Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
2.
Rheumatology (Oxford) ; 61(6): 2694-2703, 2022 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-34559194

RESUMEN

OBJECTIVE: JIA is a chronic inflammatory disease of unknown origin. The regulation of inflammatory processes involves multiple cellular steps including mRNA transcription and translation. Different miRNAs control these processes tightly. We aimed to determine the roles of specific miRNAs within JIA pathogenesis. METHODS: We performed a global miRNA expression analysis in parallel in cells from the arthritic joint and peripheral blood of oligoarticular JIA patients and healthy controls. Quantitative RT-PCR analysis was used to verify expression of miRNA in T cells. Ex vivo experiments and flow cytometric analyses were used to analyse proliferation and redox metabolism. RESULTS: Global miRNA expression analysis demonstrated a different composition of miRNA expression at the site of inflammation compared with peripheral blood. Bioinformatic analysis of predicted miRNA target genes suggest a huge overrepresentation of genes involved in metabolic and oxidative stress pathways in the inflamed joint. Despite enhanced reactive oxygen species (ROS) levels within the local inflammatory milieu, JIA T cells are hyperproliferative and reveal an overexpression of miR-23a, which is an inhibitor of Peptidyl-prolyl isomerase F (PPIF), the regulator of mitochondrial ROS escape. Mitochondrial ROS escape is diminished in JIA T cells, resulting in their prolonged survival. CONCLUSION: Our data suggest that miRNA-dependent mitochondrial ROS shuttling might be a mechanism that contributes to T cell regulation in JIA at the site of inflammation.


Asunto(s)
Artritis Juvenil , MicroARNs , Humanos , Inflamación/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/metabolismo
3.
J Neurosci Res ; 99(9): 2172-2187, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34110645

RESUMEN

Several eye diseases, for example, retinal artery occlusion, diabetic retinopathy, and glaucoma, are associated with retinal hypoxia. The lack of oxygen in the retina, especially in retinal ganglion cells (RGCs), causes cell damage up to cell degeneration and leads to blindness. Using multielectrode array recordings, an ex vivo hypoxia acute model was established to analyze the electrical activity of murine wild-type retinae under hypoxic stress conditions. Hypoxia was induced by exchanging the perfusion with oxygen-saturated medium by nitrogen-saturated medium. Hypoxic periods of 0 min (control) up to 60 min were tested on the retinae of adult female C57BL/6J mice. The electrical RGC activity vanished during hypoxia, but conditionally returned after the reestablishment of conventional test conditions. With increasing duration of hypoxia, the returning RGC activity decreased. After a hypoxic period of 30 min and a subsequent recovery time of 30 min, 59.43 ± 11.35% of the initially active channels showed a restored RGC activity. The survival rate of retinal cells after hypoxic stress was analyzed by a live/dead staining assay using two-photon laser scanning microscopy. For detailed information about molecular changes caused by hypoxia, a microarray gene expression analysis was performed. Furthermore, the effect of 2-aminoethanesulfonic acid (taurine, 1 mM) on retinae under hypoxic stress was tested. Treatment with taurine resulted in an increase in the RGC response rate after hypoxia and also increased the survival rate of retinal cells under hypoxic stress, confirming its potential as promising candidate for neuroprotective therapies of eye diseases.


Asunto(s)
Potenciales de Acción/fisiología , Hipoxia de la Célula/fisiología , Retina/fisiología , Animales , Electrodos , Femenino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Células Ganglionares de la Retina/fisiología
4.
Neurobiol Dis ; 155: 105371, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33932559

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) with episodes of inflammatory demyelination and remyelination. While remyelination has been linked with functional recovery in MS patients, there is evidence of ongoing tissue damage despite complete myelin repair. In this study, we investigated the long-term consequences of an acute demyelinating white matter CNS lesion. For this purpose, acute demyelination was induced by 5-week-cuprizone intoxication in male C57BL/6 J mice, and the tissues were examined after a 7-month recovery period. While myelination and oligodendrocyte densities appeared normal, ongoing axonal degeneration and glia cell activation were found in the remyelinated corpus callosum. Neuropathologies were paralleled by subtle gait abnormalities evaluated using DigiGait™ high speed ventral plane videography. Gene array analyses revealed increased expression levels of various inflammation related genes, among protein kinase c delta (PRKCD). Immunofluorescence stains revealed predominant microglia/macrophages PRKCD expression in both, cuprizone tissues and post-mortem MS lesions. These results support the hypothesis that chronic microglia/macrophages driven tissue injury represents a key aspect of progressive neurodegeneration and functional decline in MS.


Asunto(s)
Axones/patología , Encéfalo/patología , Mediadores de Inflamación , Esclerosis Múltiple/patología , Degeneración Nerviosa/patología , Sustancia Blanca/patología , Animales , Axones/metabolismo , Encéfalo/metabolismo , Quelantes/toxicidad , Cuprizona/toxicidad , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/psicología , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/genética , Degeneración Nerviosa/psicología , Sustancia Blanca/metabolismo
5.
Sci Rep ; 11(1): 6934, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33767215

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by an expanded polyglutamine repeat in the huntingtin gene. The neuropathology of HD is characterized by the decline of a specific neuronal population within the brain, the striatal medium spiny neurons (MSNs). The origins of this extreme vulnerability remain unknown. Human induced pluripotent stem cell (hiPS cell)-derived MSNs represent a powerful tool to study this genetic disease. However, the differentiation protocols published so far show a high heterogeneity of neuronal populations in vitro. Here, we compared two previously published protocols to obtain hiPS cell-derived striatal neurons from both healthy donors and HD patients. Patch-clamp experiments, immunostaining and RT-qPCR were performed to characterize the neurons in culture. While the neurons were mature enough to fire action potentials, a majority failed to express markers typical for MSNs. Voltage-clamp experiments on voltage-gated sodium (Nav) channels revealed a large variability between the two differentiation protocols. Action potential analysis did not reveal changes induced by the HD mutation. This study attempts to demonstrate the current challenges in reproducing data of previously published differentiation protocols and in generating hiPS cell-derived striatal MSNs to model a genetic neurodegenerative disorder in vitro.


Asunto(s)
Técnicas de Cultivo de Célula , Diferenciación Celular , Enfermedad de Huntington , Neuronas/fisiología , Potenciales de Acción , Animales , Calcio/metabolismo , Estudios de Casos y Controles , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas , Ratones Endogámicos C57BL , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/metabolismo , Ácido gamma-Aminobutírico/metabolismo
6.
Dis Model Mech ; 14(2)2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33408083

RESUMEN

Activin/myostatin signalling acts to induce skeletal muscle atrophy in adult mammals by inhibiting protein synthesis as well as promoting protein and organelle turnover. Numerous strategies have been successfully developed to attenuate the signalling properties of these molecules, which result in augmenting muscle growth. However, these molecules, in particular activin, play major roles in tissue homeostasis in numerous organs of the mammalian body. We have recently shown that although the attenuation of activin/myostatin results in robust muscle growth, it also has a detrimental impact on the testis. Here, we aimed to discover the long-term consequences of a brief period of exposure to muscle growth-promoting molecules in the testis. We demonstrate that muscle hypertrophy promoted by a soluble activin type IIB ligand trap (sActRIIB) is a short-lived phenomenon. In stark contrast, short-term treatment with sActRIIB results in immediate impact on the testis, which persists after the sessions of the intervention. Gene array analysis identified an expansion in aberrant gene expression over time in the testis, initiated by a brief exposure to muscle growth-promoting molecules. The impact on the testis results in decreased organ size as well as quantitative and qualitative impact on sperm. Finally, we have used a drug-repurposing strategy to exploit the gene expression data to identify a compound - N6-methyladenosine - that may protect the testis from the impact of the muscle growth-promoting regime. This work indicates the potential long-term harmful effects of strategies aimed at promoting muscle growth by attenuating activin/myostatin signalling. Furthermore, we have identified a molecule that could, in the future, be used to overcome the detrimental impact of sActRIIB treatment on the testis.


Asunto(s)
Receptores de Activinas Tipo II/genética , Subunidades beta de Inhibinas/genética , Miostatina/genética , Testículo/anomalías , Testículo/efectos de los fármacos , Receptores de Activinas Tipo II/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Peso Corporal , Biología Computacional , Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Subunidades beta de Inhibinas/metabolismo , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Fenotipo , Análisis de Componente Principal , Transducción de Señal , Factores de Tiempo
7.
Aquat Toxicol ; 231: 105719, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33360234

RESUMEN

Numerous environmental pollutants have the potential to accumulate in sediments, and among them are endocrine-disrupting chemicals (EDCs). It is well documented that water-borne exposure concentrations of some potent EDCs, more specifically estrogenic- active compounds (ECs), can impair the reproduction of fish. In contrast, little is known about the bioavailability and effects of sediment-associated ECs on fish. Particularly, when sediments are disturbed, e.g., during flood events, chemicals may be released from the sediment and become bioavailable. The main objectives of this study were to evaluate a) whether ECs from the sediment become bioavailable to fish when the sediment is suspended, and b) whether such exposure leads to endocrine responses in fish. Juvenile rainbow trout (Oncorhynchus mykiss) were exposed over 21 days to constantly suspended sediments in the following treatments: i) a contaminated sediment from the Luppe River, representing a "hotspot" for EC accumulation, ii) a reference sediment (exhibiting only background contamination), iii) three dilutions, 2-, 4- and 8-fold of Luppe sediment diluted with the reference sediment, and iv) a water-only control. Measured estrogenic activity using in vitro bioassays as well as target analysis of nonylphenol and estrone via LC-MS/MS in sediment, water, fish plasma, as well as bile samples, confirmed that ECs became bioavailable from the sediment during suspension. ECs were dissolved in the water phase, as indicated by passive samplers, and were readily taken up by the exposed trout. An estrogenic response of fish to Luppe sediment was indicated by increased abundance of transcripts of typical estrogen responsive genes, i.e. vitelline envelope protein α in the liver and vitellogenin induction in the skin mucus. Altered gene expression profiles of trout in response to suspended sediment from the Luppe River suggest that in addition to ECs a number of other contaminants such as dioxins, polychlorinated biphenyls (PCBs) and heavy metals were remobilized during suspension. The results of the present study demonstrated that sediments not only function as a sink for ECs but can turn into a significant source of pollution when sediments are resuspended as during flood-events. This highlights the need for sediment quality criteria considering bioavailability sediment-bound contaminants in context of flood events.


Asunto(s)
Estrógenos/toxicidad , Sedimentos Geológicos/química , Oncorhynchus mykiss/metabolismo , Animales , Disponibilidad Biológica , Exposición a Riesgos Ambientales , Femenino , Ontología de Genes , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Vitelogeninas/metabolismo , Contaminantes Químicos del Agua/toxicidad
9.
Oncogene ; 39(44): 6856-6870, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32978523

RESUMEN

Recent findings suggested a benefit of anti-EGFR therapy for basal-like muscle-invasive bladder cancer (MIBC). However, the impact on bladder cancer with substantial squamous differentiation (Sq-BLCA) and especially pure squamous cell carcinoma (SCC) remains unknown. Therefore, we comprehensively characterized pure and mixed Sq-BLCA (n = 125) on genetic and protein expression level, and performed functional pathway and drug-response analyses with cell line models and isolated primary SCC (p-SCC) cells of the human urinary bladder. We identified abundant EGFR expression in 95% of Sq-BLCA without evidence for activating EGFR mutations. Both SCaBER and p-SCC cells were sensitive to EGFR tyrosine kinase inhibitors (TKIs: erlotinib and gefitinib). Combined treatment with anti-EGFR TKIs and varying chemotherapeutics led to a concentration-dependent synergism in SCC cells according to the Chou-Talalay method. In addition, the siRNA knockdown of EGFR impaired SCaBER viability suggesting a putative "Achilles heel" of Sq-BLCA. The observed effects seem Sq-BLCA-specific since non-basal urothelial cancer cells were characterized by poor TKI sensitivity associated with a short-term feedback response potentially attenuating anti-tumor activity. Hence, our findings give further insights into a crucial, Sq-BLCA-specific role of the ERBB signaling pathway proposing improved effectiveness of anti-EGFR based regimens in combination with chemotherapeutics in squamous bladder cancers with wild-type EGFR-overexpression.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Transicionales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Transicionales/genética , Carcinoma de Células Transicionales/patología , Línea Celular Tumoral , Estudios de Cohortes , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib/farmacología , Clorhidrato de Erlotinib/uso terapéutico , Femenino , Gefitinib/farmacología , Gefitinib/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Inhibidores de Proteínas Quinasas/uso terapéutico , ARN Interferente Pequeño/metabolismo , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Receptor ErbB-3/antagonistas & inhibidores , Receptor ErbB-3/metabolismo , Receptor ErbB-4/antagonistas & inhibidores , Receptor ErbB-4/metabolismo , Transducción de Señal/efectos de los fármacos , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología
10.
Eur J Transl Myol ; 30(1): 8737, 2020 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-32499882

RESUMEN

Numerous approaches are being developed to promote post-natal muscle growth based on attenuating Myostatin/Activin signalling for clinical uses such as the treatment neuromuscular diseases, cancer cachexia and sarcopenia. However there have been concerns about the effects of inhibiting Activin on tissues other than skeletal muscle. We intraperitoneally injected mice with the Activin ligand trap, sActRIIB, in young, adult and a progeric mouse model. Treatment at any stage in the life of the mouse rapidly increased muscle mass. However at all stages of life the treatment decreased the weights of the testis. Not only were the testis smaller, but they contained fewer sperm compared to untreated mice. We found that the hypertrophic muscle phenotype was lost after the cessation of sActRIIB treatment but abnormal testis phenotype persisted. In summary, attenuation of Myostatin/Activin signalling inhibited testis development. Future use of molecules based on a similar mode of action to promote muscle growth should be carefully profiled for adverse side-effects on the testis. However the effectiveness of sActRIIB as a modulator of Activin function provides a possible therapeutic strategy to alleviate testicular seminoma development.

11.
JCI Insight ; 5(8)2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32213712

RESUMEN

Septic cardiomyopathy is a life-threatening organ dysfunction caused by sepsis. Ribonuclease 1 (RNase 1) belongs to a group of host-defense peptides that specifically cleave extracellular RNA (eRNA). The activity of RNase 1 is inhibited by ribonuclease-inhibitor 1 (RNH1). However, the role of RNase 1 in septic cardiomyopathy and associated cardiac apoptosis is completely unknown. Here, we show that sepsis resulted in a significant increase in RNH1 and eRNA serum levels compared with those of healthy subjects. Treatment with RNase 1 resulted in a significant decrease of apoptosis, induced by the intrinsic pathway, and TNF expression in murine cardiomyocytes exposed to either necrotic cardiomyocytes or serum of septic patients for 16 hours. Additionally, treatment of septic mice with RNase 1 resulted in a reduction in cardiac apoptosis, TNF expression, and septic cardiomyopathy. These data demonstrate that eRNA plays a crucial role in the pathophysiology of the organ (cardiac) dysfunction in sepsis and that RNase and RNH1 may be new therapeutic targets and/or strategies to reduce the cardiac injury and dysfunction caused by sepsis.


Asunto(s)
Cardiomiopatías/metabolismo , Ácidos Nucleicos Libres de Células/metabolismo , Ribonucleasa Pancreática/metabolismo , Sepsis/metabolismo , Animales , Apoptosis/fisiología , Cardiomiopatías/etiología , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Proteínas/metabolismo , Sepsis/complicaciones
12.
EMBO Mol Med ; 12(3): e11021, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-31943786

RESUMEN

Kidney fibrosis is characterized by expansion and activation of platelet-derived growth factor receptor-ß (PDGFR-ß)-positive mesenchymal cells. To study the consequences of PDGFR-ß activation, we developed a model of primary renal fibrosis using transgenic mice with PDGFR-ß activation specifically in renal mesenchymal cells, driving their pathological proliferation and phenotypic switch toward myofibroblasts. This resulted in progressive mesangioproliferative glomerulonephritis, mesangial sclerosis, and interstitial fibrosis with progressive anemia due to loss of erythropoietin production by fibroblasts. Fibrosis induced secondary tubular epithelial injury at later stages, coinciding with microinflammation, and aggravated the progression of hypertensive and obstructive nephropathy. Inhibition of PDGFR activation reversed fibrosis more effectively in the tubulointerstitium compared to glomeruli. Gene expression signatures in mice with PDGFR-ß activation resembled those found in patients. In conclusion, PDGFR-ß activation alone is sufficient to induce progressive renal fibrosis and failure, mimicking key aspects of chronic kidney disease in humans. Our data provide direct proof that fibrosis per se can drive chronic organ damage and establish a model of primary fibrosis allowing specific studies targeting fibrosis progression and regression.


Asunto(s)
Enfermedades Renales , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Fibroblastos/patología , Fibrosis , Humanos , Riñón/patología , Enfermedades Renales/patología , Ratones , Ratones Transgénicos , Miofibroblastos/patología
13.
Leukemia ; 34(4): 1062-1074, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31728053

RESUMEN

Classical Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) are a heterogeneous group of hematopoietic malignancies including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). The JAK2V617F mutation plays a central role in these disorders and can be found in 90% of PV and ~50-60% of ET and PMF. Hypoxia-inducible factor 1 (HIF-1) is a master transcriptional regulator of the response to decreased oxygen levels. We demonstrate the impact of pharmacological inhibition and shRNA-mediated knockdown (KD) of HIF-1α in JAK2V617F-positive cells. Inhibition of HIF-1 binding to hypoxia response elements (HREs) with echinomycin, verified by ChIP, impaired growth and survival by inducing apoptosis and cell cycle arrest in Jak2V617F-positive 32D cells, but not Jak2WT controls. Echinomycin selectively abrogated clonogenic growth of JAK2V617F cells and decreased growth, survival, and colony formation of bone marrow and peripheral blood mononuclear cells and iPS cell-derived progenitor cells from JAK2V617F-positive patients, while cells from healthy donors were unaffected. We identified HIF-1 target genes involved in the Warburg effect as a possible underlying mechanism, with increased expression of Pdk1, Glut1, and others. That was underlined by transcriptome analysis of primary patient samples. Collectively, our data show that HIF-1 is a new potential therapeutic target in JAK2V617F-positive MPN.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Equinomicina/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Madre Pluripotentes Inducidas/patología , Janus Quinasa 2/genética , Mutación , Trastornos Mieloproliferativos/patología , Adulto , Anciano , Anciano de 80 o más Años , Antibióticos Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Ciclo Celular , Proliferación Celular , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Pronóstico , Células Tumorales Cultivadas
14.
Sci Rep ; 9(1): 13939, 2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31558805

RESUMEN

The synaptic transmission in the mammalian brain is not limited to the interplay between the pre- and the postsynapse of neurons, but involves also astrocytes as well as extracellular matrix (ECM) molecules. Glycoproteins, proteoglycans and hyaluronic acid of the ECM pervade the pericellular environment and condense to special superstructures termed perineuronal nets (PNN) that surround a subpopulation of CNS neurons. The present study focuses on the analysis of PNNs in a quadruple knockout mouse deficient for the ECM molecules tenascin-C (TnC), tenascin-R (TnR), neurocan and brevican. Here, we analysed the proportion of excitatory and inhibitory synapses and performed electrophysiological recordings of the spontaneous neuronal network activity of hippocampal neurons in vitro. While we found an increase in the number of excitatory synaptic molecules in the quadruple knockout cultures, the number of inhibitory synaptic molecules was significantly reduced. This observation was complemented with an enhancement of the neuronal network activity level. The in vivo analysis of PNNs in the hippocampus of the quadruple knockout mouse revealed a reduction of PNN size and complexity in the CA2 region. In addition, a microarray analysis of the postnatal day (P) 21 hippocampus was performed unravelling an altered gene expression in the quadruple knockout hippocampus.


Asunto(s)
Brevicano/metabolismo , Potenciales Postsinápticos Excitadores , Potenciales Postsinápticos Inhibidores , Proteínas del Tejido Nervioso/metabolismo , Proteoglicanos/metabolismo , Tenascina/metabolismo , Animales , Brevicano/genética , Región CA2 Hipocampal/metabolismo , Región CA2 Hipocampal/fisiología , Células Cultivadas , Femenino , Eliminación de Gen , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Neurocano , Proteoglicanos/genética , Sinapsis/metabolismo , Sinapsis/fisiología , Tenascina/genética
15.
Transl Oncol ; 12(9): 1237-1256, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31295651

RESUMEN

Nasopharyngeal carcinoma (NPC) is a highly malignant epithelial cancer linked to EBV infection. Addition of interferon-ß (IFNß) to chemo- and radiochemotherapy has led to survival rates >90% in children and adolescents. As NPC cells are sensitive to apoptosis via tumor necrosis factor-related apoptosis inducing ligand (TRAIL), we explored the role of TRAIL and IFNß in the killing of NPC cells by natural killer (NK) cells. NPC cells, including cells of a patient-derived xenograft were exposed to NK cells in the presence or absence of IFNß. NK cells killed NPC- but not nasoepithelial cells and killing was predominately mediated via TRAIL. Incubation of NK cells with IFNß increased cytotoxicity against NPC cells. Concomitant incubation of NK- and NPC cells with IFNß before coculture reduced cytotoxicity and could be overcome by blocking the PD-1/PD-L1 axis leading to the release of intracellular TRAIL from NK cells. In conclusion, combination of IFNß and anti-PD-1, augmenting cytotoxicity of NK cells against NPC cells, could be a strategy to improve NPC-directed therapy and warrants further evaluation in vivo.

16.
Cancer Immunol Immunother ; 68(8): 1317-1329, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31312900

RESUMEN

BACKGROUND: Nasopharyngeal carcinoma (NPC) is an EBV-associated neoplasm occurring endemically in Southeast Asia and sporadically all over the world. In children and adolescents, high cure rates have been obtained using chemotherapy, radiochemotherapy and maintenance therapy with interferon beta (IFNß). The mechanism by which IFNß contributes to a low systemic relapse rate has not yet been fully revealed. PATIENTS AND METHODS: NK cells and serum samples from two patients with NPC were analyzed before and at different time points during IFNß therapy, for assessment of TRAIL expression and NK cell cytotoxicity. Cytotoxicity was measured using the calcein release assay and the contribution of different death effector pathways was analyzed using specific inhibitors. RESULTS: Treatment with IFNß induced TRAIL expression on patients' NK cells and increased their cytotoxicity against NPC targets in vitro. NK cell-mediated cytotoxicity was predominately mediated via TRAIL. IFNß also induced the production of soluble TRAIL (sTRAIL) by NK cells and its release upon contact with NPC cells. IFNß treatment increased serum levels of sTRAIL in patients. Moreover, sTRAIL concentrated from patients' serum samples induced apoptosis ex vivo in NPC cells from a patient-derived xenograft. CONCLUSION: Increased cytotoxicity of NK cells against NPC cells and increased serum levels of biologically active TRAIL in patients treated with IFNß could be a means to eliminate micrometastatic disease and explain the low systemic relapse rate in this patient group.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/fisiología , Inmunoterapia/métodos , Interferón beta/uso terapéutico , Células Asesinas Naturales/inmunología , Carcinoma Nasofaríngeo/terapia , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Adolescente , Animales , Apoptosis , Línea Celular Tumoral , Niño , Citotoxicidad Inmunológica , Femenino , Humanos , Ratones , Ratones Desnudos , Carcinoma Nasofaríngeo/inmunología , Recurrencia Local de Neoplasia , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Mol Cell Cardiol ; 134: 51-61, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31271758

RESUMEN

Reduced shear stress resulting from disturbed blood flow can impair endothelial integrity and drive the development of vascular inflammatory lesions. Metalloproteinases of the ADAM family have been implicated in the regulation of cell survival and inflammatory responses. Here we investigate the mechanism and function of ADAM15 upregulation in primary flow cultured endothelial cells. Transcriptomic analysis indicated that within the ADAM family ADAM15 mRNA is most prominently upregulated (4-fold) when endothelial cells are exposed to physiologic shear stress. This induction was confirmed in venous, arterial and microvascular endothelial cells and is associated with increased presence of ADAM15 protein in the cell lysates (5.6-fold) and on the surface (3.1-fold). The ADAM15 promoter contains several consensus sites for the transcription factor KLF2 which is also upregulated by shear stress. Induction of endothelial KLF2 by simvastatin treatment is associated with ADAM15 upregulation (1.8-fold) which is suppressed by counteracting simvastatin with geranylgeranyl pyrophosphate. KLF2 overexpression promotes ADAM15 expression (2.1-fold) under static conditions whereas KLF2 siRNA knockdown prevents ADAM15 induction by shear stress. Functionally, ADAM15 promotes survival of endothelial cells challenged by growth factor depletion or TNF stimulation as shown by ADAM15 shRNA knockdown (1.6-fold). Exposure to shear stress increases endothelial survival while additional knockdown of ADAM15 reduces survival (6.7-fold) under flow conditions. Thus, physiologic shear stress resulting from laminar flow promotes KLF2 induced ADAM15 expression which contributes to endothelial survival. The absence of ADAM15 at low shear stress or static conditions may therefore lead to increased endothelial damage and promote vascular inflammation.


Asunto(s)
Proteínas ADAM/genética , Células Endoteliales/fisiología , Proteínas de la Membrana/genética , Regulación hacia Arriba/genética , Células Cultivadas , Endotelio Vascular/fisiología , Regulación de la Expresión Génica/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , ARN Mensajero/genética , Estrés Mecánico
18.
Sci Rep ; 9(1): 8262, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31164666

RESUMEN

Post-translational modifications of core histones participate in controlling the expression of genes. Methylation of lysine 4 of histone H3 (H3K4), together with acetylation of H3K27, is closely associated with open chromatin and gene transcription. H3K4 methylation is catalyzed by KMT2 lysine methyltransferases that include the mixed-lineage leukemia 1-4 (MLL1-4) and SET1A and B enzymes. For efficient catalysis, all six require a core complex of four proteins, WDR5, RBBP5, ASH2L, and DPY30. We report that targeted disruption of Ash2l in the murine hematopoietic system results in the death of the mice due to a rapid loss of mature hematopoietic cells. However, lin-Sca1+Kit+ (LSK) cells, which are highly enriched in hematopoietic stem and multi-potent progenitor cells, accumulated in the bone marrow. The loss of Ash2l resulted in global reduction of H3K4 methylation and deregulated gene expression, including down-regulation of many mitosis-associated genes. As a consequence, LSK cells accumulated in the G2-phase of the cell cycle and were unable to proliferate and differentiate. In conclusion, Ash2l is essential for balanced gene expression and for hematopoietic stem and multi-potent progenitor cell physiology.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Células Madre/metabolismo , Factores de Transcripción/genética , Animales , Diferenciación Celular , Proliferación Celular/genética , Cromatina/genética , Regulación de la Expresión Génica/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Lisina/genética , Metilación , Ratones
19.
Stem Cell Res Ther ; 10(1): 116, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30953537

RESUMEN

BACKGROUND: The mechanisms underpinning the regenerative capabilities of mesenchymal stem cells (MSC) were originally thought to reside in their ability to recognise damaged tissue and to differentiate into specific cell types that would replace defective cells. However, recent work has shown that molecules produced by MSCs (secretome), particularly those packaged in extracellular vesicles (EVs), rather than the cells themselves are responsible for tissue repair. METHODS: Here we have produced a secretome from adipose-derived mesenchymal stem cells (ADSC) that is free of exogenous molecules by incubation within a saline solution. Various in vitro models were used to evaluate the effects of the secretome on cellular processes that promote tissue regeneration. A cardiotoxin-induced skeletal muscle injury model was used to test the regenerative effects of the whole secretome or isolated extracellular vesicle fraction in vivo. This was followed by bioinformatic analysis of the components of the protein and miRNA content of the secretome and finally compared to a secretome generated from a secondary stem cell source. RESULTS: Here we have demonstrated that the secretome from adipose-derived mesenchymal stem cells shows robust effects on cellular processes that promote tissue regeneration. Furthermore, we show that the whole ADSC secretome is capable of enhancing the rate of skeletal muscle regeneration following acute damage. We assessed the efficacy of the total secretome compared with the extracellular vesicle fraction on a number of assays that inform on tissue regeneration and demonstrate that both fractions affect different aspects of the process in vitro and in vivo. Our in vitro, in vivo, and bioinformatic results show that factors that promote regeneration are distributed both within extracellular vesicles and the soluble fraction of the secretome. CONCLUSIONS: Taken together, our study implies that extracellular vesicles and soluble molecules within ADSC secretome act in a synergistic manner to promote muscle generation.


Asunto(s)
Células Madre Mesenquimatosas/citología , Músculo Esquelético/crecimiento & desarrollo , Proteoma/genética , Regeneración/genética , Animales , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/genética , Vesículas Extracelulares/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/patología , Ratones , MicroARNs/genética , Músculo Esquelético/metabolismo , Proteínas/genética , Solubilidad
20.
Nat Commun ; 10(1): 1685, 2019 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-30976008

RESUMEN

Neonatal sepsis is characterized by hyperinflammation causing enhanced morbidity and mortality compared to adults. This suggests differences in the response towards invading threats. Here we investigate activated cord blood macrophages (CBMΦ) in comparison to adult macrophages (PBMΦ), indicating incomplete interferon gamma (IFN-γ) and interleukin 10 (IL-10)-induced activation of CBMΦ. CBMΦ show reduced expression of phagocytosis receptors and cytokine expression in addition to altered energy metabolism. In particular, IFN-γ as well as IL-10-activated CBMΦ completely fail to increase glycolysis and furthermore show reduced activation of the mTOR pathway, which is important for survival in sepsis. MTOR inhibition by rapamycin equalizes cytokine production in CBMΦ and PBMΦ. Finally, incubation of PBMΦ with cord blood serum or S100A8/A9, which is highly expressed in neonates, suppresses mTOR activation, prevents glycolysis and the expression of an PBMΦ phenotype. Thus, a metabolic alteration is apparent in CBMΦ, which might be dependent on S100A8/A9 expression.


Asunto(s)
Citotoxicidad Inmunológica , Metabolismo Energético/inmunología , Macrófagos/metabolismo , Adulto , Factores de Edad , Calgranulina A/inmunología , Calgranulina A/metabolismo , Calgranulina B/inmunología , Calgranulina B/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Sangre Fetal/citología , Glucólisis/inmunología , Voluntarios Sanos , Humanos , Recién Nacido , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-10/inmunología , Interleucina-10/metabolismo , Macrófagos/inmunología , Cultivo Primario de Células , Sepsis/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA