Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Fernanda Gomes is active.

Publication


Featured researches published by Fernanda Gomes.


Current Microbiology | 2016

Control of Bovine Mastitis: Old and Recent Therapeutic Approaches

Fernanda Gomes; Mariana Henriques

Mastitis is defined as the inflammatory response resulting of the infection of the udder tissue and it is reported in numerous species, namely in domestic dairy animals. This pathology is the most frequent disease of dairy cattle and can be potentially fatal. Mastitis is an economically important pathology associated with reduced milk production, changes in milk composition and quality, being considered one of the most costly to dairy industry. Therefore, the majority of research in the field has focused on control of bovine mastitis and many efforts are being made for the development of new and effective anti-mastitis drugs. Antibiotic treatment is an established component of mastitis control programs; however, the continuous search for new therapeutic alternatives, effective in the control and treatment of bovine mastitis, is urgent. This review will provide an overview of some conventional and emerging approaches in the management of bovine mastitis’ infections.


Current Microbiology | 2011

In vitro Activity of Daptomycin, Linezolid and Rifampicin on Staphylococcus epidermidis Biofilms

Bruna Leite; Fernanda Gomes; Pilar Teixeira; Clovis Wesley Oliveira de Souza; Elisabeth Loshchagin Pizzolitto; Rosário Oliveira

Owing to their massive use, Staphylococcus epidermidis has recently developed significant resistance to several antibiotics, and became one of the leading causes of hospital-acquired infections. Current antibiotics are typically ineffective in the eradication of bacteria in biofilm-associated persistent infections. Accordingly, the paucity of effective treatment against cells in this mode of growth is a key factor that potentiates the need for new agents active in the prevention or eradication of biofilms. Daptomycin and linezolid belong to the novel antibiotic therapies that are active against gram-positive cocci. On the other hand, rifampicin has been shown to be one of the most potent, prevalent antibiotics against S. epidermidis biofilms. Therefore, the main aim of this study was to study the susceptibility of S. epidermidis biofilm cells to the two newer antimicrobial agents previously mentioned, and compare the results obtained with the antimicrobial effect of rifampicin, widely used in the prevention/treatment of indwelling medical device infections. To this end the in vitro activities of daptomycin, linezolid, and rifampicin on S. epidermidis biofilms were accessed, using these antibiotics at MIC and peak serum concentrations. The results demonstrated that at MIC concentration, rifampicin was the most effective antibiotic tested. At peak serum concentration, both strains demonstrated similar susceptibility to rifampicin and daptomycin, with colony-forming units (CFUs) reductions of approximately 3–4 log10, with a slightly lower response to linezolid, which was also more strain dependent. However, considering all the parameters studied, daptomycin was considered the most effective antibiotic tested, demonstrating an excellent in vitro activity against S. epidermidis biofilm cells. In conclusion, this antibiotic can be strongly considered as an acceptable therapeutic option for S. epidermidis biofilm-associated infections and can represent a potential alternative to rifampicin in serious infections where rifampicin resistance becomes prevalent.


Biofouling | 2014

Mini-review: Staphylococcus epidermidis as the most frequent cause of nosocomial infections: old and new fighting strategies.

Fernanda Gomes; Pilar Teixeira; Rosário Oliveira

Staphylococcus epidermidis is nowadays regarded as the most frequent cause of nosocomial infections and indwelling medical device-associated infections. One of the features that contributes to the success of this microorganism and which is elemental to the onset of pathogenesis is its ability to form biofilms. Cells in this mode of growth are inherently more resistant to antimicrobials. Seeking to treat staphylococcal-related infections and to prevent their side effects, such as the significant morbidity and health care costs, many efforts are being made to develop of new and effective antistaphylococcal drugs. Indeed, due to its frequency and extreme resistance to treatment, staphylococcal-associated infections represent a serious burden for the public health system. This review will provide an overview of some conventional and emerging anti-biofilm approaches in the management of medical device-associated infections related to this important nosocomial pathogen.


BMC Research Notes | 2012

Confocal laser scanning microscopy analysis of S. epidermidis biofilms exposed to farnesol, vancomycin and rifampicin

Nuno Cerca; Fernanda Gomes; Sofia Pereira; Pilar Teixeira; Rosário Oliveira

BackgroundStaphylococcus epidermidis is the major bacterial species found in biofilm-related infections on indwelling medical devices. Microbial biofilms are communities of bacteria adhered to a surface and surrounded by an extracellular polymeric matrix. Biofilms have been associated with increased antibiotic tolerance to the immune system. This increased resistance to conventional antibiotic therapy has lead to the search for new antimicrobial therapeutical agents. Farnesol, a quorum-sensing molecule in Candida albicans, has been described as impairing growth of several different microorganisms and we have previously shown its potential as an adjuvant in antimicrobial therapy against S. epidermidis. However, its mechanism of action in S. epidermidis is not fully known. In this work we better elucidate the role of farnesol against S: epidermidis biofilms using confocal laser scanning microscopy (CLSM).Findings24 h biofilms were exposed to farnesol, vancomycin or rifampicin and were analysed by CLSM, after stained with a Live/Dead stain, a known indicator of cell viability, related with cell membrane integrity. Biofilms were also disrupted by sonication and viable and cultivable cells were quantified by colony forming units (CFU) plating. Farnesol showed a similar effect as vancomycin, both causing little reduction of cell viability but at the same time inducing significant changes in the biofilm structure. On the other hand, rifampicin showed a distinct action in S. epidermidis biofilms, by killing a significant proportion of biofilm bacteria.ConclusionsWhile farnesol is not very efficient at killing biofilm bacteria, it damages cell membrane, as determined by the live/dead staining, in a similar way as vancomycin. Furthermore, farnesol might induce biofilm detachment, as determined by the reduced biofilm biomass, which can partially explain the previous findings regarding its role as a possible chemotherapy adjuvant.


Enfermedades Infecciosas Y Microbiologia Clinica | 2013

Combined effect of linezolid and N-acetylcysteine against Staphylococcus epidermidis biofilms

Bruna Leite; Fernanda Gomes; Pilar Teixeira; Clovis Wesley Oliveira de Souza; Elisabeth Loshchagin Pizzolitto; Rosário Oliveira

INTRODUCTION Staphylococcus epidermidis is an organism commonly associated with infections caused by biofilms. Biofilms are less sensible to antibiotics and therefore are more difficult to eradicate. Linezolid and N-acetylcysteine (NAC), have demonstrated to be active against gram-positive microorganisms. Therefore and since linezolid and NAC have different modes of action, the main objective of this work was to investigate the single and synergistic effect of linezolid and NAC against S. epidermidis biofilms. METHODS This work reports the in vitro effect of linezolid and NAC against S. epidermidis biofilms, treated with MIC (4mgml(-1)) and 10×MIC of NAC, and MIC (1μgml(-1)) and peak serum concentration (PS=18μgml(-1)) of linezolid alone and in combination. After exposure of S. epidermidis biofilms to linezolid and/or NAC for 24h, several biofilm parameters were evaluated, namely the number of cultivable cells [colony forming unit (CFU) enumeration], total biofilm biomass and cellular activity. RESULTS When tested alone, NAC at 10×MIC was the most effective agent against S. epidermidis biofilms. However, the combination linezolid (MIC)+NAC (10×MIC) showed a synergistic effect and was the most biocidal treatment tested, promoting a 5log reduction in the number of biofilm viable cells. CONCLUSION This combination seems to be a potential candidate to combat infections caused by S. epidermidis biofilms, namely as a catheter lock solution therapy.


Microbial Drug Resistance | 2011

Virulence gene expression by Staphylococcus epidermidis biofilm cells exposed to antibiotics.

Fernanda Gomes; Pilar Teixeira; Nuno Cerca; Howard Ceri; Rosário Oliveira

Staphylococcus epidermidis have become important causes of nosocomial infections, as its pathogenesis is correlated with the ability to form biofilms on polymeric surfaces. Production of poly-N-acetylglucosamine (PNAG) is crucial for S. epidermidis biofilm formation and is synthesized by the gene products of the icaADBC gene cluster. Production of PNAG/polysaccharide intercellular adhesin and biofilm formation are regulated by the alternative sigma factor, σ(B), and is influenced by a variety of environmental conditions including disinfectants and other antimicrobial substances. The susceptibility of five S. epidermidis strains to antibiotics alone and in double combination was previously tested. Our results demonstrated that some combinations are active and present a general broad spectrum against S. epidermidis biofilms, namely rifampicin-clindamycin and rifampicin-gentamicin. In the present study, it was investigated whether the combination of rifampicin with clindamycin and gentamicin and these antibiotics alone influence the expression of specific genes (icaA and rsbU) of S. epidermidis within biofilms using real-time polymerase chain reaction. The data showed that in most cases the expression of both genes tested significantly increased after exposure to antimicrobial agents alone and in combination. Besides having a similar antimicrobial effect, rifampicin combined with clindamycin and gentamicin induced a lower expression of biofilm-related genes relatively to rifampicin alone. Associated with the advantage of combinatorial therapy in avoiding the emergence of antibiotic resistance, this study demonstrated that it can also cause a lower genetic expression of icaA and rsbU genes, which are responsible for PNAG/polysaccharide intercellular adhesin production, and consequently reduce biofilm formation recidivism, relatively to rifampicin alone.


Fems Immunology and Medical Microbiology | 2016

Bovine mastitis disease/pathogenicity: evidence of the potential role of microbial biofilms.

Fernanda Gomes; Maria J. Saavedra; Mariana Henriques

Bovine mastitis (BM) is a disease with high incidence worldwide and one of the most relevant bovine pathologies and the most costly to the dairy industry. BM is an inflammation of the udder and represents one of the most difficult veterinary diseases to control. Biofilm formation is considered a selective advantage for pathogens causing mastitis, facilitating bacterial persistence in the udder. In fact, recently some authors drew attention to the biofilm formation ability presented by several mastitis causing pathogens and to its possible relation with recurrent mastitis infections and with the increased resistance to antimicrobial agents and host immune defence system. Actually, up to now, several researchers reported the potential role of cells in this mode of growth in the previous facts mentioned. As a consequence of the presence of biofilms, the infection here focused is more difficult to treat and eradicate, making this problem a more relevant pressing issue. Thus, we believe that a deeper knowledge of these structures in mastitis can help to determine the best control strategy to be used in veterinary practice in order to reduce losses in the dairy industry and to ensure milk safety and quality. The aim of this paper was to review the existing research and consequently to provide an overview of the role of biofilms in BM infections.


The American Journal of the Medical Sciences | 2011

Farnesol as Antibiotics Adjuvant in Staphylococcus epidermidis Control In Vitro

Fernanda Gomes; Nuno Cerca; Joana Azeredo; Rosário Oliveira; Pilar Teixeira; Bruna Leite

Introduction:Farnesol is a sesquiterpenoid that has been described as impairing bacterial growth. Therefore, the goal of this study was to compare the in vitro postantimicrobial effect (PAE) of farnesol against Staphylococcus epidermidis with the corresponding values of most common practice antibiotics and also to evaluate the combined effect of farnesol with these antibiotics against planktonic and biofilm cells. Methods:After exposure of S epidermidis cells to farnesol and antibiotics at minimum inhibitory concentration for 1 hour, the cells were regrown in medium without any antimicrobial agent. Cellular viability was assessed by colony-forming units, every hour for 12 hours, and then, the PAE was determined. The combined effect of farnesol (0, 30, 100 and 300 &mgr;M) with vancomycin, tetracycline and rifampicin was also evaluated, by using these antibiotics at peak serum concentration. Results:When PAE is concerned, it was found that cells grown in 100 &mgr;M of farnesol behaved similarly to cells that had never been in contact with farnesol, whereas a clear difference was obtained with cells exposed to 300 &mgr;M of farnesol, displaying a longer PAE. Farnesol showed a combined effect with the tested antibiotics against planktonic cells, although this was not so evident against biofilm cells. Conclusions:Despite the reduced efficacy against biofilm cells, farnesol seems to be a potential adjuvant therapeutic agent to antibiotics against S epidermidis planktonic cells. Moreover, its long PAE makes farnesol a potential candidate in the prevention of biofilm formation because it showed to be very effective against planktonic cells alone as well.


American Journal of Therapeutics | 2013

Staphylococcus epidermidis biofilms control by N-acetylcysteine and rifampicin

Bruna Leite; Fernanda Gomes; Pilar Teixeira; Clovis Wesley Oliveira de Souza; Elisabeth Loshchagin Pizzolitto; Rosário Oliveira

Medical device–associated infections caused by Staphylococcus epidermidis usually involve biofilm formation and its eradication is particularly challenging. Although rifampicin has been proving to be one of the most effective antibiotics against S. epidermidis biofilms, its use as a single agent can lead to the acquisition of resistance. Therefore, we assessed the combined effect of rifampicin with N-acetylcysteine (NAC) known by its mucolytic effect, in the control of S. epidermidis biofilms. Biofilms of 2 S. epidermidis strains (9142 and 1457) were treated with 1x minimum inhibitory concentration (4 mg/mL) and 10x minimum inhibitory concentration (40 mg/mL) of NAC and 10 mg/L (peak serum) of rifampicin alone and in combination. NAC at 40 mg/L alone or in combination with rifampicin (10 mg/L) significantly reduced (4 log10) the number of biofilm cells. Considering their different modes of action, the association of NAC with rifampicin constitutes a promising therapeutic strategy in the treatment of infections associated to S. epidermidis biofilms.


Brazilian Journal of Microbiology | 2012

Farnesol in combination with N-acetylcysteine against Staphylococcus epidermidis planktonic and biofilm cells

Fernanda Gomes; Bruna Leite; Pilar Teixeira; Joana Azeredo; Rosário Oliveira

Staphylococcus epidermidis is the most frequent cause of nosocomial sepsis and catheter-related infections, in which biofilm formation is considered to be the main virulence mechanism. In biofilm environment, microbes exhibit enhanced resistance to antimicrobial agents. This fact boosted the search of possible alternatives to antibiotics. Farnesol and N-acetylcysteine (NAC) are non-antibiotic drugs that have demonstrated antibacterial properties. In this study, the effect of farnesol and NAC isolated or in combination (farnesol+NAC) was evaluated. NAC at 10 × MIC caused a total cell death in planktonic cells. On the other hand, S. epidermidis biofilms exhibited 4 log reduction in viable cell number after a 24h treatment with NAC at the former concentration. Our results demonstrated that there was a higher CFU log reduction of S. epidermidis planktonic cells when farnesol was combined with NAC at 1 × MIC relatively to each agent alone. However, these results were not relevant because NAC alone at 10 × MIC was always the condition which gave the best results, having a very high killing effect on planktonic cells and a significant bactericidal effect on biofilm cells. This study demonstrated that no synergy was observed between farnesol and NAC. However, the pronounced antibacterial effect of NAC against S. epidermidis, on both lifestyles, indicates the use of NAC as a potential therapeutic agent in alternative to antibiotics.

Collaboration


Dive into the Fernanda Gomes's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge