Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 5983, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37752135

RESUMEN

Resistance mechanisms to immune checkpoint blockade therapy (ICBT) limit its response duration and magnitude. Paradoxically, Interferon γ (IFNγ), a key cytokine for cellular immunity, can promote ICBT resistance. Using syngeneic mouse tumour models, we confirm that chronic IFNγ exposure confers resistance to immunotherapy targeting PD-1 (α-PD-1) in immunocompetent female mice. We observe upregulation of poly-ADP ribosyl polymerase 14 (PARP14) in chronic IFNγ-treated cancer cell models, in patient melanoma with elevated IFNG expression, and in melanoma cell cultures from ICBT-progressing lesions characterised by elevated IFNγ signalling. Effector T cell infiltration is enhanced in tumours derived from cells pre-treated with IFNγ in immunocompetent female mice when PARP14 is pharmacologically inhibited or knocked down, while the presence of regulatory T cells is decreased, leading to restoration of α-PD-1 sensitivity. Finally, we determine that tumours which spontaneously relapse in immunocompetent female mice following α-PD-1 therapy upregulate IFNγ signalling and can also be re-sensitised upon receiving PARP14 inhibitor treatment, establishing PARP14 as an actionable target to reverse IFNγ-driven ICBT resistance.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Melanoma , Femenino , Humanos , Animales , Ratones , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Receptor de Muerte Celular Programada 1 , Interferón gamma , Recurrencia Local de Neoplasia , Modelos Animales de Enfermedad , Poli(ADP-Ribosa) Polimerasas
2.
Cell Rep ; 39(12): 110995, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35732120

RESUMEN

Dysregulated cellular metabolism is a cancer hallmark for which few druggable oncoprotein targets have been identified. Increased fatty acid (FA) acquisition allows cancer cells to meet their heightened membrane biogenesis, bioenergy, and signaling needs. Excess FAs are toxic to non-transformed cells but surprisingly not to cancer cells. Molecules underlying this cancer adaptation may provide alternative drug targets. Here, we demonstrate that diacylglycerol O-acyltransferase 1 (DGAT1), an enzyme integral to triacylglyceride synthesis and lipid droplet formation, is frequently up-regulated in melanoma, allowing melanoma cells to tolerate excess FA. DGAT1 over-expression alone transforms p53-mutant zebrafish melanocytes and co-operates with oncogenic BRAF or NRAS for more rapid melanoma formation. Antagonism of DGAT1 induces oxidative stress in melanoma cells, which adapt by up-regulating cellular reactive oxygen species defenses. We show that inhibiting both DGAT1 and superoxide dismutase 1 profoundly suppress tumor growth through eliciting intolerable oxidative stress.


Asunto(s)
Diacilglicerol O-Acetiltransferasa , Melanoma , Animales , Diacilglicerol O-Acetiltransferasa/genética , Diacilglicerol O-Acetiltransferasa/metabolismo , Proteínas Oncogénicas/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno , Triglicéridos , Pez Cebra/metabolismo
3.
Curr Res Physiol ; 5: 216-223, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35637870

RESUMEN

Respirometry, based on oxygen uptake, is commonly employed for measuring metabolic rate. There is a growing need for metabolic rate measurements suitable for developmental studies, particularly in Danio rerio, where many important developmental stages occur at < 4 mm. However, respirometry becomes more challenging as the size of the organism reduces. Additionally, respirometry can be costly and require significant experience and technical knowledge which may prohibit uptake in non-specialist/non-physiology labs. Thus, using equipment routine in most developmental/molecular biology laboratories, we measured glucose uptake in 96-h post fertilisation (hpf) zebrafish larvae and compared it to stop-flow respirometry measures of oxygen uptake to test whether glucose uptake was a suitable alternative measure of metabolic rate. A Passing-Bablok regression revealed that within a 95% limit of agreement, the rate of glucose uptake and the rate of oxygen uptake were equivalent as measures of metabolic rate in 96 hpf Danio rerio larvae. Thus, the methodology we outline here may be a useful alternative or a complementary method for assessing metabolic rate in small organisms.

4.
J Immunol ; 205(4): 994-1008, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32641385

RESUMEN

Mucosal surfaces such as fish gills interface between the organism and the external environment and as such are major sites of foreign Ag encounter. In the gills, the balance between inflammatory responses to waterborne pathogens and regulatory responses toward commensal microbes is critical for effective barrier function and overall fish health. In mammals, IL-4 and IL-13 in concert with IL-10 are essential for balancing immune responses to pathogens and suppressing inflammation. Although considerable progress has been made in the field of fish immunology in recent years, whether the fish counterparts of these key mammalian cytokines perform similar roles is still an open question. In this study, we have generated IL-4/13A and IL-4/13B mutant zebrafish (Danio rerio) and, together with an existing IL-10 mutant line, characterized the consequences of loss of function of these cytokines. We demonstrate that IL-4/13A and IL-4/13B are required for the maintenance of a Th2-like phenotype in the gills and the suppression of type 1 immune responses. As in mammals, IL-10 appears to have a more striking anti-inflammatory function than IL-4-like cytokines and is essential for gill homeostasis. Thus, both IL-4/13 and IL-10 paralogs in zebrafish exhibit aspects of conserved function with their mammalian counterparts.


Asunto(s)
Proteínas de Peces/inmunología , Branquias/inmunología , Homeostasis/inmunología , Inflamación/inmunología , Interleucina-10/inmunología , Interleucina-4/inmunología , Pez Cebra/inmunología , Animales , Inmunidad/inmunología , Interleucina-13/inmunología , Mamíferos/inmunología
5.
Cancer Cell ; 31(5): 621-634.e6, 2017 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-28416184

RESUMEN

Aberrant WNT signaling drives colorectal cancer (CRC). Here, we identify TIAM1 as a critical antagonist of CRC progression through inhibiting TAZ and YAP, effectors of WNT signaling. We demonstrate that TIAM1 shuttles between the cytoplasm and nucleus antagonizing TAZ/YAP by distinct mechanisms in the two compartments. In the cytoplasm, TIAM1 localizes to the destruction complex and promotes TAZ degradation by enhancing its interaction with ßTrCP. Nuclear TIAM1 suppresses TAZ/YAP interaction with TEADs, inhibiting expression of TAZ/YAP target genes implicated in epithelial-mesenchymal transition, cell migration, and invasion, and consequently suppresses CRC cell migration and invasion. Importantly, high nuclear TIAM1 in clinical specimens associates with increased CRC patient survival. Together, our findings suggest that in CRC TIAM1 suppresses tumor progression by regulating YAP/TAZ activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular , Neoplasias Colorrectales/metabolismo , Células Epiteliales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Mucosa Intestinal/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfoproteínas/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células CACO-2 , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Citoplasma/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Mucosa Intestinal/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Fenotipo , Fosfoproteínas/genética , Proteolisis , Interferencia de ARN , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Transactivadores , Factores de Transcripción , Transcripción Genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Transfección , Vía de Señalización Wnt , Proteínas Señalizadoras YAP , Pez Cebra/embriología , Proteínas con Repetición de beta-Transducina/genética , Proteínas con Repetición de beta-Transducina/metabolismo
6.
J Immunol ; 197(9): 3520-3530, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27694495

RESUMEN

CD4+ T cells are at the nexus of the innate and adaptive arms of the immune system. However, little is known about the evolutionary history of CD4+ T cells, and it is unclear whether their differentiation into specialized subsets is conserved in early vertebrates. In this study, we have created transgenic zebrafish with vibrantly labeled CD4+ cells allowing us to scrutinize the development and specialization of teleost CD4+ leukocytes in vivo. We provide further evidence that CD4+ macrophages have an ancient origin and had already emerged in bony fish. We demonstrate the utility of this zebrafish resource for interrogating the complex behavior of immune cells at cellular resolution by the imaging of intimate contacts between teleost CD4+ T cells and mononuclear phagocytes. Most importantly, we reveal the conserved subspecialization of teleost CD4+ T cells in vivo. We demonstrate that the ancient and specialized tissues of the gills contain a resident population of il-4/13b-expressing Th2-like cells, which do not coexpress il-4/13a Additionally, we identify a contrasting population of regulatory T cell-like cells resident in the zebrafish gut mucosa, in marked similarity to that found in the intestine of mammals. Finally, we show that, as in mammals, zebrafish CD4+ T cells will infiltrate melanoma tumors and obtain a phenotype consistent with a type 2 immune microenvironment. We anticipate that this unique resource will prove invaluable for future investigation of T cell function in biomedical research, the development of vaccination and health management in aquaculture, and for further research into the evolution of adaptive immunity.


Asunto(s)
Enfermedades de los Peces/inmunología , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Melanoma/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Pez Cebra/inmunología , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Células Cultivadas , Branquias/inmunología , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Mamíferos , Sistema Mononuclear Fagocítico , Neoplasias Experimentales
7.
Oncotarget ; 7(26): 40348-40361, 2016 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-27248171

RESUMEN

Phenotype-guided re-profiling of approved drug molecules presents an accelerated route to developing anticancer therapeutics by bypassing the target-identification bottleneck of target-based approaches and by sampling drugs already in the clinic. Further, combinations incorporating targeted therapies can be screened for both efficacy and toxicity. Previously we have developed an oncogenic-RAS-driven zebrafish melanoma model that we now describe display melanocyte hyperplasia while still embryos. Having devised a rapid method for quantifying melanocyte burden, we show that this phenotype can be chemically suppressed by incubating V12RAS transgenic embryos with potent and selective small molecule inhibitors of either MEK or PI3K/mTOR. Moreover, we demonstrate that combining MEK inhibitors (MEKi) with dual PI3K/mTOR inhibitors (PI3K/mTORi) resulted in a super-additive suppression of melanocyte hyperplasia. The robustness and simplicity of our novel screening assay inspired us to perform a modest screen of FDA approved compounds for their ability to potentiate MEKi PD184352 or PI3K/mTORi NVPBEZ235 suppression of V12RAS-driven melanocyte hyperplasia. Through this route, we confirmed Rapamycin as a compound that could synergize with MEKi and even more so with PI3K/mTORi to suppress melanoma development, including suppressing the growth of cultured human melanoma cells. Further, we discovered two additional compounds-Disulfiram and Tanshinone-that also co-operate with MEKi to suppress the growth of transformed zebrafish melanocytes and showed activity toward cultured human melanoma cells. In conclusion, we provide proof-of-concept that our phenotype-guided screen could be used to identify compounds that affect melanoma development and prompt further evaluation of Disulfiram and Tanshinone as possible partners for combination therapy.


Asunto(s)
Reposicionamiento de Medicamentos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Abietanos/administración & dosificación , Animales , Animales Modificados Genéticamente , Apoptosis/efectos de los fármacos , Benzamidas/química , Línea Celular Tumoral , Modelos Animales de Enfermedad , Disulfiram/administración & dosificación , Inhibidores Enzimáticos/química , Regulación Neoplásica de la Expresión Génica , Humanos , Melaninas/química , Melanocitos/citología , Oligonucleótidos Antisentido/genética , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Sirolimus/química , Pez Cebra
8.
Oncotarget ; 7(26): 39654-39670, 2016 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-27166257

RESUMEN

Mutations affecting Gαq proteins are pervasive in uveal melanoma (UM), suggesting they 'drive' UM pathogenesis. The ERK1/2-MAPK pathway is critical for cutaneous melanoma development and consequently an important therapeutic target. Defining the contribution of ERK1/2-MAPK signalling to UM development has been hampered by the lack of an informative animal model that spontaneously develops UM. Towards this end, we engineered transgenic zebrafish to express oncogenic GNAQQ209P in the melanocyte lineage. This resulted in hyperplasia of uveal melanocytes, but with no evidence of malignant progression, nor perturbation of skin melanocytes. Combining expression of oncogenic GNAQQ209P with p53 inactivation resulted in earlier onset and even more extensive hyperplasia of uveal melanocytes that progressed to UM. Immunohistochemistry revealed only weak immunoreactivity to phosphorylated (p)ERK1/2 in established uveal tumours-in contrast to strong immunoreactivity in oncogenic RAS-driven skin lesions-but ubiquitous positive staining for nuclear Yes-associated protein (YAP). Moreover, no changes were observed in pERK1/2 levels upon transient knockdown of GNAQ or phospholipase C-beta (PLC-ß) inhibition in the majority of human UM cell lines we tested harbouring GNAQ mutations. In summary, our findings demonstrate a weak correlation between oncogenic GNAQQ209P mutation and sustained ERK1/2-MAPK activation, implying that ERK1/2 signalling is unlikely to be instrumental in the maintenance of GNAQQ209P-driven UMs.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Neoplasias de la Úvea/enzimología , Animales , Animales Modificados Genéticamente , Carcinógenos , Línea Celular , Linaje de la Célula , Elementos Transponibles de ADN , Perfilación de la Expresión Génica , Genes p53 , Humanos , Inmunohistoquímica , Melanocitos/metabolismo , Mutación , Transducción de Señal , Neoplasias de la Úvea/genética , Pez Cebra
9.
Cell Rep ; 10(1): 88-102, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25543140

RESUMEN

The E3 ubiquitin ligase HUWE1, deregulated in carcinoma, has been implicated in tumor formation. Here, we uncover a role for HUWE1 in cell migration and invasion through degrading the RAC activator TIAM1, implying an additional function in malignant progression. In MDCKII cells in response to HGF, HUWE1 catalyzes TIAM1 ubiquitylation and degradation predominantly at cell-cell adhesions, facilitating junction disassembly, migration, and invasion. Depleting HUWE1 or mutating the TIAM1 ubiquitylation site prevents TIAM1 degradation, antagonizing scattering, and invasion. Moreover, simultaneous depletion of TIAM1 restores migration and invasion in HUWE1-depleted cells. Significantly, we show that HUWE1 stimulates human lung cancer cell invasion through regulating TIAM1 stability. Finally, we demonstrate that HUWE1 and TIAM1 protein levels are inversely correlated in human lung carcinomas. Thus, we elucidate a critical role for HUWE1 in regulating epithelial cell-cell adhesion and provide additional evidence that ubiquitylation contributes to spatiotemporal control of RAC.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Neoplasias Pulmonares/genética , Invasividad Neoplásica/genética , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Carcinogénesis , Adhesión Celular/genética , Movimiento Celular/genética , Perros , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Neoplasias Pulmonares/patología , Células de Riñón Canino Madin Darby , Invasividad Neoplásica/patología , Proteolisis , Transducción de Señal/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética , Proteínas de Unión al GTP rac/metabolismo
10.
Hum Mol Genet ; 21(8): 1744-59, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22210625

RESUMEN

Lowe syndrome, which is characterized by defects in the central nervous system, eyes and kidneys, is caused by mutation of the phosphoinositide 5-phosphatase OCRL1. The mechanisms by which loss of OCRL1 leads to the phenotypic manifestations of Lowe syndrome are currently unclear, in part, owing to the lack of an animal model that recapitulates the disease phenotype. Here, we describe a zebrafish model for Lowe syndrome using stable and transient suppression of OCRL1 expression. Deficiency of OCRL1, which is enriched in the brain, leads to neurological defects similar to those reported in Lowe syndrome patients, namely increased susceptibility to heat-induced seizures and cystic brain lesions. In OCRL1-deficient embryos, Akt signalling is reduced and there is both increased apoptosis and reduced proliferation, most strikingly in the neural tissue. Rescue experiments indicate that catalytic activity and binding to the vesicle coat protein clathrin are essential for OCRL1 function in these processes. Our results indicate a novel role for OCRL1 in neural development, and support a model whereby dysregulation of phosphoinositide metabolism and clathrin-mediated membrane traffic leads to the neurological symptoms of Lowe syndrome.


Asunto(s)
Encéfalo/embriología , Modelos Animales de Enfermedad , Síndrome Oculocerebrorrenal , Monoéster Fosfórico Hidrolasas/genética , Proteínas de Pez Cebra/genética , Pez Cebra , Animales , Encéfalo/patología , Supervivencia Celular , Clatrina/metabolismo , Embrión no Mamífero , Desarrollo Embrionario , Endosomas/metabolismo , Perfilación de la Expresión Génica , Aparato de Golgi/metabolismo , Calor , Fosfatidilinositol 4,5-Difosfato/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Empalme de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Convulsiones/fisiopatología , Transducción de Señal , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/metabolismo
11.
Proc Natl Acad Sci U S A ; 108(3): 1099-103, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21199949

RESUMEN

T2-family acidic endoribonucleases are represented in all genomes. A physiological role for RNase T2 has yet to be defined for metazoa. RNASET2 mutation in humans is linked with a leukoencephalopathy that arises in infancy characterized by cortical cysts and multifocal white matter lesions. We now show localization of RNASET2 within lysosomes. Further, we demonstrate that loss of rnaset2 in mutant zebrafish results in accumulation of undigested rRNA within lysosomes within neurons of the brain. Further, by using high field intensity magnetic resonance microimaging, we reveal white matter lesions in these animals comparable to those observed in RNASET2-deficient infants. This correlates with accumulation of Amyloid precursor protein and astrocytes at sites of neurodegeneration. Thus we conclude that familial cystic leukoencephalopathy is a lysosomal storage disorder in which rRNA is the best candidate for the noxious storage material.


Asunto(s)
Leucoencefalopatías/genética , Enfermedades por Almacenamiento Lisosomal/genética , Lisosomas/metabolismo , Estabilidad del ARN/fisiología , ARN Ribosómico/metabolismo , Ribonucleasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Pez Cebra/genética , Animales , Encéfalo/metabolismo , Línea Celular , Clonación Molecular , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Hibridación in Situ , Imagen por Resonancia Magnética , Microscopía Electrónica de Transmisión , Neuronas/metabolismo , Neuronas/patología , Estabilidad del ARN/genética , Ribonucleasas/genética , Proteínas Supresoras de Tumor/genética
12.
Dis Model Mech ; 2(7-8): 399-411, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19470611

RESUMEN

Deregulated Ras signalling is implicated in most human neoplasia, exemplified by melanoma. Whereas Raf activation occurs almost ubiquitously in benign and malignant melanocytic neoplasms, implying an involvement in tumour initiation, phosphoinositide 3-kinase (PI3K) activation occurs predominantly in malignant neoplasms, implying an involvement in malignant progression. Here, we dissect the contributions of these two pathways to tumourigenesis in vivo, by modulating their activities in zebrafish melanocytes. Misexpression of oncogenic Ras (V12RAS) in founder fish induced frequent melanoma, beginning at larval stages, with concomitant activation of Raf-Mek-Erk and PI3K-Akt signalling. Misexpression of effector-domain mutants of V12RAS, or of various downstream effectors, confirmed a selective role for the Raf-Mek-Erk pathway in initiating neoplasia, but highlighted the requirement for additional Ras effector pathways for malignancy. The phenotype of animals with germ-line transmission of V12RAS resembled familial atypical mole and melanoma (FAMM) syndrome: melanocytes displayed hyperplasia, dysplasia, altered terminal differentiation and spontaneously progressed to invasive melanoma. Co-expressing a dominant-interfering form of PI3K abolished V12RAS-induced malignancy, demonstrating a direct role for PI3K signalling in the malignant progression of melanoma in vivo, and highlighting PI3K as a promising target for melanoma therapy.


Asunto(s)
Melanoma/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Quinasas raf/metabolismo , Animales , Diferenciación Celular , Línea Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Mutación , Invasividad Neoplásica , Isoformas de Proteínas , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Pez Cebra
13.
EMBO Rep ; 8(10): 966-73, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17823612

RESUMEN

Previous studies have shown that Wnt signals, relayed through beta-catenin and T-cell factor 4 (Tcf4), are essential for the induction and maintenance of crypts in mice. We have now generated a tcf4 (tcf7l2) mutant zebrafish by reverse genetics. We first observe a phenotypic defect at 4 weeks post-fertilization (wpf), leading to death at about 6 wpf. The phenotype comprises a loss of proliferation at the base of the intestinal folds of the middle and distal parts of the intestine. The proximal intestine represents an independent compartment, as it expresses sox2 in the epithelium and barx1 in the surrounding mesenchyme, which are early stomach markers in higher vertebrates. Zebrafish are functionally stomach-less, but the proximal intestine might share its ontogeny with the mammalian stomach. Rare adult homozygous tcf4(-/-) 'escapers' show proliferation defects in the gut epithelium, but have no other obvious abnormalities. This study underscores the involvement of Tcf4 in maintaining proliferative self-renewal in the intestine throughout life.


Asunto(s)
Proliferación Celular , Mucosa Intestinal/metabolismo , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Animales , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Inmunohistoquímica , Hibridación in Situ , Intestinos/citología , Masculino , Mutación , Fenotipo , Factores de Tiempo , Factor de Transcripción 4 , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Pez Cebra , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/fisiología
14.
BMC Cell Biol ; 7: 24, 2006 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-16787535

RESUMEN

BACKGROUND: Integrins comprise a large family of alpha,beta heterodimeric, transmembrane cell adhesion receptors that mediate diverse essential biological functions. Higher vertebrates possess a single beta1 gene, and the beta1 subunit associates with a large number of alpha subunits to form the major class of extracellular matrix (ECM) receptors. Despite the fact that the zebrafish (Danio rerio) is a rapidly emerging model organism of choice for developmental biology and for models of human disease, little is currently known about beta1 integrin sequences and functions in this organism. RESULTS: Using RT-PCR, complete coding sequences of zebrafish beta1 paralogs were obtained from zebrafish embryos or adult tissues. The results show that zebrafish possess two beta1 paralogs (beta1-1 and beta1-2) that have a high degree of identity to other vertebrate beta1 subunits. In addition, a third, more divergent, beta1 paralog is present (beta1-3), which may have altered ligand-binding properties. Zebrafish also have other divergent beta1-like transcripts, which are C-terminally truncated forms lacking the transmembrane and cytoplasmic domains. Together with beta1-3 these truncated forms comprise a novel group of beta1 paralogs, all of which have a mutation in the ADMIDAS cation-binding site. Phylogenetic and genomic analyses indicate that the duplication that gave rise to beta1-1 and beta1-2 occurred after the divergence of the tetrapod and fish lineages, while a subsequent duplication of the ancestor of beta1-2 may have given rise to beta1-3 and an ancestral truncated paralog. A very recent tandem duplication of the truncated beta1 paralogs appears to have taken place. The different zebrafish beta1 paralogs have varied patterns of temporal expression during development. Beta1-1 and beta1-2 are ubiquitously expressed in adult tissues, whereas the other beta1 paralogs generally show more restricted patterns of expression. CONCLUSION: Zebrafish have a large set of integrin beta1 paralogs. beta1-1 and beta1-2 may share the roles of the solitary beta1 subunit found in other vertebrates, whereas beta1-3 and the truncated beta1 paralogs may have acquired novel functions.


Asunto(s)
Integrina beta1/genética , Familia de Multigenes/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Evolución Molecular , Etiquetas de Secuencia Expresada , Duplicación de Gen , Regulación del Desarrollo de la Expresión Génica , Humanos , Integrina beta1/química , Integrina beta1/fisiología , Modelos Moleculares , Datos de Secuencia Molecular , Especificidad de Órganos , Filogenia , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Vertebrados/genética , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/fisiología
15.
J Biol Chem ; 281(1): 543-8, 2006 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-16249175

RESUMEN

Mutations in the canonical Wnt signaling pathway leading to its activation are known to cause the majority of intestinal tumors. However, few genes targeted by this pathway have been demonstrated to affect tumor development in vivo. Here we show that Tiam1, a selective Rac GTPase activator, is a Wnt-responsive gene expressed in the base of intestinal crypts and up-regulated in mouse intestinal tumors and human colon adenomas. Moreover, by comparing tumor development in APC mutant Min (multiple intestinal neoplasia) mice expressing or lacking Tiam1, we found that Tiam1 deficiency significantly reduces the formation and growth of polyps in vivo. However, invasion of malignant intestinal tumors is enhanced by a lack of Tiam1. In line with this, knock-down of Tiam1 reduced the growth potential of human colorectal cancer cells and their ability to form E-cadherin-based adhesions, a prerequisite for local invasion of tumor cells. Our data indicate a novel cross-talk between Tiam1-Rac and canonical Wnt-signaling pathways that influences intestinal tumor formation and progression.


Asunto(s)
Adenoma/metabolismo , Neoplasias Colorrectales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Adenoma/genética , Adenoma/patología , Animales , Células Cultivadas , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas de Neoplasias/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Transfección , Proteínas de Unión al GTP rac/metabolismo
16.
Nature ; 425(6958): 633-7, 2003 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-14534590

RESUMEN

Truncation of the tumour suppressor adenomatous polyposis coli (Apc) constitutively activates the Wnt/beta-catenin signalling pathway. Apc has a role in development: for example, embryos of mice with truncated Apc do not complete gastrulation. To understand this role more fully, we examined the effect of truncated Apc on zebrafish development. Here we show that, in contrast to mice, zebrafish do complete gastrulation. However, mutant hearts fail to loop and form excessive endocardial cushions. Conversely, overexpression of Apc or Dickkopf 1 (Dkk1), a secreted Wnt inhibitor, blocks cushion formation. In wild-type hearts, nuclear beta-catenin, the hallmark of activated canonical Wnt signalling, accumulates only in valve-forming cells, where it can activate a Tcf reporter. In mutant hearts, all cells display nuclear beta-catenin and Tcf reporter activity, while valve markers are markedly upregulated. Concomitantly, proliferation and epithelial-mesenchymal transition, normally restricted to endocardial cushions, occur throughout the endocardium. Our findings identify a novel role for Wnt/beta-catenin signalling in determining endocardial cell fate.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Válvulas Cardíacas/embriología , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , División Celular , Linaje de la Célula , Proteínas del Citoesqueleto/genética , Regulación del Desarrollo de la Expresión Génica , Genes APC , Genotipo , Válvulas Cardíacas/anomalías , Válvulas Cardíacas/citología , Válvulas Cardíacas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Mutación/genética , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Transactivadores/genética , Proteínas Wnt , Pez Cebra/genética , Proteínas de Pez Cebra/genética , beta Catenina
17.
Biochem J ; 364(Pt 1): 255-64, 2002 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11988099

RESUMEN

A highly conserved multisubunit enzymic complex, SWI/SNF, participates in the regulation of eukaryote gene expression through its ability to remodel chromatin. While a single component of SWI/SNF, Swi2 or a related protein, can perform this function in vitro, the other components appear to modulate the activity and specificity of the complex in vivo. Here we describe the cloning of hELD/OSA1, a 189 KDa human homologue of Drosophila Eld/Osa protein, a constituent of Drosophila SWI/SNF. By comparing conserved peptide sequences in Eld/Osa homologues we define three domains common to all family members. A putative DNA binding domain, or ARID (AT-rich DNA-interacting domain), may function in targetting SWI/SNF to chromatin. Two other domains unique to Eld/Osa proteins, EHD1 and EHD2, map to the C-terminus. We show that EHD2 mediates binding to Brahma-related gene 1 (BRG1), a human homologue of yeast Swi2. EHD1 and EHD2 also appear capable of interacting with each other. Using an antibody raised against EHD2 of hELD/OSA1, we detected Eld/Osa1 in endogenous SWI/SNF complexes derived from mouse brain.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/metabolismo , Secuencia de Bases , Células COS , Bovinos , Cromatina/metabolismo , Clonación Molecular , ADN/metabolismo , ADN Helicasas , ADN Complementario/metabolismo , Drosophila , Epítopos , Humanos , Immunoblotting , Ratones , Datos de Secuencia Molecular , Péptidos/química , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Transfección , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA