Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Cell ; 49(1): 158-71, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23159735

RESUMEN

How the glucocorticoid receptor (GR) activates some genes while potently repressing others remains an open question. There are three current models for suppression: transrepression via GR tethering to AP-1/NF-κB sites, direct GR association with inhibitory elements (nGREs), and GR recruitment of the corepressor GRIP1. To gain insights into GR suppression, we used genomic analyses and genome-wide profiling of GR, p65, and c-Jun in LPS-stimulated macrophages. We show that GR mediates both activation and repression at tethered sites, GREs, and GRIP1-bound elements, indicating that motif classification is insufficient to predict regulatory polarity of GR binding. Interestingly, sites of GR repression utilize GRIP1's corepressor function and display reduced histone acetylation. Together, these findings suggest that while GR occupancy confers hormone responsiveness, the receptor itself may not participate in the regulatory effects. Furthermore, transcriptional outcome is not established by sequence but is influenced by epigenetic regulators, context, and other unrecognized regulatory determinants.


Asunto(s)
Epigénesis Genética , Genoma , Inflamación/genética , Receptores de Glucocorticoides/fisiología , Acetilación , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Mapeo Cromosómico , Análisis por Conglomerados , Secuencia de Consenso , Dexametasona/farmacología , Glucocorticoides/farmacología , Histonas/metabolismo , Inflamación/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta , Factores de Transcripción/genética , Transcriptoma
2.
Proc Natl Acad Sci U S A ; 115(51): 13069-13074, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30478046

RESUMEN

MicroRNAs (miRNAs) have been extensively reported to be associated with hematological malignancies. The loss of miR-15a/16-1 at chromosome 13q14 is a hallmark of most of human chronic lymphocytic leukemia (CLL). Deletion of murine miR-15a/16-1 and miR-15b/16-2 has been demonstrated to promote B cell malignancies. Here, we evaluate the biological role of miR-15/16 clusters, crossbreeding miR-15a/16-1 and miR-15b/16-2 knockout mice. Unexpectedly, the complete deletion of both clusters promoted myeloproliferative disorders in the majority of the mice by the age of 5 months with a penetrance of 70%. These mice showed a significant enlargement of spleen and abnormal swelling of lymph nodes. Flow cytometry characterization demonstrated an expanded CD11b/Gr-1 double-positive myeloid population both in spleen and in bone marrow. The transplantation of splenocytes harvested from double-KO mice into wild-type recipient mice resulted in the development of myeloproliferative disorders, as observed in the donors. In vivo, miR-15/16 cluster deletion up-regulated the expression of Cyclin D1, Cyclin D2, and Bcl-2. Taken together, our findings identify a driver oncogenic role for miR-15/16 cluster deletion in different leukocytic cell lineages.


Asunto(s)
Leucemia Mieloide Aguda/etiología , MicroARNs/fisiología , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Ciclinas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/metabolismo , Bazo/patología
3.
Proc Natl Acad Sci U S A ; 114(23): E4641-E4647, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28533390

RESUMEN

Epithelial growth factor-like 7 (EGFL7) is a protein that is secreted by endothelial cells and plays an important role in angiogenesis. Although EGFL7 is aberrantly overexpressed in solid tumors, its role in leukemia has not been evaluated. Here, we report that levels of both EGFL7 mRNA and EGFL7 protein are increased in blasts of patients with acute myeloid leukemia (AML) compared with normal bone marrow cells. High EGFL7 mRNA expression associates with lower complete remission rates, and shorter event-free and overall survival in older (age ≥60 y) and younger (age <60 y) patients with cytogenetically normal AML. We further show that AML blasts secrete EGFL7 protein and that higher levels of EGFL7 protein are found in the sera from AML patients than in sera from healthy controls. Treatment of patient AML blasts with recombinant EGFL7 in vitro leads to increases in leukemic blast cell growth and levels of phosphorylated AKT. EGFL7 blockade with an anti-EGFL7 antibody reduced the growth potential and viability of AML cells. Our findings demonstrate that increased EGFL7 expression and secretion is an autocrine mechanism supporting growth of leukemic blasts in patients with AML.


Asunto(s)
Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Proteínas Angiogénicas/antagonistas & inhibidores , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Proteínas de Unión al Calcio , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Supervivencia sin Enfermedad , Familia de Proteínas EGF , Factores de Crecimiento Endotelial/antagonistas & inhibidores , Femenino , Humanos , Leucemia Mieloide Aguda/terapia , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Pronóstico , Proteínas/metabolismo , Proteínas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Factores de Riesgo , Regulación hacia Arriba , Adulto Joven
4.
Genes Dev ; 24(24): 2760-5, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21106671

RESUMEN

In the macrophage, toll-like receptors (TLRs) are key sensors that trigger signaling cascades to activate inflammatory programs via the NF-κB gene network. However, the genomic network targeted by TLR/NF-κB activation and the molecular basis by which it is restrained to terminate activation and re-establish quiescence is poorly understood. Here, using chromatin immunoprecipitation sequencing (ChIP-seq), we define the NF-κB cistrome, which is comprised of 31,070 cis-acting binding sites responsive to lipopolysaccharide (LPS)-induced signaling. In addition, we demonstrate that the transcriptional repressor B-cell lymphoma 6 (Bcl-6) regulates nearly a third of the Tlr4-regulated transcriptome, and that 90% of the Bcl-6 cistrome is collapsed following Tlr4 activation. Bcl-6-deficient macrophages are acutely hypersensitive to LPS and, using comparative ChIP-seq analyses, we found that the Bcl-6 and NF-κB cistromes intersect, within nucleosomal distance, at nearly half of Bcl-6-binding sites in stimulated macrophages to promote opposing epigenetic modifications of the local chromatin. These results reveal a genomic strategy for controlling the innate immune response in which repressive and inductive cistromes establish a dynamic balance between macrophage quiescence and activation via epigenetically marked cis-regulatory elements.


Asunto(s)
Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Macrófagos/inmunología , FN-kappa B/genética , Animales , Sitios de Unión , Células Cultivadas , Epigénesis Genética , Lipopolisacáridos/farmacología , Ratones , Proteínas Proto-Oncogénicas c-bcl-6 , Receptor Toll-Like 4/genética
7.
Blood Adv ; 8(4): 947-958, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38181781

RESUMEN

ABSTRACT: Acute graft-versus-host disease (GVHD) is a major complication of allogeneic hematopoietic cell transplantation (allo-HCT). Using preclinical mouse models of disease, previous work in our laboratory has linked microRNA-155 (miR-155) to the development of acute GVHD. Transplantation of donor T cells from miR-155 host gene (MIR155HG) knockout mice prevented acute GVHD in multiple murine models of disease while maintaining critical graft-versus-leukemia (GVL) response, necessary for relapse prevention. In this study, we used clustered, regularly interspaced, short palindromic repeats (CRISPR)/Cas9 genome editing to delete miR-155 in primary T cells (MIR155HGΔexon3) from human donors, resulting in stable and sustained reduction in expression of miR-155. Using the xenogeneic model of acute GVHD, we show that NOD/SCID/IL2rγnull (NSG) mice receiving MIR155HGΔexon3 human T cells provide protection from lethal acute GVHD compared with mice that received human T cells with intact miR-155. MIR155HGΔexon3 human T cells persist in the recipients displaying decreased proliferation potential, reduced pathogenic T helper-1 cell population, and infiltration into GVHD target organs, such as the liver and skin. Importantly, MIR155HGΔexon3 human T cells retain GVL response significantly improving survival in an in vivo model of xeno-GVL. Altogether, we show that CRISPR/Cas9-mediated deletion of MIR155HG in primary human donor T cells is an innovative approach to generate allogeneic donor T cells that provide protection from lethal GVHD while maintaining robust antileukemic response.


Asunto(s)
Enfermedad Injerto contra Huésped , MicroARNs , Humanos , Ratones , Animales , Incidencia , Sistemas CRISPR-Cas , Ratones Endogámicos NOD , Ratones SCID , Enfermedad Injerto contra Huésped/prevención & control , Ratones Noqueados , MicroARNs/genética
8.
Exp Hematol ; 123: 28-33.e3, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37209901

RESUMEN

Mantle cell lymphoma (MCL) is an aggressive, noncurative, mature B-cell lymphoma, with a median overall survival of 6-7 years. This underlines a need for effective therapeutic strategies to treat MCL better. Epidermal growth factor-like 7 (EGFL7) is a protein secreted by endothelial cells shown to play a critical role in angiogenesis. Our laboratory has previously demonstrated that EGFL7 supports the growth of leukemic blasts in patients with acute myeloid leukemia (AML); however, its role in MCL has not been investigated yet. In this study, we report that EGFL7 messenger RNA (mRNA) is increased in the cells of patients with MCL compared with cells from healthy controls, and patients with high EGFL7 are associated with lower overall survival rates. Furthermore, EGFL7 is increased in the plasma of patients with MCL compared with the plasma from healthy controls. We further show that EGFL7 binds to epidermal growth factor receptor (EGFR) and activates AKT signaling pathway in MCL cells and that blocking EGFL7 in MCL in patient and cell lines decreases cell growth and increases apoptosis in vitro. Finally, anti-EGFL7 treatment inhibits tumor size and prolongs survival in a mouse model of MCL. In conclusion, our study reveals a role for EGFL7 in MCL cell proliferation and highlights EGFL7 inhibition as a promising new treatment for patients with MCL.


Asunto(s)
Linfoma de Células del Manto , Animales , Ratones , Línea Celular Tumoral , Familia de Proteínas EGF/metabolismo , Células Endoteliales/metabolismo , Linfoma de Células del Manto/metabolismo , Transducción de Señal , Humanos
9.
Blood Adv ; 6(7): 2403-2408, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-34654057

RESUMEN

Acute graft-versus-host disease (aGVHD) is the second most common cause of death after allogeneic hematopoietic stem cell transplantation (allo-HSCT), underscoring the need for novel therapies. Based on previous work that endothelial cell dysfunction is present in aGVHD and that epidermal growth factor-like domain 7 (EGFL7) plays a significant role in decreasing inflammation by repressing endothelial cell activation and T-cell migration, we hypothesized that increasing EGFL7 levels after allo-HSCT will diminish the severity of aGVHD. Here, we show that treatment with recombinant EGFL7 (rEGFL7) in 2 different murine models of aGVHD decreases aGVHD severity and improves survival in recipient mice after allogeneic transplantation with respect to controls without affecting graft-versus-leukemia effect. Furthermore, we showed that rEGFL7 treatment results in higher thymocytes, T, B, and dendritic cell counts in recipient mice after allo-HSCT. This study constitutes a proof of concept of the ability of rEGFL7 therapy to reduce GHVD severity and mortality after allo-HSCT.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Trasplante de Médula Ósea/efectos adversos , Células Endoteliales , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones , Trasplante Homólogo
10.
Nat Commun ; 11(1): 3702, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32710081

RESUMEN

Spinal cord injury (SCI) causes immune dysfunction, increasing the risk of infectious morbidity and mortality. Since bone marrow hematopoiesis is essential for proper immune function, we hypothesize that SCI disrupts bone marrow hematopoiesis. Indeed, SCI causes excessive proliferation of bone marrow hematopoietic stem and progenitor cells (HSPC), but these cells cannot leave the bone marrow, even after challenging the host with a potent inflammatory stimulus. Sequestration of HSPCs in bone marrow after SCI is linked to aberrant chemotactic signaling that can be reversed by post-injury injections of Plerixafor (AMD3100), a small molecule inhibitor of CXCR4. Even though Plerixafor liberates HSPCs and mature immune cells from bone marrow, competitive repopulation assays show that the intrinsic long-term functional capacity of HSPCs is still impaired in SCI mice. Together, our data suggest that SCI causes an acquired bone marrow failure syndrome that may contribute to chronic immune dysfunction.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/etiología , Médula Ósea/metabolismo , Traumatismos de la Médula Espinal/complicaciones , Animales , Bencilaminas , Médula Ósea/patología , Células de la Médula Ósea , Trastornos de Fallo de la Médula Ósea/patología , Proliferación Celular , Quimiocina CXCL12 , Ciclamas , Modelos Animales de Enfermedad , Femenino , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Receptores CXCR4/antagonistas & inhibidores , Transducción de Señal , Traumatismos de la Médula Espinal/inmunología
11.
Clin Cancer Res ; 26(3): 669-678, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31672772

RESUMEN

PURPOSE: EGF-like domain 7 (EGFL7) is a secreted protein and recently has been shown to play an important role in acute myeloid leukemia (AML); however, the underlying mechanism by which EGFL7 promotes leukemogenesis is largely unknown. EXPERIMENTAL DESIGN: Using an antibody interaction array, we measured the ability of EGFL7 to bind directly approximately 400 proteins expressed by primary AML blasts. Primary patient samples were stimulated in vitro with recombinant EGFL7 (rEGFL7) or anti-EGFL7 blocking antibody to assess alterations in downstream signaling and the ability to effect blast differentiation and survival. We treated three independent AML models with anti-EGFL7 or IgG1 control to determine whether anti-EGFL7 could prolong survival in vivo. RESULTS: We found EGFL7 significantly binds several signaling proteins important for normal and malignant hematopoiesis including NOTCH. Stimulation of AML blasts with rEGFL7 reduced NOTCH intracellular domain and NOTCH target gene expression while treatment with an anti-EGFL7 blocking antibody resulted in reactivation of NOTCH signaling, increased differentiation, and apoptosis. Competitive ligand-binding assays showed rEGFL7 inhibits DELTA-like (DLL) 4-mediated NOTCH activation while anti-EGFL7 combined with DLL4 significantly increased NOTCH activation and induced apoptosis. Using three different AML mouse models, we demonstrated that in vivo treatment with anti-EGFL7 alone results in increased survival. CONCLUSIONS: Our data demonstrate that EGFL7 contributes to NOTCH silencing in AML by antagonizing canonical NOTCH ligand binding. Reactivation of NOTCH signaling in vivo using anti-EGFL7 results in prolonged survival of leukemic mice, supporting the use of EGFL7 as a novel therapeutic target in AML.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Proteínas de Unión al Calcio/metabolismo , Familia de Proteínas EGF/metabolismo , Leucemia Mieloide Aguda/patología , Receptores Notch/antagonistas & inhibidores , Animales , Apoptosis , Proteínas de Unión al Calcio/genética , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Familia de Proteínas EGF/genética , Femenino , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Receptores Notch/metabolismo , Transducción de Señal
14.
Leukemia ; 33(9): 2169-2182, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30858548

RESUMEN

In acute myeloid leukemia (AML), novel therapies are needed to target not only the rapidly dividing AML blasts but also the distinct population of leukemia stem cells (LSCs), which have abnormal self-renewal capacity and increased chemotherapy resistance. Elucidation of the expression and function of deregulated genes in LSCs is critical to specifically target LSCs and may consequently lead to improving outcomes of AML patients. Here, we correlated long non-coding RNA (lncRNA) expression profiles obtained from two RNA-seq datasets of 375 younger (aged <60 years) 76 older (≥60 years) adults with cytogenetically normal AML with a 'core enriched' (CE) gene expression signature (GES) associated with LSCs. We identified a LSC-specific signature of 111 lncRNAs that correlated strongly with the CE-GES. Among the top upregulated LSC-associated lncRNAs, we identified the lncRNA DANCR. Further experiments confirmed that DANCR is upregulated in functionally validated LSC-enriched populations. DANCR knock-down in LSCs resulted in decreased stem-cell renewal and quiescence. Furthermore, we showed that targeting Dancr in vivo using a primary murine model of AML (expressing both Mll partial tandem duplication/Flt3 internal tandem duplication) prolonged the survival of mice after serial transplantation. Our data suggest that LSCs have a distinct lncRNA signature with functional relevance and therapeutic potential.


Asunto(s)
Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Enfermedad Aguda , Animales , Autorrenovación de las Células/genética , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad
15.
Nat Commun ; 10(1): 5351, 2019 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-31767858

RESUMEN

Long non-coding RNAs (lncRNAs) are important regulatory molecules that are implicated in cellular physiology and pathology. In this work, we dissect the functional role of the HOXB-AS3 lncRNA in patients with NPM1-mutated (NPM1mut) acute myeloid leukemia (AML). We show that HOXB-AS3 regulates the proliferative capacity of NPM1mut AML blasts in vitro and in vivo. HOXB-AS3 is shown to interact with the ErbB3-binding protein 1 (EBP1) and guide EBP1 to the ribosomal DNA locus. Via this mechanism, HOXB-AS3 regulates ribosomal RNA transcription and de novo protein synthesis. We propose that in the context of NPM1 mutations, HOXB-AS3 overexpression acts as a compensatory mechanism, which allows adequate protein production in leukemic blasts.


Asunto(s)
Leucemia Mieloide/genética , Mutación , Proteínas Nucleares/genética , ARN Largo no Codificante/genética , ARN Ribosómico/genética , Transcripción Genética , Enfermedad Aguda , Animales , Línea Celular Tumoral , Proliferación Celular , Células HEK293 , Humanos , Células K562 , Leucemia Mieloide/patología , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Nucleofosmina , Biosíntesis de Proteínas/genética , Células THP-1 , Trasplante Heterólogo
16.
Nat Commun ; 4: 2354, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23965929

RESUMEN

Mitsugumin 53 (MG53) negatively regulates skeletal myogenesis by targeting insulin receptor substrate 1 (IRS-1). Here, we show that MG53 is an ubiquitin E3 ligase that induces IRS-1 ubiquitination with the help of an E2-conjugating enzyme, UBE2H. Molecular manipulations that disrupt the E3-ligase function of MG53 abolish IRS-1 ubiquitination and enhance skeletal myogenesis. Skeletal muscles derived from the MG53-/- mice show an elevated IRS-1 level with enhanced insulin signalling, which protects the MG53-/- mice from developing insulin resistance when challenged with a high-fat/high-sucrose diet. Muscle samples derived from human diabetic patients and mice with insulin resistance show normal expression of MG53, indicating that altered MG53 expression does not serve as a causative factor for the development of metabolic disorders. Thus, therapeutic interventions that target the interaction between MG53 and IRS-1 may be a novel approach for the treatment of metabolic diseases that are associated with insulin resistance.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Insulina/metabolismo , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , Animales , Proteínas Portadoras/genética , Diferenciación Celular , Línea Celular , Diabetes Mellitus/metabolismo , Dieta Alta en Grasa , Prueba de Tolerancia a la Glucosa , Proteínas Sustrato del Receptor de Insulina/genética , Resistencia a la Insulina , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación
17.
Cell Metab ; 15(4): 554-62, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22465074

RESUMEN

Chronic inflammation is a hallmark of atherosclerosis, but its transcriptional underpinnings are poorly understood. We show that the transcriptional repressor Bcl6 is an anti-inflammatory regulator whose loss in bone marrow of Ldlr(-/-) mice results in severe atherosclerosis and xanthomatous tendonitis, a virtually pathognomonic complication in patients with familial hypercholesterolemia. Disruption of the interaction between Bcl6 and SMRT or NCoR with a peptide inhibitor in vitro recapitulated atherogenic gene changes in mice transplanted with Bcl6-deficient bone marrow, pointing to these cofactors as key mediators of Bcl6 inflammatory suppression. Using ChIP-seq, we reveal the SMRT and NCoR corepressor cistromes, each consisting of over 30,000 binding sites with a nearly 50% overlap. While the complete cistromes identify a diversity of signaling pathways, the Bcl6-bound subcistromes for each corepressor are highly enriched for NF-κB-driven inflammatory and tissue remodeling genes. These results reveal that Bcl6-SMRT/NCoR complexes constrain immune responses and contribute to the prevention of atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Aterosclerosis/patología , Proteínas de Unión al ADN/metabolismo , Inflamación/genética , Co-Represor 2 de Receptor Nuclear/metabolismo , Animales , Aterosclerosis/complicaciones , Secuencia de Bases , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Colesterol/metabolismo , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/complicaciones , Inflamación/patología , Lipoproteínas LDL/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Co-Represor 2 de Receptor Nuclear/genética , Proteínas Proto-Oncogénicas c-bcl-6 , Tendinopatía/patología
18.
Cell Metab ; 13(3): 283-93, 2011 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-21356518

RESUMEN

How type I skeletal muscle inherently maintains high oxidative and vascular capacity in the absence of exercise is unclear. We show that nuclear receptor ERRγ is highly expressed in type I muscle and, when transgenically expressed in anaerobic type II muscles (ERRGO mice), dually induces metabolic and vascular transformation in the absence of exercise. ERRGO mice show increased expression of genes promoting fat metabolism, mitochondrial respiration, and type I fiber specification. Muscles in ERRGO mice also display an activated angiogenic program marked by myofibrillar induction and secretion of proangiogenic factors, neovascularization, and a 100% increase in running endurance. Surprisingly, the induction of type I muscle properties by ERRγ does not involve PGC-1α. Instead, ERRγ genetically activates the energy sensor AMPK in mediating the metabovascular changes in ERRGO mice. Therefore, ERRγ represents a previously unrecognized determinant that specifies intrinsic vascular and oxidative metabolic features that distinguish type I from type II muscle.


Asunto(s)
Músculo Esquelético/metabolismo , Receptores de Estrógenos/metabolismo , Transactivadores/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Metabolismo de los Lípidos/genética , Ratones , Ratones Transgénicos , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/fisiología , Neovascularización Fisiológica , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Condicionamiento Físico Animal , Proteínas Quinasas/metabolismo , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA