Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 1435: 249-272, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38175479

RESUMO

Clostridioides difficile infection (CDI), previously Clostridium difficile infection, is a symptomatic infection of the large intestine caused by the spore-forming anaerobic, gram-positive bacterium Clostridioides difficile. CDI is an important healthcare-associated disease worldwide, characterized by high levels of recurrence, morbidity, and mortality. CDI is observed at a higher rate in immunocompromised patients after antimicrobial therapy, with antibiotics disrupting the commensal microbiota and promoting C. difficile colonization of the gastrointestinal tract.A rise in clinical isolates resistant to multiple antibiotics and the reduced susceptibility to the most commonly used antibiotic molecules have made the treatment of CDI more complicated, allowing the persistence of C. difficile in the intestinal environment.Gut colonization and biofilm formation have been suggested to contribute to the pathogenesis and persistence of C. difficile. In fact, biofilm growth is considered as a serious threat because of the related antimicrobial tolerance that makes antibiotic therapy often ineffective. This is the reason why the involvement of C. difficile biofilm in the pathogenesis and recurrence of CDI is attracting more and more interest, and the mechanisms underlying biofilm formation of C. difficile as well as the role of biofilm in CDI are increasingly being studied by researchers in the field.Findings on C. difficile biofilm, possible implications in CDI pathogenesis and treatment, efficacy of currently available antibiotics in treating biofilm-forming C. difficile strains, and some antimicrobial alternatives under investigation will be discussed here.


Assuntos
Antibacterianos , Biofilmes , Clostridioides difficile , Farmacorresistência Bacteriana , Humanos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Biofilmes/efeitos dos fármacos , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/microbiologia
2.
Adv Exp Med Biol ; 1050: 97-115, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29383666

RESUMO

Clostridium difficile infection (CDI) is an important healthcare-associated disease worldwide, mainly occurring after antimicrobial therapy. Antibiotics administered to treat a number of infections can promote C. difficile colonization of the gastrointestinal tract and, thus, CDI. A rise in multidrug resistant clinical isolates to multiple antibiotics and their reduced susceptibility to the most commonly used antibiotic molecules have made the treatment of CDI more complicated, allowing the persistence of C. difficile in the intestinal environment.Gut colonization and biofilm formation have been suggested to contribute to the pathogenesis and persistence of C. difficile. In fact, biofilm growth is considered as a serious threat because of the related increase in bacterial resistance that makes antibiotic therapy often ineffective. However, although the involvement of the C. difficile biofilm in the pathogenesis and recurrence of CDI is attracting more and more interest, the mechanisms underlying biofilm formation of C. difficile as well as the role of biofilm in CDI have not been extensively described.Findings on C. difficile biofilm, possible implications in CDI pathogenesis and treatment, efficacy of currently available antibiotics in treating biofilm-forming C. difficile strains, and some antimicrobial alternatives under investigation will be discussed here.


Assuntos
Biofilmes , Clostridioides difficile/fisiologia , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/genética , Clostridioides difficile/ultraestrutura , Modelos Biológicos
3.
Adv Exp Med Biol ; 1057: 73-83, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28639245

RESUMO

OBJECTIVE: Biofilm formation has been associated with the persistence of Acinetobacter baumannii in hospital settings and its propensity to cause infection. We investigated the adhesion ability and clonality of 128 A. baumannii isolates recovered from urine and urinary catheters of patients admitted to 5 European hospitals during 1991-2013. METHODS: Isolates identification was confirmed by rpoB sequencing and by the presence of blaOXA-51. The presence of carbapenemases was detected by PCR. Clonality was determined by Sequence Group (SG) identification, Pulsed field gel electrophoresis (PFGE) and Multilocus sequence typing. Adhesion ability was defined by quantitative biofilm production assay and biofilms were characterized by Confocal Laser Microscopy and Scanning Electron Microscopy. RESULTS: The 128 isolates, either resistant (85.9%) or susceptible (14.1%) to carbapenems, and belonging to 50 different PFGE types and 24 different STs, were distributed among SG1 (67.2%), SG2 (10.2%) and other allelic profiles (22.7%). ST218 was the most frequent ST, corresponding to 54,5% of the isolates collected between 2011 and 2013. Among the 109 isolates showing resistance to at least 1 carbapenem, 55% revealed the presence of an acquired carbapenem-hydrolyzing class D - lactamases (CHDL): blaOXA-23 were the most frequent gene detected from 2008 onwards (75%). Among all the clinical isolates, 42.2% were strong biofilm producers, with the older isolates having the highest adhesion ability. Most isolates recovered later, belonging to ST218 and harbouring blaOXA-23, were homogeneously less adhesive. CONCLUSIONS: An evolution towards a decrease in adhesion ability and a CHDL content change was observed along the years in several European countries.


Assuntos
Infecções por Acinetobacter/urina , Acinetobacter baumannii/crescimento & desenvolvimento , Biofilmes , Cateteres Urinários/microbiologia , Acinetobacter baumannii/classificação , Antibacterianos , Proteínas de Bactérias , Eletroforese em Gel de Campo Pulsado , Europa (Continente) , Hospitais , Humanos , Testes de Sensibilidade Microbiana , Tipagem de Sequências Multilocus , beta-Lactamases
4.
Crit Rev Microbiol ; 42(2): 293-309, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25159044

RESUMO

Biofilm recalcitrance is a persistent problem when managing difficult to heal and infected chronic wounds. The wound biofilm is a fundamental factor in the re-occurrence and delayed healing commonly observed in non-healing and infected chronic wounds. However, there is presently no single antimicrobial agent that is completely efficacious against both the planktonic and sessile polymicrobial communities evident in at risk or infected wounds. We will review currently available antimicrobials, with particular emphasis on silver and iodine, employed to help suppress biofilms in wounds. In addition, we will also review the effect of pH on antimicrobial efficacy. Available evidence suggests that it is best to take a multifactorial approach towards controlling biofilm in chronic, "at risk" and infected wounds. This highlights the growing importance of avoiding indiscriminate or inappropriate use of antimicrobials in the treatment of chronic wounds.


Assuntos
Anti-Infecciosos Locais/uso terapêutico , Infecção dos Ferimentos/tratamento farmacológico , Animais , Anti-Infecciosos Locais/administração & dosagem , Anti-Infecciosos Locais/classificação , Biofilmes/efeitos dos fármacos , Curativos Biológicos , Humanos , Concentração de Íons de Hidrogênio , Iodo/química , Iodo/farmacologia , Prata/química , Prata/farmacologia , Resultado do Tratamento , Infecção dos Ferimentos/microbiologia
5.
Adv Exp Med Biol ; 901: 25-36, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26542603

RESUMO

The accumulation of reactive oxygen species (ROS) in microbial biofilms has been recently recognized to play a role in promoting antibiotic resistance in biofilm-growing bacteria. ROS are also over-produced when a medical device is implanted and they can promote device susceptibility to infection or aseptic loosening. High levels of ROS seem also to be responsible for the establishment of chronic wounds.In this study, a novel antioxidant polyacrylate was synthesized and investigated in terms of antimicrobial and antibiofilm activity. The polymer possesses in side-chain hydroxytyrosol (HTy), that is a polyphenolic compound extracted from olive oil wastewaters.The obtained 60 nm in size polymer nanoparticles showed good scavenging and antibacterial activity versus a strain of Staphylococcus epidermidis. Microbial adherence assays evidenced that the hydroxytyrosol-containing polymer was able to significantly reduce bacterial adhesion compared to the control. These findings open novel perspective for a successful use of this antioxidant polymer for the prevention or treatment of biofilm-based infections as those related to medical devices or chronic wounds.


Assuntos
Antibacterianos/farmacologia , Antioxidantes/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Álcool Feniletílico/análogos & derivados , Staphylococcus epidermidis/efeitos dos fármacos , Antibacterianos/química , Antioxidantes/química , Biofilmes/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Álcool Feniletílico/química , Álcool Feniletílico/farmacologia , Staphylococcus epidermidis/crescimento & desenvolvimento , Staphylococcus epidermidis/fisiologia
6.
Adv Exp Med Biol ; 830: 97-112, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25366223

RESUMO

Anaerobic bacteria can cause an infection when they encounter a permissive environment within the host. These opportunistic pathogens are seldom recovered as single isolates but more frequently are involved in polymicrobial infections, together with other anaerobes or aerobes. Nowadays it's known that some anaerobic bacteria are also able to grow as biofilm even if this feature and its role in the healthcare-associated infections (HAIs) are still poorly characterized. As consequence, the involvement of biofilm-forming anaerobic bacteria in infections related to healthcare procedures, including surgery and medical devices implantation, is underestimated.The current knowledge on the role of biofilm-growing anaerobes in HAIs has been here reviewed, with particular reference to respiratory, intestinal, intra-abdominal, wound, and urogenital tract infections. Even if the data are still scarce, the ability to form biofilm of opportunistic anaerobic species and their possible role as causative agents of HAIs should alert even more clinicians and microbiologists on the need to search for anaerobes in clinical samples when their presence can be reasonably assumed.


Assuntos
Bactérias Anaeróbias/fisiologia , Biofilmes/crescimento & desenvolvimento , Infecções Relacionadas a Cateter/microbiologia , Infecções Respiratórias/microbiologia , Infecção dos Ferimentos/microbiologia , Bacteriemia/sangue , Bacteriemia/microbiologia , Bactérias Anaeróbias/ultraestrutura , Humanos , Intestinos/microbiologia , Microscopia Eletrônica de Varredura , Sistema Urogenital/microbiologia
7.
New Microbiol ; 37(2): 119-27, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24858639

RESUMO

Acinetobacter baumannii has received much attention in recent years because of its increasing involvement in a number of severe infections and outbreaks occurring in clinical settings, and presumably related to its ability to survive and persist in hospital environments. The treatment of infections caused by A. baumannii nosocomial strains has become increasingly problematic, due to their intrinsic and/or acquired resistance to multiple classes of antibiotics. Furthermore, the demonstrated ability of nosocomial strains to grow as biofilm is believed to play a significant role in their persistence and antibiotic resistance. This review summarises current knowledge on A. baumannii biofilm formation and its clinical significance, as well as the related genetic determinants and the regulation of this process.


Assuntos
Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/fisiologia , Biofilmes , Acinetobacter baumannii/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos
8.
BMC Microbiol ; 13: 144, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23800205

RESUMO

BACKGROUND: Phylogenetic group D Escherichia coli clones (ST69, ST393, ST405) are increasingly reported as multidrug resistant strains causing extra-intestinal infections. We aim to characterize inter- and intraclonal diversity of a broad sample (isolates from different geographic locations and origins with variable antibiotic resistance profiles, 1980-2010) and their ability to adhere and form biofilm by both a modified quantitative biofilm producing assay and Field Emission Scanning Electron Microscopy (FESEM). RESULTS: High virulence scores were observed among ST69 (median 14/range 9-15) and ST393 (median 14/range 8-15) clones, particularly enriched in pap alleles, iha, kpsMTII-K5 and ompT, in contrast with ST405 (median 6/range 2-14) isolates, exhibiting frequently fyuA, malX and traT. All ST69 and ST393 and only two ST405 isolates were classified as ExPEC. Biofilm production was detected in two non-clinical ST69 and three ST393 isolates from different origins showing variable virulence profiles. Within each clonal group, and despite the high diversity of PFGE-types observed, isolates from different countries and recovered over large periods of time were clustered in a few groups sharing common virulence gene profiles among ST69 (n = 10 isolates) and ST393 (n = 9 isolates) (fimH-iha-iutA-kpsMTII-K5-(traT)-sat-(ompT)-papA-papEF-papGII-papC) or ST405 (n = 6 isolates) (fimH-traT-fyuA-malX). CONCLUSIONS: This study highlights the circulation of highly transmissible ST69, ST393 and ST405 variants among different settings. Biofilm production seems not to be directly correlated with their epidemiological success.


Assuntos
Biofilmes/crescimento & desenvolvimento , Escherichia coli/classificação , Escherichia coli/fisiologia , Variação Genética , Análise por Conglomerados , Eletroforese em Gel de Campo Pulsado , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Genótipo , Humanos , Microscopia Eletrônica de Varredura , Tipagem Molecular , Fatores de Virulência/genética
9.
Microb Ecol Health Dis ; 242013 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-24009545

RESUMO

The use of probiotic products, especially for humans, requires an unequivocal taxonomical definition of their microbial content, in order to assign the probiotic effects to well identified and characterized microbial strains. In the absence of this, the labeling of some marketed probiotics may be misleading, both in terms of microbiological contents and possible beneficial effects. Currently, the 'polyphasic taxonomy' based on the integration of phenotypic and genotypic data seems to be the most appropriate approach. In fact, even if phenotypic characters often overlap among genetically different species, the molecular methods alone are frequently not able to establish distinct boundaries among phylogenetically related species. Thus, a valid scheme for the identification of a probiotic strain should be currently based on its morphological, physiological, and biochemical features as well as on aspects of its genetic profile. It is important that the identity of specific probiotic strains appearing on the product label is the result of a carefully selected combination of suitable phenotypic and genotypic analytical methods. Only adoption of such a policy could give the right emphasis to the significance of strain-specificity and thus provide health authorities with accurate tools to better evaluate the health benefits claimed by each probiotic-based product. The most common phenotypic and genotypic methods are briefly reviewed here with the aim of highlighting the suitable techniques which can be used to differentiate among microorganisms of probiotic interest, particularly those claiming beneficial health effects for humans.

10.
Antimicrob Agents Chemother ; 56(7): 3973-6, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22491693

RESUMO

The characterization of a broad representative sample of ST131 Escherichia coli isolates from different origins and settings (1991 to 2010) revealed that this clonal group has likely diversified recently and that the expansion of particular variants has probably been favored by the capture of diverse, multidrug-resistant IncFII plasmids (pC15-1a, pEK499, pKF3-140-like). The low ability to adhere and to grow as biofilm that was detected in this study suggests unknown mechanisms for the persistence of this clonal group which need to be further explored.


Assuntos
Escherichia coli/genética , Antibacterianos/farmacologia , Biofilmes/crescimento & desenvolvimento , Farmacorresistência Bacteriana Múltipla/genética , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana , Plasmídeos/genética
11.
Microorganisms ; 10(9)2022 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-36144359

RESUMO

Poloxamer 338 (P338), a nonionic surfactant amphiphilic copolymer, is herein proposed as an anti-biofilm compound for the management of catheter-associated urinary tract infections (CAUTIs). P338's ability to disrupt Escherichia coli biofilms on silicone urinary catheters and to serve as antibiotic enhancer was evaluated for biofilm-producing E. coli Ec5FSL and Ec9FSL clinical strains, isolated from urinary catheters. In static conditions, quantitative biofilm formation assay allowed us to determine the active P338 concentration. In dynamic conditions, the BioFlux system, combined with confocal laser scanning microscopy, allowed us to investigate the P338 solution's ability to detach biofilm, alone or in combination with sub-MIC concentrations of cefoxitin (FOX). The 0.5% P338 solution was able to destroy the structure of E. coli biofilms, to reduce the volume and area fraction covered by adherent cells (41.42 ± 4.79% and 56.20 ± 9.22% reduction for the Ec5FSL and Ec9FSL biofilms, respectively), and to potentiate the activity of 1\2 MIC FOX in disaggregating biofilms (19.41 ± 7.41% and 34.66 ± 3.75% reduction in the area fraction covered by biofilm for Ec5FSL and Ec9FSL, respectively) and killing cells (36.85 ± 7.13% and 32.33 ± 4.65% increase in the biofilm area covered by dead Ec5FSL and Ec9FSL cells, respectively).

12.
Gels ; 8(8)2022 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-36005081

RESUMO

Here, we present a one-pot procedure for the preparation of hyaluronic acid (HA) sulfonated hydrogels in aqueous alkaline medium. The HA hydrogels were crosslinked using 1,4-butanedioldiglycidyl ether (BDDE) alone, or together with N-bis(2-hydroxyethyl)-2-aminoethanesulfonic acid (Bes), as a safe sulfonating agent. Conditions for the simultaneous reaction of HA with BDDE and Bes were optimized and the resulting hydrogels were characterized under different reaction times (24, 72, and 96 h). The incorporation of sulfonic groups into the HA network was proven by elemental analysis and FTIR spectroscopy and its effect on water uptake was evaluated. Compared with the non-sulfonated sample, sulfonated gels showed improved mechanical properties, with their compressive modulus increased from 15 to 70 kPa, higher stability towards hyaluronidase, and better biocompatibility to 10T1/2 fibroblasts, especially after the absorption of collagen. As main advantages, the procedure described represents an easy and reproducible methodology for the fabrication of sulfonated hydrogels, which does not require toxic chemicals and/or solvents.

13.
Nat Cell Biol ; 24(10): 1461-1474, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36109671

RESUMO

The common view is that T lymphocytes activate telomerase to delay senescence. Here we show that some T cells (primarily naïve and central memory cells) elongated telomeres by acquiring telomere vesicles from antigen-presenting cells (APCs) independently of telomerase action. Upon contact with these T cells, APCs degraded shelterin to donate telomeres, which were cleaved by the telomere trimming factor TZAP, and then transferred in extracellular vesicles at the immunological synapse. Telomere vesicles retained the Rad51 recombination factor that enabled telomere fusion with T-cell chromosome ends lengthening them by an average of ~3,000 base pairs. Thus, there are antigen-specific populations of T cells whose ageing fate decisions are based on telomere vesicle transfer upon initial contact with APCs. These telomere-acquiring T cells are protected from senescence before clonal division begins, conferring long-lasting immune protection.


Assuntos
Telomerase , Telomerase/genética , Telomerase/metabolismo , Memória Imunológica , Linfócitos T/metabolismo , Telômero/genética , Telômero/metabolismo , Senescência Celular/genética
14.
Front Immunol ; 13: 890298, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35979352

RESUMO

The composition of the intestinal microbiota plays a critical role in shaping the immune system. Modern lifestyle, the inappropriate use of antibiotics, and exposure to pollution have significantly affected the composition of commensal microorganisms. The intestinal microbiota has been shown to sustain inappropriate autoimmune responses at distant sites in animal models of disease, and may also have a role in immune-mediated central nervous system (CNS) diseases such as multiple sclerosis (MS). We studied the composition of the gut mycobiota in fecal samples from 27 persons with MS (pwMS) and in 18 healthy donors (HD), including 5 pairs of homozygous twins discordant for MS. We found a tendency towards higher fungal abundance and richness in the MS group, and we observed that MS twins showed a higher rate of food-associated strains, such as Saccharomyces cerevisiae. We then found that in pwMS, a distinct population of cells with antibacterial and antifungal activity is expanded during the remitting phase and markedly decreases during clinically and/or radiologically active disease. These cells, named MAIT (mucosal-associated invariant T cells) lymphocytes, were significantly more activated in pwMS compared to HD in response to S. cerevisiae and Candida albicans strains isolated from fecal samples. This activation was also mediated by fungal-induced IL-23 secretion by innate immune cells. Finally, immunofluorescent stainings of MS post-mortem brain tissues from persons with the secondary progressive form of the disease showed that MAIT cells cross the blood-brain barrier (BBB) and produce pro-inflammatory cytokines in the brain. These results were in agreement with the hypothesis that dysbiosis of the gut microbiota might determine the inappropriate response of a subset of pathogenic mucosal T cells and favor the development of systemic inflammatory and autoimmune diseases.


Assuntos
Microbioma Gastrointestinal , Células T Invariantes Associadas à Mucosa , Esclerose Múltipla , Animais , Encéfalo , Linfócitos T CD8-Positivos/patologia , Saccharomyces cerevisiae
15.
Neuroscientist ; 26(5-6): 487-502, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32441219

RESUMO

The gut microbiota, consisting of bacteria, fungi, archaea, viruses, and protozoa, together with their collective genomes (microbiome), plays a key role in immune system development and maturation, gut morphology, and in performing essential metabolic functions. Several factors, including lifestyle, body mass index, diet, antibiotic use, and the environment, influence the balance of the intestinal microbiota, whose alterations (the so-called dysbiosis) in recent years have been associated with the onset and/or progression of neurological and neuropsychiatric disorders. The purpose of this narrative review is to provide an overview of the possible involvement of the microbiota-gut-brain axis in the pathogenesis of diseases of the central nervous system, with a special focus on key issues and common misjudgments on the potential contribution of specific microorganisms.


Assuntos
Encéfalo/imunologia , Sistema Nervoso Central/microbiologia , Disbiose/imunologia , Microbioma Gastrointestinal/fisiologia , Doenças Neurodegenerativas/microbiologia , Animais , Encéfalo/metabolismo , Sistema Nervoso Central/imunologia , Disbiose/metabolismo , Humanos , Doenças Neurodegenerativas/imunologia , Neurônios/imunologia
16.
Pathogens ; 9(11)2020 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-33113846

RESUMO

Poloxamers are nontoxic, amphiphilic copolymers used in different formulations. Due to its surfactant properties, Poloxamer 338 (P388) is herein proposed as a strategy to avoid biofilm formation often causing recalcitrant catheter-associated urinary tract infections (CAUTI). The aim is to evaluate the ability of P388 coatings to affect the adhesion of Ec5FSL and Ec9FSL Escherichia coli strains on silicone urinary catheters. Attenuated total reflection infrared spectroscopy, atomic force microscopy, and static water contact angle measurement were employed to characterize the P388-coated silicone catheter in terms of amount of P388 layered, coating thickness, homogeneity, and hydrophilicity. In static conditions, the antifouling power of P388 was defined by comparing the E. coli cells adherent on a hydrophilic P388-adsorbed catheter segment with those on an uncoated one. A P388-coated catheter, having a homogeneous coverage of 35 nm in thickness, reduced of 0.83 log10 and 0.51 log10 the biofilm of Ec5FSL and Ec9FSL, respectively. In dynamic conditions, the percentage of cell adhesion on P388-adsorbed silicone channels was investigated by a microfluidic system, simulating the in vivo conditions of catheterized patients. As a result, both E. coli isolates were undetected. The strong and stable antifouling property against E. coli biofilm lead us to consider P388 as a promising anti-biofilm agent for CAUTIs control.

17.
Drugs ; 79(15): 1635-1655, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31468316

RESUMO

Biofilm-growing cells show an enhanced antimicrobial tolerance with respect to the same cells growing in a free-floating way. This is due to physical or chemical diffusion barriers and increased transfer of resistance markers. Thus, tissue- and medical device-related biofilms can be considered among the leading sources of antibiotic treatment failure, causing many of the deadliest chronic infections afflicting humans nowadays. To find a satisfying way to counteract this major health threat, a great effort has been made in recent years to develop safe, effective and fast-acting anti-biofilm strategies. In this review, we summarise and evaluate the most promising tools and molecules that have demonstrated their ability to modulate steps involved in biofilm formation or to disperse pre-formed biofilms, without conferring evolutionary pressure to microorganisms.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Infecções/tratamento farmacológico , Humanos
18.
APMIS ; 125(4): 392-417, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28407425

RESUMO

The use of implantable medical devices is a common and indispensable part of medical care for both diagnostic and therapeutic purposes. However, as side effect, the implant of medical devices quite often leads to the occurrence of difficult-to-treat infections, as a consequence of the colonization of their abiotic surfaces by biofilm-growing microorganisms increasingly resistant to antimicrobial therapies. A promising strategy to combat device-related infections is based on anti-infective biomaterials that either repel microbes, so they cannot attach to the device surfaces, or kill them in the surrounding areas. In general, such biomaterials are characterized by antifouling coatings, exhibiting low adhesion or even repellent properties towards microorganisms, or antimicrobial coatings, able to kill microbes approaching the surface. In this light, the present overview will address the development in the last two decades of antifouling and antimicrobial biomaterials designed to potentially limit the initial stages of microbial adhesion, as well as the microbial growth and biofilm formation on medical device surfaces.


Assuntos
Materiais Biocompatíveis/química , Biofilmes , Próteses e Implantes/microbiologia , Anti-Infecciosos/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Aderência Bacteriana/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Humanos , Infecções Relacionadas à Prótese/microbiologia , Infecções Relacionadas à Prótese/prevenção & controle
19.
Pathog Dis ; 74(2)2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26656887

RESUMO

Resistance mechanism to metronidazole is still poorly understood, even if the number of reports on Clostridium difficile strains with reduced susceptibility to this antibiotic is increasing. In this study, we investigated the ability of the C. difficile strains 7032994, 7032985 and 7032989, showing different susceptibility profiles to metronidazole but all belonging to the PCR ribotype 010, to form biofilm in vitro in presence and absence of subinhibitory concentrations of metronidazole. The quantitative biofilm production assay performed in presence of metronidazole revealed a significant increase in biofilm formation in both the susceptible strain 7032994 and the strain 7032985 exhibiting a reduced susceptibility to this antibiotic, while antibiotic pressure did not affect the biofilm-forming ability of the stable-resistant strain 7032989. Moreover, confocal microscopy analysis showed an abundant biofilm matrix production by the strains 7032994 and 7032885, when grown in presence of metronidazole, but not in the stable-resistant one. These results seem to demonstrate that subinhibitory concentrations of metronidazole are able to enhance the in vitro biofilm production of the above-mentioned PCR ribotype 010 C. difficile strains, susceptible or with reduced susceptibility to this antibiotic, suggesting a possible role of biofilm formation in the multifactorial mechanism of metronidazole resistance developed by C. difficile.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/fisiologia , Metronidazol/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Clostridioides difficile/classificação , Clostridioides difficile/ultraestrutura
20.
Adv Wound Care (New Rochelle) ; 4(7): 389-397, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26155381

RESUMO

Significance: The presence of a "pathogenic" or "highly virulent" biofilm is a fundamental risk factor that prevents a chronic wound from healing and increases the risk of the wound becoming clinically infected. There is presently no unequivocal gold standard method available for clinicians to confirm the presence of biofilms in a wound. Thus, to help support clinician practice, we devised an algorithm intended to demonstrate evidence of the presence of a biofilm in a wound to assist with wound management. Recent Advances: A variety of histological and microscopic methods applied to tissue biopsies are currently the most informative techniques available for demonstrating the presence of generic (not classified as pathogenic or commensal) biofilms and the effect they are having in promoting inflammation and downregulating cellular functions. Critical Issues: Even as we rely on microscopic techniques to visualize biofilms, they are entities which are patchy and dispersed rather than confluent, particularly on biotic surfaces. Consequently, detection of biofilms by microscopic techniques alone can lead to frequent false-negative results. Furthermore, visual identification using the naked eye of a pathogenic biofilm on a macroscopic level on the wound will not be possible, unlike with biofilms on abiotic surfaces. Future Direction: Lacking specific biomarkers to demonstrate microscopic, nonconfluent, virulent biofilms in wounds, the present focus on biofilm research should be placed on changing clinical practice. This is best done by utilizing an anti-biofilm toolbox approach, rather than speculating on unscientific approaches to identifying biofilms, with or without staining, in wounds with the naked eye. The approach to controlling biofilm should include initial wound cleansing, periodic debridement, followed by the application of appropriate antimicrobial wound dressings. This approach appears to be effective in removing pathogenic biofilms.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA