Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Inflammation ; 46(6): 2320-2331, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37581762

RESUMEN

The retina represents a highly specialized structure with the primary function to capture a light signal and to convert it into electrical impulses. Any damage or disease of the retina can cause visual impairment. Since retinal degenerative diseases are generally associated with immune cell infiltration, a local inflammatory reaction, and cytokine burn, there is a need for mechanisms to prevent the retina from damage by a deleterious immune reaction. In this study, we show that mouse retinal explants co-cultivated with stimulated spleen cells, inhibit in a dose-dependent manner the activation of T cells, and suppress the production of cytokines interleukin-2, interleukin-10, and interferon-[Formula: see text]. The immunoregulatory properties of the retina were mainly mediated by a paracrine effect since retinal explants, separated by a semipermeable membrane, or supernatants obtained after the cultivation of retinal explants, inhibited the reactivity of immune cells. A model of retinal damage was established by the application of sodium iodate which selectively destroys photoreceptors, as it was demonstrated by a decrease in the number of rhodopsin-positive cells. This process was accompanied by increased infiltration of the retina with cells of the immune system and by a local inflammatory reaction. The pharmacologically damaged retina had significantly decreased the ability to inhibit T cell activation and production of cytokines by immune cells. Overall, the results showed that the retina possesses immunoregulatory properties and inhibits the activation and functions of T cells. However, the immunomodulatory properties of the retina are decreased if the retina is damaged.


Asunto(s)
Citocinas , Retina , Animales , Ratones , Citocinas/metabolismo , Inflamación/metabolismo
2.
Stem Cell Rev Rep ; 19(5): 1360-1369, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36810951

RESUMEN

Negative impacts of nanomaterials on stem cells and cells of the immune system are one of the main causes of an impaired or slowed tissue healing. Therefore, we tested effects of four selected types of metal nanoparticles (NPs): zinc oxide (ZnO), copper oxide (CuO), silver (Ag), and titanium dioxide (TiO2) on the metabolic activity and secretory potential of mouse mesenchymal stem cells (MSCs), and on the ability of MSCs to stimulate production of cytokines and growth factors by macrophages. Individual types of nanoparticles differed in the ability to inhibit metabolic activity, and significantly decreased the production of cytokines and growth factors (interleukin-6, vascular endothelial growth factor, hepatocyte growth factor, insulin-like growth factor-1) by MSCs, with the strongest inhibitory effect of CuO NPs and the least effect of TiO2 NPs. The recent studies indicate that immunomodulatory and therapeutic effects of transplanted MSCs are mediated by macrophages engulfing apoptotic MSCs. We co-cultivated macrophages with heat-inactivated MSCs which were untreated or were preincubated with the highest nontoxic concentrations of metal NPs, and the secretory activity of macrophages was determined. Macrophages cultivated in the presence of both untreated MSCs or MSCs preincubated with NPs produced significantly enhanced and comparable levels of various cytokines and growth factors. These results suggest that metal nanoparticles inhibit therapeutic properties of MSCs by a direct negative effect on their secretory activity, but MSCs cultivated in the presence of metal NPs have preserved the ability to stimulate cytokine and growth factor production by macrophages.


Asunto(s)
Células Madre Mesenquimatosas , Nanopartículas del Metal , Ratones , Animales , Factor A de Crecimiento Endotelial Vascular/farmacología , Citocinas
3.
Toxicol In Vitro ; 87: 105536, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36528116

RESUMEN

Nanoparticles (NPs) have a wide use in various field of industry and in medicine, where they represent a promise for their antimicrobial effects. Simultaneous application of NPs and therapeutic stem cells can speed up tissue regeneration and improve healing process but there is a danger of negative impacts of NPs on stem cells. Therefore, we tested effects of four types of metal antimicrobial NPs on characteristics and function properties of mouse mesenchymal stem cells (MSCs) in vitro. All types of tested NPs, i.e. zinc oxide, silver, copper oxide and titanium dioxide, exerted negative effects on the expression of phenotypic markers, metabolic activity, differentiation potential, expression of genes for immunoregulatory molecules and on production of cytokines and growth factors by MSCs. However, there were apparent differences in the impact of individual types of NPs on tested characteristics and function properties of MSCs. The results showed that individual types of NPs influence the activity of MSCs, and thus the use of metal NPs during tissue regeneration and in combination with stem cell therapy should be well considered.


Asunto(s)
Antiinfecciosos , Células Madre Mesenquimatosas , Nanopartículas del Metal , Nanopartículas , Ratones , Animales , Nanopartículas del Metal/toxicidad , Diferenciación Celular , Cicatrización de Heridas
4.
Stem Cell Rev Rep ; 18(7): 2365-2375, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35288846

RESUMEN

An encounter of the developing immune system with an antigen results in the induction of immunological areactivity to this antigen. In the case of transplantation antigens, the application of allogeneic hematopoietic cells induces a state of neonatal transplantation tolerance. This tolerance depends on the establishment of cellular chimerism, when allogeneic cells survive in the neonatally treated recipient. Since mesenchymal stem/stromal cells (MSCs) have been shown to have low immunogenicity and often survive in allogeneic recipients, we attempted to use these cells for induction of transplantation tolerance. Newborn (less than 24 h old) C57BL/6 mice were injected intraperitoneally with 5 × 106 adipose tissue-derived MSCs isolated from allogeneic donors and the fate and survival of these cells were monitored. The impact of MSC application on the proportion of cell populations of the immune system and immunological reactivity was assessed. In addition, the survival of skin allografts in neonatally treated recipients was tested. We found that in vitro expanded MSCs did not survive in neonatal recipients, and the living MSCs were not detected few days after their application. Furthermore, there were no significant changes in the proportion of individual immune cell populations including CD4+ cell lineages, but we detected an apparent shift to the production of Th1 cytokines IL-2 and IFN-γ in neonatally treated mice. However, skin allografts in the MSC-treated recipients were promptly rejected. These results therefore show that in vitro expanded MSCs do not survive in neonatal recipients, but induce a cytokine imbalance without induction of transplantation tolerance.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Animales Recién Nacidos , Citocinas , Interleucina-2 , Ratones , Ratones Endogámicos C57BL , Tolerancia al Trasplante
5.
J Immunol Methods ; 493: 113013, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33689808

RESUMEN

Nonspecific binding of conjugated antibodies represents a critical step which could significantly influence the results of immunostaining or flow cytometry. In this respect, various staining procedures and distinct cell types can alter the results obtained with different fluorochromes. In this study, we analysed nonspecific binding of R-phycoerythrin (R-PE)-conjugated antibodies to mouse mitogen-stimulated B and T lymphocytes. The cells were fixed, permeabilized and stained using isotype control antibodies conjugated with different fluorochromes and assessed by flow cytometry. R-PE-conjugated antibodies bound to LPS-stimulated B cells, in contrast to Con A-stimulated T cells, independently of their specificity. The percentage of R-PE positive B cells varied, according to the used antibodies or the fixation/permeabilization kit. Nevertheless, up to 30% of R-PE+ B cells after staining with R-PE-conjugated isotype control antibodies was detected. Furthermore, LPS-stimulated B cells bound nonspecifically, in a dose-dependent manner, unconjugated R-PE molecules. Con A-stimulated T cells slightly bound R-PE only in high concentrations. Similarly, the antibodies conjugated with other fluorochromes showed less than 1% of nonspecific binding independently of the manufacturer of antibodies or fixation/permeabilization kits. The data demonstrated that LPS-stimulated B cells, in contrast to Con A-stimulated T cells, bind R-PE nonspecifically following formaldehyde or paraformaldehyde fixation. Therefore, the results based on the use of R-PE-conjugated antibodies should be taken with a precaution.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Linfocitos B/inmunología , Mitógenos/inmunología , Ficoeritrina/inmunología , Linfocitos T/inmunología , Animales , Sitios de Unión/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ficoeritrina/metabolismo
6.
Stem Cells Dev ; 30(8): 418-427, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33607933

RESUMEN

Mesenchymal stem cells (MSCs) represent a population of adult stem cells that have potent immunoregulatory, anti-inflammatory, and antiapoptotic properties. In addition, they have ability to migrate to the site of inflammation or injury, where they contribute to the regeneration and healing process. For these properties, MSCs have been used as therapeutic cells in several models, including treatment of damages or disorders of the ocular surface. If the damage of the ocular surface is extensive and involves a limbal region where limbal stem cell reside, MSC therapy has been proved as the effective treatment approach. Although the anti-inflammatory properties of MSCs have been well characterized, mechanisms of antiapoptotic action of MSCs are not well recognized. Using a chemically damaged cornea in a mouse model, we showed that the injury decreases expression of the gene for antiapoptotic molecule Bcl-2 and increases the expression of proapoptotic genes Bax and p53. These changes were attenuated by local transplantation of MSCs after corneal damage. The antiapoptotic effect of MSCs was tested in an in vitro model of co-cultivation of corneal explants with MSCs. The apoptosis of corneal cells in the explants was induced by proinflammatory cytokines and was significantly inhibited in the presence of MSCs. The antiapoptotic effect of MSCs was mediated by paracrine action, as confirmed by separation of the explants in inserts or by supernatants from MSCs. In addition, MSCs decreased the expression of genes for the molecules associated with endoplasmic reticulum stress Atf4, Bip, and p21, which are associated with apoptosis. The results show that MSCs inhibit the expression of proapoptotic genes and decrease the number of apoptotic cells in the damaged corneas, and this action might be one of the mechanisms of the therapeutic action of MSCs.


Asunto(s)
Apoptosis/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Queratitis/genética , Células Madre Mesenquimatosas/metabolismo , Animales , Células Cultivadas , Córnea/metabolismo , Lesiones de la Cornea/genética , Lesiones de la Cornea/metabolismo , Lesiones de la Cornea/terapia , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Femenino , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Queratitis/metabolismo , Queratitis/patología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Front Immunol ; 12: 760881, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35154069

RESUMEN

Differences in frequencies of blood cell subpopulations were reported to influence the course of infections, atopic and autoimmune diseases, and cancer. We have discovered a unique mouse strain B10.O20 containing extremely high frequency of myeloid-derived cells (MDC) in spleen. B10.O20 carries 3.6% of genes of the strain O20 on the C57BL/10 genetic background. It contains much higher frequency of CD11b+Gr1+ cells in spleen than both its parents. B10.O20 carries O20-derived segments on chromosomes 1, 15, 17, and 18. Their linkage with frequencies of blood cell subpopulations in spleen was tested in F2 hybrids between B10.O20 and C57BL/10. We found 3 novel loci controlling MDC frequencies: Mydc1, 2, and 3 on chromosomes 1, 15, and 17, respectively, and a locus controlling relative spleen weight (Rsw1) that co-localizes with Mydc3 and also influences proportion of white and red pulp in spleen. Mydc1 controls numbers of CD11b+Gr1+ cells. Interaction of Mydc2 and Mydc3 regulates frequency of CD11b+Gr1+ cells and neutrophils (Gr1+Siglec-F- cells from CD11b+ cells). Interestingly, Mydc3/Rsw1 is orthologous with human segment 6q21 that was shown previously to determine counts of white blood cells. Bioinformatics analysis of genomic sequence of the chromosomal segments bearing these loci revealed polymorphisms between O20 and C57BL/10 that change RNA stability and genes' functions, and we examined expression of relevant genes. This identified potential candidate genes Smap1, Vps52, Tnxb, and Rab44. Definition of genetic control of MDC can help to personalize therapy of diseases influenced by these cells.


Asunto(s)
Células Mieloides/fisiología , Animales , Cromosomas/genética , Biología Computacional/métodos , Femenino , Ligamiento Genético/genética , Sitios Genéticos/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología , Polimorfismo Genético/genética , Estabilidad del ARN/genética , Bazo/fisiología
8.
Cell Immunol ; 347: 103987, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31787200

RESUMEN

The knowledge of mechanisms of regulation of IL-10 production by B cells remains still very limited. We show here that highly purified mouse B cells stimulated with LPS produce significant levels of IL-10, but Bregs in our model do not express detectable level of either Foxp3 or GATA-3. Nevertheless, IL-10 production by B cells is regulated by cytokines. In activated B cells, IL-10 production was significantly enhanced by IFN-γ and decreased in the presence of IL-4 or TGF-ß. These findings are in sharp contrast with the observations in T cells, where IL-10 production correlates with GATA-3 or FoxP3 expression, and the cytokines regulate IL-10 production in a reverse manner than in activated B cells. These results thus show that the production of IL-10 by Bregs is regulated by cytokines independently of the expression of GATA-3 and FoxP3, which is clearly different from GATA-3-dependent IL-10 production by activated Th2 cells and FoxP3 expression in IL-10-producing Tregs.


Asunto(s)
Linfocitos B Reguladores/inmunología , Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción GATA3/metabolismo , Interleucina-10/biosíntesis , Animales , Células Cultivadas , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Interferón gamma/inmunología , Interleucina-4/inmunología , Lipopolisacáridos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Factor de Crecimiento Transformador beta/inmunología
9.
Stem Cell Rev Rep ; 15(6): 880-891, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31863334

RESUMEN

Retinal degenerative disorders are characterized by a local upregulation of inflammatory factors, infiltration with cells of the immune system, a vascular dysfunction and by the damage of retinal cells. There is still a lack of treatment protocols for these diseases. Mesenchymal stem cell (MSC)-based therapy using immunoregulatory, regenerative and differentiating properties of MSCs offers a promising treatment option. In this study, we analyzed the immunomodulatory properties of mouse bone marrow-derived MSCs after their intravitreal delivery to the inflammatory environment in the eye, caused by the application of pro-inflammatory cytokines IL-1ß, TNF-α and IFN-γ. The intravitreal administration of these cytokines induces an increased expression of pro-inflammatory molecules such as IL-1α, IL-6, inducible nitric oxide synthase, TNF-α and vascular endothelial growth factor in the retina. However, a significant decrease in the expression of genes for all these pro-inflammatory molecules was observed after the intravitreal injection of MSCs. We further showed that an increased infiltration of the retina with immune cells, mainly with macrophages, which was observed after pro-inflammatory cytokine application, was significantly reduced after the intravitreal application of MSCs. The similar immunosuppressive effects of MSCs were also demonstrated in vitro in cultures of cytokine-stimulated retinal explants and MSCs. Overall, the results show that intravitreal application of MSCs inhibits the early retinal inflammation caused by pro-inflammatory cytokines, and propose MSCs as a promising candidate for stem cell-based therapy of retinal degenerative diseases.


Asunto(s)
Inmunomodulación/efectos de los fármacos , Mediadores de Inflamación/farmacología , Inflamación/prevención & control , Células Madre Mesenquimatosas/citología , Retina/efectos de los fármacos , Animales , Antivirales/farmacología , Citocinas/metabolismo , Femenino , Inmunomodulación/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Retina/citología , Retina/inmunología , Retina/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
10.
Nanomaterials (Basel) ; 9(5)2019 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-31052583

RESUMEN

In the body, engineered nanoparticles (NPs) may be recognized and processed by immune cells, among which macrophages play a crucial role. We evaluated the effects of selected NPs [NM-100 (TiO2), NM-110 (ZnO), NM-200 (SiO2), and NM-300 K (Ag)] on THP-1 macrophage-like cells. The cells were exposed to subcytotoxic concentrations of NPs (1-25 µg/mL) and the expression of immunologically relevant genes (VCAM1, TNFA, CXCL8, ICAM1, CD86, CD192, and IL1B) was analyzed by RT-qPCR. The expression of selected cytokines, growth factors and surface molecules was assessed by flow cytometry or ELISA. Generation of reactive oxygen species and induction of DNA breaks were also analyzed. Exposure to diverse NPs caused substantially different molecular responses. No significant effects were detected for NM-100 treatment. NM-200 induced production of IL-8, a potent attractor and activator of neutrophils, growth factors (VEGF and IGF-1) and superoxide. NM-110 triggered a proinflammatory response, characterized by the activation of transcription factor NF-κB, an enhanced production of proinflammatory cytokines (TNF-α) and chemokines (IL-8). Furthermore, the expression of cell adhesion molecules VCAM-1 and ICAM-1 and hepatocyte growth factor (HGF), as well as superoxide production and DNA breaks, were affected. NM-300 K enhanced IL-8 production and induced DNA breaks, however, it decreased the expression of chemokine receptor (CCR2) and CD86 molecule, indicating potential immunosuppressive activity. The toxicity of ZnO and Ag NPs was probably caused by their intracellular dissolution, as indicated by transmission electron microscopy imaging. The observed effects in macrophages might further influence both innate and adaptive immune responses by promoting neutrophil recruitment via IL-8 release and enhancing the adhesion and stimulation of T cells by VCAM-1 and ICAM-1 expression.

11.
Nanotoxicology ; 13(7): 952-963, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31012774

RESUMEN

The inhalation or application of nanoparticles (NPs) has serious impacts on immunological reactivity. However, the effects of NPs on the immune system are influenced by numerous factors, which cause a high variability in the results. Here, mice were exposed to a three month continuous inhalation of copper oxide (CuO) NPs, and at different time intervals (3, 14, 42 and 93 days), the composition of cell populations of innate and adaptive immunity was evaluated in the spleen by flow cytometry. The ability of spleen cells from exposed and control mice to respond to stimulation with T- or B-cell mitogens by proliferation and by production of cytokines IL-2, IL-6, IL-10, IL-17 and IFN-γ was characterized. The results showed that the inhalation of CuO NPs predominantly affects the cells of innate immunity (changes in the proportion of eosinophils, neutrophils, macrophages and antigen-presenting cells) with a minimal effect on the percentage of T and B lymphocytes. However, the proliferative and secretory activity of T cells was already significantly enhanced after 3 days from the start of inhalation, decreased on day 14 and normalized at the later time intervals. There was no correlation between the impacts of NPs on the cells of innate and adaptive immunity. The results have shown that the inhalation of CuO NPs significantly alters the composition of cell populations of innate immunity and modulates the proliferation and production of cytokines by cells of the adaptive immune system. However, the immunomodulatory effects of inhaled NPs strongly depend on the time of inhalation.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Cobre/toxicidad , Inmunidad Innata/efectos de los fármacos , Nanopartículas/toxicidad , Administración por Inhalación , Animales , Cobre/administración & dosificación , Citocinas/biosíntesis , Femenino , Cinética , Ratones , Ratones Endogámicos ICR , Modelos Animales
12.
J Neuroimmune Pharmacol ; 14(2): 215-225, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30242613

RESUMEN

Pathogenesis of amyotrophic lateral sclerosis (ALS) involves several mechanisms resulting in a shift from a neuroprotective to a neurotoxic immune reaction. A promising tool for ALS treatment is represented by mesenchymal stem cells (MSCs), which possess both regenerative potential and immunomodulatory properties. In this study, we aimed to compare the immunomodulatory properties of MSCs isolated from the bone marrow of patients suffering from ALS and healthy donors. Moreover, the influence of proinflammatory cytokines on the immunoregulatory functions of MSCs was also evaluated. We found that MSCs from ALS patients and healthy donors comparably affected mitogen-stimulated peripheral blood mononuclear cells and reduced the percentage of T helper (Th)1, Th17 and CD8+CD25+ lymphocytes. These MSCs also equally increased the percentage of Th2 and CD4+FOXP3+ T lymphocytes. On the other hand, MSCs from ALS patients decreased more strongly the production of tumour necrosis factor-α than MSCs from healthy donors, but this difference was abrogated in the case of MSCs stimulated with cytokines. Significant differences between cytokine-treated MSCs from ALS patients and healthy donors were detected in the effects on the percentage of CD8+CD25+ and CD4+FOXP3+ T lymphocytes. In general, treatment of MSCs with cytokines results in a potentiation of their effects, but in the case of MSCs from ALS patients, it causes stagnation or even restriction of some of their immunomodulatory properties. We conclude that MSCs from ALS patients exert comparable immunomodulatory effects to MSCs from healthy donors, but respond differently to stimulation with proinflammatory cytokines. Graphical Abstract Treatment of mesenchymal stem cells (MSCs) with cytokines results in a potentiation of their effects, but in the case of MSCs from amyotrophic lateral sclerosis (ALS) patients, it causes stagnation (an equal reduction of the percentage of CD8+CD25+ T lymphocytes) or even restriction (no increase of proportion of CD4+FOXP3+ T lymphocytes) of some of their immunomodulatory properties. It means that MSCs from ALS patients exert comparable immunomodulatory effects to MSCs from healthy donors, but respond differently to stimulation with proinflammatory cytokines.


Asunto(s)
Esclerosis Amiotrófica Lateral/inmunología , Células de la Médula Ósea/inmunología , Células Madre Mesenquimatosas/inmunología , Citocinas/metabolismo , Femenino , Voluntarios Sanos , Humanos , Factores Inmunológicos/farmacología , Inmunomodulación , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Mitógenos/farmacología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
13.
Toxicol Sci ; 168(1): 190-200, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30500950

RESUMEN

Despite the wide application of nanomaterials, toxicity studies of nanoparticles (NP) are often limited to in vitro cell models, and the biological impact of NP exposure in mammals has not been thoroughly investigated. Zinc oxide (ZnO) NPs are commonly used in various consumer products. To evaluate the effects of the inhalation of ZnO NP in mice, we studied splice junction expression in the lungs as a proxy to gene expression changes analysis. Female ICR mice were treated with 6.46 × 104 and 1.93 × 106 NP/cm3 for 3 days and 3 months, respectively. An analysis of differential expression and alternative splicing events in 298 targets (splice junctions) of 68 genes involved in the processes relevant to the biological effects of ZnO NP was conducted using next-generation sequencing. Three days of exposure resulted in the upregulation of IL-6 and downregulation of BID, GSR, NF-kB2, PTGS2, SLC11A2, and TXNRD1 splice junction expression; 3 months of exposure increased the expression of splice junctions in ALDH3A1, APAF1, BID, CASP3, DHCR7, GCLC, GCLM, GSR, GSS, EHHADH, FAS, HMOX-1, IFNγ, NF-kB1, NQO-1, PTGS1, PTGS2, RAD51, RIPK2, SRXN1, TRAF6, and TXNRD1. Alternative splicing of TRAF6 and TXNRD1 was induced after 3 days of exposure to 1.93 × 106 NP/cm3. In summary, we observed changes of splice junction expression in genes involved in oxidative stress, apoptosis, immune response, inflammation, and DNA repair, as well as the induction of alternative splicing in genes associated with oxidative stress and inflammation. Our data indicate the potential negative biological effects of ZnO NP inhalation.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Nanopartículas/toxicidad , Óxido de Zinc/toxicidad , Administración por Inhalación , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Inflamación , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos ICR , Estrés Oxidativo/efectos de los fármacos
14.
Stem Cell Rev Rep ; 14(6): 801-811, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30136142

RESUMEN

Morphine is an analgesic drug therapeutically administered to relieve pain. However, this drug has numerous side effects, which include impaired healing and regeneration after injuries or tissue damages. It suggests negative effects of morphine on stem cells which are responsible for tissue regeneration. Therefore, we studied the impact of morphine on the properties and functional characteristics of human bone marrow-derived mesenchymal stem cells (MSCs). The presence of µ-, δ- and κ-opioid receptors (OR) in untreated MSCs, and the enhanced expression of OR in MSCs pretreated with proinflammatory cytokines, was demonstrated using immunoblotting and by flow cytometry. Morphine modified in a dose-dependent manner the MSC phenotype, inhibited MSC proliferation and altered the ability of MSCs to differentiate into adipocytes or osteoblasts. Furthermore, morphine rather enhanced the expression of genes for various immunoregulatory molecules in activated MSCs, but significantly inhibited the production of the vascular endothelial growth factor, hepatocyte growth factor or leukemia inhibitory factor. All of these observations are underlying the selective impact of morphine on stem cells, and offer an explanation for the mechanisms of the negative effects of opioid drugs on stem cells and regenerative processes after morphine administration or in opioid addicts.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Morfina/farmacología , Osteoblastos/metabolismo , Receptores Opioides/metabolismo , Humanos , Células Madre Mesenquimatosas/patología
15.
J Neuroimmunol ; 321: 12-23, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29957382

RESUMEN

Regulation of µ-, δ- and κ-opioid receptor protein level in spleen lymphocytes when stimulated by mitogen is not known. To answer the question whether these cells do express opioid receptor (OR) proteins, primary, fresh rat spleen lymphocytes were prepared and stimulated for 48 h with mitogenic dose of Con A. The unstimulated lymphocytes did not express µ- and δ-OR proteins in detectable amounts, however, stimulation with Con A resulted in appearance of clearly detectable immunoblot signals of both µ-OR and δ-OR. κ-OR were detected already in primary cells and increased 2.4-fold in Con A-stimulated cells. These results were supported by data obtained by flow cytometry analysis indicating a dramatic increase in number of µ-, δ- and κ-OR expressing cells after mitogen stimulation. The newly synthesized µ-, δ- and κ-OR in Con A-stimulated spleen lymphocytes were present in the cells interior and not functionally mature, at least in terms of their ability to enhance activity of trimeric G proteins determined by three different protocols of agonist-stimulated, high-affinity [35S]GTPγS binding assay. The up-regulation of µ-, δ- and κ-OR was associated with specific decrease of their cognate trimeric G proteins, Gi1α/Gi2α; the other Gα and Gß subunits were unchanged. The level of ß-arrestin-1/2 was also decreased in Con A-stimulated splenocytes. We conclude that up-regulation of OR expression level in spleen lymphocytes by Con A proceeds in conjunction with down-regulation of their intracellular signaling partners, Gi1α/Gi2α proteins and ß-arrestin-1/2. These regulatory proteins are expressed in high amounts already in unstimulated cells and decreased by mitogen stimulation.


Asunto(s)
Linfocitos/metabolismo , Receptores Opioides delta/biosíntesis , Receptores Opioides kappa/biosíntesis , Receptores Opioides mu/biosíntesis , Bazo/metabolismo , Animales , Concanavalina A/farmacología , Linfocitos/efectos de los fármacos , Masculino , Mitógenos/farmacología , Ratas , Ratas Wistar , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides mu/efectos de los fármacos , Bazo/citología , Bazo/efectos de los fármacos , Regulación hacia Arriba
16.
J Clin Pathol ; 71(8): 735-742, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29523587

RESUMEN

AIMS: Mesenchymal stem cells (MSCs) have recently been tested in clinical trials to treat severe diseases, including amyotrophic lateral sclerosis (ALS). Since autologous MSCs are frequently used for therapy, we aimed to evaluate the possible influence of the disease on characteristics and function of these cells. METHODS: MSCs were isolated from the bone marrow of patients with ALS and compared with MSCs from healthy controls (HC). The cells were tested for phenotype, growth properties, differentiation ability, metabolic activity, secretory potential, expression of genes for immunomodulatory molecules and for the ability to regulate proliferation of mitogen-stimulated peripheral blood leucocytes. MSCs from patients with ALS and HC were either unstimulated or treated with proinflammatory cytokines for 24 hours before testing. RESULTS: MSCs isolated from patients with ALS have a higher differentiation potential into adipocytes, express elevated levels of mRNA for interleukin-6, but produce less hepatocyte growth factor than MSCs from HC. On the other hand, there were no significant differences between MSCs from patients with ALS and HC in the expression of phenotypic markers, growth properties, metabolic activity, osteogenic differentiation potential and immunoregulatory properties. CONCLUSIONS: The results suggest that, in spite of some differences in cytokine production, MSCs from patients with ALS can be useful as autologous cells in therapy of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Células de la Médula Ósea/patología , Separación Celular/métodos , Células Madre Mesenquimatosas/patología , Adipogénesis , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/metabolismo , Biomarcadores/metabolismo , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Metabolismo Energético , Femenino , Humanos , Activación de Linfocitos , Masculino , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Osteogénesis , Fenotipo
17.
Biomed Pharmacother ; 97: 402-411, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29091890

RESUMEN

Immunosuppressive drugs are used to suppress graft rejection after transplantation and for the treatment of various diseases. The main limitations of their use in clinical settings are severe side effects, therefore alternative approaches are desirable. In this respect, mesenchymal stem cells (MSCs) possess a regenerative and immunomodulatory capacity that has generated considerable interest for their use in cell-based therapy. Currently, MSCs are tested in many clinical trials, including the treatment of diseases which require simultaneous immunosuppressive treatment. Since the molecular targets of immunosuppressive drugs are also present in MSCs, we investigated whether immunosuppressive drugs interact with the activity of MSCs. Human MSCs isolated from the bone marrow (BM) or adipose tissue (AT) were cultured in the presence of clinical doses of five widely used immunosuppressive drugs (cyclosporine A, mycophenolate mofetil, rapamycin, prednisone and dexamethasone), and the influence of these drugs on several factors related to the immunosuppressive properties of MSCs, including the expression of immunomodulatory enzymes, various growth factors, cytokines, chemokines, adhesion molecules and proapoptotic ligands, was assessed. Glucocorticoids, especially dexamethasone, showed the most prominent effects on both types of MSCs and suppressed the expression of the majority of the factors that were tested. A significant increase of hepatocyte growth factor production in AT-MSCs and of indoleamine 2,3-dioxygenase expression in both types of MSCs were the only exceptions. In conclusion, clinically relevant doses of inhibitors of calcineurin, mTOR and IMPDH and glucocorticoids interfere with MSC functions, but do not restrain their immunosuppressive properties. These findings should be taken into account before preparing immunosuppressive strategies combining the use of immunosuppressive drugs and MSCs.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Inmunosupresores/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Interferón gamma/agonistas , Interferón gamma/biosíntesis , Leucocitos Mononucleares/metabolismo , Células Madre Mesenquimatosas/metabolismo
18.
Stem Cells Dev ; 26(19): 1399-1408, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28728472

RESUMEN

Retinal disorders represent the main cause of decreased quality of vision and even blindness worldwide. The loss of retinal cells causes irreversible damage of the retina, and there are currently no effective treatment protocols for most retinal degenerative diseases. A promising approach for the treatment of retinal disorders is represented by stem cell-based therapy. The perspective candidates are mesenchymal stem cells (MSCs), which can differentiate into multiple cell types and produce a number of trophic and growth factors. In this study, we show the potential of murine bone marrow-derived MSCs to differentiate into cells expressing retinal markers and we identify the key supportive role of interferon-γ (IFN-γ) in the differentiation process. MSCs were cultured for 7 days with retinal extract and supernatant from T-cell mitogen concanavalin A-stimulated splenocytes, simulating the inflammatory site of retinal damage. MSCs cultured in such conditions differentiated to the cells expressing retinal cell markers such as rhodopsin, S antigen, retinaldehyde-binding protein, calbindin 2, recoverin, and retinal pigment epithelium 65. To identify a supportive molecule in the supernatants from activated spleen cells, MSCs were cultured with retinal extract in the presence of various T-cell cytokines. The expression of retinal markers was enhanced only in the presence of IFN-γ, and the supportive role of spleen cell supernatants was abrogated with the neutralization antibody anti-IFN-γ. In addition, differentiated MSCs were able to express a number of neurotrophic factors, which are important for retinal regeneration. Taken together, the results show that MSCs can differentiate into cells expressing retinal markers and that this differentiation process is supported by IFN-γ.


Asunto(s)
Diferenciación Celular , Interferón gamma/farmacología , Células Madre Mesenquimatosas/citología , Retina/citología , Animales , Calbindina 2/genética , Calbindina 2/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Femenino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Recoverina/genética , Recoverina/metabolismo , Retina/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , cis-trans-Isomerasas/genética , cis-trans-Isomerasas/metabolismo
19.
J Tissue Eng Regen Med ; 11(5): 1456-1465, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-26118469

RESUMEN

The immunosuppressive effects of systemically administered mesenchymal stem cells (MSCs) and immunosuppressive drugs have been well documented. We analysed the mechanisms underlying the therapeutic effect of MSCs applied locally in combination with non-specific immunosuppression in a mouse model of allogeneic skin transplantation. The MSC-seeded and cyclosporine A (CsA)-loaded nanofibre scaffolds were applied topically to skin allografts in a mouse model and the local immune response was assessed and characterized. MSCs migrated from the scaffold into the side of injury and were detected in the graft region and draining lymph nodes (DLNs). The numbers of graft-infiltrating macrophages and the production of nitric oxide (NO) were significantly decreased in recipients treated with MSCs and CsA, and this reduction correlated with impaired production of IFNγ in the graft and DLNs. In contrast, the proportion of alternatively activated macrophages (F4/80+ CD206+ cells) and the production of IL-10 by intragraft macrophages were significantly upregulated. The ability of MSCs to alter the phenotype of macrophages from the M1 type into an M2 population was confirmed in a co-culture system in vitro. We suggest that the topical application of MSCs in combination with CsA induces a switch in macrophages to a population with an alternatively activated 'healing' phenotype and producing elevated levels of IL-10. These alterations in macrophage phenotype and function could represent one of the mechanisms of immunosuppressive action of MSCs applied in combination with CsA. Copyright © 2015 John Wiley & Sons, Ltd.


Asunto(s)
Ciclosporina/farmacología , Supervivencia de Injerto/efectos de los fármacos , Macrófagos/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Trasplante de Piel , Aloinjertos , Animales , Diferenciación Celular , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C
20.
J Tissue Eng Regen Med ; 11(5): 1588-1597, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-26118945

RESUMEN

Damaged neural tissue is regenerated by neural stem cells (NSCs), which represent a rare and difficult-to-culture cell population. Therefore, alternative sources of stem cells are being tested to replace a shortage of NSCs. Here we show that mouse adipose tissue-derived mesenchymal stem cells (MSCs) can be effectively differentiated into cells expressing neuronal cell markers. The differentiation protocol, simulating the inflammatory site of neural injury, involved brain tissue extract, fibroblast growth factor, epidermal growth factor, supernatant from activated splenocytes and electrical stimulation under physiological conditions. MSCs differentiated using this protocol displayed neuronal cell morphology and expressed genes for neuronal cell markers, such as neurofilament light (Nf-L), medium (Nf-M) and heavy (Nf-H) polypeptides, synaptophysin (SYP), neural cell adhesion molecule (NCAM), glutamic acid decarboxylase (GAD), neuron-specific nuclear protein (NeuN), ßIII-tubulin (Tubb3) and microtubule-associated protein 2 (Mtap2), which are absent (Nf-L, Nf-H, SYP, GAD, NeuN and Mtap2) or only slightly expressed (NCAM, Tubb3 and Nf-M) in undifferentiated cells. The differentiation was further enhanced when the cells were cultured on nanofibre scaffolds. The neural differentiation of MSCs, which was detected at the level of gene expression, was confirmed by positive immunostaining for Nf-L protein. The results thus show that the simulation of conditions in an injured neural tissue and inflammatory environment, supplemented with electrical stimulation under physiological conditions and cultivation of cells on a three-dimensional (3D) nanofibre scaffold, form an effective protocol for the differentiation of MSCs into cells with neuronal markers. Copyright © 2015 John Wiley & Sons, Ltd.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Animales , Inflamación/metabolismo , Inflamación/patología , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos BALB C , Tejido Nervioso/patología , Células-Madre Neurales/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...