Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 141(2): 208-221, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28251649

RESUMEN

Intracellular purine turnover is mainly oriented to preserving the level of triphosphate nucleotides, fundamental molecules in vital cell functions that, when released outside cells, act as receptor signals. Conversely, high levels of purine bases and uric acid are found in the extracellular milieu, even in resting conditions. These compounds could derive from nucleosides/bases that, having escaped to cell reuptake, are metabolized by extracellular enzymes similar to the cytosolic ones. Focusing on purine nucleoside phosphorylase (PNP) that catalyzes the reversible phosphorolysis of purine (deoxy)-nucleosides/bases, we found that it is constitutively released from cultured rat C6 glioma cells into the medium, and has a molecular weight and enzyme activity similar to the cytosolic enzyme. Cell exposure to 10 µM ATP or guanosine triphosphate (GTP) increased the extracellular amount of all corresponding purines without modifying the levels/activity of released PNP, whereas selective activation of ATP P2Y1 or adenosine A2A metabotropic receptors increased PNP release and purine base formation. The reduction to 1% in oxygen supply (2 h) to cells decreased the levels of released PNP, leading to an increased presence of extracellular nucleosides and to a reduced formation of xanthine and uric acid. Conversely, 2 h cell re-oxygenation enhanced the extracellular amounts of both PNP and purine bases. Thus, hypoxia and re-oxygenation modulated in opposite manner the PNP release/activity and, thereby, the extracellular formation of purine metabolism end-products. In conclusion, extracellular PNP and likely other enzymes deputed to purine base metabolism are released from cells, contributing to the purinergic system homeostasis and exhibiting an important pathophysiological role.


Asunto(s)
Glioma/enzimología , Purina-Nucleósido Fosforilasa/metabolismo , Animales , Línea Celular Tumoral , Ratas
2.
Brain Behav Immun ; 46: 1-16, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25736063

RESUMEN

Post-concussion syndrome is an aggregate of symptoms that commonly present together after head injury. These symptoms, depending on definition, include headaches, dizziness, neuropsychiatric symptoms, and cognitive impairment. However, these symptoms are common, occurring frequently in non-head injured controls, leading some to question the existence of post-concussion syndrome as a unique syndrome. Therefore, some have attempted to explain post-concussion symptoms as post-traumatic stress disorder, as they share many similar symptoms and post-traumatic stress disorder does not require head injury. This explanation falls short as patients with post-concussion syndrome do not necessarily experience many key symptoms of post-traumatic stress disorder. Therefore, other explanations must be sought to explain the prevalence of post-concussion like symptoms in non-head injury patients. Many of the situations in which post-concussion syndrome like symptoms may be experienced such as infection and post-surgery are associated with systemic inflammatory responses, and even neuroinflammation. Post-concussion syndrome itself has a significant neuroinflammatory component. In this review we examine the evidence of neuroinflammation in post-concussion syndrome and the potential role systemic inflammation plays in post-concussion syndrome like symptoms. We conclude that given the overlap between these conditions and the role of inflammation in their etiologies, a new term, post-inflammatory brain syndromes (PIBS), is necessary to describe the common outcomes of many different inflammatory insults. The concept of post-concussion syndrome is in its evolution therefore, the new term post-inflammatory brain syndromes provides a better understanding of etiology of its wide-array of symptoms and the wide array of conditions they can be seen in.


Asunto(s)
Conmoción Encefálica/fisiopatología , Encéfalo/fisiopatología , Inflamación/fisiopatología , Síndrome Posconmocional/diagnóstico , Síndrome Posconmocional/fisiopatología , Conmoción Encefálica/psicología , Humanos , Inflamación/psicología , Síndrome Posconmocional/psicología
3.
Adv Exp Med Biol ; 837: 23-33, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25310956

RESUMEN

Increasing body of evidence indicates that neuron-neuroglia interaction may play a key role in determining the progression of neurodegenerative diseases including Parkinson's disease (PD), a chronic pathological condition characterized by selective loss of dopaminergic (DA) neurons in the substantia nigra. We have previously reported that guanosine (GUO) antagonizes MPP(+)-induced cytotoxicity in neuroblastoma cells and exerts neuroprotective effects against 6-hydroxydopamine (6-OHDA) and beta-amyloid-induced apoptosis of SH-SY5Y cells. In the present study we demonstrate that GUO protected C6 glioma cells, taken as a model system for astrocytes, from 6-OHDA-induced neurotoxicity. We show that GUO, either alone or in combination with 6-OHDA activated the cell survival pathways ERK and PI3K/Akt. The involvement of these signaling systems in the mechanism of the nucleoside action was strengthened by a reduction of the protective effect when glial cells were pretreated with U0126 or LY294002, the specific inhibitors of MEK1/2 and PI3K, respectively. Since the protective effect on glial cell death of GUO was not affected by pretreatment with a cocktail of nucleoside transporter blockers, GUO transport and its intracellular accumulation were not at play in our in vitro model of PD. This fits well with our data which pointed to the presence of specific binding sites for GUO on rat brain membranes. On the whole, the results described in the present study, along with our recent evidence showing that GUO when administered to rats via intraperitoneal injection is able to reach the brain and with previous data indicating that it stimulates the release of neurotrophic factors, suggest that GUO, a natural compound, by acting at the glial level could be a promising agent to be tested against neurodegeneration.


Asunto(s)
Astrocitos/efectos de los fármacos , Guanosina/farmacología , Fármacos Neuroprotectores/farmacología , Neurotoxinas/antagonistas & inhibidores , Oxidopamina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Butadienos/farmacología , Línea Celular Tumoral , Cromonas/farmacología , Fragmentación del ADN/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Glioma/patología , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Morfolinas/farmacología , Neurotoxinas/toxicidad , Nitrilos/farmacología , Proteínas de Transporte de Nucleósidos/antagonistas & inhibidores , Oxidopamina/toxicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
4.
Arch Phys Med Rehabil ; 95(3): 588-90, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24060492

RESUMEN

Postanoxic myoclonus is a rare manifestation after an anoxic event, with fewer than 150 cases reported in the literature. The condition is characterized by myoclonic jerks, which are worse on action than at rest, and postural lapses, ataxia, and dysarthria. The disability caused by postanoxic myoclonus can be profound, and treatment in the rehabilitation setting is exceptionally challenging. We present 2 patients who suffered from postanoxic myoclonus after an anoxic event, both of whom were successfully treated with a combination of levetiracetam, valproic acid, and clonazepam. These cases act as a framework for discussing the management of postanoxic myoclonus in the clinical setting.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Clonazepam/uso terapéutico , Mioclonía/tratamiento farmacológico , Piracetam/análogos & derivados , Ácido Valproico/uso terapéutico , Anciano , Anticonvulsivantes/administración & dosificación , Clonazepam/administración & dosificación , Quimioterapia Combinada , Humanos , Hipoxia/complicaciones , Levetiracetam , Masculino , Persona de Mediana Edad , Mioclonía/etiología , Piracetam/administración & dosificación , Piracetam/uso terapéutico , Ácido Valproico/administración & dosificación
5.
J Neurosci Res ; 91(2): 262-72, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23151946

RESUMEN

After ischemic stroke, early thrombolytic therapy to reestablish tissue perfusion improves outcome but triggers a cascade of deleterious cellular and molecular events. Using a collaborative approach, our groups examined the effects of guanosine (Guo) in response to ischemic reperfusion injury in vitro and in vivo. In a transient middle cerebral artery occlusion (MCAO) in rats, Guo significantly reduced infarct volume in a dose-dependent manner when given systemically either immediately before or 30 min, but not 60 min, after the onset of the 5.5-hr reperfusion period. In a separate experiment, Guo significantly reduced infarct volume after 24 hr of reperfusion when administered 5 min before reperfusion. Western blot analysis did not reveal any significant changes either in endoplasmic reticulum (ER) stress proteins (GRP 78 and 94) or HSP 70 or in levels of m-calpain. In vitro oxygen and glucose deprivation (OGD) significantly increased production of both reactive oxygen species (ROS) and interleukin-8 (IL-8) in the primary astrocytes. Guo did not alter ROS or IL-8 production when given to the astrocytes before OGD. However, Guo when added to the cells prior to or 30 min after reperfusion significantly reduced IL-8 release but not ROS formation. Our study revealed a dose- and time-dependent protective effect of Guo on reperfusion injury in vitro and vivo. The mechanisms by which Guo exerts its effect are independent of unfolded proteins in ER or the level of intracellular calcium or ROS formation. However, the effect may be induced, at least partially, by inhibiting IL-8, a marker of reperfusion-triggered proinflammatory events.


Asunto(s)
Infarto Encefálico/prevención & control , Guanosina/administración & dosificación , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Daño por Reperfusión/prevención & control , Análisis de Varianza , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Infarto Encefálico/etiología , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/deficiencia , Proteínas de Choque Térmico/metabolismo , Hipoxia , Interleucina-8/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Reperfusión/efectos adversos , Daño por Reperfusión/complicaciones , Factores de Tiempo
6.
Front Neurol ; 14: 1123407, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37251220

RESUMEN

Mild traumatic brain injuries (mTBIs) trigger a neuroinflammatory response, which leads to perturbations in the levels of inflammatory cytokines, resulting in a distinctive profile. A systematic review and meta-analysis were conducted to synthesize data related to levels of inflammatory cytokines in patients with mTBI. The electronic databases EMBASE, MEDLINE, and PUBMED were searched from January 2014 to December 12, 2021. A total of 5,138 articles were screened using a systematic approach based on the PRISMA and R-AMSTAR guidelines. Of these articles, 174 were selected for full-text review and 26 were included in the final analysis. The results of this study demonstrate that within 24 hours, patients with mTBI have significantly higher levels of Interleukin-6 (IL-6), Interleukin-1 Receptor Antagonist (IL-1RA), and Interferon-γ (IFN-γ) in blood, compared to healthy controls in majority of the included studies. Similarly one week following the injury, patients with mTBI have higher circulatory levels of Monocyte Chemoattractant Protein-1/C-C Motif Chemokine Ligand 2 (MCP-1/CCL2), compared to healthy controls in majority of the included studies. The results of the meta-analysis also confirmed these findings by demonstrating significantly elevated blood levels of IL-6, MCP-1/CCL2, and Interleukin-1 beta (IL-1ß) in the mTBI population compared to healthy controls (p < 0.0001), particularly in the acute stages (<7 days). Furthermore, it was found that IL-6, Tumor Necrosis Factor-alpha (TNF-α), IL-1RA, IL-10, and MCP-1/CCL2 were associated with poor clinical outcomes following the mTBI. Finally, this research highlights the lack of consensus in the methodology of mTBI studies that measure inflammatory cytokines in the blood, and also provides direction for future mTBI research.

8.
Arch Phys Med Rehabil ; 92(1): 31-6, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21187202

RESUMEN

OBJECTIVE: To examine the effects of body-weight supported treadmill training (BWSTT) on functional ability and quality of life in patients with progressive multiple sclerosis (MS) of high disability. DESIGN: Before-after trial. SETTING: Exercise rehabilitation research center. PARTICIPANTS: Patients with progressive MS (N=6; 5 primary progressive, 1 secondary progressive) with high disability (mean±SD expanded disability status scale, [EDSS]=6.9±1.07). All participants completed the trial. INTERVENTIONS: Subjects completed 36 sessions of BWSTT (30-min sessions, 3×wk) over 12 weeks. MAIN OUTCOME MEASURES: Outcome measures included functional ability assessed by EDSS and Multiple Sclerosis Functional Composite (MSFC). Quality of life and fatigue were assessed by the MS Quality of Life-54 (MSQoL-54) and the Modified Fatigue Impact Scale (MFIS), respectively. All tests were administered at baseline and after 12 weeks of training. RESULTS: All participants progressively improved training intensity; treadmill walking speed increased (34%; P<.001), and percent body weight support was reduced (42%; P<.001). A significant improvement in both physical (P=.02) and mental (P=.01) subscales of the MSQoL-54 was found. Fatigue was nonsignificantly reduced by 31% (P=.22); however, a large effect size (ES) was noted (ES=-.93). Functional ability remained stable with nonsignificant improvements in MSFC (P=.35; ES=.23) and EDSS (P=.36; ES=-.08) scores. CONCLUSIONS: Twelve weeks of BWSTT produces beneficial effects on quality of life and potentially reduces fatigue in patients with primary progressive MS of high disability level. Larger trials will be required to confirm these findings and to evaluate further the effects of BWSTT in progressive MS.


Asunto(s)
Terapia por Ejercicio/métodos , Esclerosis Múltiple Crónica Progresiva/rehabilitación , Calidad de Vida , Caminata , Adulto , Fatiga/fisiopatología , Fatiga/rehabilitación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Crónica Progresiva/fisiopatología , Proyectos Piloto , Factores de Tiempo
9.
Clin Neurophysiol Pract ; 5: 157-164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32939420

RESUMEN

OBJECTIVE: It is unclear why specific individuals incur chronic symptoms following a concussion. This exploratory research aims to identify and characterize any neurophysiological differences that may exist in motor cortex function in post-concussion syndrome (PCS). METHODS: Fifteen adults with PCS and 13 healthy, non-injured adults were tested. All participants completed symptom questionnaires, and transcranial magnetic stimulation (TMS) was used to measure intracortical and transcallosal excitability and inhibition in the dominant motor cortex. RESULTS: Cortical silent period (p = 0.02, g = 0.96) and ipsilateral silent period (p = 0.04, g = 0.78) were shorter in the PCS group compared to the control group which may reflect reduced GABA-mediated inhibition in PCS. Furthermore, increased corticomotor excitability was observed in the left hemisphere but not the right hemisphere. CONCLUSIONS: These data suggest that persistent neurophysiological differences are present in those with PCS. The exact contributing factors to such changes remain to be investigated by future studies. SIGNIFICANCE: This study provides novel evidence of lasting neurophysiological changes in PCS.

10.
Neurosci Lett ; 431(2): 101-5, 2008 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-18191898

RESUMEN

Deprivation of oxygen and glucose for 5h induces apoptosis in SH-SY5Y neuroblastoma cell cultures. After combined glucose and oxygen deprivation (CGOD) addition of guanosine (100 microM), a non-adenine-based purine nucleoside, significantly reduced the proportion of cells undergoing apoptosis. To determine whether guanosine was also neuroprotective in vivo, we undertook middle cerebral artery occlusion (MCAo) on male Wistar rats and administered guanosine (8mg/kg), intraperitoneally, or saline (vehicle control) daily for 7 days. Guanosine prolonged rat survival and decreased both neurological deficits and tissue damage resulting from MCAo. These data are the first to demonstrate that guanosine protects neurons from the effects of CGOD even when administered 5h after the stimulus, and is neuroprotective in experimental stroke in rats.


Asunto(s)
Guanosina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Línea Celular Transformada , Infarto Cerebral/etiología , Infarto Cerebral/prevención & control , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucosa/deficiencia , Hipoxia , Etiquetado Corte-Fin in Situ , Masculino , Ratas , Ratas Wistar , Accidente Cerebrovascular/complicaciones , Factores de Tiempo
11.
Int J MS Care ; 18(5): 221-229, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27803637

RESUMEN

Background: There is evidence of the benefits of exercise training in multiple sclerosis (MS); however, few studies have been conducted in individuals with progressive MS and severe mobility impairment. A potential exercise rehabilitation approach is total-body recumbent stepper training (TBRST). We evaluated the safety and participant-reported experience of TBRST in people with progressive MS and compared the efficacy of TBRST with that of body weight-supported treadmill training (BWSTT) on outcomes of function, fatigue, and health-related quality of life (HRQOL). Methods: Twelve participants with progressive MS (Expanded Disability Status Scale scores, 6.0-8.0) were randomized to receive TBRST or BWSTT. Participants completed three weekly sessions (30 minutes) of exercise training for 12 weeks. Primary outcomes included safety assessed as adverse events and patient-reported exercise experience assessed as postexercise response and evaluation of exercise equipment. Secondary outcomes included the Multiple Sclerosis Functional Composite, the Modified Fatigue Impact Scale, and the Multiple Sclerosis Quality of Life-54 questionnaire scores. Assessments were conducted at baseline and after 12 weeks. Results: Safety was confirmed in both exercise groups. Participants reported enjoying both exercise modalities; however, TBRST was reviewed more favorably. Both interventions reduced fatigue and improved HRQOL (P ≤ .05); there were no changes in function. Conclusions: Both TBRST and BWSTT seem to be safe, well tolerated, and enjoyable for participants with progressive MS with severe disability. Both interventions may also be efficacious for reducing fatigue and improving HRQOL. TBRST should be further explored as an exercise rehabilitation tool for patients with progressive MS.

12.
Br J Pharmacol ; 135(4): 969-76, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11861325

RESUMEN

1. Extracellular guanosine has diverse effects on many cellular components of the central nervous system, some of which may be related to its uptake into cells and others to its ability to release adenine-based purines from cells. Yet other effects of extracellular guanosine are compatible with an action on G-protein linked cell membrane receptors. 2. Specific binding sites for [(3)H]-guanosine were detected on membrane preparations from rat brain. The kinetics of [(3)H]-guanosine binding to membranes was described by rate constants of association and dissociation of 2.6122 x 10(7) M(-1) min(-1) and 1.69 min(-1), respectively. A single high affinity binding site for [(3)H]-guanosine with a K(D) of 95.4 +/- 11.9 nM and B(max) of 0.57 +/- 0.03 pmol mg(-1) protein was shown. This site was specific for guanosine, and the order of potency in displacing 50 nM [(3)H]-guanosine was: guanosine=6-thio-guanosine > inosine > 6-thio-guanine > guanine. Other naturally occurring purines, such as adenosine, hypoxanthine, xanthine caffeine, theophylline, GDP, GMP and ATP were unable to significantly displace the radiolabelled guanosine. Thus, this binding site is distinct from the well-characterized receptors for adenosine and purines. 5. The addition of GTP produced a small concentration-dependent decrease in guanosine binding, suggesting this guanosine binding site was linked to a G-protein. 6. Our results therefore are consistent with the existence of a novel cell membrane receptor site, specific for guanosine.


Asunto(s)
Encéfalo/metabolismo , Guanosina/metabolismo , Animales , Sitios de Unión , Unión Competitiva , Encéfalo/ultraestructura , Técnicas In Vitro , Cinética , Masculino , Membranas , Ensayo de Unión Radioligante , Ratas , Ratas Wistar
13.
Neuroreport ; 14(18): 2463-7, 2003 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-14663211

RESUMEN

Functional loss after spinal cord injury (SCI) is caused, in part, by demyelination of axons surviving the trauma. Administration of guanosine (8 mg/kg/day, i.p.) for 7 consecutive days, starting 5 weeks after moderate SCI in rats, improved locomotor function and spinal cord remyelination. Myelinogenesis was associated with an increase in the number of mature oligodendrocytes detected in guanosine-treated spinal cord sections in comparison with controls. These data indicate that guanosine-induced remyelination resulted, at least in part, from activation of endogenous oligodendrocyte lineage cells. These findings may have significant implications for chronic demyelinating diseases.


Asunto(s)
Guanosina/farmacología , Guanosina/uso terapéutico , Vaina de Mielina/fisiología , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Enfermedad Crónica , Femenino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/efectos de los fármacos , Fibras Nerviosas Mielínicas/metabolismo , Ratas , Ratas Wistar , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo
14.
Neuroreport ; 13(14): 1789-92, 2002 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-12395124

RESUMEN

Rat brain astrocyte and microglia cultures express different members of ATP-binding-cassette (ABC) proteins. RT-PCR analysis showed that astrocytes are equipped with P-glycoprotein (mdr1a, mdr1b), multidrug resistance-associated-protein (mrp1, mrp4, mrp5) and cystic fibrosis transmembrane conductance regulator (CFTR). No transcripts for mrp5 and CFTR were detected in microglia. The ABC protein functional activities are shown by the following results: (i) cyclosporin A (50 microM), verapamil (50 microM), probenecid (1 mM) or sulfinpyrazone (2 mM) enhanced [3H]vincristine accumulation; (ii) cyclosporin A or verapamil but not probenecid or sulfinpyrazone enhanced [3H]digoxin accumulation; (iii) glibenclamide (100 microM) inhibited 36Cl efflux from astrocytes. ATP release from glial cells was inhibited by the pretreatment with ABC protein inhibitors indicating that ABC proteins are involved in nucleotide efflux from glial cells which represent the main source of cerebral extracellular purines.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfato/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Microglía/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Membrana Celular/metabolismo , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Feto , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Ratas
15.
Neuroreport ; 14(10): 1301-4, 2003 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-12876461

RESUMEN

Transected dorsal root axons of adult rats can be induced to regenerate through the normally non-permissive environment of the dorsal root entry zone (DREZ) into the spinal cord by implanting enteric glia (EG) into the DREZ. We have now examined whether the regenerating central axons make functional connections by studying the return of function of a behavioral response, the cutaneous trunci muscle (CTM) reflex. Implantation of EG into the spinal cord DREZ led to functional recovery of the CTM reflex in 82%, 72% and 70% of animals 1, 2 and 3 months, respectively, after injury. In contrast, the CTM reflex did not recover in animals implanted with 3T3 or C6 glioma cells or with vehicle only.


Asunto(s)
Contracción Muscular/fisiología , Neuroglía/fisiología , Recuperación de la Función , Piel/inervación , Traumatismos de la Médula Espinal/fisiopatología , Células 3T3/fisiología , Células 3T3/trasplante , Animales , Carbocianinas/farmacocinética , Células Cultivadas , Femenino , Colorantes Fluorescentes/farmacocinética , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Intestinos , Ratones , Neuroglía/trasplante , Ratas , Ratas Wistar , Raíces Nerviosas Espinales/metabolismo , Raíces Nerviosas Espinales/patología , Raíces Nerviosas Espinales/fisiopatología , Factores de Tiempo , Trasplante/métodos , Células Tumorales Cultivadas/fisiología , Células Tumorales Cultivadas/trasplante
16.
Neuroreport ; 15(5): 833-6, 2004 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-15073525

RESUMEN

Apoptosis is implicated in the pathophysiology of Alzheimer's disease. Extracellular guanosine inhibits staurosporine-induced apoptosis in astrocytes. We examined whether guanosine protects SH-SY5Y human neuroblastoma cells against beta-amyloid(betaA)-induced apoptosis. Addition of betaA (fragment 25-35, 5 microM for 24 h) to SH-SY5Y cells increased the number of apoptotic cells, as evaluated by oligonucleosome ELISA. Guanosine pre-treatment decreased betaA-induced apoptosis (maximal effect after 24 h, 300 microM, p<0.05). The anti-apoptotic effect of guanosine was reduced by LY294002 (PI3K inhibitor) or PD98059 (MEK inhibitor) (p<0.05). Guanosine increased phosphorylation of Akt/PKB, and this was abolished by inhibiting PI3K or MEK, (p<0.001, 5 min). Thus, the protective effect of guanosine against betaA-induced apoptosis of SH-SY5Y cells is mediated via activation of the PI3K/Akt/PKB and MAPK pathways.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Apoptosis , Guanosina/farmacología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Western Blotting , Línea Celular Tumoral , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Flavonoides/farmacología , Humanos , Morfolinas/farmacología , Neuroblastoma , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Factores de Tiempo
17.
In Vitro Cell Dev Biol Anim ; 38(4): 188-90, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12197768

RESUMEN

The enteric nervous system is a large and complex division of the peripheral nervous system. The glia associated with it share some characteristics with the olfactory-ensheathing glia, astrocytes and Schwann cells. To facilitate studies of rat enteric glia, we have developed a method for preparing them in large quantities with a high degree of homogeneity. The enteric glia were isolated from the small intestine of Wistar rats by enzymatic digestion with dispase. The cell isolate was added to a mitotically arrested layer of 3T3 cells. Subsequent separation of the enteric glia from the 3T3 cells was done enzymatically, with unavoidable loss of many enteric glia and potential contamination of enteric glia cultures with the 3T3 cells. Therefore, 3T3 cells were cultured in Nunc 0.2-microm tissue culture inserts that could be readily removed from the wells when no longer needed. There was no loss of the enteric glia. The cultures consisted entirely of GFAP-labeled cells, presumptive enteric glia. This method permits the culturing of large numbers of highly purified enteric glia without the use of expensive growth factors and complement-mediated cytolysis.


Asunto(s)
Plexo Mientérico/citología , Neuroglía/citología , Células 3T3 , Animales , Astrocitos/citología , Separación Celular/métodos , Inmunohistoquímica/métodos , Ratones , Ratas , Ratas Wistar , Células de Schwann/citología
18.
Restor Neurol Neurosci ; 31(5): 597-617, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23760224

RESUMEN

PURPOSE: Acute spinal cord injury (SCI) triggers multiple cellular and molecular pathways; therapy aimed at only one pathway is unlikely to succeed. Anecdotal reports indicate that a novel herbal formulation (JSK-Ji-Sui-Kang) may enhance recovery in humans with SCI. We investigated whether JSK's therapeutic effects could be verified in a well-established SCI model in rats. METHODS: Therapeutic effects of JSK were tested using a standard behavioral assessment, histological, immunochemical and microarray analysis. Phytochemical fingerprinting of JSK was performed using high performance liquid chromatography coupled with photodiode array detection and electrospray ionization-mass spectrometry. JSK or vehicle was gavaged to rats 24 hours after SCI and daily thereafter for 3 weeks. RESULTS: Locomotor function significantly improved (n = 12; p < 0.05), tissue damage was reduced (p < 0.01; n = 6) and more axons and myelin were observed in JSK-treated compared with vehicle control animals. JSK significantly enhanced expression of neuroglobin, vascular endothelial growth factor and growth-associated protein 43, and reduced the expression of caspase 3, cyclooxygenase-2, RhoA (p < 0.05; n = 6) and fibrinogen (p < 0.01; n = 6). RNA microarray indicated that JSK altered transcription of genes involved in ischemic and inflammatory/immune responses and apoptosis (p < 0.05; n = 3). CONCLUSIONS: JSK appears to target multiple biochemical and cellular pathways to enhance functional recovery and improve outcomes of SCI. The results provide a basis for further investigation of JSK's effects following SCI.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Preparaciones de Plantas/uso terapéutico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Animales , Medicamentos Herbarios Chinos/química , Femenino , Preparaciones de Plantas/química , Ratas , Ratas Wistar , Traumatismos de la Médula Espinal/metabolismo , Resultado del Tratamiento
19.
Neural Regen Res ; 7(28): 2165-75, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-25538736

RESUMEN

Previous studies have shown that transplanted enteric glia enhance axonal regeneration, reduce tissue damage, and promote functional recovery following spinal cord injury. However, the mechanisms by which enteric glia mediate these beneficial effects are unknown. Neurotrophic factors can promote neuronal differentiation, survival and neurite extension. We hypothesized that enteric glia may exert their protective effects against spinal cord injury partially through the secretion of neurotrophic factors. In the present study, we demonstrated that primary enteric glia cells release nerve growth factor, brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor over time with their concentrations reaching approximately 250, 100 and 50 pg/mL of culture medium respectively after 48 hours. The biological relevance of this secretion was assessed by incubating dissociated dorsal root ganglion neuronal cultures in enteric glia-conditioned medium with and/or without neutralizing antibodies to each of these proteins and evaluating the differences in neurite growth. We discovered that conditioned medium enhances neurite outgrowth in dorsal root ganglion neurons. Even though there was no detectable amount of neurotrophin-3 secretion using ELISA analysis, the neurite outgrowth effect can be attenuated by the antibody-mediated neutralization of each of the aforementioned neurotrophic factors. Therefore, enteric glia secrete nerve growth factor, brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor and neurotrophin-3 into their surrounding environment in concentrations that can cause a biological effect.

20.
Brain Res ; 1407: 79-89, 2011 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-21774919

RESUMEN

Previously we have found that extracellular guanosine (Guo) has neuroprotective properties in in vitro and in vivo. Moreover, extracellular Guo significantly increased in the ipsilateral hemisphere within 2h following focal stroke in rats, and remained elevated for one week. Therefore, we hypothesized that Guo could be a potential candidate for a non-toxic neuroprotective agent. In the present study, we examined the effects of Guo on rats following permanent middle cerebral artery occlusion (MCAO). We also determined whether Guo can precondition neurons by modulating endoplasmic reticulum (ER) stress proteins. As most therapies employ a combination treatment regimen, we optimized the neuroprotection by combining pre- and post-MCAO treatments with Guo, attempting to reduce both ischemic cell death and improve functional recovery. A combination of 4mg/kg Guo given 30min pre-stroke and 8mg/kg Guo given 3, 24 and 48h post-stroke exerted the most significant decrease in infarct volume and sustainable improvement in neurological function. Moreover, these effects are not attributable to Guo metabolites. Measurements taken 6h post-MCAO from animals pre-treated with Guo did not reveal any significant changes in ER stress proteins (GRP 78 and 94) or HSP 70, but did reveal significantly increased levels of m-calpain. Thus, our data indicate that there is a treatment regimen for Guo as a neuroprotectant following ischemic stroke. The mechanism by which Guo confers neuroprotection may involve an increase in m-calpain, possibly resulting from a mild increase in intracellular calcium. M-calpain may be involved in the preconditioning response to ischemia by upregulating endogenous pro-survival mechanisms in neurons.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Guanosina/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Conducta Animal/efectos de los fármacos , Western Blotting , Peso Corporal/efectos de los fármacos , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Guanosina/administración & dosificación , Proteínas de Choque Térmico/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Masculino , Glicoproteínas de Membrana/metabolismo , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA