Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Tissue Res ; 374(2): 329-338, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29971480

RESUMEN

The present study attempts to elucidate the role of TRPV1 cation channel receptor on primary repair in an incision-wounded mouse cornea in vivo. Previous study revealed that blocking TRPV1 suppressed myofibroblast formation and expression of transforming growth factor ß1 (TGFß1) in cultured keratocytes or ocular fibroblasts. Male C57BL/6 (wild-type; WT) mice and male C57BL/6 Trpv1-null (KO) mice incurred a full-thickness incision injury (1.8 mm in length, limbus to limbus) in the central cornea of one eye with a surgical blade under general and topical anesthesia. The injury was not sutured. On days 0, 5, and 10, the eyes were enucleated, processed for histology, immunohistochemistry, and real-time RT-PCR gene expression analysis to evaluate the effects of the loss of TRPV1 on primary healing. Electron microscopy observation was also performed to know the effect of the loss of TRPV1 on ultrastructure of keratocytes. The results showed that the loss of Trpv1 gene delayed closure of corneal stromal incision with hindered myofibroblast transdifferentiation along with declines in the expression of collagen Ia1 and TGFß1. Inflammatory cell infiltration was not affected by the loss of TRPV1. Ultrastructurally endoplasmic reticulum of TRPV1-null keratocytes was more extensively dilated as compared with WT keratocytes, suggesting an impairment of protein secretion by TRPV1-gene knockout. These results indicate that injury-related TRPV1 signal is involved in healing of stromal incision injury in a mouse cornea by selectively stimulating TGFß-induced granulation tissue formation.


Asunto(s)
Lesiones de la Cornea/patología , Canales Catiónicos TRPV/deficiencia , Cicatrización de Heridas , Animales , Córnea/patología , Córnea/ultraestructura , Lesiones de la Cornea/metabolismo , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/patología , Canales Catiónicos TRPV/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 313(3): L615-L627, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28642261

RESUMEN

The intracellular signaling mechanisms through which TGF-ß regulates pulmonary development are incompletely understood. Canonical TGF-ß signaling involves Smad2/3 phosphorylation, Smad2/3·Smad4 complex formation and nuclear localization, and gene regulation. Here, we show that physiologically relevant TGF-ß1 levels also stimulate Smad1/5 phosphorylation, which is typically a mediator of bone morphogenetic protein (BMP) signaling, in mouse pup pulmonary artery smooth muscle cells (mPASMC) and lung fibroblasts and other interstitial lung cell lines. This cross-talk mechanism likely has in vivo relevance because mixed Smad1/5/8·Smad2/3 complexes, which are indicative of TGF-ß-stimulated Smad1/5 activation, were detected in the developing mouse lung using a proximity ligation assay. Although mixed Smad complexes have been shown not to transduce nuclear signaling, we determined that TGF-ß stimulates nuclear localization of phosphorylated Smad1/5 and induces the expression of prototypical BMP-regulated genes in the mPASMC. Small-molecule kinase inhibitor studies suggested that TGF-ß-regulated Smad1/5 phosphorylation in these cells is mediated by TGF-ß-type I receptors, not BMP-type I receptors, but possibly the accessory activin-like kinase (ALK1) receptor. Although work by others suggested that ALK1 is expressed exclusively in endothelial cells in the vasculature, we detected ALK1 mRNA and protein expression in mPASMC in vitro and in mouse pup lungs. Moreover, using an antimurine ALK1 antibody and mPASMC, we determined that ALK1 regulates Smad1/5 phosphorylation by TGF-ß. Together, these studies characterize an accessory TGF-ß-stimulated BMP R-Smad signaling mechanism in interstitial cells of the developing lung. They also indicate the importance of considering alternate Smad pathways in studies directed at determining how TGF-ß regulates newborn lung development.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Fibroblastos/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/citología , Transducción de Señal/efectos de los fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Receptores de Activinas Tipo II , Animales , Animales Recién Nacidos , Benzodioxoles/farmacología , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Humanos , Imidazoles/farmacología , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Ratones , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Piridinas/farmacología , Ratas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
3.
Lab Invest ; 96(6): 641-51, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26950486

RESUMEN

We evaluated the effects of the loss of Smad3 on the development of experimental argon laser-induced choroidal neovascularization (CNV) in mice. An in vitro angiogenesis model was also used to examine the role of transforming growth factor-ß1 (TGFß1)/Smad3 signaling in vessel-like tube formation by human umbilical vein endothelial cells (HUVECs). CNV was induced in eyes of 8-12-week-old B6.129-background Smad3-deficient (KO) mice (n=47) and wild-type (WT) mice (n=47) by argon laser irradiation. Results showed that the size of the CNV induced was significantly smaller in KO mice as compared with WT mice at day 14 as revealed by high-resolution angiography with fluorescein isothiocyanate-dextran. Immunohistochemistry and real-time reverse transcription-polymerase chain reaction of RNA extracted from laser-irradiated choroidal tissues were conducted on specimens at specific timepoints. Invasion of macrophages (F4/80+), but not neutrophils (myeloperoxidase+), and appearance of myofibroblasts (α-smooth muscle actin+) were suppressed in laser-irradiated KO tissues. mRNA expression of inflammation-related factors, that is, vascular endothelial growth factor (VEGF), macrophage-chemoattractant protein-1 (MCP-1), interleukin-6 (IL-6) and TGFß1 in choroidal tissues was suppressed by the loss of Smad3. We then examined the effects of adding a Smad3 inhibitor, SIS3, or an ALK5 inhibitor, SB431542, on tube formation promoted by TGFß1 or VEGF in HUVECs cocultured with fibroblast feeder. Further addition of SIS3 or SB431542 augmented vessel-like tube formation by HUVECs in the presence of TGFß1 or VEGF. In conclusion, lack of Smad3 attenuated the growth of laser-induced CNV with suppression of inflammation by macrophages in mice. Because blocking TGFß1/Smad3 signal stimulated the activity of angiogenesis of HUVECs in vitro, the reduction of CNV in vivo in KO mice is attributed to a decrease in growth factor levels in the tissue by the loss of Smad3.


Asunto(s)
Neovascularización Coroidal/prevención & control , Macrófagos/patología , Proteína smad3/deficiencia , Animales , Neovascularización Coroidal/etiología , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mediadores de Inflamación/metabolismo , Isoquinolinas/farmacología , Láseres de Gas/efectos adversos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Piridinas/farmacología , Pirroles/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína smad3/antagonistas & inhibidores , Proteína smad3/genética , Factor de Crecimiento Transformador beta1
4.
Biol Blood Marrow Transplant ; 21(6): 1083-90, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25771402

RESUMEN

Sclerotic skin manifestations of chronic graft-versus-host disease (ScGVHD) lead to significant morbidity, including functional disability from joint range of motion (ROM) restriction. No superior second-line therapy has been established for steroid-refractory disease. Imatinib mesylate is a multikinase inhibitor of several signaling pathways implicated in skin fibrosis with in vitro antifibrotic activity. We performed an open-label pilot phase II trial of imatinib in children and adults with corticosteroid-refractory ScGVHD. Twenty patients were enrolled in a 6-month trial. Eight received a standard dose (adult, 400 mg daily; children, 260 mg/m(2) daily). Because of poor tolerability, 12 additional patients underwent a dose escalation regimen (adult, 100 mg daily initial dose up to 200 mg daily maximum; children, initial dose 65 mg/m(2) daily up to 130 mg/m(2) daily). Fourteen patients were assessable for primary response, improvement in joint ROM deficit, at 6 months. Primary outcome criteria for partial response was met in 5 of 14 (36%), stable disease in 7 of 14 (50%), and progressive disease in 2 of 14 (14%) patients. Eleven patients (79%), including 5 with partial response and 6 with stable disease, demonstrated a positive gain in ROM (range of 3% to 94% improvement in deficit). Of 13 patients with measurable changes at 6 months, the average improvement in ROM deficit was 24.2% (interquartile range, 15.5% to 30.5%; P = .011). This trial is registered at http://clinicaltrials.gov as NCT007020689.


Asunto(s)
Antineoplásicos/uso terapéutico , Fascitis/terapia , Enfermedad Injerto contra Huésped/terapia , Trasplante de Células Madre Hematopoyéticas , Mesilato de Imatinib/uso terapéutico , Leucemia/terapia , Enfermedades de la Piel/terapia , Adolescente , Adulto , Niño , Esquema de Medicación , Fascitis/inmunología , Fascitis/patología , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Articulaciones/efectos de los fármacos , Articulaciones/inmunología , Articulaciones/patología , Leucemia/inmunología , Leucemia/patología , Masculino , Persona de Mediana Edad , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapéutico , Proyectos Piloto , Prednisona/uso terapéutico , Rango del Movimiento Articular/efectos de los fármacos , Recurrencia , Enfermedades de la Piel/inmunología , Enfermedades de la Piel/patología , Tacrolimus/uso terapéutico , Trasplante Homólogo
5.
J Biol Chem ; 288(44): 32074-92, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24056369

RESUMEN

Three homologues of TGF-ß exist in mammals as follows: TGF-ß1, TGF-ß2, and TGF-ß3. All three proteins share high homology in their amino acid sequence, yet each TGF-ß isoform has unique heterologous motifs that are highly conserved during evolution. Although these TGF-ß proteins share similar properties in vitro, isoform-specific properties have been suggested through in vivo studies and by the unique phenotypes for each TGF-ß knock-out mouse. To test our hypothesis that each of these homologues has nonredundant functions, and to identify such isoform-specific roles, we genetically exchanged the coding sequence of the mature TGF-ß1 ligand with a sequence from TGF-ß3 using targeted recombination to create chimeric TGF-ß1/3 knock-in mice (TGF-ß1(Lß3/Lß3)). In the TGF-ß1(Lß3/Lß3) mouse, localization and activation still occur through the TGF-ß1 latent associated peptide, but cell signaling is triggered through the TGF-ß3 ligand that binds to TGF-ß receptors. Unlike TGF-ß1(-/-) mice, the TGF-ß1(Lß3/Lß3) mice show neither embryonic lethality nor signs of multifocal inflammation, demonstrating that knock-in of the TGF-ß3 ligand can prevent the vasculogenesis defects and autoimmunity associated with TGF-ß1 deficiency. However, the TGF-ß1(Lß3/Lß3) mice have a shortened life span and display tooth and bone defects, indicating that the TGF-ß homologues are not completely interchangeable. Remarkably, the TGF-ß1(Lß3/Lß3) mice display an improved metabolic phenotype with reduced body weight gain and enhanced glucose tolerance by induction of beneficial changes to the white adipose tissue compartment. These findings reveal both redundant and unique nonoverlapping functional diversity in TGF-ß isoform signaling that has relevance to the design of therapeutics aimed at targeting the TGF-ß pathway in human disease.


Asunto(s)
Glucosa/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta3/metabolismo , Animales , Células COS , Chlorocebus aethiops , Técnicas de Sustitución del Gen , Glucosa/genética , Células Hep G2 , Humanos , Inflamación/genética , Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Neovascularización Fisiológica/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Porcinos , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta3/genética
6.
Lab Invest ; 94(9): 1030-41, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25068659

RESUMEN

We examined whether the loss of transient receptor potential ankyrin 1 (TRPA1), an irritant-sensing ion channel, or TRPA1 antagonist treatment affects the severity inflammation and scarring during tissue wound healing in a mouse cornea injury model. In addition, the effects of the absence of TRPA1 on transforming growth factor ß1 (TGF-ß1)-signaling activation were studied in cell culture. The lack of TRPA1 in cultured ocular fibroblasts attenuated expression of TGF-ß1, interleukin-6, and α-smooth muscle actin, a myofibroblast the marker, but suppressed the activation of Smad3, p38 MAPK, ERK, and JNK. Stroma of the healing corneas of TRPA1(-/-) knockout (KO) mice appeared more transparent compared with those of wild-type mice post-alkali burn. Eye globe diameters were measured from photographs. An examination of the corneal surface and eye globes suggested the loss of TRPA1 suppressed post-alkali burn inflammation and fibrosis/scarring, which was confirmed by histology, immunohistochemistry, and gene expression analysis. Reciprocal bone marrow transplantation between mice showed that KO corneal tissue resident cells, but not KO bone marrow-derived cells, are responsible for KO mouse wound healing with reduced inflammation and fibrosis. Systemic TRPA1 antagonists reproduced the KO phenotype of healing. In conclusion, a loss or blocking of TRPA1 in mice reduces inflammation and fibrosis/scarring in the corneal stroma during wound healing following an alkali burn. The responsible mechanism may include the inhibition of TGF-ß1-signaling cascades in fibroblasts by attenuated TRPA1 signaling. Inflammatory cells are considered to have a minimum involvement in the exhibition of the KO phenotype after injury.


Asunto(s)
Enfermedades de la Córnea/prevención & control , Fibrosis/prevención & control , Inflamación/prevención & control , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Canales de Potencial de Receptor Transitorio/fisiología , Animales , Enfermedades de la Córnea/patología , Quemaduras Oculares/fisiopatología , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/genética , Cicatrización de Heridas
7.
Breast Cancer Res ; 16(3): R57, 2014 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-24890385

RESUMEN

INTRODUCTION: Transforming growth factor-ßs (TGF-ßs) play a dual role in breast cancer, with context-dependent tumor-suppressive or pro-oncogenic effects. TGF-ß antagonists are showing promise in early-phase clinical oncology trials to neutralize the pro-oncogenic effects. However, there is currently no way to determine whether the tumor-suppressive effects of TGF-ß are still active in human breast tumors at the time of surgery and treatment, a situation that could lead to adverse therapeutic responses. METHODS: Using a breast cancer progression model that exemplifies the dual role of TGF-ß, promoter-wide chromatin immunoprecipitation and transcriptomic approaches were applied to identify a core set of TGF-ß-regulated genes that specifically reflect only the tumor-suppressor arm of the pathway. The clinical significance of this signature and the underlying biology were investigated using bioinformatic analyses in clinical breast cancer datasets, and knockdown validation approaches in tumor xenografts. RESULTS: TGF-ß-driven tumor suppression was highly dependent on Smad3, and Smad3 target genes that were specifically enriched for involvement in tumor suppression were identified. Patterns of Smad3 binding reflected the preexisting active chromatin landscape, and target genes were frequently regulated in opposite directions in vitro and in vivo, highlighting the strong contextuality of TGF-ß action. An in vivo-weighted TGF-ß/Smad3 tumor-suppressor signature was associated with good outcome in estrogen receptor-positive breast cancer cohorts. TGF-ß/Smad3 effects on cell proliferation, differentiation and ephrin signaling contributed to the observed tumor suppression. CONCLUSIONS: Tumor-suppressive effects of TGF-ß persist in some breast cancer patients at the time of surgery and affect clinical outcome. Carefully tailored in vitro/in vivo genomic approaches can identify such patients for exclusion from treatment with TGF-ß antagonists.


Asunto(s)
Neoplasias de la Mama/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética , Proteínas Supresoras de Tumor/genética , Neoplasias de la Mama/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Efrinas/metabolismo , Femenino , Humanos , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño , Receptor EphA2/metabolismo , Proteína Smad2/genética , Proteína smad3/biosíntesis , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/biosíntesis , Proteínas Supresoras de Tumor/antagonistas & inhibidores
8.
Lab Invest ; 93(2): 207-17, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23207449

RESUMEN

We investigated the effects of loss of tenascin C on the healing of the stroma using incision-injured mice corneas. Tenascin C was upregulated in the stroma following incision injury to the cornea. Wild-type (WT) and tenascin C-null (knockout (KO)) mice on a C57BL/6 background were used. Cell culture experiments were also conducted to determine the effects of the lack of tenascin C on fibrogenic gene expression in ocular fibroblasts. Histology, immunohistochemistry and real-time reverse transcription PCR were employed to evaluate the healing process in the stroma. The difference in the incidence of wound closure was statistically analyzed in hematoxylin and eosin-stained samples between WT and KO mice in addition to qualitative observation. Healing of incision injury in corneal stroma was delayed, with less appearance of myofibroblasts, less invasion of macrophages and reduction in expression of collagen Iα1, fibronectin and transforming growth factor ß1 (TGFß1) in KO mice compared with WT mice. In vitro experiments showed that the loss of tenascin C counteracted TGFß1 acceleration of mRNA expression of TGFß1, and of collagen Iα1 and of myofibroblast conversion in ocular fibroblasts. These results indicate that tenascin C modulates wound healing-related fibrogenic gene expression in ocular fibroblasts and is required for primary healing of the corneal stroma.


Asunto(s)
Lesiones de la Cornea , Sustancia Propia/fisiopatología , Regulación de la Expresión Génica/genética , Tenascina/deficiencia , Cicatrización de Heridas/fisiología , Análisis de Varianza , Animales , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Fibronectinas/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta1/metabolismo
9.
J Lipid Res ; 53(12): 2698-707, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23034213

RESUMEN

Transforming growth factor-ß (TGFß) is activated as a result of liver injury, such as cholestasis. However, its influence on endogenous metabolism is not known. This study demonstrated that TGFß regulates hepatic phospholipid and bile acid homeostasis through MAD homolog 3 (SMAD3) activation as revealed by lithocholic acid-induced experimental intrahepatic cholestasis. Lithocholic acid (LCA) induced expression of TGFB1 and the receptors TGFBR1 and TGFBR2 in the liver. In addition, immunohistochemistry revealed higher TGFß expression around the portal vein after LCA exposure and diminished SMAD3 phosphorylation in hepatocytes from Smad3-null mice. Serum metabolomics indicated increased bile acids and decreased lysophosphatidylcholine (LPC) after LCA exposure. Interestingly, in Smad3-null mice, the metabolic alteration was attenuated. LCA-induced lysophosphatidylcholine acyltransferase 4 (LPCAT4) and organic solute transporter ß (OSTß) expression were markedly decreased in Smad3-null mice, whereas TGFß induced LPCAT4 and OSTß expression in primary mouse hepatocytes. In addition, introduction of SMAD3 enhanced the TGFß-induced LPCAT4 and OSTß expression in the human hepatocellular carcinoma cell line HepG2. In conclusion, considering that Smad3-null mice showed attenuated serum ALP activity, a diagnostic indicator of cholangiocyte injury, these results strongly support the view that TGFß-SMAD3 signaling mediates an alteration in phospholipid and bile acid metabolism following hepatic inflammation with the biliary injury.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Hígado/metabolismo , Fosfolípidos/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células Cultivadas , Células Hep G2 , Hepatocitos/química , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ácido Litocólico , Hígado/efectos de los fármacos , Hígado/lesiones , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos
10.
Am J Pathol ; 178(6): 2654-64, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21641388

RESUMEN

We examined whether absence or blocking of transient receptor potential vanilloid subtype 1 (TRPV1) affects the level of inflammation and fibrosis/scarring during healing of injured tissue using an alkali burn model of cornea in mice. A cornea burn was produced with 1 N NaOH instilled into one eye of TRPV1-/- (KO) (n = 88) or TRPV1+/+ (n = 94) mice. Examinations of the corneal surface and eye globe size suggested that the loss of TRPV1 suppressed inflammation and fibrosis/scarring after alkali burn, and this was confirmed by histology, IHC, and gene expression analysis. The loss of TRPV1 inhibited inflammatory cell invasion and myofibroblast generation in association with reduction of expression of proinflammatory and profibrogenic components. Experiments of bone marrow transplantation between either genotype of mice showed that KO corneal tissue resident cells, but not KO bone marrow-derived cells, are responsible for KO-type wound healing with reduced inflammation and fibrosis. The absence of TRPV1 attenuated expression of transforming growth factor ß 1 (TGFß1) and other proinflammatory gene expression in cultured ocular fibroblasts, but did not affect TGFß1 expression in macrophages. Loss of TRPV1 inhibited myofibroblast transdifferentiation in cultured fibroblasts. Systemic TRPV1 antagonists reproduced the KO type of healing. In conclusion, absence or blocking of TRPV1 suppressed inflammation and fibrosis/scarring during healing of alkali-burned mouse cornea. TRPV1 is a potential drug target for improving the outcome of inflammatory/fibrogenic wound healing.


Asunto(s)
Quemaduras Oculares/patología , Inflamación/metabolismo , Inflamación/patología , Canales Catiónicos TRPV/metabolismo , Álcalis , Animales , Células Cultivadas , Técnicas de Cocultivo , Córnea/metabolismo , Córnea/patología , Quemaduras Oculares/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Inmunohistoquímica , Inflamación/genética , Macrófagos/metabolismo , Macrófagos/patología , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Cicatrización de Heridas/genética
11.
Lab Invest ; 90(4): 543-55, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20142803

RESUMEN

Transforming growth factor-beta (TGF-beta) signaling is known to affect salivary gland physiology by influencing branching morphogenesis, regulating ECM deposition, and controlling immune homeostasis. To study the role of TGF-beta1 in the salivary gland, we created a transgenic mouse (beta1(glo)) that conditionally overexpresses active TGF-beta1 upon genomic recombination by Cre recombinase. beta1(glo) mice were bred with an MMTV (mouse mammary tumor virus)-Cre (MC) transgenic line that expresses the Cre recombinase predominantly in the secretory cells of both the mammary and salivary glands. Although most of the double positive (beta1(glo)/MC) pups die either in utero or just after birth, clear defects in salivary gland morphogenesis such as reduced branching and increased mesenchyme could be seen. Those beta1(glo)/MC mice that survived into adulthood, however, had hyposalivation due to salivary gland fibrosis and acinar atrophy. Increased TGF-beta signaling was observed in the salivary gland with elevated phosphorylation of Smad2 and concomitant increase in ECM deposition. In particular, aberrant TGF-beta1 overexpression caused salivary gland hypofunction in this mouse model because of the replacement of normal glandular parenchyma with interstitial fibrous tissue. These results further implicate TGF-beta in pathological cases of salivary gland inflammation and fibrosis that occur with chronic infections in the glands or with the autoimmune disease, Sjögren's syndrome, or with radiation therapy given to head-and-neck cancer patients.


Asunto(s)
Enfermedades de las Glándulas Salivales/fisiopatología , Glándulas Salivales/crecimiento & desarrollo , Factor de Crecimiento Transformador beta1/fisiología , Xerostomía/fisiopatología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis/fisiopatología , Inflamación/fisiopatología , Ratones , Ratones Transgénicos , Glándulas Salivales/patología
12.
Breast Cancer Res Treat ; 121(3): 727-35, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19937272

RESUMEN

The transforming growth factor beta (TGF-beta) pathway can play either a tumor-suppressing or a tumor-promoting role in human breast carcinogenesis. In order to determine whether expression of TGF-beta signaling factors varies by age at onset and breast tumor characteristics that have prognostic significance, we undertook a study of 623 women with invasive breast carcinoma enrolled in a population-based case-control study conducted in Poland from 2000 to 2003. TGF-beta signaling factors were analyzed by immunohistochemistry in tumor tissue microarrays. We found that most tumors expressed extracellular-TGF-beta1 (78%), TGF-beta2 (91%), TGF-beta3 (93%), TGF-betaR2 (72%), and phospho-SMAD2 (61%), whereas intracellular-TGF-beta1 was expressed in 32% of tumors. Expression of TGF-beta ligands (beta1, beta2, and beta3) was associated with prognostically favorable pathological features including small size, and low grade, and these associations were similar for ER-positive and negative tumors. On the contrary, expression of the receptor TGF-betaR2 was primarily associated with small tumor size among ER-negative tumors, while expression of the transcription factor phospho-SMAD2 was associated with positive nodal status among ER-negative tumors. The greater frequency of expression of phospho-SMAD2 in cancers associated with lymph node metastases is consistent with a pro-progression role for TGF-beta. In addition, expression of extracellular-TGF-beta1 (P = 0.005), TGF-betaR2 (P = 8.2E-11), and phospho-SMAD2 (P = 1.3E-8) was strongly associated with earlier age at onset, independent of ER status. Our data provide evidence that TGF-beta signaling patterns vary by age and pathologic features of prognostic significance including ER expression. These results warrant analysis in studies of clinical outcomes accounting for age, ER status and treatment.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Distribución por Edad , Edad de Inicio , Anciano , Neoplasias de la Mama/metabolismo , Estudios de Casos y Controles , Femenino , Humanos , Incidencia , Persona de Mediana Edad , Polonia/epidemiología , Pronóstico , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Estrógenos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/metabolismo
13.
Mol Vis ; 16: 1194-205, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20664686

RESUMEN

PURPOSE: To investigate the role of tenascin-C in epithelial-mesenchymal transition (EMT) of the lens epithelium during wound healing in mice. Tenascin-C is a component of the extracellular matrix in patients having post-operative capsular opacification. METHODS: The crystalline lens was injured by needle puncture in tenascin-C null (KO, n=56) and wild-type (WT, n=56) mice in a C57BL/6 background. The animals were killed at day 2, 5, or 10 post-injury. Immunohistochemistry was employed to detect alpha-smooth muscle actin (alphaSMA), a marker of EMT, collagen type I, transforming growth factor beta1 (TGFbeta1), phospho-Smad2, phospho-adducin, and phospho-myosin light chain 9 (MLC9). The expression levels of phospho-adducin and phospho-MLC9 were used as markers for the activation of protein kinase C and Rho kinase, respectively. RESULTS: The expression of tenascin-C was upregulated in WT lens epithelial cells adjacent to the capsular break at day 5. The results showed that injury-induced EMT of the mouse lens epithelium, as evaluated by histology and the expression patterns of alphaSMA and fibronectin, was attenuated in the absence of tenascin-C. Upregulation of TGFbeta1 expression in the epithelium was also inhibited, and loss of tenascin-C attenuated the phosphorylation of Smad2 and adducin in epithelial cells adjacent to the capsular break. The expression of phospho-adducin was suppressed, while the expression level of phospho-MLC9 was unchanged, in the healing epithelium in the absence of tenascin C. CONCLUSIONS: Tenascin-C is required for injury-induced EMT in the mouse lens epithelium. The mechanism behind this might involve impaired activation of cytoplasmic signaling cascades; i.e., TGFbeta/Smad and protein kinase C-adducing signaling, in the absence of tenascin-C.


Asunto(s)
Epitelio/lesiones , Epitelio/patología , Cristalino/lesiones , Cristalino/patología , Mesodermo/patología , Tenascina/deficiencia , Animales , Proteínas de Unión a Calmodulina/metabolismo , Epitelio/metabolismo , Fibronectinas/metabolismo , Inmunohistoquímica , Cristalino/metabolismo , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , 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/metabolismo , Proteína Smad2/metabolismo , Tenascina/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
14.
J Mammary Gland Biol Neoplasia ; 14(2): 131-44, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19396528

RESUMEN

During rodent mammary gland involution there is a dramatic increase in the expression of the transforming growth factor-beta isoform, TGF-beta3. The TGF-betas are multifunctional cytokines which play important roles in wound healing and in carcinogenesis. The responses that are activated in the remodeling of the gland during involution have many similarities with the wound healing process and have been postulated to generate a mammary stroma that provides a microenvironment favoring tumor progression. In this review we will discuss the putative role of TGF-beta during involution, as well as its effects on the mammary microenvironment and possible implications for pregnancy-associated tumorigenesis.


Asunto(s)
Neoplasias de la Mama/patología , Lactancia/fisiología , Glándulas Mamarias Animales/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Apoptosis/fisiología , Neoplasias de la Mama/genética , Cicatriz/fisiopatología , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Inflamación/fisiopatología , Ratones , Paridad , Embarazo , Complicaciones Neoplásicas del Embarazo/fisiopatología , Pronóstico , Isoformas de Proteínas/fisiología , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta3/fisiología , Resultado del Tratamiento , Cicatrización de Heridas/fisiología
15.
Clin Cancer Res ; 26(3): 643-656, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31582516

RESUMEN

PURPOSE: TGFßs are overexpressed in many advanced cancers and promote cancer progression through mechanisms that include suppression of immunosurveillance. Multiple strategies to antagonize the TGFß pathway are in early-phase oncology trials. However, TGFßs also have tumor-suppressive activities early in tumorigenesis, and the extent to which these might be retained in advanced disease has not been fully explored. EXPERIMENTAL DESIGN: A panel of 12 immunocompetent mouse allograft models of metastatic breast cancer was tested for the effect of neutralizing anti-TGFß antibodies on lung metastatic burden. Extensive correlative biology analyses were performed to assess potential predictive biomarkers and probe underlying mechanisms. RESULTS: Heterogeneous responses to anti-TGFß treatment were observed, with 5 of 12 models (42%) showing suppression of metastasis, 4 of 12 (33%) showing no response, and 3 of 12 (25%) showing an undesirable stimulation (up to 9-fold) of metastasis. Inhibition of metastasis was immune-dependent, whereas stimulation of metastasis was immune-independent and targeted the tumor cell compartment, potentially affecting the cancer stem cell. Thus, the integrated outcome of TGFß antagonism depends on a complex balance between enhancing effective antitumor immunity and disrupting persistent tumor-suppressive effects of TGFß on the tumor cell. Applying transcriptomic signatures derived from treatment-naïve mouse primary tumors to human breast cancer datasets suggested that patients with breast cancer with high-grade, estrogen receptor-negative disease are most likely to benefit from anti-TGFß therapy. CONCLUSIONS: Contrary to dogma, tumor-suppressive responses to TGFß are retained in some advanced metastatic tumors. Safe deployment of TGFß antagonists in the clinic will require good predictive biomarkers.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Neoplasias Pulmonares/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Resultado del Tratamiento
16.
Am J Pathol ; 173(1): 68-76, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18502822

RESUMEN

Our previous work showed that 6 weeks after cutaneous irradiation, mice null (knockout, KO) for Smad3, a cytoplasmic downstream mediator of transforming growth factor-beta, demonstrate less epidermal acanthosis and dermal inflammation than wild-type (WT) Smad3 mice. Analysis of the kinetics of inflammation showed that 6 to 8 hours after skin irradiation, there was a transient sevenfold increase in neutrophil influx in Smad3 KO mice compared with WT. Herein we describe bone marrow transplantation and skin grafting between WT and KO mice to assess the contribution of the neutrophil genotype compared with that of irradiated skin to the induction of neutrophil migration after irradiation. Results from bone marrow transplantation showed that WT marrow transplanted into KO mice enhanced neutrophil migration 6 to 8 hours after irradiation by 3.2-fold compared with KO marrow in WT mice. KO skin grafted onto either WT or KO animals showed a sixfold elevation of neutrophils after irradiation compared with grafted WT skin. These results suggest that the genotype of the irradiated skin, rather than the inflammatory cell, controls neutrophil influx. Circulating neutrophils, increased in WT mice after injection of granulocyte colony-stimulating factor, resulted in increased neutrophil migration to the skin 6 to 8 hours after irradiation and less skin damage 6 weeks after irradiation compared with untreated WT mice. Thus, early responses, including enhanced neutrophil influx, appear to contribute to subsequent cutaneous radioprotection.


Asunto(s)
Infiltración Neutrófila/efectos de la radiación , Piel/efectos de la radiación , Proteína smad3/deficiencia , Proteína smad3/genética , Animales , Trasplante de Médula Ósea , Quimiocina CXCL1/biosíntesis , Quimiocina CXCL2/biosíntesis , Genotipo , Ratones , Ratones Noqueados , Infiltración Neutrófila/inmunología , Piel/inmunología , Trasplante de Piel
17.
Int J Oncol ; 35(2): 315-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19578745

RESUMEN

Fibrosis of normal tissues often accompanies radiation treatment of cancer. Activation of the transforming growth factor-beta (TGF-beta) signaling pathway is thought to play a major role in radiation-induced fibrosis and has prompted the development and assessment of low molecular weight inhibitors of the pathway. Previous studies with halofuginone have shown it to inhibit TGF-beta signaling in vitro and protect mice from radiation-induced leg contraction (a model for soft tissue fibrosis). The current study confirms these findings for HaCaT cells stimulated with exogenous TGF-beta treatment. Reducing the halifuginone treatment from 7 days/week (used previously) to 5 days/week post-radiation exposure provided significant protection against radiation-induced leg contraction in mice 3 and 4 months post-radiation treatment. Halofuginone treatment was shown to attenuate TGF-beta signaling molecules taken from irradiated skin including TGF-betaRII, pSmad3, Smad7, and TSP1. The latter, TSP1, a co-activator of TGF-beta may serve as a suitable biomarker for monitoring the efficacy of halofuginone should it be evaluated in a clinical setting for protection against radiation-induced fibrosis.


Asunto(s)
Contractura/prevención & control , Fibrosis/prevención & control , Piperidinas/farmacología , Quinazolinonas/farmacología , Radioterapia/efectos adversos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Línea Celular , Femenino , Humanos , Pierna , Ratones , Ratones Endogámicos C3H , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/efectos de los fármacos
18.
Liver Int ; 29(7): 997-1009, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19422482

RESUMEN

BACKGROUND: Hepatic fibrosis is characterized by a progressive accumulation of fibrillar extracellular matrix (ECM) proteins including collagen, which occurs in most types of chronic liver diseases. Transforming growth factor-beta (TGF-beta)/Smad3 signalling plays a central role in tissue fibrogenesis, acting as a potent stimulus of ECM accumulation. AIM: To evaluate the potential protective role of Smad3 deficiency in the pathogenesis of liver fibrosis induced by dimethylnitrosamine (DMN) in Smad3 null mice. METHODS: Chronic hepatitis-associated fibrosis was induced in 13 Smad3 null and 13 wild-type (WT) mice by intraperitoneal DMN administration (10 microg/g body weight/day) for three consecutive days per week for 6 weeks. The liver was excised for macroscopic examination and histological, morphometric and immunohistochemical (IHC) analyses. For IHC, alpha-smooth muscle actin (alpha-SMA), collagen types I-III, TGF-beta1, connective tissue growth factor (CTGF), Smad3, Smad7 and CD3 antibodies were used. RESULTS: At macroscopic examination, the liver of DMN-treated Smad3 WT appeared harder with a dark brown colouring and necrotic areas compared with that from null mice. Histological and morphometric evaluation revealed a significantly higher degree of hepatic fibrosis and accumulation of connective tissue in the Smad3 WT compared with null mice. IHC evaluation showed a marked increase in alpha-SMA, CTGF, collagen I-III, TGF-beta and Smad3 staining in the liver of Smad3 WT compared with that in null mice, whereas Smad7 was increased only in null mice. CONCLUSIONS: The results indicate that Smad3 loss confers resistance to the development of DMN-induced hepatic fibrosis. The reduced fibrotic response appears to be due to a reduction of fibrogenic myofibroblast activation and ECM production and accumulation. Smad3 could be a novel target for potential treatment of fibrosis complicating chronic hepatitis.


Asunto(s)
Cirrosis Hepática/prevención & control , Hígado/metabolismo , Proteína smad3/deficiencia , Actinas/metabolismo , Envejecimiento , Animales , Complejo CD3/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Dimetilnitrosamina , Colágenos Fibrilares/metabolismo , Inmunohistoquímica , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones , Ratones Noqueados , Índice de Severidad de la Enfermedad , Proteína smad3/genética , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
19.
Cytokine Growth Factor Rev ; 17(1-2): 19-27, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16290023

RESUMEN

Smads2 and 3 transduce signals of TGF-beta from the cell surface to the nucleus. We used mice with a targeted deletion of Smad3 to study the specific contributions of this signaling pathway to pathogenic effects of TGF-beta. Focusing on models involving epithelial-to-mesenchymal transition (EMT), including injury to the lens and retina of the eye and to the kidney, we have found that loss of Smad3 blocks EMT and attenuates development of fibrotic sequelae. Smad3 also plays a critical role in both the tumor suppressor and pro-metastatic effects of TGF-beta in carcinogenesis. These observations suggest that development of small molecule inhibitors of Smad3 might have clinical application in treatment of fibrotic diseases as well as late stage cancers.


Asunto(s)
Células Epiteliales/metabolismo , Mesodermo/citología , Metástasis de la Neoplasia/patología , Neoplasias/prevención & control , Proteína smad3/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Diferenciación Celular/fisiología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Fibrosis/metabolismo , Humanos , Mesodermo/metabolismo , Neoplasias/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/genética
20.
Mol Cancer Ther ; 6(1): 154-62, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17237276

RESUMEN

Synthetic triterpenoids have been developed, which are potent inducers of cytoprotective enzymes and inhibitors of inflammation, greatly improving on the weak activity of naturally occurring triterpenoids. An imidazolide triterpenoid derivative, 1-[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole (CDDO-Im or TP235), has been previously shown to potently protect against hepatic tumorigenesis, acting in part by inducing cytoprotective genes through Keap1-Nrf2-antioxidant response element (ARE) signaling. In these studies, the pharmacodynamic activity of CDDO-Im is characterized in two distinct lines of ARE reporter mice and by measuring increases in Nqo1 transcript levels as a marker of cytoprotective gene induction. Oral administration of CDDO-Im induces ARE-regulated cytoprotective genes in many tissues in the mouse, including liver, lung, kidney, intestines, brain, heart, thymus, and salivary gland. CDDO-Im induces Nqo1 RNA transcripts in some organs at doses as low as 0.3 mumol/kg body weight (orally). A structure activity evaluation of 15 additional triterpenoids (a) confirmed the importance of Michael acceptor groups on both the A and C rings, (b) showed the requirement for a nitrile group at C-2 of the A ring, and (c) indicated that substituents at C-17 dramatically affected pharmacodynamic action in vivo. In addition to CDDO-Im, other triterpenoids, particularly the methyl ester CDDO-Me (TP155) and the dinitrile TP225, are extremely potent inducers of cytoprotective genes in mouse liver, lung, small intestine mucosa, and cerebral cortex. This pharmacodynamic characterization highlights the chemopreventive promise of several synthetic triterpenoids in multiple target organs.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Factor 2 Relacionado con NF-E2/genética , Triterpenos/farmacología , Administración Oral , Animales , Antioxidantes/metabolismo , Quimioprevención , Relación Dosis-Respuesta a Droga , Genes Reporteros/genética , Imidazoles/química , Hígado/citología , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Masculino , Ratones , NAD(P)H Deshidrogenasa (Quinona) , NADPH Deshidrogenasa/genética , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Elementos de Respuesta/genética , Activación Transcripcional , Triterpenos/administración & dosificación , Triterpenos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA