Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Ann Oncol ; 31(6): 780-788, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32240793

RESUMEN

BACKGROUND: Bromodomain and extra-terminal (BET) proteins are epigenetic readers that regulate expression of genes involved in oncogenesis. CC-90010 is a novel, oral, reversible, small-molecule BET inhibitor. PATIENTS AND METHODS: CC-90010-ST-001 (NCT03220347; 2015-004371-79) is a phase I dose-escalation and expansion study of CC-90010 in patients with advanced or unresectable solid tumors and relapsed/refractory (R/R) non-Hodgkin's lymphoma (NHL). We report results from the dose escalation phase, which explored 11 dose levels and four dosing schedules, two weekly (2 days on/5 days off; 3 days on/4 days off), one biweekly (3 days on/11 days off), and one monthly (4 days on/24 days off). The primary objectives were to determine the safety, maximum tolerated dose (MTD) and/or recommended phase II dose (RP2D) and schedule. Secondary objectives were to evaluate signals of early antitumor activity, pharmacokinetics, and pharmacodynamics. RESULTS: This study enrolled 69 patients, 67 with solid tumors and two with diffuse large B-cell lymphoma (DLBCL). The median age was 57 years (range, 21-80) and the median number of prior regimens was four (range, 1-9). Treatment-related adverse events (TRAEs) were mostly mild and manageable; grade 3/4 TRAEs reported in more than two patients were thrombocytopenia (13%), anemia, and fatigue (4% each). Six patients had dose-limiting toxicities. MTDs were 15 mg (2 days on/5 days off), 30 mg (3 days on/11 days off), and 45 mg (4 days on/24 days off). The RP2D and schedule selected for expansion was 45 mg (4 days on/24 days off). As of 8 October 2019, one patient with grade 2 astrocytoma achieved a complete response, one patient with endometrial carcinoma had a partial response, and six patients had prolonged stable disease ≥11 months. CONCLUSIONS: CC-90010 is well tolerated, with single-agent activity in patients with heavily pretreated, advanced solid tumors.


Asunto(s)
Antineoplásicos , Linfoma de Células B Grandes Difuso , Linfoma no Hodgkin , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma no Hodgkin/tratamiento farmacológico , Dosis Máxima Tolerada , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Adulto Joven
2.
J Cell Biol ; 139(2): 541-52, 1997 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-9334355

RESUMEN

Members of the TGF-beta superfamily are important regulators of skeletal development. TGF-betas signal through heteromeric type I and type II receptor serine/threonine kinases. When over-expressed, a cytoplasmically truncated type II receptor can compete with the endogenous receptors for complex formation, thereby acting as a dominant-negative mutant (DNIIR). To determine the role of TGF-betas in the development and maintenance of the skeleton, we have generated transgenic mice (MT-DNIIR-4 and -27) that express the DNIIR in skeletal tissue. DNIIR mRNA expression was localized to the periosteum/perichondrium, syno-vium, and articular cartilage. Lower levels of DNIIR mRNA were detected in growth plate cartilage. Transgenic mice frequently showed bifurcation of the xiphoid process and sternum. They also developed progressive skeletal degeneration, resulting by 4 to 8 mo of age in kyphoscoliosis and stiff and torqued joints. The histology of affected joints strongly resembled human osteo-arthritis. The articular surface was replaced by bone or hypertrophic cartilage as judged by the expression of type X collagen, a marker of hypertrophic cartilage normally absent from articular cartilage. The synovium was hyperplastic, and cartilaginous metaplasia was observed in the joint space. We then tested the hypothesis that TGF-beta is required for normal differentiation of cartilage in vivo. By 4 and 8 wk of age, the level of type X collagen was increased in growth plate cartilage of transgenic mice relative to wild-type controls. Less proteoglycan staining was detected in the growth plate and articular cartilage matrix of transgenic mice. Mice that express DNIIR in skeletal tissue also demonstrated increased Indian hedgehog (IHH) expression. IHH is a secreted protein that is expressed in chondrocytes that are committed to becoming hypertrophic. It is thought to be involved in a feedback loop that signals through the periosteum/ perichondrium to inhibit cartilage differentiation. The data suggest that TGF-beta may be critical for multifaceted maintenance of synovial joints. Loss of responsiveness to TGF-beta promotes chondrocyte terminal differentiation and results in development of degenerative joint disease resembling osteoarthritis in humans.


Asunto(s)
Huesos/metabolismo , Cartílago Articular/citología , Cartílago Articular/metabolismo , Articulaciones/metabolismo , Osteoartritis/genética , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas Receptoras/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Membrana Sinovial/metabolismo , Transactivadores , Animales , Diferenciación Celular , Expresión Génica/efectos de los fármacos , Placa de Crecimiento/metabolismo , Proteínas Hedgehog , Humanos , Hipertrofia , Articulaciones/patología , Ratones , Ratones Transgénicos , Osteoartritis/patología , Osteoartritis/fisiopatología , Reacción en Cadena de la Polimerasa , Biosíntesis de Proteínas , ARN Mensajero/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Transcripción Genética , Factor de Crecimiento Transformador beta/farmacología
3.
Mol Biol Cell ; 9(7): 1903-18, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9658179

RESUMEN

Bone remodeling depends on the spatial and temporal coupling of bone formation by osteoblasts and bone resorption by osteoclasts; however, the molecular basis of these inductive interactions is unknown. We have previously shown that osteoblastic overexpression of TGF-beta2 in transgenic mice deregulates bone remodeling and leads to an age-dependent loss of bone mass that resembles high-turnover osteoporosis in humans. This phenotype implicates TGF-beta2 as a physiological regulator of bone remodeling and raises the question of how this single secreted factor regulates the functions of osteoblasts and osteoclasts and coordinates their opposing activities in vivo. To gain insight into the physiological role of TGF-beta in bone remodeling, we have now characterized the responses of osteoblasts to TGF-beta in these transgenic mice. We took advantage of the ability of alendronate to specifically inhibit bone resorption, the lack of osteoclast activity in c-fos-/- mice, and a new transgenic mouse line that expresses a dominant-negative form of the type II TGF-beta receptor in osteoblasts. Our results show that TGF-beta directly increases the steady-state rate of osteoblastic differentiation from osteoprogenitor cell to terminally differentiated osteocyte and thereby increases the final density of osteocytes embedded within bone matrix. Mice overexpressing TGF-beta2 also have increased rates of bone matrix formation; however, this activity does not result from a direct effect of TGF-beta on osteoblasts, but is more likely a homeostatic response to the increase in bone resorption caused by TGF-beta. Lastly, we find that osteoclastic activity contributes to the TGF-beta-induced increase in osteoblast differentiation at sites of bone resorption. These results suggest that TGF-beta is a physiological regulator of osteoblast differentiation and acts as a central component of the coupling of bone formation to resorption during bone remodeling.


Asunto(s)
Remodelación Ósea/fisiología , Osteoblastos/fisiología , Factor de Crecimiento Transformador beta/fisiología , Alendronato/farmacología , Animales , Remodelación Ósea/efectos de los fármacos , Resorción Ósea/genética , Recuento de Células/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Fémur , Regulación de la Expresión Génica/efectos de los fármacos , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Minerales/metabolismo , Osteoblastos/efectos de los fármacos , Osteoclastos/citología , Osteocitos/citología , Periostio/metabolismo , Fenotipo , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Factor de Crecimiento Transformador beta/sangre , Factor de Crecimiento Transformador beta/genética
4.
J Endocrinol ; 167(1): 175-82, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11018765

RESUMEN

Skeletal myoblasts are inherently programmed to leave the cell cycle and begin the differentiation process following removal of exogenous growth factors. Serum withdrawal results in a marked induction of IGF production which is essential for skeletal muscle differentiation in vitro. However, the potential role of the tyrosine kinase IGF-I receptor (thought to be the principal mediator of both IGF-I and II signaling in skeletal muscle) in the decision of myoblasts to begin differentiation following serum withdrawal is unknown. To explore the role of the IGF-I receptor in this decision by skeletal myoblasts, we functionally inactivated endogenous IGF-I receptors in mouse C2C12 cells using a dominant negative, kinase-inactive IGF-I receptor in which the ATP-binding site lysine (K) at residue 1003 has been mutated to alanine (A). Cell lines with the greatest degree of mutant IGF-I receptor expression (A/K cells) demonstrated functional inactivation of endogenous IGF-I receptors as determined by their impaired ability to phosphorylate the principal substrate of the IGF-I receptor, IRS-1, in response to treatment with IGF-I. In addition, the proliferative response of myoblasts to IGF-I was completely abolished in A/K cells. Following withdrawal of exogenous growth factors, A/K cells demonstrated a marked delay in the induction of the gene expression of myogenin, a skeletal muscle-specific transcription factor essential for differentiation, and a subsequent delay in the induction of muscle creatine kinase activity. Delayed differentiation in A/K cells was associated with prolonged phosphorylation of the cell cycle regulatory retinoblastoma (Rb) protein; it is the un- (or hypo-) phosphorylated form of Rb which is known to promote differentiation in skeletal myoblasts. Thus, the IGF-I receptor regulates the timing of myoblast differentiation induced by serum withdrawal. The delayed differentiation of skeletal myoblasts with functionally inactive IGF-I receptors may result, at least in part, from delayed induction of myogenin gene expression and prolonged phosphorylation of the Rb protein.


Asunto(s)
Músculo Esquelético/citología , Receptor IGF Tipo 1/fisiología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , División Celular/fisiología , Medio de Cultivo Libre de Suero , Ratones , Fosforilación , Proteína de Retinoblastoma/metabolismo , Transfección
5.
J Musculoskelet Neuronal Interact ; 3(4): 304-7; discussion 320-1, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15758305
7.
Dev Biol ; 178(2): 459-71, 1996 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-8812142

RESUMEN

Expression of a transfected MyoD gene induces myogenic differentiation of most cell types. In this study, we evaluated the ability of an exogenous MyoD gene to induce myogenic conversion in two pairs of matched cell lines with different degrees of differentiation within either the osteoblastic or chondrocytic lineage. We show that osteoblasts and chondrocytes are resistant to the myogenic effects of MyoD alone. However, in their less-differentiated cell line counterparts, MyoD induces expression of muscle-cell-specific markers. Less-differentiated osteoblasts can be made resistant to MyoD-induced myogenic conversion by induction of adipogenic differentiation using dexamethasone. Finally, a dominant positive form of MyoD, one which is tethered to a partner, E47, activates muscle-specific gene expression in osteoblasts. Our results suggest that the response of a cell to MyoD depends on its lineage and its degree of differentiation. Furthermore, commitment of cells to the osteoblastic or chondrocytic lineage may involve inhibition of alternative pathways, such as those leading to myoblastic differentiation. Finally, osteoblasts may express a protein(s) which interferes with the activity of MyoD by inhibiting its association with E proteins. This interference can be overcome by expression of the MyoD-E47 hybrid, suggesting that osteoblasts are otherwise competent to undergo myogenic conversion.


Asunto(s)
Cartílago/citología , Músculo Esquelético/citología , Proteína MioD/biosíntesis , Osteoblastos/citología , Animales , Cartílago/metabolismo , Diferenciación Celular , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Transferencia de Gen , Músculo Esquelético/metabolismo , Proteína MioD/genética , Osteoblastos/metabolismo , Ratas
8.
Development ; 120(5): 1085-95, 1994 May.
Artículo en Inglés | MEDLINE | ID: mdl-8026322

RESUMEN

Transforming growth factor-beta (TGF-beta) is thought to play a role in mesenchymal cell development and, specifically, in muscle differentiation, yet its precise role in the latter process remains unclear. TGF-beta has been shown to both inhibit and induce myoblast maturation in vitro, depending on the culture conditions. Whether the type I or type II TGF-beta receptor mediates the various TGF-beta effects on myogenesis is not known. In the present study, C2C12 myoblasts were transfected with an expression vector for a truncated type II TGF-beta receptor, which has been shown to act as a dominant negative inhibitor of type II receptor signaling. In contrast to the parental cells, the transfected clones did not efficiently form myotubes or induce expression of MyoD, myogenin and several other differentiation markers following incubation in low serum media. However, some muscle differentiation markers continued to be expressed in the transfected cells suggesting that at least two pathways are involved in muscle cell differentiation. These cells could still growth arrest in low serum media, showing that decreased proliferation can be dissociated from differentiation. Unlike several oncogenes known to block myogenic differentiation, expression of the truncated TGF-beta receptor did not result in myoblast transformation. Injection of the parental or the transfected C2C12 cells into the limb muscle of nude mice revealed quantitative and qualitative differences in their behavior, and suggested that myoblasts expressing the truncated TGF-beta receptor cannot fuse in vivo. Finally, retrovirus-mediated expression of MyoD in the transfected cells restored their ability to form myotubes in vitro, indicating that inhibition of myoblast differentiation by the truncated TGF-beta receptor may depend on decreased MyoD expression. We propose that TGF-beta signaling through the type II receptor is required for several distinct aspects of myogenic differentiation and that TGF-beta acts as a competence factor in this multistep process.


Asunto(s)
Expresión Génica/fisiología , Músculos/embriología , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta/fisiología , Animales , Northern Blotting , Western Blotting , Diferenciación Celular/genética , Línea Celular , Técnica del Anticuerpo Fluorescente , Ratones , Músculos/citología , Proteína MioD/genética , Miogenina/genética , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Transducción de Señal/fisiología
9.
J Biol Chem ; 270(41): 24237-45, 1995 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-7592630

RESUMEN

Transforming growth factor-beta (TGF-beta) inhibits the proliferation of epithelial cells by altering the expression or function of various components of the cell cycle machinery. Expression of one of these components, cyclin A, is inhibited by TGF-beta treatment. We have identified a 760-base pair fragment of the human cyclin A gene promoter that is sufficient to confer TGF-beta responsiveness. Using this promoter fragment, we have developed a cyclin A-based luciferase reporter assay that quantitates the growth inhibitory effect of TGF-beta in transient transfection assays. This assay was used to determine which domains of the type I (RI) and type II (RII) receptors were required for the antiproliferative effect of TGF-beta. In parallel, the functionality of chimeric receptors, between RI and RII (RI-RII or RII-RI), was tested for TGF-beta effect on gene expression using a reporter assay based on the plasminogen activator inhibitor type 1 (PAI-1) promoter. We found that TGF-beta-induced inhibition of cyclin A expression was absent in RI or RII-deficient Mv1Lu cells and that this response was restored by expression of wild-type type I or type II receptors in these cells. Furthermore, expression of a single chimeric receptor, either RI-RII or RII-RI, did not confer cyclin A regulation by TGF-beta. However, expression of two reciprocal chimeras (RI-RII and RII-RI) resulted in growth inhibition, similarly to wild-type receptors. In addition, chimeric receptors as well as mutant receptors with a deleted cytoplasmic domain and kinase-negative receptors inhibited TGF-beta responsiveness in the cyclin A reporter assay in a dominant negative fashion. Finally, in both receptor types, the juxtamembrane domain preceding the kinase domain was essential for receptor function but the cytoplasmic tail was dispensable. Our results suggest that a functional TGF-beta receptor complex is required for TGF-beta-dependent down-regulation of cyclin A gene expression and illustrate the identical receptor requirements for TGF-beta-induced growth inhibition and gene expression.


Asunto(s)
Ciclinas/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Regiones Promotoras Genéticas , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Factor de Crecimiento Transformador beta/farmacología , Animales , Secuencia de Bases , División Celular/efectos de los fármacos , Línea Celular , Chlorocebus aethiops , Ciclinas/genética , Citomegalovirus/genética , Cartilla de ADN , Vectores Genéticos , Humanos , Cinética , Luciferasas/biosíntesis , Visón , Datos de Secuencia Molecular , Mutagénesis , Reacción en Cadena de la Polimerasa , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transfección
10.
Princess Takamatsu Symp ; 24: 264-75, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-8983081

RESUMEN

Transforming growth factor-beta (TGF-beta) is a multifunctional protein that modulates cell proliferation and interaction with the extracellular matrix. Three common TGF-beta receptors are found on the cell surface. The type III receptor is a transmembrane proteoglycan with a short cytoplasmic domain and is thought not to be involved in TGF-beta induced signalling. In contrast, the type II and type I receptors are transmembrane serine/threonine kinases. The type II receptor determines the ligand specificity, whereas the type I receptor interacts with the type II receptor and may not have a ligand binding specificity by itself. Both type II and type I receptors are involved in TGF-beta induced signalling. The type II receptor, likely in conjunction with the type I receptor, is required for the antiproliferative effect of TGF-beta, whereas the type I receptor is the likely mediator of the effects of TGF-beta on the expression of several genes including some extracellular matrix proteins. To address the role of TGF-beta signalling in myoblast differentiation, we transfected a dominant negative mutant of the type II receptor in myoblasts, thus inhibiting type II receptor mediated signalling. These cells not longer had the ability to differentiate in vitro or in vivo, suggesting that TGF-beta signalling through the type II receptor provides competence for myoblastic differentiation. These studies also indicate that there are several signalling pathways involved in myoblastic differentiation, one of which is modulated by the TGF-beta signalling.


Asunto(s)
Receptores de Activinas Tipo I , Receptores de Factores de Crecimiento Transformadores beta/clasificación , Factor de Crecimiento Transformador beta/fisiología , Animales , Diferenciación Celular , Desarrollo Embrionario y Fetal , Humanos , Ratones , Morfogénesis , Familia de Multigenes , Músculos/citología , Músculos/embriología , Proteínas de Neoplasias/fisiología , Especificidad de Órganos , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteoglicanos/química , Proteoglicanos/genética , Proteoglicanos/fisiología , Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Relación Estructura-Actividad , Transfección , Factor de Crecimiento Transformador beta/farmacología
11.
Osteoarthritis Cartilage ; 10(9): 692-706, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12202122

RESUMEN

OBJECTIVE: To examine the potential of insulin, in a sustained delivery system, as a treatment for arthritis. DESIGN: The effect of insulin on matrix synthesis, matrix breakdown, and nitric oxide production in primary cartilage explants was examined. The activity of insulin on diseased cartilage from Dunkin Hartley guinea pigs, diabetic mice, and osteoarthritic patients was measured. The specificity of insulin stimulation was compared to that of IGF-I using osteoblasts and fibroblasts. Finally, the stability of insulin in a biologically relevant system was tested, and a slow-release formulation of insulin was developed and characterized. RESULTS: In articular cartilage explants, insulin stimulated proteoglycan (PG) synthesis, inhibited PG release and nitric oxide production, and overcame the detrimental effects of interleukin 1 (IL-1). The mechanism whereby insulin decreased matrix breakdown was through inhibition of aggrecanase activity. Insulin was active on cartilage at concentrations at which insulin does not cross-react with insulin-like growth factor I (IGF-I) receptors nor stimulate proliferation of other cells types. The response of cartilage to insulin did not diminish with age or disease. Insulin stimulated matrix synthesis in osteoarthritic cartilage and local treatment with insulin overcame endogenous suppression of matrix synthesis in diabetic cartilage. Poly-lactic-coglycolic acid (PLGA) was found to be an effective carrier for delivery of insulin, and PLGA-Insulin was active on articular cartilage in vitro and in vivo. CONCLUSIONS: As the incidence of arthritis increases with the aging population, an effective therapy to induce repair of cartilage is needed. Based on its biological activities, insulin appears to be an attractive protein therapeutic candidate. Maximum insulin effectiveness may require a sustained delivery system.


Asunto(s)
Insulina/administración & dosificación , Osteoartritis de la Rodilla/tratamiento farmacológico , Anciano , Animales , Disponibilidad Biológica , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Preparaciones de Acción Retardada , Diabetes Mellitus Experimental/metabolismo , Estabilidad de Medicamentos , Femenino , Cobayas , Humanos , Insulina/farmacocinética , Insulina/uso terapéutico , Masculino , Ratones , Persona de Mediana Edad , Osteoartritis de la Rodilla/metabolismo , Proteoglicanos/biosíntesis
12.
Cell Growth Differ ; 6(7): 827-36, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7547504

RESUMEN

The transforming growth factor-beta (TGF-beta) superfamily is a group of secreted growth factors that appears to play a central role in mesenchymal differentiation, including cartilage and bone formation. The present study examines the role of one member of this family, vgr-1, also called bone morphogenetic protein-6, in mesenchymal cell differentiation. This factor may be considered as a prototype for the largest subgroup of related factors within the TGF-beta superfamily, the function of which has as yet been poorly defined. vgr-1 has been localized previously to hypertrophic cartilage and has been shown to induce endochondral bone formation in vivo. To further characterize the role of vgr-1 in bone and cartilage differentiation, we stably transfected the pluripotent mesenchymal cell line ROB-C26 with a vector to overexpress vgr-1. Overexpression of this factor did not affect cell shape or morphology, but it enhanced osteoblastic differentiation in vitro and altered cellular responsiveness to retinoic acid. Furthermore, the extracellular matrix produced by these vgr-1-overexpressing cells induced ectopic bone formation in vivo and osteoblastic differentiation in vitro, similar to the matrix produced by C26 cells treated with retinoic acid. The osteoinductive effect of the matrix from vgr-1-overexpressing cells was blocked using a neutralizing vgr-1 antibody but not with a neutralizing TGF-beta 1 antibody, indicating that vgr-1 alone was required for this osteogenic effect. In contrast, the osteoinductive effect of matrix from retinoic acid-treated cells was blocked with both vgr-1 and TGF-beta 1 antibodies, suggesting that TGF-beta 1 may act prior to vgr-1 during osteoblastic differentiation. We further demonstrated that osteoinduction by vgr-1 was dependent on presentation of vgr-1 within the matrix, because the osteoinductive effect of matrix from vgr-1-overexpressing cells could not be mimicked with the addition of soluble vgr-1 to parental C26 cells. Finally, overexpression of MyoD within the C26 cells overexpressing vgr-1 converted the cells to myoblasts, indicating that vgr-1 had induced early osteoblastic.


Asunto(s)
Sustancias de Crecimiento/farmacología , Mesodermo/citología , Osteoblastos/citología , Proteínas/farmacología , Células Madre/citología , Fosfatasa Alcalina , Proteína Morfogenética Ósea 6 , Proteínas Morfogenéticas Óseas , Diferenciación Celular/efectos de los fármacos , Línea Celular/citología , Línea Celular/efectos de los fármacos , Matriz Extracelular , Expresión Génica/fisiología , Mesodermo/efectos de los fármacos , Proteína MioD/fisiología , Osteoblastos/efectos de los fármacos , Plásticos , Proteínas/genética , Células Madre/efectos de los fármacos , Transfección
13.
Cytokine ; 16(1): 10-21, 2001 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-11669582

RESUMEN

Overexpression of interleukin (IL-)17 has recently been shown to be associated with a number of pathological conditions. Because IL-17 is found at high levels in the synovial fluid surrounding cartilage in patients with inflammatory arthritis, the present study determined the direct effect of IL-17 on articular cartilage. As shown herein, IL-17 was a direct and potent inducer of matrix breakdown and an inhibitor of matrix synthesis in articular cartilage explants. These effects were mediated in part by leukemia inhibitory factor (LIF), but did not depend on interleukin-1 activity. The mechanism whereby IL-17 induced matrix breakdown in cartilage tissue appeared to be due to stimulation of activity of aggrecanase(s), not matrix metalloproteinase(s). However, IL-17 upregulated expression of matrix metalloproteinase(s) in chondrocytes cultured in monolayer. In vivo, IL-17 induced a phenotype similar to inflammatory arthritis when injected into the intra-articular space of mouse knee joints. Furthermore, a related protein, IL-17E, was found to have catabolic activity on human articular cartilage. This study characterizes the mechanism whereby IL-17 acts directly on cartilage matrix turnover. Such findings have important implications for the treatment of degenerative joint diseases such as arthritis.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Interleucina-17/farmacología , Animales , Western Blotting , Cartílago Articular/citología , Cartílago Articular/metabolismo , Bovinos , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Técnicas de Cultivo , Citocinas/farmacología , Endopeptidasas/metabolismo , Femenino , Inyecciones Intraarticulares , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/fisiología , Rótula/metabolismo , Proteoglicanos/efectos de los fármacos , Proteoglicanos/metabolismo , Porcinos , Regulación hacia Arriba
14.
EMBO J ; 20(19): 5332-41, 2001 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11574464

RESUMEN

The proinflammatory cytokine interleukin 17 (IL-17) is the founding member of a family of secreted proteins that elicit potent cellular responses. We report a novel human IL-17 homolog, IL-17F, and show that it is expressed by activated T cells, can stimulate production of other cytokines such as IL-6, IL-8 and granulocyte colony-stimulating factor, and can regulate cartilage matrix turnover. Unexpectedly, the crystal structure of IL-17F reveals that IL-17 family members adopt a monomer fold typical of cystine knot growth factors, despite lacking the disulfide responsible for defining the canonical "knot" structure. IL-17F dimerizes in a parallel manner like neurotrophins, and features an unusually large cavity on its surface. Remarkably, this cavity is located in precisely the same position where nerve growth factor binds its high affinity receptor, TrkA, suggesting further parallels between IL-17s and neurotrophins with respect to receptor recognition.


Asunto(s)
Interleucina-17/química , Receptores de Interleucina/metabolismo , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Cartílago/metabolismo , Cristalografía por Rayos X , Cistina/química , Dimerización , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Familia de Multigenes , Estructura Terciaria de Proteína , ARN Mensajero/aislamiento & purificación , Receptores de Interleucina-17 , Proteínas Recombinantes/química , Homología de Secuencia de Aminoácido , Linfocitos T/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA