Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Pediatr Diabetes ; 18(2): 95-102, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-26843101

RESUMEN

BACKGROUND AND OBJECTIVE: Matrix metalloproteinases (MMPs) mediate blood-brain barrier dysfunction in inflammatory disease states. Our objective was to compare circulating MMPs in children with diabetic ketoacidosis (DKA) to children with type 1 diabetes mellitus without DKA. RESEARCH DESIGN AND METHODS: This was a prospective study performed at five tertiary-care pediatric hospitals. We measured plasma MMP-2, MMP-3, and MMP-9 early during DKA (time 1; within 2 h of beginning intravenous fluids) and during therapy (time 2; median 8 h; range: 4-16 h). The primary outcome was MMP levels in 34 children with DKA vs. 23 children with type 1 diabetes without DKA. Secondary outcomes included correlations between MMPs and measures of DKA severity. RESULTS: In children with DKA compared with diabetes controls, circulating MMP-2 levels were lower (mean 77 vs. 244 ng/mL, p < 0.001), MMP-3 levels were similar (mean 5 vs. 4 ng/mL, p = 0.57), and MMP-9 levels were higher (mean 67 vs. 25 ng/mL, p = 0.002) early in DKA treatment. MMP-2 levels were correlated with pH at time 1 (r = 0.45, p = 0.018) and time 2 (r = 0.47, p = 0.015) and with initial serum bicarbonate at time 2 (r = 0.5, p = 0.008). MMP-9 levels correlated with hemoglobin A1c in DKA and diabetes controls, but remained significantly elevated in DKA after controlling for hemoglobin A1c (ß = -31.3, p = 0.04). CONCLUSIONS: Circulating MMP-2 levels are lower and MMP-9 levels are higher in children during DKA compared with levels in children with diabetes without DKA. Alterations in MMP expression could mediate BBB dysfunction occurring during DKA.


Asunto(s)
Diabetes Mellitus Tipo 1/sangre , Cetoacidosis Diabética/sangre , Metaloproteinasas de la Matriz/sangre , Adolescente , Estudios de Casos y Controles , Niño , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/terapia , Cetoacidosis Diabética/terapia , Femenino , Fluidoterapia/métodos , Hemoglobina Glucada/metabolismo , Humanos , Masculino , Índice de Severidad de la Enfermedad
2.
Neurotrauma Rep ; 3(1): 207-216, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35734394

RESUMEN

Traumatic brain injury (TBI) exacts significant neurological and financial costs on patients and their families. In adult patients with moderate-to-severe TBI, central auditory impairments have been reported. These auditory impairments may interfere with language receptivity, as observed in children with developmental brain injury. Although rodent models of TBI have been widely used to examine behavioral outcomes, few studies have evaluated how TBI affects higher-order central auditory processing across a range of cue complexities. Here, auditory processing was assessed using a modified acoustic startle paradigm. We used a battery of progressively complex stimuli (single-tone, silent gaps in white noise, and frequency-modulated [FM] sweeps) in adult rats that received unilateral controlled cortical impact injury. TBI subjects showed significant reductions in acoustic startle absolute responses across nearly all stimuli, regardless of cue, duration of stimuli, or cue complexity. Despite this overall reduction of startle magnitudes in injured animals, the detection of single-tone stimuli was comparable between TBI and sham-injured subjects, indicating intact hearing after TBI. TBI subjects showed deficits in rapid gap (5 ms) and FM sweep (175 ms) detection, and, in contrast to shams, they did not improve on detecting silent gaps and FM sweeps across days of testing. Our findings provide evidence for both low-level (brainstem-mediated) and higher-order central auditory processing deficits in a rodent model of TBI, which parallel sensory impairments observed in TBI patients. The present findings support the use of modified pre-pule auditory detection paradigms to investigate clinically relevant processes in TBI.

3.
J Neurotrauma ; 38(4): 385-398, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-32940130

RESUMEN

Neuroinflammation and dysfunction of the blood-brain barrier (BBB) are two prominent mechanisms of secondary injury in neurotrauma. It has been suggested that Toll-like receptors (TLRs) play important roles in initiating and propagating neuroinflammation resulting from traumatic brain injury (TBI), but potential beneficial effects of targeting these receptors in TBI have not been broadly studied. Here, we investigated the effect of targeting TLRs with proteoglycan 4 (PRG4) on post-traumatic neuroinflammation and BBB function. PRG4 is a mucinous glycoprotein with strong anti-inflammatory properties, exerting its biological effects by interfering with TLR2/4 signaling. In addition, PRG4 has the ability to inhibit activation of cluster of differentiation 44 (CD44), a cell-surface glycoprotein playing an important role in inflammation. Using the controlled cortical impact model of TBI in rats, we showed a rapid and prolonged upregulation of message for TLR2/4 and CD44 in the injured cortex. In the in vitro model of the BBB, recombinant human PRG4 (rhPRG4) crossed the endothelial monolayers through a high-capacity, saturable transport system. In rats sustaining TBI, PRG4 delivery to the brain was enhanced by post-traumatic increase in BBB permeability. rhPRG4 injected intravenously at 1 h post-TBI potently inhibited post-traumatic activation of nuclear factor kappa B and extracellular signal-regulated kinases 1/2, the two major signal transduction pathways associated with TLR2/4 and CD44, and curtailed the post-traumatic influx of monocytes. In addition, PRG4 restored normal BBB function after TBI by preventing the post-traumatic loss of tight junction protein claudin 5 and reduced neuronal death. Our observations provide support for therapeutic strategies targeting TLRs in TBI.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/complicaciones , Encefalitis/tratamiento farmacológico , Proteoglicanos/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Muerte Celular/efectos de los fármacos , Encefalitis/etiología , Encefalitis/metabolismo , Encefalitis/patología , Masculino , Modelos Animales , FN-kappa B/metabolismo , Proteoglicanos/uso terapéutico , Ratas , Ratas Long-Evans , Transducción de Señal/efectos de los fármacos
4.
Fluids Barriers CNS ; 16(1): 41, 2019 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-31856859

RESUMEN

This article highlights the scientific achievements, professional career, and personal interactions of Malcolm B. Segal who passed away in July this year. Born in 1937 in Goodmayes, Essex, UK, Segal rose to the Chairman position in the Division of Physiology at United Medical and Dental School of Guy's and St. Thomas' Hospitals, retiring in 2006 after his long professional career in biomedical science. Being trained in Hugh Davson's laboratory, Segal became one of the pioneers in research on cerebrospinal fluid physiology and the choroid plexus. During the course of his career, Segal himself trained a number of young scientists and collaborated with many colleagues around the world, making long-lasting friendships along the way. In addition to his professional accomplishments as a researcher and educator, Segal was an avid sailor and wine connoisseur, and enjoyed teaching classes on navigation and wine tasting.


Asunto(s)
Líquido Cefalorraquídeo/fisiología , Plexo Coroideo/fisiología , Fisiología/historia , Historia del Siglo XX , Historia del Siglo XXI
5.
BMC Neurosci ; 9: 97, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18834514

RESUMEN

BACKGROUND: GPR125 belongs to the family of Adhesion G protein-coupled receptors (GPCRs). A single copy of GPR125 was found in many vertebrate genomes. We also identified a Drosophila sequence, DmCG15744, which shares a common ancestor with the entire Group III of Adhesion GPCRs, and also contains Ig, LRR and HBD domains which were observed in mammalian GPR125. RESULTS: We found specific expression of GPR125 in cells of the choroid plexus using in situ hybridization and protein-specific antibodies and combined in situ/immunohistochemistry co-localization using cytokeratin, a marker specific for epithelial cells. Induction of inflammation by LPS did not change GPR125 expression. However, GPR125 expression was transiently increased (almost 2-fold) at 4 h after traumatic brain injury (TBI) followed by a decrease (approximately 4-fold) from 2 days onwards in the choroid plexus as well as increased expression (2-fold) in the hippocampus that was delayed until 1 day after injury. CONCLUSION: These findings suggest that GPR125 plays a functional role in choroidal and hippocampal response to injury.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Plexo Coroideo/patología , Proteínas de la Membrana/genética , Receptores Acoplados a Proteínas G/genética , Animales , Secuencia de Bases , Lesiones Encefálicas/inducido químicamente , Plexo Coroideo/metabolismo , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inmunohistoquímica , Hibridación in Situ , Inflamación/inducido químicamente , Inflamación/fisiopatología , Queratinas/genética , Queratinas/metabolismo , Lipopolisacáridos , Masculino , Glicoproteínas de Membrana , Proteínas de la Membrana/metabolismo , Ratones , Complejo GPIb-IX de Glicoproteína Plaquetaria , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Factores de Tiempo , Regulación hacia Arriba/genética
6.
Cerebrospinal Fluid Res ; 4: 11, 2007 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-18162136

RESUMEN

BACKGROUND: There is an increasing interest in using choroid plexus (CP) epithelial cell lines to study the properties of the blood-cerebrospinal fluid barrier (BCSFB). Currently, there are three major CP-derived cell lines available. Z310 and TR-CSFB3, two immortalized cell lines carrying the simian virus 40 large T-antigen gene, were derived from rat CP epithelium, whereas the CPC-2 cell line was derived from human CP carcinoma. Although these cell lines have previously been used in various functional studies, the expression of adherens junction (AJ) and tight junction (TJ) proteins in these epithelial cells has not been systematically studied. Accordingly, in the present study, we sought to characterize the expression of these junctional proteins in these three cell lines. METHODS: The cells were grown in six-well cell culture plates. Reverse-transcriptase polymerase chain reaction, Western blotting, and immunocytochemistry were used to characterize the expression of AJ and TJ proteins in the CP cell lines. RESULTS: Z310 and TR-CSFB3 cells expressed a TJ protein, occludin, and its cytosolic binding partner, zonula occludens 1, as well as an AJ protein, E-cadherin, and beta-catenin, a cytoplasmic protein that interacts with E-cadherin. However, the expression of occludin and E-cadherin in TR-CSFB3 cells at both the mRNA and protein level was weaker than that found in Z301 cells. The immunocytochemical analysis also demonstrated that the staining pattern for these junctional proteins in TR-CSFB3 cells was discontinuous and the staining intensity was weaker than that observed in Z310 cells. The message for claudin 1 and claudin 2 was expressed at low levels in TR-CSFB3 cells and these cells were weakly immunopositive for claudin 1. In comparison, the message for these TJ proteins could not be detected in Z310 cells. CPC-2 cells expressed occludin, which was localized to areas of cell-cell contact, but the staining pattern for this TJ protein was found to be variable and irregular. Although CPC-2 cells expressed mRNA for claudin 1, claudin 2, and claudin 11, only claudin 1 was expressed at the protein level and it was localized to the nuclei rather than to areas of cell-cell contact. An AJ protein, E-cadherin, was also found to be mislocalized in CPC-2 cells, even though its cytosolic binding partner, beta-catenin, was restricted to areas of cell-cell contact, as in normal CP. CONCLUSION: The three CP cell lines analyzed in this study vary considerably with regard to the expression of AJ and TJ proteins, which is likely reflected by different barrier properties of these in vitro models of BCSFB.

7.
Sci Rep ; 7(1): 10995, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28887487

RESUMEN

Brain-derived neurotrophic factor (BDNF), a key player in regulating synaptic strength and learning, is dysregulated following traumatic brain injury (TBI), suggesting that stimulation of BDNF signaling pathways may facilitate functional recovery. This study investigates whether CN2097, a peptidomimetic ligand which targets the synaptic scaffold protein, postsynaptic density protein 95, to enhance downstream signaling of tropomyosin-related kinase B, a receptor for BDNF, can improve neurological function after TBI. Moderate to severe TBI elicits neuroinflammation and c-Jun-N-terminal kinase (JNK) activation, which is associated with memory deficits. Here we demonstrate that CN2097 significantly reduces the post-traumatic synthesis of proinflammatory mediators and inhibits the post-traumatic activation of JNK in a rodent model of TBI. The recordings of field excitatory post-synaptic potentials in the hippocampal CA1 subfield demonstrate that TBI inhibits the expression of long-term potentiation (LTP) evoked by high-frequency stimulation of Schaffer collaterals, and that CN2097 attenuates this LTP impairment. Lastly, we demonstrate that CN2097 significantly improves the complex auditory processing deficits, which are impaired after injury. The multifunctionality of CN2097 strongly suggests that CN2097 could be highly efficacious in targeting complex secondary injury processes resulting from neurotrauma.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/rehabilitación , Receptor trkB/metabolismo , Animales , Conducta Animal , Biomarcadores , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Aprendizaje por Laberinto , Péptidos Cíclicos/farmacología , Ratas , Recuperación de la Función
8.
PLoS One ; 11(12): e0167677, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28030563

RESUMEN

The pathophysiological mechanisms underlying mild traumatic brain injury (mTBI) are not well understood, but likely involve neuroinflammation. Here the controlled cortical impact model of mTBI in rats was used to test this hypothesis. Mild TBI caused a rapid (within 6 h post-mTBI) upregulation of synthesis of TNF-α and IL-1ß in the cerebral cortex and hippocampus, followed by an increase in production of neutrophil (CXCL1-3) and monocyte (CCL2) chemoattractants. While astrocytes were not a significant source of CXC chemokines, they highly expressed CCL2. An increase in production of CXC chemokines coincided with the influx of neutrophils into the injured brain. At 6 h post-mTBI, we observed a robust influx of CCL2-expressing neutrophils across pial microvessels into the subarachnoid space (SAS) near the injury site. Mild TBI was not accompanied by any significant influx of neutrophils into the brain parenchyma until 24 h after injury. This was associated with an early induction of expression of intercellular adhesion molecule 1 on the endothelium of the ipsilateral pial, but not intraparenchymal, microvessels. At 6 h post-mTBI, we also observed a robust influx of neutrophils into the ipsilateral cistern of velum interpositum (CVI), a slit-shaped cerebrospinal fluid space located above the 3rd ventricle with highly vascularized pia mater. From SAS and CVI, neutrophils appeared to move along the perivascular spaces to enter the brain parenchyma. The monocyte influx was not observed until 24 h post-mTBI, and these inflammatory cells predominantly entered the ipsilateral SAS and CVI, with a limited invasion of brain parenchyma. These observations indicate that the endothelium of pial microvessels responds to injury differently than that of intraparenchymal microvessels, which may be associated with the lack of astrocytic ensheathment of cerebrovascular endothelium in pial microvessels. These findings also suggest that neuroinflammation represents the potential therapeutic target in mTBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/fisiopatología , Leucocitos/inmunología , Microvasos/inmunología , Piamadre/irrigación sanguínea , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Mediadores de Inflamación/metabolismo , Leucocitos/metabolismo , Masculino , Microvasos/metabolismo , Infiltración Neutrófila , Piamadre/inmunología , Ratas
9.
J Neurotrauma ; 33(1): 49-57, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25794137

RESUMEN

No routine tests currently exist to objectively diagnose mild traumatic brain injury (mTBI)/concussion. Previously reported biomarkers for mTBI represented proteins released from damaged neurons or glia. However, low levels of these proteins, and/or the complexity of assays used for their detection, limits implementation of these biomarkers in routine practice. Here, we sought to identify proteins whose synthesis is altered post-mTBI and whose blood levels could be measured using standard immunoassays. Adult patients sustaining a concussion within the past 24 h were enrolled. Controls were uninjured subjects and patients with orthopedic injury (OI). Four candidate biomarkers were identified: copeptin; galectin 3 (LGALS3); matrix metalloproteinase 9 (MMP9); and occludin (OCLN). A 3.4-fold decrease (p<0.0001) in plasma concentration of copeptin was found in mTBI patients within 8 h after accident, compared to uninjured controls. Plasma levels of LGALS3, MMP9, and OCLN increased 3.6- to 4.5-fold (p<0.0001) within the same time frame postinjury. Levels of at least two biomarkers were altered beyond their respective cut-off values in 90% of mTBI patients, whereas in none of uninjured controls were levels of two biomarkers simultaneously changed. A positive correlation (r=0.681; p<0.001) between plasma levels of LGALS3 and OCLN was also found in mTBI patients, whereas in OI patients or uninjured subjects, these variables did not correlate. This panel of biomarkers discerns, with high accuracy, patients with isolated concussion from uninjured individuals within the first 8 h after accident. These biomarkers can also aid in diagnosing concussion in the presence of OI.


Asunto(s)
Biomarcadores/sangre , Conmoción Encefálica/sangre , Galectina 3/sangre , Glicopéptidos/sangre , Metaloproteinasa 9 de la Matriz/sangre , Ocludina/sangre , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad
10.
J Neurotrauma ; 22(8): 919-28, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16083358

RESUMEN

In a number of stress conditions, the biological effects of tumor necrosis factor-alpha (TNF-alpha), such as the induction of neuronal apoptosis, are presumably attenuated by the soluble fragments of TNF receptors (sTNFRs). Within 1 h after spinal cord injury, increased synthesis and/or secretion of TNF-alpha is detectable at the injury site. However, the shedding of ectodomains of TNFRs in the traumatized spinal cord has not yet been reported. In the present study, adult Sprague-Dawley rats were subjected to acute spinal cord injury (ASCI) by applying a 25-g Walsh-Tator aneurysm clip at the C8-T1 level. Sham-injured animals underwent laminectomy and facetectomy only. A PE10 catheter was placed in the subarachnoid space to collect the samples of cerebrospinal fluid (CSF) from near the injury site. These CSF samples were analyzed by ELISA for the presence of TNF-alpha and soluble TNFR1 and TNFR2 (sTNFR1 and sTNFR2, respectively). The spinal cord tissue was analyzed by immunohistochemistry for the expression of TNF-alpha, TNFR1, and TNFR2, and by the TUNEL technique for the occurrence of neuronal death. The levels of TNFR1 and sTNFR1 in the injured tissue were determined by Western blotting. Immunohistochemistry demonstrated the increased neuronal expression of TNF-alpha and its receptors at 6 h post-ASCI. No changes in the intensity of staining were observed in the sham-injured rats. In addition, at 6 h after the injury, a significant increase in the number of TUNEL-positive neurons was observed. Numerous neurons in traumatized tissue were also immunoreactive for activated caspase-3, suggesting that the TUNEL-positive neurons were undergoing an apoptotic death. At 1 h after ASCI, TNF-alpha levels in the CSF were significantly higher than those found in the sham-injured animals, indicating the release of this cytokine into the interstitial fluid. This was followed by a significant increase, compared to the sham-injured controls, in sTNFR1 levels in the CSF at 3 and 6 h after the insult. Unlike sTNFR1, the levels of sTNFR2 in the CSF were unchanged at any time point post-ASCI. The increased shedding of TNFR1 was confirmed by Western blotting. It is concluded that the shedding of TNFR1 receptor may represent an important post-traumatic physiological response aimed at reducing the proapoptotic effect of TNF-alpha.


Asunto(s)
Apoptosis/fisiología , Degeneración Nerviosa/fisiopatología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Traumatismos de la Médula Espinal/líquido cefalorraquídeo , Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Caspasa 3 , Caspasas/metabolismo , Líquido Cefalorraquídeo/química , Líquido Cefalorraquídeo/metabolismo , Modelos Animales de Enfermedad , Femenino , Degeneración Nerviosa/líquido cefalorraquídeo , Degeneración Nerviosa/inmunología , Ratas , Ratas Sprague-Dawley , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Médula Espinal/inmunología , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/fisiopatología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/líquido cefalorraquídeo , Regulación hacia Arriba/fisiología
11.
Fluids Barriers CNS ; 12: 1, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25745556

RESUMEN

In June 2014 Dr. Joseph D. Fenstermacher celebrated his 80th birthday, which was honored by the symposium held in New London, NH, USA. This review discusses Fenstermacher's contribution to the field of fluids and barriers of the CNS. Specifically, his fundamental work on diffusion of molecules within the brain extracellular space and the research on properties of the blood-brain barrier in health and disease are described. Fenstermacher's early research on cerebrospinal fluid dynamics and the regulation of cerebral blood flow is also reviewed, followed by the discussion of his more recent work involving the use of magnetic resonance imaging.

12.
J Neurotrauma ; 21(8): 1090-102, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15319008

RESUMEN

Experimental evidence obtained in various animal models of brain injury indicates that vasopressin promotes the formation of cerebral edema. However, the molecular and cellular mechanisms underlying this vasopressin action are not fully understood. In the present study, we analyzed the temporal changes in expression of vasopressin V1a receptors after traumatic brain injury (TBI) in rats. In the intact brain, the V1a receptor was expressed in neurons located in all layers of the frontoparietal cortex. The V1a receptor-immunoreactive product was predominantly localized to neuronal nuclei and had both a diffused and punctate staining pattern. The V1a receptors were also expressed in astrocytes, especially in layer 1 of the frontoparietal cortex. In these cells, two distinctive patterns of immunopositive staining for V1a receptors were observed: a diffused cytosolic staining of cell bodies and processes and a clearly punctate staining pattern that was predominantly localized to the astrocytic cell bodies. The real-time reverse-transcriptase polymerase chain reaction analysis of changes in mRNA for the V1a receptor demonstrated that after TBI, there is an early (4 h post-TBI) increase in the number of transcripts in the ipsilateral frontoparietal cortex, when compared to the contralateral hemisphere or the sham-injured rats. This increase in the message was followed by the up-regulation of expression of the V1a receptors at the protein level. This was most evident in cortical astrocytes in the areas surrounding the lesion. The number of the V1a receptor-immunopositive astrocytes in the traumatized parenchyma gradually increased, starting at 8 h and peaking at 4-6 days after TBI. Furthermore, a redistribution of V1a receptors from the astrocytic cell bodies to the astrocytic processes was observed. In addition to astrocytes, an increased expression of V1a receptors was found in the endothelium of both blood microvessels and the large-diameter blood vessels in the frontoparietal cortex ipsilateral to injury. This increase in the V1a receptor expression was apparent between 2 and 4 days after TBI. As early as 1-2 h following the impact, there was also a striking increase in the number of the V1a receptor-immunopositive beaded axonal processes, with greatly enlarged varicosities, that were localized to various areas of the injured parenchyma. It is suggested that the increased expression of V1a receptors plays an important role in the vasopressin-mediated formation of edema in the injured brain.


Asunto(s)
Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Encéfalo/metabolismo , Receptores de Vasopresinas/metabolismo , Vasopresinas/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Axones/metabolismo , Axones/patología , Barrera Hematoencefálica/fisiología , Encéfalo/patología , Encéfalo/fisiopatología , Edema Encefálico/etiología , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Masculino , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Neuronas/metabolismo , Neuronas/patología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Vasopresinas/genética , Regulación hacia Arriba/genética
13.
Neuroreport ; 13(2): 257-9, 2002 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-11893921

RESUMEN

Increasing evidence indicates that basic fibroblast growth factor (FGF2), a well-known mitogen, can regulate the synthesis and secretion of peptide hormones. FGF2 has also been recently shown to inhibit cerebrospinal fluid (CSF) formation and increase the number of dark choroid plexus epithelial cells. These latter FGF2 actions could be mediated by vasopressin (VP) synthesized by and released from choroidal epithelium. The present study was therefore designed to determine whether VP co-localizes with fibroblast growth factor receptors (FGFRs) in the choroid plexus epithelium. Using confocal laser-scanning microscopy, we demonstrated the apical (CSF-facing) distribution of FGFRs in epithelial cells. FGFRs were expressed on clusters of VP-positive cells, with the intensity of FGFR-immunopositive staining varying from one group of cells to the other. These observations are in line with the possible mediatory role of VP in the FGF2-dependent inhibition of CSF secretion and the induction of dark epithelial cells.


Asunto(s)
Plexo Coroideo/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Vasopresinas/metabolismo , Animales , Epitelio/metabolismo , Inmunohistoquímica , Ratas , Distribución Tisular
14.
R I Med J (2013) ; 97(5): 22-6, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24791263

RESUMEN

This article provides an overview of the Brown University Traumatic Brain Injury Research Consortium (TBIRC) and summarizes the multidisciplinary basic and clinical neuroscience work being conducted by investigators at Brown University and the affiliate hospitals in association with the Norman Prince Neurosciences Institute (NPNI).


Asunto(s)
Lesiones Encefálicas , Universidades , Academias e Institutos , Humanos , Neurociencias , Investigación
15.
PLoS One ; 8(11): e79328, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24223928

RESUMEN

Several lines of evidence indicate that the blood-cerebrospinal fluid barrier (BCSFB), which primarily resides in the choroid plexus (CP), plays a significant pathophysiological role not only in neuroinflammatory diseases, such as multiple sclerosis, but also in traumatic brain injury (TBI). Here we investigated how arginine vasopressin (AVP) regulates function of the BCSFB in the context of post-traumatic neuroinflammation. It has previously been shown that AVP exacerbates various forms of brain injury, but the mechanisms underlying this AVP action are poorly understood. Type 1A AVP receptor is highly expressed on the CP epithelium and the CP synthesizes AVP. Using the controlled cortical impact model of TBI, we demonstrated decreased post-traumatic production of proinflammatory mediators by the CP and reduced influx of inflammatory cells across the BCSFB in AVP-deficient Brattleboro rats when compared with Long-Evans rats, a parental strain for Brattleboro rats. Arginine vasopressin was also found to play an important role in post-traumatic activation of c-Jun N-terminal kinase (JNK) in the CP. In the CP epithelial cell cultures, AVP augmented the tumor necrosis factor-α- and interleukin-1ß-dependent increase in synthesis of proinflammatory mediators, including neutrophil chemoattractants, an action largely dependent on the JNK signaling pathway. Under in vivo conditions, a selective JNK inhibitor decreased the post-traumatic production of neutrophil chemoattractants by the CP and reduced the influx of neutrophils across the BCSFB. These results provide evidence for the synergistic interactions between proinflammatory cytokines and AVP, a ligand for G protein-coupled receptors, and support a pathophysiological role of AVP in post-traumatic neuroinflammation.


Asunto(s)
Arginina Vasopresina/farmacología , Lesiones Encefálicas/metabolismo , Quimiocinas/biosíntesis , Plexo Coroideo/efectos de los fármacos , Interleucina-1beta/farmacología , Neutrófilos/citología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Lesiones Encefálicas/sangre , Lesiones Encefálicas/inmunología , Plexo Coroideo/inmunología , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Neutrófilos/efectos de los fármacos , Ratas
16.
J Cereb Blood Flow Metab ; 32(1): 93-104, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21829211

RESUMEN

The invasion of inflammatory cells occurring after ischemic or traumatic brain injury (TBI) has a detrimental effect on neuronal survival and functional recovery after injury. We have recently demonstrated that not only the blood-brain barrier, but also the blood-cerebrospinal fluid (CSF) barrier (BCSFB), has a role in posttraumatic recruitment of neutrophils. Here, we show that TBI results in a rapid increase in synthesis and release into the CSF of a major chemoattractant for monocytes, CCL2, by the choroid plexus epithelium, a site of the BCSFB. Using an in vitro model of the BCSFB, we also show that CCL2 is released across the apical and basolateral membranes of the choroidal epithelium, a pattern of chemokine secretion that promotes leukocyte migration across epithelial barriers. Immunohistochemical and electron microscopic analyses of choroidal tissue provide evidence for the movement of monocytes, sometimes in tandem with neutrophils, along the paracellular pathways between adjacent epithelial cells. These data further support the pathophysiological role of BCSFB in promoting the recruitment of inflammatory cells to the injured brain.


Asunto(s)
Barrera Hematoencefálica/inmunología , Lesiones Encefálicas/inmunología , Líquido Cefalorraquídeo/inmunología , Plexo Coroideo/inmunología , Monocitos/citología , Animales , Membrana Basal/inmunología , Membrana Basal/ultraestructura , Barrera Hematoencefálica/ultraestructura , Western Blotting , Lesiones Encefálicas/sangre , Lesiones Encefálicas/líquido cefalorraquídeo , Células Cultivadas , Líquido Cefalorraquídeo/citología , Quimiocina CCL2/inmunología , Quimiocina CCL2/metabolismo , Quimiotaxis de Leucocito/inmunología , Plexo Coroideo/irrigación sanguínea , Plexo Coroideo/ultraestructura , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/inmunología , Uniones Intercelulares/inmunología , Uniones Intercelulares/ultraestructura , Masculino , Microscopía Electrónica de Transmisión , Monocitos/inmunología , Monocitos/ultraestructura , Infiltración Neutrófila/inmunología , Ratas , Ratas Long-Evans , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Cereb Blood Flow Metab ; 31(1): 47-51, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20959854

RESUMEN

Previous studies have indicated that the primary targets for vasopressin actions on the injured brain are the cerebrovascular endothelium and astrocytes, and that vasopressin amplifies the posttraumatic production of proinflammatory mediators. Here, the controlled cortical impact model of traumatic brain injury in rats was used to identify the sources of vasopressin in the injured brain. Injury increased vasopressin synthesis in the hypothalamus and cerebral cortex adjacent to the posttraumatic lesion. In the cortex, vasopressin was predominantly produced by activated microglia/macrophages, and, to a lesser extent, by the cerebrovascular endothelium. These data further support the pathophysiological role of vasopressin in brain injury.


Asunto(s)
Química Encefálica/fisiología , Lesiones Encefálicas/metabolismo , Vasopresinas/biosíntesis , Animales , Arginina Vasopresina/biosíntesis , Lesiones Encefálicas/patología , Corteza Cerebral/lesiones , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Endotelio Vascular/fisiología , Hipernatremia/metabolismo , Hipotálamo/lesiones , Hipotálamo/metabolismo , Hipotálamo/patología , Inmunohistoquímica , Macrófagos/fisiología , Masculino , Microglía/fisiología , Microscopía Confocal , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Long-Evans , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Transl Stroke Res ; 2(4): 492-516, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22299022

RESUMEN

The blood-brain barrier (BBB) is formed by tightly connected cerebrovascular endothelial cells, but its normal function also depends on paracrine interactions between the brain endothelium and closely located glia. There is a growing consensus that brain injury, whether it is ischemic, hemorrhagic, or traumatic, leads to dysfunction of the BBB. Changes in BBB function observed after injury are thought to contribute to the loss of neural tissue and to affect the response to neuroprotective drugs. New discoveries suggest that considering the entire gliovascular unit, rather than the BBB alone, will expand our understanding of the cellular and molecular responses to traumatic brain injury (TBI). This review will address the BBB breakdown in TBI, the role of blood-borne factors in affecting the function of the gliovascular unit, changes in BBB permeability and post-traumatic edema formation, and the major pathophysiological factors associated with TBI that may contribute to post-traumatic dysfunction of the BBB. The key role of neuroinflammation and the possible effect of injury on transport mechanisms at the BBB will also be described. Finally, the potential role of the BBB as a target for therapeutic intervention through restoration of normal BBB function after injury and/or by harnessing the cerebrovascular endothelium to produce neurotrophic growth factors will be discussed.

19.
Psychiatr Clin North Am ; 33(4): 741-56, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21093676

RESUMEN

A complex set of molecular and functional reactions is set into motion by traumatic brain injury (TBI). New research that extends beyond pathological effects on neurons suggests a key role for the blood-brain barrier, neurovascular unit, arginine vasopressin, and neuroinflammation in the pathophysiology of TBI. The prevalence of molecular derangements in TBI holds promise for the identification and use of biomarkers to assess severity of injury, determine prognosis, and perhaps direct therapy. Hopefully, improved knowledge of these elements of pathophysiology will provide the mechanistic clues that lead to improved treatment of TBI.


Asunto(s)
Arginina Vasopresina/metabolismo , Barrera Hematoencefálica/fisiopatología , Lesiones Encefálicas/fisiopatología , Encéfalo/fisiopatología , Estrés Oxidativo/inmunología , Biomarcadores , Encéfalo/inmunología , Encéfalo/metabolismo , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/terapia , Humanos , Pronóstico , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
20.
J Neurotrauma ; 27(8): 1449-61, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20504162

RESUMEN

Arginine vasopressin (AVP) has previously been shown to promote disruption of the blood-brain barrier, exacerbate edema, and augment the loss of neural tissue in various forms and models of brain injury. However, the mechanisms underlying these AVP actions are not well understood. These mechanisms were studied in AVP-deficient Brattleboro rats (Avp(di/di)), and their parental Long-Evans strain, using a controlled cortical impact model of traumatic brain injury (TBI). The increased influx of inflammatory cells into the injured cortex in wild-type versus Avp(di/di) rats was associated with higher levels of cortical synthesis of the CXC and CC chemokines found in wild-type versus Avp(di/di) rats. These chemokines were predominantly produced by the cerebrovascular endothelium and astrocytes. In astrocyte and brain endothelial cell cultures, AVP acted synergistically with tumor necrosis factor-alpha (TNF-alpha) to increase the TNF-alpha-dependent production of CXC and CC chemokines. These AVP actions were mediated by c-Jun N-terminal kinase (JNK), as shown by Western blotting and pharmacological inhibition of JNK activity. The activity of JNK was increased in response to injury, and the differences in the magnitude of its post-traumatic activation between Avp(di/di) and wild-type rats were observed. These data demonstrate that AVP plays an important role in exacerbating the brain inflammatory response to injury.


Asunto(s)
Lesiones Encefálicas/metabolismo , Mediadores de Inflamación/metabolismo , Vasopresinas/genética , Vasopresinas/fisiología , Animales , Western Blotting , Encéfalo/patología , Lesiones Encefálicas/patología , Células Cultivadas , Quimiocina CCL2/biosíntesis , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Inmunohistoquímica , Inmunoprecipitación , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratas , Ratas Brattleboro , Ratas Long-Evans , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo , Vasopresinas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA