Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38746313

RESUMEN

Schwann cells are vital to development and maintenance of the peripheral nervous system and their dysfunction has been implicated in a range of neurological and neoplastic disorders, including NF2 -related schwannomatosis. We have developed a novel human induced pluripotent stem cell (hiPSC) model for the study of Schwann cell differentiation in health and disease. We performed transcriptomic, immunofluorescence, and morphological analysis of hiPSC derived Schwann cell precursors (SPCs) and terminally differentiated Schwann-like cells (SLCs) representing distinct stages of development. To further validate our findings, we performed integrated, cross-species analyses across multiple external datasets at bulk and single cell resolution. Our hiPSC model of Schwann cell development shared overlapping gene expression signatures with human amniotic mesenchymal stem cell (hAMSCs) derived SLCs and in vivo mouse models, but also revealed unique features that may reflect species-specific aspects of Schwann cell biology. Moreover, we have identified gene co-expression modules that are dynamically regulated during hiPSC to SLC differentiation associated with ear and neural development, cell fate determination, the NF2 gene, and extracellular matrix (ECM) organization. By cross-referencing results between multiple datasets and analyses, we have identified potential new genes that are related to NF2 for further study including: ANXA1, CDH6, COL1A1, COL8A1, MFAP5, IGFBP5, FGF1, AHNAK, CDKN2B, LOX, CAV1 , and CAV2 . Our hiPSC model further provides a tractable platform for studying Schwann cell development in the context of human disease.

2.
Proc Natl Acad Sci U S A ; 119(15): e2121098119, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35377803

RESUMEN

The pathogenesis of lung fibrosis involves hyperactivation of innate and adaptive immune pathways that release inflammatory cytokines and growth factors such as tumor growth factor (TGF)ß1 and induce aberrant extracellular matrix protein production. During the genesis of pulmonary fibrosis, resident alveolar macrophages are replaced by a population of newly arrived monocyte-derived interstitial macrophages that subsequently transition into alveolar macrophages (Mo-AMs). These transitioning cells initiate fibrosis by releasing profibrotic cytokines and remodeling the matrix. Here, we describe a strategy for leveraging the up-regulation of the mannose receptor CD206 in interstitial macrophages and Mo-AM to treat lung fibrosis. We engineered mannosylated albumin nanoparticles, which were found to be internalized by fibrogenic CD206+ monocyte derived macrophages (Mo-Macs). Mannosylated albumin nanoparticles incorporating TGFß1 small-interfering RNA (siRNA) targeted the profibrotic subpopulation of CD206+ macrophages and prevented lung fibrosis. The findings point to the potential utility of mannosylated albumin nanoparticles in delivering TGFß-siRNA into CD206+ profibrotic macrophages as an antilung fibrosis strategy.


Asunto(s)
Linfotoxina-alfa , Macrófagos Alveolares , Nanopartículas , Fibrosis Pulmonar , ARN Interferente Pequeño , Animales , Bleomicina/farmacología , Modelos Animales de Enfermedad , Linfotoxina-alfa/genética , Macrófagos Alveolares/inmunología , Receptor de Manosa , Ratones , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/terapia , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética
3.
iScience ; 24(11): 103348, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34816102

RESUMEN

The apoptosis inducing receptor CD95/Fas has multiple tumorigenic activities. In different genetically engineered mouse models tumor-expressed CD95 was shown to be critical for cell growth. Using a combination of immune-deficient and immune-competent mouse models, we now establish that loss of CD95 in metastatic triple negative breast cancer (TNBC) cells prevents tumor growth by modulating the immune landscape. CD95-deficient, but not wild-type, tumors barely grow in an immune-competent environment and show an increase in immune infiltrates into the tumor. This growth reduction is caused by infiltrating NK cells and does not involve T cells or macrophages. In contrast, in immune compromised mice CD95 k.o. cells are not growth inhibited, but they fail to form metastases. In summary, we demonstrate that in addition to its tumor and metastasis promoting activities, CD95 expression by tumor cells can exert immune suppressive activities on NK cells, providing a new target for immune therapy.

4.
Sci Rep ; 10(1): 1310, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31992798

RESUMEN

CD95/Fas is an apoptosis inducing death receptor. However, it also has multiple nonapoptotic activities that are tumorigenic. Chronic stimulation of CD95 on breast cancer cells can increase their cancer initiating capacity through activation of a type I interferon (IFN-I)/STAT1 pathway when caspases are inhibited. We now show that this activity relies on the canonical components of the CD95 death-inducing signaling complex, FADD and caspase-8, and on the activation of NF-κB. We identified caspase-2 as the antagonistic caspase that downregulates IFN-I production. Once produced, IFN-Is bind to their receptors activating both STAT1 and STAT2 resulting in upregulation of the double stranded (ds)RNA sensor proteins RIG-I and MDA5, and a release of a subset of endogenous retroviruses. Thus, CD95 is part of a complex cell autonomous regulatory network that involves activation of innate immune components that drive cancer stemness and contribute to therapy resistance.


Asunto(s)
Neoplasias de la Mama/metabolismo , Interferón Tipo I/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Receptor fas/metabolismo , Neoplasias de la Mama/etiología , Neoplasias de la Mama/patología , Caspasa 8/metabolismo , Muerte Celular , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Femenino , Humanos , Inmunidad , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Células Madre Neoplásicas/patología , ARN Bicatenario/genética , ARN Bicatenario/inmunología
5.
J Cell Biochem ; 120(2): 2226-2235, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30277585

RESUMEN

Distal-less homeobox 3 (Dlx3), a member of the Dlx family of homeobox proteins, is a transcriptional activator of runt-related transcription factor 2 (Runx2) during osteogenic differentiation. It has been demonstrated that forced expression of Runx2 induces an osteogenic program and ectopic calcification in muscles. Therefore, it would be reasonable to predict that Dlx3 also affects myogenic differentiation. The relationship between Dlx3 and myogenesis, however, remains poorly understood. Therefore, in this study, the role and regulation of Dlx3 during myogenic differentiation were investigated. Expression level of Dlx3 was downregulated in human mesenchymal stem cells (MSCs), mouse MSCs, and C2C12 cells cultured in myogenic medium. Dlx3 level was inversely correlated with myogenic differentiation 1 and the muscle-specific microRNA, microRNA-133 (miR-133). The expression level of Runx2 was closely regulated by Dlx3 even under myogenic conditions. Overexpression of Dlx3 markedly downregulated expression levels of myogenic transcription factors and myotube formation in C2C12 cells, whereas Dlx3 knockdown enhanced myogenic differentiation. The Dlx3 3'-untranslated region (3'-UTR) has two potential binding sites for miR-133. Luciferase reporter assays demonstrated that Dlx3 is a direct target of miR-133a and miR-133b, and that the two target sites are redundantly active. Taken together, these results suggest that Dlx3 is a negative regulator of myogenic differentiation and that miR-133a and miR-133b enhance myogenic differentiation, partly through inhibition of Dlx3 expression via direct targeting of the Dlx3 3'-UTR.

6.
Elife ; 72018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30324908

RESUMEN

CD95/Fas ligand binds to the death receptor CD95 to induce apoptosis in sensitive cells. We previously reported that CD95L mRNA is enriched in sequences that, when converted to si/shRNAs, kill all cancer cells by targeting critical survival genes (Putzbach et al., 2017). We now report expression of full-length CD95L mRNA itself is highly toxic to cells and induces a similar form of cell death. We demonstrate that small (s)RNAs derived from CD95L are loaded into the RNA induced silencing complex (RISC) which is required for the toxicity and processing of CD95L mRNA into sRNAs is independent of both Dicer and Drosha. We provide evidence that in addition to the CD95L transgene a number of endogenous protein coding genes involved in regulating protein translation, particularly under low miRNA conditions, can be processed to sRNAs and loaded into the RISC suggesting a new level of cell fate regulation involving RNAi.


Asunto(s)
Proteína Ligando Fas/genética , ARN Mensajero/genética , ARN Mensajero/orina , Complejo Silenciador Inducido por ARN/genética , Receptor fas/genética , Apoptosis/genética , Proteína Ligando Fas/química , Regulación de la Expresión Génica/genética , Células HCT116 , Humanos , MicroARNs/química , MicroARNs/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Complejo Silenciador Inducido por ARN/química , Receptor fas/química
7.
Int J Mol Sci ; 19(10)2018 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-30322210

RESUMEN

Distal-less homeobox 5 (Dlx5) is a negative regulator of adipogenesis. Dlx5 expression is decreased by adipogenic stimuli, but the mechanisms of Dlx5 downregulation by adipogenic stimuli have not yet been determined. Here, we tested the impact of cAMP/PKA (protein kinase A) signaling induced by 3-isobutyl-1 methyl xanthine (IBMX), forskolin, and 8-CPT-cAMP on the expression of Dlx5 in 3T3-L1 preadipocytes. Significant downregulation of Dlx5 mRNA expression and protein production levels were observed via cAMP/PKA-dependent signaling. Forced expression of cAMP-responsive element-binding protein (CREB) and CCAAT/enhancer-binding protein ß (C/EBPß) was sufficient for downregulation of Dlx5 expression and revealed that CREB functions upstream of C/EBPß. In addition, C/EBPß knockdown by siRNA rescued Dlx5 expression in IBMX-treated 3T3-L1 preadipocytes. Luciferase assays using a Dlx5-luc-2935 reporter construct demonstrated the requirement of the Dlx5 promoter region, ranging from -774 to -95 bp that contains two putative C/EBPß binding elements (site-1: -517 to -510 bp and site-2: -164 to -157 bp), in the suppression of Dlx5 transcription. Consequently, chromatin immunoprecipitation analysis confirmed the importance of site-1, but not site-2, in C/EBPß binding and transcriptional suppression of Dlx5. In conclusion, we elucidated the underling mechanism of Dlx5 downregulation in IBMX-induced adipogenesis. IBMX activated cAMP/PKA/CREB signaling and subsequently upregulated C/EBPß, which binds to the Dlx5 promoter to suppress Dlx5 transcription.


Asunto(s)
Adipocitos/citología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Homeodominio/genética , 1-Metil-3-Isobutilxantina/farmacología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis , Animales , Sitios de Unión , Proteína beta Potenciadora de Unión a CCAAT/genética , Colforsina/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Tionucleótidos/farmacología
8.
Cell Rep ; 18(10): 2373-2386, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28273453

RESUMEN

Stimulation of CD95/Fas drives and maintains cancer stem cells (CSCs). We now report that this involves activation of signal transducer and activator of transcription 1 (STAT1) and induction of STAT1-regulated genes and that this process is inhibited by active caspases. STAT1 is enriched in CSCs in cancer cell lines, patient-derived human breast cancer, and CD95high-expressing glioblastoma neurospheres. CD95 stimulation of cancer cells induced secretion of type I interferons (IFNs) that bind to type I IFN receptors, resulting in activation of Janus-activated kinases, activation of STAT1, and induction of a number of STAT1-regulated genes that are part of a gene signature recently linked to therapy resistance in five primary human cancers. Consequently, we identified type I IFNs as drivers of cancer stemness. Knockdown or knockout of STAT1 resulted in a strongly reduced ability of CD95L or type I IFN to increase cancer stemness. This identifies STAT1 as a key regulator of the CSC-inducing activity of CD95.


Asunto(s)
Interferón Tipo I/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Factor de Transcripción STAT1/metabolismo , Receptor fas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Caspasa 3/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Marcaje Isotópico , Fosforilación , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Regulación hacia Arriba
9.
J Cell Biochem ; 116(5): 730-42, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25424317

RESUMEN

MicroRNAs are novel key regulators of cellular differentiation. Dlx transcription factors play an important role in osteoblast differentiation, and Dlx5 and Dlx2 are known targets of miR-124. Therefore, in the present study, we investigated the regulatory effects of miR-124 on the osteogenic differentiation and in vivo bone formation of mesenchymal stem cells (MSCs). During osteogenic induction by BMP2, the expression levels of miR-124 were inversely correlated with those of osteogenic differentiation marker genes in human and mouse bone marrow-derived MSCs, MC3T3-E1 cells and C2C12 cells. The overexpression of a miR-124 mimic significantly decreased the expression levels of Dlx5, Dlx3, and Dlx2, whereas the silencing of miR-124 with hairpin inhibitors significantly increased the expression of these Dlx genes. Luciferase reporter assays demonstrated that miR-124 directly targets the 3'UTRs of Dlx3, Dlx5, and Dlx2. The overexpression of a miR-124 mimic suppressed the osteogenic marker gene expression levels, alkaline phosphatase activity and matrix mineralization, which were all significantly increased by the overexpression of a miR-124 inhibitor. When ectopic bone formation was induced by the subcutaneous transplantation of human bone marrow-derived MSCs in nude mice, MSCs overexpressing a miR-124 inhibitor significantly enhanced woven bone formation compared with control MSCs. However, MSCs overexpressing a miR-124 mimic exhibited increased adipocyte differentiation at the expense of ectopic bone formation. These results suggest that miR-124 is a negative regulator of osteogenic differentiation and in vivo bone formation and that the targeting of Dlx5, Dlx3, and Dlx2 genes partly contributes to this inhibitory effect exerted by miR-124.


Asunto(s)
Huesos/metabolismo , Diferenciación Celular , MicroARNs/genética , MicroARNs/metabolismo , Osteoblastos/citología , Regiones no Traducidas 3' , Adipocitos/citología , Adipocitos/metabolismo , Animales , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Huesos/citología , Células Cultivadas , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Ratones Desnudos , Osteoblastos/metabolismo , Osteogénesis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
J Cell Biochem ; 115(9): 1572-81, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24733577

RESUMEN

MicroRNAs (miRNAs), including miR-1, miR-133, and miR-206, play a crucial role in muscle development by regulating muscle cell proliferation and differentiation. The aim of the present study was to define the effect of miR-124 on myogenic differentiation of mesenchymal stem cells (MSCs). The expression level of miR-124 in skeletal muscles was much lower than those in primary cultured bone marrow-derived MSCs and the bone, fat and brain tissues obtained from C57BL/6 mice. Myogenic stimuli significantly decreased the expression levels of miR-124 in mouse bone marrow-derived MSCs and C2C12 cells. Forced expression of miR-124 suppressed the expression of myogenic marker genes such as Myf5, Myod1, myogenin and myosin heavy chain and multinucleated myotube formation. Blockade of endogenous miR-124 with a hairpin inhibitor enhanced myogenic marker gene expression and myotube formation. During myogenic differentiation of MSCs and C2C12 cells, the levels of Dlx5, a known target of miR-124, were inversely regulated with those of miR-124. Furthermore, overexpression of Dlx5 increased myogenic differentiation, whereas knockdown of Dlx5 using siRNA inhibited myogenesis in C2C12 cells. These results suggest that miR-124 is a negative regulator of myogenic differentiation of MSCs and that upregulation of Dlx5 accompanied with downregulation of miR-124 by myogenic stimuli is necessary for the proper progression of myogenic differentiation.


Asunto(s)
Fémur/citología , Proteínas de Homeodominio/genética , Células Madre Mesenquimatosas/citología , MicroARNs/genética , Desarrollo de Músculos , Tibia/citología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL
11.
BMB Rep ; 47(9): 506-11, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24393528

RESUMEN

We investigated the effects of high calorie and low calorie diets on skeletal integrity, and whether ß-adrenergic blockade (BB) attenuates bone loss induced by dietary calorie alteration. Male 6-week-old C57BL/6 mice were assigned to either an ad-lib fed control diet (CON), a high calorie diet (HIGH), or a low calorie diet (LOW) group. In each diet group, mice were treated with either vehicle (VEH) or propranolol, a ß-adrenergic antagonist. Over 12-weeks, ß-blockade mitigated body weight and fat mass increases induced by the high calorie diet. Femoral trabecular bone mineral density and the expression levels of osteogenic marker genes in bone marrow cells were reduced in HIGHVEH and LOWVEH mice, and BB significantly attenuated this decline only in HIGH mice. In summary, the magnitude of bone loss induced by low calorie diet was greater than that caused by high calorie diet in growing mice, and ß-blockade mitigated high calorie diet-induced bone loss.


Asunto(s)
Antagonistas Adrenérgicos/farmacología , Peso Corporal/efectos de los fármacos , Dieta , Propranolol/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Resorción Ósea/metabolismo , Resorción Ósea/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Osteogénesis/efectos de los fármacos , Receptores Adrenérgicos beta/química , Receptores Adrenérgicos beta/metabolismo , Tibia/química , Tibia/metabolismo
12.
J Cell Physiol ; 229(5): 640-50, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24446199

RESUMEN

Sclerostin decreases bone mass by antagonizing the Wnt signaling pathway. We examined whether obesity-induced bone loss is associated with the expression of sclerostin. Five-week-old male mice were assigned to one of two groups (n = 10 each) and fed either a control diet (10% kcal from fat; CON) or a high-fat diet (60% kcal from fat; HF) for 12 weeks. Thex final body weight and whole body fat mass of the HF mice were higher than those of the CON mice. The distal femur cancellous bone mineral density and bone formation rate was lower in HF mice than in CON mice. The percent erosion surface was higher in the HF mice than the CON mice. The serum levels and femoral osteocytic protein expression levels of tumor necrosis factor-α (TNF-α) were significantly higher in HF mice than in CON mice. Sclerostin mRNA levels and osteocytic sclerostin protein levels in femoral cortex were also higher in HF mice than in CON mice. Sclerostin expression in MLO-Y4 osteocytes increased with TNF-α treatment, and TNF-α-induced sclerostin expression was blocked by the inhibition of NF-κB activation. Chromatin immunoprecipitation and a luciferase reporter assay demonstrated that NF-κB directly binds to the NF-κB binding elements on the mouse sost promoter and stimulates sclerostin expression. These results support a model in which, in the context of obesity or other inflammatory diseases that increase the production of TNF-α, TNF-α upregulates the expression of sclerostin through NF-κB signaling pathway, thus contributing to bone loss.


Asunto(s)
Grasas de la Dieta/efectos adversos , Glicoproteínas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales , Animales , Composición Corporal/efectos de los fármacos , Densidad Ósea/efectos de los fármacos , Línea Celular , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Obesidad , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética
13.
Exp Cell Res ; 319(14): 2125-34, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23648570

RESUMEN

Distal-less homeobox 5 (Dlx5) is a pro-osteogenic but anti-adipogenic transcription factor that regulates lineage commitment in mesenchymal stem cells. Although the expression of Dlx5 is known to be decreased by adipogenic stimuli, the mechanism of Dlx5 down-regulation has not yet been clarified. MicroRNAs (miRNAs) are small regulatory RNAs that post-transcriptionally regulate many biological functions, including cell differentiation. In this study, we examined whether miRNAs are involved in down-regulation of Dlx5 following adipogenic stimuli. We screened candidate miRNAs that have a direct target site in the Dlx5 3'UTR using computational prediction programs, selected seven miRNA candidates with the highest binding score and observed their expression levels in 3T3-L1 murine pre-adipocytes. Among the miRNAs examined, only miR-124 was significantly up-regulated by 24-h incubation in adipogenic medium. Among the four components of adipogenic stimuli (1-methy-3-isobutyl xanthine, insulin, indomethacin and dexamethasone), insulin exhibited the highest stimulatory effect on miR-124 expression. Insulin significantly increased the expression of miR-124 precursors including pri-miR-124-1, pri-miR124-2 and pri-miR-124-3. LY294002, an inhibitor of phosphatidylinositol-3-kinase, prevented the regulatory effect of insulin on the expression levels of miR-124 and Dlx5. Over-expression of a miR-124 mimic decreased the expression of Dlx5 while increasing adipogenic differentiation in 3T3-L1 cells. Blocking miR-124 with anti-miR-124, a hairpin inhibitor of miR-124, increased the expression level of Dlx5 and suppressed adipogenic differentiation. When reporter assays were performed with a reporter construct containing the Dlx5 3'UTR sequence downstream of a luciferase gene, miR-124 mimic suppressed, but anti-miR-124 enhanced, luciferase activity in an miR-124 binding site-dependent manner. These results suggest that insulin-induced miR-124 plays a pivotal role in post-transcriptional regulation of Dlx5 during adipogenic differentiation and that miR-124 exerts pro-adipogenic effects by targeting Dlx5, at least in part.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Insulina/metabolismo , MicroARNs/metabolismo , Regulación hacia Arriba , Regiones no Traducidas 3' , Células 3T3 , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis , Animales , Sitios de Unión , Cromonas/farmacología , Proteínas de Homeodominio/genética , Ratones , MicroARNs/genética , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN Interferente Pequeño , Transcripción Genética
14.
J Cell Physiol ; 228(5): 1076-86, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23042144

RESUMEN

Smad ubiquitination regulatory factor 1 (Smurf1) is an E3 ubiquitin ligase that negatively regulates osteoblast differentiation. Although tumor necrosis factor-α (TNF-α) has been shown to increase Smurf1 expression, the details of the regulatory mechanisms remain unclear. Here, we investigated the molecular mechanism by which TNF-α stimulates Smurf1 expression in C2C12 and primary cultured mouse calvarial cells. TNF-α treatment rapidly induced the activation of NF-κB and MAPKs. Smurf1 induction by TNF-α was blocked by the inhibition of JNK or ERK, while the inhibition of NF-κB and p38 MAPK had no effect on Smurf1 induction. TNF-α treatment or c-Jun overexpression enhanced the activity of a luciferase reporter that contained a 2.7 kb mouse Smurf1 promoter sequence. Site-directed mutagenesis of the Smurf1 reporter and chromatin immunoprecipitation analysis demonstrated that the activating protein-1 (AP-1) binding motif at -922 bp on the mouse Smurf1 promoter mediated TNF-α/JNK/AP-1-stimulated Smurf1 transcription. Interestingly, Smurf1 expression was not observed in Runx2-null mouse calvarial cells. When Runx2 was ectopically expressed in these cells, the basal and TNF-α-induced expression of Smurf1 was restored. Overexpression of Runx2 transactivated the Smurf1 promoter in a dose-dependent manner. Reporter and chromatin immunoprecipitation assays demonstrated that the Runx2-binding motif at -202 bp functioned in Runx2-mediated Smurf1 expression. ERK activation by TNF-α treatment or constitutively active MEK1 overexpression increased Smurf1 expression in a Runx2-dependent manner. These results suggest that the JNK/AP-1 and ERK/Runx2 signaling pathways mediate TNF-α-dependent Smurf1 transcription.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal , Factor de Transcripción AP-1 , Transcripción Genética , Factor de Necrosis Tumoral alfa , Ubiquitina-Proteína Ligasas , Animales , Diferenciación Celular , Línea Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Fosforilación , Transducción de Señal , Cráneo/citología , Cráneo/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas Quinasas p38 Activadas por Mitógenos/genética
15.
J Cell Physiol ; 227(11): 3603-12, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22307523

RESUMEN

Myeloid Elf-1 like factor (MEF) is one of the Ets transcription factors known to regulate cell proliferation and differentiation. A previous report has shown that osteoblast-specific MEF transgenic mice (Col1a1-MEF TG mice) have low bone mass but high bone marrow adiposity. In the present study, we explored a previously unappreciated mechanism whereby MEF promotes adipogenesis in bone marrow. An adipogenic colony-forming unit assay showed that bone marrow cells derived from Col1a1-MEF TG mice had a higher adipogenic differentiation potential compared to those from wild-type. The levels of adipogenic marker genes expression in 3T3L1 cells were higher when co-cultured with Col1a1-MEF TG bone marrow cells than with wild-type cells. MC3T3-E1 preosteoblasts transfected with MEF secreted higher levels of 15-deoxy-delta (12, 14)-prostaglandin J(2), a potent endogenous ligand of peroxisome proliferator-activated receptor γ (PPARγ), under adipogenic conditions. MEF overexpression increased the adipogenic marker genes expression including PPARγ and lipid droplet accumulation in MC3T3-E1 preosteoblasts and 3T3L1 preadipocytes. Endogenous MEF expression levels increased as adipocyte differentiation proceeded. Knockdown of MEF by siRNA suppressed expression levels of adipogenic marker genes including PPARγ. MEF directly bound to the MEF binding element on the mouse PPARγ promoter, transactivating promoter activity. Immunohistochemical staining of tibia sections demonstrated that bone lining cells and bone marrow cells express higher levels of PPARγ protein in Col1a1-MEF TG mice than in wild-type mice. These results suggest that MEF transactivates PPARγ expression, which, in turn, enhances adipogenic differentiation. Furthermore, MEF overexpressing osteoblasts secrete higher levels of adipogenic factors, creating a marrow microenvironment that favors adipogenesis.


Asunto(s)
Adipogénesis , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , PPAR gamma , Factores de Transcripción/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Microambiente Celular/fisiología , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Osteoblastos/citología , Osteoblastos/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Prostaglandina D2/análogos & derivados , Prostaglandina D2/metabolismo , ARN Interferente Pequeño , Factores de Transcripción/genética
16.
Mol Cells ; 31(6): 573-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21533547

RESUMEN

During orthodontic tooth movement, local hypoxia and enhanced osteoclastogenesis are observed in the compression side of periodontal tissues. The receptor activator of nuclear factor-κB ligand (RANKL) is an osteoblast/stromal cell-derived factor that is essential for osteoclastogenesis. In this study, we examined the effect of hypoxia on RANKL expression in human periodontal ligament fibroblasts (PDLFs) to investigate the relationship between local hypoxia and enhanced osteoclastogenesis in the compression side of periodontal tissues. Hypoxia significantly enhanced the levels of RANKL mRNA and protein as well as hypoxia inducible factor-1α (HIF-1α) protein in PDLFs. Constitutively active HIF-1α alone significantly increased the levels of RANKL expression in PDLFs under normoxic conditions, whereas dominant negative HIF-1α blocked hypoxia-induced RANKL expression. To investigate further whether HIF-1α directly regulates RANKL transcription, a luciferase reporter assay was performed using the reporter vector containing the RANKL promoter sequence. Exposure to hypoxia or overexpression of constitutively active HIF-1α significantly increased RANKL promoter activity, whereas dominant negative HIF-1α blocked hypoxia-induced RANKL promoter activity. Furthermore, mutations of putative HIF-1α binding elements in RANKL promoter prevented hypoxia-induced RANKL promoter activity. The results of chromatin immunoprecipitation showed that hypoxia or constitutively active HIF-1α increased the DNA binding of HIF-1α to RANKL promoter. These results suggest that HIF-1α mediates hypoxia-induced up-regulation of RANKL expression and that in compression side periodontal ligament, hypoxia enhances osteoclastogenesis, at least in part, via an increased RANKL expression in PDLFs.


Asunto(s)
Fibroblastos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ligamento Periodontal/citología , Ligando RANK/genética , Hipoxia de la Célula , Células Cultivadas , Inmunoprecipitación de Cromatina , Genes Reporteros , Humanos , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Ligamento Periodontal/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Ligando RANK/metabolismo , Transcripción Genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Biochem Biophys Res Commun ; 408(3): 399-404, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21514273

RESUMEN

Tumor necrosis factor-α (TNF-α) is known to suppress adipocyte differentiation via a ß-catenin-dependent pathway. However, the mechanisms by which TNF-α induces Wnt/ß-catenin signaling pathway in adipocytes is unclear. Msx2, a homeobox transcription factor, is known to increase osteoblast differentiation through activation of the Wnt/ß-catenin pathway. Therefore, in the present study, we investigated whether TNF-α activates the Wnt/ß-catenin signaling pathway via the induction of Msx2 expression in 3T3-L1 preadipocytes. We found that TNF-α transiently increased Msx2 expression as well as the expression of canonical Wnt signaling molecules, including Wnt3a, Wnt7a, Wnt7b, Wnt10b, low-density lipoprotein receptor-related protein 5 (LRP5) and T-cell factor 1 (TCF1). Furthermore, TNF-α increased ß-catenin/TCF-dependent transcriptional activity. To better understand the role of Msx2 in Wnt signaling, we examined the effects of Msx2 overexpression and knockdown on Wnt/ß-catenin signaling. Msx2 overexpression alone significantly increased the levels of Wnt3a, Wnt7a, Wnt7b, Wnt10b, LRP5 and TCF1 expression, whereas knockdown of Msx2 using small interfering RNA prevented TNF-α-induced expression of Wnt signaling molecules. Taken together, the results of this study indicate that TNF-α enhances the Wnt/ß-catenin signaling pathway by inducing Msx2 expression, which in turn suppresses adipocytic differentiation.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Proteínas de Homeodominio/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Animales , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Humanos , Ratones , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
18.
Bone ; 49(2): 242-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21514407

RESUMEN

Nuclear factor of activated T cell (NFAT) is a key transcription factor for receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation. However, it is unclear whether NFAT plays a role in the expression of RANKL in osteoblasts. High extracellular calcium ([Ca(2+)](o)) increases intracellular calcium, enhances RANKL expression in osteoblasts/stromal cells, and induces osteoclastogenesis in a coculture of osteoblasts and hematopoietic bone marrow cells. Because intracellular calcium signaling activates the calcineurin/NFAT pathway, we examined the role of NFAT activation on high [Ca(2+)](o)-induced RANKL expression in MC3T3-E1 subclone 4 (MC4) cells. Among the family of NFAT transcription factors, expression of NFATc1 and NFATc3, but not NFATc2, NFATc4 or NFAT5, was observed in MC4 cells. High [Ca(2+)](o) increased the expression levels of NFATc1, NFATc3 and RANKL. Cyclosporin A and FK506, inhibitors of calcineurin phosphatase, blocked high [Ca(2+)](o)-induced expression of NFAT and RANKL. Knockdown of NFATc1 and NFATc3 by siRNA prevented high [Ca(2+)](o)-induced RANKL expression, whereas overexpression of NFATc1 and NFATc3 induced RANKL expression. Furthermore, overexpressed NFATc1 upregulated NFATc3 expression, but NFATc1 knockdown decreased NFATc3 expression. Chromatin immunoprecipitation and reporter assay results showed that NFATc3, but not NFATc1, directly binds to the RANKL promoter and stimulates RANKL expression. In summary, these results demonstrate that high [Ca(2+)](o) increases expression of RANKL via activation of the calcineurin/NFAT pathway in osteoblasts. In addition, high [Ca(2+)](o) induces the activation and expression of NFATc1; NFATc3 expression and activity are subsequently increased; and NFATc3 directly binds to the RANKL promoter to increase its expression.


Asunto(s)
Calcio/farmacología , Factores de Transcripción NFATC/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Animales , Western Blotting , Calcineurina/genética , Calcineurina/metabolismo , Línea Celular , Inmunoprecipitación de Cromatina , Ratones , Mutagénesis Sitio-Dirigida , Factores de Transcripción NFATC/genética , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...