Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Int J Mol Sci ; 24(17)2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37685961

RESUMEN

The aryl hydrocarbon receptor (AHR) is a transcription factor that is commonly upregulated in pancreatic ductal adenocarcinoma (PDAC). AHR hinders the shuttling of human antigen R (ELAVL1) from the nucleus to the cytoplasm, where it stabilises its target messenger RNAs (mRNAs) and enhances protein expression. Among these target mRNAs are those induced by gemcitabine. Increased AHR expression leads to the sequestration of ELAVL1 in the nucleus, resulting in chemoresistance. This study aimed to investigate the interaction between AHR and ELAVL1 in the pathogenesis of PDAC in vitro. AHR and ELAVL1 genes were silenced by siRNA transfection. The RNA and protein were extracted for quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot (WB) analysis. Direct binding between the ELAVL1 protein and AHR mRNA was examined through immunoprecipitation (IP) assay. Cell viability, clonogenicity, and migration assays were performed. Our study revealed that both AHR and ELAVL1 inter-regulate each other, while also having a role in cell proliferation, migration, and chemoresistance in PDAC cell lines. Notably, both proteins function through distinct mechanisms. The silencing of ELAVL1 disrupts the stability of its target mRNAs, resulting in the decreased expression of numerous cytoprotective proteins. In contrast, the silencing of AHR diminishes cell migration and proliferation and enhances cell sensitivity to gemcitabine through the AHR-ELAVL1-deoxycytidine kinase (DCK) molecular pathway. In conclusion, AHR and ELAVL1 interaction can form a negative feedback loop. By inhibiting AHR expression, PDAC cells become more susceptible to gemcitabine through the ELAVL1-DCK pathway.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Proteína 1 Similar a ELAV/genética , Gemcitabina , Páncreas , Hormonas Pancreáticas , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Receptores de Hidrocarburo de Aril/genética , ARN Mensajero/genética , Desoxicitidina Quinasa/efectos de los fármacos , Desoxicitidina Quinasa/metabolismo , Neoplasias Pancreáticas
2.
Int J Mol Sci ; 21(23)2020 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-33260776

RESUMEN

Aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, which mediates the effects of a variety of environmental stimuli in multiple tissues. Recent advances in AHR biology have underlined its importance in cells with high developmental potency, including pluripotent stem cells. Nonetheless, there is little data on AHR expression and its role during the initial stages of stem cell differentiation. The purpose of this study was to investigate the temporal pattern of AHR expression during directed differentiation of human embryonic stem cells (hESC) into neural progenitor, early mesoderm and definitive endoderm cells. Additionally, we investigated the effect of the AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the gene expression profile in hESCs and differentiated cells by RNA-seq, accompanied by identification of AHR binding sites by ChIP-seq and epigenetic landscape analysis by ATAC-seq. We showed that AHR is differentially regulated in distinct lineages. We provided evidence that TCDD alters gene expression patterns in hESCs and during early differentiation. Additionally, we identified novel potential AHR target genes, which expand our understanding on the role of this protein in different cell types.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias Humanas/citología , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Biomarcadores , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Cuerpos Embrioides/citología , Cuerpos Embrioides/efectos de los fármacos , Genoma Humano , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ligandos
3.
Allergy ; 74(11): 2146-2156, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31049964

RESUMEN

BACKGROUND: miR-10a-5p has been shown to regulate cancer cell proliferation and invasiveness and endothelial cell inflammatory responses. The function of miR-10a-5p in the skin has not been previously studied. The aim of the current study was to examine miR-10a-5p expression, regulation, and function in keratinocytes (KCs) in association with atopic dermatitis (AD). METHODS: The expression of miR-10a-5p and its target genes was analyzed using RT-qPCR, mRNA array analysis, in situ hybridization, and immunofluorescence. The transfection of miRNA mimics, cell cycle distribution analysis, and luciferase assays was used to study miR-10a-5p functions in human primary KCs. RESULTS: miR-10a-5p was found to be upregulated in lesional skin from patients with AD and in proliferating KCs. Array and pathway analysis of IL-1ß-stimulated KCs revealed that miR-10a-5p inhibited many genes that affect cell cycle progression and only a few inflammation-related genes. Accordingly, fewer cells in S-phase and reduced proliferation were detected as characteristics of miR-10a-5p-transfected KCs. The influence of miR-10a-5p on cell proliferation was also evident in KCs induced by AD-related cytokines, including IL-4, IL-17, and IL-1ß, as measured by the capacity to strongly suppress the expression of the proliferation marker Ki-67. Among AD-related putative direct target genes, we verified hyaluronan synthase 3, a damage-associated positive regulator of KC migration and proliferation, as a direct target of miR-10a-5p. CONCLUSIONS: miR-10a-5p inhibits KC proliferation and directly targets hyaluronan synthase 3 and thereby may modulate AD-associated processes in the skin.


Asunto(s)
Dermatitis Atópica/etiología , Dermatitis Atópica/metabolismo , Regulación de la Expresión Génica , Queratinocitos/metabolismo , MicroARNs/genética , Interferencia de ARN , Adulto , Ciclo Celular/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Citocinas/metabolismo , Dermatitis Atópica/patología , Susceptibilidad a Enfermedades , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Piel/inmunología , Piel/metabolismo , Piel/patología , Adulto Joven
4.
Int J Mol Sci ; 20(12)2019 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-31234584

RESUMEN

The aryl hydrocarbon receptor (Ahr) is a ligand-activated transcription factor primarily known for its toxicological functions. Recent studies have established its importance in many physiological processes including female reproduction, although there is limited data about the precise mechanisms how Ahr itself is regulated during ovarian follicle maturation. This study describes the expression of Ahr in ovarian granulosa cells (GCs) of immature mice in a gonadotropin-dependent manner. We show that Ahr upregulation in vivo requires both follicle stimulating hormone (FSH) and luteinizing hormone (LH) activities. FSH alone increased Ahr mRNA, but had no effect on Ahr protein level, implicating a possible LH-dependent post-transcriptional regulation. Also, the increase in Ahr protein is specific to large antral follicles in induced follicle maturation. We show that Ahr expression in GCs of mid-phase follicular maturation is downregulated by protein kinase A (PKA) signaling and activation of Ahr promoter is regulated by chromatin remodeling.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Epigénesis Genética , Hormona Folículo Estimulante/metabolismo , Regulación de la Expresión Génica , Células de la Granulosa/metabolismo , Hormona Luteinizante/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Animales , Biomarcadores , Cromatina/genética , Cromatina/metabolismo , Femenino , Ratones , Folículo Ovárico/metabolismo , Receptores de Hidrocarburo de Aril/genética , Transcripción Genética
5.
Biochim Biophys Acta Gen Subj ; 1861(8): 2146-2154, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28552560

RESUMEN

Recent studies have shown that cellular bioenergetics may be involved in stem cell differentiation. Considering that during cancerogenesis cells acquire numerous properties of stem cells, it is possible to assume that the energy metabolism in tumorigenic cells might be differently regulated. The aim of this study was to compare the mitochondrial bioenergetic profile of normal pluripotent human embryonic stem cells (hESC) and relatively nullipotent embryonal carcinoma cells (2102Ep cell line). We examined three parameters related to cellular bioenergetics: phosphotransfer system, aerobic glycolysis, and oxygen consumption. Activities and expression levels of main enzymes that facilitate energy transfer were measured. The oxygen consumption rate studies were performed to investigate the respiratory capacity of cells. 2102Ep cells showed a shift in energy distribution towards adenylate kinase network. The total AK activity was almost 3 times higher in 2102Ep cells compared to hESCs (179.85±5.73 vs 64.39±2.55mU/mg of protein) and the expression of AK2 was significantly higher in these cells, while CK was downregulated. 2102Ep cells displayed reduced levels of oxygen consumption and increased levels of aerobic glycolysis compared to hESCs. The compromised respiration of 2102Ep cells is not the result of increased mitochondrial mass, increased proton leak, and reduced respiratory reserve capacity of the cells or impairment of respiratory chain complexes. Our data showed that the bioenergetic profile of 2102Ep cells clearly distinguishes them from normal hESCs. This should be considered when this cell line is used as a reference, and highlight the importance of further research concerning energy metabolism of stem cells.


Asunto(s)
Células Madre de Carcinoma Embrionario/metabolismo , Metabolismo Energético , Células Madre Embrionarias Humanas/metabolismo , Consumo de Oxígeno , Adenilato Quinasa/análisis , Línea Celular Tumoral , Creatina Quinasa/análisis , Glucólisis , Humanos , Mitocondrias/metabolismo
6.
Cancers (Basel) ; 15(18)2023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37760608

RESUMEN

Pancreatic cancer, particularly pancreatic ductal adenocarcinoma (PDAC), has an immune suppressive environment that allows tumour cells to evade the immune system. The aryl-hydrocarbon receptor (AHR) is a transcription factor that can be activated by certain exo/endo ligands, including kynurenine (KYN) and other tryptophan metabolites. Once activated, AHR regulates the expression of various genes involved in immune responses and inflammation. Previous studies have shown that AHR activation in PDAC can have both pro-tumorigenic and anti-tumorigenic effects, depending on the context. It can promote tumour growth and immune evasion by suppressing anti-tumour immune responses or induce anti-tumour effects by enhancing immune cell function. In this study involving 30 PDAC patients and 30 healthy individuals, peripheral blood samples were analysed. PDAC patients were categorized into Low (12 patients) and High/Medium (18 patients) AHR groups based on gene expression in peripheral blood mononuclear cells (PBMCs). The Low AHR group showed distinct immune characteristics, including increased levels of immune-suppressive proteins such as PDL1, as well as alterations in lymphocyte and monocyte subtypes. Functional assays demonstrated changes in phagocytosis, nitric oxide production, and the expression of cytokines IL-1, IL-6, and IL-10. These findings indicate that AHR's expression level has a crucial role in immune dysregulation in PDAC and could be a potential target for early diagnostics and personalised therapeutics.

7.
Biol Reprod ; 86(3): 77, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22116805

RESUMEN

The aryl hydrocarbon receptor (AHR) mediates the toxicity of a variety of environmental chemicals. Apart from this, an understanding is emerging that the AHR has a fundamental role in female reproduction. Evidence suggests that AHR participates in regulation of follicle-stimulating hormone receptor (Fshr) transcript level in mouse ovary by binding to the promoter of this gene in vivo. The purpose of this study was to demonstrate the molecular interplay of the Fshr promoter involved in the transactivation by AHR in mouse granulosa cells. We found that AHR activates the Fshr promoter through the region from -209 to -99 bp. In this region, the importance of the E-box motif was revealed by site-directed mutagenesis followed by promoter analysis. By focusing on the DNA/protein interactions, we defined the fact that the intact E-box but not upstream transcription factor 1 (USF1), which is known to bind this motif, is necessary for AHR binding to mouse Fshr promoter. Furthermore, by constructing AHR mutants defective in DNA interaction, we confirmed the importance of DNA binding for AHR's ability to bind to and activate Fshr promoter. Collectively, the present study demonstrates that AHR modulates Fshr transactivation by its direct association through an E-box and not by recruitment via interaction with USFs. These observations suggest that although AHR and USF may respond to different signals, they compete on binding to the same E-box. Our data, together with that from one prior study suggesting involvement of E-box motif in AHR-mediated transcription, provide novel understanding of the way in which AHR may regulate its target genes through E-box sites.


Asunto(s)
Elementos E-Box/fisiología , Regiones Promotoras Genéticas/fisiología , Receptores de Hidrocarburo de Aril/fisiología , Receptores de HFE/fisiología , Activación Transcripcional/fisiología , Animales , Secuencia de Bases , Femenino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Datos de Secuencia Molecular , Mutación/genética , Unión Proteica/fisiología , Receptores de Hidrocarburo de Aril/genética , Factores Estimuladores hacia 5'/fisiología
8.
Methods Mol Biol ; 2383: 579-585, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34766315

RESUMEN

The unique properties of human embryonic stem (hES) cells render them invaluable for many scientific and clinical endeavors. Wider application of hES cells requires a comprehensive understanding of their biology that can be dissected using RNA interference-based gene silencing. However, commonly used transfection methods to deliver nucleic acids into a cell often lead to differentiation of hES cells. For that reason, effective transfection technique with minimal side effects is essential for studying and employing hES cells. Here, we describe a CPP-based method for targeted gene silencing in hES cells using siRNA complexed with PepFect 14 (PF14). This approach results in effective downregulation of mRNA and protein levels of a target gene without adverse effects on cell viability and pluripotency.


Asunto(s)
Células Madre Embrionarias Humanas , Silenciador del Gen , Humanos , Interferencia de ARN , ARN Interferente Pequeño/genética , Transfección
9.
PLoS One ; 16(1): e0245348, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33471801

RESUMEN

The ability of butyrate to promote differentiation of cancer cells has important implication for colorectal cancer (CRC) prevention and therapy. In this study, we examined the effect of sodium butyrate (NaBT) on the energy metabolism of colon adenocarcinoma Caco-2 cells coupled with their differentiation. NaBT increased the activity of alkaline phosphatase indicating differentiation of Caco-2 cells. Changes in the expression of pluripotency-associated markers OCT4, NANOG and SOX2 were characterized during the induced differentiation at mRNA level along with the measures that allowed distinguishing the expression of different transcript variants. The functional activity of mitochondria was studied by high-resolution respirometry. Glycolytic pathway and phosphotransfer network were analyzed using enzymatical assays. The treatment of Caco-2 cells with NaBT increased production of ATP by oxidative phosphorylation, enhanced mitochondrial spare respiratory capacity and caused rearrangement of the cellular phosphotransfer networks. The flexibility of phosphotransfer networks depended on the availability of glutamine, but not glucose in the cell growth medium. These changes were accompanied by suppressed cell proliferation and altered gene expression of the main pluripotency-associated transcription factors. This study supports the view that modulating cell metabolism through NaBT can be an effective strategy for treating CRC. Our data indicate a close relationship between the phosphotransfer performance and metabolic plasticity of CRC, which is associated with the cell differentiation state.


Asunto(s)
Antineoplásicos/farmacología , Ácido Butírico/farmacología , Neoplasias del Colon/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Células CACO-2 , Diferenciación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Humanos
10.
Mol Cell Endocrinol ; 510: 110816, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32294491

RESUMEN

Human granulosa cells acquired as leftover from IVF treatment can be used to study infertility problems and are a valuable tool in the research of follicle maturation and ovulation. There is a need for more defined and long-term culture protocols for studying the response of granulosa cells upon treatment with selected hormones/chemicals. In the current study, we tested the effect of adding FGF2, IGF2 and FSH into defined basal medium in order to find culture conditions that would support proliferation of cumulus and mural granulosa cells along with the expression of common granulosa cell type markers such as FSHR, AMHR2, LHR and CYP19A1. We found that FGF2, IGF2 together with FSH helped to retain granulosa cell marker expression while supporting cell survival at least for two weeks of culture. The defined serum-free culture conditions for long-term culturing will be valuable in providing new standards in the research of human granulosa cells.


Asunto(s)
Técnicas de Cultivo de Célula , Factor 2 de Crecimiento de Fibroblastos/farmacología , Hormona Folículo Estimulante/farmacología , Células de la Granulosa/citología , Factor II del Crecimiento Similar a la Insulina/farmacología , Biomarcadores/metabolismo , Recuento de Células , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo/farmacología , Medio de Cultivo Libre de Suero , Células del Cúmulo/efectos de los fármacos , Células del Cúmulo/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Humanos
11.
Mol Ther ; 16(1): 115-123, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28192705

RESUMEN

The tumor suppressor p14ARF is widely deregulated in many types of cancers and is believed to function as a failsafe mechanism, inhibiting proliferation and inducing apoptosis as cellular response to a high oncogene load. We have found that a 22-amino-acid-long peptide derived from the N-terminal part of p14ARF, denoted ARF(1-22), which has previously been shown to mimic the function of p14ARF, has cell-penetrating properties. This peptide is internalized to the same extent as the cell-penetrating peptide (CPP) TP10 and dose-dependently decreases proliferation in MCF-7 and MDA MB 231 cells. Uptake of the ARF(1-22) peptide is associated with low membrane disturbance, measured by deoxyglucose and lactate dehydrogenase (LDH) leakage, as compared to its scrambled peptide. Also, flow cytometric analysis of annexin V/propidium iodide (PI) binding and Hoechst staining of nuclei suggest that ARF(1-22) induces apoptosis, whereas scrambled or inverted peptide sequences have no effect. The ARF(1-22) peptide mainly translocates cells through endocytosis, and is found intact inside cells for at least 3 hours. To our knowledge, this is the first time a CPP having pro-apoptopic activity has been designed from a protein.

12.
Mol Ther ; 16(1): 115-23, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17984975

RESUMEN

The tumor suppressor p14ARF is widely deregulated in many types of cancers and is believed to function as a failsafe mechanism, inhibiting proliferation and inducing apoptosis as cellular response to a high oncogene load. We have found that a 22-amino-acid-long peptide derived from the N-terminal part of p14ARF, denoted ARF(1-22), which has previously been shown to mimic the function of p14ARF, has cell-penetrating properties. This peptide is internalized to the same extent as the cell-penetrating peptide (CPP) TP10 and dose-dependently decreases proliferation in MCF-7 and MDA MB 231 cells. Uptake of the ARF(1-22) peptide is associated with low membrane disturbance, measured by deoxyglucose and lactate dehydrogenase (LDH) leakage, as compared to its scrambled peptide. Also, flow cytometric analysis of annexin V/propidium iodide (PI) binding and Hoechst staining of nuclei suggest that ARF(1-22) induces apoptosis, whereas scrambled or inverted peptide sequences have no effect. The ARF(1-22) peptide mainly translocates cells through endocytosis, and is found intact inside cells for at least 3 hours. To our knowledge, this is the first time a CPP having pro-apoptopic activity has been designed from a protein.


Asunto(s)
Permeabilidad de la Membrana Celular , Citotoxinas/síntesis química , Citotoxinas/fisiología , Péptidos/síntesis química , Péptidos/fisiología , Proteína p14ARF Supresora de Tumor/química , Secuencia de Aminoácidos , Apoptosis/genética , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/genética , Proliferación Celular , Citotoxinas/genética , Células HeLa , Humanos , Imitación Molecular/genética , Datos de Secuencia Molecular , Ácidos Nucleicos de Péptidos/síntesis química , Ácidos Nucleicos de Péptidos/genética , Ácidos Nucleicos de Péptidos/fisiología , Péptidos/genética , Proteína p14ARF Supresora de Tumor/genética , Proteína p14ARF Supresora de Tumor/metabolismo
13.
Stem Cell Res Ther ; 10(1): 43, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30678718

RESUMEN

BACKGROUND: Human embryonic stem (hES) cells serve as an invaluable tool for research and future medicine, but their transfection often leads to unwanted side effects as the method itself may induce differentiation. On the other hand, RNA interference (RNAi)-based targeted gene silencing is a quick, cost-effective, and easy-to-perform method to address questions regarding the function of genes, especially when hypomorphic knockdowns are needed. Therefore, effective transfection method with minimal side effects is essential for applying RNAi to hES cells. Here, we report a highly promising approach for targeted gene silencing in hES cells with siRNA complexed with cell-penetrating peptide PepFect 14 (PF14). This strategy provides researchers with efficient tool for unraveling the functions of genes or addressing the differentiation of pluripotent stem cells. METHODS: We present a method for delivery of siRNA into hES cells with cell-penetrating peptide PF14. Accordingly, hES cells were transfected in ROCK inhibitor containing medium for 24 h right after EDTA passaging as small cell clumps. Fluorescently labeled siRNA and siRNAs targeting OCT4 or beta-2-microglobulin (B2M) mRNA sequences were used to evaluate the efficiency of transfection and silencing. Analyses were performed at various time points by flow cytometry, RT-qPCR, and immunofluorescence microscopy. RESULTS: Effective downregulation of OCT4 in 70% of treated hES cells at protein level was achieved, along with 90% reduction at mRNA level in bulk population of cells. The applicability of this low-cost and easy-to-perform method was confirmed by inducing silencing of another target not associated with hES cell pluripotency (B2M). Furthermore, we discovered that downregulation of OCT4 induces neuroectodermal differentiation accompanied by reduced expression of B2M during early stage of this lineage. CONCLUSIONS: The results demonstrate PF14 as a promising tool for studying gene function and regulatory networks in hES cells by using RNAi.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Silenciador del Gen/fisiología , Células Madre Embrionarias Humanas/metabolismo , Lipopéptidos/genética , ARN Interferente Pequeño/metabolismo , Péptidos de Penetración Celular/genética , Humanos , Transfección
14.
Biol Open ; 7(11)2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30385434

RESUMEN

The maintenance of the pluripotency of human embryonic stem (hES) cells requires special conditions for culturing. These conditions include specific growth factors containing media and extracellular matrix (ECM) or an appropriate substrate for adhesion. Interactions between the cells and ECM are mediated by integrins, which interact with the components of ECM in active conformation. This study focused on the characterisation of the role of integrin ß1 in the adhesion, migration and differentiation of hES cells. Blocking integrin ß1 abolished the adhesion of hES cells, decreasing their survival and pluripotency. This effect was in part rescued by the inhibition of RhoA signalling with Y-27632. The presence of Y-27632 increased the migration of hES cells and supported their differentiation into embryoid bodies. The differences in integrin ß1 recycling in the phosphorylation of the myosin light chain and in the localisation of TSC2 were observed between the hES cells growing as a single-cell culture and in a colony. The hES cells at the centre and borders of the colony were found to have differences in their morphology, migration and signalling network activity. We concluded that the availability of integrin ß1 was essential for the contraction, migration and differentiation ability of hES cells.

15.
Thromb Res ; 120(6): 911-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17376515

RESUMEN

In up to 25% haemophilia A patients, the administration of coagulation factor VIII (FVIII) preparations for treatment of haemorrhages results in production of factor VIII specific antibodies. Plasma-derived FVIII preparations contain other plasma proteins, which may modulate the immune response to FVIII. We used FVIII-deficient mice to assess the role of von Willebrand factor (VWF) and cytokine transforming growth factor beta-1 (TGF-beta1) in the immune response against FVIII. Using the FVIII and FVIII in complex with VWF purified from the plasma-derived FVIII preparation, we demonstrated that a lower concentration of FVIII antibody was induced in FVIII-VWF-treated mice compared to FVIII-treated mice (p<0.05). The addition of recombinant latent TGF-beta1 to FVIII decreased the antibody response against FVIII compared to FVIII treatment alone (p<0.01). The obtained results suggest that VWF and latent TGF-beta1 present in plasma-derived FVIII preparations reduce the immune response against FVIII. However, we cannot exlude possible modulatory effects of other plasma proteins.


Asunto(s)
Factor VIII/inmunología , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Factor de von Willebrand/inmunología , Traslado Adoptivo , Animales , Anticuerpos/sangre , Linfocitos B/citología , Linfocitos B/inmunología , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , División Celular/inmunología , Células Cultivadas , Cromatografía , Factor VIII/genética , Factor VIII/metabolismo , Factor VIII/farmacología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Endogámicos , Ratones Mutantes , Bazo/citología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Factor de von Willebrand/metabolismo , Factor de von Willebrand/farmacología
16.
Stem Cells Int ; 2016: 1580701, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27247576

RESUMEN

The lipophilic statin lovastatin decreases cholesterol synthesis and is a safe and effective treatment for the prevention of cardiovascular diseases. Growing evidence points at antitumor potential of lovastatin. Therefore, understanding the molecular mechanism of lovastatin function in different cell types is critical to effective therapy design. In this study, we investigated the effects of lovastatin on the differentiation potential of human embryonic stem (hES) cells (H9 cell line). Multiparameter flow cytometric assay was used to detect changes in the expression of transcription factors characteristic of hES cells. We found that lovastatin treatment delayed NANOG downregulation during ectodermal and endodermal differentiation. Likewise, expression of ectodermal (SOX1 and OTX2) and endodermal (GATA4 and FOXA2) markers was higher in treated cells. Exposure of hES cells to lovastatin led to a minor decrease in the expression of SSEA-3 and a significant reduction in CD133 expression. Treated cells also formed fewer embryoid bodies than control cells. By analyzing hES with and without CD133, we discovered that CD133 expression is required for proper formation of embryoid bodies. In conclusion, lovastatin reduced the heterogeneity of hES cells and impaired their differentiation potential.

17.
J Control Release ; 235: 195-204, 2016 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-27269729

RESUMEN

The skin is a difficult to access tissue for efficient delivery of large and/or charged macromolecules, including therapeutic DNA and RNA oligonucleotides. Cell-penetrating peptide PepFect6 (PF6) has been shown to be suitable transport vehicle for siRNAs in cell culture and systemically in vivo in mice. MiR-146a is known as anti-inflammatory miRNA that inhibits multiple factors from the nuclear factor (NF)-κB pathway in various cell types, including keratinocytes. In this study, PF6 was shown to form unimodal nanocomplexes with miR-146a mimic that entered into human primary keratinocytes, where miR-146a inhibited the expression of its direct targets from the NF-κB pathway and the genes known to be activated by NF-κB, C-C motif ligand (CCL)5 and interleukin (IL)-8. The transfection of miR-146a mimic with PF6 was more efficient in sub-confluent keratinocyte cultures, affected keratinocyte proliferation less and had similar effect on cell viability when compared with a lipid based agent. Subcutaneous pre-administration of PF6-miR-146a nanocomplexes attenuated ear-swelling and reduced the expression of pro-inflammatory cytokines and chemokines IL-6, CCL11, CCL24 and C-X-C motif ligand 1 (CXCL1) in a mouse model of irritant contact dermatitis. Our data demonstrates that PF6-miR-146a nanoparticles might have potential in the development of therapeutics to target inflammatory skin diseases.


Asunto(s)
Antiinflamatorios/administración & dosificación , Queratinocitos/efectos de los fármacos , Lipopéptidos/administración & dosificación , MicroARNs/administración & dosificación , Nanopartículas/administración & dosificación , Quinolinas/administración & dosificación , Animales , Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/inmunología , Dermatitis por Contacto/tratamiento farmacológico , Dermatitis por Contacto/inmunología , Modelos Animales de Enfermedad , Humanos , Irritantes , Queratinocitos/metabolismo , Lipopéptidos/química , Lipopéptidos/uso terapéutico , Ratones Endogámicos C57BL , MicroARNs/química , MicroARNs/genética , MicroARNs/uso terapéutico , Microscopía Electrónica de Transmisión , Nanopartículas/química , Nanopartículas/uso terapéutico , Nanopartículas/ultraestructura , Quinolinas/química , Quinolinas/uso terapéutico , Acetato de Tetradecanoilforbol
18.
Oncogene ; 23(14): 2523-30, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-14676827

RESUMEN

CD43 or leukosialin is a transmembrane sialoglycoprotein, whose extracellular domain participates in cell adhesiveness and the cytoplasmic tail regulates a variety of intracellular signal transduction pathways involved in cell proliferation. CD43 is abundantly expressed on the surface of hematopoietic cells, but CD43 expression is also frequently found in the tumor cells of nonhematopoietic origin. In the early stages of some tumors, the accumulation of tumor suppressor protein p53 has been described. Here, we show that the expression of CD43 causes the induction of functionally active p53 protein. Moreover, we found that the activation of p53 by CD43 is mediated by tumor suppressor protein ARF. The coexpression of CD43 and ARF in ARF-null mouse embryonic fibroblasts resulted in programmed cell death, but that was not the case when CD43 alone was expressed in these cells. These data provide the first evidence of the connection between p53- and CD43-dependent pathways.


Asunto(s)
Factor 1 de Ribosilacion-ADP/genética , Factor 1 de Ribosilacion-ADP/metabolismo , Antígenos CD , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis , Biomarcadores , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Genes Supresores de Tumor , Humanos , Leucosialina , Ratones , Modelos Biológicos
19.
Oncogene ; 23(37): 6175-85, 2004 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-15208667

RESUMEN

The p53 protein is a central player in cellular response to DNA damage. Induction of p53 by DNA-damaging agents involves elevation of its steady-state level and activation of its potency as a transcription factor. In the cell population, these responses can occur either homogeneously (where every single cell responds simultaneously and similarly to its neighbor) or heterogeneously (where only some cells of a population respond and the number of these increases with increasing dose of inducer). We have studied here the p53 response to DNA-damaging agents (camptothecin, mitomycin C) in individual cells. We show that the level of p53 protein is increased in every single cell of the population homogeneously, while the p53-dependent transcription can be subject to an on/off-type response. Depending on the structure of the target promoter, p53-dependent transcription can be regulated according to the binary or graded model. The on/off-type transcriptional activation pattern of p53 defines two distinct subpopulations of cells after DNA damage.


Asunto(s)
Mutágenos/toxicidad , Transcripción Genética/fisiología , Proteína p53 Supresora de Tumor/fisiología , Secuencia de Bases , Western Blotting , Daño del ADN , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética
20.
PLoS One ; 10(9): e0138346, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26378917

RESUMEN

Laminin isoforms laminin-511 and -521 are expressed by human embryonic stem cells (hESC) and can be used as a growth matrix to culture these cells under pluripotent conditions. However, the expression of these laminins during the induction of hESC differentiation has not been studied in detail. Furthermore, the data regarding the expression pattern of laminin chains in differentiating hESC is scarce. In the current study we aimed to fill this gap and investigated the potential changes in laminin expression during early hESC differentiation induced by retinoic acid (RA). We found that laminin-511 but not -521 accumulates in the committed cells during early steps of hESC differentiation. We also performed a comprehensive analysis of the laminin chain repertoire and found that pluripotent hESC express a more diverse range of laminin chains than shown previously. In particular, we provide the evidence that in addition to α1, α5, ß1, ß2 and γ1 chains, hESC express α2, α3, ß3, γ2 and γ3 chain proteins and mRNA. Additionally, we found that a variant of laminin α3 chain-145 kDa-accumulated in RA-treated hESC showing that these cells produce prevalently specifically modified version of α3 chain in early phase of differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/fisiología , Laminina/metabolismo , Animales , Células Cultivadas , Humanos , Ratones , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA