Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Sci (Weinh) ; 11(1): e2302203, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37967345

RESUMEN

Macrophage-myofibroblast transition (MMT) is a newly discovered pathway for mass production of pro-tumoral cancer-associated fibroblasts (CAFs) in non-small cell lung carcinoma (NSCLC) in a TGF-ß1/Smad3 dependent manner. Better understanding its regulatory signaling in tumor microenvironment (TME) may identify druggable target for the development of precision medicine. Here, by dissecting the transcriptome dynamics of tumor-associated macrophage at single-cell resolution, a crucial role of a hematopoietic transcription factor Runx1 in MMT formation is revealed. Surprisingly, integrative bioinformatic analysis uncovers Runx1 as a key regulator in the downstream of MMT-specific TGF-ß1/Smad3 signaling. Stromal Runx1 level positively correlates with the MMT-derived CAF abundance and mortality in NSCLC patients. Mechanistically, macrophage-specific Runx1 promotes the transcription of genes related to CAF signatures in MMT cells at genomic level. Importantly, macrophage-specific genetic deletion and systemic pharmacological inhibition of TGF-ß1/Smad3/Runx1 signaling effectively prevent MMT-driven CAF and tumor formation in vitro and in vivo, representing a potential therapeutic target for clinical NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Miofibroblastos/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Microambiente Tumoral
2.
Inflammation ; 46(6): 2241-2253, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37530929

RESUMEN

Our previous study had demonstrated that Runx1 promoted LPS-induced macrophage inflammatory response, however, the role of Runx1 in M2 macrophage polarization still remains largely unknown. This study was conducted to investigate the role of Runx1 in IL-4/IL-13-induced M2 macrophage polarization and its potential regulatory mechanism. We found that exposure of macrophages to IL-4/IL-13 induced a remarkable increasement in Runx1 expression level. Specifically, we established genetically modified mice lacking Runx1 in myeloid cells, including macrophages. RNA-Seq was performed to identify differentially expressed genes (DEGs) between Runx1 knockout and WT control bone marrow-derived macrophages (BMDMs). We identified 686 DEGs, including many genes which were highly expressed in M2 macrophage. In addition, bioinformatics analysis indicated that these DEGs were significantly enriched in extracellular matrix-related processes. Moreover, RT-qPCR analysis showed that there was an obvious upregulation in the relative expression levels of M2 marker genes, including Arg1, Ym1, Fizz1, CD71, Mmp9, and Tgm2, in Runx1 knockout macrophages, as compared to WT controls. Consistently, similar results were obtained in the protein and enzymatic activity levels of Arg1. Finally, we found that the STAT6 phosphorylation level was significantly enhanced in Runx1 knockout macrophages, and the STAT6 inhibitor AS1517499 partly reduced the upregulated effect of Runx1 deficiency on the M2 macrophage polarization. Taken together, Runx1 deficiency facilitates IL-4/IL-13-induced M2 macrophage polarization through enhancing STAT6 phosphorylation.


Asunto(s)
Interleucina-13 , Interleucina-4 , Animales , Ratones , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Interleucina-13/metabolismo , Interleucina-4/farmacología , Interleucina-4/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Fosforilación , Factor de Transcripción STAT6/metabolismo
3.
Food Funct ; 14(18): 8201-8216, 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37551935

RESUMEN

Activation of adipose tissue thermogenesis is a promising strategy in the treatment of obesity and obesity-related metabolic disorders. Kaempferol (KPF) is a predominant dietary flavonoid with multiple pharmacological properties, such as anti-inflammatory and antioxidant activities. In this study, we sought to characterize the role of KPF in adipocyte thermogenesis. We demonstrated that KPF-treated mice were protected from diet-induced obesity, glucose tolerance, and insulin resistance, accompanied by markedly increased energy expenditure, ex vivo oxygen consumption of white fat, and increased expression of proteins related to adaptive thermogenesis. KPF-promoted beige cell formation is a cell-autonomous effect, since the overexpression of cyclin-dependent kinase 6 (CDK6) in preadipocytes partially reversed browning phenotypes observed in KPF-treated cells. Overall, these data implicate that KPF is involved in promoting beige cell formation by suppressing CDK6 protein expression. This study provides evidence that KPF is a promising natural product for obesity treatment by boosting energy expenditure.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Quinasa 6 Dependiente de la Ciclina , Animales , Ratones , Quinasa 6 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/farmacología , Quinasa 6 Dependiente de la Ciclina/uso terapéutico , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/uso terapéutico , Tejido Adiposo Pardo/metabolismo , Quempferoles/farmacología , Adipocitos , Obesidad/tratamiento farmacológico , Obesidad/genética , Obesidad/metabolismo , Tejido Adiposo Blanco/metabolismo , Dieta Alta en Grasa/efectos adversos , Transducción de Señal , Termogénesis , Ratones Endogámicos C57BL , Metabolismo Energético
4.
Mar Drugs ; 21(6)2023 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-37367670

RESUMEN

Due to the challenge of prostate cancer (PCa) management, there has been a surge in efforts to identify more safe and effective compounds that can modulate the epithelial-mesenchymal transition (EMT) for driving metastasis. Holothurin A (HA), a triterpenoid saponin isolated from Holothuria scabra, has now been characterized for its diverse biological activities. However, the mechanisms of HA in EMT-driven metastasis of human PCa cell lines has not yet been investigated. Moreover, runt-related transcription factor 1 (RUNX1) acts as an oncogene in prostate cancer, but little is known about its role in the EMT. Thus, the purpose of this study was to determine how RUNX1 influences EMT-mediated metastasis, as well as the potential effect of HA on EMT-mediated metastasis in endogenous and exogenous RUNX1 expressions of PCa cell lines. The results demonstrated that RUNX1 overexpression could promote the EMT phenotype with increased EMT markers, consequently driving metastatic migration and invasion in PC3 cell line through the activation of Akt/MAPK signaling pathways. Intriguingly, HA treatment could antagonize the EMT program in endogenous and exogenous RUNX1-expressing PCa cell lines. A decreasing metastasis of both HA-treated cell lines was evidenced through a downregulation of MMP2 and MMP9 via the Akt/P38/JNK-MAPK signaling pathway. Overall, our approach first demonstrated that RUNX1 enhanced EMT-driven prostate cancer metastasis and that HA was capable of inhibiting the EMT and metastatic processes and should probably be considered as a candidate for metastasis PCa treatment.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias de la Próstata , Masculino , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Transducción de Señal , Neoplasias de la Próstata/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Movimiento Celular , Línea Celular Tumoral , Metástasis de la Neoplasia , Invasividad Neoplásica
5.
Int J Biol Macromol ; 238: 124026, 2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-36933589

RESUMEN

Bone morphogenetic protein 15 (BMP15) is specifically expressed in oocytes in pigs at all stages from early stages to ovulation and has an important role in oocyte maturation. However, there are few reports on the molecular mechanisms by which BMP15 affects oocyte maturation. In this study, we identified the core promoter region of BMP15 using a dual luciferase activity assay and successfully predicted the DNA binding motif of the transcription factor RUNX1. The effect of BMP15 and RUNX1 on oocyte maturation was examined using the first polar body extrusion rate, a reactive oxygen species (ROS) assay and total glutathione (GSH) content at three time points of 12, 24 and 48 h of in vitro culture of porcine isolated oocytes. Subsequently, the effect of the transcription factor RUNX1 on the TGF-ß signaling pathway (BMPR1B and ALK5) was further verified using RT-qPCR and Western blotting. We found that the overexpression of BMP15 significantly increased the first polar body extrusion rate (P < 0.01) and total glutathione content of oocytes cultured in vitro for 24 h and decreased reactive oxygen levels (P < 0.01), whereas interference with BMP15 decreased the first polar body extrusion rate (P < 0.01), increased reactive oxygen levels in oocytes cultured in vitro for 24 h (P < 0.01), and decreased glutathione content (P < 0.01). The dual luciferase activity assay and online software prediction showed that RUNX1 is a potential transcription factor binding to the core promoter region (-1203/-1423 bp) of BMP15. Overexpression of RUNX1 significantly increased the expression of BMP15 and oocyte maturation rate, while inhibition of RUNX1 decreased the expression of BMP15 and the oocyte maturation rate. Moreover, the expression of BMPR1B and ALK5 in the TGF-ß signaling pathway increased significantly after overexpression of RUNX1, whereas their expression decreased after inhibition of RUNX1. Overall, our results suggest that the transcription factor RUNX1 positively regulates the expression of BMP15 and influences oocyte maturation through the TGF-ß signaling pathway. This study provides a theoretical basis for further complementing the BMP15/TGF-ß signaling pathway to regulate mammalian oocyte maturation.


Asunto(s)
Proteína Morfogenética Ósea 15 , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Femenino , Animales , Porcinos , Proteína Morfogenética Ósea 15/genética , Proteína Morfogenética Ósea 15/metabolismo , Proteína Morfogenética Ósea 15/farmacología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Oocitos , Transducción de Señal , Glutatión/metabolismo , Oxígeno/metabolismo , Luciferasas/metabolismo , Mamíferos/metabolismo
6.
Neurobiol Dis ; 181: 106080, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36925052

RESUMEN

BACKGROUND: Ischemic stroke (IS) is the primary cause of mortality and disability worldwide. Circular RNAs (circRNAs) have been proposed as crucial regulators in IS. This study focused on the role of circPDS5B in IS and its underlying mechanism. METHOD: Transient middle cerebral artery occlusion (tMCAO) mice and glucose deprivation/reoxygenation (OGD/R)-exposed human brain microvascular endothelial cells (BMECs) were used as IS models. Expression levels of circPDS5B, heterogenous nuclear ribonucleoprotein L (hnRNPL), runt-related transcription factor-1 (Runx1), and Zinc finger protein 24 (ZNF24) were quantified by qRT-PCR. MTT, wound healing, transwell and tube formation assays were employed to evaluate the cell proliferation, migration, and angiogenesis, respectively. Moreover, RNA pull-down, and RIP assay were performed to investigate the interaction among circPDS5B, hnRNPL and vascular endothelial growth factor-A (VEGF-A). RESULTS: circPDS5B was significantly up-regulated in IS patients and tMCAO mice. Deficiency of circPDS5B relieved brain infarction and neuronal injury of tMCAO mice. OGD/R-induced apoptosis, inhibition in viability, migration, and angiogenesis in BMECs were dramatically abrogated by circPDS5B knockdown. Mechanistically, circPDS5B stabilized Runx1 and ZNF24 via recruiting hnRNPL, thereby suppressing the transcription and expression of VEGFA. hnRNPL silencing strengthened circPDS5B knockdown-mediated beneficial effect on IS. CONCLUSION: Altogether, our study showed that high expression of circPDS5B exacerbated IS through recruitment of hnRNPL to stabilize Runx1/ZNF24 and subsequently inactivate VEGFA. Our findings suggest circPDS5B may be a novel therapeutic target for IS.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo L , Accidente Cerebrovascular Isquémico , MicroARNs , Accidente Cerebrovascular , Factor A de Crecimiento Endotelial Vascular , Animales , Humanos , Ratones , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Células Endoteliales/metabolismo , Glucosa/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo L/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo L/farmacología , Infarto de la Arteria Cerebral Media/metabolismo , Accidente Cerebrovascular Isquémico/genética , Accidente Cerebrovascular Isquémico/metabolismo , MicroARNs/metabolismo , Neovascularización Fisiológica , ARN Circular/genética , ARN Circular/metabolismo , ARN Circular/farmacología , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Mol Carcinog ; 62(5): 598-612, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36727657

RESUMEN

Since its initial discovery as a natural isotopologue of dihydrogen oxide (1 H2 O), extensive research has focused on the biophysical, biochemical, and pharmacological effects of deuterated water (2 H2 O [D2 O, also referred to as "heavy water"]). Using a panel of cultured human pancreatic ductal adenocarcinoma (PDAC) cells we have profiled (i) D2 O-induced phenotypic antiproliferative and apoptogenic effects, (ii) redox- and proteotoxicity-directed stress response gene expression, and (iii) phosphoprotein-signaling related to endoplasmic reticulum (ER) and MAP-kinase stress response pathways. Differential array analysis revealed early modulation of stress response gene expression in both BxPC-3 and PANC-1 PDAC cells elicited by D2 O (90%; ≤6 h; upregulated: HMOX1, NOS2, CYP2E1, CRYAB, DDIT3, NFKBIA, PTGS1, SOD2, PTGS2; downregulated: RUNX1, MYC, HSPA8, HSPA1A) confirmed by independent RT-qPCR analysis. Immunoblot-analysis revealed rapid (≤6 h) onset of D2 O-induced MAP-kinase signaling (p-JNK, p-p38) together with ER stress response upregulation (p-eIF2α, ATF4, XBP1s, DDIT3/CHOP). Next, we tested the chemotherapeutic efficacy of D2 O-based drinking water supplementation in an orthotopic PDAC model employing firefly luciferase-expressing BxPC-3-FLuc cells in SCID mice. First, feasibility and time course of systemic deuteration (30% D2 O in drinking water; 21 days) were established using time-resolved whole-body proton magnetic resonance imaging and isotope-ratio mass spectrometry-based plasma (D/H)-analysis. D2 O-supplementation suppressed tumor growth by almost 80% with downregulated expression of PCNA, MYC, RUNX1, and HSP70 while increasing tumor levels of DDIT3/CHOP, HO-1, and p-eIF2α. Taken together, these data demonstrate for the first time that pharmacological induction of systemic deuteration significantly reduces orthotopic tumor burden in a murine PDAC xenograft model.


Asunto(s)
Carcinoma Ductal Pancreático , Agua Potable , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/uso terapéutico , Ratones SCID , Óxido de Deuterio/farmacología , Óxido de Deuterio/uso terapéutico , Línea Celular Tumoral , Neoplasias Pancreáticas/genética , Carcinoma Ductal Pancreático/genética , Proliferación Celular , Apoptosis , Neoplasias Pancreáticas
8.
Mol Cells ; 46(4): 231-244, 2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-36625319

RESUMEN

Leucine-rich repeat containing 15 (LRRC15) has been identified as a contributing factor for cartilage damage in osteoarthritis; however, its involvement in rheumatoid arthritis (RA) and the underlying mechanisms have not been well characterized. The purpose of this study was to explore the function of LRRC15 in RA-associated fibroblast-like synoviocytes (RA-FLS) and in mice with collagen-induced arthritis (CIA) and to dissect the epigenetic mechanisms involved. LRRC15 was overexpressed in the synovial tissues of patients with RA, and LRRC15 overexpression was associated with increased proliferative, migratory, invasive, and angiogenic capacities of RA-FLS and accelerated release of pro-inflammatory cytokines. LRRC15 knockdown significantly inhibited synovial proliferation and reduced bone invasion and destruction in CIA mice. Runt-related transcription factor 1 (RUNX1) transcriptionally represses LRRC15 by binding to core-binding factor subunit beta (CBF-ß). Overexpression of RUNX1 significantly inhibited the invasive phenotype of RA-FLS and suppressed the expression of proinflammatory cytokines. Conversely, the effects of RUNX1 were significantly reversed after overexpression of LRRC15 or inhibition of RUNX1-CBF-ß interactions. Therefore, we demonstrated that RUNX1-mediated transcriptional repression of LRRC15 inhibited the development of RA, which may have therapeutic effects for RA patients.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Animales , Ratones , Artritis Experimental/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Citocinas/metabolismo , Epigénesis Genética , Fibroblastos/metabolismo , Membrana Sinovial/metabolismo , Humanos
9.
Mol Biotechnol ; 64(10): 1130-1142, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35435591

RESUMEN

MiR-199a-3p was reported decreased in serum of coronary heart disease patients and human atherosclerotic plaques. This study aims to investigate the roles of miR-199a-3p in atherosclerosis (AS). AS was induced in ApoE-/- mice via high fat diet for 12 weeks. Oxidized low density lipoprotein (ox-LDL) was used to induce foaming in RAW264.7 cells. The expression level of miR-199a-3p was decreased in aortas of AS mice and ox-LDL-treated macrophages. Oil red O staining, ELISA, flow cytometry, and western blot results demonstrated that miR-199a-3p mimics restrained ox-LDL-induced lipid accumulation, foaming, and inflammation in RAW264.7 cells, while miR-199a-3p inhibitor played opposite roles. Runt-related transcription factor 1 (RUNX1), a pro-inflammatory factor, was identified as a target of miR-199a-3p, and its expression was downregulated by miR-199a-3p. RUNX1 was increased in macrophages from aortas and peripheral blood of AS mice. Ox-LDL-induced inflammation and lipid accumulation were aggravated by RUNX1, and the effects of miR-199a-3p were antagonized by ectopic expression of RUNX1 in RAW264.7 cells. The phosphorylation of signal transducer and activator of transcription 3 (STAT3) was inhibited by miR-199a-3p and enhanced by RUNX1. In conclusions, we demonstrated that miR-199a-3p alleviated ox-LDL-induced foaming and inflammation by downregulating RUNX1 expression and deactivating STAT3 signaling in macrophages. These findings may provide novel targets for treatment of AS.


Asunto(s)
Aterosclerosis , MicroARNs , Animales , Apoptosis , Aterosclerosis/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo
10.
Int J Oncol ; 60(1)2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34958111

RESUMEN

Runt­related transcription factor 1 (RUNX1), which is also known as acute myeloid leukemia 1 (AML1), has been frequently found with genomic aberrations in human leukemia. RUNX1 encodes a transcription factor that can regulate the expression of hematopoietic genes. In addition, tumor necrosis factor­related apoptosis­inducing ligand (TRAIL) performs an important function for malignant tumors in immune surveillance. However, the regulatory mechanism of TRAIL expression remain to be fully elucidated. In the present study, tetradecanoylphorbol 13­acetate­treated megakaryocytic differentiated K562 cells was used to examine the effect of RUNX1 on TRAIL expression. Luciferase assay series of TRAIL promoters for the cells co­transfected with RUNX1 and core­binding factor ß (CBFß) expression vectors were performed to evaluate the nature of TRAIL transcriptional regulation. Electrophoresis mobility shift assay of the RUNX1 consensus sequence of the TRAIL promoter with recombinant RUNX1 and CBFß proteins was also performed. BloodSpot database analysis for TRAIL expression in patients with acute myeloid leukemia were performed. The expression of TRAIL, its receptor Death receptor 4 and 5 and RUNX1 in K562 cells transfected with the RUNX1 expression vector and RUNX1 siRNA were evaluated by reverse transcription­quantitative PCR (RT­qPCR). TRAIL and RUNX1­ETO expression was also measured in Kasumi­1 cells transfected with RUNX1­ETO siRNA and in KG­1 cells transfected with RUNX1­ETO expression plasmid, both by RT­qPCR. Cell counting, lactate dehydrogenase assay and cell cycle analysis by flow cytometry were performed on Kasumi­1, KG­1, SKNO­1 and K562 cells treated with TRAIL and HDAC inhibitors sodium butyrate or valproic acid. The present study demonstrated that RUNX1 is a transcriptional regulator of TRAIL. It was initially found that the induction of TRAIL expression following the megakaryocytic differentiation of human leukemia cells was RUNX1­dependent. Subsequently, overexpression of RUNX1 was found to increase TRAIL mRNA expression by activating its promoter activity. Additional analyses revealed that RUNX1 regulated the expression of TRAIL in an indirect manner, because RUNX1 retained its ability to activate this promoter following the mutation of all possible RUNX1 consensus sites. Furthermore, TRAIL expression was reduced in leukemia cells carrying the t(8;21) translocation, where the RUNX1­ETO chimeric protein interfere with normal RUNX1 function. Exogenous treatment of recombinant TRAIL proteins was found to induce leukemia cell death. To conclude, the present study provided a novel mechanism, whereby TRAIL is a target gene of RUNX1 and TRAIL expression was inhibited by RUNX1­ETO. These results suggest that TRAIL is a promising agent for the clinical treatment of t(8;21) AML.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/efectos de los fármacos , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Humanos , Células K562/efectos de los fármacos , Células K562/metabolismo , Ratones , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transcripción Genética/genética
11.
J Virol ; 94(22)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32878886

RESUMEN

Herpes simplex virus 1 (HSV-1) and HSV-2 can efficiently establish lifelong, transcriptionally silent latency states in sensory neurons to escape host detection. While host factors have previously been associated with long-range insulators in the viral genome, it is still unknown whether host transcription factors can repress viral genes more proximately to promote latency in dorsal root ganglion (DRG) neurons. Here, we assessed whether RUNX (runt-related transcription factor) transcription factors, which are critical in the development of sensory neurons, could be binding HSV-1 genome directly to suppress viral gene expression and lytic infection. Using previously published transcriptome sequencing data, we confirmed that mouse DRG neurons highly express Runx1 mRNA. Through computational analysis of HSV-1 and HSV-2 genomes, we observed that putative RUNX consensus binding sites (CBSs) were more enriched and more closely located to viral gene transcription start sites than would be expected by chance. We further found that RUNX CBSs were significantly more enriched among genomes of herpesviruses compared to those of nonherpesviruses. Utilizing an in vitro model of HSV-1 infection, we found that overexpressed RUNX1 could bind putative binding sites in the HSV-1 genome, repress numerous viral genes spanning all three kinetic classes, and suppress productive infection. In contrast, knockdown of RUNX1 in neuroblastoma cells induced viral gene expression and increased HSV-1 infection in vitro In sum, these data support a novel role for RUNX1 in directly binding herpesvirus genome, silencing the transcription of numerous viral genes, and ultimately limiting overall infection.IMPORTANCE Infecting 90% of the global population, HSV-1 and HSV-2 represent some of the most prevalent viruses in the world. Much of their success can be attributed to their ability to establish lifelong latent infections in the dorsal root ganglia (DRG). It is still largely unknown, however, how host transcription factors are involved in establishing this latency. Here, we report that RUNX1, expressed highly in DRG, binds HSV-1 genome, represses transcription of numerous viral genes, and suppresses productive in vitro infection. Our computational work further suggests this strategy may be used by other herpesviruses to reinforce latency in a cell-specific manner.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Herpesviridae/genética , Herpesviridae/fisiología , Herpesvirus Humano 1/efectos de los fármacos , Animales , Sitios de Unión , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Ganglios Espinales/virología , Regulación Viral de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genoma Viral , Células HEK293 , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Humanos , Ratones , Células Receptoras Sensoriales/virología , Ganglio del Trigémino/virología , Activación Viral/fisiología , Latencia del Virus/fisiología
12.
Mol Cells ; 43(2): 126-138, 2020 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-31991535

RESUMEN

The transcription factor RUNX1 first came to prominence due to its involvement in the t(8;21) translocation in acute myeloid leukemia (AML). Since this discovery, RUNX1 has been shown to play important roles not only in leukemia but also in the ontogeny of the normal hematopoietic system. Although it is currently still challenging to fully assess the different parameters regulating RUNX1 dosage, it has become clear that the dose of RUNX1 can greatly affect both leukemia and normal hematopoietic development. It is also becoming evident that varying levels of RUNX1 expression can be used as markers of tumor progression not only in the hematopoietic system, but also in non-hematopoietic cancers. Here, we provide an overview of the current knowledge of the effects of RUNX1 dosage in normal development of both hematopoietic and epithelial tissues and their associated cancers.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/uso terapéutico , Neoplasias/tratamiento farmacológico , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Humanos
13.
Blood ; 118(8): 2266-74, 2011 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-21725048

RESUMEN

G-CSF signals contribute to granulocyte lineage specification. We previously found that G-CSF induces SHP2 tyrosine phosphorylation and that chemical inhibition of SHP1/SHP2 reduces CFU-G and prevents G-CSF but not M-CSF activation of ERK. We now find that SHP2 shRNA knockdown in the 32Dcl3 granulocytic line reduces ERK activation, diminishes CEBPA protein and RNA expression and promoter histone acetylation, and inhibits granulopoiesis. Exogenous, shRNA-resistant SHP2 rescues these effects of SHP2 knockdown, exogenous C/EBPα rescues granulocytic markers, and exogenous RUNX1 rescues C/EBPα. 32Dcl3 lines with knockdown of ERK1 and ERK2 retain normal levels of C/EBPα and differentiate normally in G-CSF despite also having reduced proliferation. SHP2 knockdown reduces CEBPA levels in lineage-negative murine marrow cells cultured in TPO, Flt3 ligand, and SCF, without affecting the rate of cell expansion. On transfer to IL-3, IL-6, and SCF to induce myelopoiesis, levels of granulocytic RNAs are reduced and monocyte-specific RNAs are increased by SHP2 knockdown, and there is a reduction in the percentage of CFU-G that form in methylcellulose and of granulocytes that develop in liquid culture. In summary, SHP2 is required for induction of C/EBPα expression and granulopoiesis in response to G-CSF or other cytokines independent of SHP2-mediated ERK activation.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Mielopoyesis/genética , Mielopoyesis/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Secuencia de Bases , Línea Celular , Ensayo de Unidades Formadoras de Colonias , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Citocinas/farmacología , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Factor Estimulante de Colonias de Granulocitos/farmacología , Granulocitos/citología , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Mielopoyesis/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/farmacología , ARN Interferente Pequeño/genética , Proteínas Recombinantes
14.
Blood ; 108(10): 3329-34, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16888092

RESUMEN

The Notch1-RBP-Jkappa and the transcription factor Runx1 pathways have been independently shown to be indispensable for the establishment of definitive hematopoiesis. Importantly, expression of Runx1 is down-regulated in the para-aortic splanchnopleural (P-Sp) region of Notch1- and Rbpsuh-null mice. Here we demonstrate that Notch1 up-regulates Runx1 expression and that the defective hematopoietic potential of Notch1-null P-Sp cells is successfully rescued in the OP9 culture system by retroviral transfer of Runx1. We also show that Hes1, a known effector of Notch signaling, potentiates Runx1-mediated transactivation. Together with the recent findings in zebrafish, Runx1 is postulated to be a cardinal down-stream mediator of Notch signaling in hematopoietic development throughout vertebrates. Our findings also suggest that Notch signaling may modulate both expression and transcriptional activity of Runx1.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Receptor Notch1/deficiencia , Animales , Línea Celular , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/administración & dosificación , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis , Humanos , Ratones , Ratones Mutantes , Receptor Notch1/genética , Receptor Notch1/fisiología , Transducción Genética , Vísceras/citología
15.
J Biol Chem ; 280(49): 40589-98, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16195230

RESUMEN

Osteopontin (OPN), a glycosylated phosphoprotein that binds calcium, is present in bone extracellular matrix and has been reported to modulate both mineralization and bone resorption. Targeted disruption in mice of the vitamin D receptor (VDR) or Runx2 results in marked inhibition of OPN expression in osteoblasts. In this study, we addressed possible cross-talk between VDR and Runx2 in regulating OPN transcription. 1,25-Dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) or Runx2 stimulated OPN transcription (mouse OPN promoter -777/+79) 2-3-fold. However, coexpression of Runx2 and VDR in COS-7 cells and treatment with 1,25(OH)(2)D(3) resulted in an 8-fold induction of OPN transcription, indicating for the first time functional cooperation between Runx2 and VDR in the regulation of OPN transcription. In ROS 17/2.8 and MC3T3-E1 cells that contain endogenous Runx2, AML-1/ETO, which acts as a repressor of Runx2, significantly inhibited 1,25(OH)(2)D(3) induction of OPN transcription, OPN mRNA, and protein expression. Both a Runx2 site (-136/-130) and the vitamin D response element (-757/-743) in the OPN promoter are needed for cooperative activation. Chromatin immunoprecipitation analyses showed that 1,25(OH)(2)D(3) can enhance VDR and Runx2 recruitment on the OPN promoter, further indicating cooperation between these two factors in the regulation of OPN. In osteoblastic cells, Hes-1, a downstream factor of the Notch signaling pathway, was found to enhance basal and 1,25(OH)(2)D(3)-induced OPN transcription. This enhancement was inhibited by AML-1/ETO, an inhibitor of Runx2. Immunoprecipitation assays indicated that Hes-1 and Runx2 interact and that 1,25(OH)(2)D(3) can enhance this interaction. Taken together, these findings define novel mechanisms involving the intersection of three pathways, Runx2, 1,25(OH)(2)D(3), and Notch signaling, that play a major role in the regulation of OPN in osteoblastic cells and therefore in the process of bone remodeling.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Regulación de la Expresión Génica , Receptores Notch/fisiología , Sialoglicoproteínas/genética , Transducción de Señal/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Western Blotting , Remodelación Ósea/fisiología , Células COS , Calcitriol/farmacología , Chlorocebus aethiops , Cromatina/química , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/farmacología , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/fisiología , Técnicas de Inmunoadsorción , Luciferasas/genética , Ratones , Mutagénesis Sitio-Dirigida , Proteínas de Fusión Oncogénica/farmacología , Osteoblastos/metabolismo , Osteopontina , Regiones Promotoras Genéticas/genética , ARN Mensajero/análisis , Proteína 1 Compañera de Translocación de RUNX1 , Receptor Cross-Talk/fisiología , Receptores de Calcitriol/genética , Receptores de Calcitriol/fisiología , Proteínas Recombinantes de Fusión , Elementos de Respuesta/fisiología , Factor de Transcripción HES-1 , Transcripción Genética/efectos de los fármacos , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...