Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Microb Cell Fact ; 23(1): 13, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38183102

RESUMEN

BACKGROUND: S. lividans TK24 is a popular host for the production of small molecules and the secretion of heterologous protein. Within its large genome, twenty-nine non-essential clusters direct the biosynthesis of secondary metabolites. We had previously constructed ten chassis strains, carrying deletions in various combinations of specialized metabolites biosynthetic clusters, such as those of the blue actinorhodin (act), the calcium-dependent antibiotic (cda), the undecylprodigiosin (red), the coelimycin A (cpk) and the melanin (mel) clusters, as well as the genes hrdD, encoding a non-essential sigma factor, and matAB, a locus affecting mycelial aggregation. Genome reduction was aimed at reducing carbon flow toward specialized metabolite biosynthesis to optimize the production of secreted heterologous protein. RESULTS: Two of these S. lividans TK24 derived chassis strains showed ~ 15% reduction in biomass yield, 2-fold increase of their total native secretome mass yield and enhanced abundance of several secreted proteins compared to the parental strain. RNAseq and proteomic analysis of the secretome suggested that genome reduction led to cell wall and oxidative stresses and was accompanied by the up-regulation of secretory chaperones and of secDF, a Sec-pathway component. Interestingly, the amount of the secreted heterologous proteins mRFP and mTNFα, by one of these strains, was 12 and 70% higher, respectively, than that secreted by the parental strain. CONCLUSION: The current study described a strategy to construct chassis strains with enhanced secretory abilities and proposed a model linking the deletion of specialized metabolite biosynthetic clusters to improved production of secreted heterologous proteins.


Asunto(s)
Proteómica , Streptomyces lividans , Streptomyces lividans/genética , Transporte de Proteínas , Transporte Biológico , Regulación hacia Arriba
2.
J Inflamm Res ; 15: 4995-5008, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36065319

RESUMEN

Purpose: To verify the antibacterial and immunomodulatory effects of the amylose derivative - chlorite-oxidized oxyamylose (COAM) - in a skin wound setting. Methods: In vitro antibacterial effects of COAM against opportunistic bacterial pathogens common to skin wounds, including Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA), were determined by cultivation methods. The effects of COAM on myeloid cell infiltration into full thickness skin wounds were investigated in wild-type and in transgenic CX3CR1-GFP mice. Results: On the basis of in vitro experiments, an antibacterial effect of COAM against Staphylococcus species including MRSA was confirmed. The minimum inhibitory concentration of COAM was determined as 2000 µg/mL against these bacterial strains. Control full thickness skin wounds yielded maximal neutrophil influxes and no additive effect on neutrophil influx was observed following topical COAM-treatment. However, COAM administration increased local CX3CR1 macrophage counts at days 3 and 4 and induced a trend towards better wound healing. Conclusion: Aside from its known broad antiviral impact, COAM possesses in vitro antibacterial effects specifically against Gram-positive opportunistic pathogens of the skin and modulates in vivo macrophage contents in mouse skin wounds.

3.
J Virol ; 96(6): e0006022, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35107376

RESUMEN

The impact of the host microbiota on arbovirus infections is currently not well understood. Arboviruses are viruses transmitted through the bites of infected arthropods, predominantly mosquitoes or ticks. The first site of arbovirus inoculation is the biting site in the host skin, which is colonized by a complex microbial community that could possibly influence arbovirus infection. We demonstrated that preincubation of arboviruses with certain components of the bacterial cell wall, including lipopolysaccharides (LPS) of some Gram-negative bacteria and lipoteichoic acids or peptidoglycan of certain Gram-positive bacteria, significantly reduced arbovirus infectivity in vitro. This inhibitory effect was observed for arboviruses of different virus families, including chikungunya virus of the Alphavirus genus and Zika virus of the Flavivirus genus, showing that this is a broad phenomenon. A modest inhibitory effect was observed following incubation with a panel of heat-inactivated bacteria, including bacteria residing on the skin. No viral inhibition was observed after preincubation of cells with LPS. Furthermore, a virucidal effect of LPS on viral particles was noticed by electron microscopy. Therefore, the main inhibitory mechanism seems to be due to a direct effect on the virus particles. Together, these results suggest that bacteria are able to decrease the infectivity of alphaviruses and flaviviruses. IMPORTANCE During the past decades, the world has experienced a vast increase in epidemics of alphavirus and flavivirus infections. These viruses can cause severe diseases, such as hemorrhagic fever, encephalitis, and arthritis. Several alpha- and flaviviruses, such as chikungunya virus, Zika virus, and dengue virus, are significant global health threats because of their high disease burden, their widespread (re-)emergence, and the lack of (good) anti-arboviral strategies. Despite the clear health burden, alphavirus and flavivirus infection and disease are not fully understood. A knowledge gap in the interplay between the host and the arbovirus is the potential interaction with host skin bacteria. Therefore, we studied the effect of (skin) bacteria and bacterial cell wall components on alphavirus and flavivirus infectivity in cell culture. Our results show that certain bacterial cell wall components markedly reduced viral infectivity by interacting directly with the virus particle.


Asunto(s)
Alphavirus , Arbovirus , Pared Celular , Flavivirus , Alphavirus/patogenicidad , Alphavirus/fisiología , Animales , Arbovirus/patogenicidad , Arbovirus/fisiología , Bacterias , Virus Chikungunya , Flavivirus/patogenicidad , Flavivirus/fisiología , Lipopolisacáridos , Microbiota , Virus Zika
4.
J Med Chem ; 63(22): 13851-13860, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33191744

RESUMEN

Standard literature procedures for the chemical synthesis of l-threose nucleosides generally employ l-ascorbic acid as starting material. Herein, we have explored two alternative routes that start from either l-arabitol or l-diethyl tartrate, both affording 2-O-methyl-l-threofuranose as a key building block for nucleobase incorporation. The access to multigram quantities of this glycosyl donor in a reproducible fashion allows for the preparation of 2'-deoxy-α-l-threofuranosyl phosphonate nucleosides on a large scale. This methodology was applied to the gram scale synthesis of an aryloxy amidate prodrug of phosphonomethoxydeoxythreosyl adenine. This prodrug exerted potent activity against an entecavir-resistant hepatitis B virus (HBV) strain, while leading to a significant reduction in the levels of HBV covalently closed circular DNA in a cellular assay. Furthermore, its remarkable anti-HBV efficacy was also confirmed in vivo using a hydrodynamic injection-based HBV mouse model, without relevant toxicity and systemic exposure occurring.


Asunto(s)
Antivirales/farmacología , ADN Circular/genética , Farmacorresistencia Viral/efectos de los fármacos , Guanina/análogos & derivados , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B/tratamiento farmacológico , Profármacos/farmacología , Adenina/química , Animales , ADN Circular/análisis , ADN Viral/análisis , ADN Viral/genética , Guanina/farmacología , Hepatitis B/virología , Virus de la Hepatitis B/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Nucleósidos/química , Replicación Viral
5.
Microb Cell Fact ; 17(1): 198, 2018 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-30577858

RESUMEN

BACKGROUND: The Gram-positive Streptomyces lividans TK24 is an attractive host for heterologous protein production because of its high capability to secrete proteins-which favors correct folding and facilitates downstream processing-as well as its acceptance of methylated DNA and its low endogeneous protease activity. However, current inconsistencies in protein yields urge for a deeper understanding of the burden of heterologous protein production on the cell. In the current study, transcriptomics and [Formula: see text]-based fluxomics were exploited to uncover gene expression and metabolic flux changes associated with heterologous protein production. The Rhodothermus marinus thermostable cellulase A (CelA)-previously shown to be successfully overexpressed in S. lividans-was taken as an example protein. RESULTS: RNA-seq and [Formula: see text]-based metabolic flux analysis were performed on a CelA-producing and an empty-plasmid strain under the same conditions. Differential gene expression, followed by cluster analysis based on co-expression and co-localization, identified transcriptomic responses related to secretion-induced stress and DNA damage. Furthermore, the OsdR regulon (previously associated with hypoxia, oxidative stress, intercellular signaling, and morphological development) was consistently upregulated in the CelA-producing strain and exhibited co-expression with isoenzymes from the pentose phosphate pathway linked to secondary metabolism. Increased expression of these isoenzymes matches to increased fluxes in the pentose phosphate pathway. Additionally, flux maps of the central carbon metabolism show increased flux through the tricarboxylic acid cycle in the CelA-producing strain. Redirection of fluxes in the CelA-producing strain leads to higher production of NADPH, which can only partly be attributed to increased secretion. CONCLUSIONS: Transcriptomic and fluxomic changes uncover potential new leads for targeted strain improvement strategies which may ease the secretion stress and metabolic burden associated with heterologous protein synthesis and secretion, and may help create a more consistently performing S. lividans strain. Yet, links to secondary metabolism and redox balancing should be further investigated to fully understand the S. lividans metabolome under heterologous protein production.


Asunto(s)
Familia de Multigenes/genética , Biosíntesis de Proteínas/genética , Streptomyces lividans/metabolismo , Transcriptoma/genética
6.
Front Microbiol ; 9: 3019, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30581427

RESUMEN

Fluorescent proteins are a major cell biology tool to analyze protein sub-cellular topology. Here we have applied this technology to study protein secretion in the Gram-positive bacterium Streptomyces lividans TK24, a widely used host for heterologous protein secretion biotechnology. Green and monomeric red fluorescent proteins were fused behind Sec (SPSec) or Tat (SPTat) signal peptides to direct them through the respective export pathway. Significant secretion of fluorescent eGFP and mRFP was observed exclusively through the Tat and Sec pathways, respectively. Plasmid over-expression was compared to a chromosomally integrated spSec-mRFP gene to allow monitoring secretion under high and low level synthesis in various media. Fluorimetric detection of SPSec-mRFP recorded folded states, while immuno-staining detected even non-folded topological intermediates. Secretion of SPSec-mRFP is unexpectedly complex, is regulated independently of cell growth phase and is influenced by the growth regime. At low level synthesis, highly efficient secretion occurs until it is turned off and secretory preforms accumulate. At high level synthesis, the secretory pathway overflows and proteins are driven to folding and subsequent degradation. High-level synthesis of heterologous secretory proteins, whether secretion competent or not, has a drastic effect on the endogenous secretome, depending on their secretion efficiency. These findings lay the foundations of dissecting how protein targeting and secretion are regulated by the interplay between the metabolome, secretion factors and stress responses in the S. lividans model.

7.
J Infect ; 77(2): 119-130, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29752963

RESUMEN

OBJECTIVES: The opportunistic pathogen Staphylococcus epidermidis is progressively involved in device-related infections. Since these infections involve biofilm formation, antibiotics are not effective. Conversely, a vaccine can be advantageous to prevent these infections. In view of vaccine development, predicted surface proteins were evaluated on their potential as a vaccine target. METHODS: Immunoglobulins directed against S. epidermidis surface proteins SesB, M, O, Q and R were used to firstly affirm their surface location. Further, inhibitory effects of these IgGs on biofilm formation were determined in vitro on polystyrene and polyurethane surfaces and in vivo using a subcutaneous catheter mouse model. We also examined the opsonophagocytotic capacity of these IgGs. RESULTS: Surface localization of the five Ses proteins was demonstrated both for planktonic and sessile cells, though to a variable extent. Ses-specific IgGs added to planktonic cells had a variable inhibitory effect on cell adhesion to polystyrene, while only anti-SesO IgGs decreased cell attachment to polyurethane catheters. Although phagocytic killing was only obtained after opsonization with SesB-specific IgGs, a significant reduction of in vivo formed biofilms was observed after administration of SesB-, SesM- and SesO-specific IgGs. CONCLUSIONS: Regardless of their characterization or function, S. epidermidis surface proteins can be adequate targets for vaccine development aiming the prevention of device-related infections caused by invasive S. epidermidis strains.


Asunto(s)
Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Proteínas de la Membrana/inmunología , Infecciones Estafilocócicas/prevención & control , Staphylococcus epidermidis/metabolismo , Animales , Especificidad de Anticuerpos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas , Regulación Bacteriana de la Expresión Génica/fisiología , Células HL-60 , Humanos , Inmunoglobulina G/inmunología , Conejos , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/inmunología
8.
Eur J Med Chem ; 148: 384-396, 2018 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-29477072

RESUMEN

Aminoacyl-tRNA synthetases (aaRSs) are enzymes that precisely attach an amino acid to its cognate tRNA. This process, which is essential for protein translation, is considered a viable target for the development of novel antimicrobial agents, provided species selective inhibitors can be identified. Aminoacyl-sulfamoyl adenosines (aaSAs) are potent orthologue specific aaRS inhibitors that demonstrate nanomolar affinities in vitro but have limited uptake. Following up on our previous work on substitution of the base moiety, we evaluated the effect of the N3-position of the adenine by synthesizing the corresponding 3-deazaadenosine analogues (aaS3DAs). A typical organism has 20 different aaRS, which can be split into two distinct structural classes. We therefore coupled six different amino acids, equally targeting the two enzyme classes, via the sulfamate bridge to 3-deazaadenosine. Upon evaluation of the inhibitory potency of the obtained analogues, a clear class bias was noticed, with loss of activity for the aaS3DA analogues targeting class II enzymes when compared to the equivalent aaSA. Evaluation of the available crystallographic structures point to the presence of a conserved water molecule which could have importance for base recognition within class II enzymes, a property that can be explored in future drug design efforts.


Asunto(s)
Aminoacil-ARNt Sintetasas/antagonistas & inhibidores , Antiinfecciosos/química , Tubercidina/química , Aminoácidos/química , Diseño de Fármacos , Proteínas de Escherichia coli , Ácidos Sulfónicos/química , Tubercidina/farmacología
9.
Microb Cell Fact ; 16(1): 232, 2017 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-29274637

RESUMEN

BACKGROUND: The gene encoding a thermostable cellulase of family 12 was previously isolated from a Rhodothermus marinus through functional screening. CelA is a protein of 260 aminoacyl residues with a 28-residue amino-terminal signal peptide. Mature CelA was poorly synthesized in some Escherichia coli strains and not at all in others. Here we present an alternative approach for its heterologous production as a secreted polypeptide in Streptomyces. RESULTS: CelA was successfully over-expressed as a secreted polypeptide in Streptomyces lividans TK24. To this end, CelA was fused C-terminally to the secretory signal peptide of the subtilisin inhibitor protein (Sianidis et al. in J Biotechnol. 121: 498-507, 2006) from Streptomyces venezuelae and a new cloning strategy developed. Optimal growth media and conditions that stall biomass production promote excessive CelA secretion. Under optimal growth conditions in nutrient broth medium, significant amounts of mature CelA (50-90 mg/L or 100-120 mg/g of dry cell weight) are secreted in the spent growth media after 7 days. A protocol to rapidly purify CelA to homogeneity from culture supernatants was developed and specific anti-sera raised against it. Biophysical, biochemical and immmuno-detection analyses indicate that the enzyme is intact, stable and fully functional. CelA is the most thermostable heterologous polypeptide shown to be secreted from S. lividans. CONCLUSION: This study further validates and extends the use of the S. lividans platform for production of heterologous enzymes of industrial importance and extends it to active thermostable enzymes. This study contributes to developing a platform for poly-omics analysis of protein secretion in S. lividans.


Asunto(s)
Proteínas Bacterianas/metabolismo , Celulasa/metabolismo , Expresión Génica , Rhodothermus/enzimología , Streptomyces lividans/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Celulasa/química , Celulasa/genética , Estabilidad de Enzimas , Calor , Transporte de Proteínas , Rhodothermus/genética , Streptomyces lividans/metabolismo
10.
Infect Genet Evol ; 43: 222-4, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27259364

RESUMEN

Staphylococcus epidermidis is one of the major concerns with respect to hospital-acquired infections. Therefore, a rapid and easy method to identify at species level S. epidermidis isolates out of a broad range of bacteria is necessary. Based on earlier studies, the sesC gene encoding a S. epidermidis surface protein revealed to be a highly conserved gene in this species. By means of an easy and inexpensive PCR assay, the presence of sesC was checked in 438 clinical staphylococcal isolates. Results showed that sesC is specifically present in all S. epidermidis. In conclusion, the sesC gene can be exploited as a genetic marker in order to distinguish S. epidermidis from other isolates.


Asunto(s)
Proteínas Bacterianas/genética , Infección Hospitalaria/diagnóstico , Proteínas de la Membrana/genética , Infecciones Estafilocócicas/diagnóstico , Staphylococcus epidermidis/genética , Técnicas de Tipificación Bacteriana , Infección Hospitalaria/microbiología , Cartilla de ADN/química , Expresión Génica , Marcadores Genéticos , Humanos , Reacción en Cadena de la Polimerasa/métodos , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/clasificación , Staphylococcus epidermidis/aislamiento & purificación
11.
PLoS One ; 11(1): e0146704, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26799073

RESUMEN

Staphylococcus epidermidis is the most common cause of device-associated infections. It has been shown that active and passive immunization in an animal model against protein SesC significantly reduces S. epidermidis biofilm-associated infections. In order to elucidate its role, knock-out of sesC or isolation of S. epidermidis sesC-negative mutants were attempted, however, without success. As an alternative strategy, sesC was introduced into Staphylococcus aureus 8325-4 and its isogenic icaADBC and srtA mutants, into the clinical methicillin-sensitive S. aureus isolate MSSA4 and the MRSA S. aureus isolate BH1CC, which all lack sesC. Transformation of these strains with sesC i) changed the biofilm phenotype of strains 8325-4 and MSSA4 from PIA-dependent to proteinaceous even though PIA synthesis was not affected, ii) converted the non-biofilm-forming strain 8325-4 ica::tet to a proteinaceous biofilm-forming strain, iii) impaired PIA-dependent biofilm formation by 8325-4 srtA::tet, iv) had no impact on protein-mediated biofilm formation of BH1CC and v) increased in vivo catheter and organ colonization by strain 8325-4. Furthermore, treatment with anti-SesC antibodies significantly reduced in vitro biofilm formation and in vivo colonization by these transformants expressing sesC. These findings strongly suggest that SesC is involved in S. epidermidis attachment to and subsequent biofilm formation on a substrate.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Infecciones Relacionadas con Catéteres/microbiología , Proteínas de la Membrana/genética , Staphylococcus aureus/patogenicidad , Staphylococcus epidermidis/patogenicidad , Adhesinas Bacterianas/metabolismo , Animales , Proteínas Bacterianas/genética , Catéteres Venosos Centrales/microbiología , Regulación Bacteriana de la Expresión Génica , Venas Yugulares/cirugía , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Microscopía Electrónica de Rastreo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus epidermidis/genética
12.
Oncotarget ; 5(7): 1761-9, 2014 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-24732092

RESUMEN

Spores of some species of the strictly anaerobic bacteria Clostridium naturally target and partially lyse the hypoxic cores of tumors, which tend to be refractory to conventional therapies. The anti-tumor effect can be augmented by engineering strains to convert a non-toxic prodrug into a cytotoxic drug specifically at the tumor site by expressing a prodrug-converting enzyme (PCE). Safe doses of the favored prodrug CB1954 lead to peak concentrations of 6.3 µM in patient sera, but at these concentration(s) known nitroreductase (NTR) PCEs for this prodrug show low activity. Furthermore, efficacious and safe Clostridium strains that stably express a PCE have not been reported. Here we identify a novel nitroreductase from Neisseria meningitidis, NmeNTR, which is able to activate CB1954 at clinically-achievable serum concentrations. An NmeNTR expression cassette, which does not contain an antibiotic resistance marker, was stably localized to the chromosome of Clostridium sporogenes using a new integration method, and the strain was disabled for safety and containment by making it a uracil auxotroph. The efficacy of Clostridium-Directed Enzyme Prodrug Therapy (CDEPT) using this system was demonstrated in a mouse xenograft model of human colon carcinoma. Substantial tumor suppression was achieved, and several animals were cured. These encouraging data suggest that the novel enzyme and strain engineering approach represent a promising platform for the clinical development of CDEPT.


Asunto(s)
Antineoplásicos/metabolismo , Aziridinas/metabolismo , Terapia Biológica , Carcinoma/terapia , Clostridium/enzimología , Neoplasias del Colon/terapia , Nitrorreductasas/metabolismo , Esporas Bacterianas/enzimología , Animales , Antineoplásicos/sangre , Aziridinas/sangre , Terapia Biológica/efectos adversos , Clostridium/genética , Ratones , Neisseria meningitidis/enzimología , Neisseria meningitidis/genética , Nitrorreductasas/genética , Nitrorreductasas/aislamiento & purificación , Organismos Modificados Genéticamente , Plásmidos , Profármacos/metabolismo , Ingeniería de Proteínas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Biochim Biophys Acta ; 1843(8): 1750-61, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24412306

RESUMEN

Proteins secreted by Gram-positive bacteria are released into the culture medium with the obvious benefit that they usually retain their native conformation. This property makes these host cells potentially interesting for the production of recombinant proteins, as one can take full profit of established protocols for the purification of active proteins. Several state-of-the-art strategies to increase the yield of the secreted proteins will be discussed, using Streptomyces lividans as an example and compared with approaches used in some other host cells. It will be shown that approaches such as increasing expression and translation levels, choice of secretion pathway and modulation of proteins thereof, avoiding stress responses by changing expression levels of specific (stress) proteins, can be helpful to boost production yield. In addition, the potential of multi-omics approaches as a tool to understand the genetic background and metabolic fluxes in the host cell and to seek for new targets for strain and protein secretion improvement is discussed. It will be shown that S. lividans, along with other Gram-positive host cells, certainly plays a role as a production host for recombinant proteins in an economically viable way. This article is part of a Special Issue entitled: Protein trafficking and secretion in bacteria. Guest Editors: Anastassios Economou and Ross Dalbey.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Proteínas Bacterianas/genética , Biotecnología/métodos , Medios de Cultivo , Transporte de Proteínas/genética , Proteínas Recombinantes/genética , Streptomyces lividans/genética , Streptomyces lividans/crecimiento & desarrollo , Biología de Sistemas
14.
J Biomed Mater Res A ; 102(1): 215-24, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23661274

RESUMEN

Implant-related infections are a serious complication in prosthetic surgery, substantially jeopardizing implant fixation. As porous coatings for improved osseointegration typically present an increased surface roughness, their resulting large surface area (sometimes increasing with over 700% compared to an ideal plane) renders the implant extremely susceptible to bacterial colonization and subsequent biofilm formation. Therefore, there is particular interest in orthopaedic implantology to engineer surfaces that combine both the ability to improve osseointegration and at the same time reduce the infection risk. As part of this orthopaedic coating development, the interest of in vitro studies on the interaction between implant surfaces and bacteria/biofilms is growing. In this study, the in vitro staphylococcal adhesion and biofilm formation on newly developed porous pure Ti coatings with 50% porosity and pore sizes up to 50 µm is compared to various dense and porous Ti or Ti-6Al-4V reference surfaces. Multiple linear regression analysis indicates that surface roughness and hydrophobicity are the main determinants for bacterial adherence. Accordingly, the novel coatings display a significant reduction of up to five times less bacterial surface colonization when compared to a commercial state-of-the-art vacuum plasma sprayed coating. However, the results also show that a further expansion of the porosity with over 15% and/or the pore size up to 150 µm is correlated to a significant increase in the roughness parameters resulting in an ascent of bacterial attachment. Chemically modifying the Ti surface in order to improve its hydrophilicity, while preserving the average roughness, is found to strongly decrease bacteria quantities, indicating the importance of surface functionalization to reduce the infection risk of porous coatings.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Staphylococcus aureus/fisiología , Staphylococcus epidermidis/fisiología , Titanio/química , Porosidad , Humectabilidad
15.
Math Biosci ; 246(1): 113-21, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24041624

RESUMEN

The metabolic impact exerted on a microorganism due to heterologous protein production is still poorly understood in Streptomyces lividans. In this present paper, based on exometabolomic data, a proposed genome-scale metabolic network model is used to assess this metabolic impact in S. lividans. Constraint-based modeling results obtained in this work revealed that the metabolic impact due to heterologous protein production is widely distributed in the genome of S. lividans, causing both slow substrate assimilation and a shift in active pathways. Exchange fluxes that are critical for model performance have been identified for metabolites of mouse tumor necrosis factor, histidine, valine and lysine, as well as biomass. Our results unravel the interaction of heterologous protein production with intracellular metabolism of S. lividans, thus, a possible basis for further studies in relieving the metabolic burden via metabolic or bioprocess engineering.


Asunto(s)
Genoma/fisiología , Redes y Vías Metabólicas/fisiología , Modelos Biológicos , Streptomyces lividans/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Ratones
16.
J Microbiol Methods ; 94(3): 192-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23791917

RESUMEN

Recent results with respect to the secretory production of bio-active Mycobacterium tuberculosis proteins in Streptomyces have stimulated the further exploitation of this host as a bacterial cell factory. However, the rapid isolation of a recombinant protein by conventional procedures can be a restrictive step. A previous attempt to isolate recombinant antigens fused to the widely used 6His-tag was found to be relatively incompatible with secretory production in the Streptomyces host. As an alternative, the eight-residue Strep-tag® II (WSHPQFEK), displaying intrinsic binding affinity towards streptavidin, was evaluated for the secretory production of two M. tuberculosis immunodominant antigens in Streptomyces lividans and their subsequent downstream processing. Therefore, the genes ag85A (Rv3804c, encoding the mycolyl-transferase Ag85A) and Rv2626c (encoding hypoxic response protein 1), were equipped with a 3'-Strep-tag® II-encoding sequence and placed under control of the Streptomyces venezuelae CBS762.70 subtilisin inhibitor (vsi) transcriptional, translational and signal sequences. Strep-tagged Ag85A and Rv2626c proteins were detected in the spent medium of recombinant S. lividans cultures at 48h of growth, and purified using a Strep-Tactin Superflow® matrix. Recombinant Ag85A appeared as a 30-kDa protein of which the N-terminal amino acid sequence was identical to the expected one. Rv2626c was produced in two forms of 17 and 37kDa respectively, both with the same predicted N-terminal sequence, suggesting that the 37-kDa product is an Rv2626c dimer. The obtained results indicate that the Strep-tagII is proteolytically stable in Streptomyces and does not interfere with the membrane translocation of Ag85A and Rv2626c. A comparison of reactivity of serum from tuberculosis patients versus healthy persons by ELISA showed that both S. lividans-derived antigens were recognized by sera of individuals infected with M. tuberculosis, indicating that they remained antigenetically active. To our knowledge, this is the first report showing the usefulness of an affinity peptide for detection and efficient downstream processing of recombinant proteins produced in Streptomyces. The present results add up strength to the significance of S. lividans as a valuable host to produce M. tuberculosis proteins with vaccine and diagnostic potential.


Asunto(s)
Aciltransferasas/aislamiento & purificación , Antígenos Bacterianos/aislamiento & purificación , Proteínas Bacterianas/aislamiento & purificación , Oligopéptidos/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Streptomyces lividans/metabolismo , Aciltransferasas/inmunología , Aciltransferasas/metabolismo , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Cromatografía de Afinidad/métodos , Humanos , Mycobacterium tuberculosis/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Estadísticas no Paramétricas , Tuberculosis/inmunología
17.
Adv Healthc Mater ; 1(1): 117-27, 2012 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23184696

RESUMEN

Titanium-based implants are widely used in modern clinical practice; however, complications associated with implants due to bacterial-induced infections arise frequently, caused mainly by staphylococci, streptococci, Pseudomonas spp. and coliform bacteria. Although increased hydrophilicity of the biomaterial surface is known to be beneficial in minimizing the biofilm, quantitative analyses between the actual implant parameters and bacterial development are scarce. Here, the results of in vitro studies of Staphylococcus aureus and Staphylococcus epidermidis proliferation on uncoated and coated titanium materials with different roughness, porosity, topology, and hydrophilicity are shown. The same materials have been tested in parallel with respect to human osteogenic and endothelial cell adhesion, proliferation, and differentiation. The experimental data processed by meta-analysis are indicating the possibility of decreasing the biofilm formation by 80-90% for flat substrates versus untreated plasma-sprayed porous titanium and by 65-95% for other porous titanium coatings. It is also shown that optimized surfaces would lead to 10-50% enhanced cell proliferation and differentiation versus reference porous titanium coatings. This presents an opportunity to manufacture implants with intrinsic reduced infection risk, yet without the additional use of antibacterial substances.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Oseointegración/fisiología , Prótesis e Implantes/microbiología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/prevención & control , Titanio/química , Análisis de Falla de Equipo , Humanos , Ensayo de Materiales , Infecciones Relacionadas con Prótesis/microbiología , Infecciones Relacionadas con Prótesis/prevención & control , Propiedades de Superficie
18.
Appl Microbiol Biotechnol ; 96(2): 367-72, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22797598

RESUMEN

Streptomyces lividans has shown potential as an expression system for heterologous proteins. Overexpression of proteic factors important for heterologous protein production is a valuable approach to improve yields of such proteins. Comparative transcriptomic analysis revealed that several genes were differentially expressed in strains involved in heterologous protein production. For instance, the gene-encoding phosphoenolpyruvate carboxykinase (pepck) showed a significant twofold change in recombinant S. lividans producing human tumour necrosis factor-alpha (hTNF-α). The effect of pepck overexpression on S. lividans TK24 and its hTNF-α producing recombinant was thus investigated in bench-top fermenters. Results obtained revealed that pepck overexpression resulted into a twofold increase in specific PEPCK activity during growth. This overexpression is correlated with slower growth rate, reduced excretion of pyruvate and less alkalinisation of the growth medium when compared with the control strain. After 26 h of fermentation, hTNF-α yields were enhanced (up to 1.7-fold) in the pepck-overexpressing S. lividans TK24, demonstrating that this metabolic engineering approach is indeed promising for heterologous protein production.


Asunto(s)
Proteínas Bacterianas/genética , Expresión Génica , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Streptomyces coelicolor/enzimología , Streptomyces lividans/crecimiento & desarrollo , Factor de Necrosis Tumoral alfa/genética , Proteínas Bacterianas/metabolismo , Humanos , Ingeniería Metabólica , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Streptomyces coelicolor/genética , Streptomyces lividans/genética , Streptomyces lividans/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
19.
Expert Rev Vaccines ; 11(3): 319-34, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22380824

RESUMEN

Previously considered a human commensal, Staphylococcus epidermidis is a frequent cause of nosocomial infections and the most common cause of device-related infections. Because the expression of toxins and other obvious virulence factors is less in S. epidermidis, the biofilm-forming capacity is its major virulence factor. Biofilm growth is characterized by high resistance to antimicrobial agents and host immune responses, making biofilm eradication tremendously difficult. The increasing prevalence of multidrug-resistant S. epidermidis strains additionally hampers antimicrobial therapy. Therefore, immunoprophylaxis and immunotherapy targeting factors expressed at some point in biofilm formation might offer new tools to combat S. epidermidis infections. So far, a limited number of targets have been examined for their immunotherapeutic potential. In this review, we focus on the already tested and possible targets for vaccine development, discuss the accompanying challenges and speculate on future possibilities with respect to immunotherapeutic solutions to deal with S. epidermidis infections.


Asunto(s)
Inmunoterapia/métodos , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/terapia , Vacunas Estafilocócicas/administración & dosificación , Vacunas Estafilocócicas/inmunología , Staphylococcus epidermidis/inmunología , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Infección Hospitalaria/prevención & control , Infección Hospitalaria/terapia , Farmacorresistencia Bacteriana , Humanos , Staphylococcus epidermidis/patogenicidad
20.
J Biotechnol ; 158(4): 159-67, 2012 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-21777629

RESUMEN

The biopharmaceutical market has come a long way since 1982, when the first biopharmaceutical product, recombinant human insulin, was launched. Just over 200 biopharma products have already gained approval. The global market for biopharmaceuticals which is currently valued at over US$99 billion has been growing at an impressive compound annual growth rate over the previous years. To produce these biopharmaceuticals and other industrially important heterologous proteins, different prokaryotic and eukaryotic expression systems are used. All expression systems have some advantages as well as some disadvantages that should be considered in selecting which one to use. Choosing the best one requires evaluating the options--from yield to glycosylation, to proper folding, to economics of scale-up. No host cell from which all the proteins can be universally expressed in large quantities has been found so far. Therefore, it is important to provide a variety of host-vector expression systems in order to increase the opportunities to screen for the most suitable expression conditions or host cell. In this overview, we focus on Streptomyces lividans, a Gram-positive bacterium with a proven excellence in secretion capacity, as host for heterologous protein production. We will discuss its advantages and disadvantages, and how with systems biology approaches strains can be developed to better producing cell factories.


Asunto(s)
Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Streptomyces lividans/genética , Streptomyces lividans/metabolismo , Animales , Biofarmacia/métodos , Vectores Genéticos/genética , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...