Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Immunity ; 56(12): 2816-2835.e13, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38091953

RESUMEN

Cancer cells can evade natural killer (NK) cell activity, thereby limiting anti-tumor immunity. To reveal genetic determinants of susceptibility to NK cell activity, we examined interacting NK cells and blood cancer cells using single-cell and genome-scale functional genomics screens. Interaction of NK and cancer cells induced distinct activation and type I interferon (IFN) states in both cell types depending on the cancer cell lineage and molecular phenotype, ranging from more sensitive myeloid to less sensitive B-lymphoid cancers. CRISPR screens in cancer cells uncovered genes regulating sensitivity and resistance to NK cell-mediated killing, including adhesion-related glycoproteins, protein fucosylation genes, and transcriptional regulators, in addition to confirming the importance of antigen presentation and death receptor signaling pathways. CRISPR screens with a single-cell transcriptomic readout provided insight into underlying mechanisms, including regulation of IFN-γ signaling in cancer cells and NK cell activation states. Our findings highlight the diversity of mechanisms influencing NK cell susceptibility across different cancers and provide a resource for NK cell-based therapies.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Humanos , Células Asesinas Naturales , Neoplasias/genética , Presentación de Antígeno , Genómica , Citotoxicidad Inmunológica/genética , Línea Celular Tumoral
2.
Semin Cancer Biol ; 39: 68-76, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27544796

RESUMEN

The nuclear factor-κB (NF-κB) transcription factor family plays critical roles in the pathophysiology of hematologic neoplasias, including multiple myeloma. The current review examines the roles that this transcription factor system plays in multiple myeloma cells and the nonmalignant accessory cells of the local microenvironment; as well as the evidence indicating that a large proportion of myeloma patients harbor genomic lesions which perturb diverse genes regulating the activity of NF-κB. This article also discusses the therapeutic targeting of the NF-κB pathway using proteasome inhibitors, a pharmacological class that has become a cornerstone in the therapeutic management of myeloma; and reviews some of the future challenges and opportunities for NF-κB-related research in myeloma.


Asunto(s)
Mieloma Múltiple/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Humanos , Terapia Molecular Dirigida/métodos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Transducción de Señal , Microambiente Tumoral
3.
J Immunother Cancer ; 11(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36754452

RESUMEN

BACKGROUND: The MUC1-C protein evolved in mammals to protect barrier tissues from loss of homeostasis; however, MUC1-C promotes oncogenesis in association with chronic inflammation. Aberrant expression of MUC1-C in cancers has been linked to depletion and dysfunction of T cells in the tumor microenvironment. In contrast, there is no known involvement of MUC1-C in the regulation of natural killer (NK) cell function. METHODS: Targeting MUC1-C genetically and pharmacologically in cancer cells was performed to assess effects on intracellular and cell surface expression of the MHC class I chain-related polypeptide A (MICA) and MICB ligands. The MICA/B promoters were analyzed for H3K27 and DNA methylation. Shedding of MICA/B was determined by ELISA. MUC1-C interactions with ERp5 and RAB27A were assessed by coimmunoprecipitation and direct binding studies. Exosomes were isolated for analysis of secretion. Purified NK cells were assayed for killing of cancer cell targets. RESULTS: Our studies demonstrate that MUC1-C represses expression of the MICA and MICB ligands that activate the NK group 2D receptor. We show that the inflammatory MUC1-C→NF-κB pathway drives enhancer of zeste homolog 2-mediated and DNMT-mediated methylation of the MICA and MICB promoter regions. Targeting MUC1-C genetically and pharmacologically with the GO-203 inhibitor induced intracellular and cell surface MICA/B expression but not MICA/B cleavage. Mechanistically, MUC1-C regulates the ERp5 thiol oxidoreductase that is necessary for MICA/B protease digestion and shedding. In addition, MUC1-C interacts with the RAB27A protein, which is required for exosome formation and secretion. As a result, targeting MUC1-C markedly inhibited secretion of exosomes expressing MICA/B. In concert with these results, we show that targeting MUC1-C promotes NK cell-mediated killing. CONCLUSIONS: These findings uncover pleotropic mechanisms by which MUC1-C confers evasion of cancer cells to NK cell recognition and destruction.


Asunto(s)
Exosomas , Mucina-1 , Neoplasias , Humanos , Exosomas/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Asesinas Naturales , Ligandos , Mucina-1/genética , Mucina-1/metabolismo , Neoplasias/patología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Microambiente Tumoral
4.
J Clin Invest ; 133(22)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37751299

RESUMEN

The B cell leukemia/lymphoma 2 (BCL-2) inhibitor venetoclax is effective in chronic lymphocytic leukemia (CLL); however, resistance may develop over time. Other lymphoid malignancies such as diffuse large B cell lymphoma (DLBCL) are frequently intrinsically resistant to venetoclax. Although genomic resistance mechanisms such as BCL2 mutations have been described, this probably only explains a subset of resistant cases. Using 2 complementary functional precision medicine techniques - BH3 profiling and high-throughput kinase activity mapping - we found that hyperphosphorylation of BCL-2 family proteins, including antiapoptotic myeloid leukemia 1 (MCL-1) and BCL-2 and proapoptotic BCL-2 agonist of cell death (BAD) and BCL-2 associated X, apoptosis regulator (BAX), underlies functional mechanisms of both intrinsic and acquired resistance to venetoclax in CLL and DLBCL. Additionally, we provide evidence that antiapoptotic BCL-2 family protein phosphorylation altered the apoptotic protein interactome, thereby changing the profile of functional dependence on these prosurvival proteins. Targeting BCL-2 family protein phosphorylation with phosphatase-activating drugs rewired these dependencies, thus restoring sensitivity to venetoclax in a panel of venetoclax-resistant lymphoid cell lines, a resistant mouse model, and in paired patient samples before venetoclax treatment and at the time of progression.


Asunto(s)
Antineoplásicos , Leucemia Linfocítica Crónica de Células B , Linfoma de Células B Grandes Difuso , Ratones , Animales , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/patología , Resistencia a Antineoplásicos/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteína bcl-X/genética , Proteínas Reguladoras de la Apoptosis , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo
5.
Nat Cancer ; 4(5): 754-773, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37237081

RESUMEN

Clinical progress in multiple myeloma (MM), an incurable plasma cell (PC) neoplasia, has been driven by therapies that have limited applications beyond MM/PC neoplasias and do not target specific oncogenic mutations in MM. Instead, these agents target pathways critical for PC biology yet largely dispensable for malignant or normal cells of most other lineages. Here we systematically characterized the lineage-preferential molecular dependencies of MM through genome-scale clustered regularly interspaced short palindromic repeats (CRISPR) studies in 19 MM versus hundreds of non-MM lines and identified 116 genes whose disruption more significantly affects MM cell fitness compared with other malignancies. These genes, some known, others not previously linked to MM, encode transcription factors, chromatin modifiers, endoplasmic reticulum components, metabolic regulators or signaling molecules. Most of these genes are not among the top amplified, overexpressed or mutated in MM. Functional genomics approaches thus define new therapeutic targets in MM not readily identifiable by standard genomic, transcriptional or epigenetic profiling analyses.


Asunto(s)
Mieloma Múltiple , Humanos , Mieloma Múltiple/genética , Genómica , Genoma , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética
6.
Nat Genet ; 53(8): 1196-1206, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34253920

RESUMEN

To systematically define molecular features in human tumor cells that determine their degree of sensitivity to human allogeneic natural killer (NK) cells, we quantified the NK cell responsiveness of hundreds of molecularly annotated 'DNA-barcoded' solid tumor cell lines in multiplexed format and applied genome-scale CRISPR-based gene-editing screens in several solid tumor cell lines, to functionally interrogate which genes in tumor cells regulate the response to NK cells. In these orthogonal studies, NK cell-sensitive tumor cells tend to exhibit 'mesenchymal-like' transcriptional programs; high transcriptional signature for chromatin remodeling complexes; high levels of B7-H6 (NCR3LG1); and low levels of HLA-E/antigen presentation genes. Importantly, transcriptional signatures of NK cell-sensitive tumor cells correlate with immune checkpoint inhibitor (ICI) resistance in clinical samples. This study provides a comprehensive map of mechanisms regulating tumor cell responses to NK cells, with implications for future biomarker-driven applications of NK cell immunotherapies.


Asunto(s)
Citotoxicidad Inmunológica/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Inhibidores de Puntos de Control Inmunológico/farmacología , Células Asesinas Naturales/fisiología , Células Alogénicas/fisiología , Animales , Antígenos B7/genética , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/fisiología , Pruebas Inmunológicas de Citotoxicidad/métodos , Citotoxicidad Inmunológica/fisiología , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Genoma Humano , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Ratones Endogámicos NOD , Ensayos Antitumor por Modelo de Xenoinjerto , Antígenos HLA-E
7.
Cell Rep ; 34(1): 108532, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33406420

RESUMEN

Heterobifunctional proteolysis-targeting chimeric compounds leverage the activity of E3 ligases to induce degradation of target oncoproteins and exhibit potent preclinical antitumor activity. To dissect the mechanisms regulating tumor cell sensitivity to different classes of pharmacological "degraders" of oncoproteins, we performed genome-scale CRISPR-Cas9-based gene editing studies. We observed that myeloma cell resistance to degraders of different targets (BET bromodomain proteins, CDK9) and operating through CRBN (degronimids) or VHL is primarily mediated by prevention of, rather than adaptation to, breakdown of the target oncoprotein; and this involves loss of function of the cognate E3 ligase or interactors/regulators of the respective cullin-RING ligase (CRL) complex. The substantial gene-level differences for resistance mechanisms to CRBN- versus VHL-based degraders explains mechanistically the lack of cross-resistance with sequential administration of these two degrader classes. Development of degraders leveraging more diverse E3 ligases/CRLs may facilitate sequential/alternating versus combined uses of these agents toward potentially delaying or preventing resistance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos/farmacología , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Quinasa 9 Dependiente de la Ciclina/metabolismo , Resistencia a Antineoplásicos , Edición Génica , Regulación Neoplásica de la Expresión Génica , Genes Sobrepuestos , Estudio de Asociación del Genoma Completo , Genómica/métodos , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Proteínas Oncogénicas/metabolismo , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Proteolisis , Células Tumorales Cultivadas
8.
Int J Cancer ; 124(8): 1773-7, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19101987

RESUMEN

Prostate cancer commonly affects men in the Western world. A major factor of the life-threatening course of this disease is the high rate of metastasis, predominantly to bones. Circulating tumor cells encounter platelets and may activate them, resulting in a production of microparticles (MPs). MPs are small platelet fragments expressing membrane receptors as well as cytoplasmic constituents. Here, we report that prostate cancer cells, Clone-1 (Cl-1), preincubated with platelet-derived MPs (PMPs), demonstrate increased invasion through a gelatin-coated (a denatured form of collagen) membrane of the Boyden chamber system. This effect was accompanied by an increased secretion of metalloproteinase-2 (MMP-2) as demonstrated by a gelatin zymography. Application of MMP-2/9 inhibitor reversed the PMP-induced tumor cell invasion. PMPs were shown to adhere to Cl-1 cells, but direct contact between them may not be mandatory for MMP secretion because PMP lysate induced MMP-2 production by Cl-1 cells to the same extent as did intact PMPs. PMP-induced MMP-2 secretion was inhibited by neutralization of either PKC or total intracellular tyrosine phosphorylation, but was not affected by blocking major intraplatelet cytokines. Actinomycin D (a transcription inhibitor) did not modify this effect, whereas cycloheximide (an inhibitor of protein translation) abolished the MMP-2 release. MMP-2 secretion was accompanied by a rapid and transient increase in MMP-2 mRNA level after a 2-hr coincubation of prostate cancer cells with PMPs. Thus, PMPs promote tumor invasiveness, at least in part by stimulation of MMP-2 production.


Asunto(s)
Plaquetas/metabolismo , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 2 de la Matriz/metabolismo , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología , Adhesión Celular , Técnicas de Cultivo de Célula/instrumentación , Cicloheximida/farmacología , Citoplasma/metabolismo , Dactinomicina/farmacología , Gelatina/química , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Factores de Tiempo , Regulación hacia Arriba
9.
Cardiovasc Res ; 67(1): 30-8, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15878159

RESUMEN

OBJECTIVE: Platelet activation is accompanied by the release of microparticles. However, little is known about the role of platelet-derived microparticles (PMP) in the regulation of angiogenesis and related clinical situations. The aim of our study was to evaluate the effect of PMP on angiogenesis and to analyze its mechanisms. METHODS: Both in vitro (rat aortic ring model, cell invasion test) and in vivo (agarose bead transplantation, artificial cardiac ischemia in Sabra rats) approaches were used in the study. RESULTS: A dose-dependent pro-angiogenic effect of PMP was observed in the rat aortic ring model. This effect could be eliminated by inhibition of VEGF, bFGF, and PDGF, but not heparanase. PMP exerted their effect via PI 3-kinase, Src kinase, and ERK, whereas protein kinase C and p38 were not involved. Moreover, PMP induced invasion of endothelial cells through a layer of matrigel. This effect was mediated by VEGF, heparanase, and PDGF, but not bFGF. Furthermore, PMP induced angiogenesis in an in vivo model in which agarose beads containing PMP were transplanted subcutaneously into mice. In addition, the effect of PMP on angiogenesis was evaluated in the model of in vivo chronic myocardial ischemia in rats. Ischemia induced a decrease in the number of functioning capillaries (34+/-21.5 vs. 157+/-42.0 per view field), but their amount increased after injection of PMP into the myocarium (97+/-27.3; p<0.001 vs. ischemia without PMP). CONCLUSIONS: PMP induce angiogenesis both in vitro and in vivo. Injection of PMP into the ischemic myocardium might improve the process of revascularization after chronic ischemia.


Asunto(s)
Plaquetas/fisiología , Isquemia Miocárdica/metabolismo , Neovascularización Fisiológica/fisiología , Activación Plaquetaria , Animales , Aorta , Capilares , Movimiento Celular , Citocinas/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Técnicas In Vitro , Modelos Animales , Nanoestructuras , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Ratas Endogámicas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
PLoS One ; 11(12): e0166655, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27935954

RESUMEN

Both normal wound healing and tumor angiogenesis are mitigated by the sequential, carefully orchestrated release of growth stimulators and inhibitors. These regulators are released from platelet clots formed at the sites of activated endothelium in a temporally and spatially controlled manner, and the order of their release depends on their affinity to glycosaminoglycans (GAG) such as heparan sulfate (HS) within the extracellular matrix, and platelet open canallicular system. The formation of vessel sprouts, triggered by angiogenesis regulating factors with lowest affinities for heparan sulfate (e.g. VEGF), is followed by vessel-stabilizing PDGF-B or bFGF with medium affinity for HS, and by inhibitors such as PF-4 and TSP-1 with the highest affinities for HS. The invasive wound-like edge of growing tumors has an overabundance of angiogenesis stimulators, and we propose that their abundance out-competes angiogenesis inhibitors, effectively preventing inhibition of angiogenesis and vessel maturation. We evaluate this hypothesis using an experimentally motivated agent-based model, and propose a general theoretical framework for understanding mechanistic similarities and differences between the processes of normal wound healing and pathological angiogenesis from the point of view of competitive inhibition.


Asunto(s)
Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Microambiente Tumoral , Cicatrización de Heridas , Animales , Unión Competitiva , Plaquetas/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Modelos Biológicos , Neoplasias/irrigación sanguínea , Neovascularización Fisiológica , Factor Plaquetario 4/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-sis/metabolismo , Trombospondina 1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Thromb Haemost ; 110(2): 323-30, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23765126

RESUMEN

Platelets contain chemo-attractants and mitogens that have a major role in tissue repair. Therefore we hypothesised that tissue regeneration secondary to activation of endogenous neural stem cells (eNSC) can be enhanced by delivering platelets to the ischaemic brain. To examine these potential therapeutic effects we injected platelet-poor plasma (PPP), fibroblast growth factor (FGF2) and platelet lysate (PLT) to the lateral ventricles after permanent middle cerebral artery occlusion (PMCAO) in rats. The animals were tested with the neurological severity score, and infarct volumes were measured at 90 days post-PMCAO. Immunohistochemistry was used to determine the fate of newborn cells and to count blood vessels in the ischaemic brain. Platelets significantly increased eNSC proliferation and angiogenesis in the subventricular zone (SVZ) and in the peri-lesion cortex. Functional outcome was significantly improved and injury size was significantly reduced in rats treated with PLT suggesting additional neuroprotective effects. In conclusion, local delivery of PLT to the lateral ventricles induces angiogenesis, neurogenesis and neuroprotection and reduces behavioural deficits after brain ischaemia.


Asunto(s)
Plaquetas/fisiología , Infarto de la Arteria Cerebral Media/terapia , Animales , Conducta Animal , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Inyecciones Intraventriculares , Masculino , Neovascularización Fisiológica , Células-Madre Neurales/patología , Células-Madre Neurales/fisiología , Neurogénesis , Fármacos Neuroprotectores , Ratas , Ratas Endogámicas SHR , Nicho de Células Madre
12.
Thromb Res ; 130 Suppl 1: S98-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23026678

RESUMEN

Platelets play a major role in hemostasis, but are also involved in vascular biology processes such as angiogenesis and tumor metastasis. Activated platelets release many proteins favoring wound healing and promoting angiogenesis. Microparticles (MP) are small plasma membrane vesicles shed from cells upon their activation or apoptosis. Platelet-derived microparticles (PMP) constitute the majority of the pool of MP circulating in the blood. PMP express and may transfer functional receptors, stimulate the release of cytokines, activate intracellular signaling pathways, promote angiogenesis, and are involved in tissue regeneration and cancer metastasis. We investigated the effect of PMP on cancer cells metastasis and their potential beneficial effect in an ischemic stroke model.


Asunto(s)
Plaquetas/metabolismo , Encéfalo/metabolismo , Micropartículas Derivadas de Células/metabolismo , Infarto de la Arteria Cerebral Media/sangre , Neoplasias Pulmonares/sangre , Neoplasias de la Próstata/sangre , Regeneración , Animales , Apoptosis , Plaquetas/patología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Encéfalo/fisiopatología , Proliferación Celular , Micropartículas Derivadas de Células/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/secundario , Masculino , Ratones , Invasividad Neoplásica , Neovascularización Patológica , Neovascularización Fisiológica , Neurogénesis , Activación Plaquetaria , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/patología , Ratas , Transducción de Señal
13.
J Mol Neurosci ; 47(3): 659-65, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22290563

RESUMEN

Platelet microparticles (PMP) are small subcellular fragments, shed upon platelet activation. PMP host a variety of cytokines and growth factor that were previously shown to affect angiogenesis and postischemic tissue regeneration. This study attempted to explore the effect of PMP on neural stem cell (NSC) proliferation, survival and differentiation. Cells were grown as neurospheres and treated with PMP, or relevant growth factors, sphere size and cell fates were evaluated. PMP treatment led to larger neurospheres with increased cell survival. PMP treatment was comparable with the effect of acceptable single growth factors such as fibroblastic growth factor (FGF), vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF). PMP treatment also increased the differentiation potential of NSC to glia and neurons. Specific growth factor inhibitors only partly blocked these effects, which were associated with increments in ERK and Akt phosphorylation. In this study, we show that various growth factors contained within the PMP promote neuronal cell proliferation, survival and differentiation. The results suggest a role for platelet microparticles in augmenting endogenous neural progenitor and stem cells angiogenesis and neurogenesis that might be utilized for treatment following brain injury.


Asunto(s)
Plaquetas/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Micropartículas Derivadas de Células/fisiología , Células-Madre Neurales/citología , Animales , Plaquetas/citología , Supervivencia Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Femenino , Humanos , Ratones , Células-Madre Neurales/fisiología
14.
Curr Neurovasc Res ; 9(3): 185-92, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22621230

RESUMEN

Activated platelets shed microparticles, which contain a variety of growth factors central to angiogenesis and neurogenesis. The aim of this study was to explore whether platelet derived microparticles (PMP) can boost endogenous neural stem cells dependent repair mechanisms following stroke in a rat model. To examine the effects of PMP therapy in-vivo, we delivered PMP or vehicle via a biodegradable polymer to the brain surface after permanent middle cerebral artery occlusion (PMCAO) in rats. Rats were tested with the neurological severity score and infarct volumes were measured at 90 days post-ischemia. Immunohistochemistry was used to determine the fate of newborn cells and to count blood vessels in the ischemic brain. The results show that PMP led to a dose dependent increase in cell proliferation, neurogenesis and angiogenesis at the infarct boundary zone and significantly improved behavioral deficits.


Asunto(s)
Plaquetas/química , Isquemia Encefálica/terapia , Micropartículas Derivadas de Células/fisiología , Neovascularización Fisiológica/fisiología , Neurogénesis/fisiología , Análisis de Varianza , Animales , Plaquetas/citología , Isquemia Encefálica/etiología , Bromodesoxiuridina/metabolismo , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Evaluación de la Discapacidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Infarto de la Arteria Cerebral Media/complicaciones , Neovascularización Fisiológica/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/fisiología , Neurogénesis/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Recuperación de la Función , Factores de Tiempo
15.
Br J Haematol ; 134(4): 426-31, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16822291

RESUMEN

C-reactive protein (CRP) is a strong predictor for acute cardiovascular events. Several endothelial prothrombotic effects of CRP have been recently reported. This study examined the effect of CRP on bovine aortic endothelial cell (EC) activation and capacity to recruit human platelets under flow conditions using the cone and plate(let) analyser method. Human recombinant CRP promoted platelet adhesion in a dose- and time-dependent manner, with a maximal effect at 20 microg/ml (increase of 174% over baseline, P < 0.01). Similar effects were observed following incubation of EC with sera of transgenic mice that express human CRP (10 microg/ml). Anti-intercellular adhesion molecule-1 neutralising monoclonal antibody and nitric oxide donor, sodium nitroprusside, blocked the effect of CRP, reducing adhesion from 202% to 128% (P < 0.05) and 114% (P = 0.02) respectively. The pro-adhesive effect of CRP was abolished by calphostin C (a protein kinase C inhibitor), whereas the extracellular signal-regulated kinase antagonist, PD98059, did not have any effect. CRP promoted P-selectin expression on the EC surface and blockade of P-selectin reversed CRP-induced platelet adhesion. In conclusion, CRP promoted platelet adhesion to EC. Our results emphasise the possible role of CRP in linking inflammation and thrombosis and provide a potential mechanism for the high incidence of vascular events associated with high CRP levels.


Asunto(s)
Aterosclerosis/patología , Proteína C-Reactiva/farmacología , Células Endoteliales/patología , Endotelio Vascular/patología , Análisis de Varianza , Animales , Anticuerpos Monoclonales/farmacología , Proteína C-Reactiva/genética , Bovinos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Eptifibatida , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Selectina-P/inmunología , Péptidos/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA