Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 125(2): 249-60, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25406351

RESUMEN

In the last decade there has been a rapid expansion in clinical trials using mesenchymal stromal cells (MSCs) from a variety of tissues. However, despite similarities in morphology, immunophenotype, and differentiation behavior in vitro, MSCs sourced from distinct tissues do not necessarily have equivalent biological properties. We performed a genome-wide methylation, transcription, and in vivo evaluation of MSCs from human bone marrow (BM), white adipose tissue, umbilical cord, and skin cultured in humanized media. Surprisingly, only BM-derived MSCs spontaneously formed a BM cavity through a vascularized cartilage intermediate in vivo that was progressively replaced by hematopoietic tissue and bone. Only BM-derived MSCs exhibited a chondrogenic transcriptional program with hypomethylation and increased expression of RUNX3, RUNX2, BGLAP, MMP13, and ITGA10 consistent with a latent and primed skeletal developmental potential. The humanized MSC-derived microenvironment permitted homing and maintenance of long-term murine SLAM(+) hematopoietic stem cells (HSCs), as well as human CD34(+)/CD38(-)/CD90(+)/CD45RA(+) HSCs after cord blood transplantation. These studies underscore the profound differences in developmental potential between MSC sources independent of donor age, with implications for their clinical use. We also demonstrate a tractable human niche model for studying homing and engraftment of human hematopoietic cells in normal and neoplastic states.


Asunto(s)
Linaje de la Célula , Epigénesis Genética , Células Madre Hematopoyéticas/citología , Células Madre Mesenquimatosas/citología , Nicho de Células Madre , Western Blotting , Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Condrogénesis/fisiología , Citometría de Flujo , Humanos , Osteogénesis/fisiología
2.
Biochem Biophys Res Commun ; 464(1): 126-32, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26086095

RESUMEN

Class 3 semaphorins were discovered as a family of axon guidance molecules, but are now known to be involved in diverse biologic processes. In this study, we investigated the anti-angiogenic potential of SEMA3E and SEMA3F (SEMA3E&F) in infantile hemangioma (IH). IH is a common vascular tumor that involves both vasculogenesis and angiogenesis. Our lab has identified and isolated hemangioma stem cells (HemSC), glucose transporter 1 positive (GLUT1(+)) endothelial cells (designated as GLUT1(sel) cells) based on anti-GLUT1 magnetic beads selection and GLUT1-negative endothelial cells (named HemEC). We have shown that these types of cells play important roles in hemangiogenesis. We report here that SEMA3E inhibited HemEC migration and proliferation while SEMA3F was able to suppress the migration and proliferation in all three types of cells. Confocal microscopy showed that stress fibers in HemEC were reduced by SEMA3E&F and that stress fibers in HemSC were decreased by SEMA3F, which led to cytoskeletal collapse and loss of cell motility in both cell types. Additionally, SEMA3E&F were able to inhibit vascular endothelial growth factor (VEGF)-induced sprouts in all three types of cells. Further, SEMA3E&F reduced the level of p-VEGFR2 and its downstream p-ERK in HemEC. These results demonstrate that SEMA3E&F inhibit IH cell proliferation and suppress the angiogenic activities of migration and sprout formation. SEMA3E&F may have therapeutic potential to treat or prevent growth of highly proliferative IH.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Neovascularización Patológica/prevención & control , Proteínas del Tejido Nervioso/farmacología , Semaforinas/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hemangioma/irrigación sanguínea , Hemangioma/genética , Hemangioma/patología , Humanos , Lactante , Neoplasias de Tejido Vascular/irrigación sanguínea , Neoplasias de Tejido Vascular/genética , Neoplasias de Tejido Vascular/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Recombinantes/farmacología , Transducción de Señal , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Sensors (Basel) ; 15(6): 13052-68, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26053751

RESUMEN

Cameleons are sophisticated genetically encoded fluorescent probes that allow quantifying cellular Ca2+ signals. The probes are based on Förster resonance energy transfer (FRET) between terminally located fluorescent proteins (FPs), which move together upon binding of Ca2+ to the central calmodulin myosin light chain kinase M13 domain. Most of the available cameleons consist of cyan and yellow FPs (CFP and YFP) as the FRET pair. However, red-shifted versions with green and orange or red FPs (GFP, OFP, RFP) have some advantages such as less phototoxicity and minimal spectral overlay with autofluorescence of cells and fura-2, a prominent chemical Ca2+ indicator. While GFP/OFP- or GFP/RFP-based cameleons have been successfully used to study cytosolic and mitochondrial Ca2+ signals, red-shifted cameleons to visualize Ca2+ dynamics of the endoplasmic reticulum (ER) have not been developed so far. In this study, we generated and tested several ER targeted red-shifted cameleons. Our results show that GFP/OFP-based cameleons due to miss-targeting and their high Ca2+ binding affinity are inappropriate to record ER Ca2+ signals. However, ER targeted GFP/RFP-based probes were suitable to sense ER Ca2+ in a reliable manner. With this study we increased the palette of cameleons for visualizing Ca2+ dynamics within the main intracellular Ca2+ store.


Asunto(s)
Calcio/análisis , Calcio/química , Retículo Endoplásmico/química , Colorantes Fluorescentes/química , Proteínas Luminiscentes/química , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas Luminiscentes/metabolismo , Microscopía Confocal
4.
Blood ; 119(21): 4971-80, 2012 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-22490334

RESUMEN

The interactions between hematopoietic cells and the bone marrow (BM) microenvironment play a critical role in normal and malignant hematopoiesis and drug resistance. These interactions within the BM niche are unique and could be important for developing new therapies. Here, we describe the development of extramedullary bone and bone marrow using human mesenchymal stromal cells and endothelial colony-forming cells implanted subcutaneously into immunodeficient mice. We demonstrate the engraftment of human normal and leukemic cells engraft into the human extramedullary bone marrow. When normal hematopoietic cells are engrafted into the model, only discrete areas of the BM are hypoxic, whereas leukemia engraftment results in widespread severe hypoxia, just as recently reported by us in human leukemias. Importantly, the hematopoietic cell engraftment could be altered by genetical manipulation of the bone marrow microenvironment: Extramedullary bone marrow in which hypoxia-inducible factor 1α was knocked down in mesenchymal stromal cells by lentiviral transfer of short hairpin RNA showed significant reduction (50% ± 6%; P = .0006) in human leukemic cell engraftment. These results highlight the potential of a novel in vivo model of human BM microenvironment that can be genetically modified. The model could be useful for the study of leukemia biology and for the development of novel therapeutic modalities aimed at modifying the hematopoietic microenvironment.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea/métodos , Microambiente Celular/fisiología , Hematopoyesis Extramedular/fisiología , Trasplante Heterotópico , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea/fisiología , Células Cultivadas , Microambiente Celular/genética , Hematopoyesis Extramedular/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Modelos Animales , Osteogénesis/genética , Osteogénesis/fisiología , Especificidad de la Especie
5.
BMC Cancer ; 14: 40, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-24460801

RESUMEN

BACKGROUND: Hypoxia-induced genes are potential targets in cancer therapy. Responses to hypoxia have been extensively studied in vitro, however, they may differ in vivo due to the specific tumor microenvironment. In this study gene expression profiles were obtained from fresh human lung cancer tissue fragments cultured ex vivo under different oxygen concentrations in order to study responses to hypoxia in a model that mimics human lung cancer in vivo. METHODS: Non-small cell lung cancer (NSCLC) fragments from altogether 70 patients were maintained ex vivo in normoxia or hypoxia in short-term culture. Viability, apoptosis rates and tissue hypoxia were assessed. Gene expression profiles were studied using Affymetrix GeneChip 1.0 ST microarrays. RESULTS: Apoptosis rates were comparable in normoxia and hypoxia despite different oxygenation levels, suggesting adaptation of tumor cells to hypoxia. Gene expression profiles in hypoxic compared to normoxic fragments largely overlapped with published hypoxia-signatures. While most of these genes were up-regulated by hypoxia also in NSCLC cell lines, membrane metallo-endopeptidase (MME, neprilysin, CD10) expression was not increased in hypoxia in NSCLC cell lines, but in carcinoma-associated fibroblasts isolated from non-small cell lung cancers. High MME expression was significantly associated with poor overall survival in 342 NSCLC patients in a meta-analysis of published microarray datasets. CONCLUSIONS: The novel ex vivo model allowed for the first time to analyze hypoxia-regulated gene expression in preserved human lung cancer tissue. Gene expression profiles in human hypoxic lung cancer tissue overlapped with hypoxia-signatures from cancer cell lines, however, the elastase MME was identified as a novel hypoxia-induced gene in lung cancer. Due to the lack of hypoxia effects on MME expression in NSCLC cell lines in contrast to carcinoma-associated fibroblasts, a direct up-regulation of stroma fibroblast MME expression under hypoxia might contribute to enhanced aggressiveness of hypoxic cancers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Fibroblastos/enzimología , Neoplasias Pulmonares/enzimología , Neprilisina/metabolismo , Células del Estroma/enzimología , Apoptosis , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular , Fibroblastos/patología , Perfilación de la Expresión Génica/métodos , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Neprilisina/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Células del Estroma/patología , Técnicas de Cultivo de Tejidos , Regulación hacia Arriba
6.
Int J Pharm ; 650: 123725, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38113976

RESUMEN

Proteolysis targeting chimeras (PROTACs) are a promising class of pharmaceutical agents with a unique mode of action. PROTACs enable the targeting of a broad variety of structures including transcription factors and other "undruggable" targets. The poor solubility and slow dissolution of PROTACs currently limit the extensive use of their potential. Up to date, only very limited drug delivery options have been examined to address this challenge. Therefore, we explored the potential of amorphous solid dispersions (ASDs) by spray drying a model PROTAC with different polymers. The resulting formulations were assessed in terms of purity, solid state, dissolution performance, and stability. A strong increase in supersaturation compared to the physical mixture was provided, although in both systems the PROTAC molecule itself was already in the amorphous state. Evaluation of the reasons for the superiority of the ASD formulations revealed that the major factor was the homogeneous, molecular distribution of the active pharmaceutical ingredient (API) in the polymer matrix, as well as improved wettability of the formulation containing Soluplus compared to the physical mixture. The manufactured formulations were stable over a minimum of 8 weeks when protected from light and humidity.


Asunto(s)
Química Farmacéutica , Quimera Dirigida a la Proteólisis , Composición de Medicamentos/métodos , Química Farmacéutica/métodos , Polímeros/química , Solubilidad
7.
Mod Pathol ; 26(2): 182-94, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22936065

RESUMEN

Chemokine receptors have a crucial role in the development and progression of lymphoid neoplasms. To determine the chemokine receptor expression profile in gastric mucosa-associated lymphoid tissue (MALT) lymphoma, we performed an expression analysis of 19 chemokine receptors at mRNA levels by using real-time RT-PCR, as well as of five chemokine receptors--CCR8, CCR9, CXCR4, CXCR6 and CXCR7--by immunohistochemistry on human tissue samples of Helicobacter pylori-associated gastritis, gastric MALT lymphoma and gastric extranodal diffuse large B-cell lymphoma originating from MALT lymphoma (transformed MALT lymphoma). Following malignant transformation from H. pylori-associated gastritis to MALT lymphoma, an upregulation of CCR7, CXCR3 and CXCR7, and a loss of CXCR4 were detected. The transformation of gastric MALT lymphomas to gastric extranodal diffuse large B-cell lymphoma was accompanied by upregulation of CCR1, CCR5, CCR7, CCR8, CCR9, CXCR3, CXCR6, CXCR7 and XCR1. Remarkably, CXCR4 expression was exclusively found in nodal marginal B-cell lymphomas and nodal diffuse large B-cell lymphomas but not at extranodal manifestation sites, ie, in gastric MALT lymphomas or gastric extranodal diffuse large B-cell lymphomas. Furthermore, the incidence of bone marrow infiltration (16/51 with bone marrow involvement vs 35/51 with bone marrow involvement; Spearman ρ=0467 P<0.001) positively correlated with CXCR4 expression. CXCL12, the ligand of CXCR4 and CXCR7, was expressed by epithelial, endothelial and inflammatory cells, MALT lymphoma cells and was most strongly expressed by extranodal diffuse large B-cell lymphoma cells, suggesting at least in part an autocrine signaling pathway. Our data indicate that CXCR4 expression is associated with nodal manifestation and a more advanced stage of lymphomas and hence, might serve as useful clinical prognostic marker.


Asunto(s)
Linfoma de Células B de la Zona Marginal/genética , Linfoma de Células B Grandes Difuso/genética , Receptores CXCR4/genética , Receptores CXCR/genética , Neoplasias Gástricas/genética , Regulación hacia Arriba , Progresión de la Enfermedad , Gastritis/genética , Gastritis/metabolismo , Gastritis/patología , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/patología , Helicobacter pylori , Humanos , Linfoma de Células B de la Zona Marginal/metabolismo , Linfoma de Células B de la Zona Marginal/patología , Linfoma de Células B Grandes Difuso/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
8.
Blood ; 113(26): 6716-25, 2009 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-19321860

RESUMEN

Endothelial progenitor cells are critically involved in essential biologic processes, such as vascular homeostasis, regeneration, and tumor angiogenesis. Endothelial colony-forming cells (ECFCs) are endothelial progenitor cells with robust proliferative potential. Their profound vessel-forming capacity makes them a promising tool for innovative experimental, diagnostic, and therapeutic strategies. Efficient and safe methods for their isolation and expansion are presently lacking. Based on the previously established efficacy of animal serum-free large-scale clinical-grade propagation of mesenchymal stromal cells, we hypothesized that endothelial lineage cells may also be propagated efficiently following a comparable strategy. Here we demonstrate that human ECFCs can be recovered directly from unmanipulated whole blood. A novel large-scale animal protein-free humanized expansion strategy preserves the progenitor hierarchy with sustained proliferation potential of more than 30 population doublings. By applying large-scale propagated ECFCs in various test systems, we observed vascular networks in vitro and perfused vessels in vivo. After large-scale expansion and cryopreservation phenotype, function, proliferation, and genomic stability were maintained. For the first time, proliferative, functional, and storable ECFCs propagated under humanized conditions can be explored in terms of their therapeutic applicability and risk profile.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Células 3T3/enzimología , Adulto , Animales , División Celular , Separación Celular/métodos , Células Cultivadas/citología , Células Cultivadas/enzimología , Células Cultivadas/trasplante , Células Clonales/citología , Células Clonales/enzimología , Ensayo de Unidades Formadoras de Colonias , Criopreservación , Medios de Cultivo , Células Endoteliales/enzimología , Sangre Fetal/citología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/enzimología , Humanos , Inmunofenotipificación , Recién Nacido , Ratones , Ratones Desnudos , Neovascularización Patológica/etiología , Neovascularización Patológica/patología , Neovascularización Fisiológica , Telómero/metabolismo , Telómero/ultraestructura , Trasplante Heterólogo
9.
Cytotherapy ; 13(4): 503-12, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21128706

RESUMEN

BACKGROUND AIMS: Clinical trials for therapeutic angiogenesis use blood- or bone marrow-derived hematopoietic cells, endothelial progenitor cells (EPC) and mesenchymal stromal cells (MSC) for vascular regeneration. Recently concerns have emerged that all three cell types could also contribute to atherosclerosis by foam cell formation. Therefore, we asked whether human myelomonocytic cells, EPC or MSC can accumulate lipid droplets (LD) and develop into foam cells. METHODS: LD accumulation was quantified by flow cytometry, confocal microscopy and cholesterol measurement in each of the cell types. The impact of an initial pro-angiogenic induction on subsequent foam cell formation was studied to mimic relevant settings already used in clinical trials. The phosphorylation state of intracellular signaling molecules in response to the pro-angiogenic stimulation was determined to delineate the operative mechanisms and establish a basis for interventional strategies. RESULTS: Foam cells were formed by monocytes but not by EPC or MSC after pro-angiogenic induction. Mitogen-activated protein kinase (MAPK) p38 phosphorylation was enhanced and kinase inhibition almost abrogated intracellular LD accumulation in monocytes. CONCLUSIONS: These data suggest that hematopoietic cell preparations containing monocytes bear the risk of foam cell formation after pro-angiogenic induction. Instead, EPC and MSC may drive vascular regeneration without atherogenesis aggravation. A thorough understanding of cell biology is necessary to develop new strategies combining pro-angiogenic and anti-atherogenic effects during cell therapy.


Asunto(s)
Células Espumosas/citología , Células Mieloides/citología , Neovascularización Fisiológica/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Cultivadas , Citometría de Flujo , Células Espumosas/metabolismo , Humanos , Microscopía Confocal , Células Mieloides/metabolismo , Células Madre/citología , Células Madre/metabolismo
10.
World Neurosurg ; 109: e24-e32, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28951183

RESUMEN

BACKGROUND: Recent studies have shown higher accuracy rates of image-guided pedicle screw placement compared to freehand (FH) placement. However, data focusing on the impact of spinal navigation on the rate of revision surgeries caused by misplaced pedicle screws (PS) are scarce. OBJECTIVE: This study is aimed at identifying the rate of revision surgeries for misplaced PS comparing three-dimensional (3D) fluoroscopy navigation (3DFL) with FH PS placement. METHODS: A retrospective analysis was conducted of 2232 patients (mean age, 65.3 ± 13.5 years) with 13,703 implanted PS who underwent instrumentation of the thoracolumbar spine between 2007 and 2015. Group 1 received surgery with use of 3DFL (January 2011 to December 2015), group 2 received surgery in the FH technique (April 2007 to December 2015). Because the use of 3DFL was initiated in January 2011, the examined period for 3DFL-navigated surgeries is shorter. Patients routinely received postoperative computed tomography scans and/or intraoperative control 3D scans. RESULTS: There was an overall rate of revision surgeries for malpositioned PS of 2.9%. In the 3DFL group, the rate of secondary revision surgeries was significantly lower with 1.35% (15/1112 patients) compared to 4.38% (49/1120 patients) in the FH group, respectively (odds ratio, 3.35; P < 0.01). Of all PS in the 3DFL group (30/7548 PS), 0.40% needed revision surgery (P < 0.01) compared to 1.14% in the FH group (70/6155 PS). CONCLUSIONS: We were able to show that the use of 3DFL-navigated PS placement significantly reduces the rate of revision surgeries after posterior spinal instrumentation compared to freehand PS placement.


Asunto(s)
Monitorización Neurofisiológica Intraoperatoria/tendencias , Vértebras Lumbares/cirugía , Neuronavegación/tendencias , Tornillos Pediculares , Reoperación/tendencias , Vértebras Torácicas/cirugía , Anciano , Femenino , Fluoroscopía/normas , Fluoroscopía/tendencias , Humanos , Imagenología Tridimensional/normas , Imagenología Tridimensional/tendencias , Monitorización Neurofisiológica Intraoperatoria/normas , Vértebras Lumbares/diagnóstico por imagen , Masculino , Persona de Mediana Edad , Neuronavegación/normas , Tornillos Pediculares/efectos adversos , Estudios Retrospectivos , Cirugía Asistida por Computador/normas , Cirugía Asistida por Computador/tendencias , Vértebras Torácicas/diagnóstico por imagen
11.
PLoS One ; 11(3): e0152594, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27031696

RESUMEN

RATIONALE: ABCA3 is a lipid transporter in the limiting membrane of lamellar bodies in alveolar type II cells. Mutations in the ABCA3 gene cause respiratory distress syndrome in new-borns and childhood interstitial lung disease. ABCA3 is N-terminally cleaved by an as yet unknown protease, a process believed to regulate ABCA3 activity. METHODS: The exact site where ABCA3 is cleaved was localized using mass spectrometry (MS). Proteases involved in ABCA3 processing were identified using small molecule inhibitors and siRNA mediated gene knockdown. Results were verified by in vitro digestion of a synthetic peptide substrate mimicking ABCA3's cleavage region, followed by MS analysis. RESULTS: We found that cleavage of ABCA3 occurs after Lys174 which is located in the proteins' first luminal loop. Inhibition of cathepsin L and, to a lesser extent, cathepsin B resulted in attenuation of ABCA3 cleavage. Both enzymes showed activity against the ABCA3 peptide in vitro with cathepsin L being more active. CONCLUSION: We show here that, like some other proteins of the lysosomal membrane, ABCA3 is a substrate of cathepsin L. Therefore, cathepsin L may represent a potential target to therapeutically influence ABCA3 activity in ABCA3-associated lung disease.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Péptido Hidrolasas/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Secuencia de Aminoácidos , Catepsina B/antagonistas & inhibidores , Catepsina B/genética , Catepsina B/metabolismo , Catepsina L/antagonistas & inhibidores , Catepsina L/genética , Catepsina L/metabolismo , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Humanos , Péptido Hidrolasas/química , Péptido Hidrolasas/genética , Péptidos/análisis , Estructura Terciaria de Proteína , Proteolisis , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Espectrometría de Masas en Tándem
12.
Nat Commun ; 7: 12897, 2016 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-27642082

RESUMEN

Recent studies revealed that mitochondrial Ca(2+) channels, which control energy flow, cell signalling and death, are macromolecular complexes that basically consist of the pore-forming mitochondrial Ca(2+) uniporter (MCU) protein, the essential MCU regulator (EMRE), and the mitochondrial Ca(2+) uptake 1 (MICU1). MICU1 is a regulatory subunit that shields mitochondria from Ca(2+) overload. Before the identification of these core elements, the novel uncoupling proteins 2 and 3 (UCP2/3) have been shown to be fundamental for mitochondrial Ca(2+) uptake. Here we clarify the molecular mechanism that determines the UCP2/3 dependency of mitochondrial Ca(2+) uptake. Our data demonstrate that mitochondrial Ca(2+) uptake is controlled by protein arginine methyl transferase 1 (PRMT1) that asymmetrically methylates MICU1, resulting in decreased Ca(2+) sensitivity. UCP2/3 normalize Ca(2+) sensitivity of methylated MICU1 and, thus, re-establish mitochondrial Ca(2+) uptake activity. These data provide novel insights in the complex regulation of the mitochondrial Ca(2+) uniporter by PRMT1 and UCP2/3.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Proteína Desacopladora 2/metabolismo , Calcio/metabolismo , Células HeLa , Humanos , Metilación , Procesamiento Proteico-Postraduccional , Proteína Desacopladora 3/metabolismo
13.
Br J Pharmacol ; 172(16): 4107-18, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25989290

RESUMEN

BACKGROUND AND PURPOSE: Highly vascularized ovarian carcinoma secretes the putative endocannabinoid and GPR55 agonist, L-α-lysophosphatidylinositol (LPI), into the circulation. We aimed to assess the involvement of this agonist and its receptor in ovarian cancer angiogenesis. EXPERIMENTAL APPROACH: Secretion of LPI by three ovarian cancer cell lines (OVCAR-3, OVCAR-5 and COV-362) was tested by mass spectrometry. Involvement of cancer cell-derived LPI on angiogenesis was tested in the in vivo chicken chorioallantoic membrane (CAM) assay along with the assessment of the effect of LPI on proliferation, network formation, and migration of neonatal and adult human endothelial colony-forming cells (ECFCs). Engagement of GPR55 was verified by using its pharmacological inhibitor CID16020046 and diminution of GPR55 expression by four different target-specific siRNAs. To study underlying signal transduction, Western blot analysis was performed. KEY RESULTS: Ovarian carcinoma cell-derived LPI stimulated angiogenesis in the CAM assay. Applied LPI stimulated proliferation, network formation, and migration of neonatal ECFCs in vitro and angiogenesis in the in vivo CAM. The pharmacological GPR55 inhibitor CID16020046 inhibited LPI-stimulated ECFC proliferation, network formation and migration in vitro as well as ovarian carcinoma cell- and LPI-induced angiogenesis in vivo. Four target-specific siRNAs against GPR55 prevented these effects of LPI on angiogenesis. These pro-angiogenic effects of LPI were transduced by GPR55-dependent phosphorylation of ERK1/2 and p38 kinase. CONCLUSIONS AND IMPLICATIONS: We conclude that inhibiting the pro-angiogenic LPI/GPR55 pathway appears a promising target against angiogenesis in ovarian carcinoma.


Asunto(s)
Lisofosfolípidos/metabolismo , Neovascularización Patológica/metabolismo , Neoplasias Ováricas/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Pollos , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/fisiología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Receptores de Cannabinoides , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Biol Open ; 3(12): 1164-72, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25395667

RESUMEN

Anandamide (N-arachidonyl ethanolamide, AEA) is an endogenous cannabinoid that is involved in various pathological conditions, including cardiovascular diseases and tumor-angiogenesis. Herein, we tested the involvement of classical cannabinoid receptors (CBRs) and the Ca(2+)-channel transient receptor potential vanilloid 1 (TRPV1) on cellular AEA uptake and its effect on endothelial cell proliferation and network-formation. Uptake of the fluorescence-labeled anandamide (SKM4-45-1) was monitored in human endothelial colony-forming cells (ECFCs) and a human endothelial-vein cell line (EA.hy926). Involvement of the receptors during AEA translocation was determined by selective pharmacological inhibition (AM251, SR144528, CID16020046, SB366791) and molecular interference by TRPV1-selective siRNA-mediated knock-down and TRPV1 overexpression. We show that exclusively TRPV1 contributes essentially to AEA transport into endothelial cells in a Ca(2+)-independent manner. This TRPV1 function is a prerequisite for AEA-induced endothelial cell proliferation and network-formation. Our findings point to a so far unknown moonlighting function of TRPV1 as Ca(2+)-independent contributor/regulator of AEA uptake. We propose TRPV1 as representing a promising target for development of pharmacological therapies against AEA-triggered endothelial cell functions, including their stimulatory effect on tumor-angiogenesis.

15.
PLoS One ; 8(6): e66909, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23826172

RESUMEN

Therapeutic neo-vasculogenesis in vivo can be achieved by the co-transplantation of human endothelial colony-forming progenitor cells (ECFCs) with mesenchymal stem/progenitor cells (MSPCs). The underlying mechanism is not completely understood thus hampering the development of novel stem cell therapies. We hypothesized that proteomic profiling could be used to retrieve the in vivo signaling signature during the initial phase of human neo-vasculogenesis. ECFCs and MSPCs were therefore either transplanted alone or co-transplanted subcutaneously into immune deficient mice. Early cell signaling, occurring within the first 24 hours in vivo, was analyzed using antibody microarray proteomic profiling. Vessel formation and persistence were verified in parallel transplants for up to 24 weeks. Proteomic analysis revealed significant alteration of regulatory components including caspases, calcium/calmodulin-dependent protein kinase, DNA protein kinase, human ErbB2 receptor-tyrosine kinase as well as mitogen-activated protein kinases. Caspase-4 was selected from array results as one therapeutic candidate for targeting vascular network formation in vitro as well as modulating therapeutic vasculogenesis in vivo. As a proof-of-principle, caspase-4 and general caspase-blocking led to diminished endothelial network formation in vitro and significantly decreased vasculogenesis in vivo. Proteomic profiling ex vivo thus unraveled a signaling signature which can be used for target selection to modulate neo-vasculogenesis in vivo.


Asunto(s)
Perfilación de la Expresión Génica , Neovascularización Fisiológica/genética , Proteómica/métodos , Transducción de Señal/genética , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Western Blotting , Inhibidores de Caspasas/farmacología , Caspasas/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Trasplante de Células Madre
16.
Methods Mol Biol ; 879: 381-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22610572

RESUMEN

Vessel wall-derived somatic endothelial colony-forming progenitor cells (ECFCs) are key players in vascular homeostasis and regeneration. Due to their robust proliferative potential and profound vessel-forming capacity, ECFCs are considered to represent an attractive tool for vascular regenerative medicine and a promising target for antiangiogenic tumor therapy. Here, we describe an easily applicable method for isolating ECFCs directly from unmanipulated adult human blood and an animal protein-free large-scale expansion system to generate more than 100 million functional ECFCs.


Asunto(s)
Separación Celular , Células Endoteliales/citología , Células Madre/citología , Animales , Proliferación Celular , Humanos
17.
PLoS One ; 7(9): e44468, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22970226

RESUMEN

Despite insights into the molecular pathways regulating hypoxia-induced gene expression, it is not known which cell types accomplish oxygen sensing during neo-vasculogenesis. We have developed a humanized mouse model of endothelial and mesenchymal progenitor co-transplantation to delineate the cellular compartments responsible for hypoxia response during vasculogenesis. Mesenchymal stem/progenitor cells (MSPCs) accumulated nuclear hypoxia-inducible transcription factor (HIF)-1α earlier and more sensitively than endothelial colony forming progenitor cells (ECFCs) in vitro and in vivo. Hypoxic ECFCs showed reduced function in vitro and underwent apoptosis within 24h in vivo when used without MSPCs. Surprisingly, only in MSPCs did pharmacologic or genetic inhibition of HIF-1α abrogate neo-vasculogenesis. HIF deletion in ECFCs caused no effect. ECFCs could be rescued from hypoxia-induced apoptosis by HIF-competent MSPCs resulting in the formation of patent perfused human vessels. Several angiogenic factors need to act in concert to partially substitute mesenchymal HIF-deficiency. Results demonstrate that ECFCs require HIF-competent vessel wall progenitors to initiate vasculogenesis in vivo and to bypass hypoxia-induced apoptosis. We describe a novel mechanistic role of MSPCs as oxygen sensors promoting vasculogenesis thus underscoring their importance for the development of advanced cellular therapies.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Modelos Animales , Neovascularización Fisiológica , Oxígeno/metabolismo , Animales , Apoptosis , Western Blotting , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/citología , Ratones , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
J Vis Exp ; (32)2009 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-19861942

RESUMEN

This paper introduces a novel recovery strategy for endothelial colony forming progenitor cells (ECFCs) from heparinized but otherwise unmanipulated adult human peripheral blood within a mean of 12 days. After large scale expansion >1x10(8) ECFCs can be obtained for further tests. Advantageously by using pHPL the contact of human cells with bovine serum antigens can be excluded. By flow cytometry and immunohistochemistry the isolated cells can be characterized as ECFC and their in vitro functionality to form vascular like structures can be tested in a matrigel assay. Further these cells can be subcutaneously injected in a mouse model to form functional, perfused vessels in vivo. After long term expansion and cryopreservation proliferation, function and genomic stability appear to be preserved. (3,4) This animal-protein free isolation and expansion method is easily applicable to generate a large quantity of ECFCs.


Asunto(s)
Células Sanguíneas/citología , Células Endoteliales/citología , Células Madre/citología , Adulto , Animales , Separación Celular/métodos , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Humanos , Inmunohistoquímica , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA