Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Int J Nanomedicine ; 19: 5419-5437, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38868592

RESUMO

Introduction: Acute myeloid leukemia (AML) remains difficult to treat due to its heterogeneity in molecular landscape, epigenetics and cell signaling alterations. Precision medicine is a major goal in AML therapy towards developing agents that can be used to treat patients with different 'subtypes' in combination with current chemotherapies. We have previously developed dextrin-colistin conjugates to combat the rise in multi-drug resistant bacterial infections and overcome dose-limiting nephrotoxicity. Recent evidence of colistin's anticancer activity, mediated through inhibition of intracellular lysine-specific histone demethylase 1 (LSD1/KDM1A), suggests that dextrin-colistin conjugates could be used to treat cancer cells, including AML. This study aimed to evaluate whether dextrin conjugation (which reduces in vivo toxicity and prolongs plasma half-life) could enhance colistin's cytotoxic effects in myeloid leukemia cell lines and compare the intracellular uptake and localization of the free and conjugated antibiotic. Results: Our results identified a conjugate (containing 8000 g/mol dextrin with 1 mol% succinoylation) that caused significantly increased toxicity in myeloid leukemia cells, compared to free colistin. Dextrin conjugation altered the mechanism of cell death by colistin, from necrosis to caspase 3/7-dependent apoptosis. In contrast, conjugation via a reversible ester linker, instead of an amide, had no effect on the mechanism of the colistin-induced cell death. Live cell confocal microscopy of fluorescently labelled compounds showed both free and dextrin-conjugated colistins were endocytosed and co-localized in lysosomes, and increasing the degree of modification by succinoylation of dextrin significantly reduced colistin internalization. Discussion: Whilst clinical translation of dextrin-colistin conjugates for the treatment of AML is unlikely due to the potential to promote antimicrobial resistance (AMR) and the relatively high colistin concentrations required for anticancer activity, the ability to potentiate the effectiveness of an anticancer drug by polymer conjugation, while reducing side effects and improving biodistribution of the drug, is very attractive, and this approach warrants further investigation.


Assuntos
Apoptose , Colistina , Dextrinas , Colistina/farmacologia , Colistina/química , Colistina/farmacocinética , Dextrinas/química , Dextrinas/farmacologia , Humanos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/farmacocinética , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Sobrevivência Celular/efeitos dos fármacos
2.
Front Cell Infect Microbiol ; 13: 1122340, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36798083

RESUMO

Background: The increasing prevalence of invasive fungal infections in immuno-compromised patients is a considerable cause of morbidity and mortality. With the rapid emergence of antifungal resistance and an inadequate pipeline of new therapies, novel treatment strategies are now urgently required. Methods: The antifungal activity of the alginate oligosaccharide OligoG in conjunction with nystatin was tested against a range of Candida spp. (C. albicans, C. glabrata, C. parapsilosis, C. auris, C. tropicalis and C. dubliniensis), in both planktonic and biofilm assays, to determine its potential clinical utility to enhance the treatment of candidal infections. The effect of OligoG (0-6%) ± nystatin on Candida spp. was examined in minimum inhibitory concentration (MIC) and growth curve assays. Antifungal effects of OligoG and nystatin treatment on biofilm formation and disruption were characterized using confocal laser scanning microscopy (CLSM), scanning electron microscopy (SEM) and ATP cellular viability assays. Effects on the cell membrane were determined using permeability assays and transmission electron microscopy (TEM). Results: MIC and growth curve assays demonstrated the synergistic effects of OligoG (0-6%) with nystatin, resulting in an up to 32-fold reduction in MIC, and a significant reduction in the growth of C. parapsilosis and C. auris (minimum significant difference = 0.2 and 0.12 respectively). CLSM and SEM imaging demonstrated that the combination treatment of OligoG (4%) with nystatin (1 µg/ml) resulted in significant inhibition of candidal biofilm formation on glass and clinical grade silicone surfaces (p < 0.001), with increased cell death (p < 0.0001). The ATP biofilm disruption assay demonstrated a significant reduction in cell viability with OligoG (4%) alone and the combined OligoG/nystatin (MIC value) treatment (p < 0.04) for all Candida strains tested. TEM studies revealed the combined OligoG/nystatin treatment induced structural reorganization of the Candida cell membrane, with increased permeability when compared to the untreated control (p < 0.001). Conclusions: Antimicrobial synergy between OligoG and nystatin against Candida spp. highlights the potential utility of this combination therapy in the prevention and topical treatment of candidal biofilm infections, to overcome the inherent tolerance of biofilm structures to antifungal agents.


Assuntos
Antifúngicos , Candidíase , Humanos , Antifúngicos/farmacologia , Antifúngicos/química , Nistatina/farmacologia , Nistatina/metabolismo , Alginatos/farmacologia , Alginatos/química , Alginatos/metabolismo , Candida , Candidíase/tratamento farmacológico , Candidíase/microbiologia , Candida tropicalis , Candida glabrata , Biofilmes , Oligossacarídeos/farmacologia , Oligossacarídeos/química , Trifosfato de Adenosina/metabolismo , Testes de Sensibilidade Microbiana
3.
Sci Rep ; 12(1): 4986, 2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35322119

RESUMO

In a number of chronic respiratory diseases e.g. cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD), the production of viscous mucin reduces pulmonary function and represents an effective barrier to diffusion of inhaled therapies e.g. antibiotics. Here, a 2-compartment Transwell model was developed to study impaired diffusion of the antibiotic colistin across an artificial sputum (AS) matrix/medium and to quantify its antimicrobial activity against Pseudomonas aeruginosa NH57388A biofilms (alone and in combination with mucolytic therapy). High-performance liquid chromatography coupled with fluorescence detection (HPLC-FLD) revealed that the presence of AS medium significantly reduced the rate of colistin diffusion (> 85% at 48 h; p < 0.05). Addition of alginate oligosaccharide (OligoG CF-5/20) significantly improved colistin diffusion by 3.7 times through mucin-rich AS medium (at 48 h; p < 0.05). Increased diffusion of colistin with OligoG CF-5/20 was shown (using confocal laser scanning microscopy and COMSTAT image analysis) to be associated with significantly increased bacterial killing (p < 0.05). These data support the use of this model to study drug and small molecule delivery across clinically-relevant diffusion barriers. The findings indicate the significant loss of colistin and reduced effectiveness that occurs with mucin binding, and support the use of mucolytics to improve antimicrobial efficacy and lower antibiotic exposure.


Assuntos
Fibrose Cística , Infecções por Pseudomonas , Alginatos/química , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Biofilmes , Colistina/farmacologia , Colistina/uso terapêutico , Fibrose Cística/microbiologia , Humanos , Mucinas/metabolismo , Oligossacarídeos/química , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa
4.
mSphere ; 6(1)2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33472983

RESUMO

Chronic Pseudomonas aeruginosa lung infections in cystic fibrosis (CF) evolve to generate environmentally adapted biofilm communities, leading to increased patient morbidity and mortality. OligoG CF-5/20, a low-molecular-weight inhaled alginate oligomer therapy, is currently in phase IIb/III clinical trials in CF patients. Experimental evolution of P. aeruginosa in response to OligoG CF-5/20 was assessed using a bead biofilm model allowing continuous passage (45 days; ∼245 generations). Mutants isolated after OligoG CF-5/20 treatment typically had a reduced biofilm-forming ability and altered motility profile. Genotypically, OligoG CF-5/20 provided no selective pressure on genomic mutations within morphotypes. Chronic exposure to azithromycin, a commonly prescribed antibiotic in CF patients, with or without OligoG CF-5/20 in the biofilm evolution model also had no effect on rates of resistance acquisition. Interestingly, however, cross-resistance to other antibiotics (e.g., aztreonam) was reduced in the presence of OligoG CF-5/20. Collectively, these findings show no apparent adverse effects from long-term exposure to OligoG CF-5/20, instead resulting in both fewer colonies with multidrug resistance (MDR)-associated phenotypes and improved antibiotic susceptibility of P. aeruginosaIMPORTANCE The emergence of multidrug-resistant (MDR) pathogens within biofilms in the cystic fibrosis lung results in increased morbidity. An inhalation therapy derived from alginate, OligoG CF-5/20, is currently in clinical trials for cystic fibrosis patients. OligoG CF-5/20 has been shown to alter sputum viscoelasticity, disrupt mucin polymer networks, and disrupt MDR pseudomonal biofilms. Long-term exposure to inhaled therapeutics may induce selective evolutionary pressures on bacteria within the lung biofilm. Here, a bead biofilm model with repeated exposure of P. aeruginosa to OligoG CF-5/20 (alone and in combination with azithromycin) was conducted to study these long-term effects and characterize the phenotypic and genotypic adaptations which result. These findings, over 6 weeks, show that long-term use of OligoG CF-5/20 does not lead to extensive mutational changes and may potentially decrease the pathogenicity of the bacterial biofilm and improve the susceptibility of P. aeruginosa to other classes of antibiotics.


Assuntos
Adaptação Fisiológica/genética , Alginatos/química , Biofilmes/efeitos dos fármacos , Genótipo , Fenótipo , Pseudomonas aeruginosa/efeitos dos fármacos , Antibacterianos/farmacologia , Biofilmes/crescimento & desenvolvimento , Farmacorresistência Bacteriana Múltipla , Humanos , Testes de Sensibilidade Microbiana , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/genética , Escarro/microbiologia , Fatores de Tempo
5.
Pharmaceutics ; 12(11)2020 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-33187332

RESUMO

The recent emergence of resistance to colistin, an antibiotic of last resort with dose-limiting toxicity, has highlighted the need for alternative approaches to combat infection. This study aimed to generate and characterise alginate oligosaccharide ("OligoG")-polymyxin (polymyxin B and E (colistin)) conjugates to improve the effectiveness of these antibiotics. OligoG-polymyxin conjugates (amide- or ester-linked), with molecular weights of 5200-12,800 g/mol and antibiotic loading of 6.1-12.9% w/w, were reproducibly synthesised. In vitro inflammatory cytokine production (tumour necrosis factor alpha (TNFα) ELISA) and cytotoxicity (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) of colistin (2.2-9.3-fold) and polymyxin B (2.9-27.2-fold) were significantly decreased by OligoG conjugation. Antimicrobial susceptibility tests (minimum inhibitory concentration (MIC), growth curves) demonstrated similar antimicrobial efficacy of ester- and amide-linked conjugates to that of the parent antibiotic but with more sustained inhibition of bacterial growth. OligoG-polymyxin conjugates exhibited improved selectivity for Gram-negative bacteria in comparison to mammalian cells (approximately 2-4-fold). Both OligoG-colistin conjugates caused significant disruption of Pseudomonas aeruginosa biofilm formation and induced bacterial death (confocal laser scanning microscopy). When conjugates were tested in an in vitro "time-to-kill" (TTK) model using Acinetobacter baumannii, only ester-linked conjugates reduced viable bacterial counts (~2-fold) after 4 h. Bi-functional OligoG-polymyxin conjugates have potential therapeutic benefits in the treatment of multidrug-resistant (MDR) Gram-negative bacterial infections, directly reducing toxicity whilst retaining antimicrobial and antibiofilm activities.

6.
Front Microbiol ; 10: 869, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31105667

RESUMO

Pseudomonas aeruginosa causes problematic chronic lung infections in those suffering from cystic fibrosis. This is due to its antimicrobial resistance mechanisms and its ability to form robust biofilm communities with increased antimicrobial tolerances. Using novel antimicrobials or repurposing current ones is required in order to overcome these problems. Manuka honey is a natural antimicrobial agent that has been used for many decades in the treatment of chronic surface wounds with great success, particularly those infected with P. aeruginosa. Here we aim to determine whether the antimicrobial activity of manuka honey could potentially be repurposed to inhibit pulmonary P. aeruginosa infections using two ex vivo models. P. aeruginosa isolates (n = 28) from an international panel were tested for their susceptibility to manuka honey and clinically relevant antibiotics (ciprofloxacin, ceftazidime, and tobramycin), alone and in combination, using conventional antimicrobial susceptibility testing (AST). To increase clinical applicability, two ex vivo porcine lung (EVPL) models (using alveolar and bronchiolar tissue) were used to determine the anti-biofilm effects of manuka honey alone and in combination with antibiotics. All P. aeruginosa isolates were susceptible to manuka honey, however, varying incidences of resistance were seen against antibiotics. The combination of sub-inhibitory manuka honey and antibiotics using conventional AST had no effect on activity against the majority of isolates tested. Using the two ex vivo models, 64% (w/v) manuka honey inhibited many of the isolates where abnormally high concentrations of antibiotics could not. Typically, combinations of both manuka honey and antibiotics had increased antimicrobial activity. These results highlight the potential of manuka honey as a future antimicrobial for the treatment of pulmonary P. aeruginosa isolates, clearing potential infection reservoirs within the upper airway.

7.
Artigo em Inglês | MEDLINE | ID: mdl-29977590

RESUMO

Acquisition of a mucoid phenotype by Pseudomonas sp. in the lungs of cystic fibrosis (CF) patients, with subsequent over-production of extracellular polymeric substance (EPS), plays an important role in mediating the persistence of multi-drug resistant (MDR) infections. The ability of a low molecular weight (Mn = 3200 g mol-1) alginate oligomer (OligoG CF-5/20) to modify biofilm structure of mucoid Pseudomonas aeruginosa (NH57388A) was studied in vitro using scanning electron microscopy (SEM), confocal laser scanning microscopy (CLSM) with Texas Red (TxRd®)-labelled OligoG and EPS histochemical staining. Structural changes in treated biofilms were quantified using COMSTAT image-analysis software of CLSM z-stack images, and nanoparticle diffusion. Interactions between the oligomers, Ca2+ and DNA were studied using molecular dynamics (MD) simulations, Fourier transform infrared spectroscopy (FTIR) and isothermal titration calorimetry (ITC). Imaging demonstrated that OligoG treatment (≥0.5%) inhibited biofilm formation, revealing a significant reduction in both biomass and biofilm height (P < 0.05). TxRd®-labelled oligomers readily diffused into established (24 h) biofilms. OligoG treatment (≥2%) induced alterations in the EPS of established biofilms; significantly reducing the structural quantities of EPS polysaccharides, and extracellular (e)DNA (P < 0.05) with a corresponding increase in nanoparticle diffusion (P < 0.05) and antibiotic efficacy against established biofilms. ITC demonstrated an absence of rapid complex formation between DNA and OligoG and confirmed the interactions of OligoG with Ca2+ evident in FTIR and MD modelling. The ability of OligoG to diffuse into biofilms, potentiate antibiotic activity, disrupt DNA-Ca2+-DNA bridges and biofilm EPS matrix highlights its potential for the treatment of biofilm-related infections.

8.
Artigo em Inglês | MEDLINE | ID: mdl-28630204

RESUMO

In chronic respiratory disease, the formation of dense, 3-dimensional "microcolonies" by Pseudomonas aeruginosa within the airway plays an important role in contributing to resistance to treatment. An in vitro biofilm model of pseudomonal microcolony formation using artificial-sputum (AS) medium was established to study the effects of low-molecular-weight alginate oligomers (OligoG CF-5/20) on pseudomonal growth, microcolony formation, and the efficacy of colistin. The studies employed clinical cystic fibrosis (CF) isolates (n = 3) and reference nonmucoid and mucoid multidrug-resistant (MDR) CF isolates (n = 7). Bacterial growth and biofilm development and disruption were studied using cell viability assays and image analysis with scanning electron and confocal laser scanning microscopy. Pseudomonal growth in AS medium was associated with increased ATP production (P < 0.05) and the formation (at 48 h) of discrete (>10-µm) microcolonies. In conventional growth medium, colistin retained an ability to inhibit growth of planktonic bacteria, although the MIC was increased (0.1 to 0.4 µg/ml) in AS medium compared to Mueller-Hinton (MH) medium. In contrast, in an established-biofilm model in AS medium, the efficacy of colistin was decreased. OligoG CF-5/20 (≥2%) treatment, however, induced dose-dependent biofilm disruption (P < 0.05) and led to colistin retaining its antimicrobial activity (P < 0.05). While circular dichroism indicated that OligoG CF-5/20 did not change the orientation of the alginate carboxyl groups, mass spectrometry demonstrated that the oligomers induced dose-dependent (>0.2%; P < 0.05) reductions in pseudomonal quorum-sensing signaling. These findings reinforce the potential clinical significance of microcolony formation in the CF lung and highlight a novel approach to treat MDR pseudomonal infections.


Assuntos
Alginatos/farmacologia , Antibacterianos/farmacologia , Biofilmes/crescimento & desenvolvimento , Colistina/farmacologia , Oligossacarídeos/farmacologia , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/efeitos dos fármacos , Infecções Respiratórias/tratamento farmacológico , Biofilmes/efeitos dos fármacos , Fibrose Cística/microbiologia , Farmacorresistência Bacteriana Múltipla , Sinergismo Farmacológico , Ácido Glucurônico/farmacologia , Ácidos Hexurônicos/farmacologia , Humanos , Testes de Sensibilidade Microbiana , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/crescimento & desenvolvimento , Percepção de Quorum/efeitos dos fármacos , Infecções Respiratórias/microbiologia , Escarro/microbiologia
9.
Neurochem Int ; 97: 137-45, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27058627

RESUMO

In this study we sought to investigate the hypothesis that expression of the Stromal Interaction Molecule 1 (STIM1) could provide protection against cell death induced by ER and oxidative stress. STIM1 performs an essential role in regulating store operated calcium entry (SOCE) and thereby provides an important route for replenishment of endoplasmic reticulum (ER) Ca(2+) stores. We used NG115-401L as a model neuronal cell phenotype with a predicted high susceptibility to ER stress due to SOCE deficiency and the absence of STIM1 expression. We show that STIM1 rescue vigorously re-establishes SOCE responses inducible by sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase (SERCA) blockers and Ca(2+)- linked receptors, producing a useful cell line with a simple STIM1/SOCE on/off switch. Surprisingly, we find that expressing STIM1 in NG115-401L cells appears to not have a significant impact on stored ER Ca(2+) levels. Yet, even though we find no evidence for an influence on ER Ca(2+) levels, we observed that provision of STIM1 function and rescue of SOCE activity produced a neuronal phenotype with significantly greater resistance to ER stress induced by SERCA blockade. Moreover, we also report that STIM1 expression, despite elevating mitochondrial reactive oxygen species, endows the NG115-401L neuronal cells with significant resistance to agents that mediate glutathione depletion and subsequent oxidative stress induced apoptosis. Our findings thus suggest that STIM1 warrants further investigation as a potential mediator of neuroprotective pathways against ER and oxidative stress.


Assuntos
Cálcio/metabolismo , Citoproteção/fisiologia , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo , Proteínas de Neoplasias/biossíntese , Estresse Oxidativo/fisiologia , Molécula 1 de Interação Estromal/biossíntese , Animais , Catecóis/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Citoproteção/efeitos dos fármacos , Relação Dose-Resposta a Droga , Retículo Endoplasmático/efeitos dos fármacos , Álcoois Graxos/farmacologia , Proteínas Sensoras de Cálcio Intracelular , Proteínas de Membrana/metabolismo , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Tapsigargina/farmacologia
10.
Bioelectromagnetics ; 37(2): 116-29, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26879225

RESUMO

Potential health effects of radiofrequency (RF) radiation from mobile phones arouse widespread public concern. RF fields from handheld devices near the brain might trigger or aggravate brain tumors or neurodegenerative diseases such as Parkinson's disease (PD). Aggregation of neural α-synuclein (S) is central to PD pathophysiology, and invertebrate models expressing human S have helped elucidate factors affecting the aggregation process. We have recently developed a transgenic strain of Caenorhabditis elegans carrying two S constructs: SC tagged with cyan (C) blue fluorescent protein (CFP), and SV with the Venus (V) variant of yellow fluorescent protein (YFP). During S aggregation in these SC+SV worms, CFP, and YFP tags are brought close enough to allow Foerster Resonance Energy Transfer (FRET). As a positive control, S aggregation was promoted at low Hg(2+) concentrations, whereas higher concentrations activated stress-response genes. Using two different exposure systems described previously, we tested whether RF fields (1.0 GHz CW, 0.002-0.02 W kg(-1); 1.8 GHz CW or GSM, 1.8 W kg(-1)) could influence S aggregation in SC+SV worms. YFP fluorescence in similar SV-only worms provided internal controls, which should show opposite changes due to FRET quenching during S aggregation. No statistically significant changes were observed over several independent runs at 2.5, 24, or 96 h. Although our worm model is sensitive to chemical promoters of aggregation, no similar effects were attributable to RF exposures.


Assuntos
Caenorhabditis elegans , Micro-Ondas , Doença de Parkinson/metabolismo , Agregados Proteicos , alfa-Sinucleína/química , Animais , Modelos Animais de Doenças , Radiometria
11.
Mol Pharm ; 13(3): 863-72, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26833139

RESUMO

The host- and bacteria-derived extracellular polysaccharide coating of the lung is a considerable challenge in chronic respiratory disease and is a powerful barrier to effective drug delivery. A low molecular weight 12-15-mer alginate oligosaccharide (OligoG CF-5/20), derived from plant biopolymers, was shown to modulate the polyanionic components of this coating. Molecular modeling and Fourier transform infrared spectroscopy demonstrated binding between OligoG CF-5/20 and respiratory mucins. Ex vivo studies showed binding induced alterations in mucin surface charge and porosity of the three-dimensional mucin networks in cystic fibrosis (CF) sputum. Human studies showed that OligoG CF-5/20 is safe for inhalation in CF patients with effective lung deposition and modifies the viscoelasticity of CF-sputum. OligoG CF-5/20 is the first inhaled polymer therapy, represents a novel mechanism of action and therapeutic approach for the treatment of chronic respiratory disease, and is currently in Phase IIb clinical trials for the treatment of CF.


Assuntos
Alginatos/química , Fibrose Cística/tratamento farmacológico , Mucinas/química , Muco/química , Oligossacarídeos/química , Polímeros/farmacologia , Adolescente , Adulto , Alginatos/metabolismo , Animais , Doença Crônica , Ensaios Clínicos Fase I como Assunto , Feminino , Ácido Glucurônico/química , Ácido Glucurônico/metabolismo , Ácidos Hexurônicos/química , Ácidos Hexurônicos/metabolismo , Humanos , Masculino , Microscopia de Força Atômica , Microscopia Eletrônica de Varredura , Mucinas/metabolismo , Muco/metabolismo , Oligossacarídeos/metabolismo , Polímeros/química , Ratos , Ratos Sprague-Dawley , Reologia , Espectroscopia de Infravermelho com Transformada de Fourier , Escarro/química , Suínos , Adulto Jovem
12.
J Med Chem ; 59(2): 647-54, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26730548

RESUMO

Dextrin-colistin conjugates have been developed with the aim of achieving reduced clinical toxicity associated with colistin, also known as polymyxin E, and improved targeting to sites of bacterial infection. This study investigated the in vitro ability of such dextrin-colistin conjugates to bind and modulate bacterial lipopolysaccharide (LPS), and how this binding affects its biological activity. These results showed that colistin and amylase-activated dextrin-colistin conjugate to a lesser extent induced aggregation of LPS to form a stacked bilayer structure with characteristic dimensions, although this did not cause any substantial change in its secondary structure. In biological studies, both colistin and dextrin-colistin conjugate effectively inhibited LPS-induced hemolysis and tumor necrosis factor α (TNFα) secretion in a concentration-dependent manner, but only dextrin-colistin conjugate showed no additive toxicity at higher concentrations. This study provides the first direct structural experimental evidence for the binding of dextrin-colistin conjugates and LPS and gives insight into the mode of action of dextrin-colistin conjugates.


Assuntos
Antibacterianos/química , Bactérias/química , Colistina/química , Colistina/farmacologia , Dextrinas/química , Dextrinas/farmacologia , Lipopolissacarídeos/química , Amilases/metabolismo , Animais , Antibacterianos/farmacologia , Linhagem Celular , Endotoxinas/antagonistas & inibidores , Endotoxinas/química , Eritrócitos/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Humanos , Técnicas In Vitro , Teste do Limulus , Lipopolissacarídeos/antagonistas & inibidores , Ratos , Fator de Necrose Tumoral alfa/biossíntese
13.
J Biomater Appl ; 28(1): 144-60, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22457041

RESUMO

This study investigated the effects of combined titanium nano-/micron-scale roughness, induced by hydrogen peroxide pre-treatments, on bone marrow stromal cell responses and Porphyromonas gingivalis adherence in vitro. Untreated surfaces exhibited nano-scale features, while hydrogen peroxide treatments promoted increased nano-/micron-scale roughness. Bone marrow stromal cell attachment and proliferation were maintained with 6 h and 24 h treatments, but significantly decreased on 1-week and 4-week-treated surfaces. Bone marrow stromal cells on 6 h-4 week-treated titanium demonstrated enhanced osteogenic differentiation versus untreated surfaces. P. gingivalis adherence was significantly increased on 24 h-4 week surfaces. Results suggest that 6 h but less than 24 h treatments maintain or promote bone marrow stromal cell responses while minimizing microbial adherence, potentially enhancing titanium surface bio-activation for osseointegration.


Assuntos
Materiais Revestidos Biocompatíveis , Titânio , Animais , Aderência Bacteriana , Adesão Celular , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Implantes Dentários , Peróxido de Hidrogênio , Teste de Materiais , Células-Tronco Mesenquimais/citologia , Nanoestruturas , Osseointegração , Porphyromonas gingivalis/fisiologia , Ratos , Propriedades de Superfície
14.
Burns ; 39(1): 7-15, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22871554

RESUMO

Colistin is a venerable antibiotic whose fortunes have been revived by its excellent activity, the diminishing output of novel clinically effective antibiotics and the increasing importance of MDR infection in burn surgery, both in the civilian and military arenas. This review synthesizes current evidence on the usage of colistin in burn surgery including the structure-activity relationship; dosing, pharmacokinetics/pharmacodynamic (PK/PD), analytic methods, resistance and current research efforts into the redevelopment of this antibiotic, to distil recommendations for future research and clinical efficacy.


Assuntos
Antibacterianos/uso terapêutico , Queimaduras/tratamento farmacológico , Colistina/uso terapêutico , Infecções por Bactérias Gram-Negativas/tratamento farmacológico , Queimaduras/microbiologia , Colistina/química , Farmacorresistência Bacteriana Múltipla , Humanos
15.
J Antimicrob Chemother ; 68(2): 257-74, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23054997

RESUMO

Multidrug-resistant, Gram-negative infection is a major global determinant of morbidity, mortality and cost of care. The advent of nanomedicine has enabled tailored engineering of macromolecular constructs, permitting increasingly selective targeting, alteration of volume of distribution and activity/toxicity. Macromolecules tend to passively and preferentially accumulate at sites of enhanced vascular permeability and are then retained. This enhanced permeability and retention (EPR) effect, whilst recognized as a major breakthrough in anti-tumoral targeting, has not yet been fully exploited in infection. Shared pathophysiological pathways in both cancer and infection are evident and a number of novel nanomedicines have shown promise in selective, passive, size-mediated targeting to infection. This review describes the similarities and parallels in pathophysiological pathways at molecular, cellular and circulatory levels between inflammation/infection and cancer therapy, where use of this principle has been established.


Assuntos
Antibacterianos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Infecções por Bactérias Gram-Negativas/tratamento farmacológico , Nanomedicina/métodos , Permeabilidade , Antibacterianos/administração & dosagem , Humanos
16.
Int J Pharm ; 420(1): 84-92, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21884772

RESUMO

Hyaluronan (HA) has been extensively used for various medical applications, including osteoarthritis, tissue augmentation and ocular surgery. More recently, it has been investigated for use in polymer therapeutics as a carrier for drugs and biologically active proteins, thanks to its biodegradability, biocompatibility and inherent biological properties. Such biological functions are strongly dependent on HA's chain length, yet the molecular weight of HAs used in polymer conjugates varies widely and is inconsistent with its intended application. Therefore, this study aimed to determine the ideal chain length of HA to be used in polymer conjugates for enhanced tissue repair. HA fragments (M(w) 45,000-900,000g/mol) were prepared by acid hydrolysis of rooster comb HA and their physicochemical and biological properties were characterized. Such HA fragments had a highly extended, almost rod-like solution conformation and demonstrated chain length- and concentration-dependent viscosity, while exposure to HAase caused a rapid reduction in HA viscosity, which was most significant for the native HA. Initial HA hydrolysis rate by HAase varied strongly with HA chain length and was dependent on the formation of a stable enzyme-substrate complex. When normal human dermal fibroblasts were exposed to the different HA fragments for 72h, only native (900,000g/mol) HA reduced proliferation at 1000µg/mL. Conversely, only the smallest HA fragment (70,000g/mol) reduced the proliferation of chronic wound fibroblasts, at 1000µg/mL. The 70,000g/mol HA fragment also promoted the greatest cell attachment. These observations demonstrate that low molecular weight (70,000-120,000g/mol) HA fragments would be best suited for the delivery of proteins and peptides with applications in chronic wound healing and paves the way for the rationalized development of novel HA conjugates.


Assuntos
Fibroblastos/efeitos dos fármacos , Ácido Hialurônico/farmacologia , Fragmentos de Peptídeos/farmacologia , Cicatrização/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Química Farmacêutica , Cromatografia em Gel , Difusão , Relação Dose-Resposta a Droga , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/metabolismo , Hialuronoglucosaminidase/metabolismo , Hidrólise , Cinética , Espectroscopia de Ressonância Magnética , Peso Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Reologia , Espectroscopia de Infravermelho com Transformada de Fourier , Relação Estrutura-Atividade , Tecnologia Farmacêutica/métodos , Viscosidade
17.
J Control Release ; 152(3): 411-7, 2011 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-21435363

RESUMO

Chronic wounds, such as ulceration of the lower limb, represent a significant clinical challenge in today's ageing society. With the aim of identifying improved therapeutics, we have previously described a bioresponsive, dextrin-recombinant human epidermal growth factor conjugate (dextrin-rhEGF), that (i) protects rhEGF against proteolytic degradation by human chronic wound fluid; and (ii) mediates rhEGF release by α-amylase, capable of stimulating increased proliferation/migration in normal dermal and chronic wound fibroblasts; and keratinocytes, in vitro. The aim of this study was to extend these findings, by investigating the effects of dextrin-rhEGF on wound healing in the (db/db) diabetic mouse, a widely used in vivo model of delayed wound healing. Standardised, full-thickness excisional wounds, created in the dorsal flank skin, were treated topically with succinoylated dextrin (50 µg/mL), rhEGF (10 µg/mL) or dextrin-rhEGF (1 or 10 µg/mL). Treatments were applied immediately after injury and subsequently on post-wounding, days 3 and 8. Wound healing was assessed macroscopically, in terms of initiation of neo-dermal tissue deposition and wound closure (including wound contraction and re-epithelialisation), over a 16 day period. Wound healing was assessed histologically, in terms of granulation tissue formation/maturity; cranio-caudal wound contraction and wound angiogenesis (CD31 immuno-staining), using tissues harvested at day 16. Blood samples were also analysed for α-amylase and rhEGF concentrations. In this established impaired wound healing model, the topically-applied dextrin-rhEGF significantly accelerated wound closure and neo-dermal tissue formation at the macroscopic level; and significantly increased granulation tissue deposition and angiogenesis at the histological level (p<0.05), relative to untreated, succinoylated dextrin and rhEGF alone controls. Overall, these findings support the further development of bioresponsive polymer conjugates, for tissue repair.


Assuntos
Dextrinas/química , Complicações do Diabetes/tratamento farmacológico , Portadores de Fármacos/química , Fator de Crescimento Epidérmico/administração & dosagem , Fator de Crescimento Epidérmico/uso terapêutico , Proteínas Recombinantes/administração & dosagem , Cicatrização/efeitos dos fármacos , Animais , Complicações do Diabetes/patologia , Fator de Crescimento Epidérmico/sangue , Fator de Crescimento Epidérmico/química , Fator de Crescimento Epidérmico/farmacologia , Tecido de Granulação/anatomia & histologia , Tecido de Granulação/irrigação sanguínea , Tecido de Granulação/efeitos dos fármacos , Tecido de Granulação/crescimento & desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Mutantes , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Recombinantes/sangue , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Pele/efeitos dos fármacos , Pele/patologia , alfa-Amilases/sangue
18.
Int J Pharm ; 402(1-2): 95-102, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20888405

RESUMO

Bioresponsive polymers may effectively be utilized to enhance the circulation time and stability of biologically active proteins and peptides, while reducing their immunogenicity and toxicity. Recently, dextrin-epidermal growth factor (EGF) conjugates, which make use of the Polymer-masked UnMasked Protein Therapy (PUMPT) concept, have been developed and shown potential as modulators of impaired wound healing. This study investigated the potential of PUMPT using hyaluronic acid (HA) conjugates to mask activity and enhance protein stability, while allowing restoration of biological activity following triggered degradation. HA fragments (Mw ∼90,000g/mol), obtained by acid hydrolysis of Rooster comb HA, were conjugated to trypsin as a model enzyme or to EGF as a model growth factor. Conjugates contained 2.45 and 0.98% (w/w) trypsin or EGF, respectively, and contained <5% free protein. HA conjugation did not significantly alter trypsin's activity. However, incubation of the conjugate with physiological concentrations of HAase increased its activity to ∼145% (p<0.001) that of the free enzyme. In contrast, when HA-EGF conjugates were tested in vitro, no effect on cell proliferation was seen, even in the presence of HAase. HA conjugates did not display typical masking/unmasking behavior, HA-trypsin conjugates exhibited ∼52% greater stability in the presence of elastase, compared to free trypsin, demonstrating the potential of HA conjugates for further development as modulators of tissue repair.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Ácido Hialurônico/química , Polímeros/química , Tripsina/farmacologia , Animais , Bovinos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Galinhas , Crista e Barbelas , Fator de Crescimento Epidérmico/química , Humanos , Hialuronoglucosaminidase/metabolismo , Masculino , Elastase Pancreática/metabolismo , Estabilidade Proteica , Ovinos , Fatores de Tempo , Tripsina/química
19.
J Dermatol Sci ; 59(2): 115-22, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20558044

RESUMO

BACKGROUND: The molecular pathogenesis of chronic skin wounds is complex and not fully understood. Although these wounds are often characterized as being in a state of persistent inflammation, the impact and participation of the innate immune responses in sustaining this inflammation needs further investigation. OBJECTIVE: We investigated the cytokine profiles, Toll-like receptor (TLR)-stimulating activities and the levels of the antibacterial peptide Lipocalin-2 (Lcn-2) in a series of healing and non-healing chronic venous leg ulcers (CVLUs) through a study time of 8 weeks. METHODS: Wound fluids from healing and non-healing CVLUs were run on a Human Cytokine Antibody Array, and Lcn-2 levels measured with ELISA. HEK 293 cells transfected with TLR2 or TLR4 and their respective co-receptors, and human peripheral blood monocytes were then stimulated with the wound fluids from healing and non-healing venous leg ulcers. RESULTS: Healing wounds were associated with decreasing levels of IL-1alpha, IL-1beta and MIP-1delta, whereas in non-healing wounds decreasing levels of IL-8 and MIP-1alpha were found. Accordingly, wound fluid from non-healing CVLUs contained persistent Lcn-2 levels and TLR2- and TLR4-stimulating activities, while, in healing wounds, the TLR-stimulating activities decreased over time with significantly diminished levels of Lcn-2 (p<0.005). CONCLUSIONS: Innate immune responses contribute to the chronic inflammation in non-healing CVLUs through participation of Toll-like receptors. The levels of the antimicrobial peptide Lcn-2 in wound fluids from these ulcers are elevated as a reflection of this contribution.


Assuntos
Imunidade Inata/fisiologia , Úlcera Varicosa/fisiopatologia , Cicatrização/fisiologia , Proteínas de Fase Aguda/metabolismo , Células Cultivadas , Quimiocina CCL3/metabolismo , Doença Crônica , Humanos , Interleucina-8/metabolismo , Rim/citologia , Rim/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Lipocalina-2 , Lipocalinas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
20.
Stem Cells Dev ; 19(6): 819-30, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20132052

RESUMO

Wounds within the oral mucosa, similarly to fetal wounds, exhibit rapid healing with reduced scarring. We hypothesized that a progenitor population resident within the oral mucosal lamina propria (OMLP) contributes to this preferential healing. Progenitor cells (PC) were reliably isolated from the OMLP by differential adhesion to fibronectin. Isolated colonies originating from a single cell demonstrated a rapid initial phase of proliferation, completing in excess of 50 population doublings (PDs) before entering cellular senescence. These data were supported by the expression of active telomerase within both developing colonies and expanded clones as assessed by immunocytochemistry (ICC) and the quantitative telomeric repeat amplification protocol. FACS analysis confirmed expression of the stem cell markers CD44, CD90, CD105, and CD166, but negative expression of CD34 and CD45 ruling out a hematopoietic or fibrocyte origin for these progenitors. A neural crest origin was confirmed by increased colony-forming efficiency (CFE) in the presence of Jagged 1 and the expression of a number of neural crest markers within the developing colonies by ICC and serially passaged clones by Western blotting. The multipotency of this novel PC population was demonstrated by differentiation of the cells down both mesenchymal (chondrogenic, osteoblastic, and adipogenic) and neuronal (neuron and Schwann-like cells) cell lineages. This article reports for the first time, the isolation and characterization of a novel, clonally derived PC population resident within the OMLP. The attributes of this adult stem cell (ASC) population and its accessibility lends itself to future therapeutic applications.


Assuntos
Mucosa Bucal/citologia , Células-Tronco Multipotentes/citologia , Crista Neural/citologia , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Membrana Celular/metabolismo , Proliferação de Células , Separação Celular , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mucosa Bucal/enzimologia , Células-Tronco Multipotentes/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Telomerase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA