Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 37(2): 333-348, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28077713

RESUMEN

Mitochondria play a variety of functional roles in cortical neurons, from metabolic support and neuroprotection to the release of cytokines that trigger apoptosis. In dendrites, mitochondrial structure is closely linked to their function, and fragmentation (fission) of the normally elongated mitochondria indicates loss of their function under pathological conditions, such as stroke and brain trauma. Using in vivo two-photon microscopy in mouse brain, we quantified mitochondrial fragmentation in a full spectrum of cortical injuries, ranging from severe to mild. Severe global ischemic injury was induced by bilateral common carotid artery occlusion, whereas severe focal stroke injury was induced by Rose Bengal photosensitization. The moderate and mild traumatic injury was inflicted by focal laser lesion and by mild photo-damage, respectively. Dendritic and mitochondrial structural changes were tracked longitudinally using transgenic mice expressing fluorescent proteins localized either in cytosol or in mitochondrial matrix. In response to severe injury, mitochondrial fragmentation developed in parallel with dendritic damage signified by dendritic beading. Reconstruction from serial section electron microscopy confirmed mitochondrial fragmentation. Unlike dendritic beading, fragmentation spread beyond the injury core in focal stroke and focal laser lesion models. In moderate and mild injury, mitochondrial fragmentation was reversible with full recovery of structural integrity after 1-2 weeks. The transient fragmentation observed in the mild photo-damage model was associated with changes in dendritic spine density without any signs of dendritic damage. Our findings indicate that alterations in neuronal mitochondria structure are very sensitive to the tissue damage and can be reversible in ischemic and traumatic injuries. SIGNIFICANCE STATEMENT: During ischemic stroke or brain trauma, mitochondria can either protect neurons by supplying ATP and adsorbing excessive Ca2+, or kill neurons by releasing proapoptotic factors. Mitochondrial function is tightly linked to their morphology: healthy mitochondria are thin and long; dysfunctional mitochondria are thick (swollen) and short (fragmented). To date, fragmentation of mitochondria was studied either in dissociated cultured neurons or in brain slices, but not in the intact living brain. Using real-time in vivo two-photon microscopy, we quantified mitochondrial fragmentation during acute pathological conditions that mimic severe, moderate, and mild brain injury. We demonstrated that alterations in neuronal mitochondria structural integrity can be reversible in traumatic and ischemic injuries, highlighting mitochondria as a potential target for therapeutic interventions.


Asunto(s)
Lesiones Encefálicas/diagnóstico por imagen , Isquemia Encefálica/diagnóstico por imagen , Microscopía de Fluorescencia por Excitación Multifotónica , Mitocondrias/patología , Neocórtex/diagnóstico por imagen , Neuronas/patología , Anestesia/métodos , Animales , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Dendritas/metabolismo , Dendritas/patología , Femenino , Colorantes Fluorescentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Mitocondrias/metabolismo , Neocórtex/metabolismo , Neuronas/metabolismo
2.
Brain Behav Immun ; 62: 110-123, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28119027

RESUMEN

The immune function of AMIGO2 is currently unknown. Here, we revealed novel roles of AMIGO2 in modulating T-cell functions and EAE using Amigo2-knockout (AMG2KO) mice. Amigo2 was abundantly expressed by murine T helper (Th) cells. Its deficiency impaired transplanted T-cell infiltration into the secondary lymphoid organs and dampened Th-cell activation, but promoted splenic Th-cell proliferation and abundancy therein. AMG2KO Th cells had respectively elevated T-bet in Th1- and GATA-3 in Th2-lineage during early Th-cell differentiation, accompanied with increased IFN-γ and IL-10 but decreased IL-17A production. AMG2KO mice exhibited ameliorated EAE, dampened spinal T-cell accumulation, decreased serum IL-17A levels and enhanced splenic IL-10 production. Adoptive transfer of encephalitogenic AMG2KO T cells induced milder EAE and dampened spinal Th-cell accumulation and Tnf expression. Mechanistically, Amigo2-overexpression in 293T cells dampened NF-kB transcriptional activity, while Amigo2-deficiency enhanced Akt but suppressed GSK-3ß phosphorylation and promoted nuclear translocations of NF-kB and NFAT1 in Th-cells. Collectively, our data demonstrate that AMIGO2 is important in regulating T-cell functions and EAE, and may be harnessed as a potential therapeutic target for multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Bazo/metabolismo , Células TH1/metabolismo , Células Th17/metabolismo , Traslado Adoptivo , Animales , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-17/sangre , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Bazo/inmunología , Células TH1/inmunología , Células Th17/inmunología
3.
J Neurosci ; 35(14): 5693-706, 2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25855182

RESUMEN

High mobility group (HMG) proteins concentrate in the nucleus, interacting with chromatin. Amphoterin is an HMG protein (HMGB1) that has been shown to have extranuclear functions and can be secreted from some cell types. Exogenous amphoterin can increase neurite growth, suggesting that the secreted protein may have growth promoting activities in neurons. Consistent with this, we show that depletion of amphoterin mRNA from cultured adult rat DRG neurons attenuates neurite outgrowth, pointing to autocrine or paracrine mechanisms for its growth-promoting effects. The mRNA encoding amphoterin localizes to axonal processes and we showed recently that its 3'-UTR is sufficient for axonal localization of heterologous transcripts (Donnelly et al., 2013). Here, we show that amphoterin mRNA is transported constitutively into axons of adult DRG neurons. A preconditioning nerve injury increases the levels of amphoterin protein in axons without a corresponding increase in amphoterin mRNA in the axons. A 60 nucleotide region of the amphoterin mRNA 3'-UTR is necessary and sufficient for its localization into axons of cultured sensory neurons. Amphoterin mRNA 3'-UTR is also sufficient for axonal localization in distal axons of DRG neurons in vivo. Overexpression of axonally targeted amphoterin mRNA increases axon outgrowth in cultured sensory neurons, but axon growth is not affected when the overexpressed mRNA is restricted to the cell body.


Asunto(s)
Axones/metabolismo , Regulación de la Expresión Génica/genética , Proteína HMGB1/genética , Biosíntesis de Proteínas/genética , ARN Mensajero/metabolismo , Células Receptoras Sensoriales/citología , Regiones no Traducidas 3'/genética , Animales , Axones/efectos de los fármacos , Transporte Biológico/genética , Células Cultivadas , Ganglios Espinales/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteína HMGB1/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Fotoblanqueo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Transducción Genética
4.
J Biol Chem ; 289(29): 19958-75, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24904058

RESUMEN

The Amigo protein family consists of three transmembrane proteins characterized by six leucine-rich repeat domains and one immunoglobulin-like domain in their extracellular moieties. Previous in vitro studies have suggested a role as homophilic adhesion molecules in brain neurons, but the in vivo functions remain unknown. Here we have cloned all three zebrafish amigos and show that amigo1 is the predominant family member expressed during nervous system development in zebrafish. Knockdown of amigo1 expression using morpholino oligonucleotides impairs the formation of fasciculated tracts in early fiber scaffolds of brain. A similar defect in fiber tract development is caused by mRNA-mediated expression of the Amigo1 ectodomain that inhibits adhesion mediated by the full-length protein. Analysis of differentiated neural circuits reveals defects in the catecholaminergic system. At the behavioral level, the disturbed formation of neural circuitry is reflected in enhanced locomotor activity and in the inability of the larvae to perform normal escape responses. We suggest that Amigo1 is essential for the development of neural circuits of zebrafish, where its mechanism involves homophilic interactions within the developing fiber tracts and regulation of the Kv2.1 potassium channel to form functional neural circuitry that controls locomotion.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Larva/crecimiento & desarrollo , Larva/metabolismo , Masculino , Red Nerviosa/crecimiento & desarrollo , Red Nerviosa/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Moléculas de Adhesión de Célula Nerviosa/antagonistas & inhibidores , Moléculas de Adhesión de Célula Nerviosa/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canales de Potasio Shab/genética , Canales de Potasio Shab/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
5.
J Neuroinflammation ; 12: 75, 2015 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-25895500

RESUMEN

BACKGROUND: Social deficit is one of the core symptoms of neuropsychiatric diseases, in which immune genes play an important role. Although a few immune genes have been shown to regulate social and emotional behaviors, how immune gene network(s) may jointly regulate sociability has not been investigated so far. METHODS: To decipher the potential immune-mediated mechanisms underlying social behavior, we first studied the brain microarray data of eight inbred mouse strains with known variations in social behavior and retrieved the differentially expressed immune genes. We then made a protein-protein interaction analysis of them to find the major networks and explored the potential association of these genes with the behavior and brain morphology in the mouse phenome database. To validate the expression and function of the candidate immune genes, we selected the C57BL/6 J and DBA/2 J strains among the eight inbred strains, compared their social behaviors in resident-intruder and 3-chambered social tests and the mRNA levels of these genes, and analyzed the correlations of these genes with the social behaviors. RESULTS: A group of immune genes were differentially expressed in the brains of these mouse strains. The representative C57BL/6 J and DBA/2 J strains displayed significant differences in social behaviors, DBA/2 J mice being less active in social dominance and social interaction than C57BL/6 J mice. The mRNA levels of H2-d1 in the prefrontal cortex, hippocampus, and hypothalamus and C1qb in the hippocampus of the DBA/2 J strain were significantly down-regulated as compared to those in the C57BL/6 J strain. In contrast, Polr3b in the hippocampus and Tnfsf13b in the prefrontal cortex of the DBA/2 J strain were up-regulated. Furthermore, C1qb, Cx3cl1, H2-d1, H2-k1, Polr3b, and Tnfsf13b were predicted to be associated with various behavioral and brain morphological features across the eight inbred strains. Importantly, the C1qb mRNA level was confirmed to be significantly correlated with the sociability in DBA/2 J but not in C57BL/6 J mice. CONCLUSIONS: Our study provided evidence on the association of immune gene network(s) with the brain development and behavior in animals and revealed neurobiological functions of novel brain immune genes that may contribute to social deficiency in animal models of neuropsychiatric disorders.


Asunto(s)
Encéfalo/metabolismo , Citocinas/metabolismo , Regulación de la Expresión Génica/fisiología , Conducta Social , Animales , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Encéfalo/anatomía & histología , Receptor 1 de Quimiocinas CX3C , Citocinas/genética , Perfilación de la Expresión Génica , Antígenos H-2/genética , Antígenos H-2/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Análisis por Micromatrices , Sistemas en Línea , ARN Polimerasa III/genética , ARN Polimerasa III/metabolismo , ARN Mensajero/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Especificidad de la Especie
6.
J Cell Sci ; 124(Pt 12): 2070-6, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21610098

RESUMEN

Owing to its ability to generate the clot-dissolving protease plasmin, tissue plasminogen activator (tPA) is the only approved drug for the acute treatment of ischemic stroke. However, tPA also promotes hemorrhagic transformation and excitotoxic events. High mobility group box-1 protein (HMGB-1) is a non-histone transcription factor and a pro-inflammatory cytokine, which has also been shown to bind to both tPA and plasminogen. We thus investigated the cellular and molecular effects through which HMGB-1 could influence the vascular and parenchymal effects of tPA during ischemia. We demonstrate that HMGB-1 not only increases clot lysis by tPA, but also reduces the passage of vascular tPA across the blood-brain barrier, as well as tPA-driven leakage of the blood-brain barrier. In addition, HMGB-1 prevents the pro-neurotoxic effect of tPA, by blocking its interaction with N-methyl-D-aspartate (NMDA) receptors and the attendant potentiation of NMDA-induced neuronal Ca²âº influx. In conclusion, we show in vitro that HMGB-1 can promote the beneficial effects of tPA while counteracting its deleterious properties. We suggest that derivatives of HMGB-1, devoid of pro-inflammatory properties, could be used as adjunctive therapies to improve the overall benefit of tPA-mediated thrombolysis following stroke.


Asunto(s)
Fibrinólisis/efectos de los fármacos , Proteína HMGB1/farmacología , Activador de Tejido Plasminógeno/farmacología , Animales , Biomarcadores/sangre , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Calcio/metabolismo , Bovinos , Células Cultivadas , Técnicas de Cocultivo , Dominios HMG-Box , Proteína HMGB1/metabolismo , Humanos , Ratones , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Activador de Tejido Plasminógeno/metabolismo
7.
EMBO Rep ; 12(12): 1293-9, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22056818

RESUMEN

Kv2.1 is a potassium channel α-subunit abundantly expressed throughout the brain. It is a main component of delayed rectifier current (I(K)) in several neuronal types and a regulator of excitability during high-frequency firing. Here we identify AMIGO (amphoterin-induced gene and ORF), a neuronal adhesion protein with leucine-rich repeat and immunoglobin domains, as an integral part of the Kv2.1 channel complex. AMIGO shows extensive spatial and temporal colocalization and association with Kv2.1 in the mouse brain. The colocalization of AMIGO and Kv2.1 is retained even during stimulus-induced changes in Kv2.1 localization. AMIGO increases Kv2.1 conductance in a voltage-dependent manner in HEK cells. Accordingly, inhibition of endogenous AMIGO suppresses neuronal I(K) at negative membrane voltages. In conclusion, our data indicate AMIGO as a function-modulating auxiliary subunit for Kv2.1 and thus provide new insights into regulation of neuronal excitability.


Asunto(s)
Proteínas de la Membrana/metabolismo , Subunidades de Proteína/metabolismo , Canales de Potasio Shab/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Células HEK293 , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Activación del Canal Iónico , Ratones , Neuronas/citología , Neuronas/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , Ratas
8.
J Biol Chem ; 286(26): 23200-13, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21527633

RESUMEN

Hmgb1 (high mobility group box-1; amphoterin) is highly expressed in brain during early development of vertebrate and nonvertebrate species. However, its role in brain development remains elusive. Here we have cloned the zebrafish Hmgb1 and specifically manipulated Hmgb1 expression using injection of morpholino antisense oligonucleotides or Hmgb1 cRNA. The HMGB1 knockdown morphants produced by injection of three different morpholino oligonucleotides display a characteristic phenotype with smaller size, smaller brain width, and shorter distance between the eyes. Closer examination of the phenotype reveals severe defects in the development of the forebrain that largely lacks catecholaminergic neural networks. The HMGB1 morphant is deficient in survival and proliferation of neural progenitors and displays fewer cell groups expressing the transcription factor Pax6a in the forebrain and aberrant Wnt8 signaling. The mechanism of HMGB1-dependent progenitor survival involves the neuronal transmembrane protein AMIGO (amphoterin-induced gene and orf), the expression of which is regulated by HMGB1 in vivo. Our data demonstrate that HMGB1 is a critical factor for brain development, enabling survival and proliferation of neural progenitors that will form the forebrain structures.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Proteína HMGB1/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Prosencéfalo/embriología , Proteínas de Pez Cebra/biosíntesis , Pez Cebra/embriología , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteína HMGB1/genética , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Oligodesoxirribonucleótidos Antisentido/farmacología , Prosencéfalo/citología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
9.
J Cell Biol ; 178(4): 687-700, 2007 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-17682049

RESUMEN

Matrix metalloproteinase (MMP)-2 and -9 are pivotal in remodeling many tissues. However, their functions and candidate substrates for brain development are poorly characterized. Intercellular adhesion molecule-5 (ICAM-5; Telencephalin) is a neuronal adhesion molecule that regulates dendritic elongation and spine maturation. We find that ICAM-5 is cleaved from hippocampal neurons when the cells are treated with N-methyl-d-aspartic acid (NMDA) or alpha-amino-3-hydroxy-5-methylisoxazole-propionic acid (AMPA). The cleavage is blocked by MMP-2 and -9 inhibitors and small interfering RNAs. Newborn MMP-2- and MMP-9-deficient mice brains contain more full-length ICAM-5 than wild-type mice. NMDA receptor activation disrupts the actin cytoskeletal association of ICAM-5, which promotes its cleavage. ICAM-5 is mainly located in dendritic filopodia and immature thin spines. MMP inhibitors block the NMDA-induced cleavage of ICAM-5 more efficiently in dendritic shafts than in thin spines. ICAM-5 deficiency causes retraction of thin spine heads in response to NMDA stimulation. Soluble ICAM-5 promotes elongation of dendritic filopodia from wild-type neurons, but not from ICAM-5-deficient neurons. Thus, MMPs are important for ICAM-5-mediated dendritic spine development.


Asunto(s)
Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Hipocampo/citología , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Endogámicos C57BL
10.
Front Cell Dev Biol ; 10: 865275, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35547817

RESUMEN

Protamine is an arginine-rich peptide that replaces histones in the DNA-protein complex during spermatogenesis. Protamine is clinically used in cardiopulmonary bypass surgery to neutralize the effects of heparin that is required during the treatment. Here we demonstrate that protamine and its 14-22 amino acid long fragments overcome the neurite outgrowth inhibition by chondroitin sulfate proteoglycans (CSPGs) that are generally regarded as major inhibitors of regenerative neurite growth after injuries of the adult central nervous system (CNS). Since the full-length protamine was found to have toxic effects on neuronal cells we used the in vitro neurite outgrowth assay to select a protamine fragment that retains the activity to overcome the neurite outgrowth inhibition on CSPG substrate and ended up in the 14 amino acid fragment, low-molecular weight protamine (LMWP). In contrast to the full-length protamine, LMWP displays very low or no toxicity in our assays in vitro and in vivo. We therefore started studies on LMWP as a possible drug lead in treatment of CNS injuries, such as the spinal cord injury (SCI). LMWP mimicks HB-GAM (heparin-binding growth-associated molecule; pleiotrophin) in that it overcomes the CSPG inhibition on neurite outgrowth in primary CNS neurons in vitro and inhibits binding of protein tyrosine phosphatase (PTP) sigma, an inhibitory receptor in neurite outgrowth, to its CSPG ligand. Furthermore, the chondroitin sulfate (CS) chains of the cell matrix even enhance the LMWP-induced neurite outgrowth on CSPG substrate. In vivo studies using the hemisection and hemicontusion SCI models in mice at the cervical level C5 revealed that LMWP enhances recovery when administered through intracerebroventricular or systemic route. We suggest that LMWP is a promising drug lead to develop therapies for CNS injuries.

11.
Biochim Biophys Acta ; 1799(1-2): 164-70, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19914413

RESUMEN

Extracellularly occurring HMGB1, either released during cell injury or actively secreted from cells, has profound effects on behaviour of a wide variety of cell types. Extracellular HMGB1 regulates migratory responses of many cell types, including neuron and growth cone migration, invasive migration of tumour cells, and migration of endothelial and immune cells. RAGE (Receptor for Advanced Glycation End Products) plays a key role as a cell surface receptor in most, if not all HMGB1-dependent migration mechanisms. HMGB1 binds to the distal immunoglobulin-like domain of RAGE, activating a signalling pathway that ends up in modulation of the cytoskeleton for regulation of cell motility. In addition to RAGE, proteoglycans and sulfated carbohydrate epitopes of glycolipids and glycoproteins may play a role as cell surface binding sites of HMGB1, affecting migratory behaviour of cells. In addition to physiological and pathophysiological cell migration control, HMGB1 has been widely studied as a molecule linking tissue injury to inflammatory mechanisms. HMGB1 by itself has little if any proinflammatory activity but it appears to activate innate immunity mechanisms as a complex with DNA, lipids and/or proinflammatory cytokines. The inflammation-inducing activity of HMGB1/DNA complexes may depend on both RAGE and Toll-like receptors of the immune cell surface. In addition to the receptors activating innate immunity, receptors downregulating inflammation upon HMGB1 release have been recently found, and include thrombomodulin and the CD-24/Siglec pathway.


Asunto(s)
Enfermedad , Proteína HMGB1/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Movimiento Celular , Proteína HMGB1/química , Humanos , Inflamación/metabolismo , Inflamación/patología , Proteoglicanos/metabolismo
12.
J Cell Biol ; 174(4): 569-80, 2006 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-16908672

RESUMEN

N-syndecan (syndecan-3) is a transmembrane proteoglycan that is abundantly expressed in the major axonal pathways and in the migratory routes of the developing brain. When ligated by heparin-binding (HB) growth-associated molecule (GAM; pleiotrophin), N-syndecan mediates cortactin-Src kinase-dependent neurite outgrowth. However, the functional role of N-syndecan in brain development remains unexplored. In this study, we show that N-syndecan deficiency perturbs the laminar structure of the cerebral cortex as a result of impaired radial migration. In addition, neural migration in the rostral migratory stream is impaired in the N-syndecan-null mice. We suggest that the migration defect depends on impaired HB-GAM-induced Src kinase activation and haptotactic migration. Furthermore, we show that N-syndecan interacts with EGF receptor (EGFR) at the plasma membrane and is required in EGFR-induced neuronal migration.


Asunto(s)
Movimiento Celular/fisiología , Corteza Cerebral/anomalías , Corteza Cerebral/metabolismo , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Neuronas/metabolismo , Proteoglicanos/deficiencia , Proteoglicanos/genética , Células Madre/metabolismo , Animales , Proteínas Portadoras/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Membrana Celular/metabolismo , Movimiento Celular/genética , Proliferación Celular , Corteza Cerebral/citología , Citocinas/metabolismo , Receptores ErbB/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Ratones , Ratones Noqueados , Neuritas/metabolismo , Neuritas/ultraestructura , Neuronas/citología , Técnicas de Cultivo de Órganos , Células Madre/citología , Sindecano-3 , Familia-src Quinasas/metabolismo
13.
Front Neurol ; 12: 738800, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34938257

RESUMEN

Heparin-binding growth-associated molecule (pleiotrophin) is a neurite outgrowth-promoting secretory protein that lines developing fiber tracts in juvenile CNS (central nervous system). Previously, we have shown that heparin-binding growth-associated molecule (HB-GAM) reverses the CSPG (chondroitin sulfate proteoglycan) inhibition on neurite outgrowth in the culture medium of primary CNS neurons and enhances axon growth through the injured spinal cord in mice demonstrated by two-photon imaging. In this study, we have started studies on the possible role of HB-GAM in enhancing functional recovery after incomplete spinal cord injury (SCI) using cervical lateral hemisection and hemicontusion mouse models. In vivo imaging of blood-oxygen-level-dependent (BOLD) signals associated with functional activity in the somatosensory cortex was used to assess the sensory functions during vibrotactile hind paw stimulation. The signal displays an exaggerated response in animals with lateral hemisection that recovers to the level seen in the sham-operated mice by injection of HB-GAM to the trauma site. The effect of HB-GAM treatment on sensory-motor functions was assessed by performance in demanding behavioral tests requiring integration of afferent and efferent signaling with central coordination. Administration of HB-GAM either by direct injection into the trauma site or by intrathecal injection improves the climbing abilities in animals with cervical hemisection and in addition enhances the grip strength in animals with lateral hemicontusion without affecting the spontaneous locomotor activity. Recovery of sensory signaling in the sensorimotor cortex by HB-GAM to the level of sham-operated mice may contribute to the improvement of skilled locomotion requiring integration of spatiotemporal signals in the somatosensory cortex.

14.
Mol Neurobiol ; 58(3): 1145-1161, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33099743

RESUMEN

A striking result from epidemiological studies show a correlation between low alcohol intake and lower incidence for ischemic stroke and severity of derived brain injury. Although reduced apoptosis and inflammation has been suggested to be involved, little is known about the mechanism mediating this effect in vivo. Increase in intracellular chloride concentration and derived depolarizing GABAAR-mediated transmission are common consequences following various brain injuries and are caused by the abnormal expression levels of the chloride cotransporters NKCC1 and KCC2. Downstream pro-apoptotic signaling through p75NTR may link GABAA depolarization with post-injury neuronal apoptosis. Here, we show that changes in GABAergic signaling, Cl- homeostasis, and expression of chloride cotransporters in the post-traumatic mouse brain can be significantly reduced by administration of 3% ethanol to the drinking water. Ethanol-induced upregulation of KCC2 has a positive impact on neuronal survival, preserving a large part of the cortical peri-infarct zone, as well as preventing the massive post-ischemic upregulation of the pro-apoptotic protein p75NTR. Importantly, intracortical multisite in vivo recordings showed that ethanol treatment could significantly ameliorate stroke-induced reduction in cortical activity. This surprising finding discloses a pathway triggered by low concentration of ethanol as a novel therapeutically relevant target.


Asunto(s)
Etanol/administración & dosificación , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Receptores de Factor de Crecimiento Nervioso/metabolismo , Simportadores/metabolismo , Animales , Apoptosis/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Biomarcadores/metabolismo , Infarto Encefálico/complicaciones , Infarto Encefálico/patología , Infarto Encefálico/fisiopatología , Supervivencia Celular/efectos de los fármacos , Cloruros/metabolismo , Dieta , Fenómenos Electrofisiológicos/efectos de los fármacos , Inflamación/complicaciones , Inflamación/patología , Inflamación/fisiopatología , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Recuperación de la Función/efectos de los fármacos , Factores de Tiempo , Ácido gamma-Aminobutírico/metabolismo , Cotransportadores de K Cl
15.
Cells ; 9(7)2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32708917

RESUMEN

The High Mobility Group Box 1 (HMGB1) is the most abundant nuclear nonhistone protein that is involved in transcription regulation. In addition, HMGB1 has previously been found as an extracellularly acting protein enhancing neurite outgrowth in cultured neurons. Although HMGB1 is widely expressed in the developing central nervous system of vertebrates and invertebrates, its function in the developing mouse brain is poorly understood. Here, we have analyzed developmental defects of the HMGB1 null mouse forebrain, and further examined our findings in ex vivo brain cell cultures. We find that HMGB1 is required for the proliferation and differentiation of neuronal stem cells/progenitor cells. Enhanced apoptosis is also found in the neuronal cells lacking HMGB1. Moreover, HMGB1 depletion disrupts Wnt/ß-catenin signaling and the expression of transcription factors in the developing cortex, including Foxg1, Tbr2, Emx2, and Lhx6. Finally, HMGB1 null mice display aberrant expression of CXCL12/CXCR4 and reduced RAGE signaling. In conclusion, HMGB1 plays a critical role in mammalian neurogenesis and brain development.


Asunto(s)
Encéfalo/metabolismo , Proteína HMGB1/metabolismo , Neurogénesis , Animales , Apoptosis , Encéfalo/crecimiento & desarrollo , Proliferación Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Regulación hacia Abajo , Embrión de Mamíferos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Receptores CXCR4/metabolismo , Factores de Transcripción/metabolismo
16.
Cells ; 9(2)2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-32074971

RESUMEN

The protein kinase JNK1 exhibits high activity in the developing brain, where it regulates dendrite morphology through the phosphorylation of cytoskeletal regulatory proteins. JNK1 also phosphorylates dendritic spine proteins, and Jnk1-/- mice display a long-term depression deficit. Whether JNK1 or other JNKs regulate spine morphology is thus of interest. Here, we characterize dendritic spine morphology in hippocampus of mice lacking Jnk1-/- using Lucifer yellow labelling. We find that mushroom spines decrease and thin spines increase in apical dendrites of CA3 pyramidal neurons with no spine changes in basal dendrites or in CA1. Consistent with this spine deficit, Jnk1-/- mice display impaired acquisition learning in the Morris water maze. In hippocampal cultures, we show that cytosolic but not nuclear JNK, regulates spine morphology and expression of phosphomimicry variants of JNK substrates doublecortin (DCX) or myristoylated alanine-rich C kinase substrate-like protein-1 (MARCKSL1), rescue mushroom, thin, and stubby spines differentially. These data suggest that physiologically active JNK controls the equilibrium between mushroom, thin, and stubby spines via phosphorylation of distinct substrates.


Asunto(s)
Espinas Dendríticas/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas , Animales , Proteína Doblecortina , Humanos , Ratones , Prueba del Laberinto Acuático de Morris , Transfección
17.
J Cell Biol ; 160(6): 963-73, 2003 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-12629050

RESUMEN

Ordered differential display identified a novel sequence induced in neurons by the neurite-promoting protein amphoterin. We named this gene amphoterin-induced gene and ORF (AMIGO), and also cloned two other novel genes homologous to AMIGO (AMIGO2 and AMIGO3). Together, these three AMIGOs form a novel family of genes coding for type I transmembrane proteins which contain a signal sequence for secretion and a transmembrane domain. The deduced extracellular parts of the AMIGOs contain six leucine-rich repeats (LRRs) flanked by cysteine-rich LRR NH2- and COOH-terminal domains and by one immunoglobulin domain close to the transmembrane region. A substrate-bound form of the recombinant AMIGO ectodomain promoted prominent neurite extension in hippocampal neurons, and in solution, the same AMIGO ectodomain inhibited fasciculation of neurites. A homophilic and heterophilic binding mechanism is shown between the members of the AMIGO family. Our results suggest that the members of the AMIGO protein family are novel cell adhesion molecules among which AMIGO is specifically expressed on fiber tracts of neuronal tissues and participates in their formation.


Asunto(s)
Membrana Celular/metabolismo , Conos de Crecimiento/metabolismo , Hipocampo/metabolismo , Proteínas de la Membrana/aislamiento & purificación , Proteínas de la Membrana/metabolismo , Moléculas de Adhesión de Célula Nerviosa/aislamiento & purificación , Vías Nerviosas/metabolismo , Animales , Membrana Celular/ultraestructura , Extensiones de la Superficie Celular/genética , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/ultraestructura , Células Cultivadas , ADN Complementario/análisis , ADN Complementario/genética , Feto , Conos de Crecimiento/ultraestructura , Proteína HMGB1 , Hipocampo/citología , Hipocampo/embriología , Inmunohistoquímica , Leucina/genética , Leucina/metabolismo , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Moléculas de Adhesión de Célula Nerviosa/genética , Vías Nerviosas/citología , Vías Nerviosas/embriología , Estructura Terciaria de Proteína/fisiología , Ratas , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico
18.
J Mol Histol ; 50(3): 203-216, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30903543

RESUMEN

Perineuronal net (PNN) is a highly structured portion of the CNS extracellular matrix (ECM) regulating synaptic plasticity and a range of pathologic conditions including posttraumatic regeneration and epilepsy. Here we studied Wisteria floribunda agglutinin-stained histological sections to quantify the PNN size and enrichment of chondroitin sulfates in mouse brain and spinal cord. Somatosensory cortex sections were examined during the period of PNN establishment at postnatal days 14, 21 and 28. The single cell PNN size and the chondroitin sulfate intensity were quantified for all cortex layers and specifically for the cortical layer IV which has the highest density of PNN-positive neurons. We demonstrate that the chondroitin sulfate proteoglycan staining intensity is increased between P14 and P28 while the PNN size remains unchanged. We then addressed posttraumatic changes of the PNN expression in laminae 6 and 7 of cervical spinal cord following hemisection injury. We demonstrate increase of the chondroitin sulfate content at 1.6-1.8 mm rostrally from the injury site and increase of the density of PNN-bearing cells at 0.4-1.2 mm caudally from the injury site. We further demonstrate decrease of the single cell PNN area at 0.2 mm caudally from the injury site suggesting that the PNN ECM takes part in the posttraumatic tissue rearrangement in the spinal cord. Our results demonstrate new insights on the PNN structure dynamics in the developing and posttraumatic CNS.


Asunto(s)
Encéfalo/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Plasticidad Neuronal/genética , Neuronas/metabolismo , Animales , Encéfalo/patología , Matriz Extracelular/metabolismo , Ratones , Neuronas/patología , Lectinas de Plantas/química , Lectinas de Plantas/farmacología , Receptores N-Acetilglucosamina/química , Médula Espinal/metabolismo , Médula Espinal/patología
19.
Liver Transpl ; 14(10): 1517-25, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18825712

RESUMEN

High mobility group box 1 protein (HMGB1), a cytokine actively secreted by phagocytes and passively released from necrotic cells, is an inflammatory mediator in experimental hepatic ischemia/reperfusion injury. We characterized its expression in human liver transplantation. In 20 patients, in addition to systemic samples, blood was drawn from portal and hepatic veins during and after reperfusion to assess changes within the graft. Plasma HMGB1, tumor necrosis factor alpha (TNF-alpha), and interleukin-6 (IL-6) levels were measured, and HMGB1 immunohistochemistry was performed on biopsies taken before and after reperfusion. Plasma HMGB1 was undetectable before reperfusion, and levels in systemic circulation peaked after graft reperfusion. At portal declamping, HMGB1 levels were substantially higher in the caval effluent [188 (80-371) ng/mL] than in portal venous blood [0 (0-3) ng/mL, P < 0.001]. HMGB1 release from the graft continued thereafter. HMGB1 levels were not related to TNF-alpha or IL-6 levels. HMGB1 expression was up-regulated in biopsies taken after reperfusion (P = 0.020), with intense hepatocyte and weak neutrophil staining. HMGB1 levels in hepatic venous blood correlated with graft steatosis (r = 0.497, P = 0.03) and peak postoperative alanine aminotransferase levels (r = 0.588, P = 0.008). Our results indicate that HMGB1 originates from the graft and is a marker of hepatocellular injury in human liver transplantation.


Asunto(s)
Proteína HMGB1/metabolismo , Trasplante de Hígado , Hígado/metabolismo , Trasplantes , Adulto , Anciano , Biomarcadores/sangre , Femenino , Humanos , Inmunohistoquímica , Interleucina-6/sangre , Hígado/lesiones , Masculino , Persona de Mediana Edad , Factor de Necrosis Tumoral alfa/sangre , Adulto Joven
20.
J Leukoc Biol ; 81(1): 49-58, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16980512

RESUMEN

HMGB1 (amphoterin) is a 30-kDa heparin-binding protein that mediates transendothelial migration of monocytes and has proinflammatory cytokine-like activities. In this study, we have investigated proinflammatory activities of both highly purified eukaryotic HMGB1 and bacterially produced recombinant HMGB1 proteins. Mass analyses revealed that recombinant eukaryotic HMGB1 has an intrachain disulphide bond. In mass analysis of tissue-derived HMGB1, two forms were detected: the carboxyl terminal glutamic acid residue lacking form and a full-length form. Cell culture studies indicated that both eukaryotic and bacterial HMGB1 proteins induce TNF-alpha secretion and nitric oxide release from mononuclear cells. Affinity chromatography analysis revealed that HMGB1 binds tightly to proinflammatory bacterial substances. A soluble proinflammatory substance was separated from the bacterial recombinant HMGB1 by chloroform-methanol treatment. HMGB1 interacted with phosphatidylserine in both solid-phase binding and cell culture assays, suggesting that HMGB1 may regulate phosphatidylserine-dependent immune reactions. In conclusion, HMGB1 polypeptide has a weak proinflammatory activity by itself, and it binds to bacterial substances, including lipids, that may strengthen its effects.


Asunto(s)
Proteína HMGB1/farmacología , Inflamación/tratamiento farmacológico , Macrófagos/inmunología , Animales , Baculoviridae/genética , Línea Celular , Escherichia coli/genética , Proteína HMGB1/metabolismo , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Óxido Nítrico/metabolismo , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA