Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 6(21)2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34591795

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) is a well-characterized animal model of multiple sclerosis. During the early phase of EAE, infiltrating monocytes and monocyte-derived macrophages contribute to T cell recruitment, especially CD4+ T cells, into the CNS, resulting in neuronal demyelination; however, in later stages, they promote remyelination and recovery by removal of myelin debris by phagocytosis. Signal regulatory protein α and CD47 are abundantly expressed in the CNS, and deletion of either molecule is protective in myelin oligodendrocyte glycoprotein-induced EAE because of failed effector T cell expansion and trafficking. Here we report that treatment with the function blocking CD47 Ab Miap410 substantially reduced the infiltration of pathogenic immune cells but impaired recovery from paresis. The underlying mechanism was by blocking the emergence of CD11chiMHCIIhi microglia at peak disease that expressed receptors for phagocytosis, scavenging, and lipid catabolism, which mediated clearance of myelin debris and the transition of monocytes to macrophages in the CNS. In the recovery phase of EAE, Miap410 Ab-treated mice had worsening paresis with sustained inflammation and limited remyelination as compared with control Ab-treated mice. In summary, Ab blockade of CD47 impaired resolution of CNS inflammation, thus worsening EAE.


Asunto(s)
Antígeno CD47/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Macrófagos/metabolismo , Microglía/metabolismo , Monocitos/metabolismo , Fagocitosis/genética , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Noqueados
2.
PLoS One ; 9(10): e110316, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25360666

RESUMEN

Glioblastoma multiform (GBM) remains clinical indication with significant "unmet medical need". Innovative new therapy to eliminate residual tumor cells and prevent tumor recurrences is critically needed for this deadly disease. A major challenge of GBM research has been the identification of novel molecular therapeutic targets and accurate diagnostic/prognostic biomarkers. Many of the current clinical therapeutic targets of immunotoxins and ligand-directed toxins for high-grade glioma (HGG) cells are surface sialylated glycoproteins. Therefore, methods that systematically and quantitatively analyze cell surface sialoglycoproteins in human clinical tumor samples would be useful for the identification of potential diagnostic markers and therapeutic targets for malignant gliomas. In this study, we used the bioorthogonal chemical reporter strategy (BOCR) in combination with label-free quantitative mass spectrometry (LFQ-MS) to characterize and accurately quantify the individual cell surface sialoproteome in human GBM tissues, in fetal, adult human astrocytes, and in human neural progenitor cells (NPCs). We identified and quantified a total of 843 proteins, including 801 glycoproteins. Among the 843 proteins, 606 (72%) are known cell surface or secreted glycoproteins, including 156 CD-antigens, all major classes of cell surface receptor proteins, transporters, and adhesion proteins. Our findings identified several known as well as new cell surface antigens whose expression is predominantly restricted to human GBM tumors as confirmed by microarray transcription profiling, quantitative RT-PCR and immunohistochemical staining. This report presents the comprehensive identification of new biomarkers and therapeutic targets for the treatment of malignant gliomas using quantitative sialoglycoproteomics with clinically relevant, patient derived primary glioma cells.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/metabolismo , Glioblastoma/patología , Proteómica/métodos , Sialoglicoproteínas/metabolismo , Adulto , Anciano , Biotinilación , Neoplasias Encefálicas/genética , Femenino , Perfilación de la Expresión Génica , Glioblastoma/genética , Humanos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Ácido N-Acetilneuramínico/metabolismo , Embarazo , Transporte de Proteínas , Sialoglicoproteínas/genética
3.
Cell Rep ; 3(6): 2127-41, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23727239

RESUMEN

Glial progenitor cells (GPCs) are a potential source of malignant gliomas. We used A2B5-based sorting to extract tumorigenic GPCs from human gliomas spanning World Health Organization grades II-IV. Messenger RNA profiling identified a cohort of genes that distinguished A2B5+ glioma tumor progenitor cells (TPCs) from A2B5+ GPCs isolated from normal white matter. A core set of genes and pathways was substantially dysregulated in A2B5+ TPCs, which included the transcription factor SIX1 and its principal cofactors, EYA1 and DACH2. Small hairpin RNAi silencing of SIX1 inhibited the expansion of glioma TPCs in vitro and in vivo, suggesting a critical and unrecognized role of the SIX1-EYA1-DACH2 system in glioma genesis or progression. By comparing the expression patterns of glioma TPCs with those of normal GPCs, we have identified a discrete set of pathways by which glial tumorigenesis may be better understood and more specifically targeted.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Glioma/genética , Glioma/patología , Células Madre Neoplásicas/patología , Neuroglía/patología , Neuroglía/fisiología , Adulto , Neoplasias Encefálicas/metabolismo , Diferenciación Celular/fisiología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Humanos , Persona de Mediana Edad , Neuroglía/metabolismo , Activación Transcripcional
4.
Cell ; 151(7): 1617-32, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23260147

RESUMEN

Factor-induced reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) is inefficient, complicating mechanistic studies. Here, we examined defined intermediate cell populations poised to becoming iPSCs by genome-wide analyses. We show that induced pluripotency elicits two transcriptional waves, which are driven by c-Myc/Klf4 (first wave) and Oct4/Sox2/Klf4 (second wave). Cells that become refractory to reprogramming activate the first but fail to initiate the second transcriptional wave and can be rescued by elevated expression of all four factors. The establishment of bivalent domains occurs gradually after the first wave, whereas changes in DNA methylation take place after the second wave when cells acquire stable pluripotency. This integrative analysis allowed us to identify genes that act as roadblocks during reprogramming and surface markers that further enrich for cells prone to forming iPSCs. Collectively, our data offer new mechanistic insights into the nature and sequence of molecular events inherent to cellular reprogramming.


Asunto(s)
Reprogramación Celular , Técnicas Citológicas/métodos , Células Madre Pluripotentes Inducidas/citología , Animales , Estudio de Asociación del Genoma Completo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Ratones , Factores de Transcripción/metabolismo
5.
Small ; 8(22): 3510-6, 2012 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-22915545

RESUMEN

Understanding the structure and function of glucose binding proteins (GBP) complexed with single walled carbon nanotubes (SWNTs) is important for the development of applications including fluorescent sensors and nanostructure particle tracking. Herein, circular dichroism (CD), thermal denaturation, photo-absorption spectroscopy and atomic force microscopy are used to study these nanostructures. The protein retains its glucose-binding activity after complexation and is thermally stable below 36 °C. However, the SWNT lowers the midpoint denaturation temperature (Tm) by 5 °C and 4 °C in the absence and presence of 10 mM glucose, respectively. This data highlights that using techniques such as CD and thermal denaturation may be necessary to fully characterize such protein-nanomaterial nanostructures.


Asunto(s)
Glucosa/química , Nanotecnología/métodos , Nanotubos de Carbono/química , Carbodiimidas/química , Dicroismo Circular , Calor , Microscopía de Fuerza Atómica , Nanopartículas/química , Fotoquímica/métodos , Alcohol Polivinílico , Unión Proteica , Desnaturalización Proteica , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja Corta
6.
PLoS One ; 5(7): e11729, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20668531

RESUMEN

BACKGROUND: Proteins that are required for anchorage-independent survival of tumor cells represent attractive targets for therapeutic intervention since this property is believed to be critical for survival of tumor cells displaced from their natural niches. Anchorage-independent survival is induced by growth factor receptor hyperactivation in many cell types. We aimed to identify molecules that critically regulate IGF-1-induced anchorage-independent survival. METHODS AND RESULTS: We conducted a high-throughput siRNA screen and identified PTK6 as a critical component of IGF-1 receptor (IGF-1R)-induced anchorage-independent survival of mammary epithelial cells. PTK6 downregulation induces apoptosis of breast and ovarian cancer cells deprived of matrix attachment, whereas its overexpression enhances survival. Reverse-phase protein arrays and subsequent analyses revealed that PTK6 forms a complex with IGF-1R and the adaptor protein IRS-1, and modulates anchorage-independent survival by regulating IGF-1R expression and phosphorylation. PTK6 is highly expressed not only in the previously reported Her2(+) breast cancer subtype, but also in high grade ER(+), Luminal B tumors and high expression is associated with adverse outcomes. CONCLUSIONS: These findings highlight PTK6 as a critical regulator of anchorage-independent survival of breast and ovarian tumor cells via modulation of IGF-1 receptor signaling, thus supporting PTK6 as a potential therapeutic target for multiple tumor types. The combined genomic and proteomic approaches in this report provide an effective strategy for identifying oncogenes and their mechanism of action.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Western Blotting , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Hibridación Fluorescente in Situ , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Estimación de Kaplan-Meier , Microscopía Confocal , Microscopía de Contraste de Fase , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Proteínas Tirosina Quinasas/genética , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo
7.
Nat Biotechnol ; 28(8): 848-55, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20644536

RESUMEN

Induced pluripotent stem cells (iPSCs) have been derived from various somatic cell populations through ectopic expression of defined factors. It remains unclear whether iPSCs generated from different cell types are molecularly and functionally similar. Here we show that iPSCs obtained from mouse fibroblasts, hematopoietic and myogenic cells exhibit distinct transcriptional and epigenetic patterns. Moreover, we demonstrate that cellular origin influences the in vitro differentiation potentials of iPSCs into embryoid bodies and different hematopoietic cell types. Notably, continuous passaging of iPSCs largely attenuates these differences. Our results suggest that early-passage iPSCs retain a transient epigenetic memory of their somatic cells of origin, which manifests as differential gene expression and altered differentiation capacity. These observations may influence ongoing attempts to use iPSCs for disease modeling and could also be exploited in potential therapeutic applications to enhance differentiation into desired cell lineages.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Linfocitos B/citología , Células Cultivadas , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Epigenómica , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Ratones , Músculo Esquelético/citología , Células Madre/citología , Transcripción Genética
8.
Nature ; 465(7295): 175-81, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20418860

RESUMEN

Induced pluripotent stem cells (iPSCs) have been generated by enforced expression of defined sets of transcription factors in somatic cells. It remains controversial whether iPSCs are molecularly and functionally equivalent to blastocyst-derived embryonic stem (ES) cells. By comparing genetically identical mouse ES cells and iPSCs, we show here that their overall messenger RNA and microRNA expression patterns are indistinguishable with the exception of a few transcripts encoded within the imprinted Dlk1-Dio3 gene cluster on chromosome 12qF1, which were aberrantly silenced in most of the iPSC clones. Consistent with a developmental role of the Dlk1-Dio3 gene cluster, these iPSC clones contributed poorly to chimaeras and failed to support the development of entirely iPSC-derived animals ('all-iPSC mice'). In contrast, iPSC clones with normal expression of the Dlk1-Dio3 cluster contributed to high-grade chimaeras and generated viable all-iPSC mice. Notably, treatment of an iPSC clone that had silenced Dlk1-Dio3 with a histone deacetylase inhibitor reactivated the locus and rescued its ability to support full-term development of all-iPSC mice. Thus, the expression state of a single imprinted gene cluster seems to distinguish most murine iPSCs from ES cells and allows for the prospective identification of iPSC clones that have the full development potential of ES cells.


Asunto(s)
Cromosomas de los Mamíferos/genética , Perfilación de la Expresión Génica , Silenciador del Gen , Impresión Genómica/genética , Células Madre Pluripotentes/metabolismo , Animales , Proteínas de Unión al Calcio , Línea Celular , Células Madre Embrionarias/metabolismo , Epigénesis Genética/genética , Femenino , Fibroblastos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Familia de Multigenes/genética , Proteínas Nucleares/genética , Células Madre Pluripotentes/citología , Proteínas/genética , ARN Largo no Codificante , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/genética
9.
Cell Stem Cell ; 3(2): 125-6, 2008 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-18682232

RESUMEN

In a recent paper in Nature Neuroscience, Jessberger et al. (2008) report that overexpression of ASCL1 (mash1) directs adult hippocampal progenitors to adopt an oligodendrocytic fate. The effect is specific to the hippocampal niche in vivo, indicating that cell-autonomous and niche-defined factors collaborate to instruct cell fate choices.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Giro Dentado/citología , Oligodendroglía/citología , Organogénesis/genética , Activación Transcripcional/genética , Adulto , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Giro Dentado/embriología , Giro Dentado/crecimiento & desarrollo , Humanos , Oligodendroglía/metabolismo , Transducción de Señal/genética , Nicho de Células Madre , Células Madre/citología , Células Madre/metabolismo
10.
Ann Neurol ; 59(5): 763-79, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16634042

RESUMEN

OBJECTIVE: Glial progenitor cells are abundant in adult human white matter. This study was designed to identify signaling pathways regulating their self-renewal and fate. METHODS: We compared the transcriptional profiles of freshly sorted adult human white matter progenitor cells (WMPCs), purified by A2B5-based immunomagnetic sorting, with those of the white matter from which they derived. RESULTS: We identified 132 genes differentially expressed by WMPCs; these included principal components of five receptor-defined signaling pathways, represented by platelet derived growth factor receptor alpha (PDGFRA) and type 3 fibroblast growth factor receptor (FGFR3), receptor tyrosine phosphatase-beta/zeta (RTPZ), notch, and syndecan3. WMPCs also differentially expressed the bone morphogenetic protein 4 (BMP4) inhibitors neuralin and BAMBI (BMP and activin membrane-bound inhibitor), suggesting tonic defense against BMP signaling. Differential overexpression of RTPZ was accompanied by that of its modulators pleiotrophin, NrCAM, tenascin, and the chondroitin sulfate proteoglycans, suggesting the importance of RTPZ signaling to WMPCs. When exposed to the RTPZ inhibitor bpV(phen), or lentiviral-shRNAi against RTPZ, WMPCs differentiated as oligodendrocytes. Conversely, when neuralin and BAMBI were antagonized by BMP4, astrocytic differentiation was induced, which was reversible by noggin. INTERPRETATION: The RTPZ and BMP pathways regulate the self-maintenance of adult human WMPCs, and can be modulated to induce their oligodendrocytic or astrocytic differentiation. As such, they provide targets by which to productively mobilize resident progenitor cells of the adult human brain.


Asunto(s)
Diferenciación Celular/fisiología , Expresión Génica/fisiología , Oligodendroglía/fisiología , Células Madre/fisiología , Adolescente , Adulto , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/farmacología , Moléculas de Adhesión Celular/biosíntesis , Citocinas/farmacología , Ambiente , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Femenino , Humanos , Inmunohistoquímica , Lentivirus/metabolismo , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Tirosina Fosfatasas/metabolismo , ARN/biosíntesis , ARN/aislamiento & purificación , ARN Interferente Pequeño/farmacología , Receptores de Superficie Celular/efectos de los fármacos , Receptores Notch/genética , Receptores Notch/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología
11.
J Cell Biol ; 171(6): 1023-34, 2005 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-16365168

RESUMEN

The Akt family of kinases are activated by growth factors and regulate pleiotropic cellular activities. In this study, we provide evidence for isoform-specific positive and negative roles for Akt1 and -2 in regulating growth factor-stimulated phenotypes in breast epithelial cells. Insulin-like growth factor-I receptor (IGF-IR) hyperstimulation induced hyperproliferation and antiapoptotic activities that were reversed by Akt2 down-regulation. In contrast, Akt1 down-regulation in IGF-IR-stimulated cells promoted dramatic neomorphic effects characteristic of an epithelial-mesenchymal transition (EMT) and enhanced cell migration induced by IGF-I or EGF stimulation. The phenotypic effects of Akt1 down-regulation were accompanied by enhanced extracellular signal-related kinase (ERK) activation, which contributed to the induction of migration and EMT. Interestingly, down-regulation of Akt2 suppressed the EMT-like morphological conversion induced by Akt1 down-regulation in IGF-IR-overexpressing cells and inhibited migration in EGF-stimulated cells. These results highlight the distinct functions of Akt isoforms in regulating growth factor-stimulated EMT and cell migration, as well as the importance of Akt1 in cross-regulating the ERK signaling pathway.


Asunto(s)
Movimiento Celular/fisiología , Células Epiteliales/metabolismo , Mesodermo/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Biomarcadores , Mama/enzimología , Mama/metabolismo , Línea Celular , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Mesodermo/enzimología , Morfogénesis , Isoformas de Proteínas/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal
12.
Trends Biotechnol ; 23(1): 1-3, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15629847

RESUMEN

The idea of a cell-based regeneration therapy for controlling or curing chronic human diseases is highly attractive. However, realization of this idea in the clinic has been hampered by the safety concerns associated with the transplantation of immortalized cells into human patients. An elegant study done by Roy and colleagues shows that neural progenitor cells immortalized by the ectopic expression of telomerase reverse transcriptase (TERT) can give rise to specific types of functionally competent neurons both in vitro and in vivo. Importantly, the immortalized progenitors maintained their phenotype with no evidence of transformation even several months after transplantation in mouse disease models. Although the potential use of telomerase-immortalized cells in the clinic remains controversial, Roy and colleagues work provides a compelling reason to seriously evaluate the potential use of telomerase-immortalized progenitor cells to treat neurodegenerative and other chronic human illnesses.


Asunto(s)
Trasplante de Células Madre/métodos , Células Madre/fisiología , Telomerasa/metabolismo , Animales , Supervivencia Celular , Transformación Celular Neoplásica , Humanos , Neuronas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...