Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Glia ; 72(4): 728-747, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38180164

RESUMEN

Senescence is a negative prognostic factor for outcome and recovery following traumatic brain injury (TBI). TBI-induced white matter injury may be partially due to oligodendrocyte demise. We hypothesized that the regenerative capacity of oligodendrocyte precursor cells (OPCs) declines with age. To test this hypothesis, the regenerative capability of OPCs in young [(10 weeks ±2 (SD)] and aged [(62 weeks ±10 (SD)] mice was studied in mice subjected to central fluid percussion injury (cFPI), a TBI model causing widespread white matter injury. Proliferating OPCs were assessed by immunohistochemistry for the proliferating cell nuclear antigen (PCNA) marker and labeled by 5-ethynyl-2'-deoxyuridine (EdU) administered daily through intraperitoneal injections (50 mg/kg) from day 2 to day 6 after cFPI. Proliferating OPCs were quantified in the corpus callosum and external capsule on day 2 and 7 post-injury (dpi). The number of PCNA/Olig2-positive and EdU/Olig2-positive cells were increased at 2dpi (p < .01) and 7dpi (p < .01), respectively, in young mice subjected to cFPI, changes not observed in aged mice. Proliferating Olig2+/Nestin+ cells were less common (p < .05) in the white matter of brain-injured aged mice, without difference in proliferating Olig2+/PDGFRα+ cells, indicating a diminished proliferation of progenitors with different spatial origin. Following TBI, co-staining for EdU/CC1/Olig2 revealed a reduced number of newly generated mature oligodendrocytes in the white matter of aged mice when compared to the young, brain-injured mice (p < .05). We observed an age-related decline of oligodendrogenesis following experimental TBI that may contribute to the worse outcome of elderly patients following TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Sustancia Blanca , Humanos , Anciano , Ratones , Animales , Antígeno Nuclear de Célula en Proliferación , Encéfalo , Oligodendroglía , Ratones Endogámicos C57BL
2.
FASEB J ; 33(8): 8990-8998, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31039042

RESUMEN

Poststroke recovery requires multiple repair mechanisms, including vascular remodeling and blood-brain barrier (BBB) restoration. Brain pericytes are essential for BBB repair and angiogenesis after stroke, but they also give rise to scar-forming platelet-derived growth factor receptor ß (PDGFR-ß)-expressing cells. However, many of the molecular mechanisms underlying this pericyte response after stroke still remain unknown. Regulator of G-protein signaling 5 (RGS5) has been associated with pericyte detachment from the vascular wall, but whether it regulates pericyte function and vascular stabilization in the chronic phase of stroke is not known. Using RGS5-knockout (KO) mice, we study how loss of RGS5 affects the pericyte response and vascular remodeling in a stroke model at 7 d after ischemia. Loss of RGS5 leads to a shift toward an increase in the number of perivascular pericytes and reduction in the density of parenchymal PDGFR-ß-expressing cells associated with normalized PDGFR-ß activation after stroke. The redistribution of pericytes resulted in higher pericyte coverage, increased vascular density, preservation of vessel lengths, and a significant reduction in vascular leakage in RGS5-KO mice compared with controls. Our study demonstrates RGS5 in pericytes as an important target to enhance vascular remodeling.-Roth, M., Gaceb, A., Enström, A., Padel, T., Genové, G., Özen, I., Paul, G. Regulator of G-protein signaling 5 regulates the shift from perivascular to parenchymal pericytes in the chronic phase after stroke.


Asunto(s)
Pericitos/metabolismo , Proteínas RGS/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Barrera Hematoencefálica , Capilares/metabolismo , Capilares/patología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica , Pericitos/patología , Proteínas RGS/deficiencia , Proteínas RGS/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Accidente Cerebrovascular/patología , Factores de Tiempo
3.
Int J Mol Sci ; 21(2)2020 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-31936248

RESUMEN

Traumatic brain injury (TBI) increases the risk of delayed neurodegenerative processes, including Parkinson's disease (PD). Interleukin-1beta (IL-1ß), a key pro-inflammatory cytokine, may promote secondary injury development after TBI. Conversely, neutralizing IL-1ß was found to improve functional recovery following experimental TBI. However, the mechanisms underlying the behavioral improvements observed by IL-1ß neutralization are still poorly understood. The present study investigated the role of IL-1ß on the microglia response and neuronal changes in the globus pallidus in response to diffuse TBI. Mice were subjected to sham injury or the central fluid percussion injury (cFPI) (a model of traumatic axonal injury), and were randomly administered an IL-1ß neutralizing or a control antibody at 30 min post-injury. The animals were analyzed at 2, 7, or 14 days post-injury. When compared to controls, mice subjected to cFPI TBI had increased microglia activation and dopaminergic innervation in the globus pallidus, and a decreased number of parvalbumin (PV) positive interneurons in the globus pallidus. Neutralization of IL-1ß attenuated the microglia activation, prevented the loss of PV+ interneurons and normalized dopaminergic fiber density in the globus pallidus of brain-injured animals. These findings argue for an important role for neuro-inflammation in the PD-like pathology observed in TBI.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Interleucina-1beta/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Conducta Animal/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/patología , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Globo Pálido/efectos de los fármacos , Globo Pálido/patología , Humanos , Interleucina-1beta/genética , Activación de Macrófagos/efectos de los fármacos , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología
4.
Stroke ; 49(9): 2182-2190, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30354999

RESUMEN

Background and Purpose- In ischemic stroke, breakdown of the blood-brain barrier (BBB) aggravates brain damage. Pericyte detachment contributes to BBB disruption and neurovascular dysfunction, but little is known about its regulation in stroke. Here, we investigated how loss of RGS5 (regulator of G protein signaling 5) in pericytes affects BBB breakdown in stroke and its consequences. Method- We used RGS5 knockout and control mice and applied a permanent middle cerebral occlusion model. We analyzed pericyte numbers, phenotype, and vessel morphology using immunohistochemistry and confocal microscopy. We investigated BBB breakdown by measuring endothelial coverage, tight junctions, and AQP4 (aquaporin 4) in addition to BBB permeability (fluorescent-conjugated dextran extravasation). Tissue hypoxia was assessed with pimonidazole hydrochloride and neuronal death quantified with the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Results- We demonstrate that loss of RGS5 increases pericyte numbers and their endothelial coverage, which is associated with higher capillary density and length, and significantly less BBB damage after stroke. Loss of RGS5 in pericytes results in reduced vascular leakage and preserved tight junctions and AQP4, decreased cerebral hypoxia, and partial neuronal protection in the infarct area. Conclusions- Our findings show that loss of RGS5 affects pericyte-related BBB preservation in stroke and identifies RGS5 as an important target for neurovascular protection.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Endotelio Vascular/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Neuronas/metabolismo , Pericitos/patología , Proteínas RGS/genética , Uniones Estrechas/metabolismo , Animales , Acuaporina 4/metabolismo , Barrera Hematoencefálica/patología , Permeabilidad Capilar , Muerte Celular , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Hipoxia/metabolismo , Hipoxia/patología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Infarto de la Arteria Cerebral Media/patología , Ratones Noqueados , Microscopía Confocal , Neuronas/patología , Accidente Cerebrovascular , Uniones Estrechas/patología
5.
Neurobiol Dis ; 94: 95-105, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27288154

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disease where the degeneration of the nigrostriatal pathway leads to specific motor deficits. There is an unmet medical need for regenerative treatments that stop or reverse disease progression. Several growth factors have been investigated in clinical trials to restore the dopaminergic nigrostriatal pathway damaged in PD. Platelet-derived growth factor-BB (PDGF-BB), a molecule that recruits pericytes to stabilize microvessels, was recently investigated in a phase-1 clinical trial, showing a dose-dependent increase in dopamine transporter binding in the putamen of PD patients. Interestingly, evidence is accumulating that PD is paralleled by microvascular changes, however, whether PDGF-BB modifies pericytes in PD is not known. Using a pericyte reporter mouse strain, we investigate the functional and restorative effect of PDGF-BB in a partial 6-hydroxydopamine medial forebrain bundle lesion mouse model of PD, and whether this restorative effect is accompanied by changes in pericyte features. We demonstrate that a 2-week treatment with PDGF-BB leads to behavioural recovery using several behavioural tests, and partially restores the nigrostriatal pathway. Interestingly, we find that pericytes are activated in the striatum of PD lesioned mice and that these changes are reversed by PDGF-BB treatment. The modulation of brain pericytes may contribute to the PDGF-BB-induced neurorestorative effects, PDGF-BB allowing for vascular stabilization in PD. Pericytes might be a new cell target of interest for future regenerative therapies.


Asunto(s)
Actividad Motora/efectos de los fármacos , Enfermedad de Parkinson/metabolismo , Pericitos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/farmacología , Animales , Becaplermina , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Masculino , Haz Prosencefálico Medial/efectos de los fármacos , Haz Prosencefálico Medial/metabolismo , Ratones Transgénicos , Actividad Motora/fisiología , Oxidopamina/farmacología , Enfermedad de Parkinson/patología , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo
6.
Acta Neuropathol ; 128(3): 381-96, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24848101

RESUMEN

Pericytes are located on the abluminal side of endothelial cells lining the microvasculature in all organs. They have been identified as multipotent progenitor cells in several tissues of the body including the human brain. New evidence suggests that pericytes contribute to tissue repair, but their role in the injured brain is largely unknown. Here, we investigate the role of pericytes in ischemic stroke. Using a pericyte-reporter mouse model, we provide unique evidence that regulator of G-protein signaling 5 expressing cells are activated pericytes that leave the blood vessel wall, proliferate and give rise to microglial cells after ischemic brain injury. Consistently, we show that activated pericytes express microglial markers in human stroke brain tissue. We demonstrate that human brain-derived pericytes adopt a microglial phenotype and upregulate mRNA specific for activated microglial cells under hypoxic conditions in vitro. Our study indicates that the vasculature is a novel source of inflammatory cells with a microglial phenotype in brain ischemia and hence identifies pericytes as an important new target for the development of future stroke therapies.


Asunto(s)
Encéfalo/patología , Microglía/patología , Pericitos/patología , Accidente Cerebrovascular/patología , Animales , Antígenos CD/metabolismo , Proteínas de Arabidopsis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Glucosa/deficiencia , Humanos , Transferasas Intramoleculares/metabolismo , Antígeno Ki-67/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Factores de Tiempo
7.
Front Neurol ; 15: 1378203, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38765267

RESUMEN

Introduction: Traumatic brain injury to thalamo-cortical pathways is associated with posttraumatic morbidity. Diffuse mechanical forces to white matter tracts and deep grey matter regions induce an inflammatory response and vascular damage resulting in progressive neurodegeneration. Pro-inflammatory cytokines, including interleukin-1ß (IL-1ß), may contribute to the link between inflammation and the injured capillary network after TBI. This study investigates whether IL-1ß is a key contributor to capillary alterations and changes in pericyte coverage in the thalamus and cortex after TBI. Methods: Animals were subjected to central fluid percussion injury (cFPI), a model of TBI causing widespread axonal and vascular pathology, or sham injury and randomized to receive a neutralizing anti-IL-1ß or a control, anti-cyclosporin A antibody, at 30 min post-injury. Capillary length and pericyte coverage of cortex and thalamus were analyzed by immunohistochemistry at 2- and 7-days post-injury. Results and Conclusion: Our results show that early post-injury attenuation of IL-1ß dependent inflammatory signaling prevents capillary damage by increasing pericyte coverage in the thalamus.

8.
Leuk Res ; 140: 107484, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38520796

RESUMEN

INTRODUCTION: Thrombocytopenia is among the most common complications following hematopoietic stem cell transplantation and is associated with increased mortality and morbidity with no standard treatment yet. In this multicenter and retrospective study, we aim to present our multi-center experience of Eltrombopag treatment in patients with isolated thrombocytopenia following HSCT. MATERIAL-METHOD: A total of 73 patients from 5 centers who underwent autologous or allogeneic stem cell transplantation, had no primary disease relapse, all of whom had neutrophil engraftment, complete chimerism, and who were diagnosed with Prolonged Isolated Thrombocytopenia (PIT) or Secondary Failure Of Platelet Recovery (SFPR) were included in the study. The patients were initiated on Eltrombopag at a dose of 50-150 mg. Complete response was defined as a platelet count >50×109/L for 7 consecutive days with no transfusion support. RESULTS: A total of 50.3% of the patients underwent Autologous and 49.7% Allogeneic Stem Cell Transplantation, 54.8% were diagnosed with PIT, and 45.2% were diagnosed with SFPR, and the treatment with 50-150 mg/day Eltrombopag was initiated on the median day +42. Complete response was achieved in 71.2% of these patients on the median day 23 of the treatment. No significant effects of the initial dose (50-150 mg/day) were detected in the Complete Response in the multivariate analysis on response. An insufficient number of Megakaryocytes in the bone marrow before Eltrombopag treatment was determined as an independent risk factor in determining the response (OR 3.57, 95% CI 1.21-10.55). The overall survival of the patients who did not respond to Eltrombopag was found to be significantly worse than that of patients who responded (p=0.022, HR:2.74, 95% CI 1.12-6.54). CONCLUSION: As a result of the present study, Eltrombopag treatment was found to be effective and safe in thrombocytopenia that develops following hematopoietic stem cell transplantation. It was concluded that its use may be more effective in patients with sufficient bone marrow megakaryocytes before the treatment and an initial dose of 50 mg/day may be appropriate in terms of cost, effectiveness, and toxicity. Large-scale randomized and controlled prospective studies are needed to determine the roles of Eltrombopag treatment in patients with post-transplant PIT and SFPR.


Asunto(s)
Benzoatos , Trasplante de Células Madre Hematopoyéticas , Hidrazinas , Pirazoles , Trombocitopenia , Humanos , Hidrazinas/uso terapéutico , Hidrazinas/administración & dosificación , Hidrazinas/efectos adversos , Benzoatos/uso terapéutico , Benzoatos/administración & dosificación , Benzoatos/efectos adversos , Pirazoles/uso terapéutico , Pirazoles/efectos adversos , Pirazoles/administración & dosificación , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Femenino , Masculino , Trombocitopenia/etiología , Trombocitopenia/tratamiento farmacológico , Adulto , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven , Adolescente , Anciano , Recuento de Plaquetas
9.
Biol Open ; 11(10)2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36111549

RESUMEN

Adaptive biological mechanisms to hypoxia are crucial to maintain oxygen homeostasis, especially in the brain. Pericytes, cells uniquely positioned at the blood-brain interface, respond fast to hypoxia by expressing regulator of G-protein signalling 5 (RGS5), a negative regulator of G-protein-coupled receptors. RGS5 expression in pericytes is observed in pathological hypoxic environments (e.g. tumours and ischaemic stroke) and associated with perivascular depletion of pericytes and vessel leakage. However, the regulation of RGS5 expression and its functional role in pericytes are not known. We demonstrate that RGS5 acts as a hypoxia-responsive protein in human brain pericytes that is regulated independent of hypoxia inducible factor-1α (HIF-1α), rapidly stabilized under hypoxia, but degraded under normoxic conditions. We show that RGS5 expression desensitizes pericytes to signalling of platelet-derived growth factor-BB (PDGFBB) and sphingosine 1-phosphate (S1P), and blocks chemokinesis or chemotaxis induced by these factors. Our data imply a role for RGS5 in antagonizing pericyte recruitment and retention to blood vessels during hypoxia and support RGS5 as a target in counteracting vessel leakage under pathological hypoxic conditions. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Isquemia Encefálica , Pericitos , Proteínas RGS , Accidente Cerebrovascular , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Proteínas de Unión al GTP/metabolismo , Humanos , Hipoxia/metabolismo , Oxígeno , Pericitos/metabolismo , Pericitos/patología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Accidente Cerebrovascular/metabolismo
10.
J Neurotrauma ; 39(5-6): 411-422, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35018831

RESUMEN

Cerebellar dysfunction after traumatic brain injury (TBI) is commonly suspected based on clinical symptoms, although cerebellar pathology has rarely been investigated. To address the hypothesis that the cerebellar axon-myelin unit is altered by diffuse TBI, we used the central fluid percussion injury (cFPI) model in adult mice to create widespread axonal injury by delivering the impact to the forebrain. We specifically focused on changes in myelin components (myelin basic protein [MBP], 2',3'-cyclic nucleotide 3'-phosphodiesterase [CNPase], nodal/paranodal domains [neurofascin (Nfasc), ankyrin-G], and phosphorylated neurofilaments [SMI-31, SMI-312]) in the cerebellum, remote from the impact, at two, seven, and 30 days post-injury (dpi). When compared with sham-injured controls, cerebellar MBP and CNPase protein levels were decreased at 2 dpi that remained reduced up to 30 dpi. Diffuse TBI induced different effects on neuronal (Nfasc 186, Nfasc 140) and glial (Nfasc 155) neurofascin isoforms that play a key role in the assembly of the nodes of Ranvier. Expression of Nfasc 140 in the cerebellum increased at 7 dpi, in contrast to Nfasc 155 levels, which were decreased. Although neurofascin binding partner ankyrin-G protein levels decreased acutely after cFPI, its expression levels increased at 7 dpi and remained unchanged up to 30 dpi. The TBI-induced reduction in neurofilament phosphorylation (SMI-31) observed in the cerebellum was closely associated with decreased levels of the myelin proteins MBP and CNPase. This is the first evidence of temporal and spatial structural changes in the axon-myelin unit in the cerebellum, remote from the location of the impact site, in a diffuse TBI model in mice.


Asunto(s)
Traumatismos Difusos del Encéfalo , Lesiones Traumáticas del Encéfalo , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Animales , Ancirinas/metabolismo , Axones/patología , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Cerebelo/metabolismo , Ratones , Vaina de Mielina/metabolismo
11.
Acta Neuropathol Commun ; 10(1): 129, 2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36064443

RESUMEN

Cerebellar dysfunction is commonly observed following traumatic brain injury (TBI). While direct impact to the cerebellum by TBI is rare, cerebellar pathology may be caused by indirect injury via cortico-cerebellar pathways. To address the hypothesis that degeneration of Purkinje cells (PCs), which constitute the sole output from the cerebellum, is linked to long-range axonal injury and demyelination, we used the central fluid percussion injury (cFPI) model of widespread traumatic axonal injury in mice. Compared to controls, TBI resulted in early PC loss accompanied by alterations in the size of pinceau synapses and levels of non-phosphorylated neurofilament in PCs. A combination of vDISCO tissue clearing technique and immunohistochemistry for vesicular glutamate transporter type 2 show that diffuse TBI decreased mossy and climbing fiber synapses on PCs. At 2 days post-injury, numerous axonal varicosities were found in the cerebellum supported by fractional anisotropy measurements using 9.4 T MRI. The disruption and demyelination of the cortico-cerebellar circuits was associated with poor performance of brain-injured mice in the beam-walk test. Despite a lack of direct input from the injury site to the cerebellum, these findings argue for novel long-range mechanisms causing Purkinje cell injury that likely contribute to cerebellar dysfunction after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Enfermedades Desmielinizantes , Animales , Axones/patología , Lesiones Traumáticas del Encéfalo/patología , Cerebelo/patología , Enfermedades Desmielinizantes/patología , Ratones , Células de Purkinje/patología
12.
Brain Inj ; 25(1): 113-26, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21117911

RESUMEN

PRIMARY OBJECTIVE: The present study was undertaken to evaluate whether enoant, which is rich in polyphenols, has any effect on electroencephalogram (EEG), oxidative stress and inflammation in ischemia/reperfusion (I/R) injury. METHODS: Ischemia was induced by 2-hour occlusion of bilateral common carotid artery. Animals orally received enoant. Group 1 was the ischemic control group. Group 2 was treated with enoant of 1.25 g kg⁻¹ per day for 15 days after I/R. Group 3 received the same concentration of enoant as in group 2 for 15 days before and after I/R. Group 4 was the sham operation group. EEG activities were recorded and the levels of TNF-α, IL-1ß and IL-6, TBARS and GSH were measured in the whole brain homogenate. RESULTS: There were significant changes in EEG activity in groups treated with enoant either before or after ischemia when compared with their basal EEG values. TNF-α, IL-6 and IL-1ß levels were significantly increased after I/R. GSH levels in group 3 treated with enoant in both pre- and post-ischemic periods were significantly increased and TBARS concentration was decreased compared with the ischemic group. CONCLUSION: The findings support that both pre-ischemic and post-ischemic administrations of enoant might produce neuroprotective action against cerebral ischemia.


Asunto(s)
Antioxidantes/farmacología , Bebidas , Isquemia Encefálica/tratamiento farmacológico , Electroencefalografía/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Vitis/química , Animales , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/prevención & control , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar , Reperfusión , Daño por Reperfusión/fisiopatología
13.
Cell Death Dis ; 11(2): 98, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029735

RESUMEN

Amyloid beta peptides (Aß) proteins play a key role in vascular pathology in Alzheimer's Disease (AD) including impairment of the blood-brain barrier and aberrant angiogenesis. Although previous work has demonstrated a pro-angiogenic role of Aß, the exact mechanisms by which amyloid precursor protein (APP) processing and endothelial angiogenic signalling cascades interact in AD remain a largely unsolved problem. Here, we report that increased endothelial sprouting in human-APP transgenic mouse (TgCRND8) tissue is dependent on ß-secretase (BACE1) processing of APP. Higher levels of Aß processing in TgCRND8 tissue coincides with decreased NOTCH3/JAG1 signalling, overproduction of endothelial filopodia and increased numbers of vascular pericytes. Using a novel in vitro approach to study sprouting angiogenesis in TgCRND8 organotypic brain slice cultures (OBSCs), we find that BACE1 inhibition normalises excessive endothelial filopodia formation and restores NOTCH3 signalling. These data present the first evidence for the potential of BACE1 inhibition as an effective therapeutic target for aberrant angiogenesis in AD.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Corteza Cerebral/irrigación sanguínea , Células Endoteliales/enzimología , Neovascularización Patológica , Receptor Notch3/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Inhibidores Enzimáticos/farmacología , Femenino , Técnicas In Vitro , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Densidad Microvascular , Seudópodos/enzimología , Seudópodos/patología , Transducción de Señal
14.
Epilepsy Behav ; 15(2): 142-5, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19318134

RESUMEN

Animals treated with scopolamine after fasting develop convulsions after they are allowed to eat ad libitum. This study was aimed at investigating the effect on these convulsions of liquid food intake, feeding by gavage, and placebo. Fasted mice treated with saline or scopolamine were allowed to eat solid food, slurry food or liquid food ad libitum, given placebo, or given liquid food by gavage. After 30 min, all animals were allowed to eat food pellets and observed for 30 min for the incidence and onset of convulsions. Scopolamine treatment caused convulsions only in the animals given solid food in the first 30 min; no convulsions were observed in the animals given slurry food, liquid food ad libitum, gavage, or placebo. When the animals that did not develop convulsions during the experiment were allowed to eat solid food, convulsions occurred. These findings indicate that complex mechanisms trigger scopolamine-induced convulsions in fasted animals eating solid food.


Asunto(s)
Ingestión de Alimentos/fisiología , Privación de Alimentos/fisiología , Antagonistas Muscarínicos , Escopolamina , Convulsiones/inducido químicamente , Convulsiones/fisiopatología , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C
15.
Biochimie ; 155: 16-25, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29698670

RESUMEN

Personalized and regenerative medicine is an emerging therapeutic strategy that is based on cell biology and biomedical engineering used to develop biological substitutes to maintain normal function or restore damaged tissues and organs. The secretory capacities of different cell types are now explored as such possible therapeutic regenerative agents in a variety of diseases. A secretome can comprise chemokines, cytokines, growth factors, but also extracellular matrix components, microvesicles and exosomes as well as genetic material and may differ depending on the tissue and the stimulus applied to the cell. With regard to clinical applications, the secretome of mesenchymal stem cells (MSC) is currently the most widely explored. However, other cell types such as pericytes may have similar properties as MSC and the potential therapeutic possibilities of these cells are only just beginning to emerge. In this review, we will summarize the currently available data describing the secretome of pericytes and its potential implications for tissue regeneration, whereby we especially focus on brain pericytes as potential new target cell for neuroregeneration and brain repair.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Citocinas/metabolismo , Pericitos/metabolismo , Proteoma/metabolismo , Regeneración , Animales , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Pericitos/citología , Medicina Regenerativa/métodos
16.
J Cereb Blood Flow Metab ; 38(1): 45-57, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28741407

RESUMEN

Brain pericytes not only maintain the anatomical, biochemical and immune blood-brain barrier, but display features of mesenchymal stem cells (MSCs) in vitro. MSCs have pro-regenerative properties attributed to their secretome. However, whether also brain pericytes possess such pro-regenerative capacities is largely unknown. Here we characterize the secretome and microvesicle (MV) release of human brain pericytes mediated by platelet-derived growth factor-BB (PDGF-BB)/PDGF receptor beta (PDGFRß) signalling. Upon PDGF-BB, pericytes release not only a plethora of growth factors and a panel of cytokines, but also MVs containing BDNF, FGFb, ßNGF, VEGF and PLGF, a response that is specific for PDGFRß signalling and activation of the ERK 1/2 pathway. In contrast, lipopolysaccharide (LPS), an activator of the innate immune system, stimulates the secretion of much higher amounts of mainly inflammatory cytokines and activates the NFκB pathway. Pericytes change their morphology and undergo opposite changes in surface marker expression, respectively. Our findings provide evidence that the secretome of human brain pericytes varies greatly depending on the exogenous stimulus. The differential secretory functions of pericytes may play an important role in either regulating neuroinflammation or contributing to neurorestoration and identify a possible new target cell for neuroregeneration.


Asunto(s)
Becaplermina/metabolismo , Encéfalo/metabolismo , Pericitos/metabolismo , Becaplermina/farmacología , Micropartículas Derivadas de Células/metabolismo , Citocinas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pericitos/efectos de los fármacos
17.
PLoS One ; 13(3): e0194146, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29518129

RESUMEN

Brain pericytes are important to maintain vascular integrity of the neurovascular unit under both physiological and ischemic conditions. Ischemic stroke is known to induce an inflammatory and hypoxic response due to the lack of oxygen and glucose in the brain tissue. How this early response to ischemia is molecularly regulated in pericytes is largely unknown and may be of importance for future therapeutic targets. Here we evaluate the transcriptional responses in in vitro cultured human brain pericytes after oxygen and/or glucose deprivation. Hypoxia has been widely known to stabilise the transcription factor hypoxia inducible factor 1-alpha (HIF1α) and mediate the induction of hypoxic transcriptional programs after ischemia. However, we find that the transcription factors Jun Proto-Oncogene (c-JUN), Nuclear Factor Of Kappa Light Polypeptide Gene Enhancer In B-Cells (NFκB) and signal transducer and activator of transcription 3 (STAT3) bind genes regulated after 2hours (hs) of omitted glucose and oxygen before HIF1α. Potent HIF1α responses require 6hs of hypoxia to substantiate transcriptional regulation comparable to either c-JUN or STAT3. Phosphorylated STAT3 protein is at its highest after 5 min of oxygen and glucose (OGD) deprivation, whereas maximum HIF1α stabilisation requires 120 min. We show that STAT3 regulates angiogenic and metabolic pathways before HIF1α, suggesting that HIF1α is not the initiating trans-acting factor in the response of pericytes to ischemia.


Asunto(s)
Encéfalo/metabolismo , Glucosa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/metabolismo , Pericitos/metabolismo , Factor de Transcripción STAT3/metabolismo , Transcripción Genética , Encéfalo/patología , Hipoxia de la Célula , Humanos , Pericitos/patología , Proto-Oncogenes Mas
19.
Seizure ; 15(7): 509-19, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16890459

RESUMEN

The present study was conducted to evaluate scopolamine-induced convulsions in fasted mice after food intake effects on the cortical electroencephalogram (EEG). Continuous EEG recordings were taken with Neuroscan for 10 min in freely moving mice with six chronic cortical electrode implants. Animals were weighed and deprived of food for 48 h. EEG recordings were taken at the 24th and 48th hour after their food deprivations. Later, all animals were treated with saline or scopolamine of 3mg/kg i.p. and EEG recordings were repeated for 10 min. Twenty minutes later, they were given food pellets and were allowed to eat ad libitum. All animals were observed for 60 min to determine the incidence and onset of convulsions and EEG recordings were taken simultaneously. The present results demonstrate that food deprivation causes differences in EEG in the elapsed time. The changes in EEG induced after food deprivation become different with scopolamine administration. In scopolamine treatment group, eating caused a series of high-voltage polyspikes and synchronized spikes with a predominant frequency in the 1-3 Hz range and fast activity that represents a typical epileptiform manifestation. It was concluded that the EEG properties and the behavioral patterns of these convulsions are in accordance with each other.


Asunto(s)
Electroencefalografía , Ayuno/efectos adversos , Antagonistas Muscarínicos/farmacología , Escopolamina/farmacología , Convulsiones/inducido químicamente , Animales , Electrodos Implantados , Electroencefalografía/efectos de los fármacos , Ayuno/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Convulsiones/fisiopatología
20.
PLoS One ; 10(4): e0123553, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875288

RESUMEN

Glioblastoma multiforme (GBM) is the most common brain tumor in adults. It presents an extremely challenging clinical problem, and treatment very frequently fails due to the infiltrative growth, facilitated by extensive angiogenesis and neovascularization. Pericytes constitute an important part of the GBM microvasculature. The contribution of endogenous brain pericytes to the tumor vasculature in GBM is, however, unclear. In this study, we determine the site of activation and the extent of contribution of endogenous brain pericytes to the GBM vasculature. GL261 mouse glioma was orthotopically implanted in mice expressing green fluorescent protein (GFP) under the pericyte marker regulator of G protein signaling 5 (RGS5). Host pericytes were not only activated within the glioma, but also in cortical areas overlying the tumor, the ipsilateral subventricular zone and within the hemisphere contralateral to the tumor. The host-derived activated pericytes that infiltrated the glioma were mainly localized to the tumor vessel wall. Infiltrating GFP positive pericytes co-expressed the pericyte markers platelet-derived growth factor receptor-ß (PDGFR-ß) and neuron-glial antigen 2. Interestingly, more than half of all PDGFR-ß positive pericytes within the tumor were contributed by the host brain. We did not find any evidence that RGS5 positive pericytes adopt another phenotype within glioma in this paradigm. We conclude that endogenous pericytes become activated in widespread areas of the brain in response to an orthotopic mouse glioma. Host pericytes are recruited into the tumor and constitute a major part of the tumor pericyte population.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Encéfalo/patología , Glioma/irrigación sanguínea , Microvasos/patología , Neovascularización Patológica/patología , Pericitos/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Adhesión Celular , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Femenino , Glioma/patología , Proteínas Fluorescentes Verdes/metabolismo , Inflamación/patología , Laminina/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Pericitos/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA