Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 2935, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37217509

RESUMEN

Resistance to glucocorticoids (GC) is associated with an increased risk of relapse in B-cell progenitor acute lymphoblastic leukemia (BCP-ALL). Performing transcriptomic and single-cell proteomic studies in healthy B-cell progenitors, we herein identify coordination between the glucocorticoid receptor pathway with B-cell developmental pathways. Healthy pro-B cells most highly express the glucocorticoid receptor, and this developmental expression is conserved in primary BCP-ALL cells from patients at diagnosis and relapse. In-vitro and in vivo glucocorticoid treatment of primary BCP-ALL cells demonstrate that the interplay between B-cell development and the glucocorticoid pathways is crucial for GC resistance in leukemic cells. Gene set enrichment analysis in BCP-ALL cell lines surviving GC treatment show enrichment of B cell receptor signaling pathways. In addition, primary BCP-ALL cells surviving GC treatment in vitro and in vivo demonstrate a late pre-B cell phenotype with activation of PI3K/mTOR and CREB signaling. Dasatinib, a multi-kinase inhibitor, most effectively targets this active signaling in GC-resistant cells, and when combined with glucocorticoids, results in increased cell death in vitro and decreased leukemic burden and prolonged survival in an in vivo xenograft model. Targeting the active signaling through the addition of dasatinib may represent a therapeutic approach to overcome GC resistance in BCP-ALL.


Asunto(s)
Linfoma de Burkitt , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Humanos , Glucocorticoides/farmacología , Glucocorticoides/uso terapéutico , Dasatinib/farmacología , Dasatinib/uso terapéutico , Receptores de Glucocorticoides/genética , Apoptosis , Proteómica , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Recurrencia , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral
2.
Nat Commun ; 13(1): 934, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177627

RESUMEN

The increasing use of mass cytometry for analyzing clinical samples offers the possibility to perform comparative analyses across public datasets. However, challenges in batch normalization and data integration limit the comparison of datasets not intended to be analyzed together. Here, we present a data integration strategy, CytofIn, using generalized anchors to integrate mass cytometry datasets from the public domain. We show that low-variance controls, such as healthy samples and stable channels, are inherently homogeneous, robust against stimulation, and can serve as generalized anchors for batch correction. Single-cell quantification comparing mass cytometry data from 989 leukemia files pre- and post normalization with CytofIn demonstrates effective batch correction while recapitulating the gold-standard bead normalization. CytofIn integration of public cancer datasets enabled the comparison of immune features across histologies and treatments. We demonstrate the ability to integrate public datasets without necessitating identical control samples or bead standards for fast and robust analysis using CytofIn.


Asunto(s)
Algoritmos , Conjuntos de Datos como Asunto , Citometría de Flujo/métodos , Melanoma/tratamiento farmacológico , Biología Computacional/métodos , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Melanoma/patología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Análisis de la Célula Individual , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología
3.
Anal Chim Acta ; 1184: 339023, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34625255

RESUMEN

Total triacylglycerol (TAG) level is a key clinical marker of metabolic and cardiovascular diseases. However, the roles of individual TAGs have not been thoroughly explored in part due to their extreme structural complexity. We present a targeted mass spectrometry-based method combining multiple reaction monitoring (MRM) and multiple stage mass spectrometry (MS3) for the comprehensive qualitative and semiquantitative profiling of TAGs. This method referred as TriP-MS3 - triacylglycerol profiling using MS3 - screens for more than 6,700 TAG species in a fully automated fashion. TriP-MS3 demonstrated excellent reproducibility (median interday CV âˆ¼ 0.15) and linearity (median R2 = 0.978) and detected 285 individual TAG species in human plasma. The semiquantitative accuracy of the method was validated by comparison with a state-of-the-art reverse phase liquid chromatography (RPLC)-MS (R2 = 0.83), which is the most commonly used approach for TAGs profiling. Finally, we demonstrate the utility and the versatility of the method by characterizing the effects of a fatty acid desaturase inhibitor on TAG profiles in vitro and by profiling TAGs in Caenorhabditis elegans.


Asunto(s)
Cromatografía de Fase Inversa , Plasma , Humanos , Espectrometría de Masas , Reproducibilidad de los Resultados , Triglicéridos
5.
Cytometry A ; 97(8): 782-799, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32602650

RESUMEN

The application of machine learning and artificial intelligence to high-dimensional cytometry data sets has increasingly become a staple of bioinformatic data analysis over the past decade. This is especially true in the field of cancer biology, where protocols for collecting multiparameter single-cell data in a high-throughput fashion are rapidly developed. As the use of machine learning methodology in cytometry becomes increasingly common, there is a need for cancer biologists to understand the basic theory and applications of a variety of algorithmic tools for analyzing and interpreting cytometry data. We introduce the reader to several keystone machine learning-based analytic approaches with an emphasis on defining key terms and introducing a conceptual framework for making translational or clinically relevant discoveries. The target audience consists of cancer cell biologists and physician-scientists interested in applying these tools to their own data, but who may have limited training in bioinformatics. © 2020 International Society for Advancement of Cytometry.


Asunto(s)
Inteligencia Artificial , Neoplasias , Biología Computacional , Humanos , Aprendizaje Automático , Neoplasias/diagnóstico , Proteómica
6.
Nat Commun ; 11(1): 2795, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493933

RESUMEN

The transcription factor JUN is highly expressed in pulmonary fibrosis. Its induction in mice drives lung fibrosis, which is abrogated by administration of anti-CD47. Here, we use high-dimensional mass cytometry to profile protein expression and secretome of cells from patients with pulmonary fibrosis. We show that JUN is activated in fibrotic fibroblasts that expressed increased CD47 and PD-L1. Using ATAC-seq and ChIP-seq, we found that activation of JUN rendered promoters and enhancers of CD47 and PD-L1 accessible. We further detect increased IL-6 that amplified JUN-mediated CD47 enhancer activity and protein expression. Using an in vivo mouse model of fibrosis, we found two distinct mechanisms by which blocking IL-6, CD47 and PD-L1 reversed fibrosis, by increasing phagocytosis of profibrotic fibroblasts and by eliminating suppressive effects on adaptive immunity. Our results identify specific immune mechanisms that promote fibrosis and suggest a therapeutic approach that could be used alongside conventional anti-fibrotics for pulmonary fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Inmunidad , Proteínas Proto-Oncogénicas c-jun/metabolismo , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Animales , Antígeno B7-H1/metabolismo , Lavado Broncoalveolar , Antígeno CD47/metabolismo , Fibroblastos/patología , Humanos , Terapia de Inmunosupresión , Interleucina-6/metabolismo , Macrófagos/metabolismo , Ratones , Fenotipo , Linfocitos T/inmunología
7.
Stem Cell Reports ; 14(4): 603-613, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32197115

RESUMEN

Osteoporosis and osteoporotic fractures lead to decreased life quality and high healthcare costs. Current treatments prevent losses in bone mass and fractures to some extent but have side effects. Therefore, better therapies are needed. This study investigated whether the transcription factor Jun has a specific pro-osteogenic potency and whether modulating Jun could serve as a novel treatment for osteoporosis-associated fractures. We demonstrate that ectopically transplanted whole bones and distinct osteoprogenitors increase bone formation. Perinatal Jun induction disturbs growth plate architecture, causing a striking phenotype with shortened and thickened bones. Molecularly, Jun induces hedgehog signaling in skeletal stem cells. Therapeutically, Jun accelerates bone growth and healing in a drilling-defect model. Altogether, these results demonstrate that Jun drives bone formation by expanding osteoprogenitor populations and forcing them into the bone fate, providing a rationale for future clinical applications.


Asunto(s)
Huesos/patología , Fracturas Osteoporóticas/metabolismo , Fracturas Osteoporóticas/patología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Células Madre/metabolismo , Animales , Desarrollo Óseo , Trasplante Óseo , Diferenciación Celular , Proliferación Celular , Curación de Fractura , Placa de Crecimiento/metabolismo , Proteínas Hedgehog/metabolismo , Ratones , Fenotipo , Transducción de Señal
8.
Blood Adv ; 3(19): 2825-2835, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31582391

RESUMEN

Chromosomal rearrangements involving the mixed lineage leukemia (MLL) gene, also known as KMT2A, are often observed in human leukemias and are generally associated with a poor prognosis. To model these leukemias, we applied clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing to induce MLL chromosomal rearrangements in human hematopoietic stem and progenitor cells purified from umbilical cord blood. Electroporation of ribonucleoprotein complexes containing chemically modified synthetic single guide RNAs and purified Cas9 protein induced translocations between chromosomes 9 and 11 [t(9;11)] at an efficiency >1%. Transplantation of gene-edited cells into immune-compromised mice rapidly induced acute leukemias of different lineages and often with multiclonal origins dictated by the duration of in vitro culture prior to transplantation. Breakpoint junction sequences served as biomarkers to monitor clonal selection and progression in culture and in vivo. High-dimensional cell surface and intracellular protein analysis by mass cytometry (CyTOF) revealed that gene-edited leukemias recapitulated disease-specific protein expression observed in human patients and showed that MLL-rearranged (MLLr) mixed phenotype acute leukemias (MPALs) were more similar to acute myeloid leukemias (AMLs) than to acute lymphoblastic leukemias (ALLs). Therefore, highly efficient generation of MLL chromosomal translocations in primary human blood stem cells using CRISPR/Cas9 reliably models human acute MLLr leukemia and provides an experimental platform for basic and translational studies of leukemia biology and therapeutics.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Edición Génica/métodos , Leucemia Mieloide Aguda/genética , Células Madre/metabolismo , Translocación Genética/genética , Animales , Humanos , Ratones
9.
Stem Cells Int ; 2019: 7692973, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31531025

RESUMEN

Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are dynamic cells that can sense the environment, adapting their regulatory functions to different conditions. Accordingly, the therapeutic potential of BM-MSCs can be modulated by preconditioning strategies aimed at modifying their paracrine action. Although rat BM-MSCs (rBM-MSCs) have been widely tested in preclinical research, most preconditioning studies have employed human and mouse BM-MSCs. Herein, we investigated whether rBM-MSCs modify their phenotype and paracrine functions in response to Toll-like receptor (TLR) agonists. The data showed that rBM-MSCs expressed TLR3, TLR4, and MDA5 mRNA and were able to internalize polyinosinic-polycytidylic acid (Poly(I:C)), a TLR3/MDA5 agonist. rBM-MSCs were then stimulated with Poly(I:C) or with lipopolysaccharide (LPS, a TLR4 agonist) for 1 h and were grown under normal culture conditions. LPS or Poly(I:C) stimulation did not affect the viability or the morphology of rBM-MSCs and did not modify the expression pattern of key cell surface markers. Poly(I:C) did not induce statistically significant changes in the release of several inflammatory mediators and VEGF by rBM-MSCs, although it tended to increase IL-6 and MCP-1 secretion, whereas LPS increased the release of IL-6, MCP-1, and VEGF, three factors that were constitutively secreted by unstimulated cells. The neurotrophic activity of the conditioned medium from unstimulated and LPS-preconditioned rBM-MSCs was investigated using dorsal root ganglion explants, showing that soluble factors produced by unstimulated and LPS-preconditioned rBM-MSCs can stimulate neurite outgrowth similarly, in a VEGF-dependent manner. LPS-preconditioned cells, however, were slightly more efficient in increasing the number of regrowing axons in a model of sciatic nerve transection in rats. In conclusion, LPS preconditioning boosted the production of constitutively secreted factors by rBM-MSCs, without changing their mesenchymal identity, an effect that requires further investigation in exploratory preclinical studies.

10.
Stem Cell Res ; 39: 101490, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31301488

RESUMEN

Induced pluripotent stem cell (iPSC) line were generated from erythroblasts of a Brazilian patient with familiar form of amyotrophic lateral sclerosis (ALS). NGS analysis demonstrated that patient carried a mutation in SOD1 gene, as well as a deletion in FUS gene. CytoTune™-iPS 2.0 Sendai Reprogramming Kit (containing the reprogramming factors OCT3/4, KLF4, SOX2 and cMYC) was used to generate the cell lines. The iPSCs express pluripotency markers, have normal karyotype and differentiated spontaneously in the three germ layers. The expression of Sendai virus was lost in all iPSC lines after 15 passages.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Esclerosis Amiotrófica Lateral/metabolismo , Brasil , Línea Celular , Humanos , Cariotipo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Mutación/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Factores de Transcripción SOXB1/metabolismo , Superóxido Dismutasa-1/genética
11.
Stem Cell Res ; 37: 101448, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31077962

RESUMEN

Induced pluripotent stem cell (iPSC) lines were generated from erythroblasts of two patients with amyotrophic lateral sclerosis (ALS) and two healthy individuals. One familial and one sporadic ALS patients were used, both with genetic alterations in VAPB gene. CytoTune™-iPS 2.0 Sendai Reprogramming Kit (containing the reprogramming factors OCT3/4, KLF4, SOX2 and cMYC) was used to generate the iPSC cell lines. The four iPSCs express pluripotency markers, have normal karyotype and differentiated spontaneously in the three germ layers. The expression of Sendai virus was lost in all iPSC lines after 15 passages.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Diferenciación Celular , Reprogramación Celular , Células Madre Pluripotentes Inducidas/patología , Leucocitos Mononucleares/patología , Mutación , Proteínas de Transporte Vesicular/genética , Adulto , Esclerosis Amiotrófica Lateral/patología , Células Cultivadas , Voluntarios Sanos , Heterocigoto , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Leucocitos Mononucleares/metabolismo , Masculino , Fenotipo
12.
PLoS One ; 14(1): e0210207, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30629659

RESUMEN

The process of neuronal differentiation is associated with neurite elongation and membrane biogenesis, and phosphatidylcholine (PtdCho) is the major membrane phospholipid in mammalian cells. During neuroblast differentiation, the transcription of two genes involved in PtdCho biosynthesis are stimulated: Chka gene for choline kinase (CK) alpha isoform and Pcyt1a gene for CTP:phosphocholine cytidylyltransferase (CCT) alpha isoform. Here we show that CKα is essential for neuronal differentiation. In addition, we demonstrated that KDM2B regulates CKα expression and, as a consequence, neuronal differentiation. This factor is up-regulated in the course of the neuroblasts proliferative and undifferentiated state and down-regulated during differentiation induced by retinoic acid (RA). During proliferation, KDM2B binds to the Box2 located in the Chka promoter repressing its transcription. Interestingly, KDM2B knockdown enhances the levels of CKα expression in neuroblast cells and induces neuronal differentiation even in the absence of RA. These results suggest that KDM2B is required for the appropriate regulation of CKα during neuronal differentiation and to the maintaining of the undifferentiated stage of neuroblast cells.


Asunto(s)
Colina Quinasa/genética , Proteínas F-Box/metabolismo , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/metabolismo , Neuroblastoma/genética , Tretinoina/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Colina Quinasa/metabolismo , Epigénesis Genética , Proteínas F-Box/genética , Estudios de Seguimiento , Técnicas de Silenciamiento del Gen , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Células-Madre Neurales/fisiología , Neuroblastoma/mortalidad , Neuroblastoma/patología , Pronóstico , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba
13.
J Neurosci Methods ; 313: 1-5, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30389488

RESUMEN

BACKGROUND: Animal models of optic nerve injury are often used to study central nervous system (CNS) degeneration and regeneration, and targeting the optic nerve is a powerful approach for axon-protective or remyelination therapy. However, the experimental delivery of drugs or cells to the optic nerve is rarely performed because injections into this structure are difficult in small animals, especially in mice. NEW METHOD: We investigated and developed methods to deliver drugs or cells to the mouse optic nerve through 3 different routes: a) intraorbital, b) through the optic foramen and c) transcranial. RESULTS: The methods targeted different parts of the mouse optic nerve: intraorbital proximal (intraorbital), intracranial middle (optic-foramen) or intracranial distal (transcranial) portion. COMPARISON WITH EXISTING METHODS: Most existing methods target the optic nerve indirectly. For instance, intravitreally delivered cells often cannot cross the inner limiting membrane to reach retinal neurons and optic nerve axons. Systemic delivery, eye drops and intraventricular injections do not always successfully target the optic nerve. Intraorbital and transcranial injections into the optic nerve or chiasm have been performed but these methods have not been well described. We approached the optic nerve with more selective and precise targeting than existing methods. CONCLUSIONS: We successfully targeted the murine optic nerve intraorbitally, through the optic foramen, and transcranially. Of all methods, the injection through the optic foramen is likely the most innovative and fastest. These methods offer additional approaches for therapeutic intervention to be used by those studying white matter damage and axonal regeneration in the CNS.


Asunto(s)
Modelos Animales de Enfermedad , Inyecciones/métodos , Nervio Óptico/efectos de los fármacos , Órbita , Base del Cráneo , Animales , Ratones , Ratones Endogámicos C57BL
14.
Sci Rep ; 7(1): 6171, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28733584

RESUMEN

Chagas disease cardiomyopathy is a parasite-driven inflammatory disease to which there are no effective treatments. Here we evaluated the therapeutic potential of N,N-dimethylsphingosine(DMS), which blocks the production of sphingosine-1-phosphate(S1P), a mediator of cellular events during inflammatory responses, in a model of chronic Chagas disease cardiomyopathy. DMS-treated, Trypanosoma cruzi-infected mice had a marked reduction of cardiac inflammation, fibrosis and galectin-3 expression when compared to controls. Serum concentrations of galectin-3, IFNγ and TNFα, as well as cardiac gene expression of inflammatory mediators were reduced after DMS treatment. The gene expression of M1 marker, iNOS, was decreased, while the M2 marker, arginase1, was increased. DMS-treated mice showed an improvement in exercise capacity. Moreover, DMS caused a reduction in parasite load in vivo. DMS inhibited the activation of lymphocytes, and reduced cytokines and NO production in activated macrophage cultures in vitro, while increasing IL-1ß production. Analysis by qRT-PCR array showed that DMS treatment modulated inflammasome activation induced by T. cruzi on macrophages. Altogether, our results demonstrate that DMS, through anti-parasitic and immunomodulatory actions, can be beneficial in the treatment of chronic phase of T. cruzi infection and suggest that S1P-activated processes as possible therapeutic targets for the treatment of Chagas disease cardiomyopathy.


Asunto(s)
Arginasa/genética , Cardiomiopatía Chagásica/tratamiento farmacológico , Inhibidores Enzimáticos/administración & dosificación , Óxido Nítrico Sintasa de Tipo II/genética , Esfingosina/análogos & derivados , Animales , Cardiomiopatía Chagásica/genética , Cardiomiopatía Chagásica/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Galectina 3/sangre , Regulación de la Expresión Génica/efectos de los fármacos , Interferón gamma/sangre , Activación de Linfocitos/efectos de los fármacos , Ratones , Carga de Parásitos , Esfingosina/administración & dosificación , Esfingosina/farmacología , Trypanosoma cruzi/efectos de los fármacos , Factor de Necrosis Tumoral alfa/sangre
15.
Nucleic Acids Res ; 44(9): 4163-73, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-26773060

RESUMEN

G-quadruplexes are dynamic structures folded in G-rich single-stranded DNA regions. These structures have been recognized as a potential nucleic acid based mechanism for regulating multiple cellular processes such as replication, transcription and genomic maintenance. So far, their transcriptional role in vivo during vertebrate embryonic development has not yet been addressed. Here, we performed an in silico search to find conserved putative G-quadruplex sequences (PQSs) within proximal promoter regions of human, mouse and zebrafish developmental genes. Among the PQSs able to fold in vitro as G-quadruplex, those present in nog3, col2a1 and fzd5 promoters were selected for further studies. In cellulo studies revealed that the selected G-quadruplexes affected the transcription of luciferase controlled by the SV40 nonrelated promoter. G-quadruplex disruption in vivo by microinjection in zebrafish embryos of either small ligands or DNA oligonucleotides complementary to the selected PQSs resulted in lower transcription of the targeted genes. Moreover, zebrafish embryos and larvae phenotypes caused by the presence of complementary oligonucleotides fully resembled those ones reported for nog3, col2a1 and fzd5 morphants. To our knowledge, this is the first work revealing in vivo the role of conserved G-quadruplexes in the embryonic development, one of the most regulated processes of the vertebrates biology.


Asunto(s)
G-Cuádruplex , Regulación del Desarrollo de la Expresión Génica , Transcripción Genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , ADN de Cadena Simple , Embrión no Mamífero/metabolismo , Humanos , Ratones , Regiones Promotoras Genéticas , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
16.
Mol Neurobiol ; 53(9): 6316-6331, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26567110

RESUMEN

Neuronal differentiation plays a key role during embryogenesis. However, based on the capacity of neuronal stem cells to either generate or regenerate neurons and because differentiation stops aberrant neuroblasts proliferation, neuronal differentiation is crucial during neuropathological conditions. Although phosphatidylcholine (PtdCho) has been proposed as an important molecule for neurite growth and neuronal regeneration, the identity of the molecular target has remained elusive. This study originally describes that lysophosphatidylcholine (LPtdCho), either exogenously supplied or generated by the imbalance of PtdCho metabolism through the enzymatic action of cytosolic phospholipase A2, acts as a neurotrophic-like factor. We demonstrated that LPtdCho induces neuronal differentiation by activation of the small G protein Ras followed by the Raf/MEK/ERK signaling pathway. Accordingly, LPtdCho redirects neuroblasts gene expression leading to the generation of functional mature neurons expressing ßIII-tubulin and having increased acetylcholinesterase activity and membrane biosynthesis required for neuritogenesis. These findings provide mechanistic details of the role of cytidine-5-diphosphocholine (CDP-choline) and PtdCho as neuroprotectors. Furthermore, as LPtdCho recapitulates the effect of the therapeutic agent retinoic acid, these results open new avenues for drug discovery for the treatment of neuropathological conditions.


Asunto(s)
Linaje de la Célula , Lisofosfatidilcolinas/farmacología , Neuronas/citología , Neuronas/metabolismo , Animales , Biomarcadores/metabolismo , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Linaje de la Célula/efectos de los fármacos , Humanos , Lisofosfatidilcolinas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/enzimología , Fosfatidilcolinas/metabolismo , Fosfolipasas A2 Citosólicas/metabolismo , Sistemas de Mensajero Secundario , Tretinoina/farmacología , Proteínas ras/metabolismo
17.
Toxicol Appl Pharmacol ; 287(2): 178-190, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26049102

RESUMEN

The cAMP pathway is a universal signaling pathway regulating many cellular processes including metabolic routes, growth and differentiation. However, its effects on xenobiotic biotransformation and transport systems are poorly characterized. The effect of cAMP on expression and activity of GST and MRP2 was evaluated in Caco-2 cells, a model of intestinal epithelium. Cells incubated with the cAMP permeable analog dibutyryl cyclic AMP (db-cAMP: 1,10,100 µM) for 48 h exhibited a dose-response increase in GST class α and MRP2 protein expression. Incubation with forskolin, an activator of adenylyl cyclase, confirmed the association between intracellular cAMP and upregulation of MRP2. Consistent with increased expression of GSTα and MRP2, db-cAMP enhanced their activities, as well as cytoprotection against the common substrate 1-chloro-2,4-dinitrobenzene. Pretreatment with protein kinase A (PKA) inhibitors totally abolished upregulation of MRP2 and GSTα induced by db-cAMP. In silico analysis together with experiments consisting of treatment with db-cAMP of Caco-2 cells transfected with a reporter construct containing CRE and AP-1 sites evidenced participation of these sites in MRP2 upregulation. Further studies involving the transcription factors CREB and AP-1 (c-JUN, c-FOS and ATF2) demonstrated increased levels of total c-JUN and phosphorylation of c-JUN and ATF2 by db-cAMP, which were suppressed by a PKA inhibitor. Co-immunoprecipitation and ChIP assay studies demonstrated that db-cAMP increased c-JUN/ATF2 interaction, with further recruitment to the region of the MRP2 promoter containing CRE and AP-1 sites. We conclude that cAMP induces GSTα and MRP2 expression and activity in Caco-2 cells via the PKA pathway, thus regulating detoxification of specific xenobiotics.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Glutatión Transferasa/biosíntesis , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Proteína de Unión a CREB/metabolismo , Células CACO-2 , Colforsina/farmacología , Dinitroclorobenceno/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Factor de Transcripción AP-1/metabolismo
18.
Biochim Biophys Acta ; 1841(4): 544-51, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24440820

RESUMEN

Neuronal differentiation is a complex process characterized by a halt in proliferation and extension of neurites from the cell body. This process is accompanied by changes in gene expression that mediate the redirection leading to neurite formation and function. Acceleration of membrane phospholipids synthesis is associated with neurite elongation, and phosphatidylcholine (PtdCho) is the major membrane phospholipid in mammalian cells. The transcription of two genes in particular encoding key enzymes in the CDP-choline pathway for PtdCho biosynthesis are stimulated; the Chka gene for choline kinase (CK) alpha isoform and the Pcyt1a gene for the CTP:phosphocholine cytidylyltransferase (CCT) alpha isoform. We report that the stimulation of CKα expression during retinoic acid (RA) induced differentiation depends on a promoter region that contains two CCAAT/Enhancer-binding Protein-ß (C/EBPß) sites. We demonstrate that during neuronal differentiation of Neuro-2a cells, RA induces Chka expression by a mechanism that involves ERK1/2 activation which triggers C/EBPß expression. Elevated levels of C/EBPß bind to the Chka proximal promoter (Box1) inducing CKα expression. In addition we identified a downstream sequence named Box2 which together with Box1 is required for the promoter to reach the full induction. This is the first elucidation of the mechanism by which the expression of Chka is coordinately regulated during neuronal differentiation.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Diferenciación Celular/efectos de los fármacos , Neuronas/metabolismo , Fosfolípidos/biosíntesis , Animales , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Proliferación Celular , Colina Quinasa/biosíntesis , Colina Quinasa/metabolismo , Citidililtransferasa de Colina-Fosfato/genética , Citidililtransferasa de Colina-Fosfato/metabolismo , Humanos , Ratones , Neuritas/metabolismo , Neuronas/citología , Fosfatidilcolinas/metabolismo , Fosfolípidos/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Tretinoina/farmacología
19.
IUBMB Life ; 63(9): 714-20, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21818839

RESUMEN

Neuronal differentiation is characterized by neuritogenesis and neurite outgrowth, processes, which are critically dependent on membrane biosynthesis, and therefore, on the expression and regulation of enzymes involved in phospholipid biosynthesis. During the last decade a great effort was made to clarify where membrane lipids are synthesized, how the newly synthesized membrane components reach the membrane and are inserted during neuritogenesis and to elucidate the mechanism by which the supply of new membrane components is coordinated with the demand for growth. Phosphatidylcholine is the principal and essential component for mammalian membranes. This review updates the mechanism by which phosphatidylcholine biosynthesis takes place and how it is coordinately regulated during neuronal differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Membrana Celular/metabolismo , Neuritas/fisiología , Neuronas/metabolismo , Fosfatidilcolinas/biosíntesis , Fosfatidilcolinas/metabolismo , Animales , Colina Quinasa/metabolismo , Citidililtransferasa de Colina-Fosfato/metabolismo , Diacilglicerol Colinafosfotransferasa/metabolismo , Humanos , Mamíferos , Modelos Biológicos , Neuronas/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA