Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Br J Cancer ; 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39313574

RESUMEN

BACKGROUND: In breast cancer, ErbB receptors play a critical role, and overcoming drug resistance remains a major challenge in the clinic. However, intricate regulatory mechanisms of ErbB family genes are poorly understood. Here, we demonstrate SON as an ErbB-regulatory splicing factor and a novel therapeutic target for ErbB-positive breast cancer. METHODS: SON and ErbB expression analyses using public database, patient tissue microarray, and cell lines were performed. SON knockdown assessed its impact on cell proliferation, apoptosis, kinase phosphorylation, RNA splicing, and in vivo tumour growth. RNA immunoprecipitation was performed to measure SON binding. RESULTS: SON is highly expressed in ErbB2-positive breast cancer patient samples, inversely correlating with patient survival. SON knockdown induced intron retention in selective splice sites within ErbB2 and ErbB3 transcripts, impairing effective RNA splicing and reducing protein expression. SON disruption suppressed downstream kinase signalling of ErbB2/3, including the Akt, p38, and JNK pathways, with increased vulnerability in ErbB2-positive breast cancer cells compared to ErbB2-negative cells. SON silencing in ErbB2-positive breast cancer xenografts led to tumour regression in vivo. CONCLUSION: We identified SON as a novel RNA splicing factor that plays a critical role in regulating ErbB2/3 expression, suggesting SON is an ideal therapeutic target in ErbB2-positive breast cancers.

2.
JCI Insight ; 9(5)2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38290089

RESUMEN

Rare diseases are underrepresented in biomedical research, leading to insufficient awareness. Zhu-Tokita-Takenouchi-Kim (ZTTK) syndrome is a rare disease caused by genetic alterations that result in heterozygous loss of function of SON. While patients with ZTTK syndrome live with numerous symptoms, the lack of model organisms hampers our understanding of SON and this complex syndrome. Here, we developed Son haploinsufficiency (Son+/-) mice as a model of ZTTK syndrome and identified the indispensable roles of Son in organ development and hematopoiesis. Son+/- mice recapitulated clinical symptoms of ZTTK syndrome, including growth retardation, cognitive impairment, skeletal abnormalities, and kidney agenesis. Furthermore, we identified hematopoietic abnormalities in Son+/- mice, including leukopenia and immunoglobulin deficiency, similar to those observed in human patients. Surface marker analyses and single-cell transcriptome profiling of hematopoietic stem and progenitor cells revealed that Son haploinsufficiency shifted cell fate more toward the myeloid lineage but compromised lymphoid lineage development by reducing genes required for lymphoid and B cell lineage specification. Additionally, Son haploinsufficiency caused inappropriate activation of erythroid genes and impaired erythropoiesis. These findings highlight the importance of the full gene expression of Son in multiple organs. Our model serves as an invaluable research tool for this rare disease and related disorders associated with SON dysfunction.


Asunto(s)
Hematopoyesis , Enfermedades Raras , Animales , Humanos , Ratones , Perfilación de la Expresión Génica , Hematopoyesis/genética , Mutación
3.
Cell Stem Cell ; 30(12): 1658-1673.e10, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38065069

RESUMEN

Stem cells regulate their self-renewal and differentiation fate outcomes through both symmetric and asymmetric divisions. m6A RNA methylation controls symmetric commitment and inflammation of hematopoietic stem cells (HSCs) through unknown mechanisms. Here, we demonstrate that the nuclear speckle protein SON is an essential m6A target required for murine HSC self-renewal, symmetric commitment, and inflammation control. Global profiling of m6A identified that m6A mRNA methylation of Son increases during HSC commitment. Upon m6A depletion, Son mRNA increases, but its protein is depleted. Reintroduction of SON rescues defects in HSC symmetric commitment divisions and engraftment. Conversely, Son deletion results in a loss of HSC fitness, while overexpression of SON improves mouse and human HSC engraftment potential by increasing quiescence. Mechanistically, we found that SON rescues MYC and suppresses the METTL3-HSC inflammatory gene expression program, including CCL5, through transcriptional regulation. Thus, our findings define a m6A-SON-CCL5 axis that controls inflammation and HSC fate.


Asunto(s)
Proteínas de Unión al ADN , Células Madre Hematopoyéticas , Inflamación , Metilación de ARN , Animales , Humanos , Ratones , Diferenciación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Metilación de ARN/genética
4.
bioRxiv ; 2023 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-38014320

RESUMEN

Rare diseases are underrepresented in biomedical research, leading to insufficient awareness. Zhu-Tokita-Takenouchi-Kim (ZTTK) syndrome is a rare disease caused by genetic alterations that result in heterozygous loss-of-function of SON. While ZTTK syndrome patients suffer from numerous symptoms, the lack of model organisms hamper our understanding of both SON and this complex syndrome. Here, we developed Son haploinsufficiency (Son+/-) mice as a model of ZTTK syndrome and identified the indispensable roles of Son in organ development and hematopoiesis. Son+/- mice recapitulated clinical symptoms of ZTTK syndrome, including growth retardation, cognitive impairment, skeletal abnormalities, and kidney agenesis. Furthermore, we identified hematopoietic abnormalities in Son+/- mice, similar to those observed in human patients. Surface marker analyses and single-cell transcriptome profiling of hematopoietic stem and progenitor cells revealed that Son haploinsufficiency inclines cell fate toward the myeloid lineage but compromises lymphoid lineage development by reducing key genes required for lymphoid and B cell lineage specification. Additionally, Son haploinsufficiency causes inappropriate activation of erythroid genes and impaired erythroid maturation. These findings highlight the importance of the full gene dosage of Son in organ development and hematopoiesis. Our model serves as an invaluable research tool for this rare disease and related disorders associated with SON dysfunction.

5.
Pharmaceuticals (Basel) ; 16(3)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36986479

RESUMEN

BACKGROUND: This study explores the association of neutrophil-to-lymphocyte (NLR), monocyte-to-lymphocyte (MLR), and platelet-to-lymphocyte (PLR) ratios with the 3-month treatment response and persistence of tumor necrosis factor-alpha (TNF-α) blockers in patients with ankylosing spondylitis (AS). METHODS: This retrospective cohort study investigated 279 AS patients who were newly initiated on TNF-α blockers between April 2004 and October 2019 and 171 sex- and age-matched healthy controls. Response to TNF-α blockers was defined as a reduction in the Bath AS Disease Activity Index of ≥50% or 20 mm, and persistence referred to the time interval from the initiation to discontinuation of TNF-α blockers. RESULTS: Patients with AS had significantly increased NLR, MLR, and PLR ratios as compared to controls. The frequency of non-response at 3 months was 3.7%, and TNF-α blockers' discontinuation occurred in 113 (40.5%) patients during the follow-up period. A high baseline NLR but not high baseline MLR and PLR showed an independently significant association with a higher risk of non-response at 3 months (OR = 12.3, p = 0.025) and non-persistence with TNF-α blockers (HR = 1.66, p = 0.01). CONCLUSIONS: NLR may be a potential marker for predicting the clinical response and persistence of TNF-α blockers in AS patients.

6.
J Biol Chem ; 298(6): 102013, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35525274

RESUMEN

Dysregulation of cyclin-dependent kinases (CDKs) can promote unchecked cell proliferation and cancer progression. Although focal adhesion kinase (FAK) contributes to regulating cell cycle progression, the exact molecular mechanism remains unclear. Here, we found that FAK plays a key role in cell cycle progression potentially through regulation of CDK4/6 protein expression. We show that FAK inhibition increased its nuclear localization and induced G1 arrest in B16F10 melanoma cells. Mechanistically, we demonstrate nuclear FAK associated with CDK4/6 and promoted their ubiquitination and proteasomal degradation through recruitment of CDC homolog 1 (CDH1), an activator and substrate recognition subunit of the anaphase-promoting complex/cyclosome E3 ligase complex. We found the FAK N-terminal FERM domain acts as a scaffold to bring CDK4/6 and CDH1 within close proximity. However, overexpression of nonnuclear-localizing mutant FAK FERM failed to function as a scaffold for CDK4/6 and CDH1. Furthermore, shRNA knockdown of CDH1 increased CDK4/6 protein expression and blocked FAK inhibitor-induced reduction of CDK4/6 in B16F10 cells. In vivo, we show that pharmacological FAK inhibition reduced B16F10 tumor size, correlating with increased FAK nuclear localization and decreased CDK4/6 expression compared with vehicle controls. In patient-matched healthy skin and melanoma biopsies, we found FAK was mostly inactive and nuclear localized in healthy skin, whereas melanoma lesions showed increased active cytoplasmic FAK and elevated CDK4 expression. Taken together, our data demonstrate that FAK inhibition blocks tumor proliferation by inducing G1 arrest, in part through decreased CDK4/6 protein stability by nuclear FAK.


Asunto(s)
Antígenos CD , Cadherinas , Quinasa 6 Dependiente de la Ciclina , Proteína-Tirosina Quinasas de Adhesión Focal , Melanoma , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Melanoma/genética , Melanoma/fisiopatología , Estados Unidos
7.
Cardiovasc Res ; 118(4): 1150-1163, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33839758

RESUMEN

AIMS: Vascular smooth muscle cells (VSMCs) normally exhibit a very low proliferative rate. Vessel injury triggers VSMC proliferation, in part, through focal adhesion kinase (FAK) activation, which increases transcription of cyclin D1, a key activator for cell cycle-dependent kinases (CDKs). At the same time, we also observe that FAK regulates the expression of the CDK inhibitors (CDKIs) p27 and p21. However, the mechanism of how FAK controls CDKIs in cell cycle progression is not fully understood. METHODS AND RESULTS: We found that pharmacological and genetic FAK inhibition increased p27 and p21 by reducing stability of S-phase kinase-associated protein 2 (Skp2), which targets theCDKIs for degradation. FAK N-terminal domain interacts with Skp2 and an APC/C E3 ligase activator fizzy-related 1 (Fzr1) in the nucleus, which promote ubiquitination and degradation of both Skp2 and Fzr1. Notably, overexpression of cyclin D1 alone failed to promote proliferation of genetic FAK kinase-dead (KD) VSMCs, suggesting that the FAK-Skp2-CDKI signalling axis is distinct from the FAK-cyclin D1 pathway. However, overexpression of both cyclin D1 and Skp2 enabled proliferation of FAK-KD VSMCs, implicating that FAK ought to control both activating and inhibitory switches for CDKs. In vivo, wire injury activated FAK in the cytosol, which increased Skp2 and decreased p27 and p21 levels. CONCLUSION: Both pharmacological FAK and genetic FAK inhibition reduced Skp2 expression in VSMCs upon injury, which significantly reduced intimal hyperplasia through elevated expression of p27 and p21. This study revealed that nuclear FAK-Skp2-CDKI signalling negatively regulates CDK activity in VSMC proliferation.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Músculo Liso Vascular , Proteínas Quinasas Asociadas a Fase-S , Proliferación Celular , Células Cultivadas , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo
8.
Circ Res ; 129(12): e215-e233, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34702049

RESUMEN

RATIONALE: Vascular smooth muscle cells (SMCs) exhibit remarkable plasticity and can undergo dedifferentiation upon pathological stimuli associated with disease and interventions. OBJECTIVE: Although epigenetic changes are critical in SMC phenotype switching, a fundamental regulator that governs the epigenetic machineries regulating the fate of SMC phenotype has not been elucidated. METHODS AND RESULTS: Using SMCs, mouse models, and human atherosclerosis specimens, we found that FAK (focal adhesion kinase) activation elicits SMC dedifferentiation by stabilizing DNMT3A (DNA methyltransferase 3A). FAK in SMCs is activated in the cytoplasm upon serum stimulation in vitro or vessel injury and active FAK prevents DNMT3A from nuclear FAK-mediated degradation. However, pharmacological or genetic FAK catalytic inhibition forced FAK nuclear localization, which reduced DNMT3A protein via enhanced ubiquitination and proteasomal degradation. Reduced DNMT3A protein led to DNA hypomethylation in contractile gene promoters, which increased SMC contractile protein expression. RNA-sequencing identified SMC contractile genes as a foremost upregulated group by FAK inhibition from injured femoral artery samples compared with vehicle group. DNMT3A knockdown in injured arteries reduced DNA methylation and enhanced contractile gene expression supports the notion that nuclear FAK-mediated DNMT3A degradation via E3 ligase TRAF6 (TNF [tumor necrosis factor] receptor-associated factor 6) drives differentiation of SMCs. Furthermore, we observed that SMCs of human atherosclerotic lesions exhibited decreased nuclear FAK, which was associated with increased DNMT3A levels and decreased contractile gene expression. CONCLUSIONS: This study reveals that nuclear FAK induced by FAK catalytic inhibition specifically suppresses DNMT3A expression in injured vessels resulting in maintaining SMC differentiation by promoting the contractile gene expression. Thus, FAK inhibitors may provide a new treatment option to block SMC phenotypic switching during vascular remodeling and atherosclerosis.


Asunto(s)
Desdiferenciación Celular , Proteínas Contráctiles/genética , Metilación de ADN , Quinasa 1 de Adhesión Focal/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Células Cultivadas , Proteínas Contráctiles/metabolismo , ADN Metiltransferasa 3A/genética , ADN Metiltransferasa 3A/metabolismo , Quinasa 1 de Adhesión Focal/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Proteolisis , Ubiquitinación , Regulación hacia Arriba
9.
Nat Commun ; 12(1): 5551, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34548489

RESUMEN

While dysregulation of RNA splicing has been recognized as an emerging target for cancer therapy, the functional significance of RNA splicing and individual splicing factors in brain tumors is poorly understood. Here, we identify SON as a master regulator that activates PTBP1-mediated oncogenic splicing while suppressing RBFOX2-mediated non-oncogenic neuronal splicing in glioblastoma multiforme (GBM). SON is overexpressed in GBM patients and SON knockdown causes failure in intron removal from the PTBP1 transcript, resulting in PTBP1 downregulation and inhibition of its downstream oncogenic splicing. Furthermore, SON forms a complex with hnRNP A2B1 and antagonizes RBFOX2, which leads to skipping of RBFOX2-targeted cassette exons, including the PTBP2 neuronal exon. SON knockdown inhibits proliferation and clonogenicity of GBM cells in vitro and significantly suppresses tumor growth in orthotopic xenografts in vivo. Collectively, our study reveals that SON-mediated RNA splicing is a GBM vulnerability, implicating SON as a potential therapeutic target in brain tumors.


Asunto(s)
Neoplasias Encefálicas/genética , Proteínas de Unión al ADN/genética , Glioblastoma/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Antígenos de Histocompatibilidad Menor/genética , Proteínas del Tejido Nervioso/genética , Proteína de Unión al Tracto de Polipirimidina/genética , Factores de Empalme de ARN/genética , Empalme del ARN , Proteínas Represoras/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Exones , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Glioblastoma/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Xenoinjertos , Humanos , Intrones , Ratones , Antígenos de Histocompatibilidad Menor/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Factores de Empalme de ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Análisis de Supervivencia
10.
PLoS One ; 16(5): e0251515, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019552

RESUMEN

Emerging evidence has shown that active enhancers are abundantly transcribed, generating long non-coding RNAs, called enhancer RNAs (eRNAs). While putative eRNAs are often observed from RNA sequencing, the roles of most eRNAs remain largely unknown. Previously, we identified putative enhancer regions at the MALAT1 locus that form chromatin-chromatin interactions under hypoxia, and one of these enhancers is located about 30 kb downstream of the NEAT1 gene and -20 kb upstream of the MALAT1 gene (MALAT1-20 kb enhancer). Here, we report that a novel eRNA, named eRNA of the NEAT1-MALAT1-Locus (eNEMAL), is transcribed from the MALAT1-20 kb enhancer and conserved in primates. We found that eNEMAL is upregulated in response to hypoxia in multiple breast cancer cell lines, but not in non-tumorigenic MCF10A cells. Overexpression and knockdown of eNEMAL revealed that alteration of eNEMAL level does not affect MALAT1 expression. Instead, we found that eNEMAL upregulates the long isoform of NEAT1 (NEAT1_2) without increasing the total NEAT1 transcript level in MCF7 breast cancer cells, suggesting that eNEMAL has a repressive effect on the 3'-end polyadenylation process required for generating the short isoform of NEAT1 (NEAT1_1). Altogether, we demonstrated that an eRNA transcribed from a MALAT1 enhancer regulates NEAT1 isoform expression, implicating the MALAT1-20 kb enhancer and its transcript eNEMAL in co-regulation of MALAT1 and NEAT1 in response to hypoxia in breast cancer cells.


Asunto(s)
Neoplasias de la Mama , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Sitios Genéticos , ARN Largo no Codificante , ARN Neoplásico , Regulación hacia Arriba , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética
11.
Inflammation ; 44(3): 1130-1144, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33527321

RESUMEN

While sustained nuclear factor-κB (NF-κB) activation is critical for proinflammatory molecule expression, regulators of NF-κB activity during chronic inflammation are not known. We investigated the role of focal adhesion kinase (FAK) on sustained NF-κB activation in tumor necrosis factor-α (TNF-α)-stimulated endothelial cells (ECs) both in vitro and in vivo. We found that FAK inhibition abolished TNF-α-mediated sustained NF-κB activity in ECs by disrupting formation of TNF-α receptor complex-I (TNFRC-I). Additionally, FAK inhibition diminished recruitment of receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and the inhibitor of NF-κB (IκB) kinase (IKK) complex to TNFRC-I, resulting in elevated stability of IκBα protein. In mice given TNF-α, pharmacological and genetic FAK inhibition blocked TNF-α-induced IKK-NF-κB activation in aortic ECs. Mechanistically, TNF-α activated and redistributed FAK from the nucleus to the cytoplasm, causing elevated IKK-NF-κB activation. On the other hand, FAK inhibition trapped FAK in the nucleus of ECs even upon TNF-α stimulation, leading to reduced IKK-NF-κB activity. Together, these findings support a potential use for FAK inhibitors in treating chronic inflammatory diseases.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Inflamación/enzimología , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Transporte Activo de Núcleo Celular , Animales , Células Cultivadas , Quinasa 1 de Adhesión Focal/genética , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Quinasa I-kappa B/metabolismo , Inflamación/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidor NF-kappaB alfa/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal
12.
PLoS One ; 16(2): e0247489, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33630943

RESUMEN

The gene SON is on human chromosome 21 (21q22.11) and is thought to be associated with hematopoietic disorders that accompany Down syndrome. Additionally, SON is an RNA splicing factor that plays a role in the transcription of leukemia-associated genes. Previously, we showed that mutations in SON cause malformations in human and zebrafish spines and brains during early embryonic development. To examine the role of SON in normal hematopoiesis, we reduced expression of the zebrafish homolog of SON in zebrafish at the single-cell developmental stage with specific morpholinos. In addition to the brain and spinal malformations we also observed abnormal blood cell levels upon son knockdown. We then investigated how blood production was altered when levels of son were reduced. Decreased levels of son resulted in lower amounts of red blood cells when visualized with lcr:GFP transgenic fish. There were also reduced thrombocytes seen with cd41:GFP fish, and myeloid cells when mpx:GFP fish were examined. We also observed a significant decrease in the quantity of T cells, visualized with lck:GFP fish. However, when we examined their hematopoietic stem and progenitor cells (HSPCs), we saw no difference in colony-forming capability. These studies indicate that son is essential for the proper differentiation of the innate and adaptive immune system, and further investigation determining the molecular pathways involved during blood development should elucidate important information about vertebrate HSPC generation, proliferation, and differentiation.


Asunto(s)
Embrión no Mamífero/citología , Hematopoyesis , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente/embriología , Diferenciación Celular , Proliferación Celular , Proteínas de Unión al ADN/fisiología , Enfermedades Hematológicas/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Antígenos de Histocompatibilidad Menor/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
13.
Cancer Gene Ther ; 28(9): 1000-1015, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33247227

RESUMEN

A high incidence of acute megakaryoblastic leukemia (AMKL) in Down syndrome patients implies that chromosome 21 genes have a pivotal role in AMKL development, but the functional contribution of individual genes remains elusive. Here, we report that SON, a chromosome 21-encoded DNA- and RNA-binding protein, inhibits megakaryocytic differentiation by suppressing RUNX1 and the megakaryocytic gene expression program. As megakaryocytic progenitors differentiate, SON expression is drastically reduced, with mature megakaryocytes having the lowest levels. In contrast, AMKL cells express an aberrantly high level of SON, and knockdown of SON induced the onset of megakaryocytic differentiation in AMKL cell lines. Genome-wide transcriptome analyses revealed that SON knockdown turns on the expression of pro-megakaryocytic genes while reducing erythroid gene expression. Mechanistically, SON represses RUNX1 expression by directly binding to the proximal promoter and two enhancer regions, the known +23 kb enhancer and the novel +139 kb enhancer, at the RUNX1 locus to suppress H3K4 methylation. In addition, SON represses the expression of the AP-1 complex subunits JUN, JUNB, and FOSB which are required for late megakaryocytic gene expression. Our findings define SON as a negative regulator of RUNX1 and megakaryocytic differentiation, implicating SON overexpression in impaired differentiation during AMKL development.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/metabolismo , Leucemia Megacarioblástica Aguda/metabolismo , Megacariocitos/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Diferenciación Celular , Síndrome de Down/genética , Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Leucemia Megacarioblástica Aguda/genética , Leucemia Megacarioblástica Aguda/patología , Transfección
14.
Mater Sci Eng C Mater Biol Appl ; 116: 111253, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32806227

RESUMEN

Silver nanoparticles (AgNPs) were synthesized via a green strategy using fifty-eight plant extracts that originated from Vietnam and Indonesia. Among the fifty-eight AgNP samples, we selected six AgNP samples synthesized by the extracts of Areca catechu, Hypotrachyna laevigata, Ardisia incarnata, Maesa calophylla, Maesa laxiflora and Adinandra poilanei. Remarkably, these six extracts exhibited higher 2,2-diphenyl-1-picrylhydrazyl radical scavenging activity and reducing power than the other extracts. Furthermore, the contents of total phenolic compounds and reducing sugars in the six selected extracts were also higher than those in the other extracts. The six selected AgNP samples showed strong surface plasmon resonance in the range of 416-438 nm. They were all spherical shaped with an average size from 12.5 ± 1.0 nm to 21.3 ± 4.9 nm as measured by field-emission transmission electron microscopy images. The hydrodynamic sizes were measured to be 49.5-122.6 nm with negative zeta potential values. Colloidal stability was excellent on the shelf for 28 days and in cell culture medium. The cytotoxicity assessment and generation of reactive oxygen species (ROS) in A549 and HeLa cells demonstrated that the AgNP samples prepared by Ardisia incarnata, Maesa calophylla, and Maesa laxiflora showed relatively high cytotoxicity and excess ROS generation among the six selected AgNP samples. Exposure of the AgNP samples to A549 and HeLa cells resulted in cell death, which was mostly due to necrosis but slightly due to late apoptosis. Cell cycle analysis demonstrated a significant increase in the cell population in the S phase. The green-synthesized AgNPs induced cell death, suggesting anticancer prospects that may offer new insight into the development of an anticancer nanomedicine.


Asunto(s)
Nanopartículas del Metal , Plata , Tecnología Química Verde , Células HeLa , Humanos , Parmeliaceae , Extractos Vegetales/farmacología
15.
Exp Mol Med ; 52(6): 877-886, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32514188

RESUMEN

Focal adhesion kinase (FAK) is an integrin-associated protein tyrosine kinase that is frequently overexpressed in advanced human cancers. Recent studies have demonstrated that aside from FAK's catalytic activity in cancer cells, its cellular localization is also critical for regulating the transcription of chemokines that promote a favorable tumor microenvironment (TME) by suppressing destructive host immunity. In addition to the protumor roles of FAK in cancer cells, FAK activity within cells of the TME may also support tumor growth and metastasis through various mechanisms, including increased angiogenesis and vascular permeability and effects related to fibrosis in the stroma. Small molecule FAK inhibitors have demonstrated efficacy in alleviating tumor growth and metastasis, and some are currently in clinical development phases. However, several preclinical trials have shown increased benefits from dual therapies using FAK inhibitors in combination with other chemotherapies or with immune cell activators. This review will discuss the role of nuclear FAK as a driver for tumor cell survival as well as potential therapeutic strategies to target FAK in both tumors and the TME.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Microambiente Tumoral/fisiología , Animales , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Microambiente Tumoral/genética
16.
J Biol Chem ; 294(29): 11213-11224, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31167784

RESUMEN

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long noncoding RNA overexpressed in various cancers that promotes cell growth and metastasis. Although hypoxia has been shown to up-regulate MALAT1, only hypoxia-inducible factors (HIFs) have been implicated in activation of the MALAT1 promoter in specific cell types and other molecular mechanisms associated with hypoxia-mediated MALAT1 up-regulation remain largely unknown. Here, we demonstrate that hypoxia induces cancer cell-specific chromatin-chromatin interactions between newly identified enhancer-like cis-regulatory elements present at the MALAT1 locus. We show that hypoxia-mediated up-regulation of MALAT1 as well as its antisense strand TALAM1 occurs in breast cancer cells, but not in nontumorigenic mammary epithelial cells. Our analyses on the MALAT1 genomic locus discovered three novel putative enhancers that are located upstream and downstream of the MALAT1 gene body. We found that parts of these putative enhancers are epigenetically modified to a more open chromatin state under hypoxia in breast cancer cells. Furthermore, our chromosome conformation capture experiment demonstrated that noncancerous cells and breast cancer cells exhibit different interaction profiles under both normoxia and hypoxia, and only breast cancer cells gain specific chromatin interactions under hypoxia. Although the HIF-2α protein can enhance the interaction between the promoter and the putative 3' enhancer, the gain of chromatin interactions associated with other upstream elements, such as putative -7 and -20 kb enhancers, were HIF-independent events. Collectively, our study demonstrates that cancer cell-specific chromatin-chromatin interactions are formed at the MALAT1 locus under hypoxia, implicating a novel mechanism of MALAT1 regulation in cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Hipoxia de la Célula , Cromatina/metabolismo , ARN Largo no Codificante/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Elementos de Facilitación Genéticos , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Regulación hacia Arriba
17.
Circ Res ; 125(2): 152-166, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31096851

RESUMEN

RATIONALE: Neointimal hyperplasia is characterized by excessive accumulation of vascular smooth muscle cells (SMCs) leading to occlusive disorders, such as atherosclerosis and stenosis. Blood vessel injury increases growth factor secretion and matrix synthesis, which promotes SMC proliferation and neointimal hyperplasia via FAK (focal adhesion kinase). OBJECTIVE: To understand the mechanism of FAK action in SMC proliferation and neointimal hyperplasia. METHODS AND RESULTS: Using combined pharmacological FAK catalytic inhibition (VS-4718) and SMC-specific FAK kinase-dead (Myh11-Cre-ERT2) mouse models, we report that FAK regulates SMC proliferation and neointimal hyperplasia in part by governing GATA4- (GATA-binding protein 4) cyclin D1 signaling. Inhibition of FAK catalytic activity facilitates FAK nuclear localization, which is required for proteasome-mediated GATA4 degradation in the cytoplasm. Chromatin immunoprecipitation identified GATA4 binding to the mouse cyclin D1 promoter, and loss of GATA4-mediated cyclin D1 transcription diminished SMC proliferation. Stimulation with platelet-derived growth factor or serum activated FAK and redistributed FAK from the nucleus to cytoplasm, leading to concomitant increase in GATA4 protein and cyclin D1 expression. In a femoral artery wire injury model, increased neointimal hyperplasia was observed in parallel with elevated FAK activity, GATA4 and cyclin D1 expression following injury in control mice, but not in VS-4718-treated and SMC-specific FAK kinase-dead mice. Finally, lentiviral shGATA4 knockdown in the wire injury significantly reduced cyclin D1 expression, SMC proliferation, and neointimal hyperplasia compared with control mice. CONCLUSIONS: Nuclear enrichment of FAK by inhibition of FAK catalytic activity during vessel injury blocks SMC proliferation and neointimal hyperplasia through regulation of GATA4-mediated cyclin D1 transcription.


Asunto(s)
Proliferación Celular , Ciclina D1/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Factor de Transcripción GATA4/metabolismo , Miocitos del Músculo Liso/metabolismo , Túnica Íntima/metabolismo , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Ciclina D1/genética , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Hiperplasia/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/fisiología , Túnica Íntima/patología
18.
Sci Rep ; 9(1): 7617, 2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31110200

RESUMEN

Protein tyrosine kinase (PTK) activity has been implicated in pro-inflammatory gene expression following tumor necrosis factor-α (TNF-α) or interkeukin-1ß (IL-1ß) stimulation. However, the identity of responsible PTK(s) in cytokine signaling have not been elucidated. To evaluate which PTK is critical to promote the cytokine-induced inflammatory cell adhesion molecule (CAM) expression including VCAM-1, ICAM-1, and E-selectin in human aortic endothelial cells (HAoECs), we have tested pharmacological inhibitors of major PTKs: Src and the focal adhesion kinase (FAK) family kinases - FAK and proline-rich tyrosine kinase (Pyk2). We found that a dual inhibitor of FAK/Pyk2 (PF-271) most effectively reduced all three CAMs upon TNF-α or IL-1ß stimulation compared to FAK or Src specific inhibitors (PF-228 or Dasatinib), which inhibited only VCAM-1 expression. In vitro inflammation assays showed PF-271 reduced monocyte attachment and transmigration on HAoECs. Furthermore, FAK/Pyk2 activity was not limited to CAM expression but was also required for expression of various pro-inflammatory molecules including MCP-1 and IP-10. Both TNF-α and IL-1ß signaling requires FAK/Pyk2 activity to activate ERK and JNK MAPKs leading to inflammatory gene expression. Knockdown of either FAK or Pyk2 reduced TNF-α-stimulated ERK and JNK activation and CAM expression, suggesting that activation of ERK or JNK is specific through FAK and Pyk2. Finally, FAK/Pyk2 activity is required for VCAM-1 expression and macrophage recruitment to the vessel wall in a carotid ligation model in ApoE-/- mice. Our findings define critical roles of FAK/Pyk2 in mediating inflammatory cytokine signaling and implicate FAK/Pyk2 inhibitors as potential therapeutic agents to treat vascular inflammatory disease such as atherosclerosis.


Asunto(s)
Quinasa 1 de Adhesión Focal/genética , Quinasa 2 de Adhesión Focal/genética , Expresión Génica/genética , Inflamación/genética , Interleucina-1beta/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Adhesión Celular/genética , Moléculas de Adhesión Celular/genética , Células Cultivadas , Citocinas/genética , Células Endoteliales/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/genética , Molécula 1 de Adhesión Celular Vascular/genética
19.
Arch Pharm Res ; 42(10): 926-934, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30972559

RESUMEN

The extract of Carpesium cernuum whole plant was successfully used as a green factory for the synthesis of silver nanoparticles in a one-step, one-pot process. The extract efficiently reduced silver ions to spherical silver nanoparticles. The size was measured as 13.0 ± 0.2 nm from high resolution transmission electron microscopic images. The reaction yield was determined to be 99.6% using inductively coupled plasma optical emission spectroscopy. The silver nanoparticles were highly stable for 28 days at ambient temperature without forming agglomeration or aggregation of nanoparticles. Dose-dependent antioxidant activity of the silver nanoparticles was observed in terms of the scavenging activity of 2,2-diphenyl-1-picrylhydrazyl radicals. The silver nanoparticles also exerted cytotoxicity on Mus musculus skin melanoma cells (B16F10) and human lung cancer cells (A549) in a dose-dependent manner. Specifically, the cytotoxicity of the silver nanoparticles on A549 cells was closely associated with apoptotic cell death. Cellular uptake of the silver was evaluated via inductively coupled plasma mass spectrometry, and a higher percentage of silver was taken up by A549 cells (22.6%) than by B16F10 cells (17.3%). This result indicated that higher cellular uptake of silver nanoparticles resulted in higher cytotoxicity on A549 cells. Therefore, plant extracts are capable of being valuable natural sources for the green synthesis of silver nanoparticles that exhibit potent biological activities for pharmaceutical and biomedical applications in future nanomedicine.


Asunto(s)
Antibacterianos/farmacología , Antineoplásicos Fitogénicos/farmacología , Antioxidantes/farmacología , Asteraceae/química , Tecnología Química Verde , Nanopartículas del Metal/química , Extractos Vegetales/farmacología , Plata/farmacología , Células A549 , Animales , Antibacterianos/síntesis química , Antibacterianos/química , Antineoplásicos Fitogénicos/síntesis química , Antineoplásicos Fitogénicos/química , Antioxidantes/síntesis química , Antioxidantes/química , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Tamaño de la Partícula , Picratos/antagonistas & inhibidores , Extractos Vegetales/síntesis química , Extractos Vegetales/química , Plata/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
20.
Mater Sci Eng C Mater Biol Appl ; 101: 204-216, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31029313

RESUMEN

We synthesized silver nanoparticles using thirty Chinese plant extracts via a green synthetic strategy. UV-visible spectra showed that the silver nanoparticles have an absorbance at 450 nm. Among the thirty extracts, seven extracts (Cratoxylum formosum, Phoebe lanceolata, Scurrula parasitica, Ceratostigma minus, Mucuna birdwoodiana, Myrsine africana and Lindera strychnifolia) exhibited the successful synthesis of silver nanoparticles. These seven extracts showed higher 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging activity and reducing power than the other extracts. The silver nanoparticles synthesized using these seven extracts were mostly spherical with high colloidal stability. The cytotoxicity of these seven silver nanoparticle samples on human lung cancer cells (A549) was clearly higher than that of the extracts alone. Furthermore, the cytotoxicity was affected by the presence or absence of fetal bovine serum. Moreover, the cytotoxicity of the silver nanoparticles synthesized with Cratoxylum formosum and Mucuna birdwoodiana extracts resulted in apoptotic cell death in A549 cells. The wound healing activity observed by the cell scratch method on mouse fibroblast cells (NIH3T3) suggested that the Lindera strychnifolia extract produced silver nanoparticles with decent activity. These results provide ample and systematic information for researchers on the green synthesis of silver nanoparticles using plant extracts.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Tecnología Química Verde/métodos , Nanopartículas del Metal/química , Extractos Vegetales/farmacología , Plata/farmacología , Cicatrización de Heridas/efectos de los fármacos , Células A549 , Animales , Compuestos de Bifenilo/química , Coloides/química , Depuradores de Radicales Libres/química , Humanos , Hidrodinámica , Nanopartículas del Metal/ultraestructura , Ratones , Células 3T3 NIH , Oxidación-Reducción , Tamaño de la Partícula , Picratos/química , Espectrofotometría Ultravioleta , Coloración y Etiquetado , Electricidad Estática
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA