Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(3): 432-447, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38409259

RESUMEN

Central nervous system (CNS)-resident cells such as microglia, oligodendrocytes and astrocytes are gaining increasing attention in respect to their contribution to CNS pathologies including multiple sclerosis (MS). Several studies have demonstrated the involvement of pro-inflammatory glial subsets in the pathogenesis and propagation of inflammatory events in MS and its animal models. However, it has only recently become clear that the underlying heterogeneity of astrocytes and microglia can not only drive inflammation, but also lead to its resolution through direct and indirect mechanisms. Failure of these tissue-protective mechanisms may potentiate disease and increase the risk of conversion to progressive stages of MS, for which currently available therapies are limited. Using proteomic analyses of cerebrospinal fluid specimens from patients with MS in combination with experimental studies, we here identify Heparin-binding EGF-like growth factor (HB-EGF) as a central mediator of tissue-protective and anti-inflammatory effects important for the recovery from acute inflammatory lesions in CNS autoimmunity. Hypoxic conditions drive the rapid upregulation of HB-EGF by astrocytes during early CNS inflammation, while pro-inflammatory conditions suppress trophic HB-EGF signaling through epigenetic modifications. Finally, we demonstrate both anti-inflammatory and tissue-protective effects of HB-EGF in a broad variety of cell types in vitro and use intranasal administration of HB-EGF in acute and post-acute stages of autoimmune neuroinflammation to attenuate disease in a preclinical mouse model of MS. Altogether, we identify astrocyte-derived HB-EGF and its epigenetic regulation as a modulator of autoimmune CNS inflammation and potential therapeutic target in MS.


Asunto(s)
Astrocitos , Esclerosis Múltiple , Animales , Humanos , Ratones , Antiinflamatorios , Modelos Animales de Enfermedad , Epigénesis Genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Inflamación , Proteómica
2.
N Engl J Med ; 389(9): 820-832, 2023 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-37646679

RESUMEN

BACKGROUND: Sickle cell disease is caused by a defect in the ß-globin subunit of adult hemoglobin. Sickle hemoglobin polymerizes under hypoxic conditions, producing deformed red cells that hemolyze and cause vaso-occlusion that results in progressive organ damage and early death. Elevated fetal hemoglobin levels in red cells protect against complications of sickle cell disease. OTQ923, a clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-edited CD34+ hematopoietic stem- and progenitor-cell (HSPC) product, has a targeted disruption of the HBG1 and HBG2 (γ-globin) gene promoters that increases fetal hemoglobin expression in red-cell progeny. METHODS: We performed a tiling CRISPR-Cas9 screen of the HBG1 and HBG2 promoters by electroporating CD34+ cells obtained from healthy donors with Cas9 complexed with one of 72 guide RNAs, and we assessed the fraction of fetal hemoglobin-immunostaining erythroblasts (F cells) in erythroid-differentiated progeny. The gRNA resulting in the highest level of F cells (gRNA-68) was selected for clinical development. We enrolled participants with severe sickle cell disease in a multicenter, phase 1-2 clinical study to assess the safety and adverse-effect profile of OTQ923. RESULTS: In preclinical experiments, CD34+ HSPCs (obtained from healthy donors and persons with sickle cell disease) edited with CRISPR-Cas9 and gRNA-68 had sustained on-target editing with no off-target mutations and produced high levels of fetal hemoglobin after in vitro differentiation or xenotransplantation into immunodeficient mice. In the study, three participants received autologous OTQ923 after myeloablative conditioning and were followed for 6 to 18 months. At the end of the follow-up period, all the participants had engraftment and stable induction of fetal hemoglobin (fetal hemoglobin as a percentage of total hemoglobin, 19.0 to 26.8%), with fetal hemoglobin broadly distributed in red cells (F cells as a percentage of red cells, 69.7 to 87.8%). Manifestations of sickle cell disease decreased during the follow-up period. CONCLUSIONS: CRISPR-Cas9 disruption of the HBG1 and HBG2 gene promoters was an effective strategy for induction of fetal hemoglobin. Infusion of autologous OTQ923 into three participants with severe sickle cell disease resulted in sustained induction of red-cell fetal hemoglobin and clinical improvement in disease severity. (Funded by Novartis Pharmaceuticals; ClinicalTrials.gov number, NCT04443907.).


Asunto(s)
Anemia de Células Falciformes , Sistemas CRISPR-Cas , Eritrocitos , Hemoglobina Fetal , Trasplante de Células Madre Hematopoyéticas , Animales , Ratones , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Antígenos CD34 , Hemoglobina Fetal/biosíntesis , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Hemoglobina Falciforme , Regiones Promotoras Genéticas
3.
Mol Cell Proteomics ; 23(1): 100683, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37993104

RESUMEN

Dysregulated mRNA splicing is involved in the pathogenesis of many diseases including cancer, neurodegenerative diseases, and muscular dystrophies such as myotonic dystrophy type 1 (DM1). Comprehensive assessment of dysregulated splicing on the transcriptome and proteome level has been methodologically challenging, and thus investigations have often been targeting only few genes. Here, we performed a large-scale coordinated transcriptomic and proteomic analysis to characterize a DM1 mouse model (HSALR) in comparison to wild type. Our integrative proteogenomics approach comprised gene- and splicing-level assessments for mRNAs and proteins. It recapitulated many known instances of aberrant mRNA splicing in DM1 and identified new ones. It enabled the design and targeting of splicing-specific peptides and confirmed the translation of known instances of aberrantly spliced disease-related genes (e.g., Atp2a1, Bin1, Ryr1), complemented by novel findings (Flnc and Ywhae). Comparative analysis of large-scale mRNA and protein expression data showed quantitative agreement of differentially expressed genes and splicing patterns between disease and wild type. We hence propose this work as a suitable blueprint for a robust and scalable integrative proteogenomic strategy geared toward advancing our understanding of splicing-based disorders. With such a strategy, splicing-based biomarker candidates emerge as an attractive and accessible option, as they can be efficiently asserted on the mRNA and protein level in coordinated fashion.


Asunto(s)
Distrofia Miotónica , Proteogenómica , Ratones , Animales , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Distrofia Miotónica/patología , Empalme Alternativo/genética , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Int J Cancer ; 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38938062

RESUMEN

Prognosis of glioblastoma patients is still poor despite multimodal therapy. The highly brain-infiltrating growth in concert with a pronounced therapy resistance particularly of mesenchymal glioblastoma stem-like cells (GSCs) has been proposed to contribute to therapy failure. Recently, we have shown that a mesenchymal-to-proneural mRNA signature of patient derived GSC-enriched (pGSC) cultures associates with in vitro radioresistance and gel invasion. Importantly, this pGSC mRNA signature is prognostic for patients' tumor recurrence pattern and overall survival. Two mesenchymal markers of the mRNA signature encode for IKCa and BKCa Ca2+-activated K+ channels. Therefore, we analyzed here the effect of IKCa- and BKCa-targeting concomitant to (fractionated) irradiation on radioresistance and glioblastoma spreading in pGSC cultures and in pGSC-derived orthotopic xenograft glioma mouse models. To this end, in vitro gel invasion, clonogenic survival, in vitro and in vivo residual DNA double strand breaks (DSBs), tumor growth, and brain invasion were assessed in the dependence on tumor irradiation and K+ channel targeting. As a result, the IKCa- and BKCa-blocker TRAM-34 and paxilline, respectively, increased number of residual DSBs and (numerically) decreased clonogenic survival in some but not in all IKCa- and BKCa-expressing pGSC cultures, respectively. In addition, BKCa- but not IKCa-blockade slowed-down gel invasion in vitro. Moreover, systemic administration of TRAM-34 or paxilline concomitant to fractionated tumor irradiation increased in the xenograft model(s) residual number of DSBs and attenuated glioblastoma brain invasion and (numerically) tumor growth. We conclude, that KCa-blockade concomitant to fractionated radiotherapy might be a promising new strategy in glioblastoma therapy.

5.
Eur J Appl Physiol ; 124(4): 1201-1216, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37966510

RESUMEN

PURPOSE: To investigate the influence of exercise intensity normalisation on intra- and inter-individual acute and adaptive responses to an interval training programme. METHODS: Nineteen cyclists were split in two groups differing (only) in how exercise intensity was normalised: 80% of the maximal work rate achieved in an incremental test (% W ˙ max) vs. maximal sustainable work rate in a self-paced interval training session (% W ˙ max-SP). Testing duplicates were conducted before and after an initial control phase, during the training intervention, and at the end, enabling the estimation of inter-individual variability in adaptive responses devoid of intra-individual variability. RESULTS: Due to premature exhaustion, the median training completion rate was 88.8% for the % W ˙ max group, but 100% for the % W ˙ max-SP the group. Ratings of perceived exertion and heart rates were not sensitive to how intensity was normalised, manifesting similar inter-individual variability, although intra-individual variability was minimised for the % W ˙ max-SP group. Amongst six adaptive response variables, there was evidence of individual response for only maximal oxygen uptake (standard deviation: 0.027 L·min-1·week-1) and self-paced interval training performance (standard deviation: 1.451 W·week-1). However, inter-individual variability magnitudes were similar between groups. Average adaptive responses were also similar between groups across all variables. CONCLUSIONS: To normalise completion rates of interval training, % W ˙ max-SP should be used to prescribe relative intensity. However, the variability in adaptive responses to training may not reflect how exercise intensity is normalised, underlining the complexity of the exercise dose-adaptation relationship. True inter-individual variability in adaptive responses cannot always be identified when intra-individual variability is accounted for.


Asunto(s)
Entrenamiento de Intervalos de Alta Intensidad , Consumo de Oxígeno , Humanos , Consumo de Oxígeno/fisiología , Ejercicio Físico/fisiología , Prueba de Esfuerzo , Frecuencia Cardíaca/fisiología
6.
Cancer ; 129(1): 118-129, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36308296

RESUMEN

BACKGROUND: In the EMPOWER-Lung 1 trial (ClinicalTrials.gov, NCT03088540), cemiplimab conferred longer survival than platinum-doublet chemotherapy for advanced non-small cell lung cancer (NSCLC) with programmed cell death-ligand 1 (PD-L1) ≥50%. Patient-reported outcomes were evaluated among trial participants. METHODS: Adults with NSCLC and Eastern Cooperative Oncology Group performance status 0 to 1 were randomly assigned cemiplimab 350 mg every 3 weeks or platinum-doublet chemotherapy. At baseline and day 1 of each treatment cycle, patients were administered the European Organization for Research and Treatment of Cancer Quality of Life-Core 30 (QLQ-C30) and Lung Cancer Module (QLQ-LC13) questionnaires. Mixed-model repeated measures analysis estimated overall change from baseline for PD-L1 ≥50% and intention-to-treat populations. Kaplan-Meier analysis estimated time to definitive deterioration. RESULTS: In PD-L1 ≥50% patients (cemiplimab, n = 283; chemotherapy, n = 280), baseline QLQ-C30 and QLQ-LC13 scores showed moderate-to-high functioning and low symptom burden. Change from baseline favored cemiplimab on global health status/quality of life (GHS/QOL), functioning, and most symptom scales. Risk of definitive deterioration across functioning scales was reduced versus chemotherapy; hazard ratios were 0.48 (95% CI, 0.32-0.71) to 0.63 (95% CI, 0.41-0.96). Cemiplimab showed lower risk of definitive deterioration for disease-related (dyspnea, cough, pain in chest, pain in other body parts, fatigue) and treatment-related symptoms (peripheral neuropathy, alopecia, nausea/vomiting, appetite loss, constipation, diarrhea) (nominal p < .05). Results were similar in the intention-to-treat population. CONCLUSIONS: Results support cemiplimab for first-line therapy of advanced NSCLC from the patient's perspective. Improved survival is accompanied by improvements versus platinum-doublet chemotherapy in GHS/QOL and functioning and reduction in symptom burden.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Antineoplásicos Inmunológicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Adulto , Humanos , Antígeno B7-H1 , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Pulmón , Neoplasias Pulmonares/tratamiento farmacológico , Dolor/etiología , Medición de Resultados Informados por el Paciente , Platino (Metal)/uso terapéutico , Calidad de Vida , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico
7.
Int J Mol Sci ; 23(17)2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-36077380

RESUMEN

Glioblastoma (GBM) is an obligatory lethal brain tumor with a median survival, even with the best standard of care therapy, of less than 20 months. In light of this fact, the evaluation of new GBM treatment approaches such as oncolytic virotherapy (OVT) is urgently needed. Based on our preliminary preclinical data, the YB-1 dependent oncolytic adenovirus (OAV) XVir-N-31 represents a promising therapeutic agent to treat, in particular, therapy resistant GBM. Preclinical studies have shown that XVir-N-31 prolonged the survival of GBM bearing mice. Now using an immunohumanized mouse model, we examined the immunostimulatory effects of XVir-N-31 in comparison to the wildtype adenovirus (Ad-WT). Additionally, we combined OVT with the inhibition of immune checkpoint proteins by using XVir-N-31 in combination with nivolumab, or by using a derivate of XVir-N-31 that expresses a PD-L1 neutralizing antibody. Although in vitro cell killing was higher for Ad-WT, XVir-N-31 induced a much stronger immunogenic cell death that was further elevated by blocking PD-1 or PD-L1. In vivo, an intratumoral injection of XVir-N-31 increased tumor infiltrating lymphocytes (TILs) and NK cells significantly more than Ad-WT not only in the virus-injected tumors, but also in the untreated tumors growing in the contralateral hemisphere. This suggests that for an effective treatment of GBM, immune activating properties by OAVs seem to be of greater importance than their oncolytic capacity. Furthermore, the addition of immune checkpoint inhibition (ICI) to OVT further induced lymphocyte infiltration. Consequently, a significant reduction in contralateral non-virus-injected tumors was only visible if OVT was combined with ICI. This strongly indicates that for an effective eradication of GBM cells that cannot be directly targeted by an intratumoral OV injection, additional ICI therapy is required.


Asunto(s)
Glioblastoma , Viroterapia Oncolítica , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioblastoma/metabolismo , Ratones , Receptor de Muerte Celular Programada 1
8.
Neuropathol Appl Neurobiol ; 47(6): 768-780, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33780024

RESUMEN

AIMS: In primary central nervous system tumours, epithelial-to-mesenchymal transition (EMT) gene expression is associated with increased malignancy. However, it has also been shown that EMT factors in gliomas are almost exclusively expressed by glioma vessel-associated pericytes (GA-Peris). In this study, we aimed to identify the mechanism of EMT in GA-Peris and its impact on angiogenic processes. METHODS: In glioma patients, vascular density and the expression of the pericytic markers platelet derived growth factor receptor (PDGFR)-ß and smooth muscle actin (αSMA) were examined in relation to the expression of the EMT transcription factor SLUG and were correlated with survival of patients with glioblastoma (GBM). Functional mechanisms of SLUG regulation and the effects on primary human brain vascular pericytes (HBVP) were studied in vitro by measuring proliferation, cell motility and growth characteristics. RESULTS: The number of PDGFR-ß- and αSMA-positive pericytes did not change with increased malignancy nor showed an association with the survival of GBM patients. However, SLUG-expressing pericytes displayed considerable morphological changes in GBM-associated vessels, and TGF-ß induced SLUG upregulation led to enhanced proliferation, motility and altered growth patterns in HBVP. Downregulation of SLUG or addition of a TGF-ß antagonising antibody abolished these effects. CONCLUSIONS: We provide evidence that in GA-Peris, elevated SLUG expression is mediated by TGF-ß, a cytokine secreted by most glioma cells, indicating that the latter actively modulate neovascularisation not only by modulating endothelial cells, but also by influencing pericytes. This process might be responsible for the formation of an unstructured tumour vasculature as well as for the breakdown of the blood-brain barrier in GBM.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Pericitos/efectos de los fármacos , Factores de Transcripción de la Familia Snail/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacocinética , Neoplasias Encefálicas/patología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Pericitos/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
9.
BMC Cancer ; 21(1): 1108, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34654395

RESUMEN

BACKGROUND: Diffuse gliomas are the most common malignant tumors of the central nervous system with poor treatment efficacy. Infiltration of immune cells into tumors during immunosurveillance is observed in multiple tumor entities and often associated with a favorable outcome. The aim of this study was to evaluate the infiltration of immune cells in gliomas and their association with cerebrospinal fluid (CSF) cytokine concentrations. METHODS: We applied immunohistochemistry in tumor tissue sections of 18 high-grade glioma (HGG) patients (4 anaplastic astrocytoma, IDH-wildtype WHO-III; 14 glioblastomas (GBM), IDH-wildtype WHO-IV) in order to assess and quantify leucocytes (CD45) and macrophages (CD68, CD163) within the tumor core, infiltration zone and perivascular spaces. In addition, we quantified the concentrations of 30 cytokines in the same patients' CSF and in 14 non-inflammatory controls. RESULTS: We observed a significantly higher percentage of CD68+ macrophages (21-27%) in all examined tumor areas when compared to CD45+ leucocytes (ca. 3-7%); CD163+ cell infiltration was between 5 and 15%. Compared to the tumor core, significantly more macrophages and leucocytes were detectable within the perivascular area. The brain parenchyma showing a lower tumor cell density seems to be less infiltrated by macrophages. Interleukin (IL)-7 was significantly downregulated in CSF of GBM patients compared to controls. Additionally, CD68+ macrophage infiltrates showed significant correlations with the expression of eotaxin, interferon-γ, IL-1ß, IL-2, IL-10, IL-13, IL-16 and vascular endothelial growth factor. CONCLUSIONS: Our findings suggest that the infiltration of lymphocytes is generally low in HGG, and does not correlate with cytokine concentrations in the CSF. In contrast, macrophage infiltrates in HGG are associated with CSF cytokine changes that possibly shape the tumor microenvironment. Although results point towards an escape from immunosurveillance or even exploitation of immune cells by HGG, further studies are necessary to decipher the exact role of the immune system in these tumors.


Asunto(s)
Astrocitoma/líquido cefalorraquídeo , Neoplasias Encefálicas/líquido cefalorraquídeo , Citocinas/líquido cefalorraquídeo , Glioblastoma/líquido cefalorraquídeo , Leucocitos , Macrófagos , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD , Antígenos de Diferenciación Mielomonocítica , Astrocitoma/patología , Neoplasias Encefálicas/patología , Recuento de Células , Quimiocina CCL11/líquido cefalorraquídeo , Femenino , Glioblastoma/patología , Humanos , Inmunohistoquímica , Interferón gamma/líquido cefalorraquídeo , Interleucinas/líquido cefalorraquídeo , Leucocitos/citología , Linfocitos Infiltrantes de Tumor/citología , Macrófagos/citología , Masculino , Persona de Mediana Edad , Receptores de Superficie Celular , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/líquido cefalorraquídeo
10.
Nat Methods ; 9(11): 1107-12, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23042453

RESUMEN

DNA rearrangements such as sister chromatid exchanges (SCEs) are sensitive indicators of genomic stress and instability, but they are typically masked by single-cell sequencing techniques. We developed Strand-seq to independently sequence parental DNA template strands from single cells, making it possible to map SCEs at orders-of-magnitude greater resolution than was previously possible. On average, murine embryonic stem (mES) cells exhibit eight SCEs, which are detected at a resolution of up to 23 bp. Strikingly, Strand-seq of 62 single mES cells predicts that the mm 9 mouse reference genome assembly contains at least 17 incorrectly oriented segments totaling nearly 1% of the genome. These misoriented contigs and fragments have persisted through several iterations of the mouse reference genome and have been difficult to detect using conventional sequencing techniques. The ability to map SCE events at high resolution and fine-tune reference genomes by Strand-seq dramatically expands the scope of single-cell sequencing.


Asunto(s)
Análisis de Secuencia de ADN/métodos , Intercambio de Cromátides Hermanas , Moldes Genéticos , Animales , Células Cultivadas , Genómica , Ratones
11.
J Neurosci ; 33(48): 18836-48, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24285890

RESUMEN

Axonal injury generates growth inert retraction bulbs with dynamic cytoskeletal properties that are severely compromised. Conversion of "frozen" retraction bulbs into actively progressing growth cones is a major aim in axon regeneration. Here we report that murine serum response factor (SRF), a gene regulator linked to the actin cytoskeleton, modulates growth cone actin dynamics during axon regeneration. In regeneration-competent facial motoneurons, Srf deletion inhibited axonal regeneration. In wild-type mice after nerve injury, SRF translocated from the nucleus to the cytoplasm, suggesting a cytoplasmic SRF function in axonal regeneration. Indeed, adenoviral overexpression of cytoplasmic SRF (SRF-ΔNLS-GFP) stimulated axonal sprouting and facial nerve regeneration in vivo. In primary central and peripheral neurons, SRF-ΔNLS-GFP stimulated neurite outgrowth, branch formation, and growth cone morphology. Furthermore, we uncovered a link between SRF and the actin-severing factor cofilin during axonal regeneration in vivo. Facial nerve axotomy increased the total cofilin abundance and also nuclear localization of phosphorylated cofilin in a subpopulation of lesioned motoneurons. This cytoplasmic-to-nucleus translocation of P-cofilin upon axotomy was reduced in motoneurons expressing SRF-ΔNLS-GFP. Finally, we demonstrate that cytoplasmic SRF and cofilin formed a reciprocal regulatory unit. Overexpression of cytoplasmic SRF reduced cofilin phosphorylation and vice versa: overexpression of cofilin inhibited SRF phosphorylation. Therefore, a regulatory loop consisting of SRF and cofilin might take part in reactivating actin dynamics in growth-inert retraction bulbs and facilitating axon regeneration.


Asunto(s)
Factores Despolimerizantes de la Actina/fisiología , Axones/efectos de los fármacos , Citoplasma/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Factor de Respuesta Sérica/farmacología , Actinas/metabolismo , Animales , Axotomía , Citoplasma/efectos de los fármacos , Nervio Facial/fisiología , Femenino , Proteínas Fluorescentes Verdes , Masculino , Ratones , Nervios Periféricos/citología , Nervios Periféricos/efectos de los fármacos , Fosforilación , Reacción en Cadena de la Polimerasa , Fracciones Subcelulares/metabolismo
12.
Free Neuropathol ; 52024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38455669

RESUMEN

Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. GBM displays excessive and unfunctional vascularization which may, among others, be a reason for its devastating prognosis. Pericytes have been identified as the major component of the irregular vessel structure in GBM. In vitro data suggest an epithelial-to-mesenchymal transition (EMT)-like activation of glioma-associated pericytes, stimulated by GBM-secreted TGF-ß, to be involved in the formation of a chaotic and dysfunctional tumor vasculature. This study investigated whether TGF-ß impacts the function of vessel associated mural cells (VAMCs) in vivo via the induction of the EMT transcription factor SLUG and whether this is associated with the development of GBM-associated vascular abnormalities. Upon preventing the TGF-ß-/SLUG-mediated EMT induction in VAMCs, the number of PDGFRß and αSMA positive cells was significantly reduced, regardless of whether TGF-ß secretion by GBM cells was blocked or whether SLUG was specifically knocked out in VAMCs. The reduced amount of PDGFRß+ or αSMA+ cells observed under those conditions correlated with a lower vessel density and fewer vascular abnormalities. Our data provide evidence that the SLUG-mediated modulation of VAMC activity is induced by GBM-secreted TGF-߬ and that activated VAMCs are key contributors in neo-angiogenic processes. We suggest that a pathologically altered activation of GA-Peris in the tumor microenvironment is responsible for the unstructured tumor vasculature. There is emerging evidence that vessel normalization alleviates tumor hypoxia, reduces tumor-associated edema and improves drug delivery. Therefore, avoiding the generation of an unstructured and non-functional tumor vasculature during tumor recurrence might be a promising treatment approach for GBM and identifies pericytes as a potential novel therapeutic target.

13.
Infect Dis Ther ; 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961047

RESUMEN

INTRODUCTION: This study aimed to assess the effects of a monoclonal antibody (mAb) combination on symptoms, daily function, and overall health-related quality of life. METHODS: We analyzed patient-reported outcomes data from symptomatic outpatients in a phase 1/2/3 trial. Patients with confirmed SARS-CoV-2 infection and ≥ 1 risk factor for severe COVID-19 received mAb treatment (casirivimab plus imdevimab 1200 mg) or placebo. Prespecified exploratory assessments included time to sustained symptoms resolution, usual health, and return to usual activities (assessed daily for 29 days). The trial was conducted from September 2020 to February 2021, prior to widespread COVID-19 vaccination programs and Omicron-lineage variants against which casirivimab + imdevimab is not active. RESULTS: In this analysis 736 outpatients received mAb and 1341 received placebo. Median time to sustained symptoms resolution was consistently shorter with mAb versus placebo (≥ 2 consecutive days: 14 vs 17 days, [nominal p = 0.0017]; ≥ 3 consecutive days: 17 vs 21 days, [nominal p = 0.0046]). Median time to sustained return to usual health and usual activities were both consistently shorter with mAb versus placebo (≥ 2 consecutive days: 12 vs 15 days [nominal p = 0.0001] and 9 vs 11 days [nominal p = 0.0001], respectively; ≥ 3 consecutive days: 14 vs 18 days [nominal p = 0.0003] and 10 vs 13 days [nominal p = 0.0041], respectively). CONCLUSIONS: mAb treatment against susceptible SARS-CoV-2 strains improved how patients feel and function, as evidenced by shortened time to sustained symptoms resolution and return to usual health and activities. Future studies are warranted to assess the patient experience with next generation mAbs. CLINICALTRIALS: GOV: Registration number, NCT04425629; Submission date June 11, 2020.

14.
Mol Oncol ; 18(3): 528-546, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38115217

RESUMEN

Neural stem cells (NSCs) are considered to be valuable candidates for delivering a variety of anti-cancer agents, including oncolytic viruses, to brain tumors. However, owing to the previously reported tumorigenic potential of NSC cell lines after intranasal administration (INA), here we identified the human hepatic stellate cell line LX-2 as a cell type capable of longer resistance to replication of oncolytic adenoviruses (OAVs) as a therapeutic cargo, and that is non-tumorigenic after INA. Our data show that LX-2 cells can longer withstand the OAV XVir-N-31 replication and oncolysis than NSCs. By selecting the highly migratory cell population out of LX-2, an offspring cell line with a higher and more stable capability to migrate was generated. Additionally, as a safety backup, we applied genomic herpes simplex virus thymidine kinase (HSV-TK) integration into LX-2, leading to high vulnerability to ganciclovir (GCV). Histopathological analyses confirmed the absence of neoplasia in the respiratory tracts and brains of immuno-compromised mice 3 months after INA of LX-2 cells. Our data suggest that LX-2 is a novel, robust, and safe cell line for delivering anti-cancer and other therapeutic agents to the brain.


Asunto(s)
Antivirales , Terapia Genética , Ratones , Humanos , Animales , Administración Intranasal , Línea Celular , Sistema Nervioso Central/metabolismo , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Timidina Quinasa/uso terapéutico
15.
Nat Struct Mol Biol ; 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38347147

RESUMEN

ARID1B is a SWI/SNF subunit frequently mutated in human Coffin-Siris syndrome (CSS) and it is necessary for proliferation of ARID1A mutant cancers. While most CSS ARID1B aberrations introduce frameshifts or stop codons, the functional consequence of missense mutations found in ARID1B is unclear. We here perform saturated mutagenesis screens on ARID1B and demonstrate that protein destabilization is the main mechanism associated with pathogenic missense mutations in patients with Coffin-Siris Syndrome.

16.
Nat Cancer ; 5(3): 481-499, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38233483

RESUMEN

Activating mutations in GNAQ/GNA11 occur in over 90% of uveal melanomas (UMs), the most lethal melanoma subtype; however, targeting these oncogenes has proven challenging and inhibiting their downstream effectors show limited clinical efficacy. Here, we performed genome-scale CRISPR screens along with computational analyses of cancer dependency and gene expression datasets to identify the inositol-metabolizing phosphatase INPP5A as a selective dependency in GNAQ/11-mutant UM cells in vitro and in vivo. Mutant cells intrinsically produce high levels of the second messenger inositol 1,4,5 trisphosphate (IP3) that accumulate upon suppression of INPP5A, resulting in hyperactivation of IP3-receptor signaling, increased cytosolic calcium and p53-dependent apoptosis. Finally, we show that GNAQ/11-mutant UM cells and patients' tumors exhibit elevated levels of IP4, a biomarker of enhanced IP3 production; these high levels are abolished by GNAQ/11 inhibition and correlate with sensitivity to INPP5A depletion. Our findings uncover INPP5A as a synthetic lethal vulnerability and a potential therapeutic target for GNAQ/11-mutant-driven cancers.


Asunto(s)
Melanoma , Humanos , Melanoma/tratamiento farmacológico , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/uso terapéutico , Mutación , Transducción de Señal , Inositol Polifosfato 5-Fosfatasas/genética
17.
J Transl Med ; 11: 216, 2013 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-24044901

RESUMEN

BACKGROUND: The brain cancer stem cell (CSC) model describes a small subset of glioma cells as being responsible for tumor initiation, conferring therapy resistance and tumor recurrence. In brain CSC, the PI3-K/AKT and the RAS/mitogen activated protein kinase (MAPK) pathways are found to be activated. In consequence, the human transcription factor YB-1, knowing to be responsible for the emergence of drug resistance and driving adenoviral replication, is phosphorylated and activated. With this knowledge, YB-1 was established in the past as a biomarker for disease progression and prognosis. This study determines the expression of YB-1 in glioblastoma (GBM) specimen in vivo and in brain CSC lines. In addition, the capacity of Ad-Delo3-RGD, an YB-1 dependent oncolytic adenovirus, to eradicate CSC was evaluated both in vitro and in vivo. METHODS: YB-1 expression was investigated by immunoblot and immuno-histochemistry. In vitro, viral replication as well as the capacity of Ad-Delo3-RGD to replicate in and, in consequence, to kill CSC was determined by real-time PCR and clonogenic dilution assays. In vivo, Ad-Delo3-RGD-mediated tumor growth inhibition was evaluated in an orthotopic mouse GBM model. Safety and specificity of Ad-Delo3-RGD were investigated in immortalized human astrocytes and by siRNA-mediated downregulation of YB-1. RESULTS: YB-1 is highly expressed in brain CSC lines and in GBM specimen. Efficient viral replication in and virus-mediated lysis of CSC was observed in vitro. Experiments addressing safety aspects of Ad-Delo3-RGD showed that (i) virus production in human astrocytes was significantly reduced compared to wild type adenovirus (Ad-WT) and (ii) knockdown of YB-1 significantly reduced virus replication. Mice harboring othotopic GBM developed from a temozolomide (TMZ)-resistant GBM derived CSC line which was intratumorally injected with Ad-Delo3-RGD survived significantly longer than mice receiving PBS-injections or TMZ treatment. CONCLUSION: The results of this study supported YB-1 based virotherapy as an attractive therapeutic strategy for GBM treatment which will be exploited further in multimodal treatment concepts.


Asunto(s)
Adenoviridae/metabolismo , Neoplasias Encefálicas/patología , Glioma/patología , Células Madre Neoplásicas/patología , Virus Oncolíticos/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo , Adenoviridae/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glioma/enzimología , Humanos , Ratones , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/metabolismo , Virus Oncolíticos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Temozolomida , Proteínas Supresoras de Tumor/metabolismo , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Pediatr ; 163(1): 173-8.e1, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23333130

RESUMEN

OBJECTIVE: To investigate dietary patterns and nutritional intake in children of mothers with eating disorders. STUDY DESIGN: Mothers (N = 9423) from a longitudinal general population birth cohort study, the Avon Longitudinal Study of Parents and Children, completed Food Frequency Questionnaires on their children at 3, 4, 7, and 9 years of age. Macronutrient intake was estimated, and dietary patterns were obtained using principal components analysis. Linear regression and mixed-effects models were used to assess dietary patterns and nutritional intake among children of women with lifetime anorexia nervosa (AN, n = 140), bulimia nervosa (BN, n = 170), or AN+BN (n = 71), compared with children of women without eating disorders (unexposed women, n = 9037). RESULTS: Children in the maternal AN and BN groups had higher scores on the "health conscious/vegetarian" dietary pattern compared with unexposed children. Less adherence to the "traditional" dietary pattern was observed in children of exposed mothers, with more pronounced differences in early childhood. Children of women with AN and BN had higher intake of energy and children of women with BN had higher intake of carbohydrates and starch and lower intake of fat, compared with children in the unexposed group. CONCLUSIONS: Maternal eating disorders are associated with altered offspring dietary patterns and macronutrient intake. Longitudinal changes in patterns of diet in children of women with eating disorders may increase the risk of weight gain or disordered eating later in life.


Asunto(s)
Anorexia Nerviosa , Bulimia Nerviosa , Fenómenos Fisiológicos Nutricionales Infantiles , Dieta , Ingestión de Alimentos , Niño , Preescolar , Femenino , Humanos , Estudios Longitudinales , Masculino , Madres
19.
Int J Eat Disord ; 46(7): 643-52, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23818118

RESUMEN

OBJECTIVE: Fat talking has been assumed to be a causal risk factor for body dissatisfaction in a number of prevention programs and body confidence campaigns. The aim of this paper was to assess whether fat talking meets three criteria necessary for causal risk factors, namely whether fat talking is: (a) cross-sectionally associated with body dissatisfaction; (b) prospectively associated with changes in body dissatisfaction; and (c) associated with changes in body dissatisfaction in experimental studies. METHOD: A systematic literature review was conducted using electronic databases and hand searching of relevant journals. Meta-analyses provided pooled effect size estimates, and meta-regressions were used to determine whether age, gender or risk of bias were effect modifiers of the relationship. RESULTS: Searches revealed 24 studies. There was a significant cross-sectional association (r = 0.297, 95% CI = 0.225-0.349), which differed in strength between age groups and genders. There was a prospective association between fat talking and changes in body dissatisfaction in long term (r = 0.144, 95% CI = 0.050-0.234), but not in short-term studies (r = 0.022, 95% CI = -0.131-0.174). One study showed that experimental exposure to fat talking was associated with increases in body dissatisfaction (d = 0.124). DISCUSSION: As such, there is good evidence that fat talking is a correlate of body dissatisfaction. The few prospective and experimental studies give an initial indication that fat talking is a causal risk factor for body dissatisfaction. Further work is needed to support this position.


Asunto(s)
Imagen Corporal/psicología , Autoevaluación (Psicología) , Adulto , Factores de Edad , Sesgo , Niño , Comunicación , Femenino , Humanos , Masculino , Grupo Paritario , Factores de Riesgo , Factores Sexuales
20.
Int J Eat Disord ; 46(4): 346-54, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23108538

RESUMEN

OBJECTIVE: The aim of the study was to examine how carers cope practically and emotionally with caring for individuals with anorexia nervosa who require intensive hospital care. METHOD: This study explores objective burden (time spent with caregiving and number of tasks), subjective burden (psychological distress), and social support in a sample of parents (n = 224) and partners (n = 28) from a consecutive series of patients (n = 178) admitted to inpatient units within the United Kingdom. RESULTS: Most time was spent providing emotional support and less with practical tasks. Time spent with caregiving was associated with carer distress and was fully mediated by carer burden. This was ameliorated by social support. Partners received minimal support from others, and we found similar levels of burden and distress for mothers and partners. DISCUSSION: The data indicate that professional and social support alleviates carer distress and may be of particular value for partners who are more isolated than parents. The data also suggest that time spent with practical support may be of more value than emotional support.


Asunto(s)
Adaptación Psicológica , Anorexia Nerviosa/terapia , Cuidadores/psicología , Costo de Enfermedad , Estrés Psicológico/psicología , Adulto , Estudios Transversales , Emociones , Femenino , Humanos , Masculino , Persona de Mediana Edad , Apoyo Social , Encuestas y Cuestionarios
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA