Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biomed Mater Eng ; 34(2): 143-157, 2023.
Article in English | MEDLINE | ID: mdl-35871316

ABSTRACT

BACKGROUND: Modulation of macrophage polarization is required for effective tissue repair and regenerative therapies. Therapeutic modulation of macrophages from an inflammatory M1 to a fibrotic M2 phenotype could help in diseases, such as chronic wounds, which are stalled in a prolonged and heightened inflammatory stage within the wound healing process. OBJECTIVE: This study evaluates the efficiency of a pullulan/gelatin nanofiber scaffold loaded with retinoic acid (RA) and adipose-derived mesenchymal stem cells (ASCs) to modulate M1 to M2 anti-inflammatory transition. METHODS: Scaffolds were fabricated by electrospinning, and crosslinked using ethylene glycol diglycidyl ether (EGDE). Exposure of RA and/or ASCs to cultured macrophages have been shown to promote M1 to M2 transition. Pullulan was chosen as a scaffold material due to its ability to quench reactive oxygen species, key signaling molecules that play an important role in the progression of inflammation, as well as for its excellent mechanical properties. Gelatin was chosen as an additional scaffold component due to the presence of cell-binding motifs and its biocompatibility. Scaffold compositions examined were 75:25 and 50:50, pullulan:gelatin. The scaffolds were crosslinked in 1:70 and 1:50 EGDE:EtOH. The scaffold composition was determined via FTIR. For the present study, the 75:25 pullulan:gelatin crosslinked with 1:70 EGDE:EtOH, forming nanofibers 328 ± 47.9 nm (mean ± SD) in diameter, was chosen as the scaffold composition due to its lower degradation and release rate, which allows a sustained delivery of RA. RESULTS: The scaffold composition degraded to approximately 80% after 14 days, with approximately 38% of the drug released after 7 days. THP-1 monocytic cells were induced into a M1 macrophage phenotype through stimulation with lipopolysaccharide (LPS) and gamma interferon (IFN-γ). These M1 macrophages were the exposed to scaffolds loaded with RA and ASCs, to induce differentiation to an M2 phenotype. CONCLUSION: Gene expression quantitation by qPCR showed a reduction of M1 biomarkers, tumor necrosis factor alpha (TNFα) and interleukin 1ß (IL1ß), and an increase of M2 biomarker CCL22 after 2 days of exposure, suggesting successful M1 to M2 transition.


Subject(s)
Mesenchymal Stem Cells , Tretinoin , Tretinoin/metabolism , Tretinoin/pharmacology , Gelatin , Macrophages/metabolism , Interferon-gamma/metabolism , Interferon-gamma/pharmacology
2.
PLoS One ; 14(2): e0213092, 2019.
Article in English | MEDLINE | ID: mdl-30807600

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0096681.].

3.
J Cyst Fibros ; 18(5): 622-629, 2019 09.
Article in English | MEDLINE | ID: mdl-30366849

ABSTRACT

BACKGROUND: There are no effective treatments for Burkholderia cenocepacia in patients with cystic fibrosis (CF) due to bacterial multi-drug resistance and defective host killing. We demonstrated that decreased bacterial killing in CF is caused by reduced macrophage autophagy due to defective cystic fibrosis transmembrane conductance regulator (CFTR) function. AR-12 is a small molecule autophagy inducer that kills intracellular pathogens such as Francisella. We evaluated the efficacy of AR-12 and a new analogue AR-13 in reducing bacterial burden in CF phagocytes. METHODS: Human CF and non-CF peripheral blood monocyte-derived macrophages, neutrophils, and nasal epithelial cells were exposed to CF bacterial strains in conjunction with treatment with antibiotics and/or AR compounds. RESULTS: AR-13 and not AR-12 had growth inhibition on B. cenocepacia and methicillin-resistantStaphylococcus aureus (MRSA) in media alone. There was a 99% reduction in MRSA in CF macrophages, 71% reduction in Pseudomonas aeruginosa in CF neutrophils, and 70% reduction in non-CF neutrophils using AR-13. Conversely, there was no reduction in B. cenocepacia in infected CF and non-CF macrophages using AR-13 alone, but AR-13 and antibiotics synergistically reduced B. cenocepacia in CF macrophages. AR-13 improved autophagy in CF macrophages and CF patient-derived epithelial cells, and increased CFTR protein expression and channel function in CF epithelial cells. CONCLUSIONS: The novel AR-12 analogue AR-13, in combination with antibiotics, reduced antibiotic-resistant bacterial burden in CF phagocytes, which correlated with increased autophagy and CFTR expression. AR-13 is a novel therapeutic for patients infected with B. cenocepacia and other resistant organisms that lack effective therapies.


Subject(s)
Bacterial Load/drug effects , Burkholderia cenocepacia/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis/pathology , Phagocytes/drug effects , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Autophagy/drug effects , Cell Culture Techniques , Cystic Fibrosis/microbiology , Drug Resistance, Bacterial , Humans
4.
PLoS One ; 12(10): e0186169, 2017.
Article in English | MEDLINE | ID: mdl-28982193

ABSTRACT

Members of the Burkholderia cepacia complex are virulent, multi-drug resistant pathogens that survive and replicate intracellularly in patients with cystic fibrosis (CF). We have discovered that B. cenocepacia cannot be cleared from CF macrophages due to defective autophagy, causing continued systemic inflammation and infection. Defective autophagy in CF is mediated through constitutive reactive oxygen species (ROS) activation of transglutaminase-2 (TG2), which causes the sequestration (accumulation) of essential autophagy initiating proteins. Cysteamine is a TG2 inhibitor and proteostasis regulator with the potential to restore autophagy. Therefore, we sought to examine the impact of cysteamine on CF macrophage autophagy and bacterial killing. Human peripheral blood monocyte-derived macrophages (MDMs) and alveolar macrophages were isolated from CF and non-CF donors. Macrophages were infected with clinical isolates of relevant CF pathogens. Cysteamine caused direct bacterial growth killing of live B. cenocepacia, B. multivorans, P. aeruginosa and MRSA in the absence of cells. Additionally, B. cenocepacia, B. multivorans, and P. aeruginosa invasion were significantly decreased in CF MDMs treated with cysteamine. Finally, cysteamine decreased TG2, p62, and beclin-1 accumulation in CF, leading to increased Burkholderia uptake into autophagosomes, increased macrophage CFTR expression, and decreased ROS and IL-1ß production. Cysteamine has direct anti-bacterial growth killing and improves human CF macrophage autophagy resulting in increased macrophage-mediated bacterial clearance, decreased inflammation, and reduced constitutive ROS production. Thus, cysteamine may be an effective adjunct to antibiotic regimens in CF.


Subject(s)
Bacteria/drug effects , Cysteamine/pharmacology , Cystic Fibrosis/microbiology , Drug Resistance, Microbial , Macrophages/microbiology , Autophagy , Bacteria/growth & development , Blotting, Western , Case-Control Studies , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Microscopy, Confocal , Microscopy, Electron, Transmission
5.
J Immunol ; 198(5): 1985-1994, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28093527

ABSTRACT

Macrophage intracellular pathogen killing is defective in cystic fibrosis (CF), despite abundant production of reactive oxygen species (ROS) in lung tissue. Burkholderia species can cause serious infection in CF and themselves affect key oxidase components in murine non-CF cells. However, it is unknown whether human CF macrophages have an independent defect in the oxidative burst and whether Burkholderia contributes to this defect in terms of assembly of the NADPH oxidase complex and subsequent ROS production. In this article, we analyze CF and non-CF human monocyte-derived macrophages (MDMs) for ROS production, NADPH assembly capacity, protein kinase C expression, and calcium release in response to PMA and CF pathogens. CF MDMs demonstrate a nearly 60% reduction in superoxide production after PMA stimulation compared with non-CF MDMs. Although CF MDMs generally have increased total NADPH component protein expression, they demonstrate decreased expression of the calcium-dependent protein kinase C conventional subclass α/ß leading to reduced phosphorylation of NADPH oxidase components p47 phox and p40 phox in comparison with non-CF MDMs. Ingestion of B. cenocepacia independently contributes to and worsens the overall oxidative burst deficits in CF MDMs compared with non-CF MDMs. Together, these results provide evidence for inherent deficits in the CF macrophage oxidative burst caused by decreased phosphorylation of NADPH oxidase cytosolic components that are augmented by Burkholderia These findings implicate a critical role for defective macrophage oxidative responses in persistent bacterial infections in CF and create new opportunities for boosting the macrophage immune response to limit infection.


Subject(s)
Burkholderia Infections/immunology , Burkholderia cenocepacia/immunology , Cystic Fibrosis/immunology , Macrophages/immunology , NADPH Oxidases/metabolism , Protein Kinase C/metabolism , Respiratory Burst , Animals , Calcium/metabolism , Cells, Cultured , Down-Regulation , Humans , Mice , Phosphorylation , Reactive Oxygen Species/metabolism
6.
BMC Infect Dis ; 16: 346, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27449800

ABSTRACT

BACKGROUND: Acinetobacter species are associated with increasing mortality due to emerging drug-resistance. Pediatric Acinetobacter infections are largely undefined in developed countries and clinical laboratory identification methods do not reliably differentiate between members of the Acinetobacter calcoaceticus-baumannii complex, leading to improper identification. Therefore we aimed to determine risk factors for invasive Acinetobacter infections within an academic, pediatric setting as well as defining microbiologic characteristics of predominant strains. METHODS: Twenty-four invasive Acinetobacter isolates were collected from 2009-2013. Comparative sequence analysis of the rpoB gene was performed coupled with phenotypic characterization of antibiotic resistance, motility, biofilm production and clinical correlation. RESULTS: Affected patients had a median age of 3.5 years, and 71 % had a central catheter infection source. rpoB gene sequencing revealed a predominance of A. pittii (45.8 %) and A. baumannii (33.3 %) strains. There was increasing incidence of A. pittii over the study. Two fatalities occurred in the A. pittii group. Seventeen percent of isolates were multi-drug resistant. A pittii and A. baumannii strains were similar in motility, but A pittii strains had significantly more biofilm production (P value = 0.018). CONCLUSIONS: A. pittii was the most isolated species highlighting the need for proper species identification. The isolated strains had limited acute mortality in children, but the occurrence of more multi-drug resistant strains in the future is a distinct possibility, justifying continued research and accurate species identification.


Subject(s)
Acinetobacter Infections/microbiology , Acinetobacter/isolation & purification , Cross Infection/microbiology , Academic Medical Centers , Acinetobacter/drug effects , Acinetobacter/genetics , Acinetobacter/physiology , Acinetobacter Infections/diagnosis , Acinetobacter Infections/drug therapy , Acinetobacter Infections/etiology , Adolescent , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms , Child , Child, Preschool , Cross Infection/diagnosis , Cross Infection/drug therapy , Cross Infection/etiology , Drug Resistance, Multiple, Bacterial , Female , Hospitals, Pediatric , Humans , Infant , Infant, Newborn , Male , Retrospective Studies , Risk Factors , Young Adult
7.
Epigenetics ; 11(5): 381-8, 2016 05 03.
Article in English | MEDLINE | ID: mdl-26909551

ABSTRACT

Autophagy is a biological process characterized by self-digestion and involves induction of autophagosome formation, leading to degradation of autophagic cargo. Aging is associated with the reduction of autophagy activity leading to neurodegenerative disorders, chronic inflammation, and susceptibility to infection; however, the underlying mechanism is unclear. DNA methylation by DNA methyltransferases reduces the expression of corresponding genes. Since macrophages are major players in inflammation and defense against infection we determined the differences in methylation of autophagy genes in macrophages derived from young and aged mice. We found that promoter regions of Atg5 and LC3B are hypermethylated in macrophages from aged mice and this is accompanied by low gene expression. Treatment of aged mice and their derived macrophages with methyltransferase inhibitor (2)-epigallocatechin-3-gallate (EGCG) or specific DNA methyltransferase 2 (DNMT2) siRNA restored the expression of Atg5 and LC3 in vivo and in vitro. Our study builds a foundation for the development of novel therapeutics aimed to improve autophagy in the elderly population and suggests a role for DNMT2 in DNA methylation activities.


Subject(s)
Aging/genetics , Autophagy-Related Protein 5/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Microtubule-Associated Proteins/genetics , Aging/pathology , Animals , Autophagosomes/drug effects , Autophagy/drug effects , Autophagy/genetics , Catechin/administration & dosage , Catechin/analogs & derivatives , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , Enzyme Inhibitors/administration & dosage , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice , RNA, Small Interfering/genetics
8.
Autophagy ; 11(9): 1471-83, 2015.
Article in English | MEDLINE | ID: mdl-26213203

ABSTRACT

Autophagy describes an intracellular process responsible for the lysosome-dependent degradation of cytosolic components. The ULK1/2 complex comprising the kinase ULK1/2 and the accessory proteins ATG13, RB1CC1, and ATG101 has been identified as a central player in the autophagy network, and it represents the main entry point for autophagy-regulating kinases such as MTOR and AMPK. It is generally accepted that the ULK1 complex is constitutively assembled independent of nutrient supply. Here we report the characterization of the ATG13 region required for the binding of ULK1/2. This binding site is established by an extremely short peptide motif at the C terminus of ATG13. This motif is mandatory for the recruitment of ULK1 into the autophagy-initiating high-molecular mass complex. Expression of a ULK1/2 binding-deficient ATG13 variant in ATG13-deficient cells resulted in diminished but not completely abolished autophagic activity. Collectively, we propose that autophagy can be executed by mechanisms that are dependent or independent of the ULK1/2-ATG13 interaction.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Autophagy , Mutation , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Animals , Apoptosis Regulatory Proteins , Autophagy-Related Protein-1 Homolog , Enzyme Stability , Fibroblasts/metabolism , Heat-Shock Proteins/metabolism , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Peptides/metabolism , Phagosomes/metabolism , Protein Binding , Proteolysis , Sequestosome-1 Protein
9.
PLoS One ; 9(5): e96681, 2014.
Article in English | MEDLINE | ID: mdl-24798083

ABSTRACT

Burkholderia cenocepacia is a virulent pathogen that causes significant morbidity and mortality in patients with cystic fibrosis (CF), survives intracellularly in macrophages, and uniquely causes systemic infections in CF. Autophagy is a physiologic process that involves engulfing non-functional organelles and proteins and delivering them for lysosomal degradation, but also plays a role in eliminating intracellular pathogens, including B. cenocepacia. Autophagy is defective in CF but can be stimulated in murine CF models leading to increased clearance of B. cenocepacia, but little is known about autophagy stimulation in human CF macrophages. IFN-γ activates macrophages and increases antigen presentation while also inducing autophagy in macrophages. We therefore, hypothesized that treatment with IFN-γ would increase autophagy and macrophage activation in patients with CF. Peripheral blood monocyte derived macrophages (MDMs) were obtained from CF and non-CF donors and subsequently infected with B. cenocepacia. Basal serum levels of IFN-γ were similar between CF and non-CF patients, however after B. cenocepacia infection there is deficient IFN-γ production in CF MDMs. IFN-γ treated CF MDMs demonstrate increased co-localization with the autophagy molecule p62, increased autophagosome formation, and increased trafficking to lysosomes compared to untreated CF MDMs. Electron microscopy confirmed IFN-γ promotes double membrane vacuole formation around bacteria in CF MDMs, while only single membrane vacuoles form in untreated CF cells. Bacterial burden is significantly reduced in autophagy stimulated CF MDMs, comparable to non-CF levels. IL-1ß production is decreased in CF MDMs after IFN-γ treatment. Together, these results demonstrate that IFN-γ promotes autophagy-mediated clearance of B. cenocepacia in human CF macrophages.


Subject(s)
Autophagy , Burkholderia Infections/immunology , Burkholderia cenocepacia , Cystic Fibrosis/immunology , Interferon-gamma/immunology , Macrophages/immunology , Adolescent , Adult , Azithromycin/chemistry , Child , Cystic Fibrosis/microbiology , Female , Humans , Interleukin-1beta/immunology , Lysosomes/metabolism , Macrophages/microbiology , Male , Microscopy, Confocal , Monocytes/immunology , Monocytes/microbiology , Phagosomes , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...