Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Hazard Mater ; 455: 131555, 2023 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-37156042

RESUMEN

Volatile organic compounds (VOCs) are gaseous chemicals found in ambient air and exhaled breath. In particular, highly reactive aldehydes are frequently found in polluted air and have been linked to various diseases. Thus, extensive studies have been carried out to elucidate disease-specific aldehydes released from the body to develop potential biomarkers for diagnostic purposes. Mammals possess innate sensory systems, such as receptors and ion channels, to detect these VOCs and maintain physiological homeostasis. Recently, electronic biosensors such as the electronic nose have been developed for disease diagnosis. This review aims to present an overview of natural sensory receptors that can detect reactive aldehydes, as well as electronic noses that have the potential to diagnose certain diseases. In this regard, this review focuses on eight aldehydes that are well-defined as biomarkers in human health and disease. It offers insights into the biological aspects and technological advances in detecting aldehyde-containing VOCs. Therefore, this review will aid in understanding the role of aldehyde-containing VOCs in human health and disease and the technological advances for improved diagnosis.


Asunto(s)
Receptores Odorantes , Compuestos Orgánicos Volátiles , Animales , Humanos , Odorantes , Nariz Electrónica , Aldehídos , Pruebas Respiratorias , Biomarcadores , Mamíferos
2.
Cell Death Differ ; 29(6): 1199-1210, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34974535

RESUMEN

Ninjurin1 (Ninj1), an adhesion molecule, regulates macrophage function in hyaloid regression, multiple sclerosis, and atherosclerosis. However, its biological relevance and the mechanism underlying its function in vascular network integrity have not been studied. In this study, we investigated the role of Ninj1 in physiological (postnatal vessel formation) and pathological (endotoxin-mediated inflammation and diabetes) conditions and developed a strategy to regulate Ninj1 using specific micro (mi)RNAs under pathological conditions. Ninj1-deficient mice exhibited decreased hyaloid regression, tip cell formation, retinal vascularized area, recruitment of macrophages, and endothelial apoptosis during postnatal development, resulting in delayed formation of the vascular network. Five putative miRNAs targeting Ninj1 were selected using the miRanda algorithm and comparison of expression patterns. Among them, miR-125a-5p showed a profound inhibitory effect on Ninj1 expression, and miR-125a-5p mimic suppressed the cell-to-cell and cell-to-matrix adhesion of macrophages and expression of pro-inflammatory factors mediated by Ninj1. Furthermore, miR-125a-5p mimic inhibited the recruitment of macrophages into inflamed retinas in endotoxin-induced inflammation and streptozotocin-induced diabetes in vivo. In particular, miR-125a-5p mimic significantly attenuated vascular leakage in diabetic retinopathy. Taken together, these findings suggest that Ninj1 plays a pivotal role in macrophage-mediated vascular integrity and that miR-125a-5p acts as a novel regulator of Ninj1 in the management of inflammatory diseases and diabetic retinopathy.


Asunto(s)
Moléculas de Adhesión Celular Neuronal , Retinopatía Diabética , MicroARNs , Factores de Crecimiento Nervioso , Animales , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Endotoxinas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo
3.
Int J Mol Sci ; 24(1)2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-36614120

RESUMEN

Electroceuticals refer to various forms of electronic neurostimulators used for therapy. Interdisciplinary advances in medical engineering and science have led to the development of the electroceutical approach, which involves therapeutic agents that specifically target neural circuits, to realize precision therapy for Alzheimer's disease (AD). To date, extensive studies have attempted to elucidate the disease-modifying effects of electroceuticals on areas in the brain of a patient with AD by the use of various physical stimuli, including electric, magnetic, and electromagnetic waves as well as ultrasound. Herein, we review non-invasive stimulatory systems and their effects on ß-amyloid plaques and tau tangles, which are pathological molecular markers of AD. Therefore, this review will aid in better understanding the recent technological developments, applicable methods, and therapeutic effects of electronic stimulatory systems, including transcranial direct current stimulation, 40-Hz gamma oscillations, transcranial magnetic stimulation, electromagnetic field stimulation, infrared light stimulation and ionizing radiation therapy, and focused ultrasound for AD.


Asunto(s)
Enfermedad de Alzheimer , Estimulación Transcraneal de Corriente Directa , Humanos , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Proteínas tau , Placa Amiloide/patología , Ovillos Neurofibrilares/patología
4.
Nat Commun ; 10(1): 5306, 2019 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-31757960

RESUMEN

After stroke, peripheral immune cells are activated and these systemic responses may amplify brain damage, but how the injured brain sends out signals to trigger systemic inflammation remains unclear. Here we show that a brain-to-cervical lymph node (CLN) pathway is involved. In rats subjected to focal cerebral ischemia, lymphatic endothelial cells proliferate and macrophages are rapidly activated in CLNs within 24 h, in part via VEGF-C/VEGFR3 signalling. Microarray analyses of isolated lymphatic endothelium from CLNs of ischemic mice confirm the activation of transmembrane tyrosine kinase pathways. Blockade of VEGFR3 reduces lymphatic endothelial activation, decreases pro-inflammatory macrophages, and reduces brain infarction. In vitro, VEGF-C/VEGFR3 signalling in lymphatic endothelial cells enhances inflammatory responses in co-cultured macrophages. Lastly, surgical removal of CLNs in mice significantly reduces infarction after focal cerebral ischemia. These findings suggest that modulating the brain-to-CLN pathway may offer therapeutic opportunities to ameliorate systemic inflammation and brain injury after stroke.


Asunto(s)
Infarto Encefálico/inmunología , Isquemia Encefálica/inmunología , Encéfalo/inmunología , Endotelio Linfático/inmunología , Ganglios Linfáticos/inmunología , Macrófagos/inmunología , Factor C de Crecimiento Endotelial Vascular/inmunología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/inmunología , Animales , Encéfalo/metabolismo , Infarto Encefálico/metabolismo , Isquemia Encefálica/metabolismo , Proliferación Celular , Células Endoteliales , Endotelio Linfático/metabolismo , Inflamación , Ganglios Linfáticos/metabolismo , Linfangiogénesis , Ratones , Cuello , Ratas , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Stroke ; 49(12): 3039-3049, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30571410

RESUMEN

Background and Purpose- The complexity and heterogeneity of stroke, as well as the associated comorbidities, may render neuroprotective drugs less efficacious in clinical practice. Therefore, the development of targeted therapies to specific patient subsets has become a high priority in translational stroke research. Ischemic stroke with type 2 diabetes mellitus has a nearly double mortality rate and worse neurological outcomes. In the present study, we tested our hypothesis that rFGF21 (recombinant human fibroblast growth factor 21) administration is beneficial for improving neurological outcomes of ischemic stroke with type 2 diabetes mellitus. Methods- Type 2 diabetes mellitus db/db and nondiabetic genetic control db/+ mice were subjected into permanent focal ischemia of distal middle cerebral artery occlusion, we examined the effects of poststroke administration with rFGF21 in systemic metabolic disorders, inflammatory gatekeeper PPARγ (peroxisome proliferator-activated receptor γ) activity at 3 days, mRNA expression of inflammatory cytokines and microglia/macrophage activation at 7 days in the perilesion cortex, and last neurological function deficits, ischemic brain infarction, and white matter integrity up to 14 days after stroke of db/db mice. Results- After permanent focal ischemia, diabetic db/db mice presented confounding pathological features, including metabolic dysregulation, more severe brain damage, and neurological impairment, especially aggravated proinflammatory response and white matter integrity loss. However, daily rFGF21 treatment initiated at 6 hours after stroke for 14 days significantly normalized systemic metabolic disorders, rescued PPARγ activity decline, inhibited proinflammatory cytokine mRNA expression, and M1-like microglia/macrophage activation in the brain. Importantly, rFGF21 also significantly reduced white matter integrity loss, ischemic brain infarction, and neurological function deficits up to 14 days after stroke. The potential mechanisms of rFGF21 may in part consist of potent systematic metabolic regulation and PPARγ-activation promotion-associated antiproinflammatory roles in the brain. Conclusions- Taken together, these results suggest rFGF21 might be a novel and potent candidate of the disease-modifying strategy for treating ischemic stroke with type 2 diabetes mellitus.


Asunto(s)
Isquemia Encefálica , Encéfalo/efectos de los fármacos , Citocinas/efectos de los fármacos , Diabetes Mellitus Tipo 2 , Factores de Crecimiento de Fibroblastos/farmacología , PPAR gamma/efectos de los fármacos , Accidente Cerebrovascular , Animales , Encéfalo/metabolismo , Encéfalo/patología , Citocinas/genética , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media , Macrófagos/efectos de los fármacos , Masculino , Ratones , Microglía/efectos de los fármacos , PPAR gamma/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Proteínas Recombinantes , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/patología
6.
J Neurochem ; 146(2): 160-172, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29570780

RESUMEN

Ischemic postconditioning is increasingly being investigated as a therapeutic approach for cerebral ischemia. However, the majority of studies are focused on the acute protection of neurons per se. Whether and how postconditioning affects multiple cells in the recovering neurovascular unit remains to be fully elucidated. Here, we asked whether postconditioning may modulate help-me signaling between injured neurons and reactive microglia. Rats were subjected to 100 min of focal cerebral ischemia, then randomized into a control versus postconditioning group. After 3 days of reperfusion, infarct volumes were significantly reduced in animals treated with postconditioning, along with better neurologic outcomes. Immunostaining revealed that ischemic postconditioning increased expression of vascular endothelial growth factor (VEGF) in neurons within peri-infarct regions. Correspondingly, we confirmed that VEGFR2 was expressed on Iba1-positive microglia/macrophages, and confocal microscopy showed that in postconditioned rats, these cells were polarized to a ramified morphology with higher expression of M2-like markers. Treating rats with a VEGF receptor 2 kinase inhibitor negated these effects of postconditioning on microglia/macrophage polarization. In vitro, postconditoning after oxygen-glucose deprivation up-regulated VEGF release in primary neuron cultures, and adding VEGF to microglial cultures partly shifted their M2-like markers. Altogether, our findings support the idea that after postconditioning, injured neurons may release VEGF as a 'help-me' signal that promotes microglia/macrophage polarization into potentially beneficial phenotypes.


Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Polaridad Celular/fisiología , Poscondicionamiento Isquémico/métodos , Microglía/patología , Neuronas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Infarto Encefálico/etiología , Proteínas de Unión al Calcio/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Embrión de Mamíferos , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucosa/deficiencia , Infusiones Intraventriculares , Masculino , Proteínas de Microfilamentos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
Stem Cells ; 36(5): 751-760, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29314444

RESUMEN

Oligodendrocyte precursor cells (OPCs) give rise to oligodendrocytes in cerebral white matter. However, the underlying mechanisms that regulate this process remain to be fully defined, especially in adult brains. Recently, it has been suggested that signaling via A-kinase anchor protein 12 (AKAP12), a scaffolding protein that associates with intracellular molecules such as protein kinase A, may be involved in Schwann cell homeostasis and peripheral myelination. Here, we asked whether AKAP12 also regulates the mechanisms of myelination in the CNS. AKAP12 knockout mice were compared against wild-type (WT) mice in a series of neurochemical and behavioral assays. Compared with WTs, 2-months old AKAP12 knockout mice exhibited loss of myelin in white matter of the corpus callosum, along with perturbations in working memory as measured by a standard Y-maze test. Unexpectedly, very few OPCs expressed AKAP12 in the corpus callosum region. Instead, pericytes appeared to be one of the major AKAP12-expressing cells. In a cell culture model system, conditioned culture media from normal pericytes promoted in-vitro OPC maturation. However, conditioned media from AKAP12-deficient pericytes did not support the OPC function. These findings suggest that AKAP12 signaling in pericytes may be required for OPC-to-oligodendrocyte renewal to maintain the white matter homeostasis in adult brain. Stem Cells 2018;36:751-760.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/fisiología , Células-Madre Neurales/citología , Oligodendroglía/metabolismo , Sustancia Blanca/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Envejecimiento , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular/fisiología , Células Cultivadas , Medios de Cultivo Condicionados , Ratones Noqueados , Vaina de Mielina/metabolismo , Neurogénesis/fisiología , Oligodendroglía/citología , Sustancia Blanca/citología
8.
Mol Neurobiol ; 54(9): 7353-7368, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-27815839

RESUMEN

Over the last few decades, molecular neurobiology has uncovered many genes whose deficiency in mice results in behavioral traits associated with human neuropsychiatric disorders such as autism, obsessive-compulsive disorder (OCD), and schizophrenia. However, the etiology of these common diseases remains enigmatic with the potential involvement of a battery of genes. Here, we report abnormal behavioral phenotypes of mice deficient in a cell adhesion molecule Ninjurin 1 (Ninj1), which are relevant to repetitive and anxiety behaviors of neuropsychiatric disorders. Ninj1 knockout (KO) mice exhibit compulsive grooming-induced hair loss and self-made lesions as well as increased anxiety-like behaviors. Histological analysis reveals that Ninj1 is predominantly expressed in cortico-thalamic circuits, and neuron-specific Ninj1 conditional KO mice manifest aberrant phenotypes similar to the global Ninj1 KO mice. Notably, the brains of Ninj1 KO mice display altered synaptic transmission in thalamic neurons as well as a reduced number of functional synapses. Moreover, the disruption of Ninj1 leads to glutamatergic abnormalities, including increased ionotropic glutamate receptors but reduced glutamate levels. Furthermore, chronic treatment with fluoxetine, a drug reportedly ameliorates compulsive behaviors in mice, prevents progression of hair loss and alleviates the compulsive grooming and anxiety-like behavior of Ninj1 KO mice. Collectively, our results suggest that Ninj1 could be involved in neuropsychiatric disorders associated with impairments of repetitive and anxiety behaviors.


Asunto(s)
Ansiedad/genética , Ansiedad/metabolismo , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/genética , Conducta Compulsiva/genética , Conducta Compulsiva/metabolismo , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/genética , Animales , Ansiedad/psicología , Células Cultivadas , Conducta Compulsiva/psicología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Int J Oncol ; 48(2): 821-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26677008

RESUMEN

Ninjurin1 is a transmembrane protein involved in macrophage migration and adhesion during inflammation. It was recently reported that repression of Ninjurin1 attenuated the lipopolysaccharide (LPS)-induced inflammatory response in macrophages; however, the precise mechanism by which Ninjurin1 modulates LPS-induced inflammation remains poorly understood. In the present study, we found that the interaction between Ninjurin1 and LPS contributed to the LPS-induced inflammatory response. Notably, pull-down assays using lysates from HEK293T cells transfected with human or mouse Ninjurin1 and biotinylated LPS (LPS-biotin) showed that LPS directly bound Ninjurin1. Subsequently, LPS binding assays with various truncated forms of Ninjurin1 protein revealed that amino acids (aa) 81-100 of Ninjurin1 were required for LPS binding. In addition, knockdown experiments using Ninj1 siRNA resulted in decreased nitric oxide (NO) and tumor necrosis factor-α (TNFα) secretion upon LPS treatment in Raw264.7 cells. Collectively, our results suggest that Ninjurin1 regulates the LPS-induced inflammatory response through its direct binding to LPS, thus, identifying Ninjurin1 as a putative target for the treatment of inflammatory diseases, such as sepsis and inflammation-associated carcinogenesis.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Factores de Crecimiento Nervioso/metabolismo , Aminoácidos/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/fisiología , Células HEK293 , Humanos , Macrófagos/metabolismo , Ratones , Óxido Nítrico/metabolismo , Unión Proteica/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
10.
Mol Med Rep ; 12(2): 1817-23, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25873382

RESUMEN

Reactive gliosis is a glial response to a wide range of central nervous system insults, which results in cellular and molecular changes to resting glial cells. Despite its fundamental effect on neuropathologies, the identification and characterization of the molecular mechanisms underlying this process remain to be fully elucidated. The aim of the present study was to analyze the expression profile and functions of the astrocytic neurotrophic factor, meteorin, in the progression of reactive gliosis. A mouse model of photothrombotic ischemia, and a primary astrocyte culture were used in the present study. Reverse transcription quantitative polymerase chain reaction, western blotting and immunofluorescence staining were performed to examine the expression levels of meteorin and reactive gliosis markers. Increased expression levels of meteorin were observed in reactive astrocytes in a photothrombotic ischemia mouse model, as well as in cultured astrocytes, which were stimulated by transforming growth factor-ß1. Exogenous treatment of the astrocytes with meteorin did not induce janus kinase-signal transducer and activator of transcription 3 signaling, however, silencing the expression of meteorin in the astrocytes resulted in an upregulation of reactive astrocyte markers, including glial fibrillary acidic protein and S100ß, indicating that endogenous meteorin is required for the maintenance of astrocytic homeostasis. These results suggested a novel role for meteorin as a negative feedback effector in reactive gliosis.


Asunto(s)
Astrocitos/metabolismo , Gliosis , Proteínas del Tejido Nervioso/metabolismo , Animales , Astrocitos/citología , Células Cultivadas , Modelos Animales de Enfermedad , Inmunohistoquímica , Isquemia/inducido químicamente , Isquemia/metabolismo , Isquemia/patología , Luz , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Fármacos Fotosensibilizantes/toxicidad , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba/efectos de los fármacos
11.
Int J Oncol ; 46(1): 99-106, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25338643

RESUMEN

ARD1 is an acetyltransferase with several variants derived from alternative splicing. Among ARD1 variants, mouse ARD1(225) (mARD1(225)), mouse ARD1(235) (mARD1(235)), and human ARD1(235) (hARD1(235)) have been the most extensively characterized and are known to have different biological functions. In the present study, we demonstrated that mARD1(225), mARD1(235), and hARD1(235) have conserved autoacetylation activities, and that they selectively regulate distinct roles of ARD1 variants in tumorigenesis. Using purified recombinants for ARD1 variants, we found that mARD1(225), mARD1(235), and hARD1(235) undergo similar autoacetylation with the target site conserved at the Lys136 residue. Moreover, functional investigations revealed that the role of mARD1(225) autoacetylation is completely distinguishable from that of mARD1(235) and hARD1(235). Under hypoxic conditions, mARD1(225) autoacetylation inhibited tumor angiogenesis by decreasing the stability of hypoxia-inducible factor-1α (HIF-1α). Autoacetylation stimulated the catalytic activity of mARD1(225) to acetylate Lys532 of the oxygen-dependent degradation (ODD) domain of HIF-1α, leading to the proteosomal degradation of HIF-1α. In contrast, autoacetylation of mARD1(235) and hARD1(235) contributed to cellular growth under normoxic conditions by increasing the expression of cyclin D1. Taken together, these data suggest that autoacetylation of ARD1 variants differentially regulates angiogenesis and cell proliferation in an isoform-specific manner.


Asunto(s)
Carcinogénesis/genética , Acetiltransferasa A N-Terminal/genética , Acetiltransferasa A N-Terminal/metabolismo , Acetiltransferasa E N-Terminal/genética , Acetiltransferasa E N-Terminal/metabolismo , Polimorfismo de Nucleótido Simple , Acetilación , Secuencia de Aminoácidos , Carcinogénesis/metabolismo , Proliferación Celular/genética , Células Cultivadas , Retroalimentación Fisiológica , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Datos de Secuencia Molecular , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Isoformas de Proteínas/genética , Estabilidad Proteica
12.
Nat Commun ; 5: 4952, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25229625

RESUMEN

The meninges forms a critical epithelial barrier, which protects the central nervous system (CNS), and therefore its prompt reconstruction after CNS injury is essential for reducing neuronal damage. Meningeal cells migrate into the lesion site after undergoing an epithelial-mesenchymal transition (EMT) and repair the impaired meninges. However, the molecular mechanisms of meningeal EMT remain largely undefined. Here we show that TGF-ß1 and retinoic acid (RA) released from the meninges, together with oxygen tension, could constitute the mechanism for rapid meningeal reconstruction. AKAP12 is an effector of this mechanism, and its expression in meningeal cells is regulated by integrated upstream signals composed of TGF-ß1, RA and oxygen tension. Functionally, AKAP12 modulates meningeal EMT by regulating the TGF-ß1-non-Smad-SNAI1 signalling pathway. Collectively, TGF-ß1, RA and oxygen tension can modulate the dynamic change in AKAP12 expression, causing prompt meningeal reconstruction after CNS injury by regulating the transition between the epithelial and mesenchymal states of meningeal cells.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Sistema Nervioso Central/lesiones , Regulación de la Expresión Génica , Meninges/metabolismo , Oxígeno/química , Animales , Aracnoides/metabolismo , Encéfalo/metabolismo , Línea Celular Tumoral , Células Epiteliales/citología , Transición Epitelial-Mesenquimal , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Receptores de Ácido Retinoico/metabolismo , Epitelio Pigmentado de la Retina/citología , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Tretinoina/metabolismo
13.
PLoS One ; 9(8): e105185, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25133627

RESUMEN

Arrest defective 1 (ARD1) is an acetyltransferase that is highly conserved across organisms, from yeasts to humans. The high homology and widespread expression of ARD1 across multiple species and tissues signify that it serves a fundamental role in cells. Human ARD1 (hARD1) has been suggested to be involved in diverse biological processes, and its role in cell proliferation and cancer development has been recently drawing attention. However, the subcellular localization of ARD1 and its relevance to cellular function remain largely unknown. Here, we have demonstrated that hARD1 is imported to the nuclei of proliferating cells, especially during S phase. Nuclear localization signal (NLS)-deleted hARD1 (hARD1ΔN), which can no longer access the nucleus, resulted in cell morphology changes and cellular growth impairment. Notably, hARD1ΔN-expressing cells showed alterations in the cell cycle and the expression levels of cell cycle regulators compared to hARD1 wild-type cells. Furthermore, these effects were rescued when the nuclear import of hARD1 was restored by exogenous NLS. Our results show that hARD1 nuclear translocation mediated by NLS is required for cell cycle progression, thereby contributing to proper cell proliferation.


Asunto(s)
Núcleo Celular/metabolismo , Acetiltransferasa A N-Terminal/metabolismo , Acetiltransferasa E N-Terminal/metabolismo , Señales de Localización Nuclear/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiología , Línea Celular , Células HEK293 , Células HeLa , Humanos , Acetiltransferasa A N-Terminal/genética , Acetiltransferasa E N-Terminal/genética , Señales de Localización Nuclear/genética , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Fase S/genética , Fase S/fisiología
14.
J Biol Chem ; 289(32): 21926-36, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-24917672

RESUMEN

Ninjurin1 is involved in the pathogenesis of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, by mediating leukocyte extravasation, a process that depends on homotypic binding. However, the precise regulatory mechanisms of Ninjurin1 during inflammation are largely undefined. We therefore examined the pro-migratory function of Ninjurin1 and its regulatory mechanisms in macrophages. Interestingly, Ninjurin1-deficient bone marrow-derived macrophages exhibited reduced membrane protrusion formation and dynamics, resulting in the impairment of cell motility. Furthermore, exogenous Ninjurin1 was distributed at the membrane of filopodial structures in Raw264.7 macrophage cells. In Raw264.7 cells, RNA interference of Ninjurin1 reduced the number of filopodial projections, whereas overexpression of Ninjurin1 facilitated their formation and thus promoted cell motility. Ninjurin1-induced filopodial protrusion formation required the activation of Rac1. In Raw264.7 cells penetrating an MBEC4 endothelial cell monolayer, Ninjurin1 was localized to the membrane of protrusions and promoted their formation, suggesting that Ninjurin1-induced protrusive activity contributed to transendothelial migration. Taking these data together, we conclude that Ninjurin1 enhances macrophage motility and consequent extravasation of immune cells through the regulation of protrusive membrane dynamics. We expect these findings to provide insight into the understanding of immune responses mediated by Ninjurin1.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Movimiento Celular/fisiología , Macrófagos/fisiología , Factores de Crecimiento Nervioso/fisiología , Animales , Adhesión Celular/fisiología , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular , Membrana Celular/fisiología , Células Cultivadas , Células Endoteliales/fisiología , Técnicas de Silenciamiento del Gen , Inflamación/etiología , Inflamación/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/genética , Neuropéptidos/metabolismo , Seudópodos/fisiología , Interferencia de ARN , Proteína de Unión al GTP rac1/metabolismo
15.
PLoS One ; 9(4): e94695, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24760034

RESUMEN

The repair process after CNS injury shows a well-organized cascade of three distinct stages: inflammation, new tissue formation, and remodeling. In the new tissue formation stage, various cells migrate and form the fibrotic scar surrounding the lesion site. The fibrotic scar is known as an obstacle for axonal regeneration in the remodeling stage. However, the role of the fibrotic scar in the new tissue formation stage remains largely unknown. We found that the number of A-kinase anchoring protein 12 (AKAP12)-positive cells in the fibrotic scar was increased over time, and the cells formed a structure which traps various immune cells. Furthermore, the AKAP12-positive cells strongly express junction proteins which enable the structure to function as a physical barrier. In in vivo validation, AKAP12 knock-out (KO) mice showed leakage from a lesion, resulting from an impaired structure with the loss of the junction complex. Consistently, focal brain injury in the AKAP12 KO mice led to extended inflammation and more severe tissue damage compared to the wild type (WT) mice. Accordingly, our results suggest that AKAP12-positive cells in the fibrotic scar may restrict excessive inflammation, demonstrating certain mechanisms that could underlie the beneficial actions of the fibrotic scar in the new tissue formation stage during the CNS repair process.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Sistema Nervioso Central/metabolismo , Fibrosis/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Animales , Western Blotting , Proteínas de Ciclo Celular/genética , Fibrosis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas Wistar , Cicatrización de Heridas/genética , Cicatrización de Heridas/fisiología
16.
J Biol Chem ; 289(6): 3328-38, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-24347169

RESUMEN

Ninjurin1 is a homotypic adhesion molecule that contributes to leukocyte trafficking in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. However, in vivo gene deficiency animal studies have not yet been done. Here, we constructed Ninjurin1 knock-out (KO) mice and investigated the role of Ninjurin1 on leukocyte trafficking under inflammation conditions such as EAE and endotoxin-induced uveitis. Ninjurin1 KO mice attenuated EAE susceptibility by reducing leukocyte recruitment into the injury regions of the spinal cord and showed less adhesion of leukocytes on inflamed retinal vessels in endotoxin-induced uveitis mice. Moreover, the administration of a custom-made antibody (Ab26-37) targeting the Ninjurin1 binding domain ameliorated the EAE symptoms, showing the contribution of its adhesion activity to leukocyte trafficking. In addition, we addressed the transendothelial migration (TEM) activity of bone marrow-derived macrophages and Raw264.7 cells according to the expression level of Ninjurin1. TEM activity was decreased in Ninjurin1 KO bone marrow-derived macrophages and siNinj1 Raw264.7 cells. Consistent with this, GFP-tagged mNinj1-overexpressing Raw264.7 cells increased their TEM activity. Taken together, we have clarified the contribution of Ninjurin1 to leukocyte trafficking in vivo and delineated its direct functions to TEM, emphasizing Ninjurin1 as a beneficial therapeutic target against inflammatory diseases such as multiple sclerosis.


Asunto(s)
Células de la Médula Ósea/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Encefalomielitis Autoinmune Experimental/metabolismo , Macrófagos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Células de la Médula Ósea/patología , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular , Susceptibilidad a Enfermedades , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Macrófagos/patología , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/genética
17.
Biochem Biophys Res Commun ; 428(4): 438-44, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23142597

RESUMEN

Ninjurin1 is known as an adhesion molecule promoting leukocyte trafficking under inflammatory conditions. However, the posttranslational modifications of Ninjurin1 are poorly understood. Herein, we defined the proteolytic cleavage of Ninjurin1 and its functions. HEK293T cells overexpressing the C- or N-terminus tagging mouse Ninjurin1 plasmid produced additional cleaved forms of Ninjurin1 in the lysates or conditioned media (CM). Two custom-made anti-Ninjurin1 antibodies, Ab(1-15) or Ab(139-152), specific to the N- or C-terminal regions of Ninjurin1 revealed the presence of its shedding fragments in the mouse liver and kidney lysates. Furthermore, Matrix Metalloproteinase (MMP) 9 was responsible for Ninjurin1 cleavage between Leu(56) and Leu(57). Interestingly, the soluble N-terminal Ninjurin1 fragment has structural similarity with well-known chemokines. Indeed, the CM from HEK293T cells overexpressing the GFP-mNinj1 plasmid was able to attract Raw264.7 cells in trans-well assay. Collectively, we suggest that the N-terminal ectodomain of mouse Ninjurin1, which may act as a chemoattractant, is cleaved by MMP9.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/química , Factores Quimiotácticos/química , Metaloproteinasa 9 de la Matriz/química , Factores de Crecimiento Nervioso/química , Secuencia de Aminoácidos , Animales , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Quimiocinas/química , Quimiocinas/genética , Quimiocinas/metabolismo , Factores Quimiotácticos/genética , Factores Quimiotácticos/metabolismo , Células HEK293 , Humanos , Riñón/metabolismo , Leucina/química , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
18.
Mol Cells ; 29(3): 223-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20119872

RESUMEN

Nerve injury induced protein 1, Ninj1 (Ninjurin1) is a cell surface protein that is induced by nerve injury and promotes axonal growth in the peripheral nervous system. However, the function of Ninj1 in the vascular system and central nervous system (CNS) is incompletely understood. Here we review recent studies that have shed further light on the role and regulation of Ninj1 in vascular remodeling and inflammation. Increasing evidence suggests that Ninj1 mediates cell communication and enhances the entry, migration, and activity of leukocytes such as monocytes and macrophages in developmental processes and inflammatory responses. Moreover, our recent studies show that Ninj1 regulates close interaction between leukocytes and vascular endothelial cells in vascular remodeling and inflamed CNS. Additionally, Ninj1 enhances the apoptosis-inducing activity of leukocytes and is cleaved by MMPs, resulting in loss of adhesion during tissue remodeling. The collective data described here show that Ninj1 is required for the entry, adhesion, activation, and movement of leukocytes during tissue remodeling and might be a potential therapeutic target to regulate the adhesion and trafficking of leukocytes in inflammation and leukocyte-mediated diseases such as multiple sclerosis, diabetic retinopathy, and neuropathy.


Asunto(s)
Apoptosis/fisiología , Moléculas de Adhesión Celular Neuronal/fisiología , Adhesión Celular/fisiología , Inflamación/fisiopatología , Factores de Crecimiento Nervioso/fisiología , Regeneración Nerviosa/fisiología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Vasos Sanguíneos/citología , Moléculas de Adhesión Celular Neuronal/química , Comunicación Celular/fisiología , Movimiento Celular/fisiología , Sistemas de Liberación de Medicamentos , Células Endoteliales/citología , Humanos , Inflamación/tratamiento farmacológico , Leucocitos/citología , Metaloproteinasas de la Matriz/fisiología , Factores de Crecimiento Nervioso/química , Traumatismos de los Nervios Periféricos , Estructura Terciaria de Proteína , Relación Estructura-Actividad
19.
Biochem Biophys Res Commun ; 387(2): 321-5, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19595672

RESUMEN

Ninjurin1 (nerve injury-induced protein, Ninj1) is an adhesion molecule that is essential for cell-to-cell interactions. However, little is known about the function of Ninj1 in the central nervous system (CNS). To address its role in the CNS, we analyzed the expression pattern of Ninj1 in normal rats and in an experimental autoimmune encephalomyelitis (EAE) model. Ninj1 was expressed in three major compartments of brains, meninges, the choroid plexus, and parenchymal perivascular spaces. In the EAE brains, Ninj1 was strongly expressed in myeloid cells (macrophages/monocytes and neutrophils) and partially expressed in endothelial cells (ECs). Furthermore, Ninj1 enhanced adhesion between BV2 cells (murine monocyte lineage microglia) and HBMECs (human brain microvascular endothelial cells). Collectively, our findings suggest that Ninj1 may mediate the entry of myeloid cells into the CNS in normal and EAE brains, and it is a potential therapeutic target for regulating myeloid cell trafficking across the blood-brain barrier (BBB) in CNS immune processes.


Asunto(s)
Encéfalo/patología , Moléculas de Adhesión Celular Neuronal/biosíntesis , Encefalomielitis Autoinmune Experimental/patología , Células Mieloides/patología , Factores de Crecimiento Nervioso/biosíntesis , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Adhesión Celular , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Endotelio/inmunología , Endotelio/metabolismo , Endotelio/patología , Monocitos/inmunología , Monocitos/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Ratas , Ratas Endogámicas Lew , Regulación hacia Arriba
20.
Biophys J ; 94(10): 4095-102, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18227131

RESUMEN

The distribution and movement of elemental ions in biologic tissues is critical for many cellular processes. In contrast to chemical techniques for imaging the intracellular distribution of ions, however, techniques for imaging the distribution of ions across tissues are not well developed. We used time-of-flight secondary ion mass spectrometry (TOF-SIMS) to obtain nonlabeled high-resolution analytic images of ion distribution in ischemic retinal tissues. Marked changes in Ca(2+) distribution, compared with other fundamental ions, such as Na(+), K(+), and Mg(2+), were detected during the progression of ischemia. Furthermore, the Ca(2+) redistribution pattern correlated closely with TUNEL-positive (positive for terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate nick end-labeling) cell death in ischemic retinas. After treatment with a calcium chelator, Ca(2+) ion redistribution was delayed, resulting in a decrease in TUNEL-positive cells. These results indicate that ischemia-induced Ca(2+) redistribution within retinal tissues is associated with the order of apoptotic cell death, which possibly explains the different susceptibility of various types of retinal cells to ischemia. Thus, the TOF-SIMS technique provides a tool for the study of intercellular communication by Ca(2+) ion movement.


Asunto(s)
Calcio/metabolismo , Isquemia/metabolismo , Vasos Retinianos/metabolismo , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Animales , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Vasos Retinianos/patología , Coloración y Etiquetado , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA