Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell Mol Life Sci ; 75(8): 1483-1497, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29103146

RESUMEN

Chemical modifications of RNA have been attracting increasing interest because of their impact on RNA fate and function. Therefore, the characterization of enzymes catalyzing such modifications is of great importance. The RNA cytosine methyltransferase NSUN3 was recently shown to generate 5-methylcytosine in the anticodon loop of mitochondrial tRNAMet. Further oxidation of this position is required for normal mitochondrial translation and function in human somatic cells. Because embryonic stem cells (ESCs) are less dependent on oxidative phosphorylation than somatic cells, we examined the effects of catalytic inactivation of Nsun3 on self-renewal and differentiation potential of murine ESCs. We demonstrate that Nsun3-mutant cells show strongly reduced mt-tRNAMet methylation and formylation as well as reduced mitochondrial translation and respiration. Despite the lower dependence of ESCs on mitochondrial activity, proliferation of mutant cells was reduced, while pluripotency marker gene expression was not affected. By contrast, ESC differentiation was skewed towards the meso- and endoderm lineages at the expense of neuroectoderm. Wnt3 was overexpressed in early differentiating mutant embryoid bodies and in ESCs, suggesting that impaired mitochondrial function disturbs normal differentiation programs by interfering with cellular signalling pathways. Interestingly, basal levels of reactive oxygen species (ROS) were not altered in ESCs, but Nsun3 inactivation attenuated induction of mitochondrial ROS upon stress, which may affect gene expression programs upon differentiation. Our findings not only characterize Nsun3 as an important regulator of stem cell fate but also provide a model system to study the still incompletely understood interplay of mitochondrial function with stem cell pluripotency and differentiation.


Asunto(s)
Metiltransferasas/metabolismo , Mitocondrias/enzimología , Células Madre Embrionarias de Ratones/enzimología , Placa Neural/enzimología , ARN de Transferencia de Metionina/metabolismo , 5-Metilcitosina/metabolismo , Animales , Diferenciación Celular , Línea Celular , Cuerpos Embrioides/citología , Cuerpos Embrioides/enzimología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Metiltransferasas/genética , Ratones , Mitocondrias/genética , Células Madre Embrionarias de Ratones/citología , Placa Neural/citología , Placa Neural/crecimiento & desarrollo , Fosforilación Oxidativa , ARN de Transferencia de Metionina/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Transcriptoma
2.
J Cell Mol Med ; 15(6): 1429-31, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21418521

RESUMEN

The connection of embryonic stem cell technology and developmental biology provides valuable tools to decipher the mechanisms underlying human brain development and diseases, especially among neuronal populations, that are not readily available in primary cultures. It is obviously the case of neurons forming the human cerebral cortex. In the images that are presented, the neurons were generated in vitro from human embryonic stem cells via forebrain-like progenitors. Maintained in culture for prolonged time, they acquired a mainly glutamatergic phenotype and morphological characteristics of cortical pyramidal neurons, including dendritic spines, and formed spectacular networks.


Asunto(s)
Corteza Cerebral/citología , Células Madre Embrionarias/citología , Red Nerviosa/citología , Neuronas/citología , Prosencéfalo/citología , Diferenciación Celular , Línea Celular , Corteza Cerebral/embriología , Espinas Dendríticas , Células Madre Embrionarias/fisiología , Fluorescencia , Humanos , Microscopía Confocal , Red Nerviosa/embriología , Neuronas/fisiología , Prosencéfalo/embriología
3.
World J Stem Cells ; 13(12): 1918-1927, 2021 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-35069990

RESUMEN

BACKGROUND: One of the most challenging tasks of modern biology concerns the real-time tracking and quantification of mRNA expression in living cells. On this matter, a novel platform called SmartFlareTM has taken advantage of fluorophore-linked nanoconstructs for targeting RNA transcripts. Although fluorescence emission does not account for the spatial mRNA distribution, NanoFlare technology has grown a range of theranostic applications starting from detecting biomarkers related to diseases, such as cancer, neurodegenerative pathologies or embryonic developmental disorders. AIM: To investigate the potential of SmartFlareTM in determining time-dependent mRNA expression of prominin 1 (CD133) and octamer-binding transcription factor 4 (OCT4) in single living cells through differentiation. METHODS: Brain fragments from the striatum of aborted human fetuses aged 8 wk postconception were processed to obtain neurospheres. For the in vitro differentiation, neurospheres were gently dissociated with Accutase solution. Single cells were resuspended in a basic medium enriched with fetal bovine serum, plated on poly-L-lysine-coated glass coverslips, and grown in a lapse of time from 1 to 4 wk. Live cell mRNA detection was performed using SmartFlareTM probes (CD133, Oct4, Actin, and Scramble). All the samples were incubated at 37 °C for 24 h. For nuclear staining, Hoechst 33342 was added. SmartFlareTM CD133- and OCT4-specific fluorescence signal was assessed using a semiquantitative visual approach, taking into account the fluorescence intensity and the number of labeled cells. RESULTS: In agreement with previous PCR experiments, a unique expression trend was observed for CD133 and OCT4 genes until 7 d in vitro (DIV). Fluorescence resulted in a mixture of diffuse cytoplasmic and spotted-like pattern, also detectable in the contacting neural branches. From 15 to 30 DIV, only few cells showed a scattered fluorescent pattern, in line with the differentiation progression and coherent with mRNA downregulation of these stemness-related genes. CONCLUSION: SmartFlareTM appears to be a reliable, easy-to-handle tool for investigating CD133 and OCT4 expression in a neural stem cell model, preserving cell biological properties in anticipation of downstream experiments.

4.
Stem Cells Dev ; 26(22): 1612-1625, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28946818

RESUMEN

Spinocerebellar ataxia type 6 (SCA6) is an autosomal-dominant neurodegenerative disorder that is caused by a CAG trinucleotide repeat expansion in the CACNA1A gene. As one of the few bicistronic genes discovered in the human genome, CACNA1A encodes not only the α1A subunit of the P/Q type voltage-gated Ca2+ channel CaV2.1 but also the α1ACT protein, a 75 kDa transcription factor sharing the sequence of the cytoplasmic C-terminal tail of the α1A subunit. Isoforms of both proteins contain the polyglutamine (polyQ) domain that is expanded in SCA6 patients. Although certain SCA6 phenotypes appear to be specific for Purkinje neurons, other pathogenic effects of the SCA6 polyQ mutation can affect a broad spectrum of central nervous system (CNS) neuronal subtypes. We investigated the expression and function of CACNA1A gene products in human neurons derived from induced pluripotent stem cells from two SCA6 patients. Expression levels of CACNA1A encoding α1A subunit were similar between SCA6 and control neurons, and no differences were found in the subcellular distribution of CaV2.1 channel protein. The α1ACT immunoreactivity was detected in the majority of cell nuclei of SCA6 and control neurons. Although no SCA6 genotype-dependent differences in CaV2.1 channel function were observed, they were found in the expression levels of the α1ACT target gene Granulin (GRN) and in glutamate-induced cell vulnerability.


Asunto(s)
Canales de Calcio/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Ataxias Espinocerebelosas/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Factores de Transcripción/metabolismo , Expansión de Repetición de Trinucleótido/fisiología
5.
Sci Rep ; 5: 8007, 2015 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-25620209

RESUMEN

The modulation of chromatin dynamics by ATP-dependent chromatin remodeling factors has been recognized as an important mechanism to regulate the balancing of self-renewal and pluripotency in embryonic stem cells (ESCs). Here we have studied the effects of a partial deletion of the gene encoding the chromatin remodeling factor Chd1 that generates an N-terminally truncated version of Chd1 in mouse ESCs in vitro as well as in vivo. We found that a previously uncharacterized serine-rich region (SRR) at the N-terminus is not required for chromatin assembly activity of Chd1 but that it is subject to phosphorylation. Expression of Chd1 lacking this region in ESCs resulted in aberrant differentiation properties of these cells. The self-renewal capacity and ESC chromatin structure, however, were not affected. Notably, we found that newly established ESCs derived from Chd1(Δ2/Δ2) mutant mice exhibited similar differentiation defects as in vitro generated mutant ESCs, even though the N-terminal truncation of Chd1 was fully compatible with embryogenesis and post-natal life in the mouse. These results underscore the importance of Chd1 for the regulation of pluripotency in ESCs and provide evidence for a hitherto unrecognized critical role of the phosphorylated N-terminal SRR for full functionality of Chd1.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Desarrollo Embrionario , Células Madre Embrionarias , Animales , Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Ratones , Serina/genética
6.
J Cell Mol Med ; 4(4): 284-288, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-12067463

RESUMEN

We investigated the apoptosis at the beginning of human cerebral cortex development, in the 6th week of embryogenesis, Carnegie stages 16 and 17. Attention was focused on the dorsal wall of the telencephalon to the ventricular zone of proliferation and to the postmitotic zone with beginning of neuronal migration. We identified apoptotic cells in tissue sections by propidium iodide staining, TUNEL and immunohistochemistry for Fas(APO-1/CD95). We determined the distribution and the percentage (reported to the propidium iodide stained nuclei) of apoptotic TUNEL-positive and Fas(APO-1/CD95)-positive cells. TUNEL-positive apoptotic cells in the proliferative zone were 20% in stage 16 and 60% in stage 17. TUNEL-positive apoptotic cells in the postmitotic zone were 8% in stage16 and 30% in stage 17. CD95-positive apoptotic cells in the proliferative zone were 5% in stage 16 and 2% in stage 17. There were no CD95-positive cells in the postmitotic zone. We evidentiated the presence of the suicide receptor Fas(APO-1/CD95) only on a small population of apoptotic neuroblasts in the proliferative zone. The differences between apoptotic distribution and receptors in early corticogenesis suggest that different apoptotic pathways drive the selection of neuronal populations.

7.
Stem Cells Dev ; 22(24): 3271-82, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23879205

RESUMEN

The value of human disease models, which are based on induced pluripotent stem cells (iPSCs), depends on the capacity to generate specifically those cell types affected by pathology. We describe a new iPSC-based model of Friedreich ataxia (FRDA), an autosomal recessive neurodegenerative disorder with an intronic GAA repeat expansion in the frataxin gene. As the peripheral sensory neurons are particularly susceptible to neurodegeneration in FRDA, we applied a development-based differentiation protocol to generate specifically these cells. FRDA and control iPSC lines were efficiently differentiated toward neural crest progenitors and peripheral sensory neurons. The progress of the cell lines through discrete steps of in vitro differentiation was closely monitored by expression levels of key markers for peripheral neural development. Since it had been suggested that FRDA pathology might start early during ontogenesis, we investigated frataxin expression in our development-related model. A pronounced frataxin deficit was found in FRDA iPSCs and neural crest cells compared to controls. Whereas we identified an upregulation of frataxin expression during sensory specification for control cells, this increase was not observed for FRDA peripheral sensory neurons. This early failure, aggravating frataxin deficiency in a specifically vulnerable human cell population, indicates a developmental component in FRDA.


Asunto(s)
Diferenciación Celular , Ataxia de Friedreich/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas de Unión a Hierro/biosíntesis , Ataxia de Friedreich/genética , Ataxia de Friedreich/patología , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/patología , Células Receptoras Sensoriales/metabolismo , Frataxina
8.
Stem Cells Dev ; 22(3): 397-411, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22889265

RESUMEN

Dopaminergic neurons derived from pluripotent stem cells are among the best investigated products of in vitro stem cell differentiation owing to their potential use for neurorestorative therapy of Parkinson's disease. However, the classical differentiation protocols for both mouse and human pluripotent stem cells generate a limited percentage of dopaminergic neurons and yield a considerable cellular heterogeneity comprising numerous scarcely characterized cell populations. To improve pluripotent stem cell differentiation protocols for midbrain dopaminergic neurons, we established extensive and strictly quantitative gene expression profiles, including markers for pluripotent cells, neural progenitors, non-neural cells, pan-neuronal and glial cells, neurotransmitter phenotypes, midbrain and nonmidbrain populations, floor plate and basal plate populations, as well as for Hedgehog, Fgf, and Wnt signaling pathways. The profiles were applied to discrete stages of in vitro differentiation of mouse embryonic stem cells toward the dopaminergic lineage and after transplantation into the striatum of 6-hydroxy-dopamine-lesioned rats. The comparison of gene expression in vitro with stages in the developing ventral midbrain between embryonic day 11.5 and 13.5 ex vivo revealed dynamic changes in the expression of transcription factors and signaling molecules. Based on these profiles, we propose quantitative gene expression milestones that predict the efficiency of dopaminergic differentiation achieved at the end point of the protocol, already at earlier stages of differentiation.


Asunto(s)
Diferenciación Celular , Neuronas Dopaminérgicas/metabolismo , Células Madre Embrionarias/metabolismo , Mesencéfalo/metabolismo , Células-Madre Neurales/metabolismo , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Células Cultivadas , Neuronas Dopaminérgicas/fisiología , Neuronas Dopaminérgicas/trasplante , Células Madre Embrionarias/fisiología , Expresión Génica , Genes del Desarrollo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Mesencéfalo/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Enfermedad de Parkinson Secundaria/patología , Enfermedad de Parkinson Secundaria/terapia , Ratas , Ratas Wistar , Transducción de Señal , Transcriptoma
9.
Discoveries (Craiova) ; 1(1): e1, 2013 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-32309535

RESUMEN

Discoveries is a new peer-reviewed, open access, online multidisciplinary and integrative journal publishing high impact reviews, experimental articles, perspective articles, and editorials from all areas related to medicine, biology, and chemistry, including but not limited to: Molecular and Cellular Biology, Biochemistry, Biophysics, Genomics, Proteomics, Biotechnology, Synthetic Biology, Bioengineering, Systems Biology, Bioinformatics, Translational Medicine, Medicine/ Clinical findings, Cognitive Science, Epidemiology, Global Medicine, Family Medicine, Organic/ Inorganic/ Physical Chemistry and Ethics in Science. Discoveries brings to the research community an outstanding editorial board that aims to address several of the innovations proposed above: there is no need to format the manuscript before submission, we have a rapid and efficient submission process, there is no need for a Cover Letter and we support the need for rules for validation of critical reagents, such as antibodies. Discoveries will aim to support high quality research on human subjects materials to provide relevance for non-human studies along with mechanistic insights into human biology and chemistry. We also aim to avoid requesting unnecessary experiments during the review process, without affecting the quality and conclusions of published manuscripts. In addition, we recognize the need of adopting the recommendations made by NCCD and other similar scientific guiding entities.

10.
Stem Cells Dev ; 21(7): 1016-46, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22204396

RESUMEN

A complex set of extrinsic and intrinsic signals acts in specific temporal and spatial orders to enable neural differentiation during development. These processes have been extensively studied in animal models, but human neural development remains much less understood. This lack of detailed information about human early neurogenesis is a hindrance for the differentiation of pluripotent stem cell lines into specific neuronal phenotypes. Therefore, it is important to strengthen the interspecies comparative approaches. We describe a novel model system in which in vitro differentiation of human and mouse embryonic stem (ES) cells are temporally aligned to each other and compared with mouse telencephalic neurogenesis in vivo. In this comparative model system, we tested the in vitro role of Hedgehog (Hh) signaling for ES cell-derived telencephalic differentiation. In vivo, Hh signaling mediates dorsal/ventral patterning during early stages of telencephalic development. We monitored the effect of pharmacological modulators of the Hh signaling pathway, purmorphamine-an agonist and cyclopamine-an antagonist of the Smoothened receptor (Smo), on the expression of region-specific transcription factors and signaling molecules relevant for telencephalic development in vivo. Purmorphamine strongly upregulated the expression of telencephalic ventral markers Nkx2.1, Nkx6.2, Lhx6, and Lhx8 in mouse and human cells, thus reflecting the in vivo process of the medial ganglionic eminence patterning and specification. Cyclopamine upregulated the expression of telencephalic dorsal markers, but at lower levels in human compared with mouse cells. Modulation of Smo in vitro differentially affected, in mouse and human cells, the expression of molecules of the Hh pathway, especially the Gli1 and Gli3 effectors, Sonic Hh ligand and Ptch receptors. These results provide evidence for the different default differentiation of mouse and human ES cells and prove the utility of the comparative system for optimizing the directed differentiation of human pluripotent stem cells.


Asunto(s)
Tipificación del Cuerpo , Células Madre Embrionarias/fisiología , Proteínas Hedgehog/metabolismo , Morfolinas/farmacología , Purinas/farmacología , Telencéfalo/citología , Alcaloides de Veratrum/farmacología , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Células Cultivadas , Células Madre Embrionarias/metabolismo , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Humanos , Ratones , Neurogénesis , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal , Receptor Smoothened , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
PLoS One ; 7(11): e50178, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209667

RESUMEN

Evidence from carefully conducted open label clinical trials suggested that therapeutic benefit can be achieved by grafting fetal dopaminergic (DAergic) neurons derived from ventral mesencephalon (VM) into the denervated striatum of Parkinson's disease (PD) patients. However, two double-blind trials generated negative results reporting deleterious side effects such as prominent dyskinesias. Heterogeneous composition of VM grafts is likely to account for suboptimal clinical efficacy.We consider that gene expression patterns of the VM tissue needs to be better understood by comparing the genetic signature of the surviving and functioning grafts with the cell suspensions used for transplantation. In addition, it is crucial to assess whether the grafted cells exhibit the DAergic phenotype of adult substantia nigra pars compacta (SNpc). To investigate this further, we used a GFP reporter mouse as source of VM tissue that enabled the detection and dissection of the grafts 6 weeks post implantation. A comparative gene expression analysis of the VM cell suspension and grafts revealed that VM grafts continue to differentiate post-implantation. In addition, implanted grafts showed a mature SNpc-like molecular DAergic phenotype with similar expression levels of TH, Vmat2 and Dat. However, by comparing gene expression of the adult SNpc with dissected grafts we detected a higher expression of progenitor markers in the grafts. Finally, when compared to the VM cell suspension, post-grafting there was a higher expression of markers inherent to glia and other neuronal populations.In summary, our data highlight the dynamic development of distinctive DAergic and non-DAergic gene expression markers associated with the maturation of VM grafts in vivo. The molecular signature of VM grafts and its functional relevance should be further explored in future studies aimed at the optimization of DAergic cell therapy approaches in PD.


Asunto(s)
Mesencéfalo/efectos de los fármacos , Mesencéfalo/embriología , Oxidopamina/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Adrenérgicos/farmacología , Anfetaminas/farmacología , Animales , Trasplante de Células/métodos , Pollos , Discinesias/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Marcadores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Neuroglía/metabolismo , Neuronas/metabolismo , Fenotipo , Ratas , Ratas Wistar , Células Madre/citología , Sustancia Negra/embriología , Sustancia Negra/metabolismo , Factores de Tiempo
12.
Stem Cells Dev ; 20(6): 947-58, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21166522

RESUMEN

Understanding the normal development of individual neural subtypes provides an essential framework for the design of rational approaches to embryonic stem cell differentiation for in vitro studies and cell replacement therapies. Of particular interest and a particular challenge are the cells that build-up the telencephalon. Recent research has unraveled key developmental mechanisms contributing to the generation of specific telencephalic cells. We focus on morphogens and transcription factors known to regulate distinct developmental processes. These include early anterior/posterior patterning, dorsal/ventral patterning, and generation of progenitor domains and neuronal specification into major classes of telencephalic cells: glutamatergic projection neurons, different subtypes of γ-aminobutyric acid-ergic interneurons and projection neurons, as well as cholinergic interneurons and projection neurons. Based on a comparison with in vivo telencephalic neurogenesis, we propose that the specific combinations of transcription factors expressed during development can serve as milestones for the in vitro differentiation of embryonic stem cells toward specific telencephalic neurons.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Neurogénesis , Neuronas/citología , Telencéfalo/citología , Animales , Humanos , Ratones , Cicatrización de Heridas
14.
Glia ; 55(4): 385-99, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17152062

RESUMEN

The great potential of human embryonic stem (hES) cells offers the opportunity both for studying basic developmental processes in vitro as well as for drug screening, modeling diseases, or future cell therapy. Defining protocols for the generation of human neural progenies represents a most important prerequisite. Here, we have used six hES cell lines to evaluate defined conditions for neural differentiation in suspension and adherent culture systems. Our protocol does not require fetal serum, feeder cells, or retinoic acid at any step, to induce neural fate decisions in hES cells. We monitored neurogenesis in differentiating cultures using morphological (including on-line follow up), immunocytochemical, and RT-PCR assays. For each hES cell line, in suspension or adherent culture, the same longitudinal progression of neural differentiation occurs. We showed the dynamic transitions from hES cells to neuroepithelial (NE) cells, to radial glial (RG) cells, and to neurons. Thus, 7 days after neural induction the majority of cells were NE, expressing nestin, Sox1, and Pax6. During neural proliferation and differentiation, NE cells transformed in RG cells, which acquired vimentin, BLBP, GLAST, and GFAP, proliferated and formed radial scaffolds. gamma-Aminobutyric acid (GABA)-positive and glutamate positive neurons, few oligodendrocyte progenitors and astrocytes were formed in our conditions and timing. Our system successfully generates human RG cells and could be an effective source for neuronal replacement, since RG cells predominantly generate neurons and provide them with support and guidance.


Asunto(s)
Células Madre Embrionarias/fisiología , Células Epiteliales/fisiología , Neuroglía/fisiología , Adhesión Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular , Medio de Cultivo Libre de Suero , Cartilla de ADN , Humanos , Inmunohistoquímica , Proteínas del Tejido Nervioso/biosíntesis , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Cell Mol Med ; 6(2): 223-34, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12169207

RESUMEN

Dendritic cells (DC) are cells of the hematopoietic system specialized in capturing antigens and initiating T cell-mediated immune responses. We show here that human DC generated from adherent peripheral blood mononuclear cells (PBMC) after in vitro stimulation with granulocyte macrophage colony stimulating factor (GM-CSF) and interleukin-4 (IL-4) express Fas antigen (APO-1, CD95) and can undergo apoptosis upon triggering of Fas by monoclonal antibodies. Immature monocytes-derived dendritic cells (MDDC) upregulate CD86 and HLA-DR expression and develop dendrites and veiled processes. Flow cytometry analysis revealed CD95 expression in approx. 40% of these MDDC and incubation with anti-CD95 mAb (0.5 microg/ml) induced apoptosis when compared to untreated controls. The extent of apoptosis induced by the agonist anti-Fas antibody strongly related to the percentage of cells expressing CD 95. Upon tumor necrosis factor alpha (TNF-alpha) additional stimulation, MDDC assumed a characteristic mature dendritic cells morphology showing prolonged veils, CD83 expression, and high levels of HLA-DR. These cells have downregulated their Fas receptors (to approx. 20%) and undergo apoptosis to a lesser extent when treated with anti-CD 95, as demonstrated by the hardly noticeable effect of this antibody on the viability of cultured cells as compared to controls. Thus, upon TNF-alpha induced maturation, MDDC became resistant to Fas-induced apoptosis. The apoptotic episodes surrounding the earlier stage of DC differentiation appeared to be mediated by Fas. In contrast, a Fas independent pathway is probably responsible for the apoptotic events associated with terminally differentiated DC.


Asunto(s)
Apoptosis/inmunología , Células Dendríticas/fisiología , Monocitos/fisiología , Receptor fas/fisiología , Anticuerpos Monoclonales/farmacología , Antígenos CD/metabolismo , Antígenos de Superficie/metabolismo , Apoptosis/efectos de los fármacos , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Antígenos HLA-DR/metabolismo , Humanos , Interleucina-4/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba , Receptor fas/inmunología , Receptor fas/farmacología
16.
J Cell Mol Med ; 6(3): 415-25, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12417058

RESUMEN

Dendritic cells (DCs) in the rheumatoid arthritis (RA) joint mediate the immunopathological process and act as a potent antigen presenting cell. We compared the expression of co-stimulatory and adhesion molecules on DCs in RA patients versus controls with traumatic joint lesions and evaluated the correlation between the immunophenotypical presentation of DCs and the clinical status of the disease. Samples of peripheral venous blood, synovial fluid (SF) and synovial tissue (ST) were obtained from 10 patients with RA at the time of hip or knee replacement and from 9 control patients with knee arthroscopy for traumatic lesions. Clinical status was appreciated using the DAS28 score. Blood, SF and dissociated ST cell populations were separated by centrifugation and analyzed by flow cytometry. Cells phenotypes were identified using three-color flow cytometry analysis for the following receptors HLA-DR, CD80, CD83, CD86, CD11c, CD18, CD54, CD58, CD3, CD4, CD8, CD19, CD20, CD14, CD16, CD56. HLA-DR molecules, co-stimulatory receptors CD80, CD86, CD83 and adhesion molecules CD18, CD11c, CD54, CD58, were analyzed by two-color immunofluorescence microscopy on ST serial sections. In patients with active RA (DAS28>5.1) we found a highly differentiated subpopulation of DCs in the ST and SF that expressed an activated phenotype (HLA-DR, CD86+, CD80+, CD83+, CD11c+, CD54+, CD58+). No differences were found between circulating DCs from RA patients and control patients. Our data suggest an interrelationship between clinical outcome and the immunophenotypical presentation of DCs. Clinical active RA (DAS28>5.1) is associated with high incidence of activated DCs population in the ST and SF as demonstrated by expression of adhesion and co-stimulatory molecules.


Asunto(s)
Antígenos CD/metabolismo , Artritis Reumatoide/inmunología , Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/metabolismo , Adulto , Antígenos de Diferenciación , Artritis Reumatoide/patología , Artroplastia de Reemplazo de Cadera , Artroplastia de Reemplazo de Rodilla , Diferenciación Celular , Células Dendríticas/inmunología , Femenino , Citometría de Flujo , Articulación de la Cadera/inmunología , Articulación de la Cadera/patología , Humanos , Articulación de la Rodilla/inmunología , Articulación de la Rodilla/patología , Masculino , Persona de Mediana Edad , Rumanía , Líquido Sinovial/citología , Líquido Sinovial/inmunología
17.
Rom J Intern Med ; 41(3): 255-67, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-15526509

RESUMEN

BACKGROUND: Dendritic cells (DCs) are potent antigen-presenting cells (APC) that are deeply implicated in the initiation and exacerbation of rheumatoid arthritis (RA). Active RA is associated with an activated DCs population as demonstrated by high expression of adhesion and co-stimulatory molecules. PURPOSE: To compare the expression of adhesion and co-stimulatory molecules on DCs from synovial tissue (ST) in patients (pts) with RA and the clinical status before and after treatment with disease modifying antirheumatic drugs (DMARDs). METHODS: Samples of ST were obtained from RA patients at the time of hip or knee replacement or arthroscopy. Clinical status (assessed by the American College of Rheumatology - ACR - core set and the DAS28) and co-stimulatory and adhesion molecules on DCs were evaluated before and after treatment. Immunophenotype of DCs was analyzed by two-color immunofluorescence microscopy. RESULTS: In patients with active RA we found a highly differentiated subpopulation of DCs that expressed an activated phenotype. After treatment, the expression of adhesion and co-stimulatory molecules on ST DCs was correlated with the ACR and DAS28 clinical response. CONCLUSION: Clinical outcome and the immunophenotypical presentation of ST DCs after DMARDs treatment are closely correlated in pts with RA. The co-stimulatory activity in the synovium is important in determining the course of the disease and provide new therapeutic targets for immune intervention.


Asunto(s)
Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Células Dendríticas/inmunología , Adulto , Anciano , Antirreumáticos/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/inmunología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA