Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 12(9)2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37174657

RESUMEN

Primary liver cancer is the third leading cause of cancer-related death worldwide. An increasing body of evidence suggests that the Hippo tumor suppressor pathway plays a critical role in restricting cell proliferation and determining cell fate during physiological and pathological processes in the liver. Merlin (Moesin-Ezrin-Radixin-like protein) encoded by the NF2 (neurofibromatosis type 2) gene is an upstream regulator of the Hippo signaling pathway. Targeting of Merlin to the plasma membrane seems to be crucial for its major tumor-suppressive functions; this is facilitated by interactions with membrane-associated proteins, including CD44 (cluster of differentiation 44). Mutations within the CD44-binding domain of Merlin have been reported in many human cancers. This study evaluated the relative contribution of CD44- and Merlin-dependent processes to the development and progression of liver tumors. To this end, mice with a liver-specific deletion of the Nf2 gene were crossed with Cd44-knockout mice and subjected to extensive histological, biochemical and molecular analyses. In addition, cells were isolated from mutant livers and analyzed by in vitro assays. Deletion of Nf2 in the liver led to substantial liver enlargement and generation of hepatocellular carcinomas (HCCs), intrahepatic cholangiocarcinomas (iCCAs), as well as mixed hepatocellular cholangiocarcinomas. Whilst deletion of Cd44 had no influence on liver size or primary liver tumor development, it significantly inhibited metastasis formation in Nf2-mutant mice. CD44 upregulates expression of integrin ß2 and promotes transendothelial migration of liver cancer cells, which may facilitate metastatic spreading. Overall, our results suggest that CD44 may be a promising target for intervening with metastatic spreading of liver cancer.


Asunto(s)
Neoplasias de los Conductos Biliares , Carcinoma Hepatocelular , Colangiocarcinoma , Receptores de Hialuranos , Neoplasias Hepáticas , Neurofibromatosis 2 , Animales , Humanos , Ratones , Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos , Carcinoma Hepatocelular/genética , Colangiocarcinoma/genética , Genes de la Neurofibromatosis 2 , Receptores de Hialuranos/genética , Neoplasias Hepáticas/genética , Neurofibromatosis 2/genética , Neurofibromina 2/genética , Neurofibromina 2/metabolismo
2.
Int J Mol Sci ; 23(15)2022 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-35955749

RESUMEN

Osteosarcoma is the most common type of pediatric bone tumor. Despite great advances in chemotherapy during the past decades, the survival rates of osteosarcoma patients remain unsatisfactory. Drug resistance is one of the main reasons, leading to treatment failure and poor prognosis. Previous reports correlated expression of cluster of differentiation 44 (CD44) with drug resistance and poor survival of osteosarcoma patients, however the underlying mechanisms are poorly defined. Here, we investigated the role of CD44 in the regulation of drug chemoresistance, using osteosarcoma cells isolated from mice carrying a mutation of the tumor suppressor neurofibromatosis type 2 (Nf2) gene. CD44 expression was knocked-down in the cells using CRISPR/Cas9 approach. Subsequently, CD44 isoforms and mutants were re-introduced to investigate CD44-dependent processes. Sensitivity to doxorubicin was analyzed in the osteosarcoma cells with modified CD44 expression by immunoblot, colony formation- and WST-1 assay. To dissect the molecular alterations induced by deletion of Cd44, RNA sequencing was performed on Cd44-positive and Cd44-negative primary osteosarcoma tissues isolated from Nf2-mutant mice. Subsequently, expression of candidate genes was evaluated by quantitative reverse transcription PCR (qRT-PCR). Our results indicate that CD44 increases the resistance of osteosarcoma cells to doxorubicin by up-regulating the levels of multidrug resistance (MDR) 1 protein expression, and suggest the role of proteolytically released CD44 intracellular domain, and hyaluronan interactions in this process. Moreover, high throughput sequencing analysis identified differential regulation of several apoptosis-related genes in Cd44-positive and -negative primary osteosarcomas, including p53 apoptosis effector related to PMP-22 (Perp). Deletion of Cd44 in osteosarcoma cells led to doxorubicin-dependent p53 activation and a profound increase in Perp mRNA expression. Overall, our results suggest that CD44 might be an important regulator of drug resistance and suggest that targeting CD44 can sensitize osteosarcoma to standard chemotherapy.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Ratones , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Osteosarcoma/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Nat Commun ; 12(1): 5887, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34620853

RESUMEN

TRIP6, a member of the ZYXIN-family of LIM domain proteins, is a focal adhesion component. Trip6 deletion in the mouse, reported here, reveals a function in the brain: ependymal and choroid plexus epithelial cells are carrying, unexpectedly, fewer and shorter cilia, are poorly differentiated, and the mice develop hydrocephalus. TRIP6 carries numerous protein interaction domains and its functions require homodimerization. Indeed, TRIP6 disruption in vitro (in a choroid plexus epithelial cell line), via RNAi or inhibition of its homodimerization, confirms its function in ciliogenesis. Using super-resolution microscopy, we demonstrate TRIP6 localization at the pericentriolar material and along the ciliary axoneme. The requirement for homodimerization which doubles its interaction sites, its punctate localization along the axoneme, and its co-localization with other cilia components suggest a scaffold/co-transporter function for TRIP6 in cilia. Thus, this work uncovers an essential role of a LIM-domain protein assembly factor in mammalian ciliogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Encéfalo/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Encéfalo/patología , Epéndimo/patología , Adhesiones Focales/metabolismo , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Interferencia de ARN , Transcriptoma
4.
Sci Rep ; 11(1): 12904, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34145356

RESUMEN

The process of myogenesis which operates during skeletal muscle regeneration involves the activation of muscle stem cells, the so-called satellite cells. These then give rise to proliferating progenitors, the myoblasts which subsequently exit the cell cycle and differentiate into committed precursors, the myocytes. Ultimately, the fusion of myocytes leads to myofiber formation. Here we reveal a role for the transcriptional co-regulator nTRIP6, the nuclear isoform of the LIM-domain protein TRIP6, in the temporal control of myogenesis. In an in vitro model of myogenesis, the expression of nTRIP6 is transiently up-regulated at the transition between proliferation and differentiation, whereas that of the cytosolic isoform TRIP6 is not altered. Selectively blocking nTRIP6 function results in accelerated early differentiation followed by deregulated late differentiation and fusion. Thus, the transient increase in nTRIP6 expression appears to prevent premature differentiation. Accordingly, knocking out the Trip6 gene in satellite cells leads to deregulated skeletal muscle regeneration dynamics in the mouse. Thus, dynamic changes in nTRIP6 expression contributes to the temporal control of myogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Diferenciación Celular/genética , Proteínas con Dominio LIM/genética , Desarrollo de Músculos/genética , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Expresión Génica , Inmunohistoquímica , Proteínas con Dominio LIM/metabolismo , Ratones , Mioblastos/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Factores de Transcripción/metabolismo
5.
Int J Cancer ; 147(9): 2564-2577, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32525563

RESUMEN

Merlin is a versatile tumor suppressor protein encoded by the NF2 gene. Several lines of evidence suggest that Merlin exerts its tumor suppressor activity, at least in part, by forming an inhibitory complex with cluster of differentiation 44 (CD44). Consistently, numerous NF2 mutations in cancer patients are predicted to perturb the interaction of Merlin with CD44. We hypothesized that disruption of the Merlin-CD44 complex through loss of Merlin, unleashes putative tumor- or metastasis-promoting functions of CD44. To evaluate the relevance of the Merlin-CD44 interaction in vivo, we compared tumor growth and progression in Cd44-positive and Cd44-negative Nf2-mutant mice. Heterozygous Nf2-mutant mice were prone to developing highly metastatic osteosarcomas. Importantly, while the absence of the Cd44 gene had no effect on the frequency of primary osteosarcoma development, it strongly diminished osteosarcoma metastasis formation in the Nf2-mutant mice. In vitro assays identified transendothelial migration as the most prominent cellular phenotype dependent on CD44. Adhesion to endothelial cells was blocked by interfering with integrin α4ß1 (very late antigen-4, VLA-4) on osteosarcoma cells and CD44 upregulated levels of integrin VLA-4 ß1 subunit. Among other putative functions of CD44, which may contribute to the metastatic behavior, the passage through the endothelial cells also appears to be critical in vivo, as CD44 significantly promoted formation of lung metastasis upon intravenous injection of osteosarcoma cells into immunocompromised mice. Altogether, our results strongly suggest that CD44 plays a metastasis-promoting role in the absence of Merlin.


Asunto(s)
Neoplasias Óseas/genética , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/genética , Neurofibromina 2/genética , Osteosarcoma/genética , Animales , Neoplasias Óseas/patología , Huesos/patología , Adhesión Celular/genética , Línea Celular Tumoral/trasplante , Proliferación Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Receptores de Hialuranos/genética , Pulmón/patología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Noqueados , Osteosarcoma/secundario
6.
Mol Carcinog ; 58(5): 621-626, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30582228

RESUMEN

Loss-of-function of RHAMM causes hypofertility and testicular atrophy in young mice, followed by germ cell neoplasia in situ (GCNIS) of the testis, cellular atypia, and development of the testicular germ cell tumor (TGCT) seminoma. These pathologies reflect the risk factors and phenotypes that precede seminoma development in humans and-given the high prevalence of RHAMM downregulation in human seminoma-link RHAMM dysfunction with the aetiology of male hypofertility and GCNIS-related TGCTs. The initiating event underlying these pathologies, in RHAMM mutant testis, is premature displacement of undifferentiated progenitors from the basal compartment. We hypothesized that cd44 (both cancer initiating cell- and oncogenic progression marker) will drive GCNIS development, induced by RHAMM-loss-of-function in the mouse. We report that cd44 is expressed in a specific subset of GCNIS testes. Its genetic deletion has no effect on GCNIS onset, but it ameliorates oncogenic progression. We conclude that cd44 expression, combined with RHAMM dysfunction, promotes oncogenic progression in the testis.


Asunto(s)
Carcinoma in Situ/prevención & control , Proteínas de la Matriz Extracelular/fisiología , Receptores de Hialuranos/fisiología , Infertilidad Masculina/prevención & control , Neoplasias de Células Germinales y Embrionarias/prevención & control , Lesiones Precancerosas/prevención & control , Neoplasias Testiculares/prevención & control , Animales , Biomarcadores de Tumor/genética , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Femenino , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de Células Germinales y Embrionarias/metabolismo , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Eliminación de Secuencia , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo
7.
PLoS One ; 13(12): e0207358, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30540779

RESUMEN

The adhesion molecule and co-receptor of receptor tyrosine kinases, CD44, is expressed in all cells of the immune system, but also in numerous non-immune cells. CD44 plays roles in the cellular response to different pathogens. The molecular actions of CD44 during these processes are by and large still unknown. The CD44 molecule undergoes a sequential proteolytic cleavage which leads to the release of a soluble intracellular domain (CD44-ICD). Previous reports had shown that the CD44-ICD is taken up into the nucleus where it enhances transcription of specific target genes. By RNA profiling we identified a CD44-dependent transcriptional increase of interferon-responsive genes, among them IFI16. IFI16 is important in the innate immune response. It senses and binds pathogenic DNA and, together with cGAS, activates the cGAS-cGAMP-STING pathway and induces the expression of genes relevant for the response, e.g. IFN-ß. Our results show that the enhancement of IFI16 expression depended on CD44 cleavage. A CD44-negative tumor cell line, embryonic fibroblasts and bone marrow-derived macrophages from cd44-/- mice were reduced in their response to IFN-γ, to viral DNA fragments and to Listeria monocytogenes infection. We could rescue the deficiency of CD44 negative RPM-MC cells and cd44-/- MEFs by expressing only the soluble CD44-ICD in the absence of any other CD44 domain. Expression of the CD44-ICD carrying a mutation that prevented the uptake into the nucleus, could not rescue the absence of CD44. This molecular aspect of regulation by CD44 may explain part of the immune phenotypes of mice with cd44 gene disruption.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Receptores de Hialuranos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Células Cultivadas , Diaminas/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Receptores de Hialuranos/genética , Inmunidad Innata/efectos de los fármacos , Interferón beta/genética , Interferón beta/metabolismo , Interferón gamma/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Melanoma/metabolismo , Melanoma/patología , Ratones , Mutagénesis , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Nucleares/genética , Fosfoproteínas/genética , Tiazoles/farmacología , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
8.
Stem Cell Reports ; 9(4): 1071-1080, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28943256

RESUMEN

Misoriented division of neuroprogenitors, by loss-of-function studies of centrosome or spindle components, has been linked to the developmental brain defects microcephaly and lissencephaly. As these approaches also affect centrosome biogenesis, spindle assembly, or cell-cycle progression, the resulting pathologies cannot be attributed solely to spindle misorientation. To address this issue, we employed a truncation of the spindle-orienting protein RHAMM. This truncation of the RHAMM centrosome-targeting domain does not have an impact on centrosome biogenesis or on spindle assembly in vivo. The RHAMM mutants exhibit misorientation of the division plane of neuroprogenitors, without affecting the division rate of these cells, resulting against expectation in megalencephaly associated with cerebral cortex thickening, cerebellum enlargement, and premature cerebellum differentiation. We conclude that RHAMM associates with the spindle of neuroprogenitor cells via its centrosome-targeting domain, where it regulates differentiation in the developing brain by orienting the spindle.


Asunto(s)
Cerebelo/citología , Corteza Cerebral/citología , Megalencefalia/etiología , Megalencefalia/patología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Huso Acromático/metabolismo , Animales , Diferenciación Celular , División Celular , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Expresión Génica , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Ratones , Neurogénesis , Organogénesis , Transporte de Proteínas
9.
Trends Cancer ; 3(7): 482-490, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28718403

RESUMEN

Identification of early cancer, detection of progression, and monitoring of therapeutic success remain urgent issues in clinical medicine, particularly given the increasing cancer incidence in our aging populations. New methodologies have provided enormous progress over the past decades by defining the genetic and proteomic composition of cancers, yielding putative cancer biomarkers detectable in blood or other body fluids less invasively and more cheaply than using currently available screening techniques that often involve biopsies or surgery. However, the clinical use of these new methodologies is still far off. In this review, we focus on putative soluble cancer biomarkers shed from the cell surface by metalloproteases overexpressed in numerous cancers. Although useful candidates have been identified, their validation and adoption into clinical use remain challenging.


Asunto(s)
Proteínas ADAM/metabolismo , Biomarcadores de Tumor/análisis , Detección Precoz del Cáncer/métodos , Proteínas de Neoplasias/análisis , Neoplasias/diagnóstico , Biomarcadores de Tumor/metabolismo , Líquidos Corporales/química , Membrana Celular/metabolismo , Monitoreo de Drogas/métodos , Humanos , Proteínas de Neoplasias/metabolismo , Neoplasias/patología , Péptidos/análisis , Péptidos/metabolismo , Proteolisis , Proteómica/métodos
10.
Sci Rep ; 6: 37464, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27876763

RESUMEN

Release of cytokines, growth factors and other life-essential molecules from precursors by a-disintegrin-and-metalloproteases (ADAMs) is regulated with high substrate-specificity. We hypothesized that this is achieved by cleavage-regulatory intracellular-domain (ICD)-modifications of the precursors. We show here that cleavage-stimuli-induced specific ICD-modifications cause structural substrate changes that enhance ectodomain sensitivity of neuregulin-1 (NRG1; epidermal-growth-factor) or CD44 (receptor-tyrosine-kinase (RTK) co-receptor) to chymotrypsin/trypsin or soluble ADAM. This inside-out signal transfer required substrate homodimerization and was prevented by cleavage-inhibitory ICD-mutations. In chimeras, regulation could be conferred to a foreign ectodomain, suggesting a common higher-order structure. We predict that substrate-specific protease-accessibility-regulation controls release of numerous ADAM substrates.


Asunto(s)
Proteínas ADAM/genética , Receptores de Hialuranos/genética , Neurregulina-1/genética , Proteínas ADAM/química , Animales , Quimera/genética , Receptores de Hialuranos/química , Ratones , Mutación , Células 3T3 NIH , Neurregulina-1/química , Péptido Hidrolasas/química , Péptido Hidrolasas/genética , Dominios Proteicos/genética , Especificidad por Sustrato/genética
11.
Cancer Res ; 76(21): 6382-6395, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27543603

RESUMEN

Hypofertility is a risk factor for the development of testicular germ cell tumors (TGCT), but the initiating event linking these pathologies is unknown. We hypothesized that excessive planar division of undifferentiated germ cells promotes their self-renewal and TGCT development. However, our results obtained from mouse models and seminoma patients demonstrated the opposite. Defective planar divisions of undifferentiated germ cells caused their premature exit from the seminiferous tubule niche, resulting in germ cell depletion, hypofertility, intratubular germ cell neoplasias, and seminoma development. Oriented divisions of germ cells, which determine their fate, were regulated by spindle-associated RHAMM-a function we found to be abolished in 96% of human seminomas. Mechanistically, RHAMM expression is regulated by the testis-specific polyadenylation protein CFIm25, which is downregulated in the human seminomas. These results suggested that spindle misorientation is oncogenic, not by promoting self-renewing germ cell divisions within the niche, but by prematurely displacing proliferating cells from their normal epithelial milieu. Furthermore, they suggested RHAMM loss-of-function and spindle misorientation as an initiating event underlying both hypofertility and TGCT initiation. These findings identify spindle-associated RHAMM as an intrinsic regulator of male germ cell fate and as a gatekeeper preventing initiation of TGCTs. Cancer Res; 76(21); 6382-95. ©2016 AACR.


Asunto(s)
Proteínas de la Matriz Extracelular/fisiología , Fertilidad , Receptores de Hialuranos/fisiología , Neoplasias de Células Germinales y Embrionarias/etiología , Seminoma/etiología , Huso Acromático/química , Neoplasias Testiculares/etiología , Testículo/citología , Animales , Apoptosis , División Celular , Proteínas de la Matriz Extracelular/análisis , Células HeLa , Humanos , Receptores de Hialuranos/análisis , Masculino , Ratones , Neoplasias de Células Germinales y Embrionarias/patología , Seminoma/patología , Neoplasias Testiculares/patología , Proteína p53 Supresora de Tumor/fisiología
12.
Mol Cell Biol ; 35(19): 3381-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26217011

RESUMEN

Ectodomain cleavage by A-disintegrin and -metalloproteases (ADAMs) releases many important biologically active substrates and is therefore tightly controlled. Part of the regulation occurs on the level of the enzymes and affects their cell surface abundance and catalytic activity. ADAM-dependent proteolysis occurs outside the plasma membrane but is mostly controlled by intracellular signals. However, the intracellular domains (ICDs) of ADAM10 and -17 can be removed without consequences for induced cleavage, and so far it is unclear how intracellular signals address cleavage. We therefore explored whether substrates themselves could be chosen for proteolysis via ICD modification. We report here that CD44 (ADAM10 substrate), a receptor tyrosine kinase (RTK) coreceptor required for cellular migration, and pro-NRG1 (ADAM17 substrate), which releases the epidermal growth factor (EGF) ligand neuregulin required for axonal outgrowth and myelination, are indeed posttranslationally modified at their ICDs. Tetradecanoyl phorbol acetate (TPA)-induced CD44 cleavage requires dephosphorylation of ICD serine 291, while induced neuregulin release depends on the phosphorylation of several NRG1-ICD serines, in part mediated by protein kinase Cδ (PKCδ). Downregulation of PKCδ inhibits neuregulin release and reduces ex vivo neurite outgrowth and myelination of trigeminal ganglion explants. Our results suggest that specific selection among numerous substrates of a given ADAM is determined by ICD modification of the substrate.


Asunto(s)
Receptores de Hialuranos/metabolismo , Neurregulina-1/metabolismo , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Técnicas de Cocultivo , Células HEK293 , Humanos , Receptores de Hialuranos/química , Datos de Secuencia Molecular , Neurregulina-1/química , Neuritas/fisiología , Fosforilación , Proteína Quinasa C-delta/metabolismo , Proteolisis , Células de Schwann/metabolismo , Transducción de Señal , Ganglio del Trigémino/citología
13.
J Biol Chem ; 290(28): 17041-54, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25925953

RESUMEN

Ectodomain shedding of transmembrane precursor proteins generates numerous life-essential molecules, such as epidermal growth factor receptor ligands. This cleavage not only releases the regulatory growth factor, but it is also the required first step for the subsequent processing by γ-secretase and the release of gene regulatory intracellular fragments. Signaling within the cell modifies the cytoplasmic tails of substrates, a step important in starting the specific and regulated cleavage of a large number of studied substrates. Ectodomain cleavage occurs, however, on the outside of the plasma membrane and is carried out by membrane-bound metalloproteases. How the intracellular domain modification communicates with the ectodomain of the substrate to allow for cleavage to occur is unknown. Here, we show that homodimerization of a cluster-of-differentiation-44 or of pro-neuregulin-1 monomers represents an essential pre-condition for their regulated ectodomain cleavage. Both substrates are associated with their respective metalloproteases under both basal or cleavage-stimulated conditions. These interactions only turn productive by specific intracellular signal-induced intracellular domain modifications of the substrates, which in turn regulate metalloprotease access to the substrates' ectodomain and cleavage. We propose that substrate intracellular domain modification induces a relative rotation or other positional change of the dimerization partners that allow metalloprotease cleavage in the extracellular space. Our findings fill an important gap in understanding substrate-specific inside-out signal transfer along cleaved transmembrane proteins and suggest that substrate dimerization (homo- or possibly heterodimerization) might represent a general principle in ectodomain shedding.


Asunto(s)
Receptores de Hialuranos/metabolismo , Neurregulina-1/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Proteína ADAM17 , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Bovinos , Línea Celular Tumoral , Membrana Celular/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Receptores de Hialuranos/química , Receptores de Hialuranos/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Células 3T3 NIH , Neurregulina-1/química , Neurregulina-1/genética , Multimerización de Proteína , Estructura Terciaria de Proteína , Proteolisis , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato
14.
Biol Open ; 4(4): 562-71, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25750434

RESUMEN

The postnatal mammalian ovary contains the primary follicles, each comprising an immature oocyte surrounded by a layer of somatic granulosa cells. Oocytes reach meiotic and developmental competence via folliculogenesis. During this process, the granulosa cells proliferate massively around the oocyte, form an extensive extracellular matrix (ECM) and differentiate into cumulus cells. As the ECM component hyaluronic acid (HA) is thought to form the backbone of the oocyte-granulosa cell complex, we deleted the relevant domain of the Receptor for HA Mediated Motility (RHAMM) gene in the mouse. This resulted in folliculogenesis defects and female hypofertility, although HA-induced signalling was not affected. We report that wild-type RHAMM localises at the mitotic spindle of granulosa cells, surrounding the oocyte. Deletion of the RHAMM C-terminus in vivo abolishes its spindle association, resulting in impaired spindle orientation in the dividing granulosa cells, folliculogenesis defects and subsequent female hypofertility. These data reveal the first identified physiological function for RHAMM, during oogenesis, and the importance of this spindle-associated function for female fertility.

15.
Mol Cancer Res ; 13(5): 879-90, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25652588

RESUMEN

UNLABELLED: Ectodomain cleavage (shedding) of transmembrane proteins by metalloproteases (MMP) generates numerous essential signaling molecules, but its regulation is not totally understood. CD44, a cleaved transmembrane glycoprotein, exerts both antiproliferative or tumor-promoting functions, but whether proteolysis is required for this is not certain. CD44-mediated contact inhibition and cellular proliferation are regulated by counteracting CD44 C-terminal interacting proteins, the tumor suppressor protein merlin (NF2) and ERM proteins (ezrin, radixin, moesin). We show here that activation or overexpression of constitutively active merlin or downregulation of ERMs inhibited 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced [as well as serum, hepatocyte growth factor (HGF), or platelet-derived growth factor (PDGF)] CD44 cleavage by the metalloprotease ADAM10, whereas overexpressed ERM proteins promoted cleavage. Merlin- and ERM-modulated Ras or Rac activity was not required for this function. However, latrunculin (an actin-disrupting toxin) or an ezrin mutant which is unable to link CD44 to actin, inhibited CD44 cleavage, identifying a cytoskeletal C-terminal link as essential for induced CD44 cleavage. Cellular migration, an important tumor property, depended on CD44 and its cleavage and was inhibited by merlin. These data reveal a novel function of merlin and suggest that CD44 cleavage products play a tumor-promoting role. Neuregulin, an EGF ligand released by ADAM17 from its pro-form NRG1, is predominantly involved in regulating cellular differentiation. In contrast to CD44, release of neuregulin from its pro-form was not regulated by merlin or ERM proteins. Disruption of the actin cytoskeleton however, also inhibited NRG1 cleavage. This current study presents one of the first examples of substrate-selective cleavage regulation. IMPLICATIONS: Investigating transmembrane protein cleavage and their regulatory pathways have provided new molecular insight into their important role in cancer formation and possible treatment.


Asunto(s)
Movimiento Celular/fisiología , Receptores de Hialuranos/metabolismo , Neurofibromina 2/metabolismo , Animales , Proliferación Celular/fisiología , Células Cultivadas , Fibroblastos , Humanos , Ratones , Ratones Transgénicos , Células 3T3 NIH , Células Tumorales Cultivadas
16.
PLoS One ; 9(5): e97549, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24819052

RESUMEN

Several LIM domain proteins regulate transcription. They are thought to act through their LIM protein-protein interaction domains as adaptors for the recruitment of transcriptional co-regulators. An intriguing example is nTRIP6, the nuclear isoform of the focal adhesion protein TRIP6. nTRIP6 interacts with AP-1 and enhances its transcriptional activity. nTRIP6 is also essential for the transrepression of AP-1 by the glucocorticoid receptor (GR), by mediating GR tethering to promoter-bound AP-1. Here we report on the molecular mechanism by which nTRIP6 exerts these effects. Both the LIM domains and the pre-LIM region of nTRIP6 are necessary for its co-activator function for AP-1. Discrete domains within the pre-LIM region mediate the dimerization of nTRIP6 at the promoter, which enables the recruitment of the Mediator complex subunits THRAP3 and Med1. This recruitment is blocked by GR, through a competition between GR and THRAP3 for the interaction with the LIM domains of nTRIP6. Thus, nTRIP6 both positively and negatively regulates transcription by orchestrating the recruitment of the Mediator complex to AP-1-regulated promoters.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas con Dominio LIM/metabolismo , Complejo Mediador/metabolismo , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas con Dominio LIM/química , Ratones , Regiones Promotoras Genéticas/genética , Complejo de la Endopetidasa Proteasomal , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Transporte de Proteínas , Receptores de Glucocorticoides/metabolismo , Factores de Transcripción/química , Transcripción Genética
17.
Oncotarget ; 5(8): 2176-86, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24742962

RESUMEN

The adenoviral oncoprotein E1A influences cellular regulation by interacting with a number of cellular proteins. In collaboration with complementary oncogenes, E1A fully transforms primary cells. As part of this action, E1A inhibits transcription of c-Jun:Fos target genes while promoting that of c-Jun:ATF2-dependent genes including jun. Both c-Jun and ATF2 are hyperphosphorylated in response to E1A. In the current study, E1A was fused with the ligand binding domain of the estrogen receptor (E1A-ER) to monitor the immediate effect of E1A activation. With this approach we now show that E1A activates c-Jun N-terminal kinase (JNK), the upstream kinases MKK4 and MKK7, as well as the small GTPase Rac1. Activation of the JNK pathway requires the N-terminal domain of E1A, and, importantly, is independent of transcription. In addition, it requires the presence of ERM proteins. Downregulation of signaling components upstream of JNK inhibits E1A-dependent JNK/c-Jun activation. Taking these findings together, we show that E1A activates the JNK/c-Jun signaling pathway upstream of Rac1 in a transcription-independent manner, demonstrating a novel mechanism of E1A action.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Transformación Celular Viral/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Animales , Línea Celular , Activación Enzimática/fisiología , Receptor alfa de Estrógeno/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Células 3T3 NIH , Proteínas de Fusión Oncogénica/metabolismo , ARN Interferente Pequeño , Transfección
18.
Proc Natl Acad Sci U S A ; 110(51): 20587-92, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297905

RESUMEN

Receptor tyrosine kinases participate in several signaling pathways through small G proteins such as Ras (rat sarcoma). An important component in the activation of these G proteins is Son of sevenless (SOS), which catalyzes the nucleotide exchange on Ras. For optimal activity, a second Ras molecule acts as an allosteric activator by binding to a second Ras-binding site within SOS. This allosteric Ras-binding site is blocked by autoinhibitory domains of SOS. We have reported recently that Ras activation also requires the actin-binding proteins ezrin, radixin, and moesin. Here we report the mechanism by which ezrin modulates SOS activity and thereby Ras activation. Active ezrin enhances Ras/MAPK signaling and interacts with both SOS and Ras in vivo and in vitro. Moreover, in vitro kinetic assays with recombinant proteins show that ezrin also is important for the activity of SOS itself. Ezrin interacts with GDP-Ras and with the Dbl homology (DH)/pleckstrin homology (PH) domains of SOS, bringing GDP-Ras to the proximity of the allosteric site of SOS. These actions of ezrin are antagonized by the neurofibromatosis type 2 tumor-suppressor protein merlin. We propose an additional essential step in SOS/Ras control that is relevant for human cancer as well as all physiological processes involving Ras.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Guanosina Difosfato/metabolismo , Sistema de Señalización de MAP Quinasas , Neurofibromina 2/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Proteínas Son Of Sevenless/metabolismo , Animales , Proteínas del Citoesqueleto/genética , Guanosina Difosfato/genética , Humanos , Ratones , Células 3T3 NIH , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neurofibromina 2/genética , Proteína Oncogénica p21(ras)/genética , Proteínas Son Of Sevenless/genética
20.
FEBS Lett ; 587(16): 2698-704, 2013 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-23856463

RESUMEN

The cell surface glycoprotein CD44 enhances phorbol-12-myristate 13-acetate (TPA)-induced expression of p21WAF1 by stabilizing its mRNA and enhancing the protein's half-life in several cell lines. Only the plasma membrane-anchored cytoplasmic tail of CD44 and its interacting ezrin, radixin, moesin (ERM) proteins are required for this effect. A mitogen activated kinase (MEK) inhibitor abolishes the action of CD44 on p21. Down-regulation of p21 dramatically decreased anchorage-independence of a cancer cell line, whereas CD44 expression in this background could partially rescue the phenotype.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/química , Regulación Enzimológica de la Expresión Génica , Receptores de Hialuranos/química , Ésteres del Forbol/química , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas , Unión Proteica , ARN/química , Transducción de Señal , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...