Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Eur J Cell Biol ; 103(2): 151406, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38547677

RESUMEN

Despite extensive research, targeted delivery of substances to the brain still poses a great challenge due to the selectivity of the blood-brain barrier (BBB). Most molecules require either carrier- or receptor-mediated transport systems to reach the central nervous system (CNS). These transport systems form attractive routes for the delivery of therapeutics into the CNS, yet the number of known brain endothelium-enriched receptors allowing the transport of large molecules into the brain is scarce. Therefore, to identify novel BBB targets, we combined transcriptomic analysis of human and murine brain endothelium and performed a complex screening of BBB-enriched genes according to established selection criteria. As a result, we propose the high-affinity cationic amino acid transporter 1 (SLC7A1) as a novel candidate for transport of large molecules across the BBB. Using RNA sequencing and in situ hybridization assays, we demonstrated elevated SLC7A1 gene expression in both human and mouse brain endothelium. Moreover, we confirmed SLC7A1 protein expression in brain vasculature of both young and aged mice. To assess the potential of SLC7A1 as a transporter for larger proteins, we performed internalization and transcytosis studies using a radiolabelled or fluorophore-labelled anti-SLC7A1 antibody. Our results showed that SLC7A1 internalised a SLC7A1-specific antibody in human colorectal carcinoma (HCT116) cells. Moreover, transcytosis studies in both immortalised human brain endothelial (hCMEC/D3) cells and primary mouse brain endothelial cells clearly demonstrated that SLC7A1 effectively transported the SLC7A1-specific antibody from luminal to abluminal side. Therefore, here in this study, we present for the first time the SLC7A1 as a novel candidate for transport of larger molecules across the BBB.


Asunto(s)
Barrera Hematoencefálica , Transportador de Aminoácidos Catiónicos 1 , Barrera Hematoencefálica/metabolismo , Animales , Humanos , Ratones , Transportador de Aminoácidos Catiónicos 1/metabolismo , Transportador de Aminoácidos Catiónicos 1/genética , Células Endoteliales/metabolismo , Ratones Endogámicos C57BL
2.
JCI Insight ; 7(16)2022 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-35881483

RESUMEN

The blood-brain barrier is formed by capillary endothelial cells expressing connexin 37 (Cx37), Cx40, and Cx43 and is joined by closely apposed astrocytes expressing Cx43 and Cx30. We investigated whether connexin-targeting peptides could limit barrier leakage triggered by LPS-induced systemic inflammation in mice. Intraperitoneal LPS administration increased endothelial and astrocytic Cx43 expression; elevated TNF-α, IL-1ß, IFN-γ, and IL-6 in plasma and IL-6 in the brain; and induced barrier leakage recorded over 24 hours. Barrier leakage was largely prevented by global Cx43 knockdown and Cx43/Cx30 double knockout in astrocytes, slightly diminished by endothelial Cx43 knockout, and not protected by global Cx30 knockout. Intravenous administration of Gap27 or Tat-Gap19 peptides just before LPS also prevented barrier leakage, and intravenously administered BAPTA-AM to chelate intracellular calcium was equally effective. Patch-clamp experiments demonstrated LPS-induced Cx43 hemichannel opening in endothelial cells, which was suppressed by Gap27, Gap19, and BAPTA. LPS additionally triggered astrogliosis that was prevented by intravenous Tat-Gap19 or BAPTA-AM. Cortically applied Tat-Gap19 or BAPTA-AM to primarily target astrocytes also strongly diminished barrier leakage. In vivo dye uptake and in vitro patch-clamp showed Cx43 hemichannel opening in astrocytes that was induced by IL-6 in a calcium-dependent manner. We conclude that targeting endothelial and astrocytic connexins is a powerful approach to limit barrier failure and astrogliosis.


Asunto(s)
Barrera Hematoencefálica , Conexina 43 , Animales , Barrera Hematoencefálica/metabolismo , Calcio/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Células Endoteliales/metabolismo , Gliosis/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Péptidos/metabolismo
3.
Cell Death Dis ; 11(3): 194, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32188841

RESUMEN

Radiotherapeutic treatment consists of targeted application of radiation beams to a tumor but exposure of surrounding healthy tissue is inevitable. In the brain, ionizing radiation induces breakdown of the blood-brain barrier by effects on brain microvascular endothelial cells. Damage from directly irradiated cells can be transferred to surrounding non-exposed bystander cells, known as the radiation-induced bystander effect. We investigated involvement of connexin channels and paracrine signaling in radiation-induced bystander DNA damage in brain microvascular endothelial cells exposed to focused X-rays. Irradiation caused DNA damage in the directly exposed area, which propagated over several millimeters in the bystander area. DNA damage was significantly reduced by the connexin channel-targeting peptide Gap26 and the Cx43 hemichannel blocker TAT-Gap19. ATP release, dye uptake, and patch clamp experiments showed that hemichannels opened within 5 min post irradiation in both irradiated and bystander areas. Bystander signaling involved cellular Ca2+ dynamics and IP3, ATP, ROS, and NO signaling, with Ca2+, IP3, and ROS as crucial propagators of DNA damage. We conclude that bystander effects are communicated by a concerted cascade involving connexin channels, and IP3/Ca2+, ATP, ROS, and NO as major contributors of regenerative signal expansion.


Asunto(s)
Adenosina Trifosfato/metabolismo , Encéfalo/irrigación sanguínea , Conexina 43/metabolismo , Daño del ADN , Células Endoteliales/metabolismo , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Células Endoteliales/efectos de la radiación , Células HeLa , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratas , Transducción de Señal
4.
Sci Rep ; 9(1): 16363, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31705038

RESUMEN

Multiple animal models have been created to gain insight into Alzheimer's disease (AD) pathology. Among the most commonly used models are transgenic mice overexpressing human amyloid precursor protein (APP) with mutations linked to familial AD, resulting in the formation of amyloid ß plaques, one of the pathological hallmarks observed in AD patients. However, recent evidence suggests that the overexpression of APP by itself can confound some of the reported observations. Therefore, we investigated in the present study the AppNL-G-Fmodel, an App knock-in (App-KI) mouse model that develops amyloidosis in the absence of APP-overexpression. Our findings at the behavioral, electrophysiological, and histopathological level confirmed an age-dependent increase in Aß1-42 levels and plaque deposition in these mice in accordance with previous reports. This had apparently no consequences on cognitive performance in a visual discrimination (VD) task, which was largely unaffected in AppNL-G-F mice at the ages tested. Additionally, we investigated neurophysiological functioning of several brain areas by phase-amplitude coupling (PAC) analysis, a measure associated with adequate cognitive functioning, during the VD task (starting at 4.5 months) and the exploration of home environment (at 5 and 8 months of age). While we did not detect age-dependent changes in PAC during home environment exploration for both the wild-type and the AppNL-G-F mice, we did observe subtle changes in PAC in the wild-type mice that were not present in the AppNL-G-F mice.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Ondas Encefálicas/fisiología , Cognición/fisiología , Modelos Animales de Enfermedad , Neuronas/patología , Placa Amiloide/patología , Animales , Conducta Animal , Aprendizaje Discriminativo , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Placa Amiloide/metabolismo , Percepción Visual
5.
J Exp Med ; 216(4): 916-935, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30872361

RESUMEN

Connexin43 (Cx43) function is influenced by kinases that phosphorylate specific serine sites located near its C-terminus. Stroke is a powerful inducer of kinase activity, but its effect on Cx43 is unknown. We investigated the impact of wild-type (WT) and knock-in Cx43 with serine to alanine mutations at the protein kinase C (PKC) site Cx43S368A, the casein kinase 1 (CK1) sites Cx43S325A/328Y/330A, and the mitogen-activated protein kinase (MAPK) sites Cx43S255/262/279/282A (MK4) on a permanent middle cerebral artery occlusion (pMCAO) stroke model. We demonstrate that MK4 transgenic animals exhibit a significant decrease in infarct volume that was associated with improvement in behavioral performance. An increase in astrocyte reactivity with a concomitant decrease in microglial reactivity was observed in MK4 mice. In contrast to WT, MK4 astrocytes displayed reduced Cx43 hemichannel activity. Pharmacological blockade of Cx43 hemichannels with TAT-Gap19 also significantly decreased infarct volume in WT animals. This study provides novel molecular insights and charts new avenues for therapeutic intervention associated with Cx43 function.


Asunto(s)
Infarto Cerebral/metabolismo , Conexina 43/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuroprotección/efectos de los fármacos , Neuroprotección/genética , Animales , Astrocitos/metabolismo , Conexina 43/antagonistas & inhibidores , Conexina 43/genética , Conexina 43/farmacología , Modelos Animales de Enfermedad , Uniones Comunicantes/metabolismo , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Fragmentos de Péptidos/farmacología , Fosforilación
6.
Biochim Biophys Acta Mol Cell Res ; 1865(11 Pt B): 1805-1814, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30076858

RESUMEN

Photodynamic therapy combines three non-toxic components: light, oxygen and a photosensitizer to generate singlet oxygen and/or other ROS molecules in order to target destruction of cancer cells. The damage induced in the targeted cells can furthermore propagate to non-exposed bystander cells thereby exacerbating the damage. Ca2+ signaling is strongly intertwined with ROS signaling and both play crucial roles in cell death. In this review we aimed to review current knowledge on the role of Ca2+ and ROS signaling, their effect on cell-cell propagation via connexin-linked mechanisms and the outcome in terms of cell death. In general, photodynamic therapy results in an increased cytosolic Ca2+ concentration originating from Ca2+ entry or Ca2+ release from internal stores. While photodynamic therapy can certainly induce cell death, the outcome depends on the cell type and the photosensitizer used. Connexin channels propagating the Ca2+ signal, and presumably regenerating ROS at distance, may play a role in spreading the effect to neighboring non-exposed bystander cells. Given the various cell types and photosensitizers used, there is currently no unified signaling scheme to explain the role of Ca2+ and connexins in the responses following photodynamic therapy. This article is part of a Special Issue entitled: Calcium signaling in health, disease and therapy edited by Geert Bultynck and Jan Parys.


Asunto(s)
Señalización del Calcio/efectos de la radiación , Calcio/metabolismo , Fotoquimioterapia , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Señalización del Calcio/efectos de los fármacos , Citoplasma/metabolismo , Humanos , Óxido Nítrico/metabolismo , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
7.
Neurochem Res ; 42(9): 2519-2536, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28634726

RESUMEN

Neuronal survival, electrical signaling and synaptic activity require a well-balanced micro-environment in the central nervous system. This is achieved by the blood-brain barrier (BBB), an endothelial barrier situated in the brain capillaries, that controls near-to-all passage in and out of the brain. The endothelial barrier function is highly dependent on signaling interactions with surrounding glial, neuronal and vascular cells, together forming the neuro-glio-vascular unit. Within this functional unit, connexin (Cx) channels are of utmost importance for intercellular communication between the different cellular compartments. Connexins are best known as the building blocks of gap junction (GJ) channels that enable direct cell-cell transfer of metabolic, biochemical and electric signals. In addition, beyond their role in direct intercellular communication, Cxs also form unapposed, non-junctional hemichannels in the plasma membrane that allow the passage of several paracrine messengers, complementing direct GJ communication. Within the NGVU, Cxs are expressed in vascular endothelial cells, including those that form the BBB, and are eminent in astrocytes, especially at their endfoot processes that wrap around cerebral vessels. However, despite the density of Cx channels at this so-called gliovascular interface, it remains unclear as to how Cx-based signaling between astrocytes and BBB endothelial cells may converge control over BBB permeability in health and disease. In this review we describe available evidence that supports a role for astroglial as well as endothelial Cxs in the regulation of BBB permeability during development as well as in disease states.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Conexinas/metabolismo , Neuroglía/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Barrera Hematoencefálica/patología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Comunicación Celular/fisiología , Humanos , Mediadores de Inflamación/metabolismo , Neuroglía/patología , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología
8.
Biochim Biophys Acta Mol Cell Res ; 1864(6): 1099-1120, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28193563

RESUMEN

Although radiotherapy is commonly used to treat cancer, its beneficial outcome is frequently hampered by the radiation resistance of tumor cells and adverse reactions in normal tissues. Mechanisms of cell-to-cell communication and how intercellular signals are translated into cellular responses, have become topics of intense investigation, particularly within the field of radiobiology. A substantial amount of evidence is available demonstrating that both gap junctional and paracrine communication pathways can propagate radiation-induced biological effects at the intercellular level, commonly referred to as radiation-induced bystander effects (RIBE). Multiple molecular signaling mechanisms involving oxidative stress, kinases, inflammatory molecules, and Ca2+ are postulated to contribute to RIBE. Ca2+ is a highly versatile and ubiquitous second messenger that regulates diverse cellular processes via the interaction with various signaling cascades. It furthermore provides a fast system for the dissemination of information at the intercellular level. Channels formed by transmembrane connexin (Cx) proteins, i.e. hemichannels and gap junction channels, can mediate the cell-to-cell propagation of increases in intracellular Ca2+ by ministering paracrine and direct cell-cell communication, respectively. We here review current knowledge on radiation-induced signaling mechanisms in irradiated and bystander cells, particularly focusing on the contribution of oxidative stress, Ca2+ and Cx channels. By illustrating the tight interplay between these different partners, we provide a conceptual framework for intercellular Ca2+ signaling as a key player in modulating the RIBE and the overall response to radiation.


Asunto(s)
Calcio/metabolismo , Conexinas/metabolismo , Estrés Oxidativo , Radioterapia , Señalización del Calcio , Humanos , Especies Reactivas de Oxígeno/metabolismo
9.
Cardiovasc Res ; 113(2): 195-206, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27677282

RESUMEN

AIMS: Connexins form gap-junctions (GJs) that directly connect cells, thereby coordinating vascular cell function and controlling vessel diameter and blood flow. GJs are composed of two hemichannels contributed by each of the connecting cells. Hemichannels also exist as non-junctional channels that, when open, lead to the entry/loss of ions and the escape of ATP. Here we investigated cross-talk between hemichannels and Ca2+/purinergic signalling in controlling blood vessel contraction. We hypothesized that hemichannel Ca2+ entry and ATP release contributes to smooth muscle cell (SMC) Ca2+ dynamics, thereby influencing vessel contractility. We applied several peptide modulators of hemichannel function and inhibitors of Ca2+ and ATP signalling to investigate their influence on SMC Ca2+ dynamics and vessel contractility. METHODS AND RESULTS: Confocal Ca2+ imaging studies on small mesenteric arteries (SMAs) from rat demonstrated that norepinephrine-induced SMC Ca2+ oscillations were inhibited by blocking IP3 receptors with xestospongin-C and by interfering with hemichannel function, most notably by the specific Cx43 hemichannel blocking peptide TAT-L2 and by TAT-CT9 that promotes Cx43 hemichannel opening. Evidence for hemichannel involvement in SMC function was supported by the fact that TAT-CT9 significantly increased SMC resting cytoplasmic Ca2+ concentration, indicating it facilitated Ca2+ entry, and by the observation that norepinephrine-triggered vessel ATP release was blocked by TAT-L2. Myograph tension measurements on isolated SMAs showed significant inhibition of norepinephrine-triggered contractility by the ATP receptor antagonist suramin, but the strongest effect was observed with TAT-L2 that gave ∼80% inhibition at 37 °C. TAT-L2 inhibition of vessel contraction was significantly reduced in conditional Cx43 knockout animals, indicating the effect was Cx43 hemichannel-dependent. Computational modelling suggested these results could be explained by the opening of a single hemichannel per SMC. CONCLUSIONS: These results indicate that Cx43 hemichannels contribute to SMC Ca2+ dynamics and contractility, by facilitating Ca2+ entry, ATP release, and purinergic signalling.


Asunto(s)
Adenosina Trifosfato/metabolismo , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Comunicación Celular/efectos de los fármacos , Conexina 43/antagonistas & inhibidores , Uniones Comunicantes/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Péptidos/farmacología , Vasoconstricción/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Simulación por Computador , Conexina 43/deficiencia , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/antagonistas & inhibidores , Conexinas/metabolismo , Femenino , Uniones Comunicantes/metabolismo , Genotipo , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato/agonistas , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Ratones Noqueados , Microscopía Confocal , Modelos Cardiovasculares , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Norepinefrina/farmacología , Fenotipo , Antagonistas Purinérgicos/farmacología , Ratas Wistar , Factores de Tiempo , Vasoconstrictores/farmacología , Proteína alfa-4 de Unión Comunicante
10.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 51-61, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27741412

RESUMEN

Pannexins form channels at the plasma membrane surface that establish a pathway for communication between the cytosol of individual cells and their extracellular environment. By doing so, pannexin signaling dictates several physiological functions, but equally underlies a number of pathological processes. Indeed, pannexin channels drive inflammation by assisting in the activation of inflammasomes, the release of pro-inflammatory cytokines, and the activation and migration of leukocytes. Furthermore, these cellular pores facilitate cell death, including apoptosis, pyroptosis and autophagy. The present paper reviews the roles of pannexin channels in inflammation and cell death. In a first part, a state-of-the-art overview of pannexin channel structure, regulation and function is provided. In a second part, the mechanisms behind their involvement in inflammation and cell death are discussed.


Asunto(s)
Conexinas/inmunología , Inflamasomas/inmunología , Inflamación/inmunología , Leucocitos/inmunología , Proteínas del Tejido Nervioso/inmunología , Animales , Muerte Celular/genética , Muerte Celular/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Movimiento Celular , Conexinas/química , Conexinas/genética , Citocinas/biosíntesis , Citocinas/inmunología , Citosol/inmunología , Citosol/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/patología , Leucocitos/patología , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Transducción de Señal
11.
Methods Mol Biol ; 1437: 155-69, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27207293

RESUMEN

Intercellular communication occurring via gap junction channels is considered a key mechanism for synchronizing physiological functions of cells and for the maintenance of tissue homeostasis. Gap junction channels are protein channels that are situated between neighboring cells and that provide a direct, yet selective route for the passage of small hydrophilic biomolecules and ions. Here, an electroporation method is described to load a localized area within an adherent cell monolayer with a gap junction-permeable fluorescent reporter dye. The technique results in a rapid and efficient labeling of a small patch of cells within the cell culture, without affecting cellular viability. Dynamic and quantitative information on gap junctional communication can subsequently be extracted by tracing the intercellular movement of the dye via time-lapse microscopy.


Asunto(s)
Electroporación/métodos , Colorantes Fluorescentes/metabolismo , Uniones Comunicantes/fisiología , Coloración y Etiquetado/métodos , Transfección/métodos , Animales , Carbenoxolona/farmacología , Comunicación Celular/fisiología , Recuento de Células , Línea Celular Tumoral , Supervivencia Celular , Conexina 43/antagonistas & inhibidores , Conexina 43/genética , Conexina 43/metabolismo , Electroporación/instrumentación , Colorantes Fluorescentes/efectos adversos , Humanos , Microscopía Fluorescente , Microscopía de Contraste de Fase/métodos , Ratas
12.
Glia ; 64(7): 1097-123, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26852907

RESUMEN

Efficient neuronal signaling in the central nervous system strictly depends on a well-balanced microenvironment around glial cells, synapses, and axons. Unique features of the blood-brain barrier (BBB) endothelium largely determine the composition of this micro-milieu and are dependent on the tight interplay with surrounding astrocytes and pericytes. BBB endothelial cells are endowed with a highly restrictive junctional complex that occludes the intercellular cleft, thereby preventing paracellular diffusion. The paracellular pathway is subject to extensive research as integrity loss of the junctional complex is associated with many neuropathologies, inflammation, and edema. Another important feature of the BBB endothelium is the low prevalence of nonspecific, transcytotic events, including (macro)pinocytosis, clathrin-dependent and caveolin-dependent endocytosis and the subsequent trafficking of vesicles to the opposite membrane. Although less studied, evidence is accruing that this pathway importantly contributes to increased BBB permeability, often when the junctional complex remains intact. Here, we review current knowledge on the contribution of the transcellular pathway to the BBB leak observed in different pathologic conditions. In addition, we hypothesize that nonselective, large pore connexin and pannexin channels may contribute to transcellular transport, either by providing a direct diffusion pathway across the endothelial monolayer, or indirectly, by exerting control over intracellular levels of the signaling ion Ca(2+) that is involved in many steps of the vesicular pathway. We conclude that transcytotic events at the BBB, despite being less acknowledged, cannot be simply dismissed as done in the past, but actively contribute to BBB leakage in many different pathologies. GLIA 2016;64:1097-1123.


Asunto(s)
Barrera Hematoencefálica/fisiología , Neuroglía/metabolismo , Transcitosis/fisiología , Animales , Barrera Hematoencefálica/citología , Células Endoteliales/metabolismo , Humanos , Permeabilidad
13.
Mediators Inflamm ; 2015: 257471, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26424967

RESUMEN

The coordination of tissue function is mediated by gap junctions (GJs) that enable direct cell-cell transfer of metabolic and electric signals. GJs are formed by connexin (Cx) proteins of which Cx43 is most widespread in the human body. Beyond its role in direct intercellular communication, Cx43 also forms nonjunctional hemichannels (HCs) in the plasma membrane that mediate the release of paracrine signaling molecules in the extracellular environment. Both HC and GJ channel function are regulated by protein-protein interactions and posttranslational modifications that predominantly take place in the C-terminal domain of Cx43. Matrix metalloproteases (MMPs) are a major group of zinc-dependent proteases, known to regulate not only extracellular matrix remodeling, but also processing of intracellular proteins. Together with Cx43 channels, both GJs and HCs, MMPs contribute to acute inflammation and a small number of studies reports on an MMP-Cx43 link. Here, we build further on these reports and present a novel hypothesis that describes proteolytic cleavage of the Cx43 C-terminal domain by MMPs and explores possibilities of how such cleavage events may affect Cx43 channel function. Finally, we set out how aberrant channel function resulting from cleavage can contribute to the acute inflammatory response during tissue injury.


Asunto(s)
Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Animales , Humanos , Inflamación/metabolismo
14.
Cell Mol Life Sci ; 72(15): 2823-51, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26118660

RESUMEN

The central nervous system (CNS) is composed of a highly heterogeneous population of cells. Dynamic interactions between different compartments (neuronal, glial, and vascular systems) drive CNS function and allow to integrate and process information as well as to respond accordingly. Communication within this functional unit, coined the neuro-glio-vascular unit (NGVU), typically relies on two main mechanisms: direct cell-cell coupling via gap junction channels (GJCs) and paracrine communication via the extracellular compartment, two routes to which channels composed of transmembrane connexin (Cx) or pannexin (Panx) proteins can contribute. Multiple isoforms of both protein families are present in the CNS and each CNS cell type is characterized by a unique Cx/Panx portfolio. Over the last two decades, research has uncovered a multilevel platform via which Cxs and Panxs can influence different cellular functions within a tissue: (1) Cx GJCs enable a direct cell-cell communication of small molecules, (2) Cx hemichannels and Panx channels can contribute to autocrine/paracrine signaling pathways, and (3) different structural domains of these proteins allow for channel-independent functions, such as cell-cell adhesion, interactions with the cytoskeleton, and the activation of intracellular signaling pathways. In this paper, we discuss current knowledge on their multifaceted contribution to brain development and to specific processes in the NGVU, including synaptic transmission and plasticity, glial signaling, vasomotor control, and blood-brain barrier integrity in the mature CNS. By highlighting both physiological and pathological conditions, it becomes evident that Cxs and Panxs can play a dual role in the CNS and that an accurate fine-tuning of each signaling mechanism is crucial for normal CNS physiology.


Asunto(s)
Sistema Nervioso Central/patología , Sistema Nervioso Central/fisiología , Conexinas/metabolismo , Transducción de Señal/fisiología , Animales , Sistema Nervioso Central/metabolismo , Uniones Comunicantes/metabolismo , Uniones Comunicantes/fisiología , Humanos , Fenómenos Fisiológicos del Sistema Nervioso
15.
Cold Spring Harb Protoc ; 2015(3): 284-8, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25734060

RESUMEN

Electroporation is a technique that temporarily induces pores in the plasma membranes of cells, thereby allowing plasma membrane-impermeable substances to enter the cells. This loading method requires an electrical drive circuit providing an electroporation signal, an electrode to apply the signal to a localized zone in a cell monolayer, and a special solution that has a low electrical conductivity. To avoid impairment of cell function and cell death from the electroporation procedure itself, the applied electrical signal should ideally be a high-frequency oscillating signal (50 kHz) without any direct current (DC) component. Here, we describe the detailed procedure of electroporation loading.


Asunto(s)
Señalización del Calcio , Fenómenos Fisiológicos Celulares , Electroporación/métodos , Animales , Línea Celular , Humanos
16.
Cold Spring Harb Protoc ; 2015(3): 289-92, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25734061

RESUMEN

Caged IP3 is an inactive form of the second messenger IP3, consisting of the biologically active molecule linked to a cage group through a photolabile bond. This bond is cleaved by exposure to brief "flashes" of ultraviolet (UV) light, thereby releasing the active IP3 molecule. The protection of caged IP3 against metabolic transformation in combination with a defined time point of fast photoliberation of IP3 provides an efficient way to temporally and spatially control the cytosolic release of IP3 and subsequent increase of cytoplasmic Ca(2+). These properties make it an ideal method for kinetic studies and also a well-suited procedure to initiate intercellular Ca(2+) waves from a point source of IP3. This protocol describes cell loading with membrane impermeable caged IP3 and the UV flash illumination procedure.


Asunto(s)
Inositol 1,4,5-Trifosfato/análogos & derivados , Fotólisis , Rayos Ultravioleta , Animales , Calcio/metabolismo , Línea Celular , Citosol/química , Humanos , Inositol 1,4,5-Trifosfato/efectos de la radiación
17.
Cold Spring Harb Protoc ; 2015(3): 239-49, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25734071

RESUMEN

Many cellular functions are driven by variations in the intracellular Ca(2+) concentration ([Ca(2+)]i), which may appear as a single-event transient [Ca(2+)]i elevation, repetitive [Ca(2+)]i increases known as Ca(2+) oscillations, or [Ca(2+)]i increases propagating in the cytoplasm as Ca(2+) waves. Additionally, [Ca(2+)]i changes can be communicated between cells as intercellular Ca(2+) waves (ICWs). ICWs are mediated by two possible mechanisms acting in parallel: one involving gap junctions that form channels directly linking the cytoplasm of adjacent cells and one involving a paracrine messenger, in most cases ATP, that is released into the extracellular space, leading to [Ca(2+)]i changes in neighboring cells. The intracellular messenger inositol 1,4,5-trisphosphate (IP3) that triggers Ca(2+) release from Ca(2+) stores is crucial in these two ICW propagation scenarios, and is also a potent trigger to initiate ICWs. Loading inactive, "caged" IP3 into cells followed by photolytic "uncaging" with UV light, thereby liberating IP3, is a well-established method to trigger [Ca(2+)]i changes in single cells that is also effective in initiating ICWs. We here describe a method to load cells with caged IP3 by local electroporation of monolayer cell cultures and to apply flash photolysis to increase intracellular IP3 and induce [Ca(2+)]i changes, or initiate ICWs. Moreover, the electroporation method allows loading of membrane-impermeable agents that interfere with IP3 and Ca(2+) signaling.


Asunto(s)
Señalización del Calcio , Electroporación/métodos , Inositol 1,4,5-Trifosfato/análogos & derivados , Fotólisis/efectos de la radiación , Adenosina Trifosfato/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Inositol 1,4,5-Trifosfato/efectos de la radiación
18.
Front Cell Neurosci ; 8: 306, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25374505

RESUMEN

In the brain, astrocytes represent the cellular population that expresses the highest amount of connexins (Cxs). This family of membrane proteins is the molecular constituent of gap junction channels and hemichannels that provide pathways for direct cytoplasm-to-cytoplasm and inside-out exchange, respectively. Both types of Cx channels are permeable to ions and small signaling molecules allowing astrocytes to establish dynamic interactions with neurons. So far, most pharmacological approaches currently available do not distinguish between these two channel functions, stressing the need to develop new specific molecular tools. In astrocytes two major Cxs are expressed, Cx43 and Cx30, and there is now evidence indicating that at least Cx43 operates as a gap junction channel as well as a hemichannel in these cells. Based on studies in primary cultures as well as in acute hippocampal slices, we report here that Gap19, a nonapeptide derived from the cytoplasmic loop of Cx43, inhibits astroglial Cx43 hemichannels in a dose-dependent manner, without affecting gap junction channels. This peptide, which not only selectively inhibits hemichannels but is also specific for Cx43, can be delivered in vivo in mice as TAT-Gap19, and displays penetration into the brain parenchyma. As a result, Gap19 combined with other tools opens up new avenues to decipher the role of Cx43 hemichannels in interactions between astrocytes and neurons in physiological as well as pathological situations.

19.
Biochim Biophys Acta ; 1843(10): 2211-32, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24768716

RESUMEN

For decades, studies have been focusing on the neuronal abnormalities that accompany neurodegenerative disorders. Yet, glial cells are emerging as important players in numerous neurological diseases. Astrocytes, the main type of glia in the central nervous system , form extensive networks that physically and functionally connect neuronal synapses with cerebral blood vessels. Normal brain functioning strictly depends on highly specialized cellular cross-talk between these different partners to which Ca(2+), as a signaling ion, largely contributes. Altered intracellular Ca(2+) levels are associated with neurodegenerative disorders and play a crucial role in the glial responses to injury. Intracellular Ca(2+) increases in single astrocytes can be propagated toward neighboring cells as intercellular Ca(2+) waves, thereby recruiting a larger group of cells. Intercellular Ca(2+) wave propagation depends on two, parallel, connexin (Cx) channel-based mechanisms: i) the diffusion of inositol 1,4,5-trisphosphate through gap junction channels that directly connect the cytoplasm of neighboring cells, and ii) the release of paracrine messengers such as glutamate and ATP through hemichannels ('half of a gap junction channel'). This review gives an overview of the current knowledge on Cx-mediated Ca(2+) communication among astrocytes as well as between astrocytes and other brain cell types in physiology and pathology, with a focus on the processes of neurodegeneration and reactive gliosis. Research on Cx-mediated astroglial Ca(2+) communication may ultimately shed light on the development of targeted therapies for neurodegenerative disorders in which astrocytes participate. This article is part of a Special Issue entitled: Calcium signaling in health and disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Señalización del Calcio , Calcio/metabolismo , Conexinas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Adenosina Trifosfato/metabolismo , Astrocitos/patología , Encéfalo/patología , Comunicación Celular , Conexinas/genética , Uniones Comunicantes/metabolismo , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Terapia Molecular Dirigida , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Neuronas/metabolismo , Neuronas/patología , Sinapsis/metabolismo , Sinapsis/patología
20.
FEBS Lett ; 588(8): 1259-70, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24631535

RESUMEN

Neuronal signaling in the CNS depends on the microenvironment around synapses and axons. To prevent fluctuations in blood composition affecting the interstitial fluid and CSF, two barriers, the blood-brain barrier (BBB) and blood-CSF barrier (BCSFB), are interposed between the blood and the brain/CSF compartment. Brain capillary endothelial cells (ECs) constitute the BBB whereas choroid plexus epithelial (CPE) cells form the BCSFB. The anatomical basis of these barriers is located at the level of an intercellular junctional complex that impedes paracellular diffusion. Tight and adherens junctions are known as the principal constituents of this junctional complex. Transmembrane connexins (Cxs) are the prime building blocks of plasma membrane hemichannels that combine to form intercellular gap junctions (GJ). Although Cxs co-exist within the junctional complex, their influence on tight/adherens junctions and their role in barrier function of BBB ECs and CPE has been mostly ignored. Here, we review current knowledge on the role of Cxs in the BBB, BCSFB and other interfaces that subside within the CNS. We conclude that Cxs are a rather unexplored but promising target for influencing CNS barrier function.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematonerviosa/metabolismo , Conexinas/metabolismo , Uniones Intercelulares/metabolismo , Animales , Humanos , Proteínas de Uniones Estrechas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...