Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Inflamm Res ; 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965133

RESUMEN

OBJECTIVE: We aimed to broaden our understanding of a potential interaction between B1R and TLR4, considering earlier studies suggesting that lipopolysaccharide (LPS) may trigger B1R stimulation. METHODS: We assessed the impact of DBK and LPS on the membrane potential of thoracic aortas from C57BL/6, B1R, or TLR4 knockout mice. Additionally, we examined the staining patterns of these receptors in the thoracic aortas of C57BL/6 and in endothelial cells (HBMEC). RESULTS: DBK does not affect the resting membrane potential of aortic rings in C57BL/6 mice, but it hyperpolarizes preparations in B1KO and TLR4KO mice. The hyperpolarization mechanism in B1KO mice involves B2R, and the TLR4KO response is independent of cytoplasmic calcium influx but relies on potassium channels. Conversely, LPS hyperpolarizes thoracic aorta rings in both C57BL/6 and B1KO mice, with the response unaffected by a B1R antagonist. Interestingly, the absence of B1R alters the LPS response to potassium channels. These activities are independent of nitric oxide synthase (NOS). While exposure to DBK and LPS does not alter B1R and TLR4 mRNA expression, treatment with these agonists increases B1R staining in endothelial cells of thoracic aortic rings and modifies the staining pattern of B1R and TLR4 in endothelial cells. Proximity ligation assay suggests a interaction between the receptors. CONCLUSION: Our findings provide additional support for a putative connection between B1R and TLR4 signaling. Given the involvement of these receptors and their agonists in inflammation, it suggests that drugs and therapies targeting their effects could be promising therapeutic avenues worth exploring.

2.
Glia ; 69(6): 1429-1443, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33497496

RESUMEN

Central nervous system (CNS) function depends on precise synaptogenesis, which is shaped by environmental cues and cellular interactions. Astrocytes are outstanding regulators of synapse development and plasticity through contact-dependent signals and through the release of pro- and antisynaptogenic factors. Conversely, myelin and its associated proteins, including Nogo-A, affect synapses in a inhibitory fashion and contribute to neural circuitry stabilization. However, the roles of Nogo-A-astrocyte interactions and their implications in synapse development and plasticity have not been characterized. Therefore, we aimed to investigate whether Nogo-A affects the capacity of astrocytes to induce synaptogenesis. Additionally, we assessed whether downregulation of Nogo-A signaling in an in vivo demyelination model impacts the synaptogenic potential of astrocytes. Our in vitro data show that cortical astrocytes respond to Nogo-A through RhoA pathway activation, exhibiting stress fiber formation and decreased ramified morphology. This phenotype was associated with reduced levels of GLAST protein and aspartate uptake, decreased mRNA levels of the synaptogenesis-associated genes Hevin, glypican-4, TGF-ß1 and BDNF, and decreased and increased protein levels of Hevin and SPARC, respectively. Corroborating these findings, conditioned medium from Nogo-A-treated astrocytes suppressed the formation of structurally and functionally mature synapses in cortical neuronal cultures. After cuprizone-induced acute demyelination, we observed reduced immunostaining for Nogo-A in the visual cortex accompanied by higher levels of Hevin expression in astrocytes and an increase in excitatory synapse density. Hence, we suggest that interactions between Nogo-A and astrocytes might represent an important pathway of plasticity regulation and could be a target for therapeutic intervention in demyelinating diseases in the future.


Asunto(s)
Astrocitos , Enfermedades Desmielinizantes , Humanos , Neurogénesis , Proteínas Nogo , Sinapsis
3.
Microvasc Res ; 131: 104024, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32502488

RESUMEN

Congenital toxoplasmosis is a parasitic disease that occurs due vertical transmission of the protozoan Toxoplasma gondii (T. gondii) during pregnancy. The parasite crosses the placental barrier and reaches the developing brain, infecting progenitor, glial, neuronal and vascular cell types. Although the role of Radial glia (RG) neural stem cells in the development of the brain vasculature has been recently investigated, the impact of T. gondii infection in these events is not yet understood. Herein, we studied the role of T. gondii infection on RG cell function and its interaction with endothelial cells. By infecting isolated RG cultures with T. gondii tachyzoites, we observed a cytotoxic effect with reduced numbers of RG populations together with decrease neuronal and oligodendrocyte progenitor populations. Conditioned medium (CM) from RG control cultures increased ZO-1 protein levels and organization on endothelial bEnd.3 cells membranes, which was impaired by CM from infected RG, accompanied by decreased trans-endothelial electrical resistance (TEER). ELISA assays revealed reduced levels of anti-inflammatory cytokine TGF-ß1 in CM from T. gondii-infected RG cells. Treatment with recombinant TGF-ß1 concomitantly with CM from infected RG cultures led to restoration of ZO-1 staining in bEnd.3 cells. Congenital infection in Swiss Webster mice led to abnormalities in the cortical microvasculature in comparison to uninfected embryos. Our results suggest that infection of RG cells by T. gondii negatively modulates cytokine secretion, which might contribute to endothelial loss of barrier properties, thus leading to impairment of neurovascular interaction establishment.


Asunto(s)
Diferenciación Celular , Corteza Cerebral/irrigación sanguínea , Células Endoteliales/parasitología , Células Ependimogliales/parasitología , Microvasos/parasitología , Acoplamiento Neurovascular , Toxoplasma/patogenicidad , Toxoplasmosis Cerebral/parasitología , Toxoplasmosis Congénita/parasitología , Animales , Línea Celular , Modelos Animales de Enfermedad , Impedancia Eléctrica , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Ratones Endogámicos C57BL , Microvasos/metabolismo , Microvasos/patología , Uniones Estrechas/metabolismo , Uniones Estrechas/parasitología , Uniones Estrechas/patología , Toxoplasmosis Cerebral/metabolismo , Toxoplasmosis Cerebral/patología , Toxoplasmosis Congénita/metabolismo , Toxoplasmosis Congénita/patología , Factor de Crecimiento Transformador beta1/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
4.
Am J Pathol ; 188(11): 2674-2687, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30121257

RESUMEN

Toxoplasmosis is one of the leading parasitic diseases worldwide. Some data suggest that chronic acquired toxoplasmosis could be linked to behavioral alterations in humans. The parasite infects neurons, forming immunologically silent cysts. Cerebral microcirculation homeostasis is determinant to brain functions, and pathologic states can alter capillarity or blood perfusion, leading to neurodegeneration and cognitive deficits. Albino mice were infected with Toxoplasma gondii (ME49 strain) and analyzed after 10, 40, and 180 days. Infected mice presented decreased cerebral blood flow at 10 and 40 days post infection (dpi), which were restored at 180 dpi, as shown by laser speckle contrast imaging. Intravital microscopy demonstrated that infection led to significant capillary rarefaction, accompanied by neuroinflammation, with microglial activation and increased numbers of rolling and adherent leukocytes to the wall of cerebral capillaries. Acetylcholine-induced vasodilation was altered at all time points, and blood brain barrier permeability was evident in infected animals at 40 dpi. Infection reduced angiogenesis, with a decreased number of isolectin B4-stained blood vessels and a decrease in length and branching of laminin-stained capillaries. Sulfadiazine reduced parasite load and partially repaired microvascular damages. We conclude that T. gondii latent infection causes a harmful insult in the brain, promoting neuroinflammation and microcirculatory dysfunction in the brain, with decreased angiogenesis and can contribute to a neurodegenerative process.


Asunto(s)
Barrera Hematoencefálica/patología , Endotelio Vascular/patología , Inflamación/patología , Microcirculación , Neuronas/patología , Toxoplasma/patogenicidad , Toxoplasmosis Cerebral/patología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/parasitología , Endotelio Vascular/inmunología , Endotelio Vascular/parasitología , Femenino , Inflamación/inmunología , Inflamación/parasitología , Ratones , Ratones Endogámicos C57BL , Neuronas/inmunología , Neuronas/parasitología , Toxoplasmosis Cerebral/inmunología , Toxoplasmosis Cerebral/parasitología
5.
Infect Immun ; 85(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28784928

RESUMEN

Clostridium difficile, the main cause of diarrhea in hospitalized patients, produces toxins A (TcdA) and B (TcdB), which affect intestinal epithelial cell survival, proliferation, and migration and induce an intense inflammatory response. Transforming growth factor ß (TGF-ß) is a pleiotropic cytokine affecting enterocyte and immune/inflammatory responses. However, it has been shown that exposure of intestinal epithelium to a low concentration of TcdA induces the release of TGF-ß1, which has a protective effect on epithelial resistance and a TcdA/TGF-ß signaling pathway interaction. The activation of this pathway in vivo has not been elucidated. The aim of this study was to investigate the role of the TGF-ß1 pathway in TcdA-induced damage in a rat intestinal epithelial cell line (IEC-6) and in a mouse model of an ileal loop. TcdA increased the expression of TGF-ß1 and its receptor, TßRII, in vitro and in vivo TcdA induced nuclear translocation of the transcription factors SMAD2/3, a hallmark of TGF-ß1 pathway activation, both in IEC cells and in mouse ileal tissue. The addition of recombinant TGF-ß1 (rTGF-ß) prevented TcdA-induced apoptosis/necrosis and restored proliferation and repair activity in IEC-6 cells in the presence of TcdA. Together, these data show that TcdA induces TGF-ß1 signaling pathway activation and suggest that this pathway might play a protective role against the effect of C. difficile-toxin.


Asunto(s)
Toxinas Bacterianas/toxicidad , Enterotoxinas/toxicidad , Mucosa Intestinal/microbiología , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular , Clostridioides difficile/patogenicidad , Enterotoxinas/metabolismo , Íleon/inmunología , Íleon/microbiología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Intestinos/inmunología , Intestinos/microbiología , Ratones , Factor de Crecimiento Transformador beta1/genética
6.
J Biol Chem ; 287(49): 41432-45, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23055518

RESUMEN

Assembly of synapses requires proper coordination between pre- and postsynaptic elements. Identification of cellular and molecular events in synapse formation and maintenance is a key step to understand human perception, learning, memory, and cognition. A key role for astrocytes in synapse formation and function has been proposed. Here, we show that transforming growth factor ß (TGF-ß) signaling is a novel synaptogenic pathway for cortical neurons induced by murine and human astrocytes. By combining gain and loss of function approaches, we show that TGF-ß1 induces the formation of functional synapses in mice. Further, TGF-ß1-induced synaptogenesis involves neuronal activity and secretion of the co-agonist of the NMDA receptor, D-serine. Manipulation of D-serine signaling, by either genetic or pharmacological inhibition, prevented the TGF-ß1 synaptogenic effect. Our data show a novel molecular mechanism that might impact synaptic function and emphasize the evolutionary aspect of the synaptogenic property of astrocytes, thus shedding light on new potential therapeutic targets for synaptic deficit diseases.


Asunto(s)
Astrocitos/citología , Corteza Cerebral/metabolismo , Neuronas/metabolismo , Serina/química , Sinapsis/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células Cultivadas , Cognición , Medios de Cultivo Condicionados/farmacología , Electrofisiología , Humanos , Ratones , Modelos Biológicos , Técnicas de Placa-Clamp , Transducción de Señal , Transfección
7.
Viruses ; 15(3)2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36992454

RESUMEN

Neurological effects of COVID-19 and long-COVID-19, as well as neuroinvasion by SARS-CoV-2, still pose several questions and are of both clinical and scientific relevance. We described the cellular and molecular effects of the human brain microvascular endothelial cells (HBMECs) in vitro exposure by SARS-CoV-2 to understand the underlying mechanisms of viral transmigration through the blood-brain barrier. Despite the low to non-productive viral replication, SARS-CoV-2-exposed cultures displayed increased immunoreactivity for cleaved caspase-3, an indicator of apoptotic cell death, tight junction protein expression, and immunolocalization. Transcriptomic profiling of SARS-CoV-2-challenged cultures revealed endothelial activation via NF-κB non-canonical pathway, including RELB overexpression and mitochondrial dysfunction. Additionally, SARS-CoV-2 led to altered secretion of key angiogenic factors and to significant changes in mitochondrial dynamics, with increased mitofusin-2 expression and increased mitochondrial networks. Endothelial activation and remodeling can further contribute to neuroinflammatory processes and lead to further BBB permeability in COVID-19.


Asunto(s)
COVID-19 , FN-kappa B , Humanos , FN-kappa B/metabolismo , SARS-CoV-2/metabolismo , Células Endoteliales/metabolismo , Síndrome Post Agudo de COVID-19 , COVID-19/metabolismo , Encéfalo , Barrera Hematoencefálica , Mitocondrias/metabolismo
8.
Dev Neurosci ; 34(1): 68-81, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22652705

RESUMEN

The major neural stem cell population in the developing cerebral cortex is the radial glia cells, which generate neurons and glial cells. The mechanisms that modulate the maintenance of the radial glia stem cell phenotype, or its differentiation, are not completely elucidated. We previously demonstrated that transforming growth factor-ß(1) (TGF-ß(1)) promotes radial glia differentiation into astrocytes in vitro [Glia 2007;55:1023-1033]. Here we investigated the intracellular signaling pathways involved in the TGF-ß(1)-induced radial glia fate commitment. We demonstrate that the mechanisms underlying the TGF-ß(1) effect on radial glia cell differentiation or progenitor potential maintenance diverge. Whereas radial glia differentiation into astrocytes is mediated by the activation of the MAPK signaling pathway, neurogenesis is modulated by different levels of PI3K and SMAD2/3 activity. Our work demonstrates that radial glia cells are a heterogeneous population and a potential target of TGF-ß(1), and suggests that its effect on radial glia fate commitment is mediated by the recruitment of a complex multipathway mechanism that controls astrocyte and neuronal generation in the developing cerebral cortex.


Asunto(s)
Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Neuroglía/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Análisis de Varianza , Animales , Astrocitos/citología , Benzamidas/farmacología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Dioxoles/farmacología , Flavonoides/farmacología , Inmunohistoquímica , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuroglía/citología , Neuronas/citología , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
9.
Neurochem Res ; 37(11): 2402-18, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22614925

RESUMEN

Neuron-astroglia interactions play a key role in several events of brain development, such as neuronal generation, migration, survival, and differentiation; axonal growth; and synapse formation and function. While there is compelling evidence of the effects of astrocyte factors on neurons, their effects on astrocytes have not been fully determined. In this review, we will focus on the role of neurons in astrocyte generation and maturation. Further, we highlight the great heterogeneity and diversity of astroglial and neural progenitors such as radial glia cells, and discuss the importance of the variety of cellular interactions in controlling the structural and functional organization of the brain. Finally, we present recent data on a new role of astrocytes in neuronal maturation, as mediators of the action of biolipids in the cerebral cortex. We will argue that the functional architecture of the brain depends on an intimate neuron-glia partnership, by briefly discussing the emerging view of how neuron-astrocyte dysfunctions might be associated with neurodegenerative diseases and neurological disorders.


Asunto(s)
Astrocitos/citología , Sistema Nervioso Central/citología , Neuronas/citología , Animales , Linaje de la Célula , Humanos
10.
Neurotoxicology ; 90: 145-157, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35307464

RESUMEN

Ethanol consumption during pregnancy or lactation permanently impairs the development of the central nervous system (CNS), resulting in the spectrum of fetal alcohol disorders (FASD). FASD is a general term that covers a set of deficits in the embryo caused by gestational alcohol exposure, with fetal alcohol syndrome (FAS) considered the most serious. The clinical features of FAS include facial abnormalities, short stature, low body weight, and evidence of structural and/or functional damage to the central nervous system (CNS). The prevalence of FAS carriers worldwide is about 15 for every 10,000 live births (about 119,000 children with APS born per year). Epidemiological data in the US show that the incidence of FAS exceeds other congenital syndromes such as Down syndrome and spina bifida. The deleterious effects of ethanol appear in different brain regions, varying according to the dose and period of neural development when the embryo was exposed, and include: 1) microcephaly; 2) abnormalities in cortical development, with a significant decrease in gyrification; 3) agenesis or hypoplasia of the corpus callosum; and 4) cognitive and behavioral deficits (such as impaired memory and learning, speech difficulties, and hyperactivity). Current evidence indicates that CNS blood vessels are particularly affected by teratogenic ethanol. The CNS vasculature is composed of specialized endothelial cells that establish intimate interactions with astrocytes, pericytes, and microglia, constituting the neurovascular unit of the blood-brain barrier (BBB). Together with the fact that BBB exert protective function, it can prevent the passage of substances and drugs to treat diseases that affect the CNS. Pathological changes in the BBB, such as drug abuse during pregnancy, congenital infections, or ageing processes can drastically alter the molecular structure and vascular stability, disrupting the BBB and aggravating certain neurodegenerative and neurological diseases. In this review, we address the effects of alcohol exposure on the formation of the BBB, specifically describing the cellular and molecular events induced by ethanol in the physiology of endothelial cells and glial cells, as well as their interaction during CNS development.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal , Consumo de Bebidas Alcohólicas , Barrera Hematoencefálica , Niño , Células Endoteliales , Etanol/toxicidad , Femenino , Humanos , Embarazo
11.
Fluids Barriers CNS ; 19(1): 63, 2022 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-35982454

RESUMEN

COVID-19, which is caused by Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2), has resulted in devastating morbidity and mortality worldwide due to lethal pneumonia and respiratory distress. In addition, the central nervous system (CNS) is well documented to be a target of SARS-CoV-2, and studies detected SARS-CoV-2 in the brain and the cerebrospinal fluid of COVID-19 patients. The blood-brain barrier (BBB) was suggested to be the major route of SARS-CoV-2 infection of the brain. Functionally, the BBB is created by an interactome between endothelial cells, pericytes, astrocytes, microglia, and neurons, which form the neurovascular units (NVU). However, at present, the interactions of SARS-CoV-2 with the NVU and the outcomes of this process are largely unknown. Moreover, age was described as one of the most prominent risk factors for hospitalization and deaths, along with other comorbidities such as diabetes and co-infections. This review will discuss the impact of SARS-CoV-2 on the NVU, the expression profile of SARS-CoV-2 receptors in the different cell types of the CNS and the possible role of aging in the neurological outcomes of COVID-19. A special emphasis will be placed on mitochondrial functions because dysfunctional mitochondria are also a strong inducer of inflammatory reactions and the "cytokine storm" associated with SARS-CoV-2 infection. Finally, we will discuss possible drug therapies to treat neural endothelial function in aged patients, and, thus, alleviate the neurological symptoms associated with COVID-19.


Asunto(s)
COVID-19 , Anciano , Barrera Hematoencefálica , Encéfalo , Células Endoteliales , Humanos , SARS-CoV-2
12.
bioRxiv ; 2022 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-35734080

RESUMEN

Neurological effects of COVID-19 and long-COVID-19 as well as neuroinvasion by SARS-CoV-2 still pose several questions and are of both clinical and scientific relevance. We described the cellular and molecular effects of the human brain microvascular endothelial cells (HBMECs) in vitro infection by SARS-CoV-2 to understand the underlying mechanisms of viral transmigration through the Blood-Brain Barrier. Despite the low to non-productive viral replication, SARS-CoV-2-infected cultures displayed increased apoptotic cell death and tight junction protein expression and immunolocalization. Transcriptomic profiling of infected cultures revealed endothelial activation via NF-κB non-canonical pathway, including RELB overexpression, and mitochondrial dysfunction. Additionally, SARS-CoV-2 led to altered secretion of key angiogenic factors and to significant changes in mitochondrial dynamics, with increased mitofusin-2 expression and increased mitochondrial networks. Endothelial activation and remodeling can further contribute to neuroinflammatory processes and lead to further BBB permeability in COVID-19.

13.
Res Sq ; 2022 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-35734086

RESUMEN

Neurological effects of COVID-19 and long-COVID-19 as well as neuroinvasion by SARS-CoV-2 still pose several questions and are of both clinical and scientific relevance. We described the cellular and molecular effects of the human brain microvascular endothelial cells (HBMECs) in vitro infection by SARS-CoV-2 to understand the underlying mechanisms of viral transmigration through the Blood-Brain Barrier. Despite the low to non- productive viral replication, SARS-CoV-2-infected cultures displayed increased apoptotic cell death and tight junction protein expression and immunolocalization. Transcriptomic profiling of infected cultures revealed endothelial activation via NF-κB non-canonical pathway, including RELB overexpression, and mitochondrial dysfunction. Additionally, SARS-CoV-2 led to altered secretion of key angiogenic factors and to significant changes in mitochondrial dynamics, with increased mitofusin-2 expression and increased mitochondrial networks. Endothelial activation and remodeling can further contribute to neuroinflammatory processes and lead to further BBB permeability in COVID-19.

14.
Mol Neurobiol ; 58(4): 1755-1768, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33387302

RESUMEN

Ethanol consumption during pregnancy or lactation period can induce permanent damage to the development of the central nervous system (CNS), resulting in fetal alcohol spectrum disorders (FASD). CNS development depends on proper neural cells and blood vessel (BV) development and blood-brain barrier (BBB) establishment; however, little is known about how ethanol affects these events. Here, we investigated the impact of ethanol exposure to endothelial cells (ECs) function and to ECs interaction with astrocytes in the context of BBB establishment. Cerebral cortex of newborn mice exposed in utero to ethanol (FASD model) presented a hypervascularized phenotype, revealed by augmented vessel density, length, and branch points. Further, aberrant distribution of the tight junction ZO-1 protein along BVs and increased rates of perivascular astrocytic endfeet around BVs were observed. In vitro exposure of human brain microcapillary ECs (HBMEC) to ethanol significantly disrupted ZO-1 distribution, decreased Claudin-5 and GLUT-1 expression and impaired glucose uptake, and increased nitric oxide secretion. These events were accompanied by upregulation of angiogenesis-related secreted proteins by ECs in response to ethanol exposure. Treatment of cortical astrocytes with conditioned medium (CM) from ethanol exposed ECs, upregulated astrocyte's expression of GFAP, Cx43, and Lipocalin-2 genes, as well as the pro-inflammatory genes, IL-1beta, IL-6, and TNF-alpha, which was accompanied by NF-kappa B protein nuclear accumulation. Our findings suggest that ethanol triggers a dysfunctional phenotype in brain ECs, leading to impairment of cortical vascular network formation, and promotes ECs-induced astrocyte dysfunction, which could dramatically affect BBB establishment in the developing brain.


Asunto(s)
Astrocitos/patología , Vasos Sanguíneos/embriología , Barrera Hematoencefálica/patología , Corteza Cerebral/embriología , Células Endoteliales/patología , Etanol/efectos adversos , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Animales Recién Nacidos , Células Endoteliales/metabolismo , Femenino , Trastornos del Espectro Alcohólico Fetal/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Neovascularización Fisiológica , Fenotipo , Embarazo
15.
J Neurosci Res ; 88(3): 530-41, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19746423

RESUMEN

Neurodegenerative diseases are a major constraint on the social and economic development of many countries. Evidence has suggested that phytochemicals have an impact on brain pathology; however, both their mechanisms of action and their cell targets are incompletely known. Here, we investigated the effects of the flavonoid casticin, extracted from Croton betulaster, a common plant in the state of Bahia in Brazil, on rat cerebral cortex neurons in vitro. Treatment of neural progenitors with 10 microM casticin increased the neuronal population positive for the neuronal marker beta-tubulin III and the neuronal transcriptional factor Tbr2 by approximately 20%. This event was followed by a 50% decrease in neuronal death. Pools of astrocyte (GFAP and S100beta), neural (nestin), and oligodendrocyte (Olig2 and NG2) progenitors were not affected by casticin. Neither neuronal commitment nor proliferation of progenitors was affected by casticin, suggesting a neuroprotective effect of this compound. Culture of neural progenitors on casticin-treated astrocyte monolayers increased the neuronal population by 40%. This effect was reproduced by conditioned medium derived from casticin-treated astrocytes, suggesting the involvement of a soluble factor. ELISA assays of the conditioned medium revealed a 20% increase in interleukin-6 level in response to casticin. In contrast to the direct effect, neuronal death was unaffected, but a 52% decrease in the death of nestin-positive progenitors was observed. Together our data suggest that casticin influences the neuronal population by two mechanisms: 1) directly, by decreasing neuronal death, and 2) indirectly, via astrocytes, by modulating the pool of neuronal progenitors.


Asunto(s)
Astrocitos/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Flavonoides/farmacología , Fármacos Neuroprotectores/farmacología , Células Madre/efectos de los fármacos , Animales , Astrocitos/fisiología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/fisiología , Croton , Citocinas/metabolismo , Flavonoides/química , Interleucina-6/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/química , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Extractos Vegetales/farmacología , Ratas , Ratas Wistar , Células Madre/fisiología
16.
Neurochem Res ; 35(7): 955-66, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20213345

RESUMEN

Flavonoids are naturally occurring polyphenolic compounds that are present in a variety of fruits, vegetables, cereals, tea, and wine, and are the most abundant antioxidants in the human diet. Evidence suggests that these phytochemicals might have an impact on brain pathology and aging; however, neither their mechanisms of action nor their cell targets are completely known. In the mature mammalian brain, astroglia constitute nearly half of the total cells, providing structural, metabolic, and trophic support for neurons. During the past few years, increasing knowledge of these cells has indicated that astrocytes are pivotal characters in neurodegenerative diseases and brain injury. Most of the physiological benefits of flavonoids are generally thought to be due to their antioxidant and free-radical scavenging effects; however, emerging evidence has supported the hypothesis that their mechanism of action might go beyond these properties. In this review, we focus on astrocytes as targets for flavonoids and their implications in brain development, neuroprotection, and glial tumor formation. Finally, we will briefly discuss the emerging view of astrocytes as essential characters in neurodegenerative diseases, and how a better understanding of the action of flavonoids might open new avenues to develop therapeutic approaches to these pathologies.


Asunto(s)
Astrocitos/fisiología , Encefalopatías/patología , Encéfalo/fisiología , Flavonoides/metabolismo , Animales , Anticarcinógenos/farmacología , Anticarcinógenos/uso terapéutico , Astrocitos/citología , Astrocitos/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encefalopatías/prevención & control , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Dieta , Flavonoides/farmacología , Flavonoides/uso terapéutico , Humanos , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/prevención & control , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
17.
J Neurosci Methods ; 343: 108806, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32574642

RESUMEN

BACKGROUND: Astrocytes, one of the main glial cell types, play critical roles in the central nervous system (CNS) development and function, including support of neuronal survival and differentiation, blood brain barrier formation, synapse homeostasis and injury response. Cell isolation and culture techniques have been proved to be a powerful tool to study astrocyte physiology and function. Due to financial constraints and rigid biosafety and ethics rules to use animal models, freezing techniques and the creation of cell banks emerged as alternatives to optimize the use of experimental animals. One of the main challenges, however, of these techniques is to guarantee that conserved cells keep their biological properties. NEW METHOD: In this work, we characterized morphologically and functionally murine secondary astrocyte cultures that have been submitted to freezing/thawing procedures. RESULTS: Morphological characterization of SAC (secondary astrocyte culture) and SFAC (secondary frozen-astrocyte culture) did not reveal significant differences on astrocyte morphology, confluence time and cell number along culture period. Functionally, SAC and SFAC did not reveal differences in their potential to support neuronal survival, maturation, neuritogenesis and synapse formation. CONCLUSIONS: Our results suggest that murine astrocytes that are submitted to freezing/thawing procedure maintain morphological and functional characteristics when compared with non-frozen astrocytes. Thus, this methodological approach is a valuable tool for in vitro research and might allow experimental optimization and reduction of animal use.


Asunto(s)
Astrocitos , Neuroglía , Animales , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Ratones , Neurogénesis
18.
J Peripher Nerv Syst ; 14(4): 285-93, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20021570

RESUMEN

The use of electromagnetic fields has been reported to enhance peripheral nerve regeneration. This study aimed to identify the effects of a prolonged protocol of low-frequency pulsed electromagnetic field (PEMF) on peripheral nerve regeneration. Thirty-four male Swiss mice (Mus musculus) were divided into PEMF (n = 17) and control (n = 17) groups. All animals underwent a unilateral sciatic-crush lesion, and the PEMF group was exposed to a 72-Hz, 2-G electromagnetic field for 30 min, five days a week, for three weeks. Functional analysis was carried out weekly. After three weeks, the animals were euthanized, and histological, morphometric, oxidative stress, and TGF-beta1 analyses were performed. Functional analysis showed no differences between the groups. Histological appearance was similar between PEMF and control nerves. Morphometric assessment showed that the PEMF nerves trended toward decreased regeneration. The levels of free radicals were more pronounced in PEMF nerves, but were not associated with an increase in the content of the TGF-beta1/Smad signaling pathway. Prolonged PEMF regimen leads to delayed histological peripheral nerve regeneration and increased oxidative stress but no loss of function recovery.


Asunto(s)
Magnetoterapia/métodos , Regeneración Nerviosa/fisiología , Nervio Ciático/fisiopatología , Neuropatía Ciática/fisiopatología , Neuropatía Ciática/terapia , Animales , Inmunohistoquímica , Masculino , Ratones , Vaina de Mielina/patología , Vaina de Mielina/fisiología , Compresión Nerviosa , Fibras Nerviosas Mielínicas/patología , Fibras Nerviosas Mielínicas/fisiología , Neuronas/patología , Neuronas/fisiología , Estrés Oxidativo/fisiología , Recuperación de la Función/fisiología , Nervio Ciático/patología , Neuropatía Ciática/patología , Transducción de Señal , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Resultado del Tratamiento
19.
Curr Neurovasc Res ; 16(4): 291-300, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31633476

RESUMEN

BACKGROUND: In the developing cerebral cortex, Radial Glia (RG) multipotent neural stem cell, among other functions, differentiate into astrocytes and serve as a scaffold for blood vessel development. After some time, blood vessel Endothelial Cells (ECs) become associated with astrocytes to form the neurovascular Blood-Brain Barrier (BBB) unit. OBJECTIVE: Since little is known about the mechanisms underlying bidirectional RG-ECs interactions in both vascular development and astrocyte differentiation, this study investigated the impact of interactions between RG and ECs mediated by secreted factors on EC maturation and gliogenesis control. METHODS: First, we demonstrated that immature vasculature in the murine embryonic cerebral cortex physically interacts with Nestin positive RG neural stem cells in vivo. Isolated Microcapillary Brain Endothelial Cells (MBEC) treated with the conditioned medium from RG cultures (RG-CM) displayed decreased proliferation, reduction in the protein levels of the endothelial tip cell marker Delta-like 4 (Dll4), and decreased expression levels of the vascular permeability associated gene, plasmalemma vesicle-associated protein-1 (PLVAP1). These events were also accompanied by increased levels of the tight junction protein expression, zonula occludens-1 (ZO-1). RESULTS: Finally, we demonstrated that isolated RG cells cultures treated with MBEC conditioned medium promoted the differentiation of astrocytes in a Vascular Endothelial Growth Factor-A (VEGF-A) dependent manner. CONCLUSION: These results suggest that the bidirectional interaction between RG and ECs is essential to induce vascular maturation and astrocyte generation, which may be an essential cell-cell communication mechanism to promote BBB establishment.


Asunto(s)
Astrocitos/citología , Barrera Hematoencefálica/citología , Diferenciación Celular/fisiología , Células Endoteliales/citología , Animales , Encéfalo/citología , Encéfalo/metabolismo , Permeabilidad Capilar/fisiología , Células Cultivadas , Ratones , Células-Madre Neurales/citología , Neurogénesis/fisiología
20.
Mol Neurobiol ; 56(7): 4653-4679, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30377983

RESUMEN

Transforming growth factor betas (TGF-ßs) are known as multifunctional growth factors that participate in the regulation of key events of development, disease, and tissue repair. In the brain, TGF-ß1 has been widely recognized as an injury-related cytokine, particularly associated with astrocyte scar formation in response to brain injury. In the last decade, however, evidence has indicated that in addition to its role in brain injury, TGF-ß1 might be a crucial regulator of cell survival and differentiation, brain homeostasis, angiogenesis, memory formation, and neuronal plasticity. In this review, we will discuss the emerging scenario of TGF-ß1 as a key regulator of astrocyte differentiation and function and the implications of TGF-ß1 as a novel mediator of cellular interactions in the central nervous system. First, we will discuss the cellular and molecular basis underlying the effect of TGF-ß on astrocyte generation and its impact on angiogenesis and blood-brain barrier function. Then, we will focus on the role of astrocytes in the development and remodeling of synapses and the role of TGF-ß1 as a new mediator of these events. Furthermore, we present seminal data that contributed to the emerging concept that astrocyte dysfunction might be associated with neurodegenerative diseases, with a special focus on Alzheimer's disease, and discuss the pros and cons of TGF-ß signaling deficits in these processes. Finally, we argue that understanding how astrocytic signals, such as TGF-ß1, regulate brain function might offer new insights into human learning, memory, and cognition, and ultimately, this understanding may provide new targets for the treatment of neurological diseases.


Asunto(s)
Astrocitos/metabolismo , Encefalopatías/patología , Encéfalo/metabolismo , Encéfalo/patología , Factor de Crecimiento Transformador beta1/metabolismo , Envejecimiento/metabolismo , Animales , Humanos , Neovascularización Fisiológica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA