Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
PLoS Biol ; 17(1): e2006972, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30695023

RESUMEN

Insulin provides important information to tissues about feeding behavior and energy status. Defective insulin signaling is associated with ageing, tissue dysfunction, and impaired wound healing. In the liver, insulin resistance leads to chronic damage and fibrosis, but it is unclear how tissue-repair mechanisms integrate insulin signals to coordinate an appropriate injury response or how they are affected by insulin resistance. In this study, we demonstrate that insulin resistance impairs local cellular crosstalk between the fibrotic stroma and bipotent adult liver progenitor cells (LPCs), whose paracrine interactions promote epithelial repair and tissue remodeling. Using insulin-resistant mice deficient for insulin receptor substrate 2 (Irs2), we highlight dramatic impairment of proregenerative fibroblast growth factor 7 (Fgf7) signaling between stromal niche cells and LPCs during chronic injury. We provide a detailed account of the role played by IRS2 in promoting Fgf7 ligand and receptor (Fgfr2-IIIb) expression by the two cell compartments, and we describe an insulin/IRS2-dependent feed-forward loop capable of sustaining hepatic re-epithelialization by driving FGFR2-IIIb expression. Finally, we shed light on the regulation of IRS2 and FGF7 within the fibrotic stroma and show-using a human coculture system-that IRS2 silencing shifts the equilibrium away from paracrine epithelial repair in favor of fibrogenesis. Hence, we offer a compelling insight into the contribution of insulin resistance to the pathogenesis of chronic liver disease and propose IRS2 as a positive regulator of communication between cell types and the transition between phases of stromal to epithelial repair.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Animales , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Factor 7 de Crecimiento de Fibroblastos/fisiología , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/fisiología , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Ratones , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Células Madre/fisiología
2.
Bioinformatics ; 35(20): 4089-4097, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30903689

RESUMEN

MOTIVATION: The analysis of biological samples in untargeted metabolomic studies using LC-MS yields tens of thousands of ion signals. Annotating these features is of the utmost importance for answering questions as fundamental as, e.g. how many metabolites are there in a given sample. RESULTS: Here, we introduce CliqueMS, a new algorithm for annotating in-source LC-MS1 data. CliqueMS is based on the similarity between coelution profiles and therefore, as opposed to most methods, allows for the annotation of a single spectrum. Furthermore, CliqueMS improves upon the state of the art in several dimensions: (i) it uses a more discriminatory feature similarity metric; (ii) it treats the similarities between features in a transparent way by means of a simple generative model; (iii) it uses a well-grounded maximum likelihood inference approach to group features; (iv) it uses empirical adduct frequencies to identify the parental mass and (v) it deals more flexibly with the identification of the parental mass by proposing and ranking alternative annotations. We validate our approach with simple mixtures of standards and with real complex biological samples. CliqueMS reduces the thousands of features typically obtained in complex samples to hundreds of metabolites, and it is able to correctly annotate more metabolites and adducts from a single spectrum than available tools. AVAILABILITY AND IMPLEMENTATION: https://CRAN.R-project.org/package=cliqueMS and https://github.com/osenan/cliqueMS. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Programas Informáticos , Espectrometría de Masas en Tándem , Cromatografía Liquida , Iones , Metabolómica , Redes Neurales de la Computación
3.
Genome Res ; 21(4): 515-24, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21324880

RESUMEN

Aberrant DNA methylation (DNAm) was first linked to cancer over 25 yr ago. Since then, many studies have associated hypermethylation of tumor suppressor genes and hypomethylation of oncogenes to the tumorigenic process. However, most of these studies have been limited to the analysis of promoters and CpG islands (CGIs). Recently, new technologies for whole-genome DNAm (methylome) analysis have been developed, enabling unbiased analysis of cancer methylomes. By using MeDIP-seq, we report a sequencing-based comparative methylome analysis of malignant peripheral nerve sheath tumors (MPNSTs), benign neurofibromas, and normal Schwann cells. Analysis of these methylomes revealed a complex landscape of DNAm alterations. In contrast to what has been reported for other tumor types, no significant global hypomethylation was observed in MPNSTs using methylome analysis by MeDIP-seq. However, a highly significant (P < 10(-100)) directional difference in DNAm was found in satellite repeats, suggesting these repeats to be the main target for hypomethylation in MPNSTs. Comparative analysis of the MPNST and Schwann cell methylomes identified 101,466 cancer-associated differentially methylated regions (cDMRs). Analysis showed these cDMRs to be significantly enriched for two satellite repeat types (SATR1 and ARLα) and suggests an association between aberrant DNAm of these sequences and transition from healthy cells to malignant disease. Significant enrichment of hypermethylated cDMRs in CGI shores (P < 10(-60)), non-CGI-associated promoters (P < 10(-4)) and hypomethylated cDMRs in SINE repeats (P < 10(-100)) was also identified. Integration of DNAm and gene expression data showed that the expression pattern of genes associated with CGI shore cDMRs was able to discriminate between disease phenotypes. This study establishes MeDIP-seq as an effective method to analyze cancer methylomes.


Asunto(s)
Metilación de ADN/genética , Epigenómica , Neoplasias de la Vaina del Nervio/genética , Análisis por Conglomerados , Islas de CpG/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Repeticiones de Minisatélite/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos
4.
Curr Opin Oncol ; 26(1): 100-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24275855

RESUMEN

PURPOSE OF REVIEW: We review accumulating evidence that nonalcoholic steatohepatitis (NASH), a more advanced form of nonalcoholic fatty liver disease (NAFLD), predisposes patients to the risk of developing hepatocellular carcinoma (HCC), and we summarize recent advances in the elucidation of cancer-promoting pathways in NASH. We highlight the potential role of progenitor cells and hepatic stellate cells (HSCs) in promoting the early events that could culminate in cancer, as well as the emerging contribution of the gut-liver axis in promoting inflammation, senescence, and tumor growth in NASH and HCC. Finally, we review the role of bile acid receptors, vitamin D, and protective cellular pathways such as autophagy. RECENT FINDINGS: Studies have recently uncovered roles for gut microbiota, bile acid receptors and vitamin D in regulating the progression from NAFLD to HCC. Intriguing findings linking senescence and autophagy in hepatic stellate cells to HCC have also been discovered, as well as a link between dysregulated progenitor cell regulation and HCC. SUMMARY: NAFLD is the most common cause of chronic liver disease in the United States and Western Europe. The lack of definitive mechanisms underlying development of NASH among patients with NAFLD and its progression to HCC limit diagnosis and management, but new findings are paving the way for better biomarkers and therapies.


Asunto(s)
Carcinoma Hepatocelular/etiología , Hígado Graso/complicaciones , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas/etiología , Síndrome Metabólico/complicaciones , Ácidos y Sales Biliares/metabolismo , Progresión de la Enfermedad , Hígado Graso/metabolismo , Células Estrelladas Hepáticas/fisiología , Humanos , Inflamación/complicaciones , Microbiota , Receptores de Calcitriol/metabolismo , Células Madre/fisiología , Estómago/microbiología
5.
J Vis Exp ; (158)2020 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-32364548

RESUMEN

When the liver is injured, hepatocyte numbers decrease, while cell size, nuclear size and ploidy increase. The expansion of non-parenchymal cells such as cholangiocytes, myofibroblasts, progenitors and inflammatory cells also indicate chronic liver damage, tissue remodeling and disease progression. In this protocol, we describe a simple high-throughput approach for calculating changes in the cellular composition of the liver that are associated with injury, chronic disease and cancer. We show how information extracted from two-dimensional (2D) tissue sections can be used to quantify and calibrate hepatocyte nuclear ploidy within a sample and enable the user to locate specific ploidy subsets within the liver in situ. Our method requires access to fixed/frozen liver material, basic immunocytochemistry reagents and any standard high-content imaging platform. It serves as a powerful alternative to standard flow cytometry techniques, which require disruption of freshly collected tissue, loss of spatial information and potential disaggregation bias.


Asunto(s)
Núcleo Celular/metabolismo , Hepatocitos/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Ploidias , Animales , Automatización , Calibración , Análisis de Datos , Femenino , Citometría de Flujo , Fluorescencia , Procesamiento de Imagen Asistido por Computador , Hígado/metabolismo , Ratones Endogámicos C57BL
6.
Trends Pharmacol Sci ; 28(3): 103-5, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17276519

RESUMEN

The leprosy pathogen Mycobacterium leprae attacks Schwann cells in the peripheral nervous system, causing them to demyelinate. Recent work by Tapinos et al. shows that a direct mechanism of demyelination induced by M. leprae depends on the binding of the bacterium to the receptor tyrosine kinase ErbB2 on Schwann cells and the resulting activation of the Ras-Raf-MEK-ERK pathway. These findings have relevance for the potential treatment of leprosy and they highlight parallels between the dedifferentiation signal in leprosy and that in nerve injury and cancer.


Asunto(s)
Leprostáticos/uso terapéutico , Lepra/tratamiento farmacológico , Mycobacterium leprae/metabolismo , Receptor ErbB-2/metabolismo , Humanos , Leprostáticos/economía , Lepra/metabolismo , Lepra/microbiología , Modelos Biológicos , Mycobacterium leprae/genética , Unión Proteica/efectos de los fármacos , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo
7.
Nat Commun ; 9(1): 4962, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30470740

RESUMEN

Activation of the Hippo pathway effector Yap underlies many liver cancers, however no germline or somatic mutations have been identified. Autophagy maintains essential metabolic functions of the liver, and autophagy-deficient murine models develop benign adenomas and hepatomegaly, which have been attributed to activation of the p62/Sqstm1-Nrf2 axis. Here, we show that Yap is an autophagy substrate and mediator of tissue remodeling and hepatocarcinogenesis independent of the p62/Sqstm1-Nrf2 axis. Hepatocyte-specific deletion of Atg7 promotes liver size, fibrosis, progenitor cell expansion, and hepatocarcinogenesis, which is rescued by concurrent deletion of Yap. Our results shed new light on mechanisms of Yap degradation and the sequence of events that follow disruption of autophagy, which is impaired in chronic liver disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia , Hepatocitos/citología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/fisiopatología , Hígado/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Carcinogénesis , Proteínas de Ciclo Celular , Diferenciación Celular , Femenino , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/patología , Neoplasias Hepáticas/genética , Masculino , Ratones , Fosfoproteínas/genética , Proteolisis , Factores de Transcripción , Proteínas Señalizadoras YAP
8.
Endocrinology ; 147(12): 6019-26, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16997835

RESUMEN

The peptide hormone ACTH stimulates lipolysis and suppresses leptin production in adipocytes via the G protein-coupled receptor, melanocortin 2 receptor (MC2-R). We have shown previously that peroxisome proliferator-activated receptor-gamma2 is the primary factor responsible for transactivation of the already identified murine MC2-R promoter in the differentiating 3T3-L1 adipocyte cell line. In this study we show that despite the activity of this promoter being transient during differentiation, MC2-R message remains elevated at later time points during adipogenesis. Analysis of the late transcripts reveals that they initiate from a transcriptional start site in the first intron of the murine MC2-R. The genomic sequence upstream of this start site acts as an adipocyte-specific promoter whose activation is delayed in differentiation, compared with the upstream promoter. A CCAAT/enhancer-binding protein binding site, 87 bp upstream of the transcriptional initiation site, is necessary for the activity of this promoter, and protein binding analyses reveal that this site is bound by CCAAT/enhancer-binding protein factors. Real-time PCR analysis of mRNA initiating from the two start sites shows that there is a switch in promoter usage from the 5' to the 3' promoter around d 5, indicating the complex regulation of the murine MC2-R during adipogenesis.


Asunto(s)
Adipogénesis/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Regiones Promotoras Genéticas , Receptor de Melanocortina Tipo 2/genética , Elementos Reguladores de la Transcripción/fisiología , Células 3T3-L1/citología , Tejido Adiposo/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Codón Iniciador/análisis , Proteínas de Unión al ADN/metabolismo , Exones , Intrones/fisiología , Ratones , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Isoformas de Proteínas/análisis , Receptor de Melanocortina Tipo 2/química , Receptor de Melanocortina Tipo 2/metabolismo , Análisis de Secuencia de ADN , Transcripción Genética , Activación Transcripcional
9.
J Mol Endocrinol ; 37(3): 415-20, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17170082

RESUMEN

The ACTH receptor melanocortin 2 receptor (MC2-R) is a G-protein-coupled receptor principally expressed in the adrenal cortex and the adipocyte, where it stimulates steroidogenesis and lipolysis respectively. The coding region of the murine gene is encoded by a single exon, although three upstream non-coding exons have been documented, one of which is incorporated by alternative splicing in adrenal cells. We have detected a novel transcript in adipocytes, which includes a previously unidentified 86 bp exon upstream of the coding region. This transcript appears with slower kinetics during a time course of differentiation of 3T3-L1 cells and is much more highly expressed in these cells and murine adipose tissues than in the Y1 murine adrenocortical cell line, also it is undetectable in murine foetal testes. Inclusion of this exon extends the 5' UTR to 468 bp and introduces three upstream open reading frames. These are typical features of mRNAs under translational control and imply that the MC2-R gene is regulated both transcriptionally and post-transcriptionally during adipogenesis.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/genética , Empalme Alternativo/genética , Diferenciación Celular , Biosíntesis de Proteínas/genética , Receptor de Melanocortina Tipo 2/genética , Células 3T3 , Animales , Secuencia de Bases , Exones/genética , Ratones , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Transcripción Genética/genética
10.
Sci STKE ; 2005(309): pe52, 2005 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-16278488

RESUMEN

Schwann cells are the target of Mycobacterium leprae, the pathogen responsible for leprosy. Once inside the cell, M. leprae activates the host's proliferative machinery, thereby increasing the number of cells susceptible to infection. This astonishing manipulation of the mammalian cell cycle is the subject of recent work by Tapinos and Rambukkana, who show that M. leprae drives proliferation through a novel route to extracellular signal-regulated kinase (ERK). In this Perspective, we discuss this important piece of work and highlight the noncanonical pathway used by M. leprae to induce proliferation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/fisiología , Mycobacterium leprae/fisiología , Células de Schwann/microbiología , Transducción de Señal/fisiología , Ciclo Celular , Diferenciación Celular , División Celular , Células Cultivadas/enzimología , Células Cultivadas/microbiología , Ciclina D1/fisiología , Activación Enzimática , Humanos , Integrinas/metabolismo , Laminina/metabolismo , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/fisiología , Quinasas Quinasa Quinasa PAM/fisiología , Fosforilación , Proteína Quinasa C-epsilon , Procesamiento Proteico-Postraduccional , Células de Schwann/clasificación , Células de Schwann/enzimología
11.
Mol Cell Endocrinol ; 213(2): 149-54, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15062562

RESUMEN

Two mutations in the same allele of the ACTH receptor (melanocortin 2 receptor, MC2R) associated with clinical hypersensitivity to ACTH have been described in a single case report. Using a stable Y6 cell expression system, we demonstrate that either the C21R or S247G mutations alone produce an inactive receptor with loss of ligand binding and responsiveness. However, the presence of both mutations in the same molecule leads to a receptor with a highly significant elevation in constitutive activity (basal cAMP accumulation for wild type expressing cells 199 +/- 11 pmol/mg protein; double mutant: 374 +/- 29 pmol/mg protein, P < 0.005. The co-expression of the normal MC2R allele results in the retention of a normal dose response to ACTH despite the presence of constitutive activity.


Asunto(s)
Mutación Missense , Receptor de Melanocortina Tipo 2/genética , Receptor de Melanocortina Tipo 2/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Animales , Línea Celular , AMP Cíclico/metabolismo , Humanos , Ligandos , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Ensayo de Unión Radioligante , Receptor de Melanocortina Tipo 2/química , Transfección
12.
Ann N Y Acad Sci ; 994: 111-7, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12851305

RESUMEN

Research into the functions and mechanisms of action of the melanocortin 2 receptor (MC2R) has been severely hampered by difficulties in expressing this gene in heterologous cells. This probably arises because of the need for a cofactor for cell surface expression. Using either the Y1 cell line that expresses endogenous MC2R or the Y6 cell line that expresses this putative expression factor, we have explored the mechanisms of desensitization and internalization after agonist stimulation. Protein kinase A dependence of desensitization has been demonstrated, although internalization is apparently independent of this kinase and dependent on a G protein receptor kinase. Possible underlying reasons for this paradox are discussed.


Asunto(s)
Receptores de Corticotropina/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Animales , Línea Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Endocitosis/fisiología , Humanos , Receptor de Melanocortina Tipo 2 , Receptores de Corticotropina/genética
13.
Stem Cells Dev ; 23(4): 380-92, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24188453

RESUMEN

Human embryonic stem cells and induced pluripotent stem cells have great potential in research and therapies. The current in vitro culture systems for human pluripotent stem cells (hPSCs) do not mimic the three-dimensional (3D) in vivo stem cell niche that transiently supports stem cell proliferation and is subject to changes which facilitate subsequent differentiation during development. Here, we demonstrate, for the first time, that a novel plant-derived nanofibrillar cellulose (NFC) hydrogel creates a flexible 3D environment for hPSC culture. The pluripotency of hPSCs cultured in the NFC hydrogel was maintained for 26 days as evidenced by the expression of OCT4, NANOG, and SSEA-4, in vitro embryoid body formation and in vivo teratoma formation. The use of a cellulose enzyme, cellulase, enables easy cell propagation in 3D culture as well as a shift between 3D and two-dimensional cultures. More importantly, the removal of the NFC hydrogel facilitates differentiation while retaining 3D cell organization. Thus, the NFC hydrogel represents a flexible, xeno-free 3D culture system that supports pluripotency and will be useful in hPSC-based drug research and regenerative medicine.


Asunto(s)
Celulosa/química , Hidrogeles/química , Células Madre Pluripotentes Inducidas/fisiología , Nanofibras/química , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Medios de Cultivo/química , Proteínas de Homeodominio/metabolismo , Humanos , Cariotipo , Mitocondrias/metabolismo , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Esferoides Celulares/fisiología
14.
Neuron ; 73(4): 729-42, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22365547

RESUMEN

Following damage to peripheral nerves, a remarkable process of clearance and regeneration takes place. Axons downstream of the injury degenerate, while the nerve is remodeled to direct axonal regrowth. Schwann cells are important for this regenerative process. "Sensing" damaged axons, they dedifferentiate to a progenitor-like state, in which they aid nerve regeneration. Here, we demonstrate that activation of an inducible Raf-kinase transgene in myelinated Schwann cells is sufficient to control this plasticity by inducing severe demyelination in the absence of axonal damage, with the period of demyelination/ataxia determined by the duration of Raf activation. Remarkably, activation of Raf-kinase also induces much of the inflammatory response important for nerve repair, including breakdown of the blood-nerve barrier and the influx of inflammatory cells. This reversible in vivo model identifies a central role for ERK signaling in Schwann cells in orchestrating nerve repair and is a powerful system for studying peripheral neuropathies and cancer.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Regeneración Nerviosa/genética , Traumatismos de los Nervios Periféricos/fisiopatología , Proteínas Proto-Oncogénicas c-raf/metabolismo , Células de Schwann/fisiología , Animales , Animales Recién Nacidos , Benzamidas/farmacología , Movimiento Celular/efectos de los fármacos , Ciclina D1/metabolismo , Citocinas/metabolismo , Difenilamina/análogos & derivados , Difenilamina/farmacología , Antagonistas de Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Leucocitos/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Mastocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Traumatismos de los Nervios Periféricos/patología , Proteínas Proto-Oncogénicas c-raf/genética , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptores de Estrógenos/genética , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/genética , Células de Schwann/ultraestructura , Linfocitos T/metabolismo , Linfocitos T/patología , Tamoxifeno/farmacología , Factores de Tiempo
15.
PLoS One ; 5(7): e11774, 2010 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-20668675

RESUMEN

Anchorage-independent proliferation is a hallmark of oncogenic transformation and is thought to be conducive to proliferation of cancer cells away from their site of origin. We have previously reported that primary Schwann cells expressing the SV40 Large T antigen (LT) are not fully transformed in that they maintain a strict requirement for attachment, requiring a further genetic change, such as oncogenic Ras, to gain anchorage-independence. Using the LT-expressing cells, we performed a genetic screen for anchorage-independent proliferation and identified Sensory and Motor Neuron Derived Factor (SMDF), a transmembrane class III isoform of Neuregulin 1. In contrast to oncogenic Ras, SMDF induced enhanced proliferation in normal primary Schwann cells but did not trigger cellular senescence. In cooperation with LT, SMDF drove anchorage-independent proliferation, loss of contact inhibition and tumourigenicity. This transforming ability was shared with membrane-bound class III but not secreted class I isoforms of Neuregulin, indicating a distinct mechanism of action. Importantly, we show that despite being membrane-bound signalling molecules, class III neuregulins transform via a cell intrinsic mechanism, as a result of constitutive, elevated levels of ErbB signalling at high cell density and in anchorage-free conditions. This novel transforming mechanism may provide new targets for cancer therapy.


Asunto(s)
Membrana Celular/metabolismo , Neurregulina-1/metabolismo , Isoformas de Proteínas/metabolismo , Células de Schwann/metabolismo , Animales , Antígenos Virales de Tumores/metabolismo , Western Blotting , Proliferación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Humanos , Neurregulina-1/genética , Isoformas de Proteínas/genética , Ratas , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Schwann/citología
16.
J Cell Biol ; 181(4): 625-37, 2008 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-18490512

RESUMEN

Schwann cell myelination depends on Krox-20/Egr2 and other promyelin transcription factors that are activated by axonal signals and control the generation of myelin-forming cells. Myelin-forming cells remain remarkably plastic and can revert to the immature phenotype, a process which is seen in injured nerves and demyelinating neuropathies. We report that c-Jun is an important regulator of this plasticity. At physiological levels, c-Jun inhibits myelin gene activation by Krox-20 or cyclic adenosine monophosphate. c-Jun also drives myelinating cells back to the immature state in transected nerves in vivo. Enforced c-Jun expression inhibits myelination in cocultures. Furthermore, c-Jun and Krox-20 show a cross-antagonistic functional relationship. c-Jun therefore negatively regulates the myelinating Schwann cell phenotype, representing a signal that functionally stands in opposition to the promyelin transcription factors. Negative regulation of myelination is likely to have significant implications for three areas of Schwann cell biology: the molecular analysis of plasticity, demyelinating pathologies, and the response of peripheral nerves to injury.


Asunto(s)
Vaina de Mielina/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Animales Recién Nacidos , Desdiferenciación Celular/efectos de los fármacos , Técnicas de Cocultivo , AMP Cíclico/farmacología , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Proteínas HMGB/metabolismo , MAP Quinasa Quinasa 7/metabolismo , Ratones , Proteínas de la Mielina/metabolismo , Vaina de Mielina/patología , Factor 6 de Transcripción de Unión a Octámeros/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/genética , Ratas , Factores de Transcripción SOXB1 , Células de Schwann/efectos de los fármacos , Células de Schwann/enzimología , Células de Schwann/patología , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Degeneración Walleriana/patología
17.
Genes Dev ; 22(23): 3335-48, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19056885

RESUMEN

Neurofibromatosis type 1 (NF1) patients develop neurofibromas, tumors of Schwann cell origin, as a result of loss of the Ras-GAP neurofibromin. In normal nerves, Schwann cells are found tightly associated with axons, while loss of axonal contact is a frequent and important early event in neurofibroma development. However, the molecular basis of this physical interaction or how it is disrupted in cancer remains unclear. Here we show that loss of neurofibromin in Schwann cells is sufficient to disrupt Schwann cell/axonal interactions via up-regulation of the Ras/Raf/ERK signaling pathway. Importantly, we identify down-regulation of semaphorin 4F (Sema4F) as the molecular mechanism responsible for the Ras-mediated loss of interactions. In heterotypic cocultures, Sema4F knockdown induced Schwann cell proliferation by relieving axonal contact-inhibitory signals, providing a mechanism through which loss of axonal contact contributes to tumorigenesis. Importantly, Sema4F levels were strongly reduced in a panel of human neurofibromas, confirming the relevance of these findings to the human disease. This work identifies a novel role for the guidance-molecules semaphorins in the mediation of Schwann cell/axonal interactions, and provides a molecular mechanism by which heterotypic cell-cell contacts control cell proliferation and suppress tumorigenesis. Finally, it provides a new approach for the development of therapies for NF1.


Asunto(s)
Axones/fisiología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurofibromina 1/fisiología , Células de Schwann/fisiología , Animales , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Humanos , Ratones , Ratones Transgénicos , Neurofibroma/metabolismo , Ratas , Ratas Sprague-Dawley , Células de Schwann/metabolismo , Semaforinas/fisiología
18.
J Biol Chem ; 279(22): 22803-8, 2004 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-15028712

RESUMEN

Adrenocorticotropic hormone can stimulate lipolysis and suppress leptin expression in murine adipocytes. These effects are mediated via the melanocortin 2 receptor (MC2-R), which is expressed when 3T3-L1 cells are induced to undergo adipogenesis. In this study, we have characterized the mc2-r promoter in the murine adipocyte, one of the few extra-adrenal sites of expression and a cell type that lacks steroidogenic factor 1 (SF-1), a transcription factor that is required for mc2-r expression in adrenal cells. Transcriptional regulation of the mc2-r in the absence of SF-1 was investigated by 5' deletion analysis of the murine mc2-r promoter in both undifferentiated and differentiated 3T3-L1 cells. The results revealed the presence of a 59-base pair regulatory region within the promoter containing an adipocyte-specific enhancer. The ability of this region to confer enhanced activity in the adipocyte was mapped to a peroxisome proliferator-response element (PPRE)-like sequence that bound to peroxisome proliferator-activated receptor gamma (PPARgamma) and its heterodimeric partner retinoid X receptor alpha (RXRalpha) in adipocyte nuclear extracts. Co-transfection of PPARgamma2/RXRalpha with the pMC2-R(-112/+105)GL3 reporter resulted in transcriptional activation in preadipocytes, and this response required an intact PPRE. Mutation of the PPRE to prevent PPARgamma/RXRalpha binding resulted in a complete abrogation of the pMC2-R(-112/+105)GL3 reporter activity in day 3 differentiated 3T3-L1 cells, demonstrating a key role played by this site in regulating MC2-R expression in the murine adipocyte. These data highlight a novel mechanism for mc2-r transcription, which may have significance in both adrenal and extra-adrenal sites of expression.


Asunto(s)
Adipocitos/fisiología , Receptor de Melanocortina Tipo 2/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Células 3T3-L1 , Adipocitos/citología , Animales , Diferenciación Celular/fisiología , Ratones , Regiones Promotoras Genéticas/genética , Factor Esteroidogénico 1 , Activación Transcripcional/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA