Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Philos Trans A Math Phys Eng Sci ; 382(2277): 20230299, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39005014

RESUMEN

Hysteresis in the pressure-saturation relation in unsaturated porous media, owing to surface tension on the liquid-gas interface, exhibits strong degeneracy in the resulting mass balance equation. As an extension of previous existence and uniqueness results, we prove that under physically admissible initial conditions and without mass exchange with the exterior, the unique global solution of the fluid diffusion problem exists and asymptotically converges as time tends to infinity to a possibly non-homogeneous mass distribution and an a priori unknown constant pressure.This article is part of the theme issue 'Non-smooth variational problems with applications in mechanics'.

2.
Dev Dyn ; 251(4): 577-608, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34582081

RESUMEN

Primary cilia are dynamic compartments that regulate multiple aspects of cellular signaling. The production, maintenance, and function of cilia involve more than 1000 genes in mammals, and their mutations disrupt the ciliary signaling which manifests in a plethora of pathological conditions-the ciliopathies. Skeletal ciliopathies are genetic disorders affecting the development and homeostasis of the skeleton, and encompass a broad spectrum of pathologies ranging from isolated polydactyly to lethal syndromic dysplasias. The recent advances in forward genetics allowed for the identification of novel regulators of skeletogenesis, and revealed a growing list of ciliary proteins that are critical for signaling pathways implicated in bone physiology. Among these, a group of protein kinases involved in cilia assembly, maintenance, signaling, and disassembly has emerged. In this review, we summarize the functions of cilia kinases in skeletal development and disease, and discuss the available and upcoming treatment options.


Asunto(s)
Ciliopatías , Polidactilia , Animales , Cilios/metabolismo , Ciliopatías/genética , Ciliopatías/patología , Homeostasis , Mamíferos , Polidactilia/genética , Proteínas/genética
3.
Proc Natl Acad Sci U S A ; 116(10): 4316-4325, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30782830

RESUMEN

Vertebrate primary cilium is a Hedgehog signaling center but the extent of its involvement in other signaling systems is less well understood. This report delineates a mechanism by which fibroblast growth factor (FGF) controls primary cilia. Employing proteomic approaches to characterize proteins associated with the FGF-receptor, FGFR3, we identified the serine/threonine kinase intestinal cell kinase (ICK) as an FGFR interactor. ICK is involved in ciliogenesis and participates in control of ciliary length. FGF signaling partially abolished ICK's kinase activity, through FGFR-mediated ICK phosphorylation at conserved residue Tyr15, which interfered with optimal ATP binding. Activation of the FGF signaling pathway affected both primary cilia length and function in a manner consistent with cilia effects caused by inhibition of ICK activity. Moreover, knockdown and knockout of ICK rescued the FGF-mediated effect on cilia. We provide conclusive evidence that FGF signaling controls cilia via interaction with ICK.


Asunto(s)
Cilios/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Sistemas CRISPR-Cas , Factores de Crecimiento de Fibroblastos/metabolismo , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Ratones Noqueados , Modelos Animales , Simulación del Acoplamiento Molecular , Células 3T3 NIH , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Proteómica , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Transducción de Señal
4.
Nonlinear Differ Equ Appl ; 29(6): 72, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36187358

RESUMEN

The full quasistatic thermomechanical system of PDEs, describing water diffusion with the possibility of freezing and melting in a visco-elasto-plastic porous solid, is studied in detail under the hypothesis that the pressure-saturation hysteresis relation is given in terms of the Preisach hysteresis operator. The resulting system of balance equations for mass, momentum, and energy coupled with the phase dynamics equation is shown to admit a global solution under general assumptions on the data.

5.
Cell Mol Life Sci ; 77(19): 3885-3903, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31820037

RESUMEN

Many patients with chronic myeloid leukemia in deep remission experience return of clinical disease after withdrawal of tyrosine kinase inhibitors (TKIs). This suggests signaling of inactive BCR-ABL, which allows the survival of cancer cells, and relapse. We show that TKI treatment inhibits catalytic activity of BCR-ABL, but does not dissolve BCR-ABL core signaling complex, consisting of CRKL, SHC1, GRB2, SOS1, cCBL, p85a-PI3K, STS1 and SHIP2. Peptide microarray and co-immunoprecipitation results demonstrate that CRKL binds to proline-rich regions located in C-terminal, intrinsically disordered region of BCR-ABL, that SHC1 requires pleckstrin homology, src homology and tyrosine kinase domains of BCR-ABL for binding, and that BCR-ABL sequence motif located in disordered region around phosphorylated tyrosine 177 mediates binding of three core complex members, i.e., GRB2, SOS1, and cCBL. Further, SHIP2 binds to the src homology and tyrosine kinase domains of BCR-ABL and its inositol phosphatase activity contributes to BCR-ABL-mediated phosphorylation of SHC1. Together, this study characterizes protein-protein interactions within the BCR-ABL core complex and determines the contribution of particular BCR-ABL domains to downstream signaling. Understanding the structure and dynamics of BCR-ABL interactome is critical for the development of drugs targeting integrity of the BCR-ABL core complex.


Asunto(s)
Proteínas de Fusión bcr-abl/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencias de Aminoácidos , Sitios de Unión , Línea Celular Tumoral , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/genética , Células HEK293 , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Fosforilación , Análisis por Matrices de Proteínas , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Dominios Homologos src
6.
Hum Mol Genet ; 27(6): 1093-1105, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29360984

RESUMEN

Cilia project from almost every cell integrating extracellular cues with signaling pathways. Constitutive activation of FGFR3 signaling produces the skeletal disorders achondroplasia (ACH) and thanatophoric dysplasia (TD), but many of the molecular mechanisms underlying these phenotypes remain unresolved. Here, we report in vivo evidence for significantly shortened primary cilia in ACH and TD cartilage growth plates. Using in vivo and in vitro methodologies, our data demonstrate that transient versus sustained activation of FGF signaling correlated with different cilia consequences. Transient FGF pathway activation elongated cilia, while sustained activity shortened cilia. FGF signaling extended primary cilia via ERK MAP kinase and mTORC2 signaling, but not through mTORC1. Employing a GFP-tagged IFT20 construct to measure intraflagellar (IFT) speed in cilia, we showed that FGF signaling affected IFT velocities, as well as modulating cilia-based Hedgehog signaling. Our data integrate primary cilia into canonical FGF signal transduction and uncover a FGF-cilia pathway that needs consideration when elucidating the mechanisms of physiological and pathological FGFR function, or in the development of FGFR therapeutics.


Asunto(s)
Acondroplasia/fisiopatología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Displasia Tanatofórica/fisiopatología , Acondroplasia/genética , Animales , Cartílago/metabolismo , Condrocitos/metabolismo , Cilios/patología , Cilios/fisiología , Ciliopatías/genética , Ciliopatías/fisiopatología , Factores de Crecimiento de Fibroblastos/metabolismo , Placa de Crecimiento/metabolismo , Humanos , Ratones , Células 3T3 NIH , Fenotipo , Cultivo Primario de Células , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/fisiología , Displasia Tanatofórica/genética
7.
J Cell Sci ; 131(16)2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-30012834

RESUMEN

Metastasis of breast cancer cells to distant organs is responsible for ∼50% of breast cancer-related deaths in women worldwide. SHIP2 (also known as INPPL1) is a phosphoinositide 5-phosphatase for phosphatidylinositol (3,4,5)-trisphosphate [PI(3,4,5)P3] and phosphatidylinositol (4,5)-bisphosphate [PI(4,5)P2]. Here we show, through depletion of SHIP2 in triple negative MDA-MB-231 cells and the use of SHIP2 inhibitors, that cell migration appears to be positively controlled by SHIP2. The effect of SHIP2 on migration, as observed in MDA-MB-231 cells, appears to be mediated by PI(3,4)P2. Adhesion on fibronectin is always increased in SHIP2-depleted cells. Apoptosis measured in MDA-MB-231 cells is also increased in SHIP2-depleted cells as compared to control cells. In xenograft mice, SHIP2-depleted MDA-MB-231 cells form significantly smaller tumors than those formed by control cells and less metastasis is detected in lung sections. Our data reveal a general role for SHIP2 in the control of cell migration in breast cancer cells and a second messenger role for PI(3,4)P2 in the migration mechanism. In MDA-MB-231 cells, SHIP2 has a function in apoptosis in cells incubated in vitro and in mouse tumor-derived cells, which could account for its role on tumor growth determined in vivo.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/antagonistas & inhibidores , Animales , Movimiento Celular/genética , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Hum Mol Genet ; 25(1): 9-23, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26494904

RESUMEN

Activating mutations in the fibroblast growth factor receptor 3 (FGFR3) cause the most common genetic form of human dwarfism, achondroplasia (ACH). Small chemical inhibitors of FGFR tyrosine kinase activity are considered to be viable option for treating ACH, but little experimental evidence supports this claim. We evaluated five FGFR tyrosine kinase inhibitors (TKIs) (SU5402, PD173074, AZD1480, AZD4547 and BGJ398) for their activity against FGFR signaling in chondrocytes. All five TKIs strongly inhibited FGFR activation in cultured chondrocytes and limb rudiment cultures, completely relieving FGFR-mediated inhibition of chondrocyte proliferation and maturation. In contrast, TKI treatment of newborn mice did not improve skeletal growth and had lethal toxic effects on the liver, lungs and kidneys. In cell-free kinase assays as well as in vitro and in vivo cell assays, none of the tested TKIs demonstrated selectivity for FGFR3 over three other FGFR tyrosine kinases. In addition, the TKIs exhibited significant off-target activity when screened against a panel of 14 unrelated tyrosine kinases. This was most extensive in SU5402 and AZD1480, which inhibited DDR2, IGF1R, FLT3, TRKA, FLT4, ABL and JAK3 with efficiencies similar to or greater than those for FGFR. Low target specificity and toxicity of FGFR TKIs thus compromise their use for treatment of ACH. Conceptually, different avenues of therapeutic FGFR3 targeting should be investigated.


Asunto(s)
Acondroplasia/tratamiento farmacológico , Pirroles/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Animales , Benzamidas/farmacología , Cartílago/efectos de los fármacos , Cartílago/metabolismo , Catálisis/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Condrocitos/metabolismo , Humanos , Ratones , Compuestos de Fenilurea/farmacología , Piperazinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Síndrome
9.
Hum Mol Genet ; 25(18): 3998-4011, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27466187

RESUMEN

The short rib polydactyly syndromes (SRPS) are a group of recessively inherited, perinatal-lethal skeletal disorders primarily characterized by short ribs, shortened long bones, varying types of polydactyly and concomitant visceral abnormalities. Mutations in several genes affecting cilia function cause SRPS, revealing a role for cilia function in skeletal development. To identify additional SRPS genes and discover novel ciliary molecules required for normal skeletogenesis, we performed exome sequencing in a cohort of patients and identified homozygosity for a missense mutation, p.E80K, in Intestinal Cell Kinase, ICK, in one SRPS family. The p.E80K mutation abolished serine/threonine kinase activity, resulting in altered ICK subcellular and ciliary localization, increased cilia length, aberrant cartilage growth plate structure, defective Hedgehog and altered ERK signalling. These data identify ICK as an SRPS-associated gene and reveal that abnormalities in signalling pathways contribute to defective skeletogenesis.


Asunto(s)
Anomalías Múltiples/genética , Proteínas Hedgehog/genética , Proteínas Serina-Treonina Quinasas/genética , Síndrome de Costilla Pequeña y Polidactilia/genética , Esqueleto/crecimiento & desarrollo , Anomalías Múltiples/fisiopatología , Cilios/genética , Cilios/patología , Exoma/genética , Femenino , Humanos , Lactante , Sistema de Señalización de MAP Quinasas , Linaje , Embarazo , Análisis de Secuencia de ADN , Síndrome de Costilla Pequeña y Polidactilia/patología , Transducción de Señal , Esqueleto/anomalías
10.
Stem Cells ; 35(9): 2050-2059, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28631381

RESUMEN

Human pluripotent stem cells (hPSC) require signaling provided by fibroblast growth factor (FGF) receptors. This can be initiated by the recombinant FGF2 ligand supplied exogenously, but hPSC further support their niche by secretion of endogenous FGF2. In this study, we describe a role of tyrosine kinase expressed in hepatocellular carcinoma (TEC) kinase in this process. We show that TEC-mediated FGF2 secretion is essential for hPSC self-renewal, and its lack mediates specific differentiation. Following both short hairpin RNA- and small interfering RNA-mediated TEC knockdown, hPSC secretes less FGF2. This impairs hPSC proliferation that can be rescued by increasing amounts of recombinant FGF2. TEC downregulation further leads to a lower expression of the pluripotency markers, an improved priming towards neuroectodermal lineage, and a failure to develop cardiac mesoderm. Our data thus demonstrate that TEC is yet another regulator of FGF2-mediated hPSC pluripotency and differentiation. Stem Cells 2017;35:2050-2059.


Asunto(s)
Linaje de la Célula , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/enzimología , Proteínas Tirosina Quinasas/metabolismo , Biomarcadores/metabolismo , Línea Celular , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos , Proteínas Recombinantes/farmacología
11.
Biotechnol Bioeng ; 115(4): 850-862, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29278409

RESUMEN

Fibroblast growth factors (FGFs) serve numerous regulatory functions in complex organisms, and their corresponding therapeutic potential is of growing interest to academics and industrial researchers alike. However, applications of these proteins are limited due to their low stability. Here we tackle this problem using a generalizable computer-assisted protein engineering strategy to create a unique modified FGF2 with nine mutations displaying unprecedented stability and uncompromised biological function. The data from the characterization of stabilized FGF2 showed a remarkable prediction potential of in silico methods and provided insight into the unfolding mechanism of the protein. The molecule holds a considerable promise for stem cell research and medical or pharmaceutical applications.


Asunto(s)
Diseño Asistido por Computadora , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Ingeniería de Proteínas , Estabilidad Proteica , Secuencia de Aminoácidos , Animales , Simulación por Computador , Evolución Molecular Dirigida , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Factor 2 de Crecimiento de Fibroblastos/química , Humanos , Mutación Puntual , Pliegue de Proteína
12.
Hum Mol Genet ; 24(7): 1918-28, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25510505

RESUMEN

Osteogenesis imperfecta (OI) is a genetic disorder that results in low bone mineral density and brittle bones. Most cases result from dominant mutations in the type I procollagen genes, but mutations in a growing number of genes have been identified that produce autosomal recessive forms of the disease. Among these include mutations in the genes SERPINH1 and FKBP10, which encode the type I procollagen chaperones HSP47 and FKBP65, respectively, and predominantly produce a moderately severe form of OI. Little is known about the biochemical consequences of the mutations and how they produce OI. We have identified a new OI mutation in SERPINH1 that results in destabilization and mislocalization of HSP47 and secondarily has similar effects on FKBP65. We found evidence that HSP47 and FKBP65 act cooperatively during posttranslational maturation of type I procollagen and that FKBP65 and HSP47 but fail to properly interact in mutant HSP47 cells. These results thus reveal a common cellular pathway in cases of OI caused by HSP47 and FKBP65 deficiency.


Asunto(s)
Colágeno Tipo I/biosíntesis , Proteínas del Choque Térmico HSP47/metabolismo , Osteogénesis Imperfecta/metabolismo , Procolágeno/biosíntesis , Proteínas de Unión a Tacrolimus/metabolismo , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Preescolar , Femenino , Proteínas del Choque Térmico HSP47/química , Proteínas del Choque Térmico HSP47/genética , Humanos , Masculino , Datos de Secuencia Molecular , Osteogénesis Imperfecta/genética , Linaje , Transporte de Proteínas , Alineación de Secuencia , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/genética , Adulto Joven
14.
J Proteome Res ; 15(10): 3841-3855, 2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27615514

RESUMEN

The fibroblast growth factor receptors (FGFRs) are important oncogenes promoting tumor progression in many types of cancer, such as breast, bladder, and lung cancer as well as multiple myeloma and rhabdomyosarcoma. However, little is known about how these receptors are internalized and down-regulated in cells. We have here applied proximity biotin labeling to identify proteins involved in FGFR4 signaling and trafficking. For this purpose we fused a mutated biotin ligase, BirA*, to the C-terminal tail of FGFR4 (FGFR4-BirA*) and the fusion protein was stably expressed in U2OS cells. Upon addition of biotin to these cells, proteins in proximity to the FGFR4-BirA* fusion protein became biotinylated and could be isolated and identified by quantitative mass spectrometry. We identified in total 291 proteins, including 80 proteins that were enriched in samples where the receptor was activated by the ligand (FGF1), among them several proteins previously found to be involved in FGFR signaling (e.g., FRS2, PLCγ, RSK2 and NCK2). Interestingly, many of the identified proteins were implicated in endosomal transport, and by precise annotation we were able to trace the intracellular pathways of activated FGFR4. Validating the data by confocal and three-dimensional structured illumination microscopy analysis, we concluded that FGFR4 uses clathrin-mediated endocytosis for internalization and is further sorted from early endosomes to the recycling compartment and the trans-Golgi network. Depletion of cells for clathrin heavy chain led to accumulation of FGFR4 at the cell surface and increased levels of active FGFR4 and PLCγ, while AKT and ERK signaling was diminished, demonstrating that functional clathrin-mediated endocytosis is required for proper FGFR4 signaling. Thus, this study reveals proteins and pathways involved in FGFR4 transport and signaling that provide possible targets and opportunities for therapeutic intervention in FGFR4 aberrant cancer.


Asunto(s)
Endosomas/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Biotinilación , Línea Celular Tumoral , Clatrina/metabolismo , Endocitosis , Humanos , Microscopía/métodos , Transporte de Proteínas , Transducción de Señal , Coloración y Etiquetado , Red trans-Golgi/metabolismo
15.
Biochim Biophys Acta ; 1852(5): 839-50, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25558817

RESUMEN

Aberrant fibroblast growth factor (FGF) signaling disturbs chondrocyte differentiation in skeletal dysplasia, but the mechanisms underlying this process remain unclear. Recently, FGF was found to activate canonical WNT/ß-catenin pathway in chondrocytes via Erk MAP kinase-mediated phosphorylation of WNT co-receptor Lrp6. Here, we explore the cellular consequences of such a signaling interaction. WNT enhanced the FGF-mediated suppression of chondrocyte differentiation in mouse limb bud micromass and limb organ cultures, leading to inhibition of cartilage nodule formation in micromass cultures, and suppression of growth in cultured limbs. Simultaneous activation of the FGF and WNT/ß-catenin pathways resulted in loss of chondrocyte extracellular matrix, expression of genes typical for mineralized tissues and alteration of cellular shape. WNT enhanced the FGF-mediated downregulation of chondrocyte proteoglycan and collagen extracellular matrix via inhibition of matrix synthesis and induction of proteinases involved in matrix degradation. Expression of genes regulating RhoA GTPase pathway was induced by FGF in cooperation with WNT, and inhibition of the RhoA signaling rescued the FGF/WNT-mediated changes in chondrocyte cellular shape. Our results suggest that aberrant FGF signaling cooperates with WNT/ß-catenin in suppression of chondrocyte differentiation.


Asunto(s)
Cartílago/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Western Blotting , Cartílago/citología , Cartílago/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Células Cultivadas , Condrocitos/metabolismo , Sinergismo Farmacológico , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células HEK293 , Humanos , Esbozos de los Miembros/efectos de los fármacos , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Microscopía Confocal , Modelos Biológicos , Ratas , Receptores de Factores de Crecimiento de Fibroblastos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Proteínas Wnt/genética , Proteínas Wnt/farmacología , Proteína Wnt3A/farmacología , beta Catenina/genética
16.
Cell Mol Life Sci ; 72(12): 2445-59, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25854632

RESUMEN

Fibroblast growth factors (FGFs) deliver extracellular signals that govern many developmental and regenerative processes, but the mechanisms regulating FGF signaling remain incompletely understood. Here, we explored the relationship between intrinsic stability of FGF proteins and their biological activity for all 18 members of the FGF family. We report that FGF1, FGF3, FGF4, FGF6, FGF8, FGF9, FGF10, FGF16, FGF17, FGF18, FGF20, and FGF22 exist as unstable proteins, which are rapidly degraded in cell cultivation media. Biological activity of FGF1, FGF3, FGF4, FGF6, FGF8, FGF10, FGF16, FGF17, and FGF20 is limited by their instability, manifesting as failure to activate FGF receptor signal transduction over long periods of time, and influence specific cell behavior in vitro and in vivo. Stabilization via exogenous heparin binding, introduction of stabilizing mutations or lowering the cell cultivation temperature rescues signaling of unstable FGFs. Thus, the intrinsic ligand instability is an important elementary level of regulation in the FGF signaling system.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Proliferación Celular , Condrosarcoma/metabolismo , Factores de Crecimiento de Fibroblastos/química , Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Condrosarcoma/genética , Condrosarcoma/patología , Dicroismo Circular , Femenino , Factores de Crecimiento de Fibroblastos/clasificación , Factores de Crecimiento de Fibroblastos/genética , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Estabilidad Proteica , Ratas , Temperatura , Células Tumorales Cultivadas
17.
Am J Hum Genet ; 90(4): 746-51, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22464252

RESUMEN

Acrodysostosis is a dominantly-inherited, multisystem disorder characterized by skeletal, endocrine, and neurological abnormalities. To identify the molecular basis of acrodysostosis, we performed exome sequencing on five genetically independent cases. Three different missense mutations in PDE4D, which encodes cyclic AMP (cAMP)-specific phosphodiesterase 4D, were found to be heterozygous in three of the cases. Two of the mutations were demonstrated to have occurred de novo, providing strong genetic evidence of causation. Two additional cases were heterozygous for de novo missense mutations in PRKAR1A, which encodes the cAMP-dependent regulatory subunit of protein kinase A and which has been recently reported to be the cause of a form of acrodysostosis resistant to multiple hormones. These findings demonstrate that acrodysostosis is genetically heterogeneous and underscore the exquisite sensitivity of many tissues to alterations in cAMP homeostasis.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Disostosis/genética , Exoma/genética , Discapacidad Intelectual/genética , Mutación , Osteocondrodisplasias/genética , Análisis de Secuencia de ADN , Secuencia de Bases , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Disostosis/diagnóstico por imagen , Femenino , Heterocigoto , Humanos , Discapacidad Intelectual/diagnóstico por imagen , Masculino , Datos de Secuencia Molecular , Osteocondrodisplasias/diagnóstico por imagen , Radiografía
18.
Am J Hum Genet ; 90(3): 550-7, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22387015

RESUMEN

Fibroblast growth factor receptor 2 (FGFR2) is a crucial regulator of bone formation during embryonic development. Both gain and loss-of-function studies in mice have shown that FGFR2 maintains a critical balance between the proliferation and differentiation of osteoprogenitor cells. We have identified de novo FGFR2 mutations in a sporadically occurring perinatal lethal skeletal dysplasia characterized by poor mineralization of the calvarium, craniosynostosis, dysmorphic facial features, prenatal teeth, hypoplastic pubis and clavicles, osteopenia, and bent long bones. Histological analysis of the long bones revealed that the growth plate contained smaller hypertrophic chondrocytes and a thickened hypercellular periosteum. Four unrelated affected individuals were found to be heterozygous for missense mutations that introduce a polar amino acid into the hydrophobic transmembrane domain of FGFR2. Using diseased chondrocytes and a cell-based assay, we determined that these mutations selectively reduced plasma-membrane levels of FGFR2 and markedly diminished the receptor's responsiveness to extracellular FGF. All together, these clinical and molecular findings are separate from previously characterized FGFR2 disorders and represent a distinct skeletal dysplasia.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Anomalías Craneofaciales/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Secuencia de Aminoácidos , Enfermedades del Desarrollo Óseo/metabolismo , Huesos/anomalías , Huesos/embriología , Huesos/metabolismo , Condrocitos/metabolismo , Anomalías Craneofaciales/metabolismo , Feto/anomalías , Feto/metabolismo , Factores de Crecimiento de Fibroblastos/deficiencia , Heterocigoto , Humanos , Datos de Secuencia Molecular , Mutación , Mutación Missense , Osteoblastos/metabolismo , Osteogénesis/genética , Transducción de Señal , Esqueleto
19.
J Pharmacol Exp Ther ; 353(1): 132-49, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25650377

RESUMEN

Achondroplasia (ACH), the most common form of human dwarfism, is caused by an activating autosomal dominant mutation in the fibroblast growth factor receptor-3 gene. Genetic overexpression of C-type natriuretic peptide (CNP), a positive regulator of endochondral bone growth, prevents dwarfism in mouse models of ACH. However, administration of exogenous CNP is compromised by its rapid clearance in vivo through receptor-mediated and proteolytic pathways. Using in vitro approaches, we developed modified variants of human CNP, resistant to proteolytic degradation by neutral endopeptidase, that retain the ability to stimulate signaling downstream of the CNP receptor, natriuretic peptide receptor B. The variants tested in vivo demonstrated significantly longer serum half-lives than native CNP. Subcutaneous administration of one of these CNP variants (BMN 111) resulted in correction of the dwarfism phenotype in a mouse model of ACH and overgrowth of the axial and appendicular skeletons in wild-type mice without observable changes in trabecular and cortical bone architecture. Moreover, significant growth plate widening that translated into accelerated bone growth, at hemodynamically tolerable doses, was observed in juvenile cynomolgus monkeys that had received daily subcutaneous administrations of BMN 111. BMN 111 was well tolerated and represents a promising new approach for treatment of patients with ACH.


Asunto(s)
Acondroplasia/tratamiento farmacológico , Péptido Natriurético Tipo-C/análogos & derivados , Neprilisina/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Acondroplasia/genética , Acondroplasia/fisiopatología , Animales , Presión Sanguínea/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/patología , Huesos/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Inyecciones Subcutáneas , Macaca fascicularis , Masculino , Ratones , Células 3T3 NIH , Péptido Natriurético Tipo-C/metabolismo , Péptido Natriurético Tipo-C/farmacología , Péptido Natriurético Tipo-C/uso terapéutico , Ratas , Proteínas Recombinantes/metabolismo
20.
Mutat Res Rev Mutat Res ; 759: 40-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24295726

RESUMEN

Somatic mutations in receptor tyrosine kinase FGFR3 cause excessive cell proliferation, leading to cancer or skin overgrowth. Remarkably, the same mutations inhibit chondrocyte proliferation and differentiation in developing bones, resulting in skeletal dysplasias, such as hypochondroplasia, achondroplasia, SADDAN and thanatophoric dysplasia. A similar phenotype is observed in Noonan syndrome, Leopard syndrome, hereditary gingival fibromatosis, neurofibromatosis type 1, Costello syndrome, Legius syndrome and cardiofaciocutaneous syndrome. Collectively termed RASopathies, the latter syndromes are caused by germline mutations in components of the RAS/ERK MAP kinase signaling pathway. This article considers the evidence suggesting that FGFR3 activation in chondrocytes mimics the activation of major oncogenes signaling via the ERK pathway. Subsequent inhibition of chondrocyte proliferation in FGFR3-related skeletal dysplasias and RASopathies is proposed to result from activation of defense mechanisms that originally evolved to safeguard mammalian organisms against cancer.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Neoplasias Óseas/genética , Diferenciación Celular/genética , Genes ras/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/patología , Neoplasias Óseas/patología , Proliferación Celular , Condrocitos/citología , Condrocitos/metabolismo , Mutación de Línea Germinal , Humanos , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA