Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Intervalo de año de publicación
1.
Adv Exp Med Biol ; 1427: 13-22, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37322331

RESUMEN

The carotid body is the most relevant oxygen sensor in mammalian organisms. This organ helps to detect acute changes in PO2, but it is also crucial for the organismal adaptation to a maintained hypoxemia. Profound angiogenic and neurogenic processes take place in the carotid body to facilitate this adaptation process. We have described a plethora of multipotent stem cells and restricted progenitors, from both vascular and neuronal lineages, existing in the quiescent normoxic carotid body, ready to contribute to organ growth and adaptation upon the arrival of the hypoxic stimulus. Our deep understanding of the functioning of this stunning germinal niche will very likely facilitate the management and treatment of an important group of diseases that course with carotid body over-activation and malfunction.


Asunto(s)
Cuerpo Carotídeo , Animales , Adulto , Humanos , Cuerpo Carotídeo/fisiología , Neuronas/fisiología , Células Madre Multipotentes , Neurogénesis , Hipoxia , Mamíferos
2.
Int J Mol Sci ; 24(6)2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36982650

RESUMEN

Antiparkinsonian carotid body (CB) cell therapy has been proven to be effective in rodent and nonhuman primate models of Parkinson's disease (PD), exerting trophic protection and restoration of the dopaminergic nigrostriatal pathway. These neurotrophic actions are mediated through the release of high levels of glial-cell-line-derived neurotrophic factor (GDNF) by the CB transplant. Pilot clinical trials have also shown that CB autotransplantation can improve motor symptoms in PD patients, although its effectiveness is affected by the scarcity of the grafted tissue. Here, we analyzed the antiparkinsonian efficacy of in vitro-expanded CB dopaminergic glomus cells. Intrastriatal xenografts of rat CB neurospheres were shown to protect nigral neurons from degeneration in a chronic MPTP mouse PD model. In addition, grafts performed at the end of the neurotoxic treatment resulted in the repair of striatal dopaminergic terminals through axonal sprouting. Interestingly, both neuroprotective and reparative effects induced by in vitro-expanded CB cells were similar to those previously reported by the use of CB transplants. This action could be explained because stem-cell-derived CB neurospheres produce similar amounts of GDNF compared to native CB tissue. This study provides the first evidence that in vitro-expanded CB cells could be a clinical option for cell therapy in PD.


Asunto(s)
Cuerpo Carotídeo , Enfermedad de Parkinson , Ratones , Ratas , Humanos , Animales , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Cuerpo Carotídeo/metabolismo , Enfermedad de Parkinson/terapia , Enfermedad de Parkinson/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Trasplante de Células , Sustancia Negra/metabolismo , Modelos Animales de Enfermedad , Cuerpo Estriado/metabolismo
3.
Int J Mol Sci ; 24(2)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36675105

RESUMEN

Neuroblastoma is a neural crest cell-derived pediatric tumor characterized by high inter- and intra-tumor heterogeneity, and by a poor outcome in advanced stages. Patient-derived xenografts (PDXs) have been shown to be useful models for preserving and expanding original patient biopsies in vivo, and for studying neuroblastoma biology in a more physiological setting. The maintenance of genetic, histologic, and phenotypic characteristics of the original biopsy along serial PDX passages in mice is a major concern regarding this model. Here we analyze consecutive PDX passages in mice, at both transcriptomic and histological levels, in order to identify potential changes or highlight similarities to the primary sample. We studied temporal changes using mRNA and miRNA expression and correlate those with neuroblastoma aggressiveness using patient-derived databases. We observed a shortening of tumor onset and an increase in proliferative potential in the PDXs along serial passages. This behavior correlates with changes in the expression of genes related to cell proliferation and neuronal differentiation, including signaling pathways described as relevant for neuroblastoma malignancy. We also identified new genes and miRNAs that can be used to stratify patients according to survival, and which could be potential new players in neuroblastoma aggressiveness. Our results highlight the usefulness of the PDX neuroblastoma model and reflect phenotypic changes that might be occurring in the mouse environment. These findings could be useful for understanding the progression of tumor aggressiveness in this pathology.


Asunto(s)
MicroARNs , Neuroblastoma , Humanos , Animales , Ratones , Pase Seriado , Neuroblastoma/metabolismo , Transcriptoma , Perfilación de la Expresión Génica , Proliferación Celular , MicroARNs/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Physiol ; 601(5): 1017-1036, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36647759

RESUMEN

The carotid body (CB) is a prototypical acute oxygen (O2 )-sensing organ that mediates reflex hyperventilation and increased cardiac output in response to hypoxaemia. CB overactivation, secondary to the repeated stimulation produced by the recurrent episodes of intermittent hypoxia, is believed to contribute to the pathogenesis of sympathetic hyperactivity present in sleep apnoea patients. Although CB functional plasticity induced by chronic intermittent hypoxia (CIH) has been demonstrated, the underlying mechanisms are not fully elucidated. Here, we show that CIH induces a small increase in CB volume and rearrangement of cell types in the CB, characterized by a mobilization of immature quiescent neuroblasts, which enter a process of differentiation into mature, O2 -sensing and neuron-like, chemoreceptor glomus cells. Prospective isolation of individual cell classes has allowed us to show that maturation of CB neuroblasts is paralleled by an upregulation in the expression of specific glomus cell genes involved in acute O2 -sensing. CIH enhances mitochondrial responsiveness to hypoxia in maturing neuroblasts as well as in glomus cells. These data provide novel perspectives on the pathogenesis of CB-mediated sympathetic overflow that may lead to the development of new pharmacological strategies of potential applicability in sleep apnoea patients. KEY POINTS: Obstructive sleep apnoea is a frequent condition in the human population that predisposes to severe cardiovascular and metabolic alterations. Activation of the carotid body, the main arterial oxygen-sensing chemoreceptor, by repeated episodes of hypoxaemia induces exacerbation of the carotid body-mediated chemoreflex and contributes to sympathetic overflow characteristic of sleep apnoea patients. In rats, chronic intermittent hypoxaemia induces fast neurogenesis in the carotid body with rapid activation of neuroblasts, which enter a process of proliferation and maturation into O2 -sensing chemoreceptor glomus cells. Maturing carotid body neuroblasts and glomus cells exposed to chronic intermittent hypoxia upregulate genes involved in acute O2 sensing and enhance mitochondrial responsiveness to hypoxia. These findings provide novel perspectives on the pathogenesis of carotid body-mediated sympathetic hyperactivation. Pharmacological modulation of carotid body fast neurogenesis could help to ameliorate the deleterious effects of chronic intermittent hypoxaemia in sleep apnoea patients.


Asunto(s)
Cuerpo Carotídeo , Apnea Obstructiva del Sueño , Ratas , Humanos , Animales , Cuerpo Carotídeo/metabolismo , Hipoxia , Oxígeno/metabolismo , Neurogénesis
5.
STAR Protoc ; 3(2): 101260, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35345594

RESUMEN

The existence of a subpopulation of undifferentiated cells with stem-like properties has been suggested in neuroblastoma tumors, but a definitive biomarker for their successful isolation is missing. Here we describe an in vitro culture system for the enrichment in undifferentiated stem-like tumor cells for subsequent functional assays. We make use of clonal non-adherent cell culture conditions together with cell sorting with specific expression markers. This protocol allows for the differential study of heterogeneous cell population in neuroblastoma tumors. For complete details on the use and execution of this protocol, please refer to Vega et al. (2019).


Asunto(s)
Neuroblastoma , Técnicas de Cultivo de Célula/métodos , Línea Celular , Separación Celular , Células Cultivadas , Humanos , Neuroblastoma/genética
6.
Int J Mol Sci ; 21(21)2020 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-33153142

RESUMEN

The carotid body (CB), a neural-crest-derived organ and the main arterial chemoreceptor in mammals, is composed of clusters of cells called glomeruli. Each glomerulus contains neuron-like, O2-sensing glomus cells, which are innervated by sensory fibers of the petrosal ganglion and are located in close contact with a dense network of fenestrated capillaries. In response to hypoxia, glomus cells release transmitters to activate afferent fibers impinging on the respiratory and autonomic centers to induce hyperventilation and sympathetic activation. Glomus cells are embraced by interdigitating processes of sustentacular, glia-like, type II cells. The CB has an extraordinary structural plasticity, unusual for a neural tissue, as it can grow several folds its size in subjects exposed to sustained hypoxia (as for example in high altitude dwellers or in patients with cardiopulmonary diseases). CB growth in hypoxia is mainly due to the generation of new glomeruli and blood vessels. In recent years it has been shown that the adult CB contains a collection of quiescent multipotent stem cells, as well as immature progenitors committed to the neurogenic or the angiogenic lineages. Herein, we review the main properties of the different cell types in the CB germinal niche. We also summarize experimental data suggesting that O2-sensitive glomus cells are the master regulators of CB plasticity. Upon exposure to hypoxia, neurotransmitters and neuromodulators released by glomus cells act as paracrine signals that induce proliferation and differentiation of multipotent stem cells and progenitors, thus causing CB hypertrophy and an increased sensory output. Pharmacological modulation of glomus cell activity might constitute a useful clinical tool to fight pathologies associated with exaggerated sympathetic outflow due to CB overactivation.


Asunto(s)
Cuerpo Carotídeo/citología , Neurotransmisores/fisiología , Nicho de Células Madre/fisiología , Adaptación Fisiológica/fisiología , Animales , Diferenciación Celular/fisiología , Humanos , Hipoxia/metabolismo , Hipoxia/fisiopatología , Neurogénesis/fisiología , Neuronas/fisiología , Neurotransmisores/metabolismo , Oxígeno/metabolismo
7.
Cancers (Basel) ; 12(11)2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33233777

RESUMEN

Neuroblastoma (NB) is one of the most common pediatric cancers and presents a poor survival rate in affected children. Current pretreatment risk assessment relies on a few known molecular parameters, like the amplification of the oncogene MYCN. However, a better molecular knowledge about the aggressive progression of the disease is needed to provide new therapeutical targets and prognostic markers and to improve patients' outcomes. The human protein kinase VRK1 phosphorylates various signaling molecules and transcription factors to regulate cell cycle progression and other processes in physiological and pathological situations. Using neuroblastoma tumor expression data, tissue microarrays from fresh human samples and patient-derived xenografts (PDXs), we have determined that VRK1 kinase expression stratifies patients according to tumor aggressiveness and survival, allowing the identification of patients with worse outcome among intermediate risk. VRK1 associates with cell cycle signaling pathways in NB and its downregulation abrogates cell proliferation in vitro and in vivo. Through the analysis of ChIP-seq and methylation data from NB tumors, we show that VRK1 is a MYCN gene target, however VRK1 correlates with NB aggressiveness independently of MYCN gene amplification, synergizing with the oncogene to drive NB progression. Our study also suggests that VRK1 inhibition may constitute a novel cell-cycle-targeted strategy for anticancer therapy in neuroblastoma.

10.
Int J Mol Sci ; 21(1)2019 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-31861671

RESUMEN

Neuroblastoma is the most frequent extracranial solid tumour in children, causing 10% of all paediatric oncology deaths. It arises in the embryonic neural crest due to an uncontrolled behaviour of sympathetic nervous system progenitors, giving rise to heterogeneous tumours. Low local or systemic tissue oxygen concentration has emerged as a cellular stimulus with important consequences for tumour initiation, evolution and progression. In neuroblastoma, several evidences point towards a role of hypoxia in tumour initiation during development, tumour cell differentiation, survival and metastatic spreading. However, the heterogeneous nature of the disease, its developmental origin and the lack of suitable experimental models have complicated a clear understanding of the effect of hypoxia in neuroblastoma tumour progression and the molecular mechanisms implicated. In this review, we have compiled available evidences to try to shed light onto this important field. In particular, we explore the effect of hypoxia in neuroblastoma cell transformation and differentiation. We also discuss the experimental models available and the emerging alternatives to study this problem, and we present hypoxia-related therapeutic avenues being explored in the field.


Asunto(s)
Redes Reguladoras de Genes , Metástasis de la Neoplasia/genética , Neuroblastoma/genética , Diferenciación Celular , Niño , Progresión de la Enfermedad , Humanos , Hipoxia Tumoral
11.
EBioMedicine ; 49: 82-95, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31685444

RESUMEN

BACKGROUND: Neuroblastoma is a paediatric tumour originated from sympathoadrenal precursors and characterized by its heterogeneity and poor outcome in advanced stages. Intra-tumoral cellular heterogeneity has emerged as an important feature in neuroblastoma, with a potential major impact on tumour aggressiveness and response to therapy. CD44 is an adhesion protein involved in tumour progression, metastasis and stemness in different cancers; however, there has been controversies about the significance of CD44 expression in neuroblastoma and its relationship with tumour progression. METHODS: We have performed transcriptomic analysis on patient tumour samples studying the outcome of patients with high CD44 expression. Adhesion, invasion and proliferation assays were performed in sorted CD44high neuroblastoma cells. Tumoursphere cultures have been used to enrich in undifferentiated stem-like cells and to asses self-renewal and differentiation potential. We have finally performed in vivo tumorigenic assays on cell line-derived or Patient-derived xenografts. FINDINGS: We show that high CD44 expression is associated with low survival in high-grade human neuroblastoma, independently of MYCN amplification. CD44 is expressed in a cell population with neural crest stem-like features, and with the capacity to generate multipotent, undifferentiated tumourspheres in culture. These cells are more invasive and proliferative in vitro. CD44 positive cells obtained from tumours are more tumorigenic and metastatic, giving rise to aggressive neuroblastic tumours at high frequency upon transplantation. INTERPRETATION: We describe an unexpected intra-tumoural heterogeneity within cellular entities expressing CD44 in neuroblastoma, and propose that CD44 has a role in neural crest stem-like undifferentiated cells, which can contribute to tumorigenesis and malignancy in this type of cancer. FUNDING: Research supported by grants from the "Asociación Española contra el Cáncer" (AECC), the Spanish Ministry of Science and Innovation SAF program (SAF2016-80412-P), and the European Research Council (ERC Starting Grant to RP).


Asunto(s)
Receptores de Hialuranos/metabolismo , Cresta Neural/patología , Células-Madre Neurales/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Ratones SCID , Células Madre Multipotentes/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Esferoides Celulares/patología , Análisis de Supervivencia
12.
Adv Exp Med Biol ; 1123: 19-38, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31016593

RESUMEN

Somatic stem cells confer plasticity to adult tissues, permitting their maintenance, repair and adaptation to a changing environment. Adult germinal niches supporting somatic stem cells have been thoroughly characterized throughout the organism, including in central and peripheral nervous systems. Stem cells do not reside alone within their niches, but they are rather accompanied by multiple progenitor cells that not only contribute to the progression of stem cell lineage but also regulate their behavior. Understanding the mechanisms underlying these interactions within the niche is crucial to comprehend associated pathologies and to use stem cells in cell therapy. We have described a stunning germinal niche in the adult peripheral nervous system: the carotid body. This is a chemoreceptor organ with a crucial function during physiological adaptation to hypoxia. We have shown the presence of multipotent stem cells within this niche, escorted by multiple restricted progenitor cell types that contribute to niche physiology and hence organismal adaptation to the lack of oxygen. Herein, we discuss new and existing data about the nature of all these stem and progenitor cell types present in the carotid body germinal niche, discussing their role in physiology and their clinical relevance for the treatment of diverse pathologies.


Asunto(s)
Cuerpo Carotídeo/citología , Células Madre Multipotentes/citología , Linaje de la Célula , Humanos , Nicho de Células Madre
13.
Proc Natl Acad Sci U S A ; 116(16): 8000-8009, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30926666

RESUMEN

Neural stem cells continuously generate newborn neurons that integrate into and modify neural circuitry in the adult hippocampus. The molecular mechanisms that regulate or perturb neural stem cell proliferation and differentiation, however, remain poorly understood. Here, we have found that mouse hippocampal radial glia-like (RGL) neural stem cells express the synaptic cochaperone cysteine string protein-α (CSP-α). Remarkably, in CSP-α knockout mice, RGL stem cells lose quiescence postnatally and enter into a high-proliferation regime that increases the production of neural intermediate progenitor cells, thereby exhausting the hippocampal neural stem cell pool. In cell culture, stem cells in hippocampal neurospheres display alterations in proliferation for which hyperactivation of the mechanistic target of rapamycin (mTOR) signaling pathway is the primary cause of neurogenesis deregulation in the absence of CSP-α. In addition, RGL cells lose quiescence upon specific conditional targeting of CSP-α in adult neural stem cells. Our findings demonstrate an unanticipated cell-autonomic and circuit-independent disruption of postnatal neurogenesis in the absence of CSP-α and highlight a direct or indirect CSP-α/mTOR signaling interaction that may underlie molecular mechanisms of brain dysfunction and neurodegeneration.


Asunto(s)
Proteínas del Choque Térmico HSP40 , Proteínas de la Membrana , Células-Madre Neurales/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Cultivadas , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Hipocampo/citología , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Neurogénesis/genética , Lipofuscinosis Ceroideas Neuronales , Transducción de Señal/genética
14.
Cell Mol Life Sci ; 76(6): 1027-1039, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30498994

RESUMEN

Oxygen constitutes a vital element for the survival of every single cell in multicellular aerobic organisms like mammals. A complex homeostatic oxygen-sensing system has evolved in these organisms, including detectors and effectors, to guarantee a proper supply of the element to every cell. The carotid body represents the most important peripheral arterial chemoreceptor organ in mammals and informs about hypoxemic situations to the effectors at the brainstem cardiorespiratory centers. To optimize organismal adaptation to maintained hypoxemic situations, the carotid body has evolved containing a niche of adult tissue-specific stem cells with the capacity to differentiate into both neuronal and vascular cell types in response to hypoxia. These neurogenic and angiogenic processes are finely regulated by the niche and by hypoxia itself. Our recent data on the cellular and molecular mechanisms underlying the functioning of this niche might help to comprehend a variety of different diseases coursing with carotid body failure, and might also improve our capacity to use these stem cells for the treatment of neurological disease. Herein, we review those data about the recent characterization of the carotid body niche, focusing on the study of the phenotype and behavior of multipotent stem cells within the organ, comparing them with other well-documented neural stem cells within the adult nervous system.


Asunto(s)
Células Madre Adultas/fisiología , Cuerpo Carotídeo/fisiología , Células-Madre Neurales/fisiología , Sistema Nervioso Periférico/fisiología , Nicho de Células Madre , Adaptación Fisiológica/fisiología , Adulto , Humanos , Hipoxia , Células Madre Multipotentes/fisiología
15.
EMBO Rep ; 19(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29335248

RESUMEN

Unlike other neural peripheral organs, the adult carotid body (CB) has a remarkable structural plasticity, as it grows during acclimatization to hypoxia. The CB contains neural stem cells that can differentiate into oxygen-sensitive glomus cells. However, an extended view is that, unlike other catecholaminergic cells of the same lineage (sympathetic neurons or chromaffin cells), glomus cells can divide and thus contribute to CB hypertrophy. Here, we show that O2-sensitive mature glomus cells are post-mitotic. However, we describe an unexpected population of pre-differentiated, immature neuroblasts that express catecholaminergic markers and contain voltage-dependent ion channels, but are unresponsive to hypoxia. Neuroblasts are quiescent in normoxic conditions, but rapidly proliferate and differentiate into mature glomus cells during hypoxia. This unprecedented "fast neurogenesis" is stimulated by ATP and acetylcholine released from mature glomus cells. CB neuroblasts, which may have evolved to facilitate acclimatization to hypoxia, could contribute to the CB oversensitivity observed in highly prevalent human diseases.


Asunto(s)
Adaptación Fisiológica/genética , Cuerpo Carotídeo/crecimiento & desarrollo , Diferenciación Celular/genética , Hipoxia , Neurogénesis/genética , Adenosina Trifosfato/metabolismo , Cuerpo Carotídeo/metabolismo , Proliferación Celular/genética , Humanos , Hipoxia/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Oxígeno/metabolismo
16.
Oncotarget ; 8(52): 89775-89792, 2017 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-29163787

RESUMEN

Pediatric tumors arise upon oncogenic transformation of stem/progenitor cells during embryonic development. Given this scenario, the existence of non-tumorigenic stem cells included within the aberrant tumoral niche, with a potential role in tumor biology, is an intriguing and unstudied possibility. Here, we describe the presence and function of non-tumorigenic neural crest-derived progenitor cells in aggressive neuroblastoma (NB) tumors. These cells differentiate into neural crest typical mesectodermal derivatives, giving rise to tumor stroma and promoting proliferation and tumor aggressiveness. Furthermore, an analysis of gene expression profiles in stage 4/M NB revealed a neural crest stem cell (NCSC) gene signature that was associated to stromal phenotype and high probability of relapse. Thus, this NCSC gene expression signature could be used in prognosis to improve stratification of stage 4/M NB tumors. Our results might facilitate the design of new therapies by targeting NCSCs and their contribution to tumor stroma.

17.
Cell Rep ; 19(3): 471-478, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28423311

RESUMEN

Adult stem cell plasticity, or the ability of somatic stem cells to cross boundaries and differentiate into unrelated cell types, has been a matter of debate in the last decade. Neural-crest-derived stem cells (NCSCs) display a remarkable plasticity during development. Whether adult populations of NCSCs retain this plasticity is largely unknown. Herein, we describe that neural-crest-derived adult carotid body stem cells (CBSCs) are able to undergo endothelial differentiation in addition to their reported role in neurogenesis, contributing to both neurogenic and angiogenic processes taking place in the organ during acclimatization to hypoxia. Moreover, CBSC conversion into vascular cell types is hypoxia inducible factor (HIF) dependent and sensitive to hypoxia-released vascular cytokines such as erythropoietin. Our data highlight a remarkable physiological plasticity in an adult population of tissue-specific stem cells and could have impact on the use of these cells for cell therapy.


Asunto(s)
Células Madre Adultas/fisiología , Cuerpo Carotídeo/citología , Mamíferos/metabolismo , Células-Madre Neurales/fisiología , Plasticidad Neuronal , Células Madre Adultas/citología , Células Madre Adultas/efectos de los fármacos , Animales , Vasos Sanguíneos/citología , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Eritropoyetina/farmacología , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones Transgénicos , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos
18.
J Mol Cell Cardiol ; 105: 12-23, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28223221

RESUMEN

Endothelial progenitor cells (EPCs) constitute a promising alternative in cardiovascular regenerative medicine due to their assigned role in angiogenesis and vascular repair. In response to injury, EPCs promote vascular remodeling by replacement of damaged endothelial cells and/or by secreting angiogenic factors over the damaged tissue. Nevertheless, such mechanisms need to be further characterized. In the current approach we have evaluated the initial response of early EPCs (eEPCs) from healthy individuals after direct contact with the factors released by carotid arteries complicated with atherosclerotic plaques (AP), in order to understand the mechanisms underlying the neovascularization and remodeling properties assigned to these cells. Herein, we found that the AP secretome stimulated eEPCs proliferation and mobilization ex vivo, and such increase was accompanied by augmented permeability, cell contraction and also an increase of cell-cell adhesion in association with raised vinculin levels. Furthermore, a comparative mass spectrometry analysis of control versus stimulated eEPCs revealed a differential expression of proteins in the AP treated cells, mostly involved in cell migration, proliferation and vascular remodeling. Some of these protein changes were also detected in the eEPCs isolated from atherosclerotic patients compared to eEPCs from healthy donors. We have shown, for the first time, that the AP released factors activate eEPCs ex vivo by inducing their mobilization together with the expression of vasculogenic related markers. The present approach could be taken as a ex vivo model to study the initial activation of vascular cells in atherosclerosis and also to evaluate strategies looking to potentiate the mobilization of EPCs prior to clinical applications.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Placa Aterosclerótica/metabolismo , Proteoma , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Humanos , Permeabilidad , Placa Aterosclerótica/patología , Proteómica/métodos
19.
Gut ; 66(7): 1306-1320, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-26984852

RESUMEN

OBJECTIVE: Pathological neovascularisation is intimately involved in portal hypertension (PH). Here, we determined the contribution of vascular stem/progenitor cells (VSPCs) to neovessel growth in PH and whether the RNA-binding protein cytoplasmic polyadenylation element binding protein-4 (CPEB4) was behind the mechanism controlling VSPC function. DESIGN: To identify and monitor VSPCs in PH rats (portal vein-ligated), we used a combinatorial approach, including sphere-forming assay, assessment of self-renewal, 5-bromo-2'-desoxyuridine label retention technique, in vitro and in vivo stem/progenitor cell (SPC) differentiation and vasculogenic capability, cell sorting, as well as immunohistochemistry, immunofluorescence and confocal microscopy expression analysis. We also determined the role of CPEB4 on VSPC proliferation using genetically engineered mouse models. RESULTS: We demonstrated the existence in the mesenteric vascular bed of VSPCs displaying capability to form cellular spheres in suspension culture, self-renewal ability, expression of molecules commonly found in SPCs, slow-cycling features, in addition to other cardinal properties exhibited by SPCs, like capacity to differentiate into endothelial cells and pericytes with remarkable vasculogenic activity. Such VSPCs showed, after PH induction, an early switch in proliferation, and differentiated in vivo into endothelial cells and pericytes, contributing, structurally and functionally, to abnormal neovessel formation. Quantification of VSPC-dependent neovessel formation in PH further illustrated the key role played by VSPCs. We also demonstrated that CPEB4 regulates the proliferation of the activated VSPC progeny upon PH induction. CONCLUSIONS: These findings demonstrate that VSPC-derived neovessel growth (ie, vasculogenesis) and angiogenesis cooperatively stimulate mesenteric neovascularisation in PH and identify VSPC and CPEB4 as potential therapeutic targets.


Asunto(s)
Hipertensión Portal/patología , Neovascularización Patológica , Proteínas de Unión al ARN/metabolismo , Células Madre/citología , Animales , Diferenciación Celular , Proliferación Celular , Ratones , Ratas
20.
Dev Growth Differ ; 58(5): 456-62, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27101323

RESUMEN

The discovery of neural stem cells has revealed a much higher structural and functional plasticity in the adult nervous system than previously anticipated. Progenitor cells are able to give rise to new neurons and glial cells when needed, thanks to their surveillance of the environment from the germinal niches. Multiple different factors define neural stem cell niches, including cellular and non-cellular components. Innervation of neurogenic centers is crucial, as it allows the functional connection between stem cell behavior and surrounding neuronal activity. Although the association between organismal behavior and neurogenesis is well documented, much less is known about the cellular and molecular mechanisms by which neurons control stem cell activity. In this review we discuss the existing data on this type of regulation from the three best characterized germinal niches in the adult nervous system: the subventricular zone, the hippocampal subgranular zone, and the carotid body. In all cases, neuronal activity modulates stem cell behavior either by neurotransmitter spillover or by synaptic-like contacts. Currently, the molecular mechanisms underlying mature neuron-stem cell interaction are being clarified. Functional consequences and potential clinical relevance of these phenomena are also discussed.


Asunto(s)
Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Transducción de Señal/fisiología , Nicho de Células Madre/fisiología , Animales , Humanos , Células-Madre Neurales/citología , Neuronas/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...