Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Biol Chem ; 298(6): 101926, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35413288

RESUMO

Skeletal muscle dynamically regulates systemic nutrient homeostasis through transcriptional adaptations to physiological cues. In response to changes in the metabolic environment (e.g., alterations in circulating glucose or lipid levels), networks of transcription factors and coregulators are recruited to specific genomic loci to fine-tune homeostatic gene regulation. Elucidating these mechanisms is of particular interest as these gene regulatory pathways can serve as potential targets to treat metabolic disease. The zinc-finger transcription factor Krüppel-like factor 15 (KLF15) is a critical regulator of metabolic homeostasis; however, its genome-wide distribution in skeletal muscle has not been previously identified. Here, we characterize the KLF15 cistrome in vivo in skeletal muscle and find that the majority of KLF15 binding is localized to distal intergenic regions and associated with genes related to circadian rhythmicity and lipid metabolism. We also identify critical interdependence between KLF15 and the nuclear receptor PPARδ in the regulation of lipid metabolic gene programs. We further demonstrate that KLF15 and PPARδ colocalize genome-wide, physically interact, and are dependent on one another to exert their transcriptional effects on target genes. These findings reveal that skeletal muscle KLF15 plays a critical role in metabolic adaptation through its direct actions on target genes and interactions with other nodal transcription factors such as PPARδ.


Assuntos
Fatores de Transcrição Kruppel-Like , Metabolismo dos Lipídeos , Músculo Esquelético , PPAR delta , Animais , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Metabolismo dos Lipídeos/genética , Camundongos , Músculo Esquelético/metabolismo , PPAR delta/genética , PPAR delta/metabolismo
2.
J Appl Toxicol ; 43(11): 1748-1760, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37408164

RESUMO

Lysine-specific demethylase 1 (LSD1) inhibitors are being developed for cancer therapy, but their bioeffects on vasculatures are not clear. In this study, we compared the influences of ORY-1001 (an LSD1 inhibitor being advanced into clinical trials) and 199 (a novel LSD1 inhibitor recently developed by us) to human umbilical vein endothelial cells (HUVECs) in vitro and further verified the bioeffects of ORY-1001 to zebrafish (Danio rerio) larvae in vivo. The results showed that up to 10 µM ORY-1001 or 199 did not significantly affect the cellular viability of HUVECs but substantially reduced the release of inflammatory interleukin-8 (IL-8) and IL-6. The signaling molecule in vasculatures, NO, was also increased in HUVECs. As the mechanism, the protein levels of endothelial NO synthase (eNOS) or p-eNOS, and their regulators Kruppel-like factor 2 (KLF2) or KLF4, were also increased after drug treatment. In vivo, 24 h treatment with up to 100 nM ORY-1001 reduced blood speed without changing morphologies or locomotor activities in zebrafish larvae. ORY-1001 treatment reduced the expression of il8 but promoted the expression of klf2a and nos in the zebrafish model. These data show that LSD1 inhibitors were not toxic but capable to inhibit inflammatory responses and affect the function of blood vessels through the up-regulation of the NOS-KLF pathway.

3.
J Biol Chem ; 297(2): 100932, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34217701

RESUMO

A complex network of transcription factors regulates genes involved in establishing and maintaining key biological properties of the human airway epithelium. However, detailed knowledge of the contributing factors is incomplete. Here we characterize the role of Krüppel-like factor 5 (KLF5), in controlling essential pathways of epithelial cell identity and function in the human lung. RNA-seq following siRNA-mediated depletion of KLF5 in the Calu-3 lung epithelial cell line identified significant enrichment of genes encoding chemokines and cytokines involved in the proinflammatory response and also components of the junctional complexes mediating cell adhesion. To determine direct gene targets of KLF5, we defined the cistrome of KLF5 using ChIP-seq in both Calu-3 and 16HBE14o- lung epithelial cell lines. Occupancy site concordance analysis revealed that KLF5 colocalized with the active histone modification H3K27ac and also with binding sites for the transcription factor CCAAT enhancer-binding protein beta (C/EBPß). Depletion of KLF5 increased both the expression and secretion of cytokines including IL-1ß, a response that was enhanced following exposure to Pseudomonas aeruginosa lipopolysaccharide. Calu-3 cells exhibited faster rates of repair after KLF5 depletion compared with negative controls in wound scratch assays. Similarly, CRISPR-mediated KLF5-null 16HBE14o- cells also showed enhanced wound closure. These data reveal a pivotal role for KLF5 in coordinating epithelial functions relevant to human lung disease.


Assuntos
Células Epiteliais , Imunidade Inata , Fatores de Transcrição Kruppel-Like , Linhagem Celular , Humanos
4.
J Appl Toxicol ; 42(4): 651-659, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34633093

RESUMO

Recently, we reported that titanium dioxide (TiO2 ) materials activated endothelial cells via Kruppel-like factor (KLF)-mediated nitric oxide (NO) dysfunction, but the roles of physical properties of materials are not clear. In this study, we prepared nanobelts from P25 particles and compared their adverse effects to human umbilical vein endothelial cells (HUVECs). TiO2 nanobelts had belt-like morphology but comparable surface areas as P25 particles. When applied to HUVECs, P25 particles or nanobelts did not induce cytotoxicity, although nanobelts were much more effective to increase intracellular Ti element concentrations compared the same amounts of P25 particles. Only nanobelts significantly induced THP-1 adhesion onto HUVECs. Consistently, nanobelts were more significant to induce the expression of intracellular adhesion molecule-1 (ICAM1) and the release of soluble ICAM-1 (sICAM-1), indicating that nanobelts were more potent to induce endothelial activation in vitro. As the mechanisms for endothelial activation, both P25 and nanobelts reduced the generation of intracellular NO as well as the expression of NO regulators KLF2 and KLF4. Combined, the results from this study indicated that the different morphologies of P25 particles and nanobelts only changed their internalization into HUVECs but showed minimal impact on KLF-mediated NO signaling pathways.


Assuntos
Fatores de Transcrição Kruppel-Like , Óxido Nítrico , Células Endoteliais da Veia Umbilical Humana , Humanos , Fatores de Transcrição Kruppel-Like/genética , Óxido Nítrico/metabolismo , Transdução de Sinais
5.
J Appl Toxicol ; 41(9): 1467-1478, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33438233

RESUMO

Morphology plays a vital role in determining the biological effects of silica nanoparticles (NPs), but its influence on the toxicity of silica NPs in endothelial cells (ECs) is still inconclusive. We synthesized five kinds of Santa Barbara 15 amorphous (SBA-15) particles with different shapes and added them to human umbilical vein endothelial cells (HUVEC). After 24 After incubation and treatment with 100 ml, more than 80% of the cells are still alive. The microgram/ml of SBA-15 indicates that SBA-15 has high biocompatibility. Fibrous SBA-15 (5) leads to the highest Si element concentration in HUVEC. No NP reduces the release of NO, and NO is an important signaling molecule in the vascular system. Only the aggregated spherical SBA-15 (3) will moderately reduce the endothelial nitric oxide synthase (eNOS) protein. Regarding transcription factors regulating eNOS, we found that all SBA-15 types significantly increased Kruppel-like factor 2 (KLF2) protein, irregular SBA-15 (1), non-aggregated spherical SBA-15 (2) and aggregation The spherical SBA-15 (3) greatly reduces KLF4 by more than 50%. Overall, our results indicate that SBA-15 with different morphologies can be internalized into HUVEC and only cause moderate cytotoxicity. All silica NPs have the smallest effect on the NO-eNOS pathway, but the irregular spherical SBA-15 reduces the eNOS modifier KLF4. The rod-shaped SBA-15 (4) seems to have higher biocompatibility because they are internalized and have negligible adverse effects on HUVEC. These results provide new evidence for the toxic effects of different forms of silica nanoparticles on HUVEC.


Assuntos
Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Nanopartículas/toxicidade , Dióxido de Silício/toxicidade , Sobrevivência Celular , Humanos , Fator 4 Semelhante a Kruppel/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Teste de Materiais , Microscopia Eletrônica de Varredura , Nanopartículas/química , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais/efeitos dos fármacos , Dióxido de Silício/química , Termodinâmica , Difração de Raios X
6.
J Biol Chem ; 293(36): 14012-14021, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30026232

RESUMO

Adipose tissue stores energy and plays an important role in energy homeostasis. CCAAT/enhancer-binding protein ß (C/EBPß) is an important early transcription factor for 3T3-L1 preadipocyte differentiation, facilitating mitotic clonal expansion (MCE) and transactivating C/EBPα and peroxisome proliferator-activated receptor-γ (PPARγ) to promote adipogenesis. C/EBPß is induced early, but the expression of antimitotic C/EBPα and PPARγ is not induced until ∼48 h. The delayed expression of C/EBPα and PPARγ is thought to ensure MCE progression, but the molecular mechanism for this delay remains elusive. Here, we show that the zinc-finger transcription factor Krüppel-like factor 10 (KLF10) is induced after adipogenic induction and that its expression positively correlates with that of C/EBPß but inversely correlates with expression of C/EBPα and PPARγ. C/EBPß bound to the KLF10 promoter and transactivated its expression during MCE. KLF10 overexpression in 3T3-L1 preadipocyte repressed adipogenesis and decreased C/EBPα and PPARγ expression, whereas siRNA-mediated down-regulation of KLF10 enhanced adipogenesis and increased C/EBPα and PPARγ expression. Luciferase assays revealed an inhibitory effect of KLF10 on C/EBPα promoter activity. Using promoter deletion and mutation analysis, we identified a KLF10-binding site within the proximal promoter region of C/EBPα. Furthermore, KLF10 interacted with and recruited histone deacetylase 1 (HDAC1) to the C/EBPα promoter, decreasing acetylated histone H4 on the C/EBPα promoter and inactivating C/EBPα transcription. Because C/EBPα can transactivate PPARγ, our results suggest a mechanism by which expression of C/EBPα and PPARγ is delayed via KLF10 expression and shed light on the negative feedback loop for C/EBPß-regulated adipogenesis in 3T3-L1 preadipocyte.


Assuntos
Adipogenia , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Fatores de Transcrição de Resposta de Crescimento Precoce/genética , Fatores de Transcrição Kruppel-Like/genética , Ativação Transcricional , Células 3T3-L1 , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/antagonistas & inibidores , Diferenciação Celular , Fatores de Transcrição de Resposta de Crescimento Precoce/metabolismo , Retroalimentação Fisiológica , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , PPAR gama/metabolismo , Fatores de Tempo
7.
Toxicol Appl Pharmacol ; 374: 11-19, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31047983

RESUMO

It has been shown before that exposure to nanomaterials (NMs) might promote the formation of foam cells, the key cells involved in all stages of atherosclerosis. However, to our best knowledge, previous studies, particularly in vitro studies, only investigated the transformation of macrophages into foam cells, whereas the importance of smooth muscle cells (SMCs) was overlooked. The present study investigated the toxicity of pristine multi-walled carbon nanotubes (MWCNTs; Code XFM19) and carboxylated MWCNTs (Code XFM21) to human aortic smooth muscle cells (HASMCs). The results showed that exposure to both types of MWCNTs significantly reduced mitochondrial activity but might not damage lysosomes. MWCNT exposure had minimal impact on cytokine release but significantly promoted lipid accumulation, which was significantly inhibited when the cells were pre-incubated with ER stress inhibitors or antioxidants. The mRNA levels of ER stress markers DDIT3 and XBP-1 s and protein levels of chop and p-chop were induced particularly by XFM21, accompanying with increased SREBF1 and SREBF2 mRNA as well as FASN protein, the key regulators involved in de novo lipogenesis. In addition, the mRNA levels of KLF4 and KLF5 and protein levels of KLF were induced after exposure to both types of MWCNTs, associated with an increase of CD68 protein levels. We concluded that MWCNTs might promote lipid accumulation in HASMCs through the induction of ER stress leading to de novo lipogenesis, as well as the activation of KLF pathway resulting in SMC transformation.


Assuntos
Aorta/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Nanotubos de Carbono/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Glutationa , Humanos , Fator 4 Semelhante a Kruppel , Metabolismo dos Lipídeos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio
8.
Toxicol Appl Pharmacol ; 382: 114758, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31521728

RESUMO

Gold (Au) nanomaterials (NMs), particularly those with PEG surface functionalization, are generally considered to be biocompatible for biomedical applications due to relatively low cytotoxicity. Herein, we investigated the toxicity of PEGylated Au nanorods (NRs) to human umbilical vein endothelial cells (HUVECs), a commonly used in vitro model for human endothelium. We found a previously unknown effect that up to 10 µg/mL Au NRs, albeit not cytotoxic, decreased the mRNA and protein levels of kruppel-like factor 2 (KLF2), a transcription factor with well-documented vasoprotective effects. The results from PCR array showed that a number of genes associated with risk of cardiovascular diseases were altered by Au NRs, and several genes are downstream genes of KLF2 according to ingenuity pathway analysis (IPA). These effects could be observed with or without the presence of inflammatory stimuli lipopolysaccharide (LPS), which suggests a pre-existing inflammatory state is not required for Au NRs to alter KLF2 signaling pathway. We further identified that Au NRs significantly decreased eNOS mRNA/p-eNOS proteins as well as increased MCP-1 mRNA/sMCP-1 release, which are targets of KLF2. Combined, our data revealed a novel pathway that PEGylated Au NPs at non-cytotoxic concentrations might alter KLF leading to the increase of risk of cardiovascular diseases in human endothelial cells. Given the importance of KLF in vascular homeostasis, our data indicate that it is necessary to evaluate the influence of engineered NPs to KLF signaling pathways, especially for NPs with biomedical uses.


Assuntos
Ouro/toxicidade , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Nanotubos/toxicidade , Polietilenoglicóis/toxicidade , Transdução de Sinais/efeitos dos fármacos , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Transdução de Sinais/fisiologia , Transcriptoma/efeitos dos fármacos , Transcriptoma/fisiologia
9.
J Biol Chem ; 291(40): 21271-21282, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27539853

RESUMO

Macrophages are the predominant innate immune cells recruited to tissues following injury or infection. These early-responding, pro-inflammatory macrophages play an essential role in the amplification of inflammation. However, macrophage pro-inflammatory gene expression should be tightly regulated to avert host tissue damage. In this study, we identify the Kruppel-like transcription factor 6 (KLF6)-B cell leukemia/lymphoma 6 (BCL6) signaling axis as a novel regulator of macrophage inflammatory gene expression and function. Utilizing complementary gain- and loss-of-function studies, we observed that KLF6 is essential for macrophage motility under ex vivo and in vivo conditions. Concordant with these observations, myeloid-specific deficiency of KLF6 significantly attenuates macrophage pro-inflammatory gene expression, recruitment, and progression of inflammation. At the molecular level, KLF6 suppresses BCL6 mRNA and protein expression by elevating PR domain-containing 1 with ZNF domain (PRDM1) levels in macrophages. Interestingly, pharmacological or genetic inhibition of BCL6 in KLF6-deficient macrophages completely abrogated the attenuation of pro-inflammatory cytokine/chemokine expression and cellular motility. Collectively, our observations reveal that KLF6 repress BCL6 to enhance macrophage inflammatory gene expression and function.


Assuntos
Quimiocinas/biossíntese , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , Macrófagos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/biossíntese , Proteínas Proto-Oncogênicas/metabolismo , Animais , Células Cultivadas , Quimiocinas/genética , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Fator 6 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-6/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
J Biol Chem ; 291(37): 19374-86, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27451392

RESUMO

Glucocorticoid receptor (GR) signaling has recently been shown to play a direct role in the regulation of cardiomyocyte function. In this study, we investigated the potential role of KLF13 as a downstream effector of GR action utilizing both in vivo and in vitro approaches. Our data show that KLF13 mRNA and protein levels are significantly diminished in the hearts of mice lacking GR in cardiomyocytes. Glucocorticoid administration up-regulated Klf13 mRNA in the mouse heart, in isolated primary cardiomyocytes, and in immortal cardiomyocyte cell lines. Glucocorticoid Klf13 gene expression was abolished by treatment with a GR antagonist (RU486) or by knockdown of GR in cardiomyocytes. Moreover, glucocorticoid induction of Klf13 mRNA was resistant to de novo protein synthesis inhibition, demonstrating that Klf13 is a direct glucocorticoid receptor gene target. A glucocorticoid responsive element (GRE) was identified in the Klf13 gene and its function was verified by chromatin immunoprecipitation in HL-1 cells and mouse hearts. Functional studies showed that GR regulation of Klf13 is critical to protect cardiomyocytes from DNA damage and cell death induced by cobalt(II) chloride hexahydrate (CoCl2·6H2O) and the antineoplastic drug doxorubicin. These results established a novel role for GR and KLF13 signaling in adult cardiomyocytes with potential clinical implications for the prevention of cardiotoxicity induced heart failure.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA , Insuficiência Cardíaca/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Animais , Proteínas de Ciclo Celular/genética , Morte Celular/efeitos dos fármacos , Cobalto/efeitos adversos , Cobalto/farmacologia , Doxorrubicina/efeitos adversos , Doxorrubicina/farmacologia , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Transgênicos , Miocárdio/patologia , Miócitos Cardíacos/patologia , Receptores de Glucocorticoides/genética , Proteínas Repressoras/genética , Elementos de Resposta
11.
J Biol Chem ; 291(35): 18084-95, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27402841

RESUMO

Regenerative medicine holds great promise for the treatment of degenerative retinal disorders. Krüppel-like factors (KLFs) are transcription factors that have recently emerged as key tools in regenerative medicine because some of them can function as epigenetic reprogrammers in stem cell biology. Here, we show that KLF16, one of the least understood members of this family, is a POU4F2 independent transcription factor in retinal ganglion cells (RGCs) as early as embryonic day 15. When overexpressed, KLF16 inhibits RGC neurite outgrowth and enhances RGC growth cone collapse in response to exogenous ephrinA5 ligands. Ephrin/EPH signaling regulates RGC connectivity. The EphA5 promoter contains multiple GC- and GT-rich KLF-binding sites, which, as shown by ChIP-assays, bind KLF16 in vivo In electrophoretic mobility shift assays, KLF16 binds specifically to a single KLF site near the EphA5 transcription start site that is required for KLF16 transactivation. Interestingly, methylation of only six of 98 CpG dinucleotides within the EphA5 promoter blocks its transactivation by KLF16 but enables transactivation by KLF2 and KLF15. These data demonstrate a role for KLF16 in regulation of RGC neurite outgrowth and as a methylation-sensitive transcriptional regulator of EphA5 expression. Together, these data identify differential low level methylation as a novel mechanism for regulating KLF16-mediated EphA5 expression across the retina. Because of the critical role of ephrin/EPH signaling in patterning RGC connectivity, understanding the role of KLFs in regulating neurite outgrowth and Eph receptor expression will be vital for successful restoration of functional vision through optic nerve regenerative therapies.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Neuritos/metabolismo , Receptor EphA5/biossíntese , Elementos de Resposta/fisiologia , Células Ganglionares da Retina/metabolismo , Transdução de Sinais/fisiologia , Ativação Transcricional/fisiologia , Animais , Metilação de DNA , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Receptor EphA5/genética , Células Ganglionares da Retina/citologia , Transcrição Gênica/fisiologia
12.
J Biol Chem ; 291(31): 16048-58, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27226561

RESUMO

The Lgals3 gene encodes a multifunctional ß-galactoside-binding protein, galectin-3. Galectin-3 has been implicated in a broad range of biological processes from chemotaxis and inflammation to fibrosis and apoptosis. The role of galectin-3 as a modulator of inflammation has been studied intensively, and recent evidence suggests that it may serve as a protective factor in obesity and other metabolic disorders. Despite considerable interest in galectin-3, little is known about its physiological regulation at the transcriptional level. Here, using knockout mice, chromatin immunoprecipitations, and cellular and molecular analyses, we show that the zinc finger transcription factor Krüppel-like factor 3 (KLF3) directly represses galectin-3 transcription. We find that galectin-3 is broadly up-regulated in KLF3-deficient mouse tissues, that KLF3 occupies regulatory regions of the Lgals3 gene, and that KLF3 directly binds its cognate elements (CACCC boxes) in the galectin-3 promoter and represses its activation in cellular assays. We also provide mechanistic insights into the regulation of Lgals3, demonstrating that C-terminal binding protein (CtBP) is required to drive optimal KLF3-mediated silencing. These findings help to enhance our understanding of how expression of the inflammatory modulator galectin-3 is controlled, opening up avenues for potential therapeutic interventions in the future.


Assuntos
Galectina 3/biossíntese , Inativação Gênica , Mediadores da Inflamação/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Repressoras/metabolismo , Elementos de Resposta , Transcrição Gênica , Animais , Galectina 3/genética , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Proteínas Repressoras/genética
13.
J Biol Chem ; 290(15): 9929-40, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25713074

RESUMO

Erythroid Kruppel-like factor (EKLF or KLF1) is a transcription factor crucial for red cell development that is directly involved in regulation of a large number of erythroid genes. EKLF serves mostly as an activator of expression of these genes; however, it can act also as a repressor. Here, we present evidence that EKLF interacts with proteins from the PIAS (protein inhibitor of activated STAT) family that convey repressive activity to EKLF in the absence of sumoylation. Our studies identify PIAS3 as a transcriptional corepressor of EKLF for at least a subset of its target genes during erythropoiesis (e.g. ß-globin, α-hemoglobin stabilizing protein). We demonstrate an interaction between EKLF and PIAS proteins confirmed by in vivo coimmunoprecipitation assays with both exogenous and endogenous proteins. We identified an LXXLL signature motif located near the N terminus of PIAS proteins that, although not involved in the EKLF-PIAS3 interaction, is required for the transrepression activity. Knockdown of endogenous PIAS3 accelerates differentiation of both murine erythroleukemia cells, as well as fetal liver cells, whereas an increase in PIAS3 levels inhibits this increase. Using chromatin immunoprecipitation assays, we show that PIAS3 preferentially occupies the ß-globin promoter in undifferentiated murine erythroleukemia cells. Together these results demonstrate that an interaction between EKLF and PIAS3 provides a novel mode of regulation of EKLF activity in the absence of sumolylation and furthermore shows an important involvement of PIAS proteins in erythropoiesis.


Assuntos
Fatores de Transcrição Kruppel-Like/genética , Mutação , Proteínas Inibidoras de STAT Ativados/genética , Ativação Transcricional , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Western Blotting , Células COS , Diferenciação Celular/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Humanos , Células K562 , Fatores de Transcrição Kruppel-Like/metabolismo , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patologia , Camundongos , Microscopia Confocal , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sumoilação , Globinas beta/genética , Globinas beta/metabolismo
14.
J Biol Chem ; 290(13): 8591-605, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25659434

RESUMO

Krüppel-like factor 3 (KLF3/BKLF), a member of the Krüppel-like factor (KLF) family of transcription factors, is a widely expressed transcriptional repressor with diverse biological roles. Although there is considerable understanding of the molecular mechanisms that allow KLF3 to silence the activity of its target genes, less is known about the signal transduction pathways and post-translational modifications that modulate KLF3 activity in response to physiological stimuli. We observed that KLF3 is modified in a range of different tissues and found that the serine/threonine kinase homeodomain-interacting protein kinase 2 (HIPK2) can both bind and phosphorylate KLF3. Mass spectrometry identified serine 249 as the primary phosphorylation site. Mutation of this site reduces the ability of KLF3 to bind DNA and repress transcription. Furthermore, we also determined that HIPK2 can phosphorylate the KLF3 co-repressor C-terminal binding protein 2 (CtBP2) at serine 428. Finally, we found that phosphorylation of KLF3 and CtBP2 by HIPK2 strengthens the interaction between these two factors and increases transcriptional repression by KLF3. Taken together, our results indicate that HIPK2 potentiates the activity of KLF3.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Oxirredutases do Álcool , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Proteínas Correpressoras , DNA/química , Ensaio de Desvio de Mobilidade Eletroforética , Fatores de Transcrição Kruppel-Like/química , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Transcrição Gênica , Ativação Transcricional
15.
J Biol Chem ; 289(9): 5914-24, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24407292

RESUMO

The mammalian heart, the body's largest energy consumer, has evolved robust mechanisms to tightly couple fuel supply with energy demand across a wide range of physiologic and pathophysiologic states, yet, when compared with other organs, relatively little is known about the molecular machinery that directly governs metabolic plasticity in the heart. Although previous studies have defined Kruppel-like factor 15 (KLF15) as a transcriptional repressor of pathologic cardiac hypertrophy, a direct role for the KLF family in cardiac metabolism has not been previously established. We show in human heart samples that KLF15 is induced after birth and reduced in heart failure, a myocardial expression pattern that parallels reliance on lipid oxidation. Isolated working heart studies and unbiased transcriptomic profiling in Klf15-deficient hearts demonstrate that KLF15 is an essential regulator of lipid flux and metabolic homeostasis in the adult myocardium. An important mechanism by which KLF15 regulates its direct transcriptional targets is via interaction with p300 and recruitment of this critical co-activator to promoters. This study establishes KLF15 as a key regulator of myocardial lipid utilization and is the first to implicate the KLF transcription factor family in cardiac metabolism.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Metabolismo dos Lipídeos , Proteínas Musculares/metabolismo , Miocárdio/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Miocárdio/patologia , Proteínas Nucleares/genética , Oxirredução , Fatores de Transcrição/genética
16.
J Biol Chem ; 289(13): 9089-99, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24532790

RESUMO

The S100 protein family represents the largest subgroup of calcium binding EF-hand type proteins. These proteins have been reported to be involved in a wide range of biological functions that are related to normal cell development and tumorigenesis. S100A14 is a recently identified member of the S100 protein family and differentially expressed in a number of different human malignancies. However, the transcriptional regulation of S100A14 and its role in breast cancer needs to be further investigated. Here, we determined that 12-O-tetradecanoylphorbol-13-acetate (TPA) up-regulated the expression of KLF4 and facilitated its binding directly to two conserved GC-rich DNA segments within the S100A14 promoter, which is essential for the transactivation of KLF4 induced S100A14 expression. Furthermore, stable silencing of KLF4 significantly suppressed breast cancer cell migration induced by TPA. Collectively, these results offer insights into the fact that TPA provokes cell motility through regulating the expression and function of S100A14 in a KLF4-dependent manner.


Assuntos
Neoplasias da Mama/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Movimento Celular/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Animais , Sequência de Bases , Sítios de Ligação , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Tempo
17.
J Biol Chem ; 289(47): 32742-56, 2014 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-25288800

RESUMO

It is increasingly important to understand the molecular basis for the plasticity of neoplastic cells and their capacity to transition between differentiated and stemlike phenotypes. Kruppel-like factor-9 (KLF9), a member of the large KLF transcription factor family, has emerged as a regulator of oncogenesis, cell differentiation, and neural development; however, the molecular basis for the diverse contextual functions of KLF9 remains unclear. This study focused on the functions of KLF9 in human glioblastoma stemlike cells. We established for the first time a genome-wide map of KLF9-regulated targets in human glioblastoma stemlike cells and show that KLF9 functions as a transcriptional repressor and thereby regulates multiple signaling pathways involved in oncogenesis and stem cell regulation. A detailed analysis of one such pathway, integrin signaling, showed that the capacity of KLF9 to inhibit glioblastoma cell stemness and tumorigenicity requires ITGA6 repression. These findings enhance our understanding of the transcriptional networks underlying cancer cell stemness and differentiation and identify KLF9-regulated molecular targets applicable to cancer therapeutics.


Assuntos
Diferenciação Celular/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Integrina alfa6/genética , Fatores de Transcrição Kruppel-Like/genética , Animais , Antibióticos Antineoplásicos/farmacologia , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Integrina alfa6/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos SCID , Regiões Promotoras Genéticas/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Carga Tumoral/genética
18.
J Biol Chem ; 289(22): 15798-809, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24759103

RESUMO

Sphingosine kinase 1 (SK1) is an FGF-inducible gene responsible for generation of sphingosine-1-phosphate, a critical lipid signaling molecule implicated in diverse endothelial cell functions. In this study, we identified SK1 as a target of the canonical FGF2/FGF receptor 1 activation pathway in endothelial cells and sought to identify novel transcriptional pathways that mediate lipid signaling. Studies using the 1.9-kb SK1 promoter and deletion mutants revealed that basal and FGF2-stimulated promoter activity occurred through two GC-rich regions located within 633 bp of the transcription start site. Screening for GC-rich binding transcription factors that could activate this site demonstrated that KLF14, a gene implicated in obesity and the metabolic syndrome, binds to this region. Congruently, overexpression of KLF14 increased basal and FGF2-stimulated SK1 promoter activity by 3-fold, and this effect was abrogated after mutation of the GC-rich sites. In addition, KLF14 siRNA transfection decreased SK1 mRNA and protein levels by 3-fold. Congruently, SK1 mRNA and protein levels were decreased in livers from KLF14 knock-out mice. Combined, luciferase, gel shift, and chromatin immunoprecipitation assays showed that KLF14 couples to p300 to increase the levels of histone marks associated with transcriptional activation (H4K8ac and H3K14ac), while decreasing repressive marks (H3K9me3 and H3K27me3). Collectively, the results demonstrate a novel mechanism whereby SK1 lipid signaling is regulated by epigenetic modifications conferred by KLF14 and p300. Thus, this is the first description of the activity and mechanisms underlying the function of KLF14 as an activator protein and novel regulator of lipid signaling.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Metabolismo dos Lipídeos/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição Sp/metabolismo , Animais , Cromatina/metabolismo , Células Endoteliais/citologia , Epigênese Genética/fisiologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células HEK293 , Histonas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fígado/citologia , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Fatores de Transcrição Sp/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Ativação Transcricional/fisiologia
19.
J Biol Chem ; 289(36): 25306-16, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25037223

RESUMO

The histone deacetylase inhibitor (HDACi) sodium butyrate promotes differentiation of colon cancer cells as evidenced by induced expression and enzyme activity of the differentiation marker intestinal alkaline phosphatase (ALPi). Screening of a panel of 33 colon cancer cell lines identified cell lines sensitive (42%) and resistant (58%) to butyrate induction of ALP activity. This differential sensitivity was similarly evident following treatment with the structurally distinct HDACi, MS-275. Resistant cell lines were significantly enriched for those harboring the CpG island methylator phenotype (p = 0.036, Chi square test), and resistant cell lines harbored methylation of the ALPi promoter, particularly of a CpG site within a critical KLF/Sp regulatory element required for butyrate induction of ALPi promoter activity. However, butyrate induction of an exogenous ALPi promoter-reporter paralleled up-regulation of endogenous ALPi expression across the cell lines, suggesting the presence or absence of a key transcriptional regulator is the major determinant of ALPi induction. Through microarray profiling of sensitive and resistant cell lines, we identified KLF5 to be both basally more highly expressed as well as preferentially induced by butyrate in sensitive cell lines. KLF5 overexpression induced ALPi promoter-reporter activity in resistant cell lines, KLF5 knockdown attenuated butyrate induction of ALPi expression in sensitive lines, and butyrate selectively enhanced KLF5 binding to the ALPi promoter in sensitive cells. These findings demonstrate that butyrate induction of the cell differentiation marker ALPi is mediated through KLF5 and identifies subsets of colon cancer cell lines responsive and refractory to this effect.


Assuntos
Fosfatase Alcalina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Fatores de Transcrição Kruppel-Like/metabolismo , Fosfatase Alcalina/genética , Benzamidas/farmacologia , Sítios de Ligação/genética , Western Blotting , Ácido Butírico/farmacologia , Diferenciação Celular/genética , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ilhas de CpG/genética , Metilação de DNA , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Células HCT116 , Células HT29 , Humanos , Fatores de Transcrição Kruppel-Like/genética , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Piridinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Biol Chem ; 289(5): 2755-64, 2014 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-24338020

RESUMO

Rosiglitazone, a well known insulin sensitizer, stimulates adipocyte differentiation via the activation of peroxisome proliferator-activated receptor γ (PPARγ). Previous two-dimensional proteomics studies using C3H10T1/2 murine mesenchymal pluripotent stem cells revealed that prolyl hydroxylase domain protein (PHD) levels significantly increased during rosiglitazone-induced adipocyte differentiation (RIAD). In this study, we investigated the functional role played by PHD during RIAD. Three PHD isoforms (PHD1, 2, and 3) were found to be up-regulated in C3H10T1/2 cells during RIAD, whereas PHD knockdown and treatment with PHD inhibitors (dimethyloxalyl glycine or ethyl-3,4-dihydroxybenzoate) blocked RIAD. PHD inhibition was found to be associated with increases in the levels of anti-adipogenic proteins such as GATA-3, KLF-2, and transcriptional coactivator with PDZ binding motif (TAZ), with their reduced ubiquitination, suggesting that PHDs evoke the ubiquitination/proteasomal degradation of anti-adipogenic proteins. On the other hand, MG-132 (a proteasomal inhibitor) prevented the degradation of anti-adipogenic proteins and retarded RIAD. PPARγ antagonists (bisphenol A diglycidyl ether or GW9662) blunted the effects of rosiglitazone on PHD regulation. Furthermore, putative PPARγ binding sites were identified in the promoter region of PHDs by ChIP-PCR, implying that rosiglitazone may induce PHD up-regulation directly by PPARγ activation. Consistent with in vitro results, oral administration of rosiglitazone to ob/ob mice for 2 weeks increased adipose PHD levels and decreased anti-adipogenic protein levels by increasing their ubiquitination. These results suggest that rosiglitazone increases PHD expression in a PPARγ-dependent manner and that this leads to the commitment of anti-adipogenic proteins to the ubiquitination-proteasomal pathway and to the subsequent induction of adipocyte differentiation.


Assuntos
Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Tiazolidinedionas/farmacologia , Aciltransferases , Adipócitos/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Fator de Transcrição GATA3/metabolismo , Hipoglicemiantes/farmacologia , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , PPAR gama/metabolismo , Pró-Colágeno-Prolina Dioxigenase/genética , Regiões Promotoras Genéticas/fisiologia , Proteômica , Rosiglitazona , Fatores de Transcrição/metabolismo , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA