ([68Ga]Ga-NOTA-3TPA14 and [68Ga]Ga-NOTA-3CMP75) in a TNBC mouse model showed precise tumor-targeting capacity in vivo with high signal-to-background ratios. (68Ga)Ga-NOTA-3CMP75 exhibited higher tumor uptake compared to (68Ga)Ga-NOTA-3TPA14. Furthermore, imaging data correlated perfectly with the immunohistochemistry staining results. In conclusion, we found a promising candidate for nanobody-based PET imaging to be further investigated as a diagnostic tool in TNBC.

6.
Nature ; 608(7922): 267-268, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35948705
7.
Microb Cell Fact ; 21(1): 133, 2022 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-35780105

RESUMO

BACKGROUND: Bacterial type III secretion systems (T3SSs) assemble a multiprotein complex termed the injectisome, which acts as a molecular syringe for translocation of specific effector proteins into the cytoplasm of host cells. The use of injectisomes for delivery of therapeutic proteins into mammalian cells is attractive for biomedical applications. With that aim, we previously generated a non-pathogenic Escherichia coli strain, called Synthetic Injector E. coli (SIEC), which assembles functional injectisomes from enteropathogenic E. coli (EPEC). The assembly of injectisomes in EPEC is assisted by the lytic transglycosylase EtgA, which degrades the peptidoglycan layer. As SIEC lacks EtgA, we investigated whether expression of this transglycosylase enhances the protein translocation capacity of the engineered bacterium. RESULTS: The etgA gene from EPEC was integrated into the SIEC chromosome under the control of the inducible tac promoter, generating the strain SIEC-eEtgA. The controlled expression of EtgA had no effect on the growth or viability of bacteria. Upon induction, injectisome assembly was ~ 30% greater in SIEC-eEtgA than in the parental strain, as determined by the level of T3SS translocon proteins, the hemolytic activity of the bacterial strain, and the impairment in flagellar motility. The functionality of SIEC-eEtgA injectisomes was evaluated in a derivative strain carrying a synthetic operon (eLEE5), which was capable of delivering Tir effector protein into the cytoplasm of HeLa cells triggering F-actin polymerization beneath the attached bacterium. Lastly, using ß-lactamase as a reporter of T3SS-protein injection, we determined that the protein translocation capacity was ~ 65% higher in the SIEC-EtgA strain than in the parental SIEC strain. CONCLUSIONS: We demonstrate that EtgA enhances the assembly of functional injectisomes in a synthetic injector E. coli strain, enabling the translocation of greater amounts of proteins into the cytoplasm of mammalian cells. Accordingly, EtgA expression may boost the protein translocation of SIEC strains programmed as living biotherapeutics.


Assuntos
Engenharia Celular , Escherichia coli Enteropatogênica , Proteínas de Escherichia coli , Glicosiltransferases , Engenharia Celular/métodos , Escherichia coli Enteropatogênica/química , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Glicosiltransferases/metabolismo , Células HeLa , Humanos , Transporte Proteico
8.
Microb Biotechnol ; 15(9): 2309-2323, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35695013

RESUMO

The ability of T7 RNA polymerase (RNAPT7 ) fusions to cytosine deaminases (CdA) for entering C➔T changes in any DNA segment downstream of a T7 promoter was exploited for hyperdiversification of defined genomic portions of Pseudomonas putida KT2440. To this end, test strains were constructed in which the chromosomally encoded pyrF gene (the prokaryotic homologue of yeast URA3) was flanked by T7 transcription initiation and termination signals and also carried plasmids expressing constitutively either high-activity (lamprey's) or low-activity (rat's) CdA-RNAPT7 fusions. The DNA segment-specific mutagenic action of these fusions was then tested in strains lacking or not uracil-DNA glycosylase (UDG), that is ∆ung/ung+ variants. The resulting diversification was measured by counting single nucleotide changes in clones resistant to 5-fluoroorotic acid (5FOA), which otherwise is transformed by wild-type PyrF into a toxic compound. Although the absence of UDG dramatically increased mutagenic rates with both CdA-RNAPT7 fusions, the most active variant - pmCDA1 - caused extensive appearance of 5FOA-resistant colonies in the wild-type strain not limited to C➔T but including also a range of other changes. Furthermore, the presence/absence of UDG activity swapped cytosine deamination preference between DNA strands. These qualities provided the basis of a robust system for continuous evolution of preset genomic portions of P. putida and beyond.


Assuntos
Evolução Molecular Direcionada , Genes Bacterianos , Mutagênese , Pseudomonas putida , Citosina/metabolismo , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , DNA Bacteriano/genética , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Evolução Molecular Direcionada/métodos , Loci Gênicos , Genômica , Mutação , Plasmídeos/genética , Pseudomonas putida/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
9.
Front Immunol ; 13: 863831, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35547740

RESUMO

The emergence of SARS-CoV-2 variants that escape from immune neutralization are challenging vaccines and antibodies developed to stop the COVID-19 pandemic. Thus, it is important to establish therapeutics directed toward multiple or specific SARS-CoV-2 variants. The envelope spike (S) glycoprotein of SARS-CoV-2 is the key target of neutralizing antibodies (Abs). We selected a panel of nine nanobodies (Nbs) from dromedary camels immunized with the receptor-binding domain (RBD) of the S, and engineered Nb fusions as humanized heavy chain Abs (hcAbs). Nbs and derived hcAbs bound with subnanomolar or picomolar affinities to the S and its RBD, and S-binding cross-competition clustered them in two different groups. Most of the hcAbs hindered RBD binding to its human ACE2 (hACE2) receptor, blocked cell entry of viruses pseudotyped with the S protein and neutralized SARS-CoV-2 infection in cell cultures. Four potent neutralizing hcAbs prevented the progression to lethal SARS-CoV-2 infection in hACE2-transgenic mice, demonstrating their therapeutic potential. Cryo-electron microscopy identified Nb binding epitopes in and out the receptor binding motif (RBM), and showed different ways to prevent virus binding to its cell entry receptor. The Nb binding modes were consistent with its recognition of SARS-CoV-2 RBD variants; mono and bispecific hcAbs efficiently bound all variants of concern except omicron, which emphasized the immune escape capacity of this latest variant.


Assuntos
COVID-19 , Anticorpos de Domínio Único , Animais , Microscopia Crioeletrônica , Epitopos/química , Humanos , Camundongos , Pandemias , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus
10.
Environ Sci Process Impacts ; 24(9): 1430-1442, 2022 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-35080575

RESUMO

The speciation of both redox reactive and volatile selenium (Se) compounds, barely reported in pristine aquatic environments, has never been investigated in remote alpine lakes, considered as sensitive ecosystems to detect the effect of global change. This work presents an integrated investigation on Se distribution and speciation conducted in 20 high altitude pristine lakes from the central-western Pyrenees. Five seasonal sampling campaigns were carried out after snowmelt (June/July) and in early fall (October) for the period 2017-2019. Concentrations of total dissolved Se (TDSe) ranged from 7 to 78 ng L-1, with selenate being ubiquitously observed in most cases (median of 61% of TDSe). Selenite was only occasionally detected up to 4 ng L-1, therefore a fraction of TDSe was presumably in the forms of elemental Se(0) and/or selenides. Depth profiles obtained in different lakes showed the occurrence of such Se(-II, 0) pools in bottom hypoxic to anoxic waters. The production of volatile Se compounds presented a low median total concentration (TVSe) of 33 pg L-1 (range 3-120 pg L-1), mainly in the form of dimethylselenide in subsurface samples (median of 82% of TVSe). The Se concentration in lake waters was significantly correlated with the sulphate concentration (ρ = 0.93, p < 0.0001), demonstrating that it is influenced by erosion and dissolution of Se and S-enriched parent bedrocks. In addition, for Se depleted alpine lake-bedrock systems, long-range transport and wet atmospheric depositions represent a major source of Se for lake waters.


Assuntos
Lagos , Selênio , Ecossistema , Ácido Selênico , Ácido Selenioso , Selênio/análise , Espanha , Sulfatos
11.
Microb Biotechnol ; 15(5): 1374-1391, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34755474

RESUMO

The probiotic strain Escherichia coli Nissle 1917 (EcN) is a common bacterial chassis in synthetic biology developments for therapeutic applications given its long track record of safe administration in humans. Chromosomal integration of the genes of interest (GOIs) in the engineered bacterium offers significant advantages in genetic stability and to control gene dose, but common methodologies relying on the transformation of EcN are inefficient. In this work, we implement in EcN the use of bacterial conjugation in combination with markerless genome engineering to efficiently insert multiple GOIs at different loci of EcN chromosome, leaving no antibiotic resistance genes, vector sequences or scars in the modified bacterium. The resolution of cointegrants that leads to markerless insertion of the GOIs requires expression of I-SceI endonuclease and its efficiency is enhanced by λ Red proteins. We show the potential of this strategy by integrating different genes encoding fluorescent and bioluminescent reporters (i.e. GFP, mKate2, luxCDABE) both individually and sequentially. We also demonstrate its application for gene deletions in EcN (ΔflhDC) and to replace the endogenous regulation of chromosomal locus (i.e. flhDC) by heterologous regulatory elements (e.g. tetR-Ptet) in order to have an ectopic control of gene expression in EcN with an external inducer to alter bacterial behaviour (e.g. flagellar motility). Whole-genome sequencing confirmed the introduction of the designed modifications without off-target alterations in the genome. This straightforward approach accelerates the generation of multiple modifications in EcN chromosome for the generation of living bacterial therapeutics.


Assuntos
Infecções por Escherichia coli , Proteínas de Escherichia coli , Probióticos , Cromossomos , Conjugação Genética , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Humanos
12.
Ann N Y Acad Sci ; 1506(1): 98-117, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34786712

RESUMO

Synthetic biology has the potential to transform cell- and gene-based therapies for a variety of diseases. Sophisticated tools are now available for both eukaryotic and prokaryotic cells to engineer cells to selectively achieve therapeutic effects in response to one or more disease-related signals, thus sparing healthy tissue from potentially cytotoxic effects. This report summarizes the Keystone eSymposium "Synthetic Biology: At the Crossroads of Genetic Engineering and Human Therapeutics," which took place on May 3 and 4, 2021. Given that several therapies engineered using synthetic biology have entered clinical trials, there was a clear need for a synthetic biology symposium that emphasizes the therapeutic applications of synthetic biology as opposed to the technical aspects. Presenters discussed the use of synthetic biology to improve T cell, gene, and viral therapies, to engineer probiotics, and to expand upon existing modalities and functions of cell-based therapies.


Assuntos
Congressos como Assunto/tendências , Engenharia Genética/tendências , Terapia Genética/tendências , Relatório de Pesquisa , Biologia Sintética/tendências , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Baseada em Transplante de Células e Tecidos/tendências , Marcação de Genes/métodos , Marcação de Genes/tendências , Engenharia Genética/métodos , Terapia Genética/métodos , Humanos , Células Matadoras Naturais/imunologia , Aprendizado de Máquina/tendências , Biologia Sintética/métodos , Linfócitos T/imunologia
13.
Commun Biol ; 4(1): 1169, 2021 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-34621006

RESUMO

ssDNA recombineering has been exploited to hyperdiversify genomically-encoded nanobodies displayed on the surface of Escherichia coli for originating new binding properties. As a proof-of-principle a nanobody recognizing the antigen TirM from enterohaemorrhagic E. coli (EHEC) was evolved towards the otherwise not recognized TirM antigen from enteropathogenic E. coli (EPEC). To this end, E. coli cells displaying this nanobody fused to the intimin outer membrane-bound domain were subjected to multiple rounds of mutagenic oligonucleotide recombineering targeting the complementarity determining regions (CDRs) of the cognate VHH gene sequence. Binders to the EPEC-TirM were selected upon immunomagnetic capture of bacteria bearing active variants and nanobodies identified with a new ability to strongly bind the new antigen. The results highlight the power of combining evolutionary properties of bacteria in vivo with oligonucleotide synthesis in vitro for the sake of focusing diversification to specific segments of a gene (or protein thereof) of interest.


Assuntos
Anticorpos Antibacterianos/imunologia , DNA Bacteriano/genética , DNA de Cadeia Simples/genética , Escherichia coli/imunologia , Anticorpos de Domínio Único/imunologia , DNA Bacteriano/metabolismo , DNA de Cadeia Simples/metabolismo
14.
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
15.
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
16.
Sci Rep ; 11(1): 3318, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33558635

RESUMO

Despite unprecedented global efforts to rapidly develop SARS-CoV-2 treatments, in order to reduce the burden placed on health systems, the situation remains critical. Effective diagnosis, treatment, and prophylactic measures are urgently required to meet global demand: recombinant antibodies fulfill these requirements and have marked clinical potential. Here, we describe the fast-tracked development of an alpaca Nanobody specific for the receptor-binding-domain (RBD) of the SARS-CoV-2 Spike protein with potential therapeutic applicability. We present a rapid method for nanobody isolation that includes an optimized immunization regimen coupled with VHH library E. coli surface display, which allows single-step selection of Nanobodies using a simple density gradient centrifugation of the bacterial library. The selected single and monomeric Nanobody, W25, binds to the SARS-CoV-2 S RBD with sub-nanomolar affinity and efficiently competes with ACE-2 receptor binding. Furthermore, W25 potently neutralizes SARS-CoV-2 wild type and the D614G variant with IC50 values in the nanomolar range, demonstrating its potential as antiviral agent.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Afinidade de Anticorpos/genética , COVID-19/imunologia , SARS-CoV-2/imunologia , Anticorpos de Domínio Único/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Enzima de Conversão de Angiotensina 2/imunologia , Animais , COVID-19/virologia , Camelídeos Americanos/imunologia , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Imunização , Masculino , Testes de Neutralização , Biblioteca de Peptídeos , Ligação Proteica/genética , SARS-CoV-2/química , SARS-CoV-2/isolamento & purificação , Glicoproteína da Espícula de Coronavírus/genética , Transfecção
17.
Nat Commun ; 11(1): 6436, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33353963

RESUMO

In vivo mutagenesis systems accelerate directed protein evolution but often show restricted capabilities and deleterious off-site mutations on cells. To overcome these limitations, here we report an in vivo platform to diversify specific DNA segments based on protein fusions between various base deaminases (BD) and the T7 RNA polymerase (T7RNAP) that recognizes a cognate promoter oriented towards the target sequence. Transcriptional elongation of these fusions generates transitions C to T or A to G on both DNA strands and in long DNA segments. To delimit the boundaries of the diversified DNA, the catalytically dead Cas9 (dCas9) is tethered with custom-designed crRNAs as a "roadblock" for BD-T7RNAP elongation. Using this T7-targeted dCas9-limited in vivo mutagenesis (T7-DIVA) system, rapid molecular evolution of the antibiotic resistance gene TEM-1 is achieved. While the efficiency is demonstrated in E. coli, the system can be adapted to a variety of bacterial and eukaryotic hosts.


Assuntos
Aminoidrolases/metabolismo , Proteína 9 Associada à CRISPR/metabolismo , Genoma Bacteriano , Proteínas Recombinantes de Fusão/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Evolução Molecular Direcionada , Escherichia coli/genética , Testes de Sensibilidade Microbiana , Mutagênese/genética , Mutação/genética , RNA Guia de Cinetoplastídeos/genética , Proteínas Virais/metabolismo , beta-Lactamases/metabolismo
18.
PLoS Biol ; 18(12): e3000986, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33378358

RESUMO

Clustering of the enteropathogenic Escherichia coli (EPEC) type III secretion system (T3SS) effector translocated intimin receptor (Tir) by intimin leads to actin polymerisation and pyroptotic cell death in macrophages. The effect of Tir clustering on the viability of EPEC-infected intestinal epithelial cells (IECs) is unknown. We show that EPEC induces pyroptosis in IECs in a Tir-dependent but actin polymerisation-independent manner, which was enhanced by priming with interferon gamma (IFNγ). Mechanistically, Tir clustering triggers rapid Ca2+ influx, which induces lipopolysaccharide (LPS) internalisation, followed by activation of caspase-4 and pyroptosis. Knockdown of caspase-4 or gasdermin D (GSDMD), translocation of NleF, which blocks caspase-4 or chelation of extracellular Ca2+, inhibited EPEC-induced cell death. IEC lines with low endogenous abundance of GSDMD were resistant to Tir-induced cell death. Conversely, ATP-induced extracellular Ca2+ influx enhanced cell death, which confirmed the key regulatory role of Ca2+ in EPEC-induced pyroptosis. We reveal a novel mechanism through which infection with an extracellular pathogen leads to pyroptosis in IECs.


Assuntos
Cálcio/metabolismo , Proteínas de Escherichia coli/metabolismo , Piroptose/fisiologia , Receptores de Superfície Celular/metabolismo , Actinas/metabolismo , Adesinas Bacterianas/metabolismo , Adesinas Bacterianas/fisiologia , Análise por Conglomerados , Escherichia coli Enteropatogênica/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/metabolismo , Infecções por Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiologia , Células HeLa , Humanos , Mucosa Intestinal/metabolismo , Intestinos/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Transporte Proteico , Receptores de Superfície Celular/fisiologia , Transdução de Sinais/fisiologia , Sistemas de Secreção Tipo III/metabolismo
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...