Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Toxicol Rep ; 6: 529-537, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31249786

RESUMEN

We previously used a chemical genetics approach with the larval zebrafish to identify small molecule inhibitors of tissue regeneration. This led to the discovery that glucocorticoids (GC) block early stages of tissue regeneration by the inappropriate activation of the glucocorticoid receptor (GR). We performed a microarray analysis to identify the changes in gene expression associated with beclomethasone dipropionate (BDP) exposure during epimorphic fin regeneration. Oncofetal cripto-1 showed > eight-fold increased expression in BDP-treated regenerates. We hypothesized that the mis-expression of cripto-1 was essential for BDP to block regeneration. Expression of cripto-1 was not elevated in GR morphants in the presence of BDP indicating that cripto-1 induction was GR-dependent. Partial translational suppression of Cripto-1 in the presence of BDP restored tissue regeneration. Retinoic acid exposure prevented increased cripto-1 expression and permitted regeneration in the presence of BDP. We demonstrated that BDP exposure increased cripto-1 expression in mouse embryonic stem cells and that regulation of cripto-1 by GCs is conserved in mammals.

3.
Oncogenesis ; 7(4): 35, 2018 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-29674704

RESUMEN

BET inhibitors exhibit broad activity in cancer models, making predictive biomarkers challenging to define. Here we investigate the biomarkers of activity of the clinical BET inhibitor GSK525762 (I-BET; I-BET762) across cancer cell lines and demonstrate that KRAS mutations are novel resistance biomarkers. This finding led us to combine BET with RAS pathway inhibition using MEK inhibitors to overcome resistance, which resulted in synergistic effects on growth and survival in RAS pathway mutant models as well as a subset of cell lines lacking RAS pathway mutations. GSK525762 treatment up-regulated p-ERK1/2 levels in both RAS pathway wild-type and mutant cell lines, suggesting that MEK/ERK pathway activation may also be a mechanism of adaptive BET inhibitor resistance. Importantly, gene expression studies demonstrated that the BET/MEK combination uniquely sustains down-regulation of genes associated with mitosis, leading to prolonged growth arrest that is not observed with either single agent therapy. These studies highlight a potential to enhance the clinical benefit of BET and MEK inhibitors and provide a strong rationale for clinical evaluation of BET/MEK combination therapies in cancer.

4.
Sci Signal ; 8(375): ra42, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25943352

RESUMEN

Receptor tyrosine kinase (RTK) signaling promotes the growth and progression of glioblastoma (GBM), a highly aggressive type of brain tumor. We previously reported that decreased miR-218 expression in GBM directly promotes RTK activity by increasing the expression of key RTKs and their signaling mediators, including the RTK epidermal growth factor receptor (EGFR), phospholipase C-γ1 (PLCγ1), and the kinases PIK3CA and ARAF. However, increased RTK signaling usually activates negative feedback mechanisms to maintain homeostasis. We found that decreased miR-218 expression in GBM cells also increased the expression of genes encoding additional upstream and downstream components of RTK signaling pathways, including the RTK platelet-derived growth factor receptor α (PDGFRα) and the kinases ribosomal S6 kinase 2 (RSK2) and S6 kinase 1 (S6K1), that collectively overrode the negative feedback mechanism. Furthermore, increased RTK signaling itself suppressed miR-218 expression. Mass spectrometry and DNA pull-down identified binding of signal transducer and activator of transcription 3 (STAT3) along with the transcriptional repressor BCL2-associated transcription factor 1 (BCLAF1) directly to the miR-218 locus. These data identify previously unknown feedback loops by which miR-218 repression promotes increased RTK signaling in high-grade gliomas.


Asunto(s)
Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , MicroARNs/metabolismo , ARN Neoplásico/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Línea Celular Tumoral , Receptores ErbB/genética , Glioblastoma/genética , Glioblastoma/patología , Humanos , MicroARNs/genética , ARN Neoplásico/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
5.
Mol Cancer Res ; 13(8): 1227-37, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25934697

RESUMEN

UNLABELLED: Glioblastoma multiforme (GBM) is notoriously resistant to therapy, and the development of a durable cure will require the identification of broadly relevant regulators of GBM cell tumorigenicity and survival. Here, we identify Sprouty2 (SPRY2), a known regulator of receptor tyrosine kinases (RTK), as one such regulator. SPRY2 knockdown reduced proliferation and anchorage-independent growth in GBM cells and slowed xenograft tumor growth in mice. SPRY2 knockdown also promoted cell death in response to coinhibition of the epidermal growth factor receptor (EGFR) and the c-MET receptor in GBM cells, an effect that involved regulation of the ability of the p38 mitogen-activated protein kinase (MAPK) to drive cell death in response to inhibitors. Analysis of data from clinical tumor specimens further demonstrated that SPRY2 protein is definitively expressed in GBM tissue, that SPRY2 expression is elevated in GBM tumors expressing EGFR variant III (EGFRvIII), and that elevated SPRY2 mRNA expression portends reduced GBM patient survival. Overall, these results identify SPRY2 and the pathways it regulates as novel candidate biomarkers and therapeutic targets in GBM. IMPLICATIONS: SPRY2, counter to its roles in other cancer settings, promotes glioma cell and tumor growth and cellular resistance to targeted inhibitors of oncogenic RTKs, thus making SPRY2 and the cell signaling processes it regulates potential novel therapeutic targets in glioma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Resistencia a Antineoplásicos , Glioblastoma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Adulto , Animales , Biomarcadores de Tumor , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Proliferación Celular , Fosfatasa 1 de Especificidad Dual/metabolismo , Fosfatasas de Especificidad Dual/metabolismo , Receptores ErbB/metabolismo , Femenino , Glioblastoma/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Trasplante de Neoplasias , Fosforilación , Proteínas Serina-Treonina Quinasas , ARN Mensajero/metabolismo
6.
Nature ; 513(7517): 251-5, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25043030

RESUMEN

Clear cell renal cell carcinoma (ccRCC), the most common form of kidney cancer, is characterized by elevated glycogen levels and fat deposition. These consistent metabolic alterations are associated with normoxic stabilization of hypoxia-inducible factors (HIFs) secondary to von Hippel-Lindau (VHL) mutations that occur in over 90% of ccRCC tumours. However, kidney-specific VHL deletion in mice fails to elicit ccRCC-specific metabolic phenotypes and tumour formation, suggesting that additional mechanisms are essential. Recent large-scale sequencing analyses revealed the loss of several chromatin remodelling enzymes in a subset of ccRCC (these included polybromo-1, SET domain containing 2 and BRCA1-associated protein-1, among others), indicating that epigenetic perturbations are probably important contributors to the natural history of this disease. Here we used an integrative approach comprising pan-metabolomic profiling and metabolic gene set analysis and determined that the gluconeogenic enzyme fructose-1,6-bisphosphatase 1 (FBP1) is uniformly depleted in over six hundred ccRCC tumours examined. Notably, the human FBP1 locus resides on chromosome 9q22, the loss of which is associated with poor prognosis for ccRCC patients. Our data further indicate that FBP1 inhibits ccRCC progression through two distinct mechanisms. First, FBP1 antagonizes glycolytic flux in renal tubular epithelial cells, the presumptive ccRCC cell of origin, thereby inhibiting a potential Warburg effect. Second, in pVHL (the protein encoded by the VHL gene)-deficient ccRCC cells, FBP1 restrains cell proliferation, glycolysis and the pentose phosphate pathway in a catalytic-activity-independent manner, by inhibiting nuclear HIF function via direct interaction with the HIF inhibitory domain. This unique dual function of the FBP1 protein explains its ubiquitous loss in ccRCC, distinguishing FBP1 from previously identified tumour suppressors that are not consistently mutated in all tumours.


Asunto(s)
Carcinoma de Células Renales/enzimología , Fructosa-Bifosfatasa/metabolismo , Neoplasias Renales/enzimología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/fisiopatología , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Fructosa-Bifosfatasa/química , Fructosa-Bifosfatasa/genética , Glucólisis , Humanos , Neoplasias Renales/genética , Neoplasias Renales/fisiopatología , Modelos Moleculares , NADP/metabolismo , Estructura Terciaria de Proteína , Porcinos
7.
J Cell Sci ; 127(Pt 16): 3555-67, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24951116

RESUMEN

Information from multiple signaling axes is integrated in the determination of cellular phenotypes. Here, we demonstrate this aspect of cellular decision making in glioblastoma multiforme (GBM) cells by investigating the multivariate signaling regulatory functions of the protein tyrosine phosphatase SHP2 (also known as PTPN11). Specifically, we demonstrate that the ability of SHP2 to simultaneously drive ERK1/2 and antagonize STAT3 pathway activities produces qualitatively different effects on the phenotypes of proliferation and resistance to EGFR and c-MET co-inhibition. Whereas the ERK1/2 and STAT3 pathways independently promote proliferation and resistance to EGFR and c-MET co-inhibition, SHP2-driven ERK1/2 activity is dominant in driving cellular proliferation and SHP2-mediated antagonism of STAT3 phosphorylation prevails in the promotion of GBM cell death in response to EGFR and c-MET co-inhibition. Interestingly, the extent of these SHP2 signaling regulatory functions is diminished in glioblastoma cells that express sufficiently high levels of the EGFR variant III (EGFRvIII) mutant, which is commonly expressed in GBM. In cells and tumors that express EGFRvIII, SHP2 also antagonizes the phosphorylation of EGFRvIII and c-MET and drives expression of HIF-1α and HIF-2α, adding complexity to the evolving understanding of the regulatory functions of SHP2 in GBM.


Asunto(s)
Proliferación Celular , Glioblastoma/enzimología , Sistema de Señalización de MAP Quinasas , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Femenino , Gefitinib , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/fisiopatología , Humanos , Indoles/administración & dosificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Desnudos , Fosforilación/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Quinazolinas/administración & dosificación , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Sulfonas/administración & dosificación
8.
Int J Mol Sci ; 15(2): 2172-90, 2014 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24481065

RESUMEN

Glioblastomas (GBM) are very aggressive and malignant brain tumors, with frequent relapses despite an appropriate treatment combining surgery, chemotherapy and radiotherapy. In GBM, hypoxia is a characteristic feature and activation of Hypoxia Inducible Factors (HIF-1α and HIF-2α) has been associated with resistance to anti-cancer therapeutics. Int6, also named eIF3e, is the "e" subunit of the translation initiation factor eIF3, and was identified as novel regulator of HIF-2α. Eukaryotic initiation factors (eIFs) are key factors regulating total protein synthesis, which controls cell growth, size and proliferation. The functional significance of Int6 and the effect of Int6/EIF3E gene silencing on human brain GBM has not yet been described and its role on the HIFs is unknown in glioma cells. In the present study, we show that Int6/eIF3e suppression affects cell proliferation, cell cycle and apoptosis of various GBM cells. We highlight that Int6 inhibition induces a diminution of proliferation through cell cycle arrest and increased apoptosis. Surprisingly, these phenotypes are independent of global cell translation inhibition and are accompanied by decreased HIF expression when Int6 is silenced. In conclusion, we demonstrate here that Int6/eIF3e is essential for proliferation and survival of GBM cells, presumably through modulation of the HIFs.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Factor 3 de Iniciación Eucariótica/genética , Glioblastoma/genética , Glioblastoma/mortalidad , Apoptosis/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Factor 3 de Iniciación Eucariótica/metabolismo , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Modelos Biológicos , Interferencia de ARN
9.
Proc Natl Acad Sci U S A ; 111(1): 291-6, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24368849

RESUMEN

Glioblastoma multiforme (GBM) and the mesenchymal GBM subtype in particular are highly malignant tumors that frequently exhibit regions of severe hypoxia and necrosis. Because these features correlate with poor prognosis, we investigated microRNAs whose expression might regulate hypoxic GBM cell survival and growth. We determined that the expression of microRNA-218 (miR-218) is decreased significantly in highly necrotic mesenchymal GBM, and orthotopic tumor studies revealed that reduced miR-218 levels confer GBM resistance to chemotherapy. Importantly, miR-218 targets multiple components of receptor tyrosine kinase (RTK) signaling pathways, and miR-218 repression increases the abundance and activity of multiple RTK effectors. This elevated RTK signaling also promotes the activation of hypoxia-inducible factor (HIF), most notably HIF2α. We further show that RTK-mediated HIF2α regulation is JNK dependent, via jun proto-oncogene. Collectively, our results identify an miR-218-RTK-HIF2α signaling axis that promotes GBM cell survival and tumor angiogenesis, particularly in necrotic mesenchymal tumors.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Mesodermo/metabolismo , MicroARNs/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/farmacología , Supervivencia Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Hipoxia , Ratones , Ratones Desnudos , Persona de Mediana Edad , Necrosis , Trasplante de Neoplasias , Neovascularización Patológica , Análisis de Secuencia por Matrices de Oligonucleótidos , Proto-Oncogenes Mas , Transducción de Señal , Adulto Joven
10.
Cancer Discov ; 4(1): 53-60, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24189146

RESUMEN

Inactivation of the von-Hippel Lindau (VHL) tumor suppressor gene occurs in 90% of human clear cell renal cell carcinomas (ccRCC) and leads to the stable expression of the hypoxia-inducible factors HIF1α and HIF2α. The constitutive expression of HIF1α in a majority of VHL-deficient tumors is counterintuitive, given that HIF1α functions as a tumor suppressor in ccRCC, whereas HIF2α clearly enhances tumor growth. We demonstrate here that miR-30c-2-3p and miR-30a-3p specifically bind and inhibit expression of HIF2A transcripts, and that the locus encoding miR-30c-2-3p and miR-30a-3p is selectively repressed in "H1H2" VHL-deficient tumors expressing both HIF1α and HIF2α proteins. Inhibiting miR-30a-3p expression increases HIF2α levels in H1H2 ccRCC cells and promotes cellular proliferation, angiogenesis, and xenograft tumor growth. Our results indicate that miR-30c-2-3p and miR-30a-3p repression enhances HIF2α expression and suggests a mechanism whereby the tumor-suppressive effects of constitutive HIF1α expression are attenuated in VHL-deficient H1H2 tumors.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , MicroARNs/metabolismo , Animales , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/genética , Ratones , Ratones Desnudos , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-22634227

RESUMEN

Since their characterization, glucocorticoids (GCs), the most commonly prescribed immunomodulatory drugs, have undergone numerous structural modifications designed to enhance their activity. In vivo assessment of these corticosteroid analogs is essential to understand the difference in molecular signaling of the ligands that share the corticosteroid backbone. Our research identified a novel function of GCs as modulators of tissue regeneration and demonstrated that GCs activate the glucocorticoid receptor (GR) to inhibit early stages of tissue regeneration in zebrafish (Danio rerio). We utilized this phenomenon to assess the effect of different GC analogs on tissue regeneration and identified that some GCs such as beclomethasone dipropionate (BDP) possess inhibitory properties, while others, such as dexamethasone and hydrocortisone have no effect on regeneration. We performed in silico molecular docking and dynamic studies and demonstrated that type and size of substitution at the C17 position of the cortisol backbone confer a unique stable conformation to GR on ligand binding that is critical for inhibitory activity. In the field of tissue regeneration, our study is one of the first Structure Activity Relationship (SAR) investigations performed in vertebrates demonstrating that the in vivo tissue regeneration model is a powerful tool to probe structure function relationships, to understand regenerative biology, and to assist in rational drug design.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Regeneración/efectos de los fármacos , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Beclometasona/farmacología , Bases de Datos de Proteínas , Dexametasona/farmacología , Embrión no Mamífero/metabolismo , Glucocorticoides/farmacología , Humanos , Hidrocortisona/farmacología , Ligandos , Modelos Animales , Conformación Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Unión Proteica , Receptores de Glucocorticoides/genética , Alineación de Secuencia , Relación Estructura-Actividad , Pez Cebra/embriología , Pez Cebra/genética
12.
J Clin Invest ; 122(4): 1427-43, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22426208

RESUMEN

Localized tissue hypoxia is a consequence of vascular compromise or rapid cellular proliferation and is a potent inducer of compensatory angiogenesis. The oxygen-responsive transcriptional regulator hypoxia-inducible factor 2α (HIF-2α) is highly expressed in vascular ECs and, along with HIF-1α, activates expression of target genes whose products modulate vascular functions and angiogenesis. However, the mechanisms by which HIF-2α regulates EC function and tissue perfusion under physiological and pathological conditions are poorly understood. Using mice in which Hif2a was specifically deleted in ECs, we demonstrate here that HIF-2α expression is required for angiogenic responses during hindlimb ischemia and for the growth of autochthonous skin tumors. EC-specific Hif2a deletion resulted in increased vessel formation in both models; however, these vessels failed to undergo proper arteriogenesis, resulting in poor perfusion. Analysis of cultured HIF-2α-deficient ECs revealed cell-autonomous increases in migration, invasion, and morphogenetic activity, which correlated with HIF-2α-dependent expression of specific angiogenic factors, including delta-like ligand 4 (Dll4), a Notch ligand, and angiopoietin 2. By stimulating Dll4 signaling in cultured ECs or restoring Dll4 expression in ischemic muscle tissue, we rescued most of the HIF-2α-dependent EC phenotypes in vitro and in vivo, emphasizing the critical role of Dll4/Notch signaling as a downstream target of HIF-2α in ECs. These results indicate that HIF-1α and HIF-2α fulfill complementary, but largely nonoverlapping, essential functions in pathophysiological angiogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Circulación Colateral/fisiología , Células Endoteliales/metabolismo , Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Neovascularización Patológica/fisiopatología , Neoplasias Cutáneas/irrigación sanguínea , Proteínas Adaptadoras Transductoras de Señales , Angiopoyetina 2/genética , Angiopoyetina 2/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Unión al Calcio , Hipoxia de la Célula , Movimiento Celular , Células Cultivadas/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neovascularización Fisiológica/fisiología , Receptores Notch/fisiología , Proteínas Recombinantes de Fusión/fisiología , Recuperación de la Función , Neoplasias Cutáneas/inducido químicamente , Cicatrización de Heridas/fisiología
13.
PLoS One ; 5(10)2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20957046

RESUMEN

BACKGROUND: The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that mediates the toxicity and biological activity of dioxins and related chemicals. The AhR influences a variety of processes involved in cellular growth and differentiation, and recent studies have suggested that the AhR is a potential target for immune-mediated diseases. METHODOLOGY/PRINCIPAL FINDINGS: During a screen for molecules that activate the AhR, leflunomide, an immunomodulatory drug presently used in the clinic for the treatment of rheumatoid arthritis, was identified as an AhR agonist. We aimed to determine whether any biological activity of leflunomide could be attributed to a previously unappreciated interaction with the AhR. The currently established mechanism of action of leflunomide involves its metabolism to A771726, possibly by cytochrome P450 enzymes, followed by inhibition of de novo pyrimidine biosynthesis by A771726. Our results demonstrate that leflunomide, but not its metabolite A771726, caused nuclear translocation of AhR into the nucleus and increased expression of AhR-responsive reporter genes and endogenous AhR target genes in an AhR-dependent manner. In silico Molecular Docking studies employing AhR ligand binding domain revealed favorable binding energy for leflunomide, but not for A771726. Further, leflunomide, but not A771726, inhibited in vivo epimorphic regeneration in a zebrafish model of tissue regeneration in an AhR-dependent manner. However, suppression of lymphocyte proliferation by leflunomide or A771726 was not dependent on AhR. CONCLUSIONS: These data reveal that leflunomide, an anti-inflammatory drug, is an agonist of the AhR. Our findings link AhR activation by leflunomide to inhibition of fin regeneration in zebrafish. Identification of alternative AhR agonists is a critical step in evaluating the AhR as a therapeutic target for the treatment of immune disorders.


Asunto(s)
Antiinflamatorios/farmacología , Isoxazoles/farmacología , Receptores de Hidrocarburo de Aril/agonistas , Animales , Secuencia de Bases , Línea Celular , Citocromo P-450 CYP1A2/metabolismo , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Genes Reporteros , Leflunamida , Ligandos , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Regeneración , Pez Cebra/genética , Pez Cebra/fisiología
14.
Biol Reprod ; 82(6): 1227-36, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20181618

RESUMEN

Spermatogenesis, a process involving the differentiation of spermatogonial stem cells into mature spermatozoa, takes place throughout masculine life. A complex system in the testis, including endocrine signaling, physical interactions between germ and somatic cells, spermatocyte meiosis, and timely release of spermatozoa, controls this cycle. We demonstrate herein that decreased O(2) levels and Epas1 activation are critical components of spermatogenesis. Postnatal Epas1 ablation leads to male infertility, with reduced testis size and weight. While immature spermatogonia and spermatocytes are present in Epas1(Delta/Delta) testes, spermatid and spermatozoan numbers are dramatically reduced. This is not due to germ cell-intrinsic defects. Rather, Epas(Delta/Delta) Sertoli cells exhibit decreased ability to form tight junctions, thereby disrupting the blood-testis barrier necessary for proper spermatogenesis. Reduced numbers of tight junction complexes are due to decreased expression of multiple genes encoding tight junction proteins, including TJP1 (ZO1), TJP2 (ZO2), and occludin. Furthermore, Epas1(Delta/Delta) testes exhibit disrupted basement membranes surrounding the seminiferous tubules, causing the premature release of incompletely differentiated germ cells. We conclude that low O(2) levels in the male gonad regulate germ cell homeostasis in this organ via EPAS1.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Barrera Hematotesticular/metabolismo , Espermatogénesis , Espermatogonias/metabolismo , Testículo/metabolismo , Animales , Membrana Basal/química , Membrana Basal/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Barrera Hematotesticular/patología , Masculino , Proteínas de la Membrana/análisis , Ratones , Ocludina , Tamaño de los Órganos , Fosfoproteínas/análisis , Túbulos Seminíferos/química , Túbulos Seminíferos/patología , Células de Sertoli/química , Células de Sertoli/patología , Espermátides/metabolismo , Espermátides/patología , Espermatogonias/patología , Testículo/patología , Uniones Estrechas/química , Uniones Estrechas/patología , Proteína de la Zonula Occludens-1 , Proteína de la Zonula Occludens-2
15.
J Biol Chem ; 284(48): 33642-53, 2009 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-19801676

RESUMEN

Zebrafish have the remarkable ability to regenerate body parts including the heart and fins by a process referred to as epimorphic regeneration. Recent studies have illustrated that similar to adult zebrafish, early life stage larvae also possess the ability to regenerate the caudal fin. A comparative microarray analysis was used to determine the degree of conservation in gene expression among the regenerating adult caudal fin, adult heart, and larval fin. Results indicate that these tissues respond to amputation/injury with strikingly similar genomic responses. Comparative analysis revealed raldh2, a rate-limiting enzyme for the synthesis of retinoic acid, as one of the most highly induced genes across the three regeneration platforms. In situ localization and functional studies indicate that raldh2 expression is critical for the formation of wound epithelium and blastema. Patterning during regenerative outgrowth was considered to be the primary function of retinoic acid signaling; however, our results suggest that it is also required for early stages of tissue regeneration. Expression of raldh2 is regulated by Wnt and fibroblast growth factor/ERK signaling.


Asunto(s)
Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Retinal-Deshidrogenasa/genética , Proteínas de Pez Cebra/genética , Animales , Butadienos/farmacología , Análisis por Conglomerados , Embrión no Mamífero/embriología , Embrión no Mamífero/lesiones , Embrión no Mamífero/metabolismo , Extremidades/embriología , Extremidades/crecimiento & desarrollo , Extremidades/fisiología , Femenino , Hibridación in Situ , Larva/genética , Larva/crecimiento & desarrollo , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Nitrilos/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Pirroles/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Regeneración/efectos de los fármacos , Regeneración/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
17.
Biochem Pharmacol ; 77(4): 498-507, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18938144

RESUMEN

The origins of molecular toxicology can be traced to understanding the interactions between halogenated aromatic hydrocarbons and the aryl hydrocarbon receptor (AHR). The physiological consequences of activation of the aryl hydrocarbon receptor are diverse, and we are just beginning to understand the importance of the AHR signal transduction pathway in homeostasis and disease. The many downstream targets that mediate these biological responses remain undefined. Studies have exploited the power of the zebrafish model to elucidate the mechanisms by which AHR activation disrupts biological signaling. Recent genomic analysis performed in a zebrafish tissue regeneration model revealed functional cross talk between AHR and the well-established Wnt/beta-catenin signal transduction pathway. This review focuses on the development of the zebrafish model of AHR biology and the application of in vivo toxicogenomics to unravel molecular mechanisms.


Asunto(s)
Receptores de Hidrocarburo de Aril/fisiología , Regeneración/fisiología , Transducción de Señal , Proteínas Wnt/metabolismo , Animales , Extremidades/fisiología , Humanos , Modelos Biológicos , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Regeneración/genética , Reproducción/efectos de los fármacos , Reproducción/fisiología , Proteínas Wnt/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Pez Cebra/fisiología
18.
FASEB J ; 22(8): 3087-96, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18495758

RESUMEN

Exposure to dioxins, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), causes a wide array of toxicities in vertebrates, which are mostly considered to be mediated through the inappropriate activation of the aryl hydrocarbon receptor (AHR) signaling pathway. Although transcriptional regulation by AHR is widely studied, the molecular mechanisms responsible for the adverse outcomes after AHR activation are largely unknown. To identify the important downstream events of AHR activation, we employed the zebrafish caudal fin regeneration model, where AHR activation blocks the regenerative process. Comparative toxicogenomic analysis revealed that both adult and larval fins respond to TCDD during regeneration with misexpression of Wnt signaling pathway members and Wnt target genes. R-Spondin1, a novel ligand for the Wnt coreceptor, was highly induced, and we hypothesized that misexpression of R-Spondin1 is necessary for AHR activation to block regeneration. Partial antisense repression of R-Spondin1 reversed the inhibitory effect of TCDD, and tissue regeneration was restored. This finding demonstrates that inhibition of regeneration by TCDD is mediated by misinduction of R-Spondin1. Because R-Spondin1 signals through the Wnt coreceptor LRP6, we further demonstrated that the TCDD-mediated block in regeneration is also LRP6 dependent. Collectively, these results indicate that inappropriate regulation of R-Spondin/LRP6 is absolutely required for TCDD to inhibit fin regeneration.


Asunto(s)
Receptores de Hidrocarburo de Aril/fisiología , Regeneración/fisiología , Trombospondinas/fisiología , Proteínas Wnt/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/fisiología , Animales , Secuencia de Bases , ADN/genética , Contaminantes Ambientales/toxicidad , Expresión Génica/efectos de los fármacos , Marcación de Gen , Proteínas HMGB/deficiencia , Proteínas HMGB/genética , Proteínas HMGB/fisiología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Dibenzodioxinas Policloradas/toxicidad , Receptor Cross-Talk , Receptores de LDL/antagonistas & inhibidores , Receptores de LDL/genética , Receptores de LDL/fisiología , Regeneración/efectos de los fármacos , Factor de Transcripción SOX9 , Trombospondinas/genética , Toxicogenética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
19.
J Biol Chem ; 282(48): 35202-10, 2007 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-17848559

RESUMEN

Identifying the molecular pathways that are required for regeneration remains one of the great challenges of regenerative medicine. Although genetic mutations have been useful for identifying some molecular pathways, small molecule probes of regenerative pathways might offer some advantages, including the ability to disrupt pathway function with precise temporal control. However, a vertebrate regeneration model amenable to rapid throughput small molecule screening is not currently available. We report here the development of a zebrafish early life stage fin regeneration model and its use in screening for small molecules that modulate tissue regeneration. By screening 2000 biologically active small molecules, we identified 17 that specifically inhibited regeneration. These compounds include a cluster of glucocorticoids, and we demonstrate that transient activation of the glucocorticoid receptor is sufficient to block regeneration, but only if activation occurs during wound healing/blastema formation. In addition, knockdown of the glucocorticoid receptor restores regenerative capability to nonregenerative, glucocorticoid-exposed zebrafish. To test whether the classical anti-inflammatory action of glucocorticoids is responsible for blocking regeneration, we prevented acute inflammation following amputation by antisense repression of the Pu.1 gene. Although loss of Pu.1 prevents the inflammatory response, regeneration is not affected. Collectively, these results indicate that signaling from exogenous glucocorticoids impairs blastema formation and limits regenerative capacity through an acute inflammation-independent mechanism. These studies also demonstrate the feasibility of exploiting chemical genetics to define the pathways that govern vertebrate regeneration.


Asunto(s)
Técnicas Genéticas , Regeneración , Animales , Antiinflamatorios/farmacología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Relación Dosis-Respuesta a Droga , Extremidades/embriología , Glucocorticoides/metabolismo , Macrófagos/citología , Masculino , Modelos Anatómicos , Modelos Biológicos , Neutrófilos/metabolismo , Transducción de Señal , Cicatrización de Heridas , Pez Cebra
20.
Toxicol Sci ; 95(1): 215-26, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17003102

RESUMEN

Adult zebra fish completely regenerate their caudal (tail) fin following partial amputation. Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) inhibits this regenerative process. Proper regulation of transcription, innervation, vascularization, and extracellular matrix (ECM) composition is essential for complete fin regeneration. Previous microarray studies suggest that genes involved in ECM regulation are misexpressed following activation of the aryl hydrocarbon receptor. To investigate whether TCDD blocks regeneration by impairing ECM remodeling, male zebra fish were i.p. injected with 50 ng/g TCDD or vehicle, and caudal fins were amputated. By 3 days postamputation (dpa), the vascular network in the regenerating fin of TCDD-exposed fish was disorganized compared to vehicle-exposed animals. Furthermore, immunohistochemical staining revealed that axonal outgrowth was impacted by TCDD as early as 3 dpa. Histological analysis demonstrated that TCDD exposure leads to an accumulation of collagen at the end of the fin ray just distal to the amputation site by 3 dpa. Mature lepidotrichial-forming cells (fin ray-forming cells) were not observed in the fins of TCDD-treated fish. The capacity to metabolize ECM was also altered by TCDD exposure. Quantitative real-time PCR studies revealed that the aryl hydrocarbon pathway is active and that matrix-remodeling genes are expressed in the regenerate following TCDD exposure.


Asunto(s)
Carcinógenos Ambientales/toxicidad , Matriz Extracelular/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/agonistas , Regeneración/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Proteínas de Pez Cebra/metabolismo , Pez Cebra/fisiología , Animales , Colágeno/metabolismo , Citocromo P-450 CYP1A1/biosíntesis , Electroforesis en Gel de Poliacrilamida , Inducción Enzimática/efectos de los fármacos , Extremidades/anatomía & histología , Extremidades/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Masculino , Metaloproteinasas de la Matriz/metabolismo , Reacción en Cadena de la Polimerasa , Proteoglicanos/metabolismo , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Factores de Tiempo , Activador de Tejido Plasminógeno/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA