Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Intervalo de año de publicación
1.
Int Rev Neurobiol ; 146: 259-279, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31349930

RESUMEN

Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons in the substantia nigra, the depletion of striatal dopamine and the presence of Lewy aggregates containing alpha-synuclein. Clinically, there are motor impairments involving cardinal movement symptoms, bradykinesia, resting tremor, muscle rigidity, and postural abnormalities, along with non-motor symptoms such as sleep, behavior and mood disorders. The current treatment for PD focuses on restoring dopaminergic neurotransmission by l-3,4-dihydroxyphenylalanine (levodopa), which loses therapeutic efficacy and induces disabling abnormal involuntary movements known as levodopa-induced dyskinesia (LID) after several years. Evidence indicates that the pathophysiology of both PD and LID disorders is also associated with the dysfunctional activity of the serotonergic (5-HT) neurons that may be responsible for motor and non-motor disturbances. The main population of 5-HT neurons is located in the dorsal raphe nuclei (DRN), which provides extensive innervation to almost the entire neuroaxis and controls multiple functions in the brain. The degeneration of DRN 5-HT neurons occurs in early PD. These neurons can also take exogenous levodopa to transform it into dopamine, which may disturb neuron activity. This review will provide an overview of the underlying mechanisms responsible for 5-HT dysfunction and its clinical relevance in PD and dyskinesia.


Asunto(s)
Encéfalo/fisiología , Encéfalo/fisiopatología , Discinesia Inducida por Medicamentos/fisiopatología , Enfermedad de Parkinson/fisiopatología , Neuronas Serotoninérgicas/fisiología , Animales , Humanos , Levodopa/efectos adversos , Modelos Neurológicos
2.
J Basic Clin Physiol Pharmacol ; 29(3): 241-246, 2018 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-29902911

RESUMEN

BACKGROUND: In previous studies, we have observed that specific N-methyl-d-aspartic acid (NMDA) antagonists and non-NMDA antagonists injected within the nucleus accumbens septi (NAS) induced an anxiolytic-like effect in the plus maze test in rats. In the present study, the effect of intracanalicular blockade of NMDA receptors using dizocilpine in the plus maze was studied in male rats bilaterally cannulated NAS. METHODS: Rats were divided into five groups that received either 1 µL injections of saline or dizocilpine (MK-801, [5R,10S]-[+]-5-methyl-10,11-dihydro-5H-dibenzo [a,d] cyclohepten-5,10-imine) in different doses (0.5, 1, 2, or 4 µg) 15 min before testing. RESULTS: Time spent in the open arm increased under dizocilpine treatment with the two higher doses (2 and 4 µg, p<0.05), extreme arrivals were increased by the three higher doses (1 µg, p<0.05; 2 and 4 µg, p<0.01), and open arm entries by the three higher doses (1, 2, and 4 µg, p<0.05). A dose-effect relationship was observed in all cases. CONCLUSIONS: We conclude that dizocilpine-glutamatergic blockade in the accumbens lead to an anxiolytic-like effect and a behavioral disinhibition related to an increase in some motoric parameters, showing specific behavioral patterns.


Asunto(s)
Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Ansiolíticos/administración & dosificación , Ansiolíticos/farmacología , Conducta Animal/efectos de los fármacos , Maleato de Dizocilpina/administración & dosificación , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
3.
Mol Neurobiol ; 55(1): 359-369, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28875428

RESUMEN

Previous studies from our laboratory show that intraperitoneal injections of 1-metyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP, 20 mg/kg) daily within 2-h intervals for 5 days in mice induce Parkinson's disease (PD)-like symptoms on the 8th day. A significant decrease in dopamine (DA) and its metabolites 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA) along with a marked decrease in the number of tyrosine hydroxylase (TH)-positive cells in the substantia nigra pars compacta (SNpc) and striatum (STr) confirms the validity of this model for studying PD. Since cerebrolysin (CBL) is a well-balanced composition of several neurotrophic factors and active peptide fragments, in the present investigation we examined the timed release of CBL using titanate nanospheres (TiNS) in treating PD in our mouse model. Our observations show that TiNS-CBL (in a dose of 3 ml/kg, i.v.) given after 2 days of MPTP administration for 5 days resulted in a marked increase in TH-positive cells in the SNpc and STr as compared to normal CBL. Also, TiNS-CBL resulted in significantly higher levels of DA, DOPAC, and HVA in SNpc and STr on the 8th day as compared to normal CBL therapy. TiNS-CBL also thwarted increased α-synuclein levels in the brain and in the cerebrospinal fluid (CSF) as well as neuronal nitric oxide synthase (nNOS) in the in PD brain as compared to untreated group. Behavioral function was also significantly improved in MPTP-treated animals that received TiNS-CBL. These observations are the first to demonstrate that timed release of TiNS-CBL has far more superior neuroprotective effects in PD than normal CBL.


Asunto(s)
Aminoácidos/administración & dosificación , Encéfalo/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Nanosferas/administración & dosificación , Trastornos Parkinsonianos/tratamiento farmacológico , Titanio/administración & dosificación , Aminoácidos/farmacocinética , Animales , Encéfalo/patología , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/farmacocinética , Liberación de Fármacos/efectos de los fármacos , Liberación de Fármacos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Nanosferas/metabolismo , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología
4.
Mol Neurobiol ; 55(1): 43-59, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28842826

RESUMEN

Exposure to an enriched environment (EE) has neuroprotective benefits and improves recovery from brain injury due to, among other, increased neurotrophic factor expression. Through these neurotrophins, important cortical and hippocampal changes occur. Vandetanib acts as a tyrosine kinase inhibitor of cell receptors, among others, the vascular endothelial growth factor receptor (VEGFR). Our aim was to investigate the effectiveness of EE counteracting cognitive and cellular effects after tyrosine kinase receptor blockade. Animals were reared under standard laboratory condition or EE; both groups received vandetanib or vehicle. Visuospatial learning was tested with Morris water maze. Neuronal, interneuronal, and vascular densities were measured by inmunohistochemistry and histochemistry techniques. Quantifications were performed in the hippocampus and in the visual cortex. Brain-derived neurotrophic factor (BDNF), tyrosine kinase B receptor (TrkB), Akt, and Erk were measured by Western blot technique. Vandetanib produces a significant decrease in vascular and neuronal densities and reduction in the expression of molecules involved in survival and proliferation processes such as phospho-Akt/Akt and phospho-Erk/Erk. These results correlated to a cognitive impairment in visuospatial test. On the other hand, animals reared in an EE are able to reverse the negative effects, activating PI3K-AKT and MAP kinase pathways mediated by BDNF-TrkB binding. Present results provide novel and consistent evidences about the usefulness of living in EE as a strategy to improve deleterious effects of blocking neurotrophic pathways by vandetanib and the notable role of the BDNF-TrkB pathway to balance the neurovascular unit and cognitive effects.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ambiente , Inhibidores de Proteínas Quinasas/toxicidad , Receptor trkB/antagonistas & inhibidores , Receptor trkB/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Ratas , Ratas Long-Evans , Transducción de Señal/efectos de los fármacos
5.
Int Rev Neurobiol ; 137: 123-165, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29132541

RESUMEN

More than 5.5 million Americans of all ages are suffering from Alzheimer's disease (AD) till today for which no suitable therapy has been developed so far. Thus, there is an urgent need to explore novel therapeutic measures to contain brain pathology in AD. The hallmark of AD includes amyloid-beta peptide (AßP) deposition and phosphorylation of tau in AD brain. Recent evidences also suggest a marked decrease in neurotrophic factors in AD. Thus, exogenous supplement of neurotrophic factors could be one of the possible ways for AD therapy. Human postmortem brain in AD shows alterations in histamine receptors as well, indicating an involvement of the amine in AD-induced brain pathology. In this review, we focused on role of histamine 3 and 4 receptor-modulating drugs in the pathophysiology of AD. Moreover, antibodies to histamine and tau appear to be also beneficial in reducing brain pathology, blood-brain barrier breakdown, and edema formation in AD. Interestingly, TiO2-nanowired delivery of cerebrolysin-a balanced composition of several neurotrophic factors attenuated AßP deposition and reduced tau phosphorylation in AD brain leading to neuroprotection. Coadministration of cerebrolysin with histamine antibodies or tau antibodies has further enhanced neuroprotection in AD. These novel observations strongly suggest a role of nanomedicine in AD that requires further investigation.


Asunto(s)
Enfermedad de Alzheimer/terapia , Aminoácidos/administración & dosificación , Anticuerpos/administración & dosificación , Histamínicos/administración & dosificación , Titanio , Proteínas tau/inmunología , Animales , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Humanos
6.
Int Rev Neurobiol ; 137: 99-122, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29132545

RESUMEN

Parkinson's disease (PD) is the second most frequent neurodegenerative disorder, but current therapies are only symptomatic. Experimental models are necessary to go deeper in the comprehension of pathophysiological mechanism and to assess new therapeutic strategies. The unilateral 6-hydroxydopamine (6-OHDA) lesion either in medial forebrain bundle (MFB) or into the striatum in rats affords to study various stages of PD depending on the evolution time lapsed. A promising alternative to address the neurodegenerative process is the use of neurotrophic factors; but its clinical use has been limited due to its short half-life and rapid degradation after in vivo administration, along with difficulties for crossing the blood-brain barrier (BBB). Tyrosine hydroxylase (TH) immunostaining revealed a significant decrease of the TH-immunopositive striatal volume in 6-OHDA group from rostral to caudal one. The loss of TH-ir neurons and axodendritic network (ADN) was higher in caudal sections showing a selective vulnerability of the topological distributed dopaminergic system. In addition to a remarkable depletion of dopamine in the nigrostriatal system, the administration of 6-OHDA into MFB induces some other neuropathological changes such as an increase of glial fibrillary acidic protein (GFAP) positive cells in substantia nigra (SN) as well as in striatum. Intrastriatal implantation of micro- or nanosystems delivering neurotrophic factor in parkinsonized rats for bypassing BBB leads to a significative functional and morphological recovery. Neurorestorative morphological changes (preservation of the TH-ir cells and ADN) along the rostrocaudal axis of caudoputamen complex and SN have been probed supporting a selective recovering after the treatment as well. Others innovative therapeutic strategies, such as the intranasal delivery, have been recently assessed in order to search the NTF effects. In addition some others methodological key points are reviewed.


Asunto(s)
Nanopartículas/administración & dosificación , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Humanos , Fármacos Neuroprotectores/uso terapéutico
7.
Int Rev Neurobiol ; 137: 65-98, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29132544

RESUMEN

The possibility that histamine influences the spinal cord pathophysiology following trauma through specific receptor-mediated upregulation of neuronal nitric oxide synthase (nNOS) was examined in a rat model. A focal spinal cord injury (SCI) was inflicted by a longitudinal incision into the right dorsal horn of the T10-11 segments. The animals were allowed to survive 5h. The SCI significantly induced breakdown of the blood-spinal cord barrier to protein tracers, reduced the spinal cord blood flow at 5h, and increased the edema formation and massive upregulation of nNOS expression. Pretreatment with histamine H1 receptor antagonist mepyramine (1mg, 5mg, and 10mg/kg, i.p., 30min before injury) failed to attenuate nNOS expression and spinal cord pathology following SCI. On the other hand, blockade of histamine H2 receptors with cimetidine or ranitidine (1mg, 5mg, or 10mg/kg) significantly reduced these early pathophysiological events and attenuated nNOS expression in a dose-dependent manner. Interestingly, TiO2-naowire delivery of cimetidine or ranitidine (5mg doses) exerted superior neuroprotective effects on SCI-induced nNOS expression and cord pathology. It appears that effects of ranitidine were far superior than cimetidine at identical doses in SCI. On the other hand, pretreatment with histamine H3 receptor agonist α-methylhistamine (1mg, 2mg, or 5mg/kg, i.p.) that inhibits histamine synthesis and release in the central nervous system thwarted the spinal cord pathophysiology and nNOS expression when used in lower doses. Interestingly, histamine H3 receptor antagonist thioperamide (1mg, 2mg, or 5mg/kg, i.p.) exacerbated nNOS expression and cord pathology after SCI. These novel observations suggest that blockade of histamine H2 receptors or stimulation of histamine H3 receptors attenuates nNOS expression and induces neuroprotection in SCI. Taken together, our results are the first to demonstrate that histamine-induced pathophysiology of SCI is mediated via nNOS expression involving specific histamine receptors.


Asunto(s)
Nanocables/administración & dosificación , Óxido Nítrico Sintasa de Tipo I/metabolismo , Receptores Histamínicos/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/prevención & control , Regulación hacia Arriba/fisiología , Animales , Cimetidina/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos , Antagonistas de los Receptores Histamínicos H1/administración & dosificación , Antagonistas de los Receptores H2 de la Histamina/administración & dosificación , Humanos , Masculino , Pirilamina/administración & dosificación , Ratas , Flujo Sanguíneo Regional/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
8.
Int Rev Neurobiol ; 137: 47-63, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29132543

RESUMEN

Influence of iron oxide magnetic nanoparticles (IOMNPs, 10nm in diameter, 0.25 or 0.50mg/mL in 100µL, i.v.) on the blood-brain barrier (BBB) permeability, edema formation, and neuronal or glial changes within 4-24h after administration was examined in normal rats and after a focal spinal cord injury (SCI). Furthermore, effect of cerebrolysin, a balanced composition of several neurotrophic factors, and active peptide fragments was also evaluated on IOMNP-induced changes in central nervous system (CNS) pathology. The SCI was inflicted in rats by making a longitudinal incision into the right dorsal horn of the T10-11 segments and allowed to survive 4 or 24h after trauma. Cerebrolysin (2.5mL/kg, i.v.) was given either 30min before IOMNP injection in the 4-h SCI group or 4h after injury in the 24-h survival groups. Control group received cerebrolysin in identical situation following IOMNP administration. In all groups, leakage of serum albumin in the CNS as a marker of BBB breakdown and activation of astrocytes using glial fibrillary acidic protein was evaluated by immunohistochemistry. The neuronal injury was examined by Nissl staining. The IOMNPs alone in either low or high doses did not induce CNS pathology either following 4 or 24h after administration. However, administration of IOMNPs in SCI group slightly enhanced the pathological changes in the CNS after 24h but not 4h after trauma. Cerebrolysin treatment markedly attenuated IOMNP-induced aggravation of SCI-induced cord pathology and induced significant neuroprotection. These observations are the first to show that IOMNPs are safe for the CNS and cerebrolysin treatment prevented CNS pathology following a combination of trauma and IOMNP injection. This indicated that cerebrolysin might be used as adjunct therapy during IOMNP administration in disease conditions, not reported earlier.


Asunto(s)
Aminoácidos/uso terapéutico , Compuestos Férricos/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Traumatismos de la Médula Espinal/terapia , Administración Intravenosa , Animales , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Nanopartículas/administración & dosificación , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/inducido químicamente , Traumatismos de la Médula Espinal/patología , Resultado del Tratamiento
9.
CNS Neurol Disord Drug Targets ; 15(9): 1188-1197, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27667557

RESUMEN

Cerebral syndromes related to high-altitude exposure are becoming more frequent as the number of trips to high altitudes has increased in the last decade. The commonest symptom is headache, followed by acute mountain sickness (AMS) and high-altitude cerebral edema (HACE), which can be fatal. The pathophysiology of these syndromes is not fully understood. The classical "tight-fit hypothesis" posits that there are some anatomical variations that would obstruct the sinovenous outflow and worsen vasogenic edema and intracranial hypertension reactive to hypoxia. This could explain microhemorrhages seen in autopsies. However, recent magnetic resonance imaging studies have demonstrated some components of cytotoxic edema in HACE absent in AMS, suggesting a dysfunction in water balance at the cellular level. Currently, the "red-ox theory" supports trigemino-vascular system activation by free radicals formed after hypoxia and the consequent oxidative stress cascades. Apart from trigemino-vascular system activation, free radicals can also provoke membrane destabilisation mediated by lipid peroxidation, inflammation, and local hypoxia inducible factor-1α and vascular endothelial growth factor activation, resulting in gross blood-brain barrier (BBB) dysfunction. Besides alterations in endothelial cells such as increased pinocytotic vesicles and disassembly of interendothelial tight junction proteins, capillary permeability may also increase with subsequent swelling of astrocyte end-feet. In conclusion, although the pathophysiology of AMS and HACE is not completely understood, recent evidence proposes a multifactorial entity, with brain swelling and compromise of the BBB considered to play an important role. A fuller comprehension of these processes is crucial to reduce and prevent BBB alterations during high-altitude exposure.


Asunto(s)
Altitud , Barrera Hematoencefálica/metabolismo , Animales , Barrera Hematoencefálica/lesiones , Barrera Hematoencefálica/patología , Humanos
10.
CNS Neurol Disord Drug Targets ; 15(9): 1045-1071, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27592625

RESUMEN

The blood-brain barrier (BBB) plays a pivotal role in the maintenance of central nervous system function in health and disease. Thus, in almost all neurodegenerative, traumatic or metabolic insults BBB breakdown occurs, allowing entry of serum proteins into the brain fluid microenvironment with subsequent edema formation and cellular injury. Accordingly, pharmacological restoration of BBB function will lead to neurorepair. However, brain injury which occurs following blast, bullet wounds, or knife injury appears to initiate different sets of pathophysiological responses. Moreover, other local factors at the time of injury such as cold or elevated ambient temperatures could also impact the final outcome. Obviously, drug therapy applied to different kinds of brain trauma occurring at either cold or hot environments may respond differently. This is largely due to the fact that internal defense mechanisms of the brain, gene expression, release of neurochemicals and binding of drugs to specific receptors are affected by external ambient temperature changes. These factors may also affect BBB function and development of edema formation after brain injury. In this review, the effects of seasonal exposure to heat and cold on traumatic brain injury using different models i.e., concussive brain injury and cerebral cortical lesion, on BBB dysfunction in relation to drug therapy are discussed. Our observations clearly suggest that closed head injury and open brain injury are two different entities and the external hot or cold environments affect both of them remarkably. Thus, effective pharmacological therapeutic strategies should be designed with these views in mind, as military personnel often experience blunt or penetrating head injuries in either cold or hot environments.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/fisiopatología , Frío , Calor , Fármacos Neuroprotectores/farmacología , Animales , Barrera Hematoencefálica/patología , Lesiones Encefálicas/patología , Ambiente , Humanos , Fármacos Neuroprotectores/uso terapéutico
11.
CNS Neurol Disord Drug Targets ; 15(9): 1092-1117, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27538949

RESUMEN

The blood-brain barrier (BBB) is a physiological regulator of transport of essential items from blood to brain for the maintenance of homeostasis of the central nervous system (CNS) within narrow limits. The BBB is also responsible for export of harmful or metabolic products from brain to blood to keep the CNS fluid microenvironment healthy. However, noxious insults to the brain caused by trauma, ischemia or environmental/chemical toxins alter the BBB function to small as well as large molecules e.g., proteins. When proteins enter the CNS fluid microenvironment, development of brain edema occurs due to altered osmotic balance between blood and brain. On the other hand, almost all neurodegenerative diseases and traumatic insults to the CNS and subsequent BBB dysfunction lead to edema formation and cell injury. To treat these brain disorders suitable drug therapy reaching their brain targets is needed. However, due to edema formation or only a focal disruption of the BBB e.g., around brain tumors, many drugs are unable to reach their CNS targets in sufficient quantity. This results in poor therapeutic outcome. Thus, new technology such as nanodelivery is needed for drugs to reach their CNS targets and be effective. In this review, use of nanowires as a possible novel tool to enhance drug delivery into the CNS in various disease models is discussed based on our investigations. These data show that nanowired delivery of drugs may have superior neuroprotective ability to treat several CNS diseases effectively indicating their role in future therapeutic strategies.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Fármacos del Sistema Nervioso Central/administración & dosificación , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Sistema Nervioso Central/lesiones , Sistemas de Liberación de Medicamentos , Nanocables , Animales , Barrera Hematoencefálica/metabolismo , Sistema Nervioso Central/efectos de los fármacos , Enfermedades del Sistema Nervioso Central/metabolismo , Humanos
12.
Mol Neurobiol ; 52(2): 867-81, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26133300

RESUMEN

Military personnel are often subjected to sleep deprivation (SD) during combat operations. Since SD is a severe stress and alters neurochemical metabolism in the brain, a possibility exists that acute or long-term SD will influence blood-brain barrier (BBB) function and brain pathology. This hypothesis was examined in young adult rats (age 12 to 14 weeks) using an inverted flowerpot model. Rats were placed over an inverted flowerpot platform (6.5 cm diameter) in a water pool where the water levels are just 3 cm below the surface. In this model, animals can go to sleep for brief periods but cannot achieve deep sleep as they would fall into water and thus experience sleep interruption. These animals showed leakage of Evans blue in the cerebellum, hippocampus, caudate nucleus, parietal, temporal, occipital, cingulate cerebral cortices, and brain stem. The ventricular walls of the lateral and fourth ventricles were also stained blue, indicating disruption of the BBB and the blood-cerebrospinal fluid barrier (BCSFB). Breakdown of the BBB or the BCSFB fluid barrier was progressive in nature from 12 to 48 h but no apparent differences in BBB leakage were seen between 48 and 72 h of SD. Interestingly, rats treated with metal nanoparticles, e.g., Cu or Ag, showed profound exacerbation of BBB disruption by 1.5- to 4-fold, depending on the duration of SD. Measurement of plasma and brain serotonin showed a close correlation between BBB disruption and the amine level. Repeated treatment with the serotonin 5-HT3 receptor antagonist ondansetron (1 mg/kg, s.c.) 4 and 8 h after SD markedly reduced BBB disruption and brain pathology after 12 to 24 h SD but not following 48 or 72 h after SD. However, TiO2-nanowired ondansetron (1 mg/kg, s.c) in an identical manner induced neuroprotection in rats following 48 or 72 h SD. However, plasma and serotonin levels were not affected by ondansetron treatment. Taken together, our observations are the first to show that (i) SD could induce BBB disruption and brain pathology, (ii) nanoparticles exacerbate SD-induced brain damage, and (iii) serotonin 5-HT3 receptor antagonist ondansetron is neuroprotective in SD that is further potentiated byTiO2-nanowired delivery, not reported earlier.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/prevención & control , Cobre/toxicidad , Nanopartículas/toxicidad , Fármacos Neuroprotectores/farmacología , Ondansetrón/farmacología , Agonistas del Receptor de Serotonina 5-HT3/farmacología , Plata/toxicidad , Privación de Sueño/fisiopatología , Animales , Proteínas Sanguíneas/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/patología , Encéfalo/fisiopatología , Química Encefálica/efectos de los fármacos , Edema Encefálico/etiología , Edema Encefálico/fisiopatología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/fisiopatología , Trastornos del Conocimiento/prevención & control , Colorantes/farmacocinética , Cobre/administración & dosificación , Evaluación Preclínica de Medicamentos , Implantes de Medicamentos , Azul de Evans/farmacocinética , Fatiga/etiología , Fatiga/fisiopatología , Fatiga/prevención & control , Radioisótopos de Yodo/farmacocinética , Masculino , Nanocables , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Ondansetrón/administración & dosificación , Ondansetrón/uso terapéutico , Ratas , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante , Trastornos de la Sensación/etiología , Trastornos de la Sensación/fisiopatología , Trastornos de la Sensación/prevención & control , Serotonina/análisis , Agonistas del Receptor de Serotonina 5-HT3/administración & dosificación , Agonistas del Receptor de Serotonina 5-HT3/uso terapéutico , Plata/administración & dosificación , Privación de Sueño/complicaciones , Factores de Tiempo
13.
Mol Neurobiol ; 52(2): 882-98, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26126513

RESUMEN

Increased levels of ubiquitin and heat shock protein (HSP) 72 kD are often seen in spinal cord injury (SCI). However, their roles in cell injury or survival are not well known. Thus, we have investigated the possible relationship between ubiquitin and HSP expressions in relation to cell injury in healthy animals, or following nanoparticle (NP) intoxication in SCI animals. A focal SCI was inflicted on the T10-11 segments over the right dorsal horn; animals were allowed to survive from 5 to 8 h after trauma. Separate groups of rats were exposed to SiO2, Ag, or Cu NPs for 7 days and subjected to SCI on the eighth day. A marked increase in ubiquitin or HSP immunoreactive cells occurred in the T9 to T12 segments 5 h after the injury, which further extended to the T4 and L5 after 8 h of survival. At this time, a marked increase in blood-spinal cord barrier (BSCB) permeability to Evans blue and radioiodine, accompanied by an intense edema formation, was observed. Changes were further exacerbated in NP-treated traumatized rats. The most marked expressions of ubiquitin and HSP in SCI were seen in rats treated with SiO2, followed by Ag, and Cu NPs. Treatment with H-290/51 (50 mg/kg p.o., 30 to 60 min after injury) or carfilzomib (1 mg/kg, i.v., 30 to 60 min after trauma) significantly reduced the ubiquitin or HSP expressions, as well as the BSCB breakdown, the edema formation, and the cell injury in the cord both 5 and 8 h after the injury, in normal animals. However, a double dose of H-290/51 (100 mg/kg) or carfilzomib (2 mg/kg) is needed to reduce cord pathology or ubiquitin and HSP expressions in traumatized animals treated with NPs. H-290/51 showed superior beneficial effects in reducing cord pathology in SCI than carfilzomib. These observations are the first to demonstrate that (i) NP-treated traumatized animals induce a widespread BSCB leakage, edema formation, and cord pathology as well as an overexpression of ubiquitin and HSP, (ii) high doses of antioxidant compounds or proteasome inhibitors are required for neuroprotection in the NP-exposed traumatized group, and (iii) ubiquitin and HSP expressions play a key role in neuronal injury in SCI, not reported earlier.


Asunto(s)
Antioxidantes/uso terapéutico , Cobre/toxicidad , Proteínas del Choque Térmico HSP72/biosíntesis , Indoles/uso terapéutico , Nanopartículas/toxicidad , Proteínas del Tejido Nervioso/biosíntesis , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , Inhibidores de Proteasoma/uso terapéutico , Plata/toxicidad , Traumatismos de la Médula Espinal/tratamiento farmacológico , Ubiquitina/biosíntesis , Animales , Antioxidantes/farmacología , Cobre/administración & dosificación , Evaluación Preclínica de Medicamentos , Edema/etiología , Edema/prevención & control , Proteínas del Choque Térmico HSP72/genética , Indoles/farmacología , Masculino , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/farmacología , Oligopéptidos/farmacología , Inhibidores de Proteasoma/farmacología , Ratas , Ratas Wistar , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/toxicidad , Plata/administración & dosificación , Médula Espinal/metabolismo , Médula Espinal/patología , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/etiología , Traumatismos de la Médula Espinal/metabolismo , Vértebras Torácicas , Ubiquitina/genética , Regulación hacia Arriba
14.
Mol Neurobiol ; 52(2): 837-45, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26126514

RESUMEN

Previous studies from our laboratory showed that topical application of growth hormone (GH) induced neuroprotection 5 h after spinal cord injury (SCI) in a rat model. Since nanodelivery of drugs exerts superior neuroprotective effects, a possibility exists that nanodelivery of GH will induce long-term neuroprotection after a focal SCI. SCI induces GH deficiency that is coupled with insulin-like growth factor-1 (IGF-1) reduction in the plasma. Thus, an exogenous supplement of GH in SCI may enhance the IGF-1 levels in the cord and induce neuroprotection. In the present investigation, we delivered TiO2-nanowired growth hormone (NWGH) after a longitudinal incision of the right dorsal horn at the T10-11 segments in anesthetized rats and compared the results with normal GH therapy on IGF-1 and GH contents in the plasma and in the cord in relation to blood-spinal cord barrier (BSCB) disruption, edema formation, and neuronal injuries. Our results showed a progressive decline in IGF-1 and GH contents in the plasma and the T9 and T12 segments of the cord 12 and 24 h after SCI. Marked increase in the BSCB breakdown, as revealed by extravasation of Evans blue and radioiodine, was seen at these time points after SCI in association with edema and neuronal injuries. Administration of NWGH markedly enhanced the IGF-1 levels and GH contents in plasma and cord after SCI, whereas normal GH was unable to enhance IGF-1 or GH levels 12 or 24 h after SCI. Interestingly, NWGH was also able to reduce BSCB disruption, edema formation, and neuronal injuries after trauma. On the other hand, normal GH was ineffective on these parameters at all time points examined. Taken together, our results are the first to demonstrate that NWGH is quite effective in enhancing IGF-1 and GH levels in the cord and plasma that may be crucial in reducing pathophysiology of SCI.


Asunto(s)
Hormona del Crecimiento/uso terapéutico , Factor I del Crecimiento Similar a la Insulina/análisis , Nanocables , Fármacos Neuroprotectores/uso terapéutico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Administración Tópica , Animales , Sistemas de Liberación de Medicamentos , Implantes de Medicamentos , Edema/etiología , Edema/prevención & control , Azul de Evans/farmacocinética , Hormona del Crecimiento/administración & dosificación , Hormona del Crecimiento/análisis , Hormona del Crecimiento/farmacocinética , Bombas de Infusión , Infusión Espinal , Radioisótopos de Yodo/farmacocinética , Masculino , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacocinética , Permeabilidad , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/análisis , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Médula Espinal/irrigación sanguínea , Médula Espinal/química , Médula Espinal/patología , Traumatismos de la Médula Espinal/sangre , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/fisiopatología , Vértebras Torácicas
15.
CNS Neurol Disord Drug Targets ; 14(3): 386-99, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25714976

RESUMEN

We have shown previously that heat stroke produced by whole body hyperthermia (WBH) for 4 h at 38°C in diabetic rats exacerbates blood-brain barrier breakdown, brain edema formation and neuronal cell injury as compared to healthy animals after identical heat exposure. In this combination of diabetes and WBH, normal therapeutic measures do not induce sufficient neuroprotection. Thus, we investigated whether nanowired mesenchymal cells (MSCs) when delivered systemically may have better therapeutic effects on brain damage in diabetic rats after WBH. Diabetes induced by streptozotocin administration (75 mg/kg, i.p, daily for 3 days) in rats resulted in clinical symptoms of the disease within 4 to 6 weeks (blood glucose level 20 to 30 mmoles/l as compared to saline control groups (4 to 6 mmoles/l). When subjected to WBH, these diabetic rats showed a 4-to 6-fold exacerbation of blood-brain barrier breakdown to Evans blue and radioiodine, along with brain edema formation and neuronal cell injury. Intravenous administration of rat MSCs (1x10(6)) to diabetic rats one week before WBH slightly reduced brain pathology, whereas TiO2 nanowired MSCs administered in an identical manner resulted in almost complete neuroprotection. On the other hand, MSCs alone significantly reduced brain pathology in saline-treated rats after WBH. These observations indicate that nanowired delivery of stem cells has superior therapeutic potential in heat stroke with diabetes, pointing to novel clinical perspectives in the future.


Asunto(s)
Encéfalo/patología , Diabetes Mellitus Experimental/patología , Golpe de Calor/patología , Golpe de Calor/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Axones/fisiología , Axones/ultraestructura , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/fisiopatología , Edema Encefálico/patología , Edema Encefálico/fisiopatología , Permeabilidad Capilar/fisiología , Diabetes Mellitus Experimental/fisiopatología , Marcha/fisiología , Golpe de Calor/fisiopatología , Masculino , Trasplante de Células Madre Mesenquimatosas/instrumentación , Células Madre Mesenquimatosas/patología , Células Madre Mesenquimatosas/fisiología , Nanopartículas del Metal , Vaina de Mielina/fisiología , Vaina de Mielina/ultraestructura , Ratas Sprague-Dawley , Titanio , Caminata/fisiología
16.
Pharmacol Rep ; 65(3): 566-78, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950579

RESUMEN

BACKGROUND: The effect of the agonism on γ-aminobutyric acid (GABA) receptors was studied within medial prefrontal cortex (mPFC), amygdala (AMY) and ventral hipocampus (VH) in the plus-maze test in male rats bilaterally cannulated. These structures send glutamatergic projections to the nucleus accumbens septi (NAS), in which interaction and integration between these afferent pathways has been described. In a previous study of our group, blockade of glutamatergic transmission within NAS induced an anxiolytic like effect. METHODS: Three rat groups received either saline or dipotassium chlorazepate (1 or 2 µg/1 µl solution) 15 min before testing. Time spent in the open arms (TSOA), time per entry (TPE), extreme arrivals (EA), open and closed arms entries (OAE, CAE) and relationship between open- and closed-arms quotient (OCAQ) were recorded. RESULTS: In the AMY injected group TSOA, OAE and EA were increased by the higher doses of dipotassium chlorazepate (p < 0.01). In the mPFC, TPE was decreased by both doses (p < 0.05). Injection within ventral hippocampus (VH) decreased TSOA, OAE and OCAQ with lower doses (p < 0.05). When the three studied saline groups were compared, TSOA, OAE, EA and OCAQ were enhanced in the VH group when compared to mPFC and AMY (p < 0.001). Insertion of inner canula (p < 0.001, p < 0.01, p < 0.01) and saline injection showed an increasing significant difference (p < 0.001 in all cases) with the action of guide cannula alone within VH in TSOA, OAE and EA. CONCLUSION: We conclude that the injection of dipotassium chlorazepate has a differential effect depending of the brain area, leading to facilitatory and inhibitory effects on anxiety processing.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Clorazepato Dipotásico/administración & dosificación , Fármacos actuantes sobre Aminoácidos Excitadores/administración & dosificación , Hipocampo/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Animales , Ansiolíticos/administración & dosificación , Ansiedad/tratamiento farmacológico , Agonistas del GABA/administración & dosificación , Masculino , Ratas
17.
J Signal Transduct ; 2012: 597915, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22852079

RESUMEN

The angiogenesis process is a key event for glioma survival, malignancy and growth. The start of angiogenesis is mediated by a cascade of intratumoural events: alteration of the microvasculature network; a hypoxic microenvironment; adaptation of neoplastic cells and synthesis of pro-angiogenic factors. Due to a chaotic blood flow, a consequence of an aberrant microvasculature, tissue hypoxia phenomena are induced. Hypoxia inducible factor 1 is a major regulator in glioma invasiveness and angiogenesis. Clones of neoplastic cells with stem cell characteristics are selected by HIF-1. These cells, called "glioma stem cells" induce the synthesis of vascular endothelial growth factor. This factor is a pivotal mediator of angiogenesis. To elucidate the role of these angiogenic mediators during glioma growth, we have used a rat endogenous glioma model. Gliomas induced by prenatal ENU administration allowed us to study angiogenic events from early to advanced tumour stages. Events such as microvascular aberrations, hypoxia, GSC selection and VEGF synthesis may be studied in depth. Our data showed that for the treatment of gliomas, developing anti-angiogenic therapies could be aimed at GSCs, HIF-1 or VEGF. The ENU-glioma model can be considered to be a useful option to check novel designs of these treatment strategies.

18.
Brain Res ; 1473: 141-54, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22824331

RESUMEN

VEGF is the major angiogenic and vascular permeability factor in health and disease. Vascular development depends on function, and in sensory areas is experience-dependent. Our aim was to investigate, qualitatively and quantitatively, the effects of intracortical infusion and neutralisation of VEGF during the first days of the critical visual period, when peak levels of endogenous VEGF secretion are reached. VEGF was intracortically delivered into middle cortical layers of P18 Long-Evans rats. Another cohort received anti-VEGF. Vehicle (PBS)-infused and non-operated animals were used as controls. Various immunopathological analyses were performed: Endothelial Barrier Antigen (EBA) for the BBB integrity and GFAP for astroglial response. Vascular density was measured by Butyryl Cholinesterase Histochemistry, neuronal density by NeuN immunohistochemistry and apoptosis by TUNEL staining. VEGF levels were measured by Western Blot. Decreased vascular permeability was evoked in VEGF-infused rats whilst EBA expression remained constant, suggesting a preserved BBB function. When VEGF was blocked, tissue showed a higher degree of extravasation and a decreased number of EBA-positive vessels surrounding the injury. Lesion induced by cannula implantation annulled the normal increase in vascular density and the decrease in neuronal density during this time. VEGF rescued in part the vascular increase, and also prevented physiological and pathological neuronal death. VEGF blockade induced a higher amount of neural loss and lower astrocytic reaction. Our results support the role of VEGF as extending beyond vascularization, preventing physiological and pathological neuronal death, not only in the injured hemisphere but also in the intact one suggesting a process of transhemispheric diaschisis.


Asunto(s)
Período Crítico Psicológico , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Corteza Visual , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Western Blotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Inyecciones Intraventriculares , Microscopía Confocal , Neovascularización Fisiológica/fisiología , Ratas , Ratas Long-Evans , Corteza Visual/efectos de los fármacos , Corteza Visual/crecimiento & desarrollo , Corteza Visual/metabolismo , Corteza Visual/patología
19.
Pharmacol Rep ; 64(1): 54-63, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22580520

RESUMEN

It has been recognized that the stress-related peptides are involved in anxiety states. Angiotensin II receptor blockade by systemic administration of the AT(1) receptor antagonists has been proposed as a new treatment possibility for anxiety disorders. For better understanding of the related mechanisms, in this study we evaluated effects of bilateral intraamygdaloid injections of 2 (LOS 2) and 4 (LOS 4) µg of losartan (LOS), a selective AT(1) receptor antagonist, on the behavior of the not stressed and acutely stressed rats in an elevated "plus" maze. Under non-stress conditions, LOS 4 increased time spent in the open arms (p < 0.01), number of extreme open arm arrivals (p < 0.05), time per entry (p < 0.01), and the number of total arm entries (p < 0.05) showing thus considerable anxiolytic activity. The open arm extreme arrivals were increased by LOS 4 in both not stressed (p < 0.05) and stressed (p < 0.05) rats. When no stressed and stressed LOS 4 animals were compared, time per entry and the number of closed arm entries (p < 0.05, both) were decreased in the latter group. Moreover, the LOS 4 stressed rats had significantly increased open/closed arm quotient (p < 0.05) as compared to the both control and LOS 4 non-stress group (p < 0.05, both). These findings suggest that the AT(1) receptor blockade in amygdala is important for the anxiolytic action of LOS (and probably other AT(1) receptor blockers) under both non-stress and stress conditions.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Ansiolíticos/farmacología , Conducta Animal/efectos de los fármacos , Losartán/farmacología , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratas , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo
20.
Rev. neurol. (Ed. impr.) ; 52(12): 743-750, 16 jun., 2011. ilus
Artículo en Español | IBECS | ID: ibc-91667

RESUMEN

Introducción. Una subpoblación de células neoplásicas con características de células madre ha sido descrita en glioblastomas multiformes humanos. Estas células desempeñan un papel fundamental en el proceso de angiogénesis y malignización tumoral, estando involucradas en la infiltración del parénquima normal adyacente. Las denominadas células madre gliales podrían ser las responsables de la recidiva tras la exéresis. Esto se debe a su capacidad de supervivencia después de tratamientos de quimio/radioterapia. Desarrollo. En este trabajo revisamos el papel de las células madre gliales en relación con el proceso de angiogénesis. También revisamos algunos hallazgos que relacionan la aparición de estas células durante la angiogénesis en un modeloexperimental de glioma endógeno de rata. Las células fueron caracterizadas utilizando anticuerpos frente a los antígenos CD133, nestina y el factor de crecimiento del endotelio vascular (VEGF). Las células nestina+ se encontraron en todos los estadios de desarrollo del tumor, mientras que las CD133+ sólo estaban presentes a partir de estadios intermedios correspondientes a la sobreexpresión del VEGF. Este momento se conoce como ‘activación de la angiogénesis’ (angiogenic switch). También observamos que algunas de las células nestina+ coexpresaban el antígeno CD133. Las células madre gliales, en nuestro modelo experimental de glioma, al igual que en glioblastomas multiformes humanos, se distribuyen formando nichos en áreas perivasculares e hipóxicas intratumorales. Conclusión. Numerosas evidencias corroboran la hipótesis de que las células madre gliales guardan una estrecha relación con el inicio de la angiogénesis, el fenómeno de hipoxia intratumoral y el entramado microvascular neoplásico (AU)


Introduction. A subpopulation of neoplastic cells with characteristics of stem cells has been described on human multiformglioblastomas. These cells play a pivotal role in tumour angiogenesis and malignancy being involved in infiltration of adjacent normal parenchyma. The named glial stem cells could be responsible for recurrences after surgery. This is due to their survival capacity after quimio/radiotherapy treatments. Development. In this work we review the role of glial stem cells in relationship with angiogenesis process. We also review some findings related to the appearance of these cells during angiogenesis in a rat endogenous experimental model of gliomas. These cells were characterized by antibodies against the antigens CD133, nestin and the vascular endothelial growth factor (VEGF). Nestin+ cells were found in every stage of tumour development, whereas CD133+ cells were only present since intermediates stages corresponding with VEGF overexpression. This moment is known as start of angiogenesis or ‘angiogenic switch’. We also found that some nestin+ cells co-expressed CD133 antigen. Glial stem cells are distributed in the experimental glioma model as well as in human multiform glioblastomas, shaping niches into perivascular or intratumoral hypoxic areas. Conclusion. Many evidences corroborate the hypothesis that glial stem cells have a close relationship with angiogenic switch, intratumor hypoxia and neoplastic microvascular network (AU)


Asunto(s)
Humanos , Neovascularización Patológica/fisiopatología , Glioblastoma/patología , Glioma/patología , Neoplasias del Sistema Nervioso/patología , Neurregulina-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...