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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 27(9)2022 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-35566074

RESUMEN

Traumatic Brain Injury (TBI), is one of the most common causes of neurological damage in young populations. It is widely considered as a risk factor for neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's (PD) disease. These diseases are characterized in part by the accumulation of disease-specific misfolded proteins and share common pathological features, such as neuronal death, as well as inflammatory and oxidative damage. Nano formulation of Pomegranate seed oil [Nano-PSO (Granagard TM)] has been shown to target its active ingredient to the brain and thereafter inhibit memory decline and neuronal death in mice models of AD and genetic Creutzfeldt Jacob disease. In this study, we show that administration of Nano-PSO to mice before or after TBI application prevents cognitive and behavioral decline. In addition, immuno-histochemical staining of the brain indicates that preventive Nano-PSO treatment significantly decreased neuronal death, reduced gliosis and prevented mitochondrial damage in the affected cells. Finally, we examined levels of Sirtuin1 (SIRT1) and Synaptophysin (SYP) in the cortex using Western blotting. Nano-PSO consumption led to higher levels of SIRT1 and SYP protein postinjury. Taken together, our results indicate that Nano-PSO, as a natural brain-targeted antioxidant, can prevent part of TBI-induced damage.


Asunto(s)
Enfermedad de Alzheimer , Lesiones Traumáticas del Encéfalo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Cognición , Ratones , Aceites de Plantas/química , Sirtuina 1/metabolismo
2.
Neurobiol Dis ; 124: 439-453, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30471415

RESUMEN

Traumatic brain injury (TBI) is a neurodegenerative disorder for which no effective pharmacological treatment is available. Glucagon-like peptide 1 (GLP-1) analogues such as Exenatide have previously demonstrated neurotrophic and neuroprotective effects in cellular and animal models of TBI. However, chronic or repeated administration was needed for efficacy. In this study, the pharmacokinetics and efficacy of PT302, a clinically available sustained-release Exenatide formulation (SR-Exenatide) were evaluated in a concussive mild (m)TBI mouse model. A single subcutaneous (s.c.) injection of PT302 (0.6, 0.12, and 0.024 mg/kg) was administered and plasma Exenatide concentrations were time-dependently measured over 3 weeks. An initial rapid regulated release of Exenatide in plasma was followed by a secondary phase of sustained-release in a dose-dependent manner. Short- and longer-term (7 and 30 day) cognitive impairments (visual and spatial deficits) induced by weight drop mTBI were mitigated by a single post-injury treatment with Exenatide delivered by s.c. injection of PT302 in clinically translatable doses. Immunohistochemical evaluation of neuronal cell death and inflammatory markers, likewise, cross-validated the neurotrophic and neuroprotective effects of SR-Exenatide in this mouse mTBI model. Exenatide central nervous system concentrations were 1.5% to 2.0% of concomitant plasma levels under steady-state conditions. These data demonstrate a positive beneficial action of PT302 in mTBI. This convenient single, sustained-release dosing regimen also has application for other neurological disorders, such as Alzheimer's disease, Parkinson's disease, multiple system atrophy and multiple sclerosis where prior preclinical studies, likewise, have demonstrated positive Exenatide actions.


Asunto(s)
Conmoción Encefálica/patología , Exenatida/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos ICR , Ratas , Ratas Sprague-Dawley
4.
Addict Biol ; 24(3): 414-425, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-29423994

RESUMEN

Synthetic cannabinoids are psychoactive substances designed to mimic the euphorigenic effects of the natural cannabis. Novel unregulated compounds appear once older compounds become illegal. It has been previously reported that synthetic cannabinoids are different than Δ9 -tetrahydrocannabinol (Δ9 -THC) as they have chemical structures unrelated to Δ9 -THC, different metabolism and, often, greater toxicity. This study aimed to investigate the effects of three novel synthetic cannabinoids and pure Δ9 -THC on body temperature, nociceptive threshold, anxiety, memory function, locomotor and exploratory parameters, and depression. We performed a battery of behavioural and motor tests starting 50 minutes post i.p. injection of each drug to adult ICR mice. The synthetic cannabinoids that were used are AB-FUBINACA, AB-CHMINACA and PB-22. All synthetic cannabinoids and Δ9 -THC caused hypothermia, but only Δ9 -THC induced a clear antinociceptive effect. All synthetic cannabinoids and Δ9 -THC caused decreased anxiety levels, spatial memory deficits and decreased exploratory behaviour as measured in the elevated plus maze, Y-maze and staircase paradigm, respectively. However, all synthetic cannabinoids but not Δ9 -THC demonstrated decreased locomotor activity in the staircase test. Moreover, only AB-FUBINACA and Δ9 -THC affected the gait balance and grip strength of the mice as was assessed by the latency time to fall from a rod. In the forced swimming test, PB-22 caused elevated depression-like behaviour while AB-FUBINACA induced a reversed effect. These results suggest varied effects among different synthetic cannabinoids and Δ9 -THC. Further studies are needed to characterize the overall effects and differences between these synthetic cannabinoids and Δ9 -THC.


Asunto(s)
Dronabinol/farmacología , Indazoles/farmacología , Psicotrópicos/farmacología , Valina/análogos & derivados , Animales , Ansiedad/fisiopatología , Temperatura Corporal/efectos de los fármacos , Depresión/fisiopatología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones Endogámicos ICR , Actividad Motora/efectos de los fármacos , Nocicepción/efectos de los fármacos , Umbral Sensorial/efectos de los fármacos , Valina/farmacología
5.
Mol Cell Neurosci ; 83: 74-82, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28690173

RESUMEN

BACKGROUND: Traumatic brain injury is the most common cause of death or chronic disability among people under-35-years-old. There is no effective pharmacological treatment currently existing for TBI. Hyperbaric oxygen therapy (HBOT) is defined as the inhalation of pure oxygen in a hyperbaric chamber that is pressurized higher than 1atm. HBOT offers physiological and mechanical effects by inducing a state of increased pressure and hyperoxia. HBOT has been proposed as an effective treatment for moderate traumatic brain injury (mTBI), yet the exact therapeutic window and mechanism that underlies this effect is not completely understood. METHODS: HBOT was administrated for 4 consecutive days, post a mouse closed head weight drop moderate TBI (mTBI) in 2 different time lines: immediate treatment - initiated 3h post-injury and delayed treatment - initiated 7days post-injury. Behavioral cognitive tests and biochemical changes were assessed. RESULTS: The results were similar for both the immediate and the delayed treatments. mTBI mice exhibited impairment in learning abilities, whereas mTBI mice treated with HBO displayed significant improvement compared with the mTBI group, performing similar to the sham groups. mTBI mice had a decline in myelin basic protein, an increase in neuronal loss (NeuN staining), and an increase in the number of reactive astrocytes (GFAP). The HBO treated mice in both groups did not exhibit these changes and remained similar to the sham group. CONCLUSIONS: The delayed HBOT has a potential to serve as a neuroprotective treatment for mTBI with a long therapeutic window. Further research is needed for fully understanding the cellular changes.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Oxigenoterapia Hiperbárica/métodos , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Unión al ADN , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos ICR , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo
6.
Cephalalgia ; 36(10): 915-23, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26566937

RESUMEN

BACKGROUND: Chronic post-traumatic headache (PTH) is one of the most common symptoms of mild traumatic brain injury (mTBI) but its underlying mechanisms remain unknown. Inflammatory degranulation of dural mast cells (MCs) is thought to promote headache, and may play a role in PTH. Whether mTBI is associated with persistent degranulation of dural MCs is yet to be determined. METHODS: Histochemistry was used to evaluate time course changes in dural MC density and degranulation level in concussive head trauma and blast mouse models of mTBI. The effects of sumatriptan and the MC stabilizer cromolyn sodium on concussion-evoked dural MC degranulation were also investigated. RESULTS: Concussive head injury evoked persistent MC degranulation for at least 30 days. Blast trauma gave rise to a delayed MC degranulation response commencing at seven days that also persisted for at least 30 days. Neither sumatriptan nor cromolyn treatment reduced concussion-evoked persistent MC degranulation. CONCLUSIONS: mTBI evoked by closed head injury or blast exposure is associated with persistent dural MC degranulation. Such a response in mTBI patients may contribute to PTH. Amelioration of PTH by sumatriptan may not involve inhibition of dural MC degranulation. If persistent dural MC degranulation contributes to PTH, then cromolyn treatment may not be effective.


Asunto(s)
Conmoción Encefálica/patología , Degranulación de la Célula/fisiología , Duramadre/patología , Mastocitos/patología , Cefalea Postraumática/patología , Animales , Traumatismos por Explosión/complicaciones , Traumatismos por Explosión/metabolismo , Traumatismos por Explosión/patología , Conmoción Encefálica/complicaciones , Conmoción Encefálica/metabolismo , Duramadre/metabolismo , Masculino , Mastocitos/metabolismo , Ratones , Ratones Endogámicos ICR , Cefalea Postraumática/etiología , Cefalea Postraumática/metabolismo
7.
Alzheimers Dement ; 12(1): 34-48, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26327236

RESUMEN

INTRODUCTION: Blast traumatic brain injury (B-TBI) affects military and civilian personnel. Presently, there are no approved drugs for blast brain injury. METHODS: Exendin-4 (Ex-4), administered subcutaneously, was evaluated as a pretreatment (48 hours) and postinjury treatment (2 hours) on neurodegeneration, behaviors, and gene expressions in a murine open field model of blast injury. RESULTS: B-TBI induced neurodegeneration, changes in cognition, and genes expressions linked to dementia disorders. Ex-4, administered preinjury or postinjury, ameliorated B-TBI-induced neurodegeneration at 72 hours, memory deficits from days 7-14, and attenuated genes regulated by blast at day 14 postinjury. DISCUSSION: The present data suggest shared pathologic processes between concussive and B-TBI, with end points amenable to beneficial therapeutic manipulation by Ex-4. B-TBI-induced dementia-related gene pathways and cognitive deficits in mice somewhat parallel epidemiologic studies of Barnes et al. who identified a greater risk in US military veterans who experienced diverse TBIs, for dementia in later life.


Asunto(s)
Traumatismos por Explosión/tratamiento farmacológico , Conmoción Encefálica/tratamiento farmacológico , Trastornos del Conocimiento/prevención & control , Péptido 1 Similar al Glucagón/agonistas , Péptidos/uso terapéutico , Ponzoñas/uso terapéutico , Animales , Traumatismos por Explosión/patología , Conmoción Encefálica/metabolismo , Conmoción Encefálica/patología , Cognición/efectos de los fármacos , Exenatida , Expresión Génica/efectos de los fármacos , Inyecciones Subcutáneas , Masculino , Ratones , Ratones Endogámicos ICR , Fármacos Neuroprotectores/administración & dosificación , Péptidos/farmacología , Ponzoñas/farmacología
8.
J Neurochem ; 135(6): 1203-1217, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25982185

RESUMEN

Traumatic brain injury (TBI), a brain dysfunction for which there is no present effective treatment, is often caused by a concussive impact to the head and affects an estimated 1.7 million Americans annually. Our laboratory previously demonstrated that exendin-4, a long-lasting glucagon-like peptide 1 receptor (GLP-1R) agonist, has neuroprotective effects in cellular and animal models of TBI. Here, we demonstrate neurotrophic and neuroprotective effects of a different GLP-1R agonist, liraglutide, in neuronal cultures and a mouse model of mild TBI (mTBI). Liraglutide promoted dose-dependent proliferation in SH-SY5Y cells and in a GLP-1R over-expressing cell line at reduced concentrations. Pre-treatment with liraglutide rescued neuronal cells from oxidative stress- and glutamate excitotoxicity-induced cell death. Liraglutide produced neurotrophic and neuroprotective effects similar to those of exendin-4 in vitro. The cAMP/PKA/pCREB pathway appears to play an important role in this neuroprotective activity of liraglutide. Furthermore, our findings in cell culture were well-translated in a weight drop mTBI mouse model. Post-treatment with a clinically relevant dose of liraglutide for 7 days in mice ameliorated memory impairments caused by mTBI when evaluated 7 and 30 days post trauma. These data cross-validate former studies of exendin-4 and suggest that liraglutide holds therapeutic potential for the treatment of mTBI. Exendin-4, a long-lasting glucagon-like peptide 1 receptor (GLP-1R) agonist, has neuroprotective effects in cellular and animal models of traumatic brain injury (TBI). Here, we demonstrate neurotrophic and neuroprotective effects of a different GLP-1R agonist, liraglutide, in neuronal cultures and a mouse model of mild TBI (mTBI). Liraglutide promoted dose-dependent proliferation in SH-SY5Y cells and in a GLP-1R over-expressing cell line at reduced concentrations. Pretreatment with liraglutide rescued neuronal cells from oxidative stress- and glutamate excitotoxicity-induced cell death. Liraglutide produced neurotrophic and neuroprotective effects similar to those of exendin-4 in vitro, likely involving the cAMP/PKA/pCREB pathway. Our findings in cell culture were well-translated in a weight-drop mTBI mouse model. Post-treatment with a clinically relevant dose of liraglutide for 7 days in mice ameliorated memory impairments caused by mTBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Péptido 1 Similar al Glucagón/farmacología , Liraglutida/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Péptidos/farmacología , Ponzoñas/farmacología , Animales , Conmoción Encefálica/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Exenatida , Hipoglucemiantes/farmacología , Ratones , Neuronas/metabolismo , Receptores de Glucagón/efectos de los fármacos
9.
J Neuroinflammation ; 12: 45, 2015 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-25879458

RESUMEN

BACKGROUND: The treatment of traumatic brain injury (TBI) represents an unmet medical need, as no effective pharmacological treatment currently exists. The development of such a treatment requires a fundamental understanding of the pathophysiological mechanisms that underpin the sequelae resulting from TBI, particularly the ensuing neuronal cell death and cognitive impairments. Tumor necrosis factor-alpha (TNF-α) is a cytokine that is a master regulator of systemic and neuroinflammatory processes. TNF-α levels are reported to become rapidly elevated post TBI and, potentially, can lead to secondary neuronal damage. METHODS: To elucidate the role of TNF-α in TBI, particularly as a drug target, the present study evaluated (i) time-dependent TNF-α levels and (ii) markers of apoptosis and gliosis within the brain and related these to behavioral measures of 'well being' and cognition in a mouse closed head 50 g weight drop mild TBI (mTBI) model in the presence and absence of post-treatment with an experimental TNF-α synthesis inhibitor, 3,6'-dithiothalidomide. RESULTS: mTBI elevated brain TNF-α levels, which peaked at 12 h post injury and returned to baseline by 18 h. This was accompanied by a neuronal loss and an increase in astrocyte number (evaluated by neuronal nuclei (NeuN) and glial fibrillary acidic protein (GFAP) immunostaining), as well as an elevation in the apoptotic death marker BH3-interacting domain death agonist (BID) at 72 h. Selective impairments in measures of cognition, evaluated by novel object recognition and passive avoidance paradigms - without changes in well being, were evident at 7 days after injury. A single systemic treatment with the TNF-α synthesis inhibitor 3,6'-dithiothalidomide 1 h post injury prevented the mTBI-induced TNF-α elevation and fully ameliorated the neuronal loss (NeuN), elevations in astrocyte number (GFAP) and BID, and cognitive impairments. Cognitive impairments evident at 7 days after injury were prevented by treatment as late as 12 h post mTBI but were not reversed when treatment was delayed until 18 h. CONCLUSIONS: These results implicate that TNF-α in mTBI induced secondary brain damage and indicate that pharmacologically limiting the generation of TNF-α post mTBI may mitigate such damage, defining a time-dependent window of up to 12 h to achieve this reversal.


Asunto(s)
Lesiones Encefálicas/complicaciones , Encéfalo/patología , Trastornos del Conocimiento , Neuronas/enzimología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/terapia , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Fosfopiruvato Hidratasa/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Talidomida/análogos & derivados , Talidomida/química , Talidomida/uso terapéutico , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
10.
Alzheimers Dement ; 10(1 Suppl): S62-75, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24529527

RESUMEN

Traumatic brain injury (TBI), either as an isolated injury or in conjunction with other injuries, is an increasingly common event. An estimated 1.7 million injuries occur within the USA each year and 10 million people are affected annually worldwide. Indeed, nearly one third (30.5%) of all injury-related deaths in the USA are associated with TBI, which will soon outpace many common diseases as the major cause of death and disability. Associated with a high morbidity and mortality and no specific therapeutic treatment, TBI has become a pressing public health and medical problem. The highest incidence of TBI occurs in young adults (15-24 years age) and in the elderly (≥75 years of age). Older individuals are particularly vulnerable to these types of injury, often associated with falls, and have shown increased mortality and worse functional outcome after lower initial injury severity. In addition, a new and growing form of TBI, blast injury, associated with the detonation of improvised explosive devices in the war theaters of Iraq and Afghanistan, are inflicting a wave of unique casualties of immediate impact to both military personnel and civilians, for which long-term consequences remain unknown and may potentially be catastrophic. The neuropathology underpinning head injury is becoming increasingly better understood. Depending on severity, TBI induces immediate neuropathologic effects that, for the mildest form, may be transient; however, with increasing severity, these injuries cause cumulative neural damage and degeneration. Even with mild TBI, which represents the majority of cases, a broad spectrum of neurologic deficits, including cognitive impairments, can manifest that may significantly influence quality of life. Further, TBI can act as a conduit to longer term neurodegenerative disorders. Prior studies of glucagon-like peptide-1 (GLP-1) and long-acting GLP-1 receptor agonists have demonstrated neurotrophic/neuroprotective activities across a broad spectrum of cellular and animal models of chronic neurodegenerative (Alzheimer's and Parkinson's diseases) and acute cerebrovascular (stroke) disorders. In view of the mechanisms underpinning these disorders as well as TBI, we review the literature and recent studies assessing GLP-1 receptor agonists as a potential treatment strategy for mild to moderate TBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Incretinas/farmacología , Incretinas/uso terapéutico , Neuronas/efectos de los fármacos , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/epidemiología , Lesiones Encefálicas/patología , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Receptor del Péptido 1 Similar al Glucagón , Humanos , Receptores de Glucagón/agonistas
11.
Neurobiol Dis ; 54: 1-11, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23454194

RESUMEN

Warfare has long been associated with traumatic brain injury (TBI) in militarized zones. Common forms of TBI can be caused by a physical insult to the head-brain or by the effects of a high velocity blast shock wave generated by the detonation of an explosive device. While both forms of trauma are distinctly different regarding the mechanism of trauma induction, there are striking similarities in the cognitive and emotional status of survivors. Presently, proven effective therapeutics for the treatment of either form of TBI are unavailable. To be able to develop efficacious therapies, studies involving animal models of physical- and blast-TBI are required to identify possible novel or existing medicines that may be of value in the management of clinical events. We examined indices of cognition and anxiety-like behavior and the hippocampal gene transcriptome of mice subjected to both forms of TBI. We identified common behavioral deficits and gene expression regulations, in addition to unique injury-specific forms of gene regulation. Molecular pathways presented a pattern similar to that seen in gene expression. Interestingly, pathways connected to Alzheimer's disease displayed a markedly different form of regulation depending on the type of TBI. While these data highlight similarities in behavioral outcomes after trauma, the divergence in hippocampal transcriptome observed between models suggests that, at the molecular level, the TBIs are quite different. These models may provide tools to help define therapeutic approaches for the treatment of physical- and blast-TBIs. Based upon observations of increasing numbers of personnel displaying TBI related emotional and behavioral changes in militarized zones, the development of efficacious therapies will become a national if not a global priority.


Asunto(s)
Traumatismos por Explosión/complicaciones , Lesiones Encefálicas/complicaciones , Cognición/fisiología , Hipocampo/fisiopatología , Transcriptoma , Animales , Traumatismos por Explosión/fisiopatología , Traumatismos por Explosión/psicología , Lesiones Encefálicas/fisiopatología , Lesiones Encefálicas/psicología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos ICR , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
AIMS Neurosci ; 10(1): 33-51, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37077956

RESUMEN

Background: In the fear memory network, the hippocampus modulates contextual aspects of fear learning while mutual connections between the amygdala and the medial prefrontal cortex are widely involved in fear extinction. G-protein-coupled receptors (GPCRs) are involved in the regulation of fear and anxiety, so the regulation of GPCRs in fear signaling pathways can modulate the mechanisms of fear memory acquisition, consolidation and extinction. Various studies suggested a role of M-type K+ channels in modulating fear expression and extinction, although conflicting data prevented drawing of clear conclusions. In the present work, we examined the impact of M-type K+ channel blockade or activation on contextual fear acquisition and extinction. In addition, regarding the pivotal role of the hippocampus in contextual fear conditioning (CFC) and the involvement of the axon initial segment (AIS) in neuronal plasticity, we investigated whether structural alterations of the AIS in hippocampal neurons occurred during contextual fear memory acquisition and short-time extinction in mice in a behaviorally relevant context. Results: When a single systemic injection of the M-channel blocker XE991 (2 mg/kg, IP) was carried out 15 minutes before the foot shock session, fear expression was significantly reduced. Expression of c-Fos was increased following CFC, mostly in GABAergic neurons at day 1 and day 2 post-fear training in CA1 and dentate gyrus hippocampal regions. A significantly longer AIS segment was observed in GABAergic neurons of the CA1 hippocampal region at day 2. Conclusions: Our results underscore the role of M-type K + channels in CFC and the importance of hippocampal GABAergic neurons in fear expression.

13.
Biomedicines ; 11(5)2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37239152

RESUMEN

Traumatic brain injury (TBI) is considered the most common neurological disorder among people under the age of 50. In modern combat zones, a combination of TBI and organophosphates (OP) can cause both fatal and long-term effects on the brain. We utilized a mouse closed-head TBI model induced by a weight drop device, along with OP exposure to paraoxon. Spatial and visual memory as well as neuron loss and reactive astrocytosis were measured 30 days after exposure to mild TBI (mTBI) and/or paraoxon. Molecular and cellular changes were assessed in the temporal cortex and hippocampus. Cognitive and behavioral deficits were most pronounced in animals that received a combination of paraoxon exposure and mTBI, suggesting an additive effect of the insults. Neuron survival was reduced in proximity to the injury site after exposure to paraoxon with or without mTBI, whereas in the dentate gyrus hilus, cell survival was only reduced in mice exposed to paraoxon prior to sustaining a mTBI. Neuroinflammation was increased in the dentate gyrus in all groups exposed to mTBI and/or to paraoxon. Astrocyte morphology was significantly changed in mice exposed to paraoxon prior to sustaining an mTBI. These results provide further support for assumptions concerning the effects of OP exposure following the Gulf War. This study reveals additional insights into the potentially additive effects of OP exposure and mTBI, which may result in more severe brain damage on the modern battlefield.

14.
Exp Neurol ; 352: 114022, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35202640

RESUMEN

BACKGROUND: Cognitive deficits are the most enduring and debilitating sequelae of mild traumatic brain injury (mTBI). However, relatively little is known about whether the cognitive effects of mTBI vary with respect to time post-injury, biological sex, and injury location. OBJECTIVES: The aim of this study was to assess the effect of the side and site of mTBI and to determine whether these effects are sexually dimorphic. METHODS: Male and female ICR mice were subjected to either a sham procedure or mTBI to the temporal lobes (right-sided or left-sided) or to the frontal lobes (bilateral) using a weight-drop model. After recovery, mice underwent a battery of behavioral tests at two post-injury time points. RESULTS: Different mTBI impact locations produced dissociable patterns of memory deficits; the extent of these deficits varied across sexes, time points, and memory domains. In both sexes, frontal mTBI mice exhibited a delayed onset of spatial memory deficits. Additionally, the performance of the frontal and left temporal injured males and females was more variable than that of controls. Interestingly, only in females does the effect of mTBI on visual recognition memory depend on the time post-injury. Moreover, only in females does spatial recognition memory remain relatively intact after mTBI to the left temporal lobe. CONCLUSION: This study showed that different mTBI impact sites produce dissociable and sex-specific patterns of cognitive deficits in mice. The results emphasize the importance of considering the injury site/side and biological sex when evaluating the cognitive sequelae of mTBI.


Asunto(s)
Conmoción Encefálica , Animales , Conmoción Encefálica/complicaciones , Cognición , Femenino , Masculino , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos ICR , Lóbulo Temporal
15.
J Neurochem ; 118(6): 1032-42, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21740439

RESUMEN

Mild traumatic brain injury (mTBI) patients do not show clear structural brain defects and, in general, do not require hospitalization, but frequently suffer from long-lasting cognitive, behavioral and emotional difficulties. Although there is no current effective treatment or cure for mTBI, tumor necrosis factor-alpha (TNF-α), a cytokine fundamental in the systemic inflammatory process, represents a potential drug target. TNF-α levels increase after mTBI and may induce or exacerbate secondary damage to brain tissue. The present study evaluated the efficacy of the experimental TNF-α synthesis inhibitor, 3,6'-dithiothalidomide, on recovery of mice from mTBI in a closed head weight-drop model that induces an acute elevation in brain TNF-α and an impairment in cognitive performance, as assessed by the Y-maze, by novel object recognition and by passive avoidance paradigms at 72 h and 7 days after injury. These impairments were fully ameliorated in mice that received a one time administration of 3,6'-dithiothalidomide at either a low (28 mg/kg) or high (56 mg/kg) dose provided either 1 h prior to injury, or at 1 or 12 h post-injury. Together, these results implicate TNF-α as a drug target for mTBI and suggests that 3,6'-dithiothalidomide may act as a neuroprotective drug to minimize impairment.


Asunto(s)
Conducta Animal/fisiología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/psicología , Talidomida/análogos & derivados , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Línea Celular , Inflamación/inducido químicamente , Inflamación/patología , Inflamación/prevención & control , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Masculino , Aprendizaje por Laberinto , Memoria/fisiología , Ratones , Ratones Endogámicos ICR , Reconocimiento en Psicología/efectos de los fármacos , Talidomida/uso terapéutico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
16.
J Mol Neurosci ; 71(1): 178-186, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32901372

RESUMEN

The present paper shows how cinnamon extract (CE) consumption mitigates neuronal loss and memory impairment following traumatic brain injury (TBI), one of the world's most common neurodegenerative diseases. TBI patients suffer short- and long-term behavioral, cognitive, and emotional impairments, including difficulties in concentration, memory loss, and depression. Research shows that CE application can mitigate cognitive and behavioral impairments in animal models for Alzheimer's and Parkinson's disease, whose pathophysiology is similar to that of TBI. This study builds on prior research by showing similar results in TBI mice models. After drinking CE for a week, mice were injured using our 70-g weight drop TBI device. For 2 weeks thereafter, the mice continued drinking CE alongside standard lab nutrition. Subsequently, the mice underwent behavioral tests to assess their memory, motor activity, and anxiety. The mice brains were harvested for immunohistochemistry staining to evaluate overall neuronal survival. Our results show that CE consumption almost completely mitigates memory impairment and decreases neuronal loss after TBI. Mice that did not consume CE demonstrated impaired memory. Our results also show that CE consumption attenuated neuronal loss in the temporal cortex and the dentate gyrus. Mice that did not consume CE suffered a significant neuronal loss. There were no significant differences in anxiety levels and motor activity between all groups. These findings show a new therapeutic approach to improve cognitive function and decrease memory loss after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Cinnamomum zeylanicum , Trastornos del Conocimiento/prevención & control , Trastornos de la Memoria/prevención & control , Fitoterapia , Extractos Vegetales/uso terapéutico , Administración Oral , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/etiología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Recuento de Células , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Prueba de Laberinto Elevado , Conducta Exploratoria , Locomoción/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos ICR , Neuronas/patología , Extractos Vegetales/farmacología , Reconocimiento en Psicología/efectos de los fármacos , Agua
17.
Neurotrauma Rep ; 2(1): 381-390, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34723249

RESUMEN

Current literature details an array of contradictory results regarding the effect of radiofrequency electromagnetic radiation (RF-EMR) on health, both in humans and in animal models. The present study was designed to ascertain the conflicting data published regarding the possible impact of cellular exposure (radiation) on male and female mice as far as spatial memory, anxiety, and general well-being is concerned. To increase the likelihood of identifying possible "subtle" effects, we chose to test it in already cognitively impaired (following mild traumatic brain injury; mTBI) mice. Exposure to cellular radiation by itself had no significant impact on anxiety levels or spatial/visual memory in mice. When examining the dual impact of mTBI and cellular radiation on anxiety, no differences were found in the anxiety-like behavior as seen at the elevated plus maze (EPM). When exposed to both mTBI and cellular radiation, our results show improvement of visual memory impairment in both female and male mice, but worsening of the spatial memory of female mice. These results do not allow for a decisive conclusion regarding the possible hazards of cellular radiation on brain function in mice, and the mTBI did not facilitate identification of subtle effects by augmenting them.

18.
Sci Rep ; 11(1): 23559, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34876621

RESUMEN

Traumatic brain injury (TBI) is a brain dysfunction without present treatment. Previous studies have shown that animals fed ketogenic diet (KD) perform better in learning tasks than those fed standard diet (SD) following brain injury. The goal of this study was to examine whether KD is a neuroprotective in TBI mouse model. We utilized a closed head injury model to induce TBI in mice, followed by up to 30 days of KD/SD. Elevated levels of ketone bodies were confirmed in the blood following KD. Cognitive and behavioral performance was assessed post injury and molecular and cellular changes were assessed within the temporal cortex and hippocampus. Y-maze and Novel Object Recognition tasks indicated that mTBI mice maintained on KD displayed better cognitive abilities than mTBI mice maintained on SD. Mice maintained on SD post-injury demonstrated SIRT1 reduction when compared with uninjured and KD groups. In addition, KD management attenuated mTBI-induced astrocyte reactivity in the dentate gyrus and decreased degeneration of neurons in the dentate gyrus and in the cortex. These results support accumulating evidence that KD may be an effective approach to increase the brain's resistance to damage and suggest a potential new therapeutic strategy for treating TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/dietoterapia , Dieta Cetogénica , Animales , Ansiedad , Astrocitos/patología , Lesiones Traumáticas del Encéfalo/sangre , Lesiones Traumáticas del Encéfalo/psicología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Disfunción Cognitiva/dietoterapia , Disfunción Cognitiva/psicología , Modelos Animales de Enfermedad , Traumatismos Cerrados de la Cabeza/sangre , Traumatismos Cerrados de la Cabeza/dietoterapia , Traumatismos Cerrados de la Cabeza/psicología , Hipocampo/metabolismo , Hipocampo/patología , Cuerpos Cetónicos/sangre , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos ICR , Neuronas/patología , Reconocimiento en Psicología , Sirtuina 1/metabolismo
19.
Neurobiol Dis ; 38(2): 299-303, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20138993

RESUMEN

Insulin-like growth factor-1 (IGF-1) was suggested as a potential neuroprotective treatment for traumatic brain injury (TBI) induced damage (cognitive as well as cellular). The main goal of the present study was to evaluate the role of the IGF-1R activation in spatial memory outcome following mild traumatic brain injury. mTBI-induced phosphorylation of IGF-1R, AKT and ERK1/2, in mice hippocampus, which was inhibited when mice were pretreated with the selective IGF-1R inhibitor AG1024. IGF-1 administration prevented spatial memory deficits following mTBI. Surprisingly, blocking the IGF-1R signaling in mTBI mice did not augment the spatial memory deficit. In addition, this data imply an intriguing and complex role of the IGF-1 signaling axis in the cellular and behavioral events following mTBI.


Asunto(s)
Lesiones Encefálicas/metabolismo , Hipocampo/metabolismo , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Recuerdo Mental/efectos de los fármacos , Recuerdo Mental/fisiología , Ratones , Ratones Endogámicos ICR , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Conducta Espacial/efectos de los fármacos , Conducta Espacial/fisiología
20.
J Neurotrauma ; 37(20): 2169-2179, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32434427

RESUMEN

Victims of mild traumatic brain injury (mTBI) usually do not display clear morphological brain defects, but frequently have long-lasting cognitive deficits, emotional difficulties, and behavioral disturbances. In the present study we used diffusion magnetic resonance imaging (dMRI) combined with graph theory measurements to investigate the effects of mTBI on brain network connectivity. We employed a non-invasive closed-head weight-drop mouse model to produce mTBI. Mice were scanned at two time points, 24 h before the injury and either 7 or 30 days following the injury. Connectivity matrices were computed for each animal at each time point, and these were subsequently used to extract graph theory measures reflecting network integration and segregation, on both the global (i.e., whole brain) and local (i.e., single regions) levels. We found that cluster coefficient, reflecting network segregation, decreased 7 days post-injury and then returned to baseline level 30 days following the injury. Global efficiency, reflecting network integration, demonstrated opposite patterns in the left and right hemispheres, with an increase of right hemisphere efficiency at 7 days and then a decrease in efficiency following 30 days, and vice versa in the left hemisphere. These findings suggest a possible compensation mechanism acting to moderate the influence of mTBI on the global network. Moreover, these results highlight the importance of tracking the dynamic changes in mTBI over time, and the potential of structural connectivity as a promising approach for studying network integrity and pathology progression in mTBI.


Asunto(s)
Conmoción Encefálica/fisiopatología , Red Nerviosa/fisiopatología , Vías Nerviosas/fisiopatología , Animales , Mapeo Encefálico/métodos , Imagen de Difusión Tensora , Modelos Animales de Enfermedad , Procesamiento de Imagen Asistido por Computador , Masculino , Ratones , Ratones Endogámicos ICR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA