Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Atherosclerosis ; 373: 17-28, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37121163

RESUMEN

BACKGROUND AND AIMS: Sprouty1 (Spry1) regulates the differentiation of vascular smooth muscle cells (VSMC), and our aim was to determine its role in atherogenesis. A significant proportion of cells within atherosclerotic lesions are derived from migration and pathological adaptation of medial VSMC. METHODS: We used global Spry1 null mouse, and Myh11-CreERT2, ROSA26-STOPfl/fl-tdTomato-Spry1fl/fl mice to allow for lineage tracing and conditional Spry1 deletion in VSMC. Atherosclerosis was induced by injection of a mutant form of mPCSK9D377Y-AAV followed by Western diet. Human aortic VSMC (hVSMC) with shRNA targeting of Spry1 were also analyzed. RESULTS: Global loss of Spry1 increased inflammatory markers ICAM1 and Cox2 in VSMC. Conditional deletion of Spry1 in VSMC had no effect on early lesion development, despite increased Sca1high cells. After 26 weeks of Western diet, mice with VSMC deletion of Spry1 had increased plaque burden, with reduced collagen content and smooth muscle alpha actin (SMA) in the fibrous cap. Lineage tracing via tdTomato marking Cre-recombined cells indicated that VSMC with loss of Spry1 had decreased migration into the lesion, noted by decreased proportions of tdTomato+ and tdTomato+/SMA + cells. Loss-of-function of Spry1 in hVSMC increased mesenchymal and activation markers, including KLF4, PDGFRb, ICAM1, and Cox2. Loss of Spry1 enhanced the effects of PDGFBB and TNFa on hVSMC. CONCLUSIONS: Loss of Spry1 in VSMC aggravated plaque formation at later stages, and increased markers of instability. Our results indicate that Spry1 suppresses the mesenchymal and inflammatory phenotype of VSMC, and its expression in VSMC is protective against chronic atherosclerotic disease.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Animales , Humanos , Ratones , Aterosclerosis/genética , Aterosclerosis/prevención & control , Aterosclerosis/metabolismo , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fenotipo , Placa Aterosclerótica/metabolismo
2.
J Cell Biochem ; 119(4): 3267-3279, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29105817

RESUMEN

Signals from growth factors or mechanical stimuli converge to promote vascular smooth muscle cell (VSMC) migration and proliferation, key events in the pathogenesis of intimal hyperplasia upon vascular injury. Spry1, a regulator of receptor tyrosine kinases (RTK), plays a role in maintaining the contractile phenotype of VSMC. The aim of the current study was to determine the role of Spry1 in VSMC proliferation in vitro and injury induced neointimal hyperplasia in vivo. VSMC proliferation and neointima formation were evaluated in cultured human aortic SMC (hAoSMC) and ligation-induced injury of mouse carotid arteries from Spry1 gene targeted mice, and their corresponding wild type littermates. Human Spry1 or non-targeting control lentiviral shRNAs were used to knock down Spry1 in hAoSMC. Time course cell cycle analysis showed a reduced fraction of S-phase cells at 12 and 24 h after growth medium stimulation in Spry1 shRNA transduced hAoSMC. Consistent with reduced S-phase entry, the induction of cyclinD1 and the levels of pRbS807/S811, pH3Ser10, and pCdc2 were also reduced, while the cell cycle inhibitor p27Kip1 was maintained in Spry1 knockdown hAoSMC. In vivo, loss of Spry1 attenuated carotid artery ligation-induced neointima formation in mice, and this effect was accompanied by a decrease in cell proliferation similar to the in vitro results. Our findings demonstrate that loss of Spry1 attenuates mitogen-induced VSMC proliferation, and thus injury-induced neointimal hyperplasia likely via insufficient activation of Akt signaling causing decreased cyclinD1 and increased p27Kip1 and a subsequent decrease in Rb and cdc2 phosphorylation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Traumatismos de las Arterias Carótidas/complicaciones , Proteínas de la Membrana/genética , Músculo Liso Vascular/citología , Neointima/genética , Fosfoproteínas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Ciclo Celular , Proliferación Celular , Células Cultivadas , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
3.
J Cell Biochem ; 117(10): 2346-56, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26950413

RESUMEN

Sef (similar expression to fgf), also know as IL17RD, is a transmembrane protein shown to inhibit fibroblast growth factor signaling in developmental and cancer contexts; however, its role as a tumor suppressor remains to be fully elucidated. Here, we show that Sef regulates epithelial-mesenchymal transition (EMT) in breast cancer cell lines. Sef expression was highest in the normal breast epithelial cell line MCF10A, intermediate expression in MCF-7 cells and lowest in MDA-MB-231 cells. Knockdown of Sef increased the expression of genes associated with EMT, and promoted cell migration, invasion, and a fibroblastic morphology of MCF-7 cells. Overexpression of Sef inhibited the expression of EMT marker genes and inhibited cell migration and invasion in MCF-7 cells. Induction of EMT in MCF10A cells by TGF-ß and TNF-α resulted in downregulation of Sef expression concomitant with upregulation of EMT gene expression and loss of epithelial morphology. Overexpression of Sef in MCF10A cells partially blocked cytokine-induced EMT. Sef was shown to block ß-catenin mediated luciferase reporter activity and to cause a decrease in the nuclear localization of active ß-catenin. Furthermore, Sef was shown to co-immunoprecipitate with ß-catenin. In a mouse orthotopic xenograft model, Sef overexpression in MDA-MB-231 cells slowed tumor growth and reduced expression of EMT marker genes. Together, these data indicate that Sef plays a role in the negative regulation of EMT in a ß-catenin dependent manner and that reduced expression of Sef in breast tumor cells may be permissive for EMT and the acquisition of a more metastatic phenotype. J. Cell. Biochem. 117: 2346-2356, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Receptores de Interleucina/metabolismo , Animales , Apoptosis , Western Blotting , Neoplasias de la Mama/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas
4.
J Cell Biochem ; 117(9): 2182-93, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26910604

RESUMEN

Our objective was to characterize lipid profiles in cell models of adipocyte differentiation in comparison to mouse adipose tissues in vivo. A novel lipid extraction strategy was combined with global lipid profiling using direct infusion and sequential precursor ion fragmentation, termed MS/MS(ALL) . Perirenal and inguinal white adipose tissue and interscapular brown adipose tissues from adult C57BL/6J mice were analyzed. 3T3-L1 preadipocytes, ear mesenchymal progenitor cells, and brown adipose-derived BAT-C1 cells were also characterized. Over 3000 unique lipid species were quantified. Principal component analysis showed that perirenal versus inguinal white adipose tissues varied in lipid composition of triacyl- and diacylglycerols, sphingomyelins, glycerophospholipids and, notably, cardiolipin CL 72:3. In contrast, hexosylceramides and sphingomyelins distinguished brown from white adipose. Adipocyte differentiation models showed broad differences in lipid composition among themselves, upon adipogenic differentiation, and with adipose tissues. Palmitoyl triacylglycerides predominate in 3T3-L1 differentiation models, whereas cardiolipin CL 72:1 and SM 45:4 were abundant in brown adipose-derived cell differentiation models, respectively. MS/MS(ALL) data suggest new lipid biomarkers for tissue-specific lipid contributions to adipogenesis, thus providing a foundation for using in vitro models of adipogenesis to reflect potential changes in adipose tissues in vivo. J. Cell. Biochem. 117: 2182-2193, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Adipocitos Blancos/metabolismo , Adipogénesis/fisiología , Tejido Adiposo Blanco/metabolismo , Metabolismo de los Lípidos/fisiología , Modelos Biológicos , Células 3T3-L1 , Adipocitos Blancos/citología , Tejido Adiposo Blanco/citología , Animales , Ratones
5.
J Bone Miner Res ; 29(5): 1217-31, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24127237

RESUMEN

Sef (similar expression to fgf genes) is a feedback inhibitor of fibroblast growth factor (FGF) signaling and functions in part by binding to FGF receptors and inhibiting their activation. Genetic studies in mice and humans indicate an important role for fibroblast growth factor signaling in bone growth and homeostasis. We, therefore, investigated whether Sef had a function role in skeletal acquisition and remodeling. Sef expression is increased during osteoblast differentiation in vitro, and LacZ staining of Sef+/- mice showed high expression of Sef in the periosteum and chondro-osseous junction of neonatal and adult mice. Mice with a global deletion of Sef showed increased cortical bone thickness, bone volume, and increased periosteal perimeter by micro-computed tomography (micro-CT). Histomorphometric analysis of cortical bone revealed a significant increase in osteoblast number. Interestingly, Sef-/- mice showed very little difference in trabecular bone by micro-CT and histomorphometry compared with wild-type mice. Bone marrow cells from Sef-/- mice grown in osteogenic medium showed increased proliferation and increased osteoblast differentiation compared with wild-type bone marrow cells. Bone marrow cells from Sef-/- mice showed enhanced FGF2-induced activation of the ERK pathway, whereas bone marrow cells from Sef transgenic mice showed decreased FGF2-induced signaling. FGF2-induced acetylation and stability of Runx2 was enhanced in Sef-/- bone marrow cells, whereas overexpression of Sef inhibited Runx2-responsive luciferase reporter activity. Bone marrow from Sef-/- mice showed enhanced hematopoietic lineage-dependent and osteoblast-dependent osteoclastogenesis and increased bone resorptive activity relative to wild-type controls in in vitro assays, whereas overexpression of Sef inhibited osteoclast differentiation. Taken together, these studies indicate that Sef has specific roles in osteoblast and osteoclast lineages and that its absence results in increased osteoblast and osteoclast activity with a net increase in cortical bone mass.


Asunto(s)
Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de la Membrana/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Periostio/metabolismo , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Tamaño de los Órganos , Osteoblastos/patología , Osteoclastos/patología , Periostio/patología
6.
Angiogenesis ; 16(4): 861-75, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23955631

RESUMEN

Angiogenesis is mediated by signaling through receptor tyrosine kinases (RTKs), Src family kinases and adhesion receptors such as integrins, yet the mechanism how these signaling pathways regulate one another remains incompletely understood. The RTK modulator, Sprouty4 (Spry4) inhibits endothelial cell functions and angiogenesis, but the mechanisms remain to be fully elucidated. In this study, we demonstrate that Spry4 regulates angiogenesis in part by regulating endothelial cell migration. Overexpression of Spry4 in human endothelial cells inhibited migration and adhesion on vitronectin (VTN), whereas knockdown of Spry4 enhanced these behaviors. These activities were shown to be c-Src-dependent and Ras-independent. Spry4 disrupted the crosstalk between vascular endothelial growth factor-2 and integrin αVß3, the receptor for VTN. Spry4 overexpression resulted in decreased integrin ß3 protein levels in a post-transcriptional manner in part by modulating its tyrosine phosphorylation by c-Src. Conversely, knockdown of Spry4 resulted in increased integrin ß3 protein levels and tyrosine phosphorylation. Moreover, in vivo analysis revealed that Spry4 regulated integrin ß3 levels in murine embryos and yolk sacs. Our findings identify an unanticipated role for Spry4 in regulating c-Src activity and integrin ß3 protein levels, which contributes to the regulation of migration and adhesion of endothelial cells. Thus, targeting Spry4 may be exploited as a target in anti-angiogenesis therapies.


Asunto(s)
Células Endoteliales/citología , Integrina beta3/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neovascularización Fisiológica/fisiología , Proteínas del Tejido Nervioso/fisiología , Familia-src Quinasas/metabolismo , Animales , Aorta/citología , Proteína Tirosina Quinasa CSK , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Embrión de Mamíferos/citología , Células Endoteliales/metabolismo , Activación Enzimática , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfaVbeta3/fisiología , Integrina beta3/fisiología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Fosforilación , Fosfotirosina/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Vasos Retinianos/crecimiento & desarrollo , Regulación hacia Arriba , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Vitronectina/metabolismo , Saco Vitelino/citología
7.
PLoS One ; 8(3): e58746, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23554919

RESUMEN

BACKGROUND: Changes in the vascular smooth muscle cell (VSMC) contractile phenotype occur in pathological states such as restenosis and atherosclerosis. Multiple cytokines, signaling through receptor tyrosine kinases (RTK) and PI3K/Akt and MAPK/ERK pathways, regulate these phenotypic transitions. The Spry proteins are feedback modulators of RTK signaling, but their specific roles in VSMC have not been established. METHODOLOGY/PRINCIPAL FINDINGS: Here, we report for the first time that Spry1, but not Spry4, is required for maintaining the differentiated state of human VSMC in vitro. While Spry1 is a known MAPK/ERK inhibitor in many cell types, we found that Spry1 has little effect on MAPK/ERK signaling but increases and maintains Akt activation in VSMC. Sustained Akt signaling is required for VSMC marker expression in vitro, while ERK signaling negatively modulates Akt activation and VSMC marker gene expression. Spry4, which antagonizes both MAPK/ERK and Akt signaling, suppresses VSMC differentiation marker gene expression. We show using siRNA knockdown and ChIP assays that FoxO3a, a downstream target of PI3K/Akt signaling, represses myocardin promoter activity, and that Spry1 increases, while Spry4 decreases myocardin mRNA levels. CONCLUSIONS: Together, these data indicate that Spry1 and Spry4 have opposing roles in VSMC phenotypic modulation, and Spry1 maintains the VSMC differentiation phenotype in vitro in part through an Akt/FoxO/myocardin pathway.


Asunto(s)
Aorta/metabolismo , Factores de Transcripción Forkhead/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transactivadores/metabolismo , Diferenciación Celular , Línea Celular , Células Cultivadas , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Expresión Génica , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Miocitos del Músculo Liso/citología , Proteínas del Tejido Nervioso/genética , Fenotipo , Fosfoproteínas/genética , Transducción de Señal , Transcripción Genética
8.
Open Med Imaging J ; 7: 19-27, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-24563682

RESUMEN

The quantitative analysis of blood vessel volumes from magnetic resonance angiograms (MRA) or µCT images is difficult and time-consuming. This fact, when combined with a study that involves multiple scans of multiple subjects, can represent a significant portion of research time. In order to enhance analysis options and to provide an automated and fast analysis method, we developed a software plugin for the ImageJ and Fiji image processing frameworks that enables the quick and reproducible volume quantification of blood vessel segments. The novel plugin named Volume Calculator (VolCal), accepts any binary (thresholded) image and produces a three-dimensional schematic representation of the vasculature that can be directly manipulated by the investigator. Using MRAs of the mouse hindlimb ischemia model, we demonstrate quick and reproducible blood vessel volume calculations with 95 - 98% accuracy. In clinical settings this software may enhance image interpretation and the speed of data analysis and thus enhance intervention decisions for example in peripheral vascular disease or aneurysms. In summary, we provide a novel, fast and interactive quantification of blood vessel volumes for single blood vessels or sets of vessel segments with particular focus on collateral formation after an ischemic insult.

9.
J Vasc Res ; 48(6): 485-94, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21832838

RESUMEN

BACKGROUND: Transforming growth factor-ß (TGF-ß) plays an important role in vascular homeostasis through effects on vascular smooth muscle cells (SMC). Fine-tuning of TGF-ß signaling occurs at the level of ALK receptors or Smads, and is regulated with cell type specificity. METHODS: Our goal was to understand TGF-ß signaling in regulating SMC differentiation marker expression in human SMC. Activation of Smads was characterized, and loss- and gain-of-function reagents used to define ALK pathways. In addition, Smad-independent mechanisms were determined. RESULTS: TGF-ß type I receptors, ALK1 and ALK5, are expressed in human SMC, and TGF-ß1 phosphorylates Smad1/5/8 and Smad2/3 in a time- and dosage-dependent pattern. ALK5 activity, not bone morphogenetic protein type I receptors, is required for Smad phosphorylation. Endoglin, a TGF-ß type III receptor, is a TGF-ß1 target in SMC, yet endoglin does not modify TGF-ß1 responsiveness. ALK5, not ALK1, is required for TGF-ß1-induction of SMC differentiation markers, and ALK5 signals through an ALK5/Smad3- and MAP kinase-dependent pathway. CONCLUSION: The definition of the specific signaling downstream of TGF-ß regulating SMC differentiation markers will contribute to a better understanding of vascular disorders involving changes in SMC phenotype.


Asunto(s)
Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Receptores de Activinas Tipo II/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Aorta/citología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Endoglina , Expresión Génica/fisiología , Humanos , Músculo Liso Vascular/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Smad1/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
10.
Genesis ; 48(9): 563-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20645309

RESUMEN

The regulatory elements of the Tie2/Tek promoter are commonly used in mouse models to direct transgene expression to endothelial cells. Tunica intima endothelial kinase 2 (Tie2) is also expressed in hematopoietic cells, although this has not been fully characterized. We determine the lineages of adult hematopoietic cells derived from Tie2-expressing populations using Tie2-Cre;Rosa26R-EYFP mice. In Tie2-Cre;Rosa26R-EYFP mice, analysis of bone marrow cells showed Cre-mediated recombination in 85% of the population. In adult bone marrow and spleen, we analyzed subclasses of early hematopoietic progenitors, T cells, monocytes, granulocytes, and B cells. We found that ∼ 84% of each lineage was EYFP(+), and nearly all cells that come from Tie2-expressing lineages are CD45(+), confirming widespread contribution to definitive hematopoietic cells. In addition, more than 82% of blood cells within the embryonic yolk sac were of Tie2(+) origin. Our findings of high levels of Tie2-Cre recombination in the hematopoietic lineage have implications for the use of the Tie2-Cre mouse as a lineage-restricted driver strain.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Recombinación Genética/genética , Animales , Proteínas Bacterianas/metabolismo , Células de la Médula Ósea/metabolismo , Cartilla de ADN/genética , Citometría de Flujo , Integrasas/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Receptor TIE-2 , Bazo/metabolismo
11.
FASEB J ; 24(9): 3264-73, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20410440

RESUMEN

Development of bone and adipose tissue are linked processes arising from a common progenitor cell, but having an inverse relationship in disease conditions such as osteoporosis. Cellular differentiation of both tissues relies on growth factor cues, and we focus this study on Sprouty1 (Spry1), an inhibitor of growth factor signaling. We tested whether Spry1 can modify the development of fat cells through its activity in regulating growth factors known to be important for adipogenesis. We utilized conditional expression and genetic-null mouse models of Spry1 in adipocytes using the fatty acid binding promoter (aP2). Conditional deletion of Spry1 results in 10% increased body fat and decreased bone mass. This phenotype was rescued on Spry1 expression, which results in decreased body fat and increased bone mass. Ex vivo bone marrow experiments indicate Spry1 in bone marrow and adipose progenitor cells favors differentiation of osteoblasts at the expense of adipocytes by suppressing CEBP-beta and PPARgamma while up regulating TAZ. Age and gender-matched littermates expressing only Cre recombinase were used as controls. Spry1 is a critical regulator of adipocyte differentiation and mesenchymal stem cell (MSC) lineage allocation, potentially acting through regulation of CEBP-beta and TAZ.


Asunto(s)
Diferenciación Celular , Proteínas de la Membrana/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Fosfoproteínas/fisiología , Absorciometría de Fotón , Proteínas Adaptadoras Transductoras de Señales , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/genética , Adipogénesis/fisiología , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Células Cultivadas , Hipertrofia/genética , Hipertrofia/fisiopatología , Immunoblotting , Inmunohistoquímica , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteogénesis/genética , Osteogénesis/fisiología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , Microtomografía por Rayos X
12.
Dev Biol ; 318(2): 323-34, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18471809

RESUMEN

Haploinsufficiency of the transcription factor TWIST1 is associated with Saethre-Chotzen Syndrome and is manifested by craniosynostosis, which is the premature closure of the calvaria sutures. Previously, we found that Twist1 forms functional homodimers and heterodimers that have opposing activities. Our data supported a model that within the calvaria sutures Twist1 homodimers (T/T) reside in the osteogenic fronts while Twist1/E protein heterodimers (T/E) are in the mid-sutures. Twist1 haploinsufficiency alters the balance between these dimers, favoring an increase in homodimer formation throughout the sutures. The data we present here further supports this model and extends it to integrate the Twist1 dimers with the pathways that are known to regulate cranial suture patency. This data provides the first evidence of a functional link between Twist1 and the FGF pathway, and indicates that differential regulation of FGF signaling by T/T and T/E dimers plays a central role in governing cranial suture patency. Furthermore, we show that inhibition of FGF signaling prevents craniosynostosis in Twist1(+/-) mice, demonstrating that inhibition of a signaling pathway that is not part of the initiating mutation can prevent suture fusion in a relevant genetic model of craniosynostosis.


Asunto(s)
Suturas Craneales/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Craneosinostosis/metabolismo , Dimerización , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Osteoblastos/citología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
13.
Am J Pathol ; 171(3): 1023-36, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17675579

RESUMEN

Notch functions as an oncogene or tumor inhibitor in various cancers, and decreases in Notch2 expression are associated with increasing grade of human breast cancer. We constitutively activated Notch signaling with intracellular domain (ICD) expression in the human adenocarcinoma line MDA-MB-231. Notch2 signaling increased apoptosis, whereas Notch4ICD (int3) significantly increased cell proliferation and growth. Cells with activated Notch2 or Notch4 were injected into nu/nu mice for analysis of in vivo tumor xenograft phenotype. Tumor growth was significantly altered depending on the receptor activated. Notch2ICD potently suppressed tumor take and growth, leading to a 60% decrease in tumors and significantly smaller, necrotic tumors. Despite this, Notch2ICD tumors were highly vascularized, although the vessels were smaller and comprised a more immature network compared with Notch4ICD tumors. Notch4ICD tumors were highly aggressive and well vascularized, indicating a role for Notch4 signaling in the promotion of the malignant phenotype in addition to its transforming ability. Although both NotchICD groups expressed angiogenic factors, Notch4ICD had selective vascular endothelial growth factor-D in both tumor and host stroma, suggesting a differential regulation of cytokines that may impact vascular recruitment and autocrine tumor signaling. Our results demonstrate that Notch2 signaling is a potent inhibitory signal in human breast cancer xenografts.


Asunto(s)
Apoptosis/fisiología , Neoplasias de la Mama , Trasplante de Neoplasias , Receptor Notch2/metabolismo , Transducción de Señal/fisiología , Trasplante Heterólogo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptor Notch2/genética , Receptor Notch4 , Receptores Notch/genética , Receptores Notch/metabolismo , Trasplante Heterólogo/patología , Trasplante Heterólogo/fisiología
14.
Arterioscler Thromb Vasc Biol ; 25(1): 77-83, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15514205

RESUMEN

OBJECTIVE: Periostin mRNA is among the most strongly upregulated transcripts in rat carotid arteries after balloon injury. The goal of the present study was to gain insight into the significance of periostin in the vasculature. METHODS AND RESULTS: Periostin expression after injury was localized to smooth muscle cells of the neointima and the adventitia. The expression of periostin in smooth muscle cells in vitro was not regulated by cytokines such as fibroblast growth factor-2 (FGF-2). In contrast, stimulation of MC3T3-E1 osteoblastic cells, NIH3T3 fibroblasts, or mesenchymal C3H10T1/2 cells with FGF-2 reduced periostin mRNA levels to <5% of controls, whereas conversely bone morphogenetic protein-2 (BMP-2) increased periostin mRNA levels. Periostin expression was induced and maintained during retinoic acid-induced smooth muscle cell differentiation in A404 cells. In addition, overexpression of periostin in C3H10T1/2 cells caused an increase in cell migration that could be blocked with an anti-periostin antibody. CONCLUSIONS: Periostin expression is associated with smooth muscle cell differentiation in vitro and promotes cell migration. Unlike other mesenchymally derived cell lines, periostin expression is not regulated by FGF-2 in smooth muscle cells. This distinction may be useful in discriminating smooth muscle and fibroblast lineages.


Asunto(s)
Vasos Sanguíneos/citología , Traumatismos de las Arterias Carótidas/genética , Moléculas de Adhesión Celular/biosíntesis , Diferenciación Celular/genética , Movimiento Celular/genética , Células 3T3/química , Células 3T3/metabolismo , Animales , Vasos Sanguíneos/química , Vasos Sanguíneos/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Arterias Carótidas/química , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Cateterismo/efectos adversos , Moléculas de Adhesión Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/fisiología , Clonación Molecular/métodos , Regulación de la Expresión Génica/fisiología , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C3H , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/química , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Células 3T3 NIH/química , Células 3T3 NIH/metabolismo , Osteosarcoma/genética , Osteosarcoma/patología , Ratas , Ratas Sprague-Dawley , Homología de Secuencia de Aminoácido
15.
Circ Res ; 96(2): 261-8, 2005 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-15618538

RESUMEN

Collagen triple helix repeat containing 1 (Cthrc1) was identified in a screen for differentially expressed sequences in balloon-injured versus normal arteries. Cthrc1 expression was not detectable in normal arteries. However, on injury it was transiently expressed by fibroblasts of the remodeling adventitia and by smooth muscle cells of the neointima. It was also found in the matrix of calcifying human atherosclerotic plaques. CTHRC1 is a secreted 28-kDa protein that is glycosylated and highly conserved from lower chordates to mammals. A short collagen motif with 12 Gly-X-Y repeats appears to be responsible for trimerization of the protein and this renders the molecule susceptible to cleavage by collagenase. Cthrc1 mRNA expression levels are increased in response to transforming growth factor-beta and bone morphogenetic protein-4. Cell migration assays performed with CTHRC1-overexpressing fibroblasts and smooth muscle cells demonstrate that increased CTHRC1 levels are associated with enhanced migratory ability. Furthermore, CTHRC1 overexpression caused a dramatic reduction in collagen type I mRNA and protein levels. Our data indicate that the novel molecule CTHRC1 is transiently expressed in the arterial wall in response to injury where it may contribute to vascular remodeling by limiting collagen matrix deposition and promoting cell migration.


Asunto(s)
Aorta/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Glicoproteínas/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Aorta/lesiones , Biopolímeros , Células CHO , Calcinosis/metabolismo , Traumatismos de las Arterias Carótidas/etiología , Traumatismos de las Arterias Carótidas/genética , Estenosis Carotídea/etiología , Cateterismo/efectos adversos , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Colagenasas/metabolismo , Cricetinae , Cricetulus , Evolución Molecular , Proteínas de la Matriz Extracelular , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Datos de Secuencia Molecular , Mioblastos/metabolismo , Mioblastos/patología , Miocitos del Músculo Liso/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/fisiología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Especificidad de la Especie , Factor de Crecimiento Transformador beta/fisiología , Vertebrados/genética , Vertebrados/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA