Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Environ Toxicol ; 37(12): 2855-2864, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36052886

RESUMEN

Circular RNA (circRNA) has been confirmed to be involved in regulating sepsis-induced acute kidney injury (AKI). Our research aims to explore circ-ZNF644 role in the development of sepsis-induced AKI. Lipopolysaccharide (LPS) was used to induce kidney tubular epithelial cell (HK2) injury. ELISA assay was performed to measure the concentrations of inflammation factors. Cell functions were determined by cell counting kit 8 assay, EdU assay and flow cytometry. Protein expression was evaluated by Western blot analysis. Quantitative real-time PCR was used to detect relative expression of circ-ZNF644, miR-335-5p and homeodomain-interacting protein kinase 1 (HIPK1). RNA interaction was confirmed by dual-luciferase reporter assay and RIP assay. LPS enhanced HK2 cell inflammation, oxidative stress, apoptosis, and reduced proliferation. Circ-ZNF644 was overexpressed in sepsis-induced AKI patients. Circ-ZNF644 knockdown suppressed LPS-induced HK2 cell injury, and this effect could be revoked by miR-335-5p inhibitor. MiR-335-5p was sponged by circ-ZNF644, and its expression was downregulated in sepsis-induced AKI patients. HIPK1 was targeted by miR-335-5p, and its expression could be suppressed by circ-ZNF644 knockdown. MiR-335-5p had an inhibition effect on HK2 cell injury induced by LPS, and HIPK1 overexpression could reverse this effect. Circ-ZNF644 knockdown relieved LPS-induced HK2 cell injury through the miR-335-5p/HIPK1 axis, confirming that circ-ZNF644 contributed to sepsis-induced AKI.


Asunto(s)
Lesión Renal Aguda , MicroARNs , Proteínas Serina-Treonina Quinasas , ARN Circular , Sepsis , Humanos , Lesión Renal Aguda/genética , Apoptosis/genética , Proliferación Celular/genética , Regulación hacia Abajo , Inflamación , Lipopolisacáridos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Sepsis/genética , ARN Circular/genética
2.
Acta Biochim Biophys Sin (Shanghai) ; 53(6): 719-728, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-33856026

RESUMEN

Acute kidney injury (AKI) is a complex syndrome with an abrupt decrease of kidney function, which is associated with high morbidity and mortality. Sepsis is the common cause of AKI. Mounting evidence has demonstrated that long non-coding RNAs (lncRNAs) play critical roles in the development and progression of sepsis-induced AKI. In this study, we aimed to illustrate the function and mechanism of lncRNA SNHG14 in lipopolysaccharide (LPS)-induced AKI. We found that SNHG14 was highly expressed in the plasma of sepsis patients with AKI. SNHG14 inhibited cell proliferation and autophagy and promoted cell apoptosis and inflammatory cytokine production in LPS-stimulated HK-2 cells. Functionally, SNHG14 acted as a competing endogenous RNA (ceRNA) to negatively regulate miR-495-3p expression in HK-2 cells. Furthermore, we identified that HIPK1 is a direct target of miR-495-3p in HK-2 cells. We also revealed that the SNHG14/miR-495-3p/HIPK1 interaction network regulated HK-2 cell proliferation, apoptosis, autophagy, and inflammatory cytokine production upon LPS stimulation. In addition, we demonstrated that the SNHG14/miR-495-3p/HIPK1 interaction network regulated the production of inflammatory cytokines (TNF-α, IL-6, and IL-1ß) via modulating NF-κB/p65 signaling in LPS-challenged HK-2 cells. In conclusion, our findings suggested a novel therapeutic axis of SNHG14/miR-495-3p/HIPK1 to treat sepsis-induced AKI.


Asunto(s)
Lesión Renal Aguda/sangre , Lesión Renal Aguda/inducido químicamente , Células Epiteliales/metabolismo , Lipopolisacáridos/efectos adversos , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Largo no Codificante/sangre , Sepsis/sangre , Transducción de Señal/genética , Apoptosis/genética , Autofagia/genética , Estudios de Casos y Controles , Línea Celular Transformada , Proliferación Celular/genética , Citocinas/biosíntesis , Células Epiteliales/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Túbulos Renales/citología , MicroARNs/genética , Proteínas Serina-Treonina Quinasas/genética , ARN Largo no Codificante/genética , Sepsis/complicaciones , Transfección
3.
Biochim Biophys Acta ; 1853(7): 1564-73, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25820028

RESUMEN

The tumor suppressor protein programmed cell death 4 (Pdcd4) is a highly conserved RNA-binding protein that inhibits the translation of specific mRNAs. Here, we have identified the homeobox-interacting protein kinase-2 (Hipk2) mRNA as a novel translational target of Pdcd4. Unlike most other protein kinases Hipk2 is constitutively active after being synthesized by the ribosome and its expression and activity are thought to be mainly controlled by modulation of the half-life of the kinase. Our work provides the first evidence that Hipk2 expression is also controlled on the level of translation. We show that Hipk2 stimulates the translation of its own mRNA and that Pdcd4 suppresses the translation of Hipk2 mRNA by interfering with this auto-regulatory feedback mechanism. We also show that the translation of the related kinase Hipk1 is controlled by a similar feedback loop and that Hipk2 also stimulates the translation of Hipk1 mRNA. Taken together, our work describes a novel mechanism of translational suppression by Pdcd4 and shows for the first time that Hipk2 controls its own synthesis by an auto-regulatory feedback mechanism. Furthermore, the effect of Hipk2 on the translation of Hipk1 RNA suggests that Hipk2 and Pdcd4 can act in similar manner to control the translation of other mRNAs.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Portadoras/genética , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al ARN/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/química , Proteínas Portadoras/metabolismo , Línea Celular , Pollos , Factor 4A Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica , Humanos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Codorniz , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Relación Estructura-Actividad
4.
OMICS ; 28(3): 111-124, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38498023

RESUMEN

Homeodomain-interacting protein kinase 1 (HIPK1) is majorly found in the nucleoplasm. HIPK1 is associated with cell proliferation, tumor necrosis factor-mediated cellular apoptosis, transcription regulation, and DNA damage response, and thought to play significant roles in health and common diseases such as cancer. Despite this, HIPK1 remains an understudied molecular target. In the present study, based on a systematic screening and mapping approach, we assembled 424 qualitative and 44 quantitative phosphoproteome datasets with 15 phosphosites in HIPK1 reported across multiple studies. These HIPK1 phosphosites were not currently attributed to any functions. Among them, Tyr352 within the kinase domain was identified as the predominant phosphosite modulated in 22 differential datasets. To analyze the functional association of HIPK1 Tyr352, we first employed a stringent criterion to derive its positively and negatively correlated protein phosphosites. Subsequently, we categorized the correlated phosphosites in known interactors, known/predicted kinases, and substrates of HIPK1, for their prioritized validation. Bioinformatics analysis identified their significant association with biological processes such as the regulation of RNA splicing, DNA-templated transcription, and cellular metabolic processes. HIPK1 Tyr352 was also identified to be upregulated in Her2+ cell lines and a subset of pancreatic and cholangiocarcinoma tissues. These data and the systems biology approach undertaken in the present study serve as a platform to explore the functional role of other phosphosites in HIPK1, and by extension, inform cancer drug discovery and oncotherapy innovation. In all, this study highlights the comprehensive phosphosite map of HIPK1 kinase and the first of its kind phosphosite-centric analysis of HIPK1 kinase based on global-level phosphoproteomics datasets derived from human cellular differential experiments across distinct experimental conditions.


Asunto(s)
Neoplasias , Proteínas Serina-Treonina Quinasas , Humanos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Transcripción Genética , Fosforilación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo
5.
Adv Sci (Weinh) ; 10(18): e2300585, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37098980

RESUMEN

Inhibition of pathological cardiac hypertrophy is recognized as an important therapeutic strategy for heart failure, although effective targets are still lacking in clinical practice. Homeodomain interacting protein kinase 1 (HIPK1) is a conserved serine/threonine kinase that can respond to different stress signals, however, whether and how HIPK1 regulates myocardial function is not reported. Here, it is observed that HIPK1 is increased during pathological cardiac hypertrophy. Both genetic ablation and gene therapy targeting HIPK1 are protective against pathological hypertrophy and heart failure in vivo. Hypertrophic stress-induced HIPK1 is present in the nucleus of cardiomyocytes, while HIPK1 inhibition prevents phenylephrine-induced cardiomyocyte hypertrophy through inhibiting cAMP-response element binding protein (CREB) phosphorylation at Ser271 and inactivating CCAAT/enhancer-binding protein ß (C/EBPß)-mediated transcription of pathological response genes. Inhibition of HIPK1 and CREB forms a synergistic pathway in preventing pathological cardiac hypertrophy. In conclusion, HIPK1 inhibition may serve as a promising novel therapeutic strategy to attenuate pathological cardiac hypertrophy and heart failure.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Insuficiencia Cardíaca , Humanos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Cardiomegalia/prevención & control , Cardiomegalia/genética , Miocitos Cardíacos , Proteínas Serina-Treonina Quinasas/metabolismo , Insuficiencia Cardíaca/metabolismo
6.
Biofactors ; 48(1): 135-147, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34856026

RESUMEN

Noncoding RNAs (ncRNAs) such as microRNAs (miRNAs), long noncoding RNAs (lncRNA), and circular RNAs are closely related to the biological processes related to obesity. As a miRNA that widely present in different cell types, miR497 is proved to be involved in cell development. However, research on the role of miR-497 as a key factor in regulating the development of adipocytes is still in gap. The role of miR-497 in the apoptosis and proliferation of mouse-derived adipocytes was detected by RNA-seq analysis, RT-qPCR, Western blot, immunofluorescence, and dual-luciferase reporter assay. Using miR-497 mimics to treat 3T3-L1 cells, we found that miR-497 targeted Bcl-2 to promote adipocyte apoptosis through the mitochondrial pathway, and this effect was consistent in the apoptosis model composed of palmitic acid (PA) and hydrogen peroxide (H2 O2 ). LncRNA homeodomain-interacting protein kinase 1 (lnc-hipk1) sponged miR-148b to weaken its silencing of Bcl-2, forming the competitive endogenous RNAs (CeRNAs) regulatory network. Furthermore, overexpression of lnc-hipk1 inhibited the apoptosis of adipocytes by targeting miR-497/Bcl-2. Co-treatment of miR-497 and lnc-hipk1 showed that lnc-hipk1 reversed the apoptosis of adipocytes caused by miR-497 overexpression. And in vivo experiments further confirmed that this effect was also achieved by the CeRNA system of lnc-hipk1/miR-497/Bcl-2. In summary, lnc-hipk1 targets miR-497/Bcl-2 to regulate adipocyte apoptosis through the mitochondrial pathway. This research enriches the research content of ncRNAs and CeRNA in adipocyte development, and provides new targets for the treatment of obesity and other metabolic syndromes.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Apoptosis/genética , Proliferación Celular/genética , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
7.
G3 (Bethesda) ; 11(12)2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34849772

RESUMEN

Homeodomain-interacting protein kinases (HIPKs) are a family of four conserved proteins essential for vertebrate development, as demonstrated by defects in the eye, brain, and skeleton that culminate in embryonic lethality when multiple HIPKs are lost in mice. While HIPKs are essential for development, functional redundancy between the four vertebrate HIPK paralogues has made it difficult to compare their respective functions. Because understanding the unique and shared functions of these essential proteins could directly benefit the fields of biology and medicine, we addressed the gap in knowledge of the four vertebrate HIPK paralogues by studying them in the fruit fly Drosophila melanogaster, where reduced genetic redundancy simplifies our functional assessment. The single hipk present in the fly allowed us to perform rescue experiments with human HIPK genes that provide new insight into their individual functions not easily assessed in vertebrate models. Furthermore, the abundance of genetic tools and established methods for monitoring specific developmental pathways and gross morphological changes in the fly allowed for functional comparisons in endogenous contexts. We first performed rescue experiments to demonstrate the extent to which each of the human HIPKs can functionally replace Drosophila Hipk for survival and morphological development. We then showed the ability of each human HIPK to modulate Armadillo/ß-catenin levels, JAK/STAT activity, proliferation, growth, and death, each of which have previously been described for Hipks, but never all together in comparable tissue contexts. Finally, we characterized novel developmental phenotypes induced by human HIPKs to gain insight to their unique functions. Together, these experiments provide the first direct comparison of all four vertebrate HIPKs to determine their roles in a developmental context.


Asunto(s)
Drosophila melanogaster , Proteínas de Homeodominio , Proteínas Quinasas , Animales , Animales Modificados Genéticamente , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Fosforilación , Proteínas Quinasas/metabolismo
8.
N Biotechnol ; 54: 28-33, 2020 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-31425885

RESUMEN

Stable cell lines can continuously produce a recombinant protein without the need to repeatedly engineer the genome. In a previous study HIPK1, Homeodomain-interacting Protein Kinase 1, was found to be a target of the microRNA miR-22 that, when repressed, improved expression of both an intracellular and a secreted protein. In this report, HEK293 cells stably over-expressing miR-22 were compared with HEK293 with knockout of HIPK1, executed by CRISPR/Cas9, for their ability to improve recombinant protein expression. In this model case of luciferase, over-expression of miR-22 improved overall activity 2.4-fold while the HIPK1 knockout improved overall activity 4.7-fold.


Asunto(s)
MicroARNs/genética , Biosíntesis de Proteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Expresión Génica , Técnicas de Inactivación de Genes , Células HEK293 , Humanos
9.
Theranostics ; 8(21): 5801-5813, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30613263

RESUMEN

Increasing evidence demonstrates the existence of two inter-convertible states of breast cancer stem cells (BCSCs) with distinct behaviors in proliferation and mobility, and the BCSC heterogeneity is accurately regulated by sophisticated mechanisms including microRNAs. The microRNA-200 family including miR-200c/141 cluster was reported to affect cancer cell invasion and metastasis by regulating epithelial to mesenchymal transition (EMT). However, the effect of miR-200 family on BCSC heterogeneity is uncertain. Thus, we investigated whether the miR-200c/141 cluster had different effects on breast tumor growth and metastasis by switching the two states of BCSC. Methods: The spontaneous mammary tumor mouse model with miR-200c/141 conditional knockout was utilized for analyzing the role of miR-200c/141 cluster in vivo. The effect of miR-200c/141 cluster on BCSCs was performed by CD24/CD29 staining and ALDEFLUOR assay. miR-200c/141 target expression and EMT-related marker expression were verified in tumor sections, primary cells and breast cancer cell lines by qRT-PCR or western blotting. Statistical analysis was determined using two-way ANOVA and Student's t-test. All values were presented as the mean ± s.e.m. Results: The deletion of miR-200c/141 cluster regulated BCSC heterogeneity and promoted the EMT-like BCSC generation, which resulted in increased tumor metastasis and inhibited tumor growth by directly upregulating the target gene homeodomain-interacting protein kinase 1 (HIPK1) and sequential ß-catenin activation. Conclusions: Our results indicated that miR-200c/141 played biphasic roles in breast tumor progression via affecting the BCSC heterogeneity, suggesting targeting BCSC heterogeneity to simultaneously restrict breast cancer initiation and metastasis could be a promising therapeutic strategy for breast cancer.


Asunto(s)
Proteínas Portadoras/metabolismo , Neoplasias Mamarias Animales/patología , MicroARNs/metabolismo , Células Madre Neoplásicas/fisiología , Mapas de Interacción de Proteínas , Proteínas Quinasas/metabolismo , beta Catenina/metabolismo , Animales , Western Blotting , Antígeno CD24/análisis , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Integrina beta1/análisis , Ratones , MicroARNs/genética , Células Madre Neoplásicas/química , Proteínas Serina-Treonina Quinasas , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Biotechnol J ; 13(2)2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28987030

RESUMEN

Protein expression from human embryonic kidney cells (HEK 293) is an important tool for structural and clinical studies. It is previously shown that microRNAs (small, noncoding RNAs) are effective means for improved protein expression from these cells, and by conducting a high-throughput screening of the human microRNA library, several microRNAs are identified as potential candidates for improving expression. From these, miR-22-3p is chosen for further study since it increased the expression of luciferase, two membrane proteins and a secreted fusion protein with minimal effect on the cells' growth and viability. Since each microRNA can interact with several gene targets, it is of interest to identify the repressed genes for understanding and exploring the improved expression mechanism for further implementation. Here, the authors describe a novel approach for identification of the target genes by integrating the differential gene expression analysis with information obtained from our previously conducted high-throughput siRNA screening. The identified genes were validated as being involved in improving luciferase expression by using siRNA and qRT-PCR. Repressing the target gene, HIPK1, is found to increase luciferase and GPC3 expression 3.3- and 2.2-fold, respectively.


Asunto(s)
MicroARNs/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , ARN Interferente Pequeño/genética , Proliferación Celular , Supervivencia Celular , Células HEK293 , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Análisis por Micromatrices , Proteínas Serina-Treonina Quinasas/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Reproducibilidad de los Resultados , Transfección
11.
Microrna ; 7(3): 178-186, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29793420

RESUMEN

The Homeodomain-Interacting Protein Kinases (HIPKs) HIPK1, HIPK2 and HIPK3 are Ser/Thr kinases which interact with homeobox proteins and other transcription factors, acting as transcriptional coactivators or corepressors. HIPKs contribute to regulate several biological processes, such as signal transduction, apoptosis, embryonic development, DNA-damage response, and cellular proliferation, in response to various extracellular stimuli. Recently it has emerged that, in addition to their role in cancer, fibrosis and diabetes, HIPKs may also be involved in other human diseases, including Amyotrophic Lateral Sclerosis (ALS), Rett syndrome, cerebellar diseases, and retinal vascular dysfunction. METHODS: Here, we update our previous paper concerning the regulation of HIPK proteins expression by microRNAs (miRNAs), pointing out the most recent findings about new cellular mechanisms and diseases which are affected by the interplay between HIPKs and miRNAs. CONCLUSION: Recently, it has emerged that HIPKs and their related miRNAs are involved in diabetic nephropathy, gastric cancer chemoresistance, cervical cancer progression, and recombinant protein expression in cultured cells. Interestingly, circular RNAs (circRNAs) deriving from HIPK2 and HIPK3 loci also modulate cellular proliferation and viability by sponging several miRNAs, thus emerging as new putative therapeutic targets for diabetes-associated retinal vascular dysfunction, astrogliosis and cancer.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MicroARNs/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Biomarcadores de Tumor/genética , Proteínas Portadoras/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
12.
J Mol Biol ; 429(1): 64-78, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27884605

RESUMEN

Cyclic AMP-response element-binding protein (CREB) plays key transcriptional roles in cell metabolism, proliferation, and survival. Ser133 phosphorylation by protein kinase A (PKA) is a well-characterized CREB activation mechanism. Homeodomain-interacting protein kinase (HIPK) 2, a nuclear serine/threonine kinase, activates CREB through Ser271 phosphorylation; however, the regulatory mechanism remains uncharacterized. Transfection of CREB in HEK293 cells together with the kinase demonstrated that HIPK2 phosphorylated CREB at Ser271 but not Ser133; likewise, PKA phosphorylated CREB at Ser133 but not Ser271, suggesting two distinct CREB regulatory mechanisms by HIPK2 and PKA. In vitro kinase assay revealed that HIPK2, and HIPK1 and HIPK3, directly phosphorylated CREB. Cells exposed to 10µM sodium arsenite increased the stability of HIPK1 and HIPK2 proteins, leading to CREB activation via Ser271 phosphorylation. Phospho-Ser271 CREB showed facilitated interaction with the TFIID subunit coactivator TAF4 assessed by immunoprecipitation. Furthermore, a focused gene array between cells transfected with CREB alone and CREB plus HIPK2 over empty vector-transfected control displayed 14- and 32-fold upregulation of cyclin A1, respectively, while no upregulation was displayed by HIPK2 alone. These results suggest that the HIPK2-phospho-Ser271 CREB axis is a new arsenic-responsive CREB activation mechanism in parallel with the PKA-phospho-Ser133 CREB axis.


Asunto(s)
Arsénico/metabolismo , Proteínas Portadoras/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Activación Transcripcional , Línea Celular , Perfilación de la Expresión Génica , Humanos , Fosforilación , Serina/metabolismo
13.
Biochim Biophys Acta Gene Regul Mech ; 1860(7): 751-760, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28495617

RESUMEN

LIM-domain proteins, containing multiple cysteine-rich zinc finger-like motifs, have been shown to play diverse roles in several cellular processes. A common theme is that they mediate important protein-protein interactions that are key to their function. Androgen receptor-associated protein 55 (ARA55) belongs to this family of bridging proteins containing four C-terminal LIM domains. It has a dual role with functions both at focal adhesions and in the nucleus, apparently shuttling between the two compartments. In the present work, we have expanded our understanding of its nuclear functions by showing that it interacts with three nuclear regulators not previously linked to ARA55. We first identified ARA55 as a novel interaction partner of the nuclear kinase HIPK1 and found that ARA55, like HIPK1, also interacts with the transcription factor c-Myb. In search of a function for these associations, we observed that the coactivator p300 not only binds to c-Myb, but to ARA55 as well. When combined, c-Myb, p300, HIPK1 and ARA55 caused strong synergistic activation of a chromatinized reporter gene. In parallel, all partners, including p300, were efficiently recruited to chromatin at the c-Myb-bound promoter. Consistent with this cooperation, we found that c-Myb and ARA55 share a common set of target genes in an osteosarcoma cellular context. We propose that ARA55 and HIPK1 assist c-Myb in recruiting the coactivator and acetyltransferase p300 to chromatin.


Asunto(s)
Cromatina/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Animales , Células COS , Línea Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Chlorocebus aethiops , Proteínas de Unión al ADN/metabolismo , Genes Reporteros/genética , Células HEK293 , Humanos , Células K562 , Regiones Promotoras Genéticas/genética , Unión Proteica/genética
14.
Cell Cycle ; 12(12): 1879-91, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23676219

RESUMEN

HIPK1 (homeodomain interacting protein kinase 1) is a serine/threonine kinase that belongs to the CMGC superfamily. Emerging data point to the role of HIPK1 in cancer, but it is still not clear whether it acts as a tumor suppressor or promoter. Here we identified HIPK1 as a kinase that is significantly overexpressed in colorectal cancer (CRC) and whose expression is stage-dependent. Being abundantly expressed at the onset of the disease, the HIPK1 level gradually decreased as tumor stage progressed. To further uncover how this factor regulates tumorigenesis and establish whether it constitutes an early factor necessary for neoplastic transformation or for cellular defense, we studied the effect of its overexpression in vitro by investigating various cancer-related signaling cascades. We found that HIPK1 mostly regulates the p53 signaling pathway both in HCT116 and HeLa cells. By phosphorylating p53 on its serine-15, HIPK1 favored its transactivation potential, which led to a rise in p21 protein level and a decline in cell proliferation. Assuming that HIPK1 could impede CRC growth by turning on the p53/p21 pathway, we then checked p21 mRNA levels in patients. Interestingly, p21 transcripts were only increased in a subset of patients expressing high levels of HIPK1. Unlike the rest of the cohort, the majority of these patients hosted a native p53 protein, meaning that such a pro-survival pathway (HIPK1+ > p53 > p21) is active in patients, and that HIPK1 acts rather as a tumor suppressor.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Western Blotting , Neoplasias Colorrectales/genética , Células HCT116 , Células HeLa , Humanos , Inmunoprecipitación , Técnicas In Vitro , Proteínas Serina-Treonina Quinasas/genética , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda