Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Int J Pharm ; 650: 123696, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38086494

RESUMEN

Immunological safety of nanofibers remains poorly reported within the scientific literature and lacks specific in vitro testing models distinct from those used to test nanoparticles. To address the challenges of currently used conventional setups being described in the literature, we developed a novel in vitro model for nanofiber mats immunogenicity testing, which enables standardization of tested surface area, excludes nanofiber mat edges, and ensures stable contacts of cells with nanofibers during the experiment. The effect of nanofibers was assessed on peripheral blood mononuclear cells (PBMCs) by measuring their metabolic activity using MTS cell proliferation assay, where key performance parameters, i.e. cell number, phytohemagglutinin-L (PHA-L) concentration, incubation time and cell lysis were optimized. Repeatability of results obtained with non-activated and PHA-L-activated PBMCs in contact with differently thick polycaprolactone nanofiber mats was compared using both models. Our model provided more reproducible results with lower variability, exhibiting its higher reliability and accuracy than the conventional one. Furthermore, results showed the presence of thicker mats resulted in reduced metabolic activity and PBMC proliferation without any observed cytotoxicity, providing additional insights into their non-immunogenic characteristics. The developed model enables more accurate biological assessment that can support new guidelines for in vitro nanofiber testing and formulation.


Asunto(s)
Nanofibras , Nanopartículas , Leucocitos Mononucleares , Reproducibilidad de los Resultados
2.
Pharmaceutics ; 15(4)2023 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-37111731

RESUMEN

Conventional treatments for chronic wounds are often ineffective, thus new therapeutic approaches are needed, such as the delivery of immunomodulatory drugs that can reduce inflammation, restore immune cell function, and facilitate tissue regeneration. A potential drug for such an approach is simvastatin, which has major drawbacks including poor solubility and chemical instability. With the aim of developing a dressing for wound healing, simvastatin and an antioxidant were incorporated into alginate/poly(ethylene oxide) nanofibers by green electrospinning without the use of organic solvents, thanks to their prior encapsulation into liposomes. The composite liposome-nanofiber formulations exhibited fibrillar morphology (160-312 nm) and unprecedentedly high phospholipid and drug content (76%). Transmission electron microscopy revealed dried liposomes as bright ellipsoidal spots homogeneously distributed over the nanofibers. After nanofiber hydration, the liposomes reconstituted in two size populations (~140 and ~435 nm), as revealed by cutting-edge MADLS® analysis. Lastly, in vitro assays demonstrated that composite liposome-nanofiber formulations are superior to liposomal formulations due to a better safety profile in keratinocytes and peripheral blood mononuclear cells. Furthermore, both formulations exhibited similarly advantageous immunomodulatory effects, measured as decreased inflammation in vitro. A synergistic combination of the two nanodelivery systems shows promise for the development of efficient dressings for chronic wound treatment.

3.
J Control Release ; 355: 371-384, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36738969

RESUMEN

Biofilm-associated diseases such as periodontitis are widespread and challenging to treat which calls for new strategies for their effective management. Probiotics represent a promising approach for targeted treatment of dysbiosis in biofilm and modulation of host immune response. In this interdisciplinary study, nanofibers with two autochthonous Bacillus strains 27.3.Z and 25.2.M were developed. The strains were isolated from the oral microbiota of healthy individuals, and their genomes were sequenced and screened for genes associated with antimicrobial and immunomodulatory activities, virulence factors, and transferability of resistance to antibiotics. Spores of two Bacillus strains were incorporated individually or in combination into hydrophilic poly(ethylene oxide) (PEO) and composite PEO/alginate nanofibers. The nanofiber mats were characterised by a high loading of viable spores (> 7 log CFU/mg) and they maintained viability during electrospinning and 6 months of storage at room temperature. Spores were rapidly released from PEO nanofibers, while presence of alginate in the nanofibers prolonged their release. All formulations exhibited swelling, followed by transformation of the nanofiber mat into a hydrogel and polymer erosion mediating spore release kinetics. The investigated Bacillus strains released metabolites, which were not cytotoxic to peripheral blood mononuclear cells (PBMCs) in vitro. Moreover, their metabolites exhibited antibacterial activity against two periodontopathogens, an antiproliferative effect on PBMCs, and inhibition of PBMC expression of proinflammatory cytokines. In summary, the developed nanofiber-based delivery system represents a promising therapeutic approach to combat biofilm-associated disease on two fronts, namely via modulation of the local microbiota with probiotic bacteria and host immune response with their metabolites.


Asunto(s)
Bacillus , Nanofibras , Humanos , Leucocitos Mononucleares , Bacillus/genética , Antibacterianos/farmacología , Polietilenglicoles , Alginatos
4.
Life (Basel) ; 12(6)2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35743928

RESUMEN

Human skeletal stem cells (hSSCs) were recently identified as podoplanin (PDPN)/CD73/CD164-positive and CD146-negative cells that decline with age, and play a role in the pathogenesis of osteoarthritis (OA). The aim of this study was to identify the hSSC-like properties of bone-derived mesenchymal stem/stromal cells (MSCs) of patients with late and early OA. Methods: First, we performed gene expression profiling for the hSSC markers in 32 patients with late and early OA, and donors without OA. Having identified the low expression of hSSC markers in late OA patients, we further performed trilineage differentiation and immunophenotyping for hSSC makers in the selected subsets from each donor group. Results: Our results show no differences in osteogenesis, chondrogenesis, and adipogenesis between the MSCs from the three groups. However, the immunophenotyping shows lower CD164 in MSCs from early OA patients in comparison with late and no OA subjects (p = 0.002 and p = 0.017). Conclusions: Our study shows that the in vitro hSSC-like properties of bone-derived MSCs are similar in patients with early and late OA, and in donors without OA. However, the lower percentage of CD164-positive MSCs in early OA patients indicates the potential of CD164 as a marker of the onset of OA.

6.
Stem Cell Rev Rep ; 14(3): 346-369, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29556896

RESUMEN

The musculoskeletal system includes tissues that have remarkable regenerative capabilities. Bone and muscle sustain micro-damage throughout the lifetime, yet they continue to provide the body with the support that is needed for everyday activities. Our current understanding is that the regenerative capacity of the musculoskeletal system can be attributed to the mesenchymal stem/ stromal cells (MSCs) that reside within its different anatomical compartments. These MSCs can replenish various tissues with progenitor cells to form functional cells, such as osteoblasts, chondrocytes, myocytes, and others. However, with aging and in certain disorders of the musculoskeletal system such as osteoarthritis or osteoporosis, this regenerative capacity of MSCs appears to be lost or diverted for the production of other non-functional cell types, such as adipocytes and fibroblasts. In this review, we shed light on the tissue sources and subpopulations of MSCs in the musculoskeletal system that have been identified in animal models, discuss the mechanisms of their anti-inflammatory action as a prerequisite for their tissue regeneration and their current applications in regenerative medicine. While providing up-to-date evidence of the role of MSCs in different musculoskeletal pathologies, in particular in osteoporosis and osteoarthritis, we share some thoughts on their potential as diagnostic markers in musculoskeletal health and disease.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Sistema Musculoesquelético/citología , Sistema Musculoesquelético/metabolismo , Medicina Regenerativa/métodos , Condrocitos/citología , Condrocitos/metabolismo , Humanos , Trasplante de Células Madre Mesenquimatosas , Membrana Sinovial/citología , Membrana Sinovial/metabolismo , Ingeniería de Tejidos/métodos
7.
Biotechnol Prog ; 34(4): 1045-1058, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29536646

RESUMEN

Inflammation plays a major role in progression of rheumatoid arthritis, a disease treated with antagonists of tumor necrosis factor-alpha (TNF-α) and interleukin 1ß (IL-1ß). New in vitro testing systems are needed to evaluate efficacies of new anti-inflammatory biological drugs, ideally in a patient-specific manner. To address this need, we studied microspheroids containing 10,000 human osteoarthritic primary chondrocytes (OACs) or chondrogenically differentiated mesenchymal stem cells (MSCs), obtained from three donors. Hypothesizing that this system can recapitulate clinically observed effects of anti-inflammatory drugs, spheroids were exposed to TNF-α, IL-1ß, or to supernatant containing secretome from activated macrophages (MCM). The anti-inflammatory efficacies of anti-TNF-α biologicals adalimumab, infliximab, and etanercept, and the anti-IL-1ß agent anakinra were assessed in short-term microspheroid and long-term macrospheroid cultures (100,000 OACs). While gene and protein expressions were evaluated in microspheroids, diameters, amounts of DNA, glycosaminoglycans, and hydroxiproline were measured in macrospheroids. The tested drugs significantly decreased the inflammation induced by TNF-α or IL-1ß. The differences in potency of anti-TNF-α biologicals at 24 h and 3 weeks after their addition to inflamed spheroids were comparable, showing high predictability of short-term cultures. Moreover, the data obtained with microspheroids grown from OACs and chondrogenically differentiated MSCs were comparable, suggesting that MSCs could be used for this type of in vitro testing. We propose that in vitro gene expression measured after the first 24 h in cultures of chondrogenically differentiated MSCs can be used to determine the functionality of anti-TNF-α drugs in personalized and preclinical studies. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1045-1058, 2018.


Asunto(s)
Condrocitos/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Células Madre Mesenquimatosas/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Diferenciación Celular/fisiología , Células Cultivadas , Condrocitos/citología , Condrogénesis/fisiología , Humanos , Interleucina-1beta/inmunología , Células Madre Mesenquimatosas/citología , Factor de Necrosis Tumoral alfa/inmunología
8.
Biologicals ; 45: 96-101, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27773423

RESUMEN

In vitro cell-based models are important tools for assessing efficacies of new leads in early phases of drug development. Human osteoarthritic chondrocytes (OACs), obtained from biomedical waste material, represent a valuable, relatively accessible cellular source that could be used for this purpose. By employing reverse transcription-polymerase chain reaction (qRT-PCR) we compared gene expression profiles of key anabolic, catabolic and inflammatory genes of freshly isolated vs. monolayer cultured OACs (passages P0-P2) and non-stimulated vs. tumor necrosis factor alpha (TNF-α) stimulated P2 OACs. After expansion of OACs in monolayer cultures, the expression of almost all analyzed genes significantly decreased. The subsequent addition of TNF-α to OACs at P2 significantly increased expressions of all catabolic and inflammatory genes, leaving the anabolic profile almost unchanged. TNF-α-treated OACs were later utilized for efficacy testing of anti-TNF-α drugs infliximab and etanercept and both significantly reduced the expressions of all catabolic and inflammatory genes tested.


Asunto(s)
Condrocitos/metabolismo , Etanercept , Infliximab , Osteoartritis de la Rodilla/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Anciano , Evaluación Preclínica de Medicamentos/métodos , Etanercept/química , Etanercept/farmacología , Femenino , Humanos , Infliximab/química , Infliximab/farmacología , Masculino , Persona de Mediana Edad
9.
Int Immunopharmacol ; 34: 146-154, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26945833

RESUMEN

Bisphenols (BPs) are widely spread pollutants that act as estrogen-like endocrine disruptors and are potentially affecting human health on a long run. We explored the effects of BPA, BPF and BPAF, on in vitro differentiation and maturation of MDDCs. Monocytes were treated with 17ß-estradiol (E2) and each BP at the beginning of their differentiation into iMDDCs. We found that 10 and 50 µM of BPA and BPF, 10 and 30µM of BPAF and 10 and 50 nM of E2 did not affect cell viability. However, 50 µM of BPA and BPF, as well as 10 and 30 µM of BPAF, significantly decreased the endocytotic capacity of iMDDCs. Both, BPA (50 µM) and BPAF (30 µM) decreased the expression of CD1a and increased the amount of DC-SIGN molecules on iMDDCs. The E2 pre-treatment moderately decreased expression of CD80, CD86 and CD83 co-stimulatory molecules while increasing the numbers of HLA-DR on mMDDCs. Only BPAF significantly influenced the expression of CD80 and CD86 (both decreased), as well as CD83 and HLA-DR molecules (both increased) on mMDDCs. In addition, BPAF modulated DC maturation signaling pathways by lowering the phosphorylation of p65 NF-κB (nuclear factor-kappaB) and ERK (extracellular signal regulated kinase) 1/2 proteins. Consequently, the in vitro proliferation of allogeneic T cells, stimulated with differently pre-treated iMDDCs and mMDDCs, was significantly reduced only in case of BPAF.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Células Dendríticas/efectos de los fármacos , Estradiol/farmacología , Monocitos/efectos de los fármacos , Fenoles/farmacología , Linfocitos T/inmunología , Animales , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Células Dendríticas/fisiología , Endocitosis/efectos de los fármacos , Humanos , Lectinas Tipo C/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Monocitos/fisiología , FN-kappa B/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos
10.
PLoS One ; 10(5): e0125791, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25950799

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is among the most aggressive cancers with a poor prognosis in spite of a plethora of established diagnostic and prognostic biomarkers and treatment modalities. Therefore, the current goal is the detection of novel biomarkers, possibly detectable in the blood of GBM patients that may enable an early diagnosis and are potential therapeutic targets, leading to more efficient interventions. EXPERIMENTAL PROCEDURES: MicroRNA profiling of 734 human and human-associated viral miRNAs was performed on blood plasma samples from 16 healthy individuals and 16 patients with GBM, using the nCounter miRNA Expression Assay Kits. RESULTS: We identified 19 miRNAs with significantly different plasma levels in GBM patients, compared to the healthy individuals group with the difference limited by a factor of 2. Additionally, 11 viral miRNAs were found differentially expressed in plasma of GBM patients and 24 miRNA levels significantly correlated with the patients' survival. Moreover, the overlap between the group of candidate miRNAs for diagnostic biomarkers and the group of miRNAs associated with survival, consisted of ten miRNAs, showing both diagnostic and prognostic potential. Among them, hsa miR 592 and hsa miR 514a 3p have not been previously described in GBM and represent novel candidates for selective biomarkers. The possible signalling, induced by the revealed miRNAs is discussed, including those of viral origin, and in particular those related to the impaired immune response in the progression of GBM. CONCLUSION: The GBM burden is reflected in the alteration of the plasma miRNAs pattern, including viral miRNAs, representing the potential for future clinical application. Therefore proposed biomarker candidate miRNAs should be validated in a larger study of an independent cohort of patients.


Asunto(s)
Neoplasias Encefálicas/sangre , Glioblastoma/sangre , MicroARNs/genética , Análisis de Supervivencia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/virología , Estudios de Casos y Controles , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/virología , Humanos , MicroARNs/sangre , Pronóstico
11.
Eur J Pharm Biopharm ; 88(1): 64-74, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24931341

RESUMEN

The aim of our work was to produce a modern nanomaterial with incorporated blood-derived growth factors, produced by electrospinning, applicable in treatment of chronic wounds. Platelet-rich plasma was chosen as a natural source of growth factors. Results showed that platelet-rich plasma stimulates keratinocyte and fibroblast cell growth in vitro. Its optimal concentration in growth medium was 2% (v/v) for both types of skin cells, while higher concentrations caused alterations in cell morphology, with reduced cell mobility and proliferation. In the next step hydrophilic nanofibers loaded with platelet-rich plasma were produced from chitosan and poly(ethylene oxide), using electrospinning. The morphology of nanofibers was stable in aqueous conditions for 72 h. It was shown that electrospinning does not adversely affect the biological activity of platelet-rich plasma. The effects of nanofibers with incorporated platelet-rich plasma on cell proliferation, survival, morphology and mobility were examined. Nanofibers limited cell mobility, changed morphology and stimulated cell proliferation. Despite of the small amount of blood-derived growth factors introduced in cell culture via platelet-rich plasma-loaded nanofibers, such nanofibrillar support significantly induced cell proliferation, indicating synergistic effect of nanotopography and incorporated growth factors. The overall results confirm favorable in vitro properties of produced nanofibers, indicating their high potential as a nanomaterial suitable for delivery of platelet-rich plasma in wound healing applications.


Asunto(s)
Administración Cutánea , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Nanofibras/química , Plasma Rico en Plaquetas/química , Piel/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Quitosano/administración & dosificación , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Microscopía Electrónica de Rastreo , Nanotecnología , Polietilenglicoles/administración & dosificación , Polímeros/química
12.
PLoS One ; 9(3): e92969, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24667745

RESUMEN

OBJECTIVES: Bone tissue regeneration requires a source of viable, proliferative cells with osteogenic differentiation capacity. Periodontal surgeries represent an opportunity to procure small amounts of autologous tissues for primary cell isolation. Our objective was to assess the potential of human alveolar bone as a source of autologous osteogenic cells for tissue engineering and biomaterials and drug testing studies. MATERIALS AND METHODS: Alveolar bone tissue was obtained from 37 patients undergoing routine periodontal surgery. Tissue harvesting and cell isolation procedures were optimized to isolate viable cells. Primary cells were subcultured and characterized with respect to their growth characteristics, gene expression of osteogenic markers, alkaline phosphatase activity and matrix mineralization, under osteogenic stimulation. RESULTS: Alveolar bone cells were successfully isolated from 28 of the 30 samples harvested with bone forceps, and from 2 of the 5 samples obtained by bone drilling. The yield of cells in primary cultures was variable between the individual samples, but was not related to the site of tissue harvesting and the patient age. In 80% of samples (n = 5), the primary cells proliferated steadily for eight subsequent passages, reaching cumulative numbers over 10(10) cells. Analyses confirmed stable gene expression of alkaline phosphatase, osteopontin and osteocalcin in early and late cell passages. In osteogenic medium, the cells from late passages increased alkaline phosphatase activity and accumulated mineralized matrix, indicating a mature osteoblastic phenotype. CONCLUSIONS: Primary alveolar bone cells exhibited robust proliferation and retained osteogenic phenotype during in vitro expansion, suggesting that they can be used as an autologous cell source for bone regenerative therapies and various in vitro studies.


Asunto(s)
Proceso Alveolar/citología , Proceso Alveolar/fisiología , Procedimientos Quirúrgicos Orales , Osteogénesis , Fenotipo , Adulto , Anciano , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Separación Celular , Femenino , Humanos , Masculino , Persona de Mediana Edad , Regeneración , Ingeniería de Tejidos , Adulto Joven
13.
J Leukoc Biol ; 95(1): 33-46, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23924658

RESUMEN

Lately, there is increasing evidence that emphasizes the regulatory functions of IFN-γ, which serve as negative-feedback mechanisms after, e.g., pathogen clearance, to prevent unnecessary tissue destruction. Inflammatory processes involving Th1 and cytotoxic responses are characterized by high, local IFN-γ concentrations, followed by resolution and immune silencing. Although this is a well-known course of events, extensive attempts to address potential differential effects of IFN-γ in the manner of its availability (quantitatively) in the environment do not exist. We demonstrate that high doses of IFN-γ do not induce DC maturation and activation but instead, induce specific regulatory characteristics in DCs. Considering their phenotype, high doses of IFN-γ extensively induce the expression of ILT-4 and HLA-G inhibitory molecules. Interestingly, the well-known priming effect of IFN-γ for IL-12p70 production is lost at these conditions, and the DC cytokine profile is switched toward an increased IL-10/IL-12p70 ratio upon subsequent stimulation with CD40L. Furthermore, such DCs are capable of silencing cellular immune responses and activation of cytotoxic CD8+ T lymphocytes, resulting in reduced cell proliferation and down-regulation of granzyme B expression. Additionally, we find that in this manner, immune regulation mediated by IFN-γ is not mainly a result of increased enzymatic activity of IDO in DCs but rather, a result of HLA-G signaling, which can be reversed by blocking mAb. Altogether, our results identify a novel mechanism by which a Th1-like environment programs the functional status of DCs to silence ongoing cytotoxic responses to prevent unwanted tissue destruction and inflammation.


Asunto(s)
Citotoxicidad Inmunológica/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Interferón gamma/farmacología , Antígenos CD/metabolismo , Comunicación Autocrina , Citocinas/biosíntesis , Células Dendríticas/metabolismo , Antígenos HLA-G/metabolismo , Humanos , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/metabolismo , Fenotipo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células TH1/inmunología , Células TH1/metabolismo
14.
Biochem Med (Zagreb) ; 23(1): 43-63, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23457765

RESUMEN

Bone and immune system are functionally interconnected. Immune and bone cells derive from same progenitors in the bone marrow, they share a common microenvironment and are being influenced by similar mediators. The evidence on increased bone resorption associated with inappropriate activation of T cells such as during inflammation, is well established. However, the molecular mechanisms beyond this clinical observation have begun to be intensively studied with the advancement of osteoimmunology. Now days, we have firm evidence on the influence of numerous proinflammatory cytokines on bone cells, with the majority of data focused on osteoclasts, the bone resorbing cells. It has been shown that some proinflammatory cytokines could possess osteoclastogenic and/or anti-osteoclastogenic properties and can target osteoclasts directly or via receptor activator of nuclear factor kappaB (RANK)/RANK ligand(RANKL)/osteoprotegerin (OPG) system. Several studies have reported opposing data regarding (anti)osteoclastogenic properties of these cytokines. Therefore, the first part of this review is summarizing current evidence on the influence of pro-inflammatory cytokines on osteoclasts and thus on bone resorption. In the second part, the evidence on the role of pro-inflammatory cytokines in osteoporosis and osteoarthritis is reviewed to show that unravelling the mechanisms beyond such complex bone diseases, is almost impossible without considering skeletal and immune systems as an indivisible integrated system.


Asunto(s)
Enfermedades Óseas/inmunología , Citocinas/farmacología , Sistema Inmunológico/inmunología , Mediadores de Inflamación/farmacología , Osteoclastos/efectos de los fármacos , Enfermedades Óseas/metabolismo , Enfermedades Óseas/patología , Humanos , Osteoclastos/metabolismo
15.
J Neuroinflammation ; 9: 144, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22734718

RESUMEN

BACKGROUND: In immune-mediated diseases of the central nervous system, astrocytes exposed to interferon-γ (IFN-γ) can express major histocompatibility complex (MHC) class II molecules and antigens on their surface. MHC class II molecules are thought to be delivered to the cell surface by membrane-bound vesicles. However, the characteristics and dynamics of this vesicular traffic are unclear, particularly in reactive astrocytes, which overexpress intermediate filament (IF) proteins that may affect trafficking. The aim of this study was to determine the mobility of MHC class II vesicles in wild-type (WT) astrocytes and in astrocytes devoid of IFs. METHODS: The identity of MHC class II compartments in WT and IF-deficient astrocytes 48 h after IFN-γ activation was determined immunocytochemically by using confocal microscopy. Time-lapse confocal imaging and Alexa Fluor546-dextran labeling of late endosomes/lysosomes in IFN-γ treated cells was used to characterize the motion of MHC class II vesicles. The mobility of vesicles was analyzed using ParticleTR software. RESULTS: Confocal imaging of primary cultures of WT and IF-deficient astrocytes revealed IFN-γ induced MHC class II expression in late endosomes/lysosomes, which were specifically labeled with Alexa Fluor546-conjugated dextran. Live imaging revealed faster movement of dextran-positive vesicles in IFN-γ-treated than in untreated astrocytes. Vesicle mobility was lower in IFN-γ-treated IF-deficient astrocytes than in WT astrocytes. Thus, the IFN-γ-induced increase in the mobility of MHC class II compartments is IF-dependent. CONCLUSIONS: Since reactivity of astrocytes is a hallmark of many CNS pathologies, it is likely that the up-regulation of IFs under such conditions allows a faster and therefore a more efficient delivery of MHC class II molecules to the cell surface. In vivo, such regulatory mechanisms may enable antigen-presenting reactive astrocytes to respond rapidly and in a controlled manner to CNS inflammation.


Asunto(s)
Astrocitos/metabolismo , Compartimento Celular/fisiología , Antígenos de Histocompatibilidad Clase II/metabolismo , Interferón gamma/fisiología , Proteínas de Filamentos Intermediarios/fisiología , Animales , Células Cultivadas , Interferón gamma/genética , Proteínas de Filamentos Intermediarios/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
16.
Int Rev Immunol ; 31(3): 202-22, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22587021

RESUMEN

A molecule with a relatively simple chemical structure, resveratrol has been found to interact with multiple molecular targets, many of them associated with inflammation and immunity. Indeed, it has been shown to act directly on central players of both innate and adaptive immunity, such as macrophages, lymphocytes, and dendritic cells. In addition, there is very little evidence suggesting significant deleterious side effects of resveratrol, further highlighting its potential future use as a therapeutic agent. This review provides an up-to-date discussion on recent advances regarding anti-inflammatory effects of resveratrol, mechanisms of action, and its potential for therapeutic use.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Sistema Inmunológico/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Estilbenos/farmacología , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/patología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Modelos Inmunológicos , Resveratrol , Transducción de Señal/inmunología , Estilbenos/uso terapéutico
17.
PLoS One ; 7(1): e28761, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253695

RESUMEN

BACKGROUND: Different immunotherapy approaches for the treatment of cancer and autoimmune diseases are being developed and tested in clinical studies worldwide. Their resulting complex experimental data should be properly evaluated, therefore reliable normal healthy control baseline values are indispensable. METHODOLOGY/PRINCIPAL FINDINGS: To assess intra- and inter-individual variability of various biomarkers, peripheral blood of 16 age and gender equilibrated healthy volunteers was sampled on 3 different days within a period of one month. Complex "crossomics" analyses of plasma metabolite profiles, antibody concentrations and lymphocyte subset counts as well as whole genome expression profiling in CD4+T and NK cells were performed. Some of the observed age, gender and BMI dependences are in agreement with the existing knowledge, like negative correlation between sex hormone levels and age or BMI related increase in lipids and soluble sugars. Thus we can assume that the distribution of all 39.743 analysed markers is well representing the normal Caucasoid population. All lymphocyte subsets, 20% of metabolites and less than 10% of genes, were identified as highly variable in our dataset. CONCLUSIONS/SIGNIFICANCE: Our study shows that the intra-individual variability was at least two-fold lower compared to the inter-individual one at all investigated levels, showing the importance of personalised medicine approach from yet another perspective.


Asunto(s)
Técnicas Citológicas/métodos , Salud , Subgrupos Linfocitarios/metabolismo , Población Blanca , Adulto , Anticuerpos/inmunología , Índice de Masa Corporal , Linfocitos T CD4-Positivos/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Células Asesinas Naturales/metabolismo , Masculino , Persona de Mediana Edad , Variaciones Dependientes del Observador , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Componente Principal , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Adulto Joven
18.
Immunology ; 134(4): 409-18, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22044067

RESUMEN

A novel lectin, isolated from the basidiomycete mushroom Clitocybe nebularis and termed C. nebularis lectin (CNL), exhibits an immunostimulatory effect on the most potent antigen-presenting cells, the dendritic cells (DCs). Treatment of human monocyte-derived DCs with CNL in doses from 1 to 10 µg/ml resulted in a dose-dependent induction of overall DC maturation characteristics. Exposure of DCs to CNL for 48 hr resulted in extensive up-regulation of co-stimulatory molecules CD80 and CD86, as well as of the maturation marker CD83 and HLA-DR molecules. Such CNL-matured DCs (CNL-DCs) were capable of inducing a T helper type 1-polarized response in naive CD4+ CD45RA+ T cells in 5-day allogeneic co-cultures. The allostimulatory potential of CNL-DCs was significantly increased relative to untreated controls, as was their capacity to produce several pro-inflammatory cytokines such as interleukin-6, interleukin-8 and tumour necrosis factor-α. By using a specific Toll-like receptor 4 (TLR4) signalling inhibitor, CLI-095, as well as Myd88 inhibitory peptide, we have shown that DC activation by CNL is completely dependent on the TLR4 activation pathway. Furthermore, activation of TLR4 by CNL was confirmed via TLR4 reporter assay. Measurement of p65 nuclear factor-κB and p38 mitogen-activated protein kinase (MAPK) phosphorylation levels following CNL stimulation of DCs revealed primarily an increase in nuclear factor-κB activity, with less effect on the induction of p38 MAPK signalling than of lipopolysaccharide-matured DCs. The CNL had the ability to activate human DCs in such a way as to subsequently direct T helper type 1 T-cell responses. Our results encourage the use of mushroom-derived lectins for use in therapeutic strategies with aims such as to strengthen anti-tumour immune responses.


Asunto(s)
Presentación de Antígeno/inmunología , Basidiomycota/química , Células Dendríticas/inmunología , Lectinas/inmunología , Ricina/inmunología , Receptor Toll-Like 4/inmunología , Presentación de Antígeno/efectos de los fármacos , Antígeno B7-1/inmunología , Antígeno B7-2/inmunología , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Relación Dosis-Respuesta Inmunológica , Humanos , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Interleucina-8/biosíntesis , Interleucina-8/inmunología , Lectinas/química , Lectinas/aislamiento & purificación , Lectinas/farmacología , Ricina/química , Ricina/aislamiento & purificación , Ricina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células TH1/inmunología , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
19.
J Immunol ; 186(8): 4794-804, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21398612

RESUMEN

Endosomal TLRs play an important role in innate immune response as well as in autoimmune processes. In the therapy of systemic lupus erythematosus, antimalarial drugs chloroquine, hydroxychloroquine, and quinacrine have been used for a long time. Their suppression of endosomal TLR activation has been attributed to the inhibition of endosomal acidification, which is a prerequisite for the activation of these receptors. We discovered that chloroquine inhibits only activation of endosomal TLRs by nucleic acids, whereas it augments activation of TLR8 by a small synthetic compound, R848. We detected direct binding of antimalarials to nucleic acids by spectroscopic experiments and determined their cellular colocalization. Further analysis revealed that other nucleic acid-binding compounds, such as propidium iodide, also inhibited activation of endosomal TLRs and colocalized with nucleic acids to endosomes. We found that imidazoquinolines, which are TLR7/8 agonists, inhibit TLR9 and TLR3 even in the absence of TLR7 or TLR8, and their mechanism of inhibition is similar to the antimalarials. In contrast to bafilomycin, none of the tested antimalarials and imidazoquinolines inhibited endosomal proteolysis or increased the endosomal pH, confirming that inhibition of pH acidification is not the underlying cause of inhibition. We conclude that the direct binding of inhibitors to nucleic acids mask their TLR-binding epitope and may explain the efficiency of those compounds in the treatment of autoimmune diseases.


Asunto(s)
Antimaláricos/farmacología , Endosomas/metabolismo , Imidazoles/farmacología , Quinolinas/farmacología , Receptores Toll-Like/antagonistas & inhibidores , Animales , Antimaláricos/metabolismo , Unión Competitiva , Línea Celular , Células Cultivadas , Cloroquina/farmacología , Medios de Cultivo/química , Medios de Cultivo/farmacología , Citocinas/metabolismo , Endosomas/química , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Imidazoles/metabolismo , Ácidos Nucleicos/metabolismo , Quinacrina/farmacología , Quinolinas/metabolismo , Suero/química , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/agonistas , Receptor Toll-Like 8/genética , Receptor Toll-Like 8/metabolismo , Receptor Toll-Like 9/antagonistas & inhibidores , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/agonistas , Receptores Toll-Like/genética , Transfección
20.
J Leukoc Biol ; 89(6): 893-905, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21447648

RESUMEN

DC-SIGN is a C-type lectin selectively expressed by certain types of DCs, including monocyte-derived DCs. Many reports have described the impact of DC-SIGN engagement with concomitant TLR signaling in tailoring of the DC maturation process, but so far, none has addressed the importance of DC-SIGN engagement during their differentiation from blood progenitors. We therefore examined the role of DC-SIGN engagement limited to the stage of IL-4-guided differentiation of DCs from human peripheral blood monocytes but not during maturation. We used two different anti-DC-SIGN antibodies with reported DC-SIGN-engaging activities. In cultures with DC-SIGN ligands, the resulting iDCs displayed abrogated expression of differentiation markers CD1a and DC-SIGN. Without further DC-SIGN activation, such DCs matured with low CD80/CD86 and high ILT3 expression, along with the appearance of macrophage marker CD14. Additionally, treated DCs indicated a tolerogenic potential by possessing a low, allostimulatory capacity and inducing naïve, allogeneic CD4(+) T cells to produce low levels of IFN-γ. Upon activation, IL-10 production was greatly increased by such DCs; however, the use of IL-10-blocking antibodies could not completely reverse alternative DC activation. This suggests an alternative activation response that is a result of a different elementary state of DCs generated with concomitant ligation of DC-SIGN. During differentiation, IL-4-induced pSTAT6 was reduced by DC-SIGN ligands. Furthermore, during LPS-induced maturation, treated DCs displayed lowered activation levels of p38 MAPK, STAT1, as well as STAT6, compared with controls. Collectively, evidence is presented confirming a crucial role for DC-SIGN signaling in DC generation from monocytes.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Células Dendríticas/citología , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Monocitos/citología , Receptores de Superficie Celular/metabolismo , Western Blotting , Moléculas de Adhesión Celular/genética , Proliferación Celular , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/inmunología , Endocitosis , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-4/farmacología , Lectinas Tipo C/genética , Ligadura , Lipopolisacáridos/farmacología , Activación de Linfocitos , Monocitos/inmunología , Monocitos/metabolismo , ARN Mensajero/genética , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA