Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cells Int ; 2021: 8818356, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33828592

RESUMEN

Mouse embryonic stem cells (mESCs) and mouse epiblast stem cells (mEpiSCs) are the pluripotent stem cells (PSCs), derived from the inner cell mass (ICM) of preimplantation embryos at embryonic day 3.5 (E3.5) and postimplantation embryos at E5.5-E7.5, respectively. Depending on their environment, PSCs can exist in the so-called naïve (ESCs) or primed (EpiSCs) states. Exposure to EpiSC or human ESC (hESC) culture condition can convert mESCs towards an EpiSC-like state. Here, we show that the undifferentiated epiblast state is however not stabilized in a sustained manner when exposing mESCs to hESC or EpiSC culture condition. Rather, prolonged exposure to EpiSC condition promotes a transition to a primitive streak- (PS-) like state via an unbiased epiblast-like intermediate. We show that the Brachyury-positive PS-like state is likely promoted by endogenous WNT signaling, highlighting a possible species difference between mouse epiblast-like stem cells and human Embryonic Stem Cells.

2.
Arterioscler Thromb Vasc Biol ; 40(3): 697-713, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31826651

RESUMEN

OBJECTIVE: Long noncoding RNAs (lncRNAs) are an emergent class of molecules with diverse functional roles, widely expressed in human physiology and disease. Although some lncRNAs have been identified in cardiovascular disease, their potential as novel targets in the prevention of atherosclerosis is unknown. We set out to discover important lncRNAs in unstable plaque and gain insight into their functional relevance. Approach and Results: Analysis of RNA sequencing previously performed on stable and unstable atherosclerotic plaque identified a panel of 47 differentially regulated lncRNAs. We focused on LINC01272, a lncRNA upregulated in unstable plaque previously detected in inflammatory bowel disease, which we termed PELATON (plaque enriched lncRNA in atherosclerotic and inflammatory bowel macrophage regulation). Here, we demonstrate that PELATON is highly monocyte- and macrophage-specific across vascular cell types, and almost entirely nuclear by cellular fractionation (90%-98%). In situ hybridization confirmed enrichment of PELATON in areas of plaque inflammation, colocalizing with macrophages around the shoulders and necrotic core of human plaque sections. Consistent with its nuclear localization, and despite containing a predicted open reading frame, PELATON did not demonstrate any protein-coding potential in vitro. Functionally, knockdown of PELATON significantly reduced phagocytosis, lipid uptake and reactive oxygen species production in high-content analysis, with a significant reduction in phagocytosis independently validated. Furthermore, CD36, a key mediator of phagocytic oxLDL (oxidized low-density lipoprotein) uptake was significantly reduced with PELATON knockdown. CONCLUSIONS: PELATON is a nuclear expressed, monocyte- and macrophage-specific lncRNA, upregulated in unstable atherosclerotic plaque. Knockdown of PELATON affects cellular functions associated with plaque progression.


Asunto(s)
Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica , ARN Largo no Codificante/metabolismo , Anciano , Anciano de 80 o más Años , Antígenos CD36/genética , Antígenos CD36/metabolismo , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Células Cultivadas , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Metabolismo de los Lípidos , Macrófagos/patología , Masculino , Necrosis , Fagocitosis , ARN Largo no Codificante/genética , Especies Reactivas de Oxígeno/metabolismo , Rotura Espontánea
3.
Front Cell Dev Biol ; 7: 228, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31681761

RESUMEN

The early mammalian embryo is characterized by the presence of three germ layers-the outer ectoderm, middle mesoderm and inner endoderm. The mesoderm is organized into paraxial, intermediate and lateral plate mesoderm. The musculature, vasculature and heart of the adult body are the major derivatives of mesoderm. Tracing back the developmental process to generate these specialized tissues has sparked much interest in the field of regenerative medicine focusing on generating specialized tissues to treat patients with degenerative diseases. Several Long Non-Coding RNAs (lncRNAs) have been identified as regulators of development, proliferation and differentiation of various tissues of mesodermal origin. A better understanding of lncRNAs that can regulate the development of these tissues will open potential avenues for their therapeutic utility and enhance our knowledge about disease progression and development. In this review, we aim to summarize the functions and mechanisms of lncRNAs regulating the early mesoderm differentiation, development and homeostasis of skeletal muscle and cardiovascular system with an emphasis on their therapeutic potential.

4.
Cell Commun Signal ; 17(1): 143, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31703702

RESUMEN

BACKGROUND: Excessive TGF-ß signalling has been shown to underlie pulmonary hypertension (PAH). Human pulmonary artery smooth muscle cells (HPASMCs) can release extracellular vesicles (EVs) but their contents and significance have not yet been studied. Here, we aimed to analyse the contents and biological relevance of HPASMC-EVs and their transport to human pulmonary arterial endothelial cells (HPAECs), as well as the potential alteration of these under pathological conditions. METHODS: We used low-input RNA-Seq to analyse the RNA cargoes sorted into released HPASMC-EVs under basal conditions. We additionally analysed the effects of excessive TGF-ß signalling, using TGF-ß1 and BMP4, in the transcriptome of HPASMCs and their EVs. We then, for the first time, optimised Cre-loxP technology for its use with primary cells in vitro, directly visualising HPASMC-to-HPAEC communication and protein markers on cells taking up EVs. Furthermore we could analyse alteration of this transport with excessive TGF-ß signalling, as well as by other cytokines involved in PAH: IL-1ß, TNF-α and VEGFA. RESULTS: We were able to detect transcripts from 2417 genes in HPASMC-EVs. Surprisingly, among the 759 enriched in HPASMC-EVs compared to their donor cells, we found Zeb1 and 2 TGF-ß superfamily ligands, GDF11 and TGF-ß3. Moreover, we identified 90 genes differentially expressed in EVs from cells treated with TGF-ß1 compared to EVs in basal conditions, including a subset involved in actin and ECM remodelling, among which were bHLHE40 and palladin. Finally, using Cre-loxP technology we showed cell-to-cell transfer and translation of HPASMC-EV Cre mRNA from HPASMC to HPAECs, effectively evidencing communication via EVs. Furthermore, we found increased number of smooth-muscle actin positive cells on HPAECs that took up HPASMC-EVs. The uptake and translation of mRNA was also higher in activated HPAECs, when stimulated with TGF-ß1 or IL-1ß. CONCLUSIONS: HPASMC-EVs are enriched in RNA transcripts that encode genes that could contribute to vascular remodelling and EndoMT during development and PAH, and TGF-ß1 up-regulates some that could enhance this effects. These EVs are functionally transported, increasingly taken up by activated HPAECs and contribute to EndoMT, suggesting a potential effect of HPASMC-EVs in TGF-ß signalling and other related processes during PAH development.


Asunto(s)
Vesículas Extracelulares/metabolismo , Hipertensión Pulmonar/patología , Miocitos del Músculo Liso/patología , Arteria Pulmonar/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Remodelación Vascular , Proteínas Morfogenéticas Óseas/metabolismo , Endotelio Vascular/patología , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Interleucina-1beta/metabolismo , Fenotipo , Factor de Crecimiento Transformador beta3/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
5.
J Cell Mol Med ; 23(4): 2526-2535, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30677223

RESUMEN

Marfan syndrome (MFS) is an autosomal dominant genetic disorder caused by mutations in the FBN1 gene. Although many peripheral tissues are affected, aortic complications, such as dilation, dissection and rupture, are the leading causes of MFS-related mortality. Aberrant TGF-beta signalling plays a major role in the pathophysiology of MFS. However, the contributing mechanisms are still poorly understood. Here, we aimed at identifying novel aorta-specific pathways involved in the pathophysiology of MFS. For this purpose, we employed the Fbn1 under-expressing mgR/mgR mouse model of MFS. We performed RNA-sequencing of aortic tissues of 9-week-old mgR/mgR mice compared with wild-type (WT) mice. With a false discovery rate <5%, our analysis revealed 248 genes to be differentially regulated including 20 genes previously unrelated with MFS-related pathology. Among these, we identified Igfbp2, Ccl8, Spp1, Mylk2, Mfap4, Dsp and H19. We confirmed the expression of regulated genes by quantitative real-time PCR. Pathway classification revealed transcript signatures involved in chemokine signalling, cardiac muscle contraction, dilated and hypertrophic cardiomyopathy. Furthermore, our immunoblot analysis of aortic tissues revealed altered regulation of pSmad2 signalling, Perk1/2, Igfbp2, Mfap4, Ccl8 and Mylk2 protein levels in mgR/mgR vs WT mice. Together, our integrative systems approach identified several novel factors associated with MFS-aortic-specific pathophysiology that might offer potential novel therapeutic targets for MFS.


Asunto(s)
Aorta Torácica/metabolismo , Proteínas Portadoras/genética , Proteínas de la Matriz Extracelular/genética , Fibrilina-1/genética , Glicoproteínas/genética , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Síndrome de Marfan/genética , Osteopontina/genética , Animales , Aorta Torácica/fisiopatología , Proteínas Portadoras/metabolismo , Quimiocina CCL8/genética , Quimiocina CCL8/metabolismo , Desmoplaquinas/genética , Desmoplaquinas/metabolismo , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Fibrilina-1/deficiencia , Regulación de la Expresión Génica , Ontología de Genes , Glicoproteínas/metabolismo , Humanos , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Síndrome de Marfan/metabolismo , Síndrome de Marfan/fisiopatología , Ratones , Ratones Transgénicos , Anotación de Secuencia Molecular , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Osteopontina/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Biología de Sistemas/métodos , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
6.
J Bone Miner Res ; 30(5): 796-808, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25407900

RESUMEN

MicroRNAs play important roles during cell reprogramming and differentiation. In this study, we identified the miR-497∼195 cluster, a member of the miR-15 family, as strongly upregulated with age of postnatal bone development in vivo and late differentiation stages of primary osteoblasts cultured in vitro. Early expression of miR-195-5p inhibits differentiation and mineralization. Microarray analyses along with quantitative PCR demonstrate that miR-195-5p alters the gene regulatory network of osteoblast differentiation and impairs the induction of bone morphogenetic protein (BMP) responsive genes. Applying reporter gene and Western blot assays, we show that miR-195-5p interferes with the BMP/Smad-pathway in a dose-dependent manner. Systematically comparing the changes in mRNA levels in response to miR-195-5p overexpression with the changes observed in the natural course of osteoblast differentiation, we demonstrate that microRNAs of the miR-15 family affect several target genes involved in BMP signaling. Predicted targets including Furin, a protease that cleaves pro-forms, genes encoding receptors such as Acvr2a, Bmp1a, Dies1, and Tgfbr3, molecules within the cascade like Smad5, transcriptional regulators like Ski and Zfp423 as well as Mapk3 and Smurf1 were validated by quantitative PCR. Taken together, our data strongly suggest that miR-497∼195 cluster microRNAs act as intracellular antagonists of BMP signaling in bone cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/genética , MicroARNs/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Transducción de Señal/genética , Envejecimiento/genética , Animales , Animales Recién Nacidos , Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Biología Computacional , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Células 3T3 NIH , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Recombinantes/farmacología , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología
7.
Int J Biochem Cell Biol ; 45(3): 696-705, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23262291

RESUMEN

Osteoblastic differentiation is controlled by complex interplay of several signaling pathways and associated key transcription factors, as well as by microRNAs (miRNAs). In our current study, we found miR-181a to be highly upregulated during BMP induced osteoblastic differentiation of C2C12 and MC3T3 cells. Overexpression of miR-181a led to upregulation of key markers of osteoblastic differentiation as well as enhanced ALP levels and Alizarin red staining, indicating the importance of this miRNA for osteoblastic differentiation. Further, we show that miR-181 isoforms (181a, 181b, 181c) are expressed during different stages of mouse calvarial and tibial development, implying their role in both endochondral and intramembranous ossification. We found several direct and indirect targets of miR-181a to be downregulated by global mRNA expression profiling. Our results demonstrate that miR-181a promotes osteoblastic differentiation via repression of TGF-ß signaling molecules by targeting the negative regulator of osteoblastic differentiation Tgfbi (Tgf-beta induced) and TßR-I/Alk5 (TGF-ß type I receptor). Furthermore, our findings suggest that Rgs4 and Gata6 are direct targets of miR-181a. Taken together, we provide evidence for a crucial functional link between a specific miRNA, miR-181a and osteoblastic differentiation.


Asunto(s)
Diferenciación Celular , MicroARNs/genética , Osteoblastos , Factor de Crecimiento Transformador beta/genética , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Factor de Transcripción GATA6/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , MicroARNs/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas RGS/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba
8.
Stem Cells Dev ; 21(16): 2987-3000, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22724507

RESUMEN

Bone morphogenetic protein (BMP) signaling is known to support differentiation of human embryonic stem cells (hESCs) into mesoderm and extraembryonic lineages, whereas other signaling pathways can largely influence this lineage specification. Here, we set out to reinvestigate the influence of ACTIVIN/NODAL and fibroblast growth factor (FGF) pathways on the lineage choices made by hESCs during BMP4-driven differentiation. We show that BMP activation, coupled with inhibition of both ACTIVIN/NODAL and FGF signaling, induces differentiation of hESCs, specifically to ßhCG hormone-secreting multinucleated syncytiotrophoblast and does not support induction of embryonic and extraembryonic lineages, extravillous trophoblast, and primitive endoderm. It has been previously reported that FGF2 can switch BMP4-induced hESC differentiation outcome to mesendoderm. Here, we show that FGF inhibition alone, or in combination with either ACTIVIN/NODAL inhibition or BMP activation, supports hESC differentiation to hCG-secreting syncytiotrophoblast. We show that the inhibition of the FGF pathway acts as a key in directing BMP4-mediated hESC differentiation to syncytiotrophoblast.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Trofoblastos/citología , Activinas/metabolismo , Animales , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/genética , Benzamidas/farmacología , Factor de Transcripción CDX2 , Diferenciación Celular/genética , Fusión Celular , Línea Celular , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Dioxoles/farmacología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Endodermo/citología , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Ratones , Modelos Biológicos , Proteína Nodal/antagonistas & inhibidores , Proteína Nodal/metabolismo , Placenta/efectos de los fármacos , Placenta/metabolismo , Embarazo , Pirroles/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Proteínas Wnt/metabolismo
9.
PLoS One ; 6(10): e25163, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21998639

RESUMEN

BACKGROUND: Bone morphogenetic proteins (BMPs) are involved in a plethora of cellular processes in embryonic development and adult tissue homeostasis. Signaling specificity is achieved by dynamic processes involving BMP receptor oligomerization and endocytosis. This allows for spatiotemporal control of Smad dependent and non-Smad pathways. In this study, we investigate the spatiotemporal regulation within the BMP-induced Smad transcriptional pathway. METHODOLOGY/PRINCIPAL FINDINGS: Here we discriminate between Smad signaling events that are dynamin-dependent (i.e., require an intact endocytic pathway) and dynamin-independent. Inhibition of dynamin-dependent endocytosis in fluorescence microscopy and fractionation studies revealed a delay in Smad1/5/8 phosphorylation and nuclear translocation after BMP-2 stimulation of C2C12 cells. Using whole genome microarray and qPCR analysis, we identified two classes of BMP-2 induced genes that are differentially affected by inhibition of endocytosis. Thus, BMP-2 induced gene expression of Id1, Id3, Dlx2 and Hey1 is endocytosis-dependent, whereas BMP-2 induced expression of Id2, Dlx3, Zbtb2 and Krt16 is endocytosis-independent. Furthermore, we demonstrate that short term inhibition of endocytosis interferes with osteoblast differentiation as measured by alkaline phosphatase (ALP) production and qPCR analysis of osteoblast marker gene expression. CONCLUSIONS/SIGNIFICANCE: Our study demonstrates that dynamin-dependent endocytosis is crucial for the concise spatial activation of the BMP-2 induced signaling cascade. Inhibition of endocytic processes during BMP-2 stimulation leads to altered Smad1/5/8 signaling kinetics and results in differential target gene expression. We show that interfering with the BMP-2 induced transcriptional network by endocytosis inhibition results in an attenuation of osteoblast differentiation. This implies that selective sensitivity of gene expression to endocytosis provides an additional mechanism for the cell to respond to BMP in a context specific manner. Moreover, we suggest a novel Smad dependent signal cascade induced by BMP-2, which does not require endocytosis.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Osteoblastos/citología , Osteoblastos/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Dinaminas/metabolismo , Endocitosis/efectos de los fármacos , Endocitosis/genética , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Hidrazonas/farmacología , Cinética , Ratones , Familia de Multigenes/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
10.
Sci Signal ; 3(107): mr1, 2010 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-20124549

RESUMEN

The family of bone morphogenetic proteins (BMPs) comprises approximately 30 secreted cytokines that signal through transmembrane serine/threonine kinase receptors. The BMP signaling pathways are fine-tuned on multiple levels: Extracellular antagonists modify ligand activity; several co-receptors enhance or inhibit downstream signaling events through multiple mechanisms; and intracellular molecules further regulate the signaling outcome and mediate crosstalk with other pathways. BMPs affect structures and processes throughout the entire body, ranging from embryonic patterning and development through stem cells and their niches, to tissue homeostasis and regeneration. This comprehensive involvement in various tissues had not been expected by Marshall Urist, who initially discovered the ability of an unknown factor in bone to induce bone growth in muscle and subsequently suggested the name "bone morphogenetic protein." Today, recombinant BMPs are used in clinical practice for the treatment of bone and kidney disorders, and new genetically modified BMPs are emerging as promising tools in regenerative medicine and tissue engineering. Clearly, the functions of BMPs within the body are more versatile than initially suspected. To discuss modern trends in BMP signaling, leaders in the field met for the First International BMP Workshop in Berlin in September 2009. Here, we summarize new insights on the roles of BMPs in various tissues and highlight recent findings in cell, structural, and developmental biology as well as the therapeutic potential of BMPs. Finally, we conclude that BMPs today deserve to be called body morphogenetic proteins.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Huesos/fisiología , Transducción de Señal/fisiología , Desarrollo Óseo , Proteínas Morfogenéticas Óseas/metabolismo , Huesos/metabolismo , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Tracto Gastrointestinal/crecimiento & desarrollo , Tracto Gastrointestinal/metabolismo , Humanos , Riñón/crecimiento & desarrollo , Riñón/metabolismo , Modelos Biológicos , Morfogénesis , Páncreas/crecimiento & desarrollo , Páncreas/metabolismo
11.
BMC Genomics ; 8: 98, 2007 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-17425807

RESUMEN

BACKGROUND: TGF-beta is one of the key cytokines implicated in various disease processes including cancer. TGF-beta inhibits growth and promotes apoptosis in normal epithelial cells and in contrast, acts as a pro-tumour cytokine by promoting tumour angiogenesis, immune-escape and metastasis. It is not clear if various actions of TGF-beta on normal and tumour cells are due to differential gene regulations. Hence we studied the regulation of gene expression by TGF-beta in normal and cancer cells. RESULTS: Using human 19 K cDNA microarrays, we show that 1757 genes are exclusively regulated by TGF-beta in A549 cells in contrast to 733 genes exclusively regulated in HPL1D cells. In addition, 267 genes are commonly regulated in both the cell-lines. Semi-quantitative and real-time qRT-PCR analysis of some genes agrees with the microarray data. In order to identify the signalling pathways that influence TGF-beta mediated gene regulation, we used specific inhibitors of p38 MAP kinase, ERK kinase, JNK kinase and integrin signalling pathways. The data suggest that regulation of majority of the selected genes is dependent on at least one of these pathways and this dependence is cell-type specific. Interestingly, an integrin pathway inhibitor, RGD peptide, significantly affected TGF-beta regulation of Thrombospondin 1 in A549 cells. CONCLUSION: These data suggest major differences with respect to TGF-beta mediated gene regulation in normal and transformed cells and significant role of non-canonical TGF-beta pathways in the regulation of many genes by TGF-beta.


Asunto(s)
Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Factor de Crecimiento Transformador beta/metabolismo , Análisis por Conglomerados , Humanos , Integrinas/antagonistas & inhibidores , Integrinas/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...