Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Muscle Nerve ; 64(6): 765-769, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34486132

RESUMEN

INTRODUCTION/AIMS: Klf10 is a member of the Krüppel-like family of transcription factors, which is implicated in mediating muscle structure (fiber size, organization of the sarcomere), muscle metabolic activity (respiratory chain), and passive force. The aim of this study was to further characterize the roles of Klf10 in the contractile properties of skeletal muscle fibers. METHODS: Fifty-two single fibers were extracted from female wild-type (WT) and Klf10 knockout (KO) oxidative (soleus) and glycolytic (extensor digitorum longus [EDL]) skinned muscles. Each fiber was immersed successively in relaxing (R), washing (W), and activating (A) solutions. Calcium was included in the activating solution to induce a maximum contraction of the fiber. The maximum force (Fmax ) was measured and normalized to the cross-sectional area to obtain the maximum stress (Stressmax ). After a steady state in contraction was reached, a quick stretch-release was performed; the force at the maximum stretch (Fstretch ) was measured and the stiffness was assessed. RESULTS: Deletion of the Klf10 gene induced changes in the contractile parameters (Fmax , Stressmax , Stiffness), which were lower and higher for soleus and EDL fibers compared with littermates, respectively. These measurements also revealed changes in the proportion and resistance of attached cross-bridges. DISCUSSION: Klf10 plays a major role in the homeostasis of the contractile behavior of skeletal muscle fibers in a muscle fiber type-specific manner. These findings further implicate important roles for Klf10 in skeletal muscle function and shed new light on understanding the molecular processes regulating the contractility of skeletal muscle fibers.


Asunto(s)
Contracción Muscular , Fibras Musculares Esqueléticas , Animales , Factores de Transcripción de la Respuesta de Crecimiento Precoz/análisis , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Factores de Transcripción de Tipo Kruppel/análisis , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Contracción Muscular/fisiología , Fibras Musculares de Contracción Rápida/fisiología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético , Factores de Transcripción/genética
2.
Proc Natl Acad Sci U S A ; 115(41): E9580-E9589, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30257941

RESUMEN

Triple-negative breast cancer (TNBC) accounts for a disproportionately high number of deaths due to a lack of targeted therapies and an increased likelihood of distant recurrence. Estrogen receptor beta (ERß), a well-characterized tumor suppressor, is expressed in 30% of TNBCs, and its expression is associated with improved patient outcomes. We demonstrate that therapeutic activation of ERß elicits potent anticancer effects in TNBC through the induction of a family of secreted proteins known as the cystatins, which function to inhibit canonical TGFß signaling and suppress metastatic phenotypes both in vitro and in vivo. These data reveal the involvement of cystatins in suppressing breast cancer progression and highlight the value of ERß-targeted therapies for the treatment of TNBC patients.


Asunto(s)
Cistatinas/metabolismo , Receptor beta de Estrógeno/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular Tumoral , Cistatinas/genética , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/genética , Femenino , Humanos , Ratones , Factor de Crecimiento Transformador beta/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Proteínas Supresoras de Tumor/agonistas , Proteínas Supresoras de Tumor/genética
3.
J Cell Physiol ; 235(7-8): 5679-5688, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31975377

RESUMEN

Deletion of TGFß inducible early gene-1 (TIEG) in mice results in an osteopenic phenotype that exists only in female animals. Molecular analyses on female TIEG knockout (KO) mouse bones identified increased expression of sclerostin, an effect that was confirmed at the protein level in serum. Sclerostin antibody (Scl-Ab) therapy has been shown to elicit bone beneficial effects in multiple animal model systems and human clinical trials. For these reasons, we hypothesized that Scl-Ab therapy would reverse the low bone mass phenotype of female TIEG KO mice. In this study, wildtype (WT) and TIEG KO female mice were randomized to either vehicle control (Veh, n = 12/group) or Scl-Ab therapy (10 mg/kg, 1×/wk, s.c.; n = 12/group) and treated for 6 weeks. Following treatment, bone imaging analyses revealed that Scl-Ab therapy significantly increased cancellous and cortical bone in the femur of both WT and TIEG KO mice. Similar effects also occurred in the vertebra of both WT and TIEG KO animals. Additionally, histomorphometric analyses revealed that Scl-Ab therapy resulted in increased osteoblast perimeter/bone perimeter in both WT and TIEG KO animals, with a concomitant increase in P1NP, a serum marker of bone formation. In contrast, osteoclast perimeter/bone perimeter and CTX-1 serum levels were unaffected by Scl-Ab therapy, irrespective of mouse genotype. Overall, our findings demonstrate that Scl-Ab therapy elicits potent bone-forming effects in both WT and TIEG KO mice and effectively increases bone mass in female TIEG KO mice.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Enfermedades Óseas Metabólicas/genética , Proteínas de Unión al ADN/genética , Osteogénesis/genética , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/sangre , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Anticuerpos/farmacología , Densidad Ósea/genética , Desarrollo Óseo/genética , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Enfermedades Óseas Metabólicas/inmunología , Enfermedades Óseas Metabólicas/patología , Femenino , Fémur/crecimiento & desarrollo , Fémur/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Ratones Noqueados , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Fenotipo
4.
Breast Cancer Res Treat ; 179(1): 241-249, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31571071

RESUMEN

PURPOSE: Significant controversy exists regarding the expression patterns of estrogen receptor beta (ERß) in normal and diseased breast tissue. To address this issue, we have validated two ERß antibodies, optimized the IHC protocols for both antibodies and now report the expression patterns of ERß in normal and malignant breast tissues. METHODS: ERß antibody specificity was determined using western blot and IHC analysis. ERß protein expression patterns were assessed via IHC in normal breast tissue and invasive breast carcinoma. Further, we report the detailed protocol of the ERß IHC assay developed in our CAP/CLIA certified laboratory to provide a standardized method for future studies. RESULTS: We have confirmed the specificity of two independent ERß monoclonal antibodies, one that detects total (i.e., full length plus splice variants 2-5, which do not include the ligand binding domain) ERß protein (PPZ0506) and one that detects only the full-length form, which includes the ligand binding domain, of ERß (PPG5/10). Using these two antibodies, we demonstrate that ERß is highly expressed in normal human breast tissue as well as in 20-30% of invasive breast cancers. Further, these two antibodies exhibited similar staining patterns across multiple different tissues and were highly concordant with regard to determining ERß positivity in breast cancers. CONCLUSIONS: ERß protein was shown to be abundant in the majority of normal breast epithelial cells and is present in 20-30% of breast cancers. Use of these two antibodies, along with their standardized IHC protocols, provide a reference for future studies aimed at determining the utility of ERß as a prognostic and/or predictive biomarker in various tissues of benign or malignant states.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Neoplasias de la Mama/diagnóstico , Mama/metabolismo , Receptor beta de Estrógeno/metabolismo , Empalme Alternativo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Detección Precoz del Cáncer , Receptor beta de Estrógeno/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Sensibilidad y Especificidad
5.
Nat Immunol ; 9(3): 245-53, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18278048

RESUMEN

Transforming growth factor-beta (TGF-beta) signaling in naive T cells induces expression of the transcription factor Foxp3, a 'master' regulator of regulatory T cells (T(reg) cells). However, the molecular mechanisms leading to Foxp3 induction remain unclear. Here we show that Itch-/- T cells were resistant to TGF-beta treatment and had less Foxp3 expression. The E3 ubiquitin ligase Itch associated with and promoted conjugation of ubiquitin to the transcription factor TIEG1. Itch cooperated with TIEG1 to induce Foxp3 expression, which was reversed by TIEG1 deficiency. Functionally, 'TGF-beta-converted' T(reg) cells generated from TIEG1-deficient mice were unable to suppress airway inflammation in vivo. These results suggest TIEG and Itch contribute to a ubiquitin-dependent nonproteolytic pathway that regulates inducible Foxp3 expression and the control of allergic responses.


Asunto(s)
Factores de Transcripción de la Respuesta de Crecimiento Precoz/fisiología , Factores de Transcripción Forkhead/metabolismo , Inflamación/inmunología , Factores de Transcripción de Tipo Kruppel/fisiología , Proteínas Represoras/fisiología , Sistema Respiratorio/inmunología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/inmunología , Hipersensibilidad/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Represoras/genética , Transducción de Señal/inmunología , Células Th2/inmunología , Transfección , Ubiquitina-Proteína Ligasas/genética
6.
Nucleic Acids Res ; 45(9): 5170-5182, 2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28201653

RESUMEN

We have previously demonstrated that TGFß Inducible Early Gene-1 (TIEG1), also known as KLF10, plays important roles in mediating skeletal development and homeostasis in mice. TIEG1 has also been identified in clinical studies as one of a handful of genes whose altered expression levels or allelic variations are associated with decreased bone mass and osteoporosis in humans. Here, we provide evidence for the first time that TIEG1 is involved in regulating the canonical Wnt signaling pathway in bone through multiple mechanisms of action. Decreased Wnt signaling in the absence of TIEG1 expression is shown to be in part due to impaired ß-catenin nuclear localization resulting from alterations in the activity of AKT and GSK-3ß. We also provide evidence that TIEG1 interacts with, and serves as a transcriptional co-activator for, Lef1 and ß-catenin. Changes in Wnt signaling in the setting of altered TIEG1 expression and/or activity may in part explain the observed osteopenic phenotype of TIEG1 KO mice as well as the known links between TIEG1 expression levels/allelic variations and patients with osteoporosis.


Asunto(s)
Huesos/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Huesos/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ligandos , Cloruro de Litio/farmacología , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Cráneo/citología , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
7.
Nucleic Acids Res ; 45(1): 127-141, 2017 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-27651452

RESUMEN

Proper temporal epigenetic regulation of gene expression is essential for cell fate determination and tissue development. The Bromodomain-containing Protein-4 (BRD4) was previously shown to control the transcription of defined subsets of genes in various cell systems. In this study we examined the role of BRD4 in promoting lineage-specific gene expression and show that BRD4 is essential for osteoblast differentiation. Genome-wide analyses demonstrate that BRD4 is recruited to the transcriptional start site of differentiation-induced genes. Unexpectedly, while promoter-proximal BRD4 occupancy correlated with gene expression, genes which displayed moderate expression and promoter-proximal BRD4 occupancy were most highly regulated and sensitive to BRD4 inhibition. Therefore, we examined distal BRD4 occupancy and uncovered a specific co-localization of BRD4 with the transcription factors C/EBPb, TEAD1, FOSL2 and JUND at putative osteoblast-specific enhancers. These findings reveal the intricacies of lineage specification and provide new insight into the context-dependent functions of BRD4.


Asunto(s)
Linaje de la Célula/genética , Epigénesis Genética , Células Epiteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteínas Nucleares/genética , Osteoblastos/metabolismo , Osteocitos/metabolismo , Factores de Transcripción/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas de Ciclo Celular , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/citología , Antígeno 2 Relacionado con Fos/genética , Antígeno 2 Relacionado con Fos/metabolismo , Perfilación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Osteoblastos/citología , Osteocitos/citología , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal , Factores de Transcripción de Dominio TEA , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción
8.
J Cell Physiol ; 233(4): 3540-3551, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29044507

RESUMEN

TIEG knockout (KO) mice exhibit a female-specific osteopenic phenotype and altered expression of TIEG in humans is associated with osteoporosis. Gene expression profiling studies identified sclerostin as one of the most highly up-regulated transcripts in the long bones of TIEG KO mice relative to WT littermates suggesting that TIEG may regulate SOST expression. TIEG was shown to substantially suppress SOST promoter activity and the regulatory elements through which TIEG functions were identified using promoter deletion and chromatin immunoprecipitation assays. Knockdown of TIEG in IDG-SW3 osteocyte cells using shRNA and CRISPR-Cas9 technology resulted in increased SOST expression and delayed mineralization, mimicking the results obtained from TIEG KO mouse bones. Given that TIEG is an estrogen regulated gene, and as changes in the hormonal milieu affect SOST expression, we performed ovariectomy (OVX) and estrogen replacement therapy (ERT) studies in WT and TIEG KO mice followed by miRNA and mRNA sequencing of cortical and trabecular compartments of femurs. SOST expression levels were considerably higher in cortical bone compared to trabecular bone. In cortical bone, SOST expression was increased following OVX only in WT mice and was suppressed following ERT in both genotypes. In contrast, SOST expression in trabecular bone was decreased following OVX and significantly increased following ERT. Interestingly, a number of miRNAs that are predicted to target sclerostin exhibited inverse expression levels in response to OVX and ERT. These data implicate important roles for TIEG and estrogen-regulated miRNAs in modulating SOST expression in bone.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Estrógenos/farmacología , Glicoproteínas/metabolismo , Osteocitos/efectos de los fármacos , Esqueleto/metabolismo , Factores de Transcripción/deficiencia , Proteínas Adaptadoras Transductoras de Señales , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Huesos/metabolismo , Femenino , Marcadores Genéticos/fisiología , Péptidos y Proteínas de Señalización Intercelular , Ratones Noqueados , Osteocitos/metabolismo , Ovariectomía/métodos , Esqueleto/efectos de los fármacos
9.
J Biol Chem ; 291(47): 24594-24606, 2016 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-27758858

RESUMEN

Perturbations in skeletal development and bone degeneration may result in reduced bone mass and quality, leading to greater fracture risk. Bone loss is mitigated by bone protective therapies, but there is a clinical need for new bone-anabolic agents. Previous work has demonstrated that Ezh2 (enhancer of zeste homolog 2), a histone 3 lysine 27 (H3K27) methyltransferase, suppressed differentiation of osteogenic progenitors. Here, we investigated whether inhibition of Ezh2 can be leveraged for bone stimulatory applications. Pharmacologic inhibition and siRNA knockdown of Ezh2 enhanced osteogenic commitment of MC3T3 preosteoblasts. Next generation RNA sequencing of mRNAs and real time quantitative PCR profiling established that Ezh2 inactivation promotes expression of bone-related gene regulators and extracellular matrix proteins. Mechanistically, enhanced gene expression was linked to decreased H3K27 trimethylation (H3K27me3) near transcriptional start sites in genome-wide sequencing of chromatin immunoprecipitations assays. Administration of an Ezh2 inhibitor modestly increases bone density parameters of adult mice. Furthermore, Ezh2 inhibition also alleviated bone loss in an estrogen-deficient mammalian model for osteoporosis. Ezh2 inhibition enhanced expression of Wnt10b and Pth1r and increased the BMP-dependent phosphorylation of Smad1/5. Thus, these data suggest that inhibition of Ezh2 promotes paracrine signaling in osteoblasts and has bone-anabolic and osteoprotective potential in adults.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Osteoporosis/metabolismo , Comunicación Paracrina , Animales , Línea Celular , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Metilación/efectos de los fármacos , Ratones , Osteoblastos/patología , Osteoporosis/patología , Ovariectomía , ARN Interferente Pequeño/farmacología , Receptor de Hormona Paratiroídea Tipo 1 , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
10.
Muscle Nerve ; 55(3): 410-416, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27421714

RESUMEN

INTRODUCTION: Transforming growth factor-beta (TGF-ß)-inducible early gene-1 (TIEG1) is a transcription factor that is highly expressed in skeletal muscle. The purpose of this study was to characterize the structural properties of both fast-twitch (EDL) and slow-twitch (soleus) muscles in the hindlimb of TIEG1-deficient (TIEG1-/- ) mice. METHODS: Ten slow and 10 fast muscles were analyzed from TIEG1-/- and wild-type (WT) mice using MRI texture (MRI-TA) and histological analyses. RESULTS: MRI-TA could discriminate between WT slow and fast muscles. Deletion of the TIEG1 gene led to changes in the texture profile within both muscle types. Specifically, muscle isolated from TIEG1-/- mice displayed hypertrophy, hyperplasia, and a modification of fiber area distribution. CONCLUSIONS: We demonstrated that TIEG1 plays an important role in the structural properties of skeletal muscle. This study further implicates important roles for TIEG1 in the development of skeletal muscle and suggests that defects in TIEG1 expression and/or function may be associated with muscle disease. Muscle Nerve 55: 410-416, 2017.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Fibras Musculares de Contracción Rápida/fisiología , Fibras Musculares de Contracción Lenta/fisiología , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Femenino , Miembro Posterior/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis de Componente Principal , ARN Mensajero/metabolismo , Factores de Transcripción/genética
11.
Biochem Biophys Res Commun ; 470(3): 528-533, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26801561

RESUMEN

Deletion of TIEG1/KLF10 in mice results in an osteopenic skeletal phenotype with significant decreases in both bone mineral density and content throughout the skeleton. Calvarial osteoblasts isolated from TIEG1 knockout (KO) mice display numerous changes in gene expression and exhibit significant delays in their mineralization rates relative to wild-type (WT) controls. Here, we demonstrate that loss of TIEG1 expression in osteoblasts results in decreased levels of Osterix mRNA. Suppression of TIEG1 expression in WT osteoblasts leads to decreased Osterix expression while restoration of TIEG1 expression in TIEG1 KO osteoblasts results in increased levels of Osterix. Transient transfection and chromatin immunoprecipitation assays reveal that TIEG1 directly binds to and activates the Osterix promoter and demonstrate that the zinc finger-containing DNA binding domain of TIEG1 is necessary for this regulation. Furthermore, we reveal that TIEG1 expression is essential for the induction of Osterix expression by important bone-related cytokines such as TGFß and BMP2 in osteoblast cells. Taken together, these data implicate an important role for TIEG1 in regulating the expression of Osterix, a master regulator of osteoblast differentiation and bone formation, and suggest that decreased expression of Osterix, as well as impaired TGFß and BMP2 signaling, contribute to the observed osteopenic bone phenotype of TIEG1 KO mice.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Proteínas de Unión al ADN/metabolismo , Osteoblastos/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Enfermedades Óseas Metabólicas/patología , Células Cultivadas , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Osteoblastos/patología , Transducción de Señal , Factor de Transcripción Sp7
12.
Biol Reprod ; 95(3): 62, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27488034

RESUMEN

Endometriosis is a highly prevalent, chronic, heterogeneous, fibro-inflammatory disease that remains recalcitrant to conventional therapy. We previously showed that loss of KLF11, a transcription factor implicated in uterine disease, results in progression of endometriosis. Despite extensive homology, co-expression, and human disease association, loss of the paralog Klf10 causes a unique inflammatory, cystic endometriosis phenotype in contrast to fibrotic progression seen with loss of Klf11. We identify here for the first time a novel role for KLF10 in endometriosis. In an animal endometriosis model, unlike wild-type controls, Klf10(-/-) animals developed cystic lesions with massive immune infiltrate and minimal peri-lesional fibrosis. The Klf10(-/-) disease progression phenotype also contrasted with prolific fibrosis and minimal immune cell infiltration seen in Klf11(-/-) animals. We further found that lesion genotype rather than that of the host determined each unique disease progression phenotype. Mechanistically, KLF10 regulated CD40/CD154-mediated immune pathways. Both inflammatory as well as fibrotic phenotypes are the commonest clinical manifestations in chronic fibro-inflammatory diseases such as endometriosis. The complementary, paralogous Klf10 and Klf11 models therefore offer novel insights into the mechanisms of inflammation and fibrosis in a disease-relevant context. Our data suggests that divergence in underlying gene dysregulation critically determines disease-phenotype predominance rather than the conventional paradigm of inflammation being precedent to fibrotic scarring. Heterogeneity in clinical progression and treatment response are thus likely from disparate gene regulation profiles. Characterization of disease phenotype-associated gene dysregulation offers novel approaches for developing targeted, individualized therapy for recurrent and recalcitrant chronic disease.


Asunto(s)
Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Endometriosis/genética , Endometrio/metabolismo , Epigénesis Genética , Factores de Transcripción de Tipo Kruppel/genética , Adolescente , Adulto , Animales , Línea Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/patología , Femenino , Regulación de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Persona de Mediana Edad , Virus Diminuto del Ratón , Adulto Joven
13.
J Biol Chem ; 288(8): 5291-302, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23300083

RESUMEN

Runx2 and Axin2 regulate craniofacial development and skeletal maintenance. Runx2 is essential for calvarial bone development, as Runx2 haploinsufficiency causes cleidocranial dysplasia. In contrast, Axin2-deficient mice develop craniosynostosis because of high ß-catenin activity. Axin2 levels are elevated in Runx2(-/-) calvarial cells, and Runx2 represses transcription of Axin2 mRNA, suggesting a direct relationship between these factors in vivo. Here we demonstrate that Runx2 binds several regions of the Axin2 promoter and that Runx2-mediated repression of Axin2 transcription depends on Hdac3. To determine whether Runx2 contributes to the etiology of Axin2 deficiency-induced craniosynostosis, we generated Axin2(-/-):Runx2(+/-) mice. These double mutant mice had longer skulls than Axin2(-/-) mice, indicating that Runx2 haploinsufficiency rescued the craniosynostosis phenotype of Axin2(-/-) mice. Together, these studies identify a key mechanistic pathway for regulating intramembranous bone development within the skull that involves Runx2- and Hdac3-mediated suppression of Axin2 to prevent the untimely closure of the calvarial sutures.


Asunto(s)
Proteína Axina/genética , Proteína Axina/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Craneosinostosis/genética , Regulación de la Expresión Génica , Osteoblastos/citología , Células 3T3 , Animales , Células de la Médula Ósea , Huesos/metabolismo , Craneosinostosis/metabolismo , Expresión Génica , Histona Desacetilasas/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Osteoblastos/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Cráneo/patología , Factores de Tiempo , Proteínas Wnt/metabolismo
14.
Eur J Immunol ; 43(1): 258-69, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23065757

RESUMEN

Bone marrow-derived macrophages (BMMs) treated with granulocyte-macrophage colony-stimulating factor (GM-CSF) or macrophage colony-stimulating factor (M-CSF), differentiate into GM-CSF-induced mouse bone marrow-derived macrophages (GM-BMMs) or M-CSF-induced mouse bone marrow-derived macrophages (M-BMMs), which have an M1 or M2 profile, respectively. GM-BMMs produce large amounts of proinflammatory cytokines and mediate resistance to pathogens, whereas M-BMMs produce antiinflammatory cytokines that contribute to tissue repair and remodeling. M-BMMs stimulated with lipopolysaccharide (LPS) are in an antiinflammatory state, with an IL-12(low) IL-10(high) phenotype. However, the regulation of this process remains unclear. Klf10 belongs to the family of Krüppel-like transcription factors and was initially described as a TGF-ß inducible early gene 1. IL-12p40 is upregulated in LPS-stimulated M-BMMs from Klf10-deficient mice, but downregulated during Klf10 overexpression. Klf11, another member of the Krüppel-like factor family, can also repress the production of IL-12p40. Furthermore, Klf10 binds to the CACCC element of the IL-12p40 promoter and inhibits its transcription. We have therefore identified Klf10 as a transcription factor that regulates the expression of IL-12p40 in M-BMMs.


Asunto(s)
Células de la Médula Ósea/inmunología , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Macrófagos/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Factores de Transcripción de la Respuesta de Crecimiento Precoz/inmunología , Regulación de la Expresión Génica/genética , Mediadores de Inflamación/metabolismo , Subunidad p40 de la Interleucina-12/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Transcripción Genética/inmunología , Transgenes/genética
15.
BMC Cancer ; 14: 749, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25288324

RESUMEN

BACKGROUND: The role and clinical value of ERß1 expression is controversial and recent data demonstrates that many ERß antibodies are insensitive and/or non-specific. Therefore, we sought to comprehensively characterize ERß1 expression across all sub-types of breast cancer using a validated antibody and determine the roles of this receptor in mediating response to multiple forms of endocrine therapy both in the presence and absence of ERα expression. METHODS: Nuclear and cytoplasmic expression patterns of ERß1 were analyzed in three patient cohorts, including a retrospective analysis of a prospective adjuvant tamoxifen study and a triple negative breast cancer cohort. To investigate the utility of therapeutically targeting ERß1, we generated multiple ERß1 expressing cell model systems and determined their proliferative responses following anti-estrogenic or ERß-specific agonist exposure. RESULTS: Nuclear ERß1 was shown to be expressed across all major sub-types of breast cancer, including 25% of triple negative breast cancers and 33% of ER-positive tumors, and was associated with significantly improved outcomes in ERα-positive tamoxifen-treated patients. In agreement with these observations, ERß1 expression sensitized ERα-positive breast cancer cells to the anti-cancer effects of selective estrogen receptor modulators (SERMs). However, in the absence of ERα expression, ERß-specific agonists potently inhibited cell proliferation rates while anti-estrogenic therapies were ineffective. CONCLUSIONS: Using a validated antibody, we have confirmed that nuclear ERß1 expression is commonly present in breast cancer and is prognostic in tamoxifen-treated patients. Using multiple breast cancer cell lines, ERß appears to be a novel therapeutic target. However, the efficacy of SERMs and ERß-specific agonists differ as a function of ERα expression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Antagonistas de Estrógenos/farmacología , Receptor beta de Estrógeno/metabolismo , Tamoxifeno/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/genética , Femenino , Humanos , Células MCF-7 , Persona de Mediana Edad
16.
Brain ; 136(Pt 4): 1274-87, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23408111

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARγ) is emerging as a major regulator in neurological diseases. However, the role of (PPARγ) and its co-regulators in cerebrovascular endothelial dysfunction after stroke is unclear. Here, we have demonstrated that (PPARγ) activation by pioglitazone significantly inhibited both oxygen-glucose deprivation-induced cerebral vascular endothelial cell death and middle cerebral artery occlusion-triggered cerebrovascular damage. Consistent with this finding, selective (PPARγ) genetic deletion in vascular endothelial cells resulted in increased cerebrovascular permeability and brain infarction in mice after focal ischaemia. Moreover, we screened for (PPARγ) co-regulators using a genome-wide and high-throughput co-activation system and revealed KLF11 as a novel (PPARγ) co-regulator, which interacted with (PPARγ) and regulated its function in mouse cerebral vascular endothelial cell cultures. Interestingly, KLF11 was also found as a direct transcriptional target of (PPARγ). Furthermore, KLF11 genetic deficiency effectively abolished pioglitazone cytoprotection in mouse cerebral vascular endothelial cell cultures after oxygen-glucose deprivation, as well as pioglitazone-mediated cerebrovascular protection in a mouse middle cerebral artery occlusion model. Mechanistically, we demonstrated that KLF11 enhanced (PPARγ) transcriptional suppression of the pro-apoptotic microRNA-15a (miR-15a) gene, resulting in endothelial protection in cerebral vascular endothelial cell cultures and cerebral microvasculature after ischaemic stimuli. Taken together, our data demonstrate that recruitment of KLF11 as a novel (PPARγ) co-regulator plays a critical role in the cerebrovascular protection after ischaemic insults. It is anticipated that elucidating the coordinated actions of KLF11 and (PPARγ) will provide new insights into understanding the molecular mechanisms underlying (PPARγ) function in the cerebral vasculature and help to develop a novel therapeutic strategy for the treatment of stroke.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Células Endoteliales/metabolismo , PPAR gamma/metabolismo , Proteínas Represoras/fisiología , Accidente Cerebrovascular/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Infarto Encefálico/metabolismo , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Ratones Transgénicos , MicroARNs/genética , PPAR gamma/deficiencia , PPAR gamma/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/fisiopatología
17.
Blood ; 118(24): 6450-60, 2011 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-21828131

RESUMEN

Emerging evidence demonstrates that proangiogenic cells (PACs) originate from the BM and are capable of being recruited to sites of ischemic injury where they contribute to neovascularization. We previously determined that among hematopoietic progenitor stem cells, common myeloid progenitors (CMPs) and granulocyte-macrophage progenitor cells (GMPs) differentiate into PACs and possess robust angiogenic activity under ischemic conditions. Herein, we report that a TGF-ß1-responsive Krüppel- like factor, KLF10, is strongly expressed in PACs derived from CMPs and GMPs, ∼ 60-fold higher than in progenitors lacking PAC markers. KLF10(-/-) mice present with marked defects in PAC differentiation, function, TGF-ß responsiveness, and impaired blood flow recovery after hindlimb ischemia, an effect rescued by wild-type PACs, but not KLF10(-/-) PACs. Overexpression studies revealed that KLF10 could rescue PAC formation from TGF-ß1(+/-) CMPs and GMPs. Mechanistically, KLF10 targets the VEGFR2 promoter in PACs which may underlie the observed effects. These findings may be clinically relevant because KLF10 expression was also found to be significantly reduced in PACs from patients with peripheral artery disease. Collectively, these observations identify TGF-ß1 signaling and KLF10 as key regulators of functional PACs derived from CMPs and GMPs and may provide a therapeutic target during cardiovascular ischemic states.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Diferenciación Celular , Factores de Transcripción de la Respuesta de Crecimiento Precoz/fisiología , Factores de Transcripción de Tipo Kruppel/fisiología , Neovascularización Fisiológica , Transducción de Señal , Factor de Crecimiento Transformador beta1/fisiología , Animales , Proteínas de Unión al ADN/genética , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Regulación de la Expresión Génica , Células Progenitoras de Granulocitos y Macrófagos/citología , Células Progenitoras de Granulocitos y Macrófagos/fisiología , Miembro Posterior , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/fisiología , Enfermedad Arterial Periférica/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Flujo Sanguíneo Regional , Factor de Crecimiento Transformador beta1/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 32(12): 2981-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23042817

RESUMEN

OBJECTIVE: Endothelial cell (EC) inflammatory status is critical to many vascular diseases. Emerging data demonstrate that mutations of Krüppel-like factor-11 (KLF11), a gene coding maturity-onset diabetes mellitus of the young type 7 (MODY7), contribute to the development of neonatal diabetes mellitus. However, the function of KLF11 in the cardiovascular system still remains to be uncovered. In this study, we aimed to investigate the role of KLF11 in vascular endothelial inflammation. METHODS AND RESULTS: KLF11 is highly expressed in vascular ECs and induced by proinflammatory stimuli. Adenovirus-mediated KLF11 overexpression inhibits expression of tumor necrosis factors-α-induced adhesion molecules. Moreover, small interfering RNA-mediated KLF11 knockdown augments the proinflammatory status in ECs. KLF11 inhibits promoter activity of adhesion molecules induced by tumor necrosis factor-α and nuclear factor-κB p65 overexpression. Mechanistically, KLF11 potently inhibits nuclear factor-κB signaling pathway via physical interaction with p65. Furthermore, KLF11 knockdown results in increased binding of p65 to vascular cell adhesion molecule-1 and E-selectin promoters. At the whole organism level, KLF11(-/-) mice exhibit a significant increase in leukocyte recruitment to ECs after lipopolysaccharide administration. CONCLUSIONS: Taken together, our data demonstrate for the first time that KLF11 is a suppressor of EC inflammatory activation, suggesting that KLF11 constitutes a novel potential molecular target for inhibition of vascular inflammatory diseases.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/fisiología , Diabetes Mellitus/fisiopatología , Endotelio Vascular/fisiología , FN-kappa B/fisiología , Proteínas Represoras/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/fisiología , Animales , Proteínas Reguladoras de la Apoptosis , Línea Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Selectina E/metabolismo , Endotelio Vascular/citología , Humanos , Leucocitos/efectos de los fármacos , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Unión Proteica , ARN Interferente Pequeño/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , eIF-2 Quinasa/metabolismo
19.
J Immunol ; 186(10): 5638-47, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21471442

RESUMEN

Earlier, we demonstrated the essential role of Kruppel-like transcription factor, TIEG1, in TGF-ß-induced regulatory T cell (Treg) development. In this article, we demonstrate that IL-6, which promotes Th17 development, abrogated TIEG1 nuclear translocation and inhibited TGF-ß-induced Treg development. Tyrosine kinase Tyk2-mediated phosphorylation of TIEG1 at Tyr179 promoted noncanonical K-27-linked polyubiquitination, which inhibited TIEG1 nuclear translocation. To test the role of TIEG1-regulated Treg/Th17 development in antitumor immunity, we analyzed TRAMP-C2 tumor growth in TIEG1(-/-) mice. The defective Treg development and elevated Th17 response resulted in enhanced immune reactivity in the tumor and inhibition of TRAMP-C2 tumor growth in TIEG1(-/-) mice. Thus, our results uncovered a novel regulatory mechanism that modulates Tregs and may regulate tumor progression.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Traslado Adoptivo , Animales , Western Blotting , Núcleo Celular/metabolismo , Citometría de Flujo , Factores de Transcripción Forkhead/metabolismo , Inmunoprecipitación , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Reacción en Cadena de la Polimerasa , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , TYK2 Quinasa/metabolismo , Células Th17/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitinación
20.
Front Med (Lausanne) ; 10: 1047166, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36926316

RESUMEN

Triple negative breast cancer (TNBC) is an aggressive sub-type of the disease which accounts for a disproportionately high percentage of breast cancer morbidities and mortalities. For these reasons, a better understanding of TNBC biology is required and the development of novel therapeutic approaches are critically needed. Estrogen receptor beta (ERß) is a reported tumor suppressor that is expressed in approximately 20% of primary TNBC tumors, where it is associated with favorable prognostic features and patient outcomes. Previous studies have shown that ERß mediates the assembly of co-repressor complexes on DNA to inhibit the expression of multiple growth promoting genes and to suppress the ability of oncogenic transcription factors to drive cancer progression. To further elucidate the molecular mechanisms by which ERß elicits its anti-cancer effects, we developed MDA-MB-231 cells that inducibly express a mutant form of ERß incapable of directly binding DNA. We demonstrate that disruption of ERß's direct interaction with DNA abolishes its ability to regulate the expression of well characterized immediate response genes and renders it unable to suppress TNBC cell proliferation. Loss of DNA binding also diminishes the ability of ERß to suppress oncogenic NFκB signaling even though it still physically associates with NFκB and other critical co-factors. These findings enhance our understanding of how ERß functions in this disease and provide a model system that can be utilized to further investigate the mechanistic processes by which ERß elicits its anti-cancer effects.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA