Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genomics ; 114(6): 110498, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36174881

RESUMEN

Diabetic retinopathy is one of the microvascular complications in diabetic patients and the leading cause of blindness worldwide. The levels of METTL3, lncRNA SNHG7, KHSRP, MKL1, endothelial and mesenchymal markers were determined by RT-qPCR or western blot assays in vitro and in vivo. H&E staining was used to observe the retinal structure in a mouse model of DR. The expression levels of METTL3 and SNHG7 were significantly downregulated in DR patients, DR mice and high glucose-induced HRMECs cells. Notably, METTL3 installed the m6A modification and enhanced the stability of SNHG7. Besides, METTL3 inhibited HRMECs EndoMT by promoting the expression of SNHG7. Additionally, SNHG7 was found to weaken MKL1 mRNA stability by binding to the RNA-binding protein KHSRP. Furthermore, we verified that METTL3 regulated EndoMT in DR through the SNHG7/MKL1 axis. We conclude that METTL3 regulates endothelial-mesenchymal transition in DR via the SNHG7/KHSRP/MKL1 axis, providing a new target for DR treatment.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , ARN Largo no Codificante , Ratones , Animales , Transferasas , Retinopatía Diabética/genética , ARN Largo no Codificante/genética , Metiltransferasas/genética
2.
Stem Cells ; 39(1): 43-54, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33075202

RESUMEN

There is wide variability in the propensity of somatic cells to reprogram into pluripotency in response to the Yamanaka factors. How to segregate these variabilities to enrich for cells of specific traits that reprogram efficiently remains challenging. Here we report that the variability in reprogramming propensity is associated with the activity of the MKL1/SRF transcription factor and concurs with small cell size as well as rapid cell cycle. Reprogramming progressive cells can be prospectively identified by their low activity of a widely used synthetic promoter, CAG. CAGlow cells arise and expand during cell cycle acceleration in the early reprogramming culture of both mouse and human fibroblasts. Our work illustrates a molecular scenario underlying the distinct reprogramming propensities and demonstrates a convenient practical approach for their enrichment.


Asunto(s)
Técnicas de Reprogramación Celular , Reprogramación Celular , Regiones Promotoras Genéticas , Factores de Transcripción , Animales , Ratones , Ratones Transgénicos , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
3.
J Cell Sci ; 132(16)2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31363007

RESUMEN

Papillary thyroid cancer (PTC), the most common thyroid malignancy, has a strong propensity for cervical lymph node metastasis (LNM), which increases the risk of locoregional recurrence and decreases survival probability in some high-risk groups. Hence, there is a pressing requirement for a reliable biomarker to predict LNM in thyroid cancer. In the present study, MKL1 (also known as MRTFA) expression was significantly increased in PTC patients with LNM compared with those without. Further receiver operating characteristic (ROC) analysis showed that MKL1 expression had a diagnostic value in the differentiation of LNM in PTC. Furthermore, Kaplan-Meier analysis revealed that high MKL1 expression was associated with significantly decreased survival in PTC. Additionally, our study indicated that MKL1 promoted the migration and invasion of PTC cells. MKL1 interacted with and recruited Smad3 to the promoter of MMP2 to activate MMP2 transcription upon treatment with TGF-ß. Moreover, there was significant correlation between expression of TGF-ß, MKL1 and MMP2 in our clinical cohort of specimens from individuals with PTC. Our results suggest that the detection of MKL1 expression could be used to predict cervical LNM and inform post-operative follow-up in individuals with PTC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides , Transactivadores/biosíntesis , Supervivencia sin Enfermedad , Femenino , Humanos , Metástasis Linfática , Masculino , Tasa de Supervivencia , Cáncer Papilar Tiroideo/metabolismo , Cáncer Papilar Tiroideo/mortalidad , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/mortalidad , Neoplasias de la Tiroides/patología
4.
BMC Cancer ; 21(1): 1184, 2021 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-34742274

RESUMEN

BACKGROUND: Histone modification plays essential roles in hepatocellular carcinoma (HCC) pathogenesis, but the regulatory mechanisms remain poorly understood. In this study, we aimed to analyze the roles of Megakaryoblastic leukemia 1 (MKL1) and its regulation of COMPASS (complex of proteins associated with Set1) in HCC cells. METHODS: MKL1 expression in clinical tissues and cell lines were detected by bioinformatics, qRT-PCR and western blot. MKL1 expression in HCC cells were silenced with siRNA, followed by cell proliferation evaluation via Edu staining and colony formation, migration and invasion using the Transwell system, and apoptosis by Hoechst staining. HCC cell tumorigenesis was assessed by cancer cell line-based xenograft model, combined with H&E staining and IHC assays. RESULTS: MKL1 expression was elevated in HCC cells and clinical tissues which was correlated with poor prognosis. MKL1 silencing significantly repressed proliferation, migration, invasion and colony formation but enhanced apoptosis in HepG2 and Huh-7 cells. MKL1 silencing also inhibited COMPASS components and p65 protein expression in HepG2 and Huh-7 cells. HepG2 cell tumorigenesis in nude mice was severely impaired by MKL1 knockdown, resulted into suppressed Ki67 expression and cell proliferation. CONCLUSION: MKL1 promotes HCC pathogenesis by regulating hepatic cell proliferation, migration and apoptosis via the COMPASS complex and NF-κB signaling.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Neoplasias Hepáticas/metabolismo , FN-kappa B/metabolismo , Transactivadores/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Apoptosis/genética , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Silenciador del Gen , Células Hep G2 , Xenoinjertos , Código de Histonas , Humanos , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Pronóstico , ARN Interferente Pequeño , Transactivadores/genética , Ensayo de Tumor de Célula Madre
5.
J Cell Mol Med ; 24(18): 10338-10342, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32770617

RESUMEN

This study was aimed to explore the correlation of intercellular adhesion molecule-1 (ICAM-1) K469E and megakaryoblastic leukaemia factor-1 (MKL-1) -184C/T polymorphisms with the susceptibility to coronary heart disease (CHD) in the Chinese Han population. 100 CHD patients and 91 healthy people that had no blood connection with each other were enrolled in this case-control study. ICAM-1 and MKL-1 polymorphisms were genotyped by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) approach. Multiple logistic regression was used to analyse the correlation between polymorphisms of ICAM-1 and MKL-1 and CHD susceptibility. Differences of genotype and allele frequencies of the two SNPs between case and control groups were analysed by chi-square test. Odds ratios (ORs) and 95% confidence intervals (CIs) were indicated relative susceptibility of CHD. The distributions of ICAM-1 and MKL-1 polymorphisms in each group conformed to Hardy-Weinberg equilibrium (HWE). After adjusting for traditional risk factors, the TT genotype frequency of MKL-1 -184C/T polymorphism was found significantly higher in case group than in control group (P < .05). Meanwhile, T allele frequency increased in case group compared with control group, and the differences had statistical significance (P = .04, OR = 2.34, 95% CI = 1.34-5.26). Logistic regression analysis in this study proved that smoking, hypertension, diabetes and triglyceride (TG) were all risk factors for CHD ICAM-1 K469E polymorphism has no association with the onset of CHD. But MKL-1 -184C/T polymorphism is associated with the risk of CHD and T allele might be a susceptibility factor for CHD.


Asunto(s)
Enfermedad Coronaria/genética , Predisposición Genética a la Enfermedad , Molécula 1 de Adhesión Intercelular/genética , Polimorfismo de Nucleótido Simple/genética , Transactivadores/genética , Estudios de Casos y Controles , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Factores de Riesgo
6.
Cell Commun Signal ; 18(1): 107, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32646440

RESUMEN

BACKGROUND: Foxp3+CD4+ regulatory T cells (Treg) constitutes a key event in autoimmune diseases. STAT5b is the critical link between the IL-2/15 and FOXP3, the master regulator of Treg cells. METHODS: The CD3+T cell and Foxp3+CD4+ regulatory T cells were overexpressioned or knockdown MKL-1 and STAT5a and tested for Treg cell development and function. Direct interaction of MKL-1 and STAT5a were analyzed by coimmunoprecipitation assays, Luciferase assay, Immunofluoresence Staining and Yeast two-hybrid screening. The effect of MKL-1 and STAT5a on the Treg genes expression was analyzed by qPCR and western blotting and Flow cytometry. RESULTS: However, the molecular mechanisms mediating STAT5b-dependent Treg genes expression and Treg cell phenotype and function in autoimmune diseases are not well defined. Here, we report that the MKL-1 is a coactivator for the major Treg genes transcription factor STAT5b, which is required for human Treg cell phenotype and function. The N terminus of STAT5b, which contains a basic coiled-coil protein-protein interaction domain, binds the C-terminal activation domain of MKL-1 and enhances MKL-1 mediated transcriptional activation of Treg-specific, CArG containing promoters, including the Treg-specific genes Foxp3. Suppression of endogenous STAT5b expression by specific small interfering RNA attenuates MKL-1 transcriptional activation in cultured human cells. The STAT5b-MKL-1 interaction identifies a role of Treg-specific gene regulation and regulated mouse Treg cell development and function and suggests a possible mechanism for the protective effects of autoimmune disease Idiopathic Thrombocytopenic Purpura (ITP). CONCLUSIONS: Our studies demonstrate for the first time that MKL-1 is a coactivator for STAT5b, the regulator of Treg cell development and function. Video abstract.


Asunto(s)
Factor de Transcripción STAT5/metabolismo , Linfocitos T Reguladores/inmunología , Transactivadores/metabolismo , Amidas/farmacología , Secuencia de Bases , Biomarcadores/metabolismo , Complejo CD3/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Janus Quinasa 3/metabolismo , Recuento de Linfocitos , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Púrpura Trombocitopénica Idiopática/inmunología , Piridinas/farmacología , Factor de Transcripción STAT5/química , Factor de Respuesta Sérica/metabolismo , Transducción de Señal/efectos de los fármacos , Transactivadores/química , Tirfostinos/farmacología , Proteína de Unión al GTP rhoA/metabolismo
7.
Int J Mol Sci ; 21(10)2020 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-32408494

RESUMEN

Acute leukemias are genetic diseases caused by translocations or mutations, which dysregulate hematopoiesis towards malignant transformation. However, the molecular mode of action is highly versatile and ranges from direct transcriptional to post-transcriptional control, which includes RNA-binding proteins (RBPs) as crucial regulators of cell fate. RBPs coordinate RNA dynamics, including subcellular localization, translational efficiency and metabolism, by binding to their target messenger RNAs (mRNAs), thereby controlling the expression of the encoded proteins. In view of the growing interest in these regulators, this review summarizes recent research regarding the most influential RBPs relevant in acute leukemias in particular. The reported RBPs, either dysregulated or as components of fusion proteins, are described with respect to their functional domains, the pathways they affect, and clinical aspects associated with their dysregulation or altered functions.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Enfermedad Aguda , Animales , Humanos , Leucemia/metabolismo , Conformación de Ácido Nucleico , Unión Proteica , Dominios Proteicos , ARN Mensajero/química , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo
8.
J Mol Cell Cardiol ; 133: 26-35, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31128166

RESUMEN

We have previously reported that promoter polymorphism of myocardin-related transcription factor A (MRTF-A) is associated with coronary atherosclerosis. However, the contribution of MRTF-A to the development of atherosclerosis remains unknown. Macrophages are known to be important mediators of atherosclerosis. It has been demonstrated that local proliferation and survival of macrophages are atherogenic. In this study, we found that MRTF-A was highly expressed in lesional macrophages in human carotid atherosclerotic plaque. We then investigated the role of macrophagic MRTF-A in the pathogenesis of atherosclerosis. ApoE null MRTF-A transgenic mice (ApoE-/-/MRTF-Atg/+), in which human MRTF-A was specifically overexpressed in monocytes/macrophages, were established and fed with normal diet to examine the progression of atherosclerosis. We found that ApoE-/-/MRTF-Atg/+ aggravated atherosclerosis and lesional macrophages were more prominently accumulated in the aortic sinus of ApoE-/-/MRTF-Atg/+ than in that of ApoE-/- littermates. We also found that MRTF-A promoted proliferation and mitigated apoptosis of macrophages both in vitro and in vivo, and down regulated the expression of cyclin-dependent kinase inhibitors. From these findings, we conclude that MRTF-A modulates functional properties of pro-atherogenic macrophages. Our study may play a valuable role in understanding the pathological role of macrophagic MRTF-A in the progression of atherosclerosis.


Asunto(s)
Aterosclerosis/etiología , Aterosclerosis/metabolismo , Susceptibilidad a Enfermedades , Macrófagos/metabolismo , Transactivadores/genética , Animales , Apoptosis/genética , Aterosclerosis/patología , Biomarcadores , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Inmunohistoquímica , Macrófagos/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal , Transactivadores/metabolismo
9.
J Cell Biochem ; 120(5): 7814-7824, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30426547

RESUMEN

Megakaryoblastic leukemia 1 (MKL1) was closely related to the pathogenesis of various human malignant cancers. MiR34a was reported to be closely related to cancer cell proliferation. Forkhead box protein 3 (FOXP3) was a transcription factor that played a different role in different cancer types. CDK6 was involved in cell cycle progression and was upregulated in several types of cancers. The present study investigated the effects of MKL1/miR34a/FOXP3 axis on cell proliferation in MGC803 gastric cancer cells. Our results demonstrated that overexpression of MKL1 promoted proliferation of MGC80-3 cells, MKL1 directly binding to the promoter of CDK6 to increase its expression. Knockdown of FOXP3 promoted proliferation of MGC80-3 cells and MKL1 inhibited the expression of FOXP3 via miR-34a. The finding can contribute to elucidating the regulatory mechanism involved in the cell cycle progression of gastric cancer cells and may aid in screening potential gene targets for the biological therapy of gastric cancer.

10.
J Cell Sci ; 130(13): 2111-2118, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28576971

RESUMEN

The complex comprising serum response factor (SRF) and megakaryoblastic leukemia 1 protein (Mkl1) promotes myofibroblast differentiation during wound healing. SRF-Mkl1 is sensitive to the mechanical properties of the extracellular environment; but how cells sense and transduce mechanical cues to modulate SRF-Mkl1-dependent gene expression is not well understood. Here, we demonstrate that the nuclear lamina-associated inner nuclear membrane protein Emerin stimulates SRF-Mkl1-dependent gene activity in a substrate stiffness-dependent manner. Specifically, Emerin was required for Mkl1 nuclear accumulation and maximal SRF-Mkl1-dependent gene expression in response to serum stimulation of cells grown on stiff substrates but was dispensable on more compliant substrates. Focal adhesion area was also reduced in cells lacking Emerin, consistent with a role for Emerin in sensing substrate stiffness. Expression of a constitutively active form of Mkl1 bypassed the requirement for Emerin in SRF-Mkl1-dependent gene expression and reversed the focal adhesion defects evident in EmdKO fibroblasts. Together, these data indicate that Emerin, a conserved nuclear lamina protein, couples extracellular matrix mechanics and SRF-Mkl1-dependent transcription.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas Nucleares/genética , Factor de Respuesta Sérica/genética , Transactivadores/genética , Cicatrización de Heridas/genética , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Adhesiones Focales/genética , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Noqueados , Miofibroblastos/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Lámina Nuclear/genética , Transducción de Señal/genética
11.
Cell Commun Signal ; 16(1): 5, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29391067

RESUMEN

BACKGROUND: Long non-coding RNAs (lncRNAs) have emerged as a new and crucial layer of gene regulation in recent years and regulate various biological processes such as carcinogenesis and metastasis. LncRNA HOTAIR, an oncogenic lncRNA, is involved in human tumorigenesis and dysregulated in cervical cancer. Megakaryoblastic leukemia 1 (MKL1), as a transcription coactivity factor, involved in cancer metastasis and cell differentiation. However, the precise mechanism of biological roles of HOTAIR and MKL1 in cancer cells remain unclear. METHODS: The expression levels of HOTAIR and MKL1 were measured by quantitative PCR (qPCR), immunoblotting, in situ hybridization (ISH) and immunohistochemistry (IHC). Wound-healing and transwell assays were used to examine the invasive abilities of HeLa cells. Luciferase reporter assays and CHIP were used to determine how MKL1 regulates HOTAIR. Tissue microarray and immunohistochemical staining were used to assess the correlation between HOTAIR and MKL1 in Cervical cancer tissues in vivo. RESULT: In this study, we have identified that MKL1 had a role in the induction of migration and invasion in cervical cancer cells. Moreover, the expression level of MKL1, as the targeting gene of miR206, was decreased after HOTAIR inhibition in HeLa cells. Agreement with it, Highly level of MKL1 correlation with HOTAIR is validated in cervical cancer tissues. Importantly, HOTAIR is observed to participate in the silencing of miR206 expression. Interestingly, HOTAIR inhibition could also accelerate the expression of MKL1 in cytoplasm. What is more, MKL1 can activate the transcription of HOTAIR through binding the CArG box in the promoter of HOTAIR. CONCLUSION: These elucidates that the phenotypic effects of migration and invasion observed after HOTAIR inhibition, at least in part, through the regulation of MKL1 via inhibition of miR206 expression in HeLa cells. These data indicate the existence of a positive feedback loop between HOTAIR and MKL1. Together, these findings suggest that MKL1 is an important player in the functions of HOTAIR in the migration and invasion of cancer cells.


Asunto(s)
MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Transactivadores/metabolismo , Regiones no Traducidas 3' , Antagomirs/metabolismo , Secuencia de Bases , Movimiento Celular , Femenino , Células HeLa , Humanos , MicroARNs/antagonistas & inhibidores , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , Transactivadores/química , Transactivadores/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
12.
Exp Cell Res ; 357(1): 135-144, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28499590

RESUMEN

Epithelial-mesenchymal transition (EMT) plays an important role in breast cancer cell metastasis. Both (megakaryoblastic leukemia)/myocardin-like 1 (MKL-1) and Signal transducer and activator of transcription 3 (STAT3) have been implicated in the control of cellular metabolism, survival and growth. Our previous study has shown that cooperativity of MKL-1 and STAT3 promoted breast cancer cell migration. Herein, we demonstrate a requirement for MKL-1 and STAT3 in miRNA-mediated cellular EMT to affect breast cancer cell migration. Here we show that cooperativity of MKL-1 and STAT3 promoted the EMT of MCF-7 cells. Importantly, MKL-1 and STAT3 promoted the expression of Vimentin via its promoter CArG box. Interestingly, miR-93-5p inhibits the EMT of breast cancer cells through suppressing the expression of MKL-1 and STAT3 via targeted their 3'UTR. These results demonstrated a novel pathway through which miR-93-5p regulates MKL-1 and STAT3 to affect EMT controlling breast cancer cell migration.


Asunto(s)
Neoplasias de la Mama/genética , Transición Epitelial-Mesenquimal/fisiología , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , Factor de Transcripción STAT3/genética , Transactivadores/genética , Neoplasias de la Mama/metabolismo , Humanos , Células MCF-7 , Regiones Promotoras Genéticas/genética
13.
Exp Cell Res ; 350(1): 267-278, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27919746

RESUMEN

The functional properties of actin-regulating formin proteins are diverse and in many cases cell-type specific. FHOD1, a formin expressed predominantly in cells of mesenchymal lineage, bundles actin filaments and participates in maintenance of cell shape, migration and cellular protrusions. FHOD1 participates in cancer-associated epithelial to mesenchymal transition (EMT) in oral squamous cell carcinoma and breast cancer. The role of FHOD1 in melanomas has not been characterized. Here, we show that FHOD1 expression is typically strong in cutaneous melanomas and cultured melanoma cells while the expression is low or absent in benign nevi. By using shRNA to knockdown FHOD1 in melanoma cells, we discovered that FHOD1 depleted cells are larger, rounder and have smaller focal adhesions and inferior migratory capacity as compared to control cells. Importantly, we found FHOD1 depleted cells to have reduced colony-forming capacity and attenuated tumor growth in vivo, a finding best explained by the reduced proliferation rate caused by cell cycle arrest. Unexpectedly, FHOD1 depletion did not prevent invasive growth at the tumor margins. These results suggest that FHOD1 participates in key cellular processes that are dysregulated in malignancy, but may not be essential for melanoma cell invasion.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas Fetales/metabolismo , Melanoma/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Cutáneas/metabolismo , Fibras de Estrés/metabolismo , Actinas/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal/fisiología , Forminas , Humanos , Ratones , Activación Transcripcional/fisiología , Regulación hacia Arriba , Melanoma Cutáneo Maligno
14.
J Neurosci ; 36(6): 2027-43, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26865625

RESUMEN

Growth-associated protein 43 (GAP43), a protein kinase C (PKC)-activated phosphoprotein, is often implicated in axonal plasticity and regeneration. In this study, we found that GAP43 can be induced by the endotoxin lipopolysaccharide (LPS) in rat brain astrocytes both in vivo and in vitro. The LPS-induced astrocytic GAP43 expression was mediated by Toll-like receptor 4 and nuclear factor-κB (NF-κB)- and interleukin-6/signal transducer and activator of transcription 3 (STAT3)-dependent transcriptional activation. The overexpression of the PKC phosphorylation-mimicking GAP43(S41D) (constitutive active GAP43) in astrocytes mimicked LPS-induced process arborization and elongation, while application of a NF-κB inhibitory peptide TAT-NBD or GAP43(S41A) (dominant-negative GAP43) or knockdown of GAP43 all inhibited astrogliosis responses. Moreover, GAP43 knockdown aggravated astrogliosis-induced microglial activation and expression of proinflammatory cytokines. We also show that astrogliosis-conditioned medium from GAP43 knock-down astrocytes inhibited GAP43 phosphorylation and axonal growth, and increased neuronal damage in cultured rat cortical neurons. These proneurotoxic effects of astrocytic GAP43 knockdown were accompanied by attenuated glutamate uptake and expression of the glutamate transporter excitatory amino acid transporter 2 (EAAT2) in LPS-treated astrocytes. The regulation of EAAT2 expression involves actin polymerization-dependent activation of the transcriptional coactivator megakaryoblastic leukemia 1 (MKL1), which targets the serum response elements in the promoter of rat Slc1a2 gene encoding EAAT2. In sum, the present study suggests that astrocytic GAP43 mediates glial plasticity during astrogliosis, and provides beneficial effects for neuronal plasticity and survival and attenuation of microglial activation. SIGNIFICANCE STATEMENT: Astrogliosis is a complex state in which injury-stimulated astrocytes exert both protective and harmful effects on neuronal survival and plasticity. In this study, we demonstrated for the first time that growth-associated protein 43 (GAP43), a well known growth cone protein that promotes axonal regeneration, can be induced in rat brain astrocytes by the proinflammatory endotoxin lipopolysaccharide via both nuclear factor-κB and signal transducer and activator of transcription 3-mediated transcriptional activation. Importantly, LPS-induced GAP43 mediates plastic changes of astrocytes while attenuating astrogliosis-induced microglial activation and neurotoxicity. Hence, astrocytic GAP43 upregulation may serve to indicate beneficial astrogliosis after CNS injury.


Asunto(s)
Astrocitos/efectos de los fármacos , Proteína GAP-43/biosíntesis , Proteína GAP-43/genética , Gliosis/genética , Microglía/efectos de los fármacos , FN-kappa B/genética , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/patología , Factor de Transcripción STAT3/genética , Receptor Toll-Like 4/genética , Animales , Citocinas/biosíntesis , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Activación de Macrófagos/efectos de los fármacos , Neuronas , Fosforilación , Ratas , Ratas Sprague-Dawley , Transactivadores/biosíntesis , Transactivadores/genética , Factores de Transcripción
15.
Biochem Biophys Res Commun ; 487(3): 500-508, 2017 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-28385531

RESUMEN

Responding to pro-metastatic cues such as low oxygen tension, cancer cells develop several different strategies to facilitate migration and invasion. During this process, expression levels of matrix metalloproteinases (MMPs) are up-regulated so that cancer cells can more easily enter or exit the circulation. In this report we show that message levels of the transcriptional modulator MKL1 were elevated in malignant forms of ovarian cancer tissues in humans when compared to more benign forms accompanying a similar change in MMP2 expression. MKL1 silencing blocked hypoxia-induced migration and invasion of ovarian cancer cells (SKOV-3) in vitro. Over-expression of MKL1 activated while MKL1 depletion repressed MMP2 transcription in SKOV-3 cells. MKL1 was recruited to the MMP2 promoter by NF-κB in response to hypoxia. Mechanistically, MKL1 recruited a histone methyltransferase, SET1, and a chromatin remodeling protein, BRG1, and coordinated their interaction to alter the chromatin structure surrounding the MMP2 promoter leading to transcriptional activation. Both BRG1 and SET1 were essential for hypoxia-induced MMP2 trans-activation. Finally, expression levels of SET1 and BRG1 were positively correlated with ovarian cancer malignancies in humans. Together, our data suggest that MKL1 promotes ovarian cancer cell migration and invasion by epigenetically activating MMP2 transcription.


Asunto(s)
Movimiento Celular , Epigénesis Genética , Metaloproteinasa 2 de la Matriz/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Transactivadores/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasias Ováricas/metabolismo , Transactivadores/genética
16.
BMC Nephrol ; 18(1): 320, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29065889

RESUMEN

BACKGROUND: Caveolae are membrane invaginations measuring 50-100 nm. These organelles, composed of caveolin and cavin proteins, are important for cellular signaling and survival. Caveolae play incompletely defined roles in human kidneys. Induction of caveolin-1/CAV1 in diseased tubules has been described previously, but the responsible mechanism remains to be defined. METHODS: Healthy and atrophying human kidneys were stained for caveolar proteins, (caveolin 1-3 and cavin 1-4) and examined by electron microscopy. Induction of caveolar proteins was studied in isolated proximal tubules and primary renal epithelial cells. These cells were challenged with hypoxia or H2O2. Primary tubular cells were also subjected to viral overexpression of megakaryoblastic leukemia 1 (MKL1) and MKL1 inhibition by the MKL1 inhibitor CCG-1423. Putative coregulators of MKL1 activity were investigated by Western blotting for suppressor of cancer cell invasion (SCAI) and filamin A (FLNA). Finally, correlative bioinformatic studies of mRNA expression of caveolar proteins and MKL1 were performed. RESULTS: In healthy kidneys, caveolar proteins were expressed by the parietal epithelial cells (PECs) of Bowman's capsule, endothelial cells and vascular smooth muscle. Electron microscopy confirmed caveolae in the PECs. No expression was seen in proximal tubules. In contrast, caveolar proteins were expressed in proximal tubules undergoing atrophy. Caveolar proteins were also induced in cultures of primary epithelial tubular cells. Expression was not enhanced by hypoxia or free radical stress (H2O2), but proved sensitive to inhibition of MKL1. Viral overexpression of MKL1 induced caveolin-1/CAV1, caveolin-2/CAV2 and SDPR/CAVIN2. In kidney tissue, the mRNA level of MKL1 correlated with the mRNA levels for caveolin-1/CAV1, caveolin-2/CAV2 and the archetypal MKL1 target tenascin C (TNC), as did the MKL1 coactivator FLNA. Costaining for TNC as readout for MKL1 activity demonstrated overlap with caveolin-1/CAV1 expression in PECs as well as in atrophic segments of proximal tubules. CONCLUSIONS: Our findings support the view that MKL1 contributes to the expression of caveolar proteins in healthy kidneys and orchestrates the induction of tubular caveolar proteins in renal injury.


Asunto(s)
Lesión Renal Aguda/metabolismo , Caveolina 1/biosíntesis , Túbulos Renales Proximales/metabolismo , Proteínas de Unión al ARN/biosíntesis , Transactivadores/fisiología , Lesión Renal Aguda/inducido químicamente , Caveolas/efectos de los fármacos , Caveolas/metabolismo , Caveolas/ultraestructura , Caveolina 1/genética , Células Cultivadas , Expresión Génica , Humanos , Peróxido de Hidrógeno/toxicidad , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Túbulos Renales/ultraestructura , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/ultraestructura , Proteínas de Unión al ARN/genética
17.
Dev Biol ; 406(2): 109-16, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26386146

RESUMEN

Myocardin-Related Transcription Factors A and B (MRTF-A and MRTF-B) are highly homologous proteins that function as powerful coactivators of serum response factor (SRF), a ubiquitously expressed transcription factor essential for cardiac development. The SRF/MRTF complex binds to CArG boxes found in the control regions of genes that regulate cytoskeletal dynamics and muscle contraction, among other processes. While SRF is required for heart development and function, the role of MRTFs in the developing or adult heart has not been explored. Through cardiac-specific deletion of MRTF alleles in mice, we show that either MRTF-A or MRTF-B is dispensable for cardiac development and function, whereas deletion of both MRTF-A and MRTF-B causes a spectrum of structural and functional cardiac abnormalities. Defects observed in MRTF-A/B null mice ranged from reduced cardiac contractility and adult onset heart failure to neonatal lethality accompanied by sarcomere disarray. RNA-seq analysis on neonatal hearts identified the most altered pathways in MRTF double knockout hearts as being involved in cytoskeletal organization. Together, these findings demonstrate redundant but essential roles of the MRTFs in maintenance of cardiac structure and function and as indispensible links in cardiac cytoskeletal gene regulatory networks.


Asunto(s)
Redes Reguladoras de Genes/fisiología , Corazón/embriología , Morfogénesis/fisiología , Sarcómeros/fisiología , Factor de Respuesta Sérica/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Citoesqueleto/fisiología , Ecocardiografía , Corazón/fisiología , Técnicas Histológicas , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Reacción en Cadena en Tiempo Real de la Polimerasa , Sarcómeros/metabolismo , Análisis de Secuencia de ARN , Transactivadores/deficiencia , Factores de Transcripción/deficiencia
18.
J Cell Sci ; 127(Pt 5): 1079-91, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24424023

RESUMEN

Cellular transformation into myofibroblasts is a central physiological process enabling tissue repair. Its deregulation promotes fibrosis and carcinogenesis. TGF-ß is the main inducer of the contractile gene program that drives myofibroblast differentiation from various precursor cell types. Crucial regulators of this transcriptional program are serum response factor (SRF) and its cofactor MKL1 (also known as MRTF-A). However, the exact mechanism of the crosstalk between TGF-ß signaling and MKL1 remains unclear. Here, we report the discovery of a novel MKL1 variant/isoform, MKL1_S, transcribed from an alternative promoter and uncover a novel translation start for the published human isoform, MKL1_L. Using a human adipose-derived mesenchymal stem cell differentiation model, we show that TGF-ß specifically upregulates MKL1_S during the initial phase of myofibroblast differentiation. We identified a functional N-terminal motif in MKL1_S that allows specific induction of a group of genes including the extracellular matrix (ECM) modifiers MMP16 and SPOCK3/testican-3. We propose that TGF-ß-mediated induction of MKL1_S initiates progression to later stages of differentiation towards a stationary myofibroblast.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Miofibroblastos/fisiología , Proteínas de Fusión Oncogénica/metabolismo , Factor de Crecimiento Transformador beta1/fisiología , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Secuencia de Bases , Núcleo Celular/metabolismo , Codón Iniciador , Proteínas de Unión al ADN/genética , Células HEK293 , Células HeLa , Humanos , Metaloproteinasa 16 de la Matriz/genética , Metaloproteinasa 16 de la Matriz/metabolismo , Datos de Secuencia Molecular , Proteínas de Fusión Oncogénica/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Transactivadores , Transcripción Genética , Regulación hacia Arriba
19.
Biochem Biophys Res Commun ; 481(1-2): 71-76, 2016 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-27825966

RESUMEN

Amyloid ß (Aß) provokes severe inflammation response in the central nervous system, which is a key risk factor for the progression of Alzheimer's disease (AD). Anti-inflammation medications shed light on treating AD. In this study, we found hypericin is a potent anti-AD constituent through anti-inflammation. Pretreatment with hypericin (5 µM and 15 µM) significantly suppresses oligomeric Aß42 (oAß42)-induced expression of interleukin-1ß (IL-1ß), interleukin-6 (IL-6), tumor necrosis factor α (TNF α) and inducible nitric oxide synthase (iNOS) and production of NO in microglia without cytotoxicity. We further found that hypericin ameliorates inflammatory response by suppressing MKL1, which is the essential cofactor of p65 during the transcription process. In an Aß injection AD mouse model, animals orally administrated hypericin (50 mg/kg) for seven days significantly decreased pro-inflammatory cytokines expression and NO production in hippocampus, meanwhile, hypericin improved oAß42-induced learning and memory impairment in mice in the Morris water maze test. Therefore, hypericin could be considered as a potential candidate for treating AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/inmunología , Inflamación/inmunología , Inflamación/prevención & control , Microglía/inmunología , Perileno/análogos & derivados , Transactivadores/inmunología , Enfermedad de Alzheimer/inducido químicamente , Péptidos beta-Amiloides , Animales , Antracenos , Línea Celular , Cognición , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/inmunología , Trastornos del Conocimiento/prevención & control , Inflamación/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Fragmentos de Péptidos , Perileno/farmacología , Resultado del Tratamiento
20.
J Mol Cell Cardiol ; 79: 157-68, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25446180

RESUMEN

Elevation of intracellular cAMP concentration has numerous vascular protective effects that are in part mediated via actin cytoskeleton-remodelling and subsequent regulation of gene expression. However, the mechanisms are incompletely understood. Here we investigated whether cAMP-induced actin-cytoskeleton remodelling modulates VSMC behaviour by inhibiting expression of CCN1. In cultured rat VSMC, CCN1-silencing significantly inhibited BrdU incorporation and migration in a wound healing assay. Recombinant CCN1 enhanced chemotaxis in a Boyden chamber. Adding db-cAMP, or elevating cAMP using forskolin, significantly inhibited CCN1 mRNA and protein expression in vitro; transcriptional regulation was demonstrated by measuring pre-spliced CCN1 mRNA and CCN1-promoter activity. Forskolin also inhibited CCN1 expression in balloon injured rat carotid arteries in vivo. Inhibiting RhoA activity, which regulates actin-polymerisation, by cAMP-elevation or pharmacologically with C3-transferase, or inhibiting its downstream kinase, ROCK, with Y27632, significantly inhibited CCN1 expression. Conversely, expression of constitutively active RhoA reversed the inhibitory effects of forskolin on CCN1 mRNA. Furthermore, CCN1 mRNA levels were significantly decreased by inhibiting actin-polymerisation with latrunculin B or increased by stimulating actin-polymerisation with Jasplakinolide. We next tested the role of the actin-dependent SRF co-factor, MKL1, in CCN1 expression. Forskolin inhibited nuclear translocation of MKL1 and binding of MKL1 to the CCN1 promoter. Constitutively-active MKL1 enhanced basal promoter activity of wild-type but not SRE-mutated CCN1; and prevented forskolin inhibition. Furthermore, pharmacological MKL-inhibition with CCG-1423 significantly inhibited CCN1 promoter activity as well as mRNA and protein expression. Our data demonstrates that cAMP-induced actin-cytoskeleton remodelling regulates expression of CCN1 through MKL1: it highlights a novel cAMP-dependent mechanism controlling VSMC behaviour.


Asunto(s)
Citoesqueleto de Actina/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , AMP Cíclico/farmacología , Proteína 61 Rica en Cisteína/genética , Transactivadores/metabolismo , Adenosina/farmacología , Aminopiridinas/farmacología , Animales , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Proliferación Celular/efectos de los fármacos , Colforsina/farmacología , Proteína 61 Rica en Cisteína/metabolismo , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Humanos , Masculino , Mitógenos/farmacología , Modelos Biológicos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Ratas Sprague-Dawley , Factor de Respuesta Sérica/metabolismo , Factores de Transcripción , Transcripción Genética/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA