Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 158, 2023 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-37403174

RESUMEN

BACKGROUND: Inflammation is a fundamental biological response to injury and infection, which if unregulated can contribute to the pathophysiology of many diseases. The vagus nerve, which primarily originates from the dorsal motor nucleus (DMN), plays an important role in rapidly dampening inflammation by regulating splenic function. However, direct vagal innervation of the spleen, which houses the majority of immune and inflammatory cells, has not been established. As an alternative to direct innervation, an anti-inflammatory reflex pathway has been proposed which involves the vagus nerve, the sympathetic celiac ganglion, and the neurotransmitter norepinephrine. Although sympathetic regulation of inflammation has been shown, the interaction of the vagus nerve and the celiac ganglia requires a unique interaction of parasympathetic and sympathetic inputs, making this putative mechanism of brain-spleen interaction controversial. BODY: As neuropeptides can be expressed at relatively high levels in neurons, we reasoned that DMN neuropeptide immunoreactivity could be used to determine their target innervation. Employing immunohistochemistry, subdiaphragmatic vagotomy, viral tract tracing, CRISPR-mediated knock-down, and functional assays, we show that cocaine and amphetamine-regulated transcript (CART) peptide-expressing projection neurons in the caudal DMN directly innervate the spleen. In response to lipopolysaccharide (LPS) stimulation, CART acts to reduce inflammation, an effect that can be augmented by intrasplenic administration of a synthetic CART peptide. These in vivo effects could be recapitulated in cultured splenocytes, suggesting that these cells express the as yet unidentified CART receptor(s). CONCLUSION: Our results provide evidence for direct connections between the caudal DMN and spleen. In addition to acetylcholine, these neurons express the neuropeptide CART that, once released, acts to suppress inflammation by acting directly upon splenocytes.


Asunto(s)
Neuropéptidos , Bazo , Humanos , Bazo/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo , Nervio Vago , Inflamación/metabolismo
2.
J Neurotrauma ; 39(19-20): 1279-1288, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35481812

RESUMEN

Epigenetic information is not permanently encoded in the DNA sequence, but rather consists of reversible, heritable modifications that regulate the gene expression profile of a cell. Epigenetic modifications can result in cellular changes that can be long lasting and include DNA methylation, histone methylation, histone acetylation, and RNA methylation. As epigenetic modifications are reversible, the enzymes that add (epigenetic writers), the proteins that decode (epigenetic readers), and the enzymes that remove (epigenetic erasers) these modifications can be targeted to alter cellular function and disease biology. While epigenetic modifications and their contributions are intense topics of current research in the context of a number of diseases, including cancer, inflammatory diseases, and Alzheimer disease, the study of epigenetics in the context of traumatic brain injury (TBI) is in its infancy. In this review, we will summarize the experimental and clinical findings demonstrating that TBI triggers epigenetic modifications, with a focus on changes in DNA methylation, histone methylation, and the translational utility of the universal methyl donor S-adenosylmethionine (SAM). Finally, we will review the evidence for using methyl donors as possible treatments for TBI-associated pathology and outcome.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Histonas , Lesiones Traumáticas del Encéfalo/genética , Epigénesis Genética , Histonas/genética , Histonas/metabolismo , Humanos , ARN , S-Adenosilmetionina/metabolismo
3.
eNeuro ; 9(1)2022.
Artículo en Inglés | MEDLINE | ID: mdl-34903525

RESUMEN

Mild traumatic brain injury (mTBI) can initiate complex pathophysiological changes in the brain. Numerous cellular and molecular mechanisms underlying these pathologic changes are altered after injury, including those involved in energy utilization, excitotoxicity, ionic disturbances, and inflammation. It is thought that targeting multiple mechanisms may be necessary to produce meaningful reductions in brain pathology and to improve outcome. Previous studies have reported that the anti-diabetic drug metformin can also affect inflammatory, cell survival, and metabolic outcomes, possibly by acting on multiple targets and/or pathways. We therefore questioned whether metformin treatment can reduce pathology after repeat mild closed head injury (rmCHI) in male C57Bl/6 mice. We found that metformin, administered acutely after each head impact, resulted in markedly reduced white matter damage, astrogliosis, loss of hippocampal parvalbumin neurons, and improved mitochondrial function. In addition, both motor and cognitive functions were significantly improved when tested after discontinuation of the treatment. These studies suggest that metformin may be beneficial as a treatment for repeat concussion.


Asunto(s)
Conmoción Encefálica , Traumatismos Cerrados de la Cabeza , Metformina , Animales , Encéfalo , Conmoción Encefálica/tratamiento farmacológico , Modelos Animales de Enfermedad , Masculino , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL
4.
Exp Neurol ; 331: 113372, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32504636

RESUMEN

In the adult brain, self-renewing radial-glia like (RGL) progenitor cells have been shown to reside in the subventricular zone and the subgranular zone of the hippocampus. A large body of evidence shows that experiences such as learning, enriched environment and stress can alter proliferation and differentiation of RGL progenitor cells. The progenitor cells present in the subgranular zone of the hippocampus divide to give rise to newborn neurons that migrate to the dentate gyrus where they differentiate into adult granule neurons. These newborn neurons have been found to have a unique role in certain types of hippocampus-dependent learning and memory, including goal-directed behaviors that require pattern separation. Experimental traumatic brain injury (TBI) in rodents has been shown to alter hippocampal neurogenesis, including triggering the acute loss of newborn neurons, as well as progenitor cell hyper-proliferation. In this review, we discuss the role of hippocampal neurogenesis in learning and memory. Furthermore, we review evidence for the molecular mechanisms that contribute to newborn neuron loss, as well as increased progenitor cell proliferation after TBI. Finally, we discuss strategies aimed at enhancing neurogenesis after TBI and their possible therapeutic benefits.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Hipocampo , Neurogénesis/fisiología , Animales , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Hipocampo/patología , Hipocampo/fisiopatología , Humanos
5.
Exp Neurol ; 327: 113207, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31962129

RESUMEN

One of the consistent pathologies associated with both clinical and experimental traumatic brain injury is axonal injury, especially following mild traumatic brain injury (or concussive injury). Several lines of experimental evidence have demonstrated a role for NAD+ metabolism in axonal degeneration. One of the enzymes that metabolizes NAD+ in axons is Sarm1 (Sterile Alpha and TIR Motif Containing 1), and its activity is thought to play a key role in axonal degeneration. Using a Sarm1 knock-out mouse, we examined if loss of Sarm1 offers axonal injury protection and improves cognitive outcome after repeated mild closed head injury (rmCHI). Our results indicate that rmCHI caused white matter damage that can be observed in the corpus callosum, cingulum bundle, alveus of the hippocampus, and fimbria of the fornix of wild-type mice. These pathological changes were markedly reduced in injured Sarm1-/- mice. Interestingly, the activation of astrocytes and microglia was also attenuated in the areas with white matter damage, suggesting reduced inflammation. Associated with these improved pathological outcomes, injured Sarm1-/- mice performed significantly better in both motor and cognitive tasks. Taken together, our results suggest that strategies aimed at inhibiting Sarm1 and/or restoring NAD+ levels in injured axons may have therapeutic utility.


Asunto(s)
Proteínas del Dominio Armadillo/genética , Axones/metabolismo , Encéfalo/metabolismo , Cognición/fisiología , Proteínas del Citoesqueleto/genética , Traumatismos Cerrados de la Cabeza/genética , Sustancia Blanca/metabolismo , Animales , Proteínas del Dominio Armadillo/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Axones/patología , Encéfalo/patología , Proteínas del Citoesqueleto/metabolismo , Traumatismos Cerrados de la Cabeza/metabolismo , Traumatismos Cerrados de la Cabeza/patología , Masculino , Ratones , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Actividad Motora/fisiología , Neuronas/metabolismo , Neuronas/patología , Reconocimiento en Psicología/fisiología , Sustancia Blanca/patología
6.
J Neurotrauma ; 37(2): 227-235, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31530217

RESUMEN

Both clinical and experimental studies have reported that mild traumatic brain injury (mTBI) can result in cognitive impairments in the absence of overt brain damage. Whether these impairments result from neuronal dysfunction/altered plasticity is an area that has received limited attention. In this study, we recorded activity of neurons in the cornu Ammonis (CA)1 subfield of the hippocampus in sham and mild lateral fluid percussion injured (mFPI) rats while these animals were performing an object location task. Electrophysiology results showed that the number of excitatory neurons encoding spatial information (i.e., place cells) was reduced in mFPI rats, and that these cells had broader and less stable place fields. Additionally, the in-field firing rate of place cells in sham operated, but not in mFPI, animals increased when objects within the testing arena were moved. Immunostaining indicated no visible damage or overall neuronal loss in mFPI brain sections. However, a reduction in the number of parvalbumin-positive inhibitory neurons in the CA1 subfield of mFPI animals was observed, suggesting that this reduction could have influenced place cell physiology. Alterations in spatial information content, place cell stability, and activity in mFPI rats coincided with poor performance in the object location task. These results indicate that altered place cell physiology may underlie the hippocampus-dependent cognitive impairments that result from mTBI.


Asunto(s)
Conmoción Encefálica/fisiopatología , Región CA1 Hipocampal/fisiopatología , Neuronas/patología , Navegación Espacial/fisiología , Animales , Masculino , Ratas , Ratas Sprague-Dawley
7.
Exp Neurol ; 323: 113081, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31655049

RESUMEN

Phosphatase and tensin homolog (PTEN)-induced kinase 1 (Pink1) is involved in mitochondrial quality control, which is essential for maintaining energy production and minimizing oxidative damage from dysfunctional/depolarized mitochondria. Pink1 mutations are the second most common cause of autosomal recessive Parkinson's disease (PD). In addition to characteristic motor impairments, PD patients also commonly exhibit cognitive impairments. As the hippocampus plays a prominent role in cognition, we tested if loss of Pink1 in mice influences learning and memory. While wild-type mice were able to perform a contextual discrimination task, age-matched Pink1 knockout (Pink1-/-) mice showed an impaired ability to differentiate between two similar contexts. Similarly, Pink1-/- mice performed poorly in a delayed alternation task as compared to age-matched controls. Poor performance in these cognitive tasks was not the result of overt hippocampal pathology. However, a significant reduction in hippocampal tyrosine hydroxylase (TH) protein levels was detected in the Pink1-/- mice. This decrease in hippocampal TH levels was also associated with reduced DOPA decarboxylase and dopamine D2 receptor levels, but not post-synaptic dopamine D1 receptor levels. These presynaptic changes appeared to be selective for dopaminergic fibers as hippocampal dopamine beta hydroxylase, choline acetyltransferase, and tryptophan hydroxylase levels were unchanged in Pink1-/- mice. Administration of the dopamine D1 receptor agonist SKF38393 to Pink1-/- mice was found to improve performance in the context discrimination task. Taken together, our results show that Pink1 loss may alter dopamine signaling in the hippocampus, which could be a contributing mechanism for the observed learning and memory impairments.


Asunto(s)
Hipocampo/metabolismo , Aprendizaje/fisiología , Memoria/fisiología , Proteínas Quinasas/deficiencia , Tirosina 3-Monooxigenasa/metabolismo , Animales , Dopamina/metabolismo , Antagonistas de Dopamina/farmacología , Aprendizaje/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos Parkinsonianos/metabolismo , Receptores de Dopamina D1/metabolismo
8.
J Neurotrauma ; 36(13): 2147-2152, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30672378

RESUMEN

In the majority of cases, the cognitive and behavioral impairments resulting from a mild traumatic brain injury (TBI) (also referred to as concussion) wane within days to weeks. In contrast, these impairments can persist for months to years after repetitive mild TBI (rmTBI). The cellular and molecular mechanisms underlying these impairments are not well understood. In the present study, we examined the consequences of rmTBI (three weight drops each separated by 72 h) on brain tissue respiration, pathology, and cognitive performance in mice. The transcription factor nuclear factor-erythroid 2-realted factor 2 (Nrf2) has been demonstrated to enhance the expression of numerous cytoprotective genes. Carnosic acid (CA) has been shown to activate Nrf2 and suppress the proinflammatory transcription factor nuclear factor kappa B (NF-κB). Because contemporaneous activation of cytoprotective genes and inhibition of proinflammatory genes can be beneficial, we questioned whether CA can be used to mitigate the pathobiology of rmTBI. The rmTBI increased hippocampal adenosine triphosphate-linked tissue respiration and proton leak that were unaffected by CA treatment. The rmTBI also caused significant motor and cognitive dysfunction, as tested using the foot fault, Barnes maze, and novel object recognition tasks. These impairments occurred in the absence of visible neuronal or dendritic loss. Post-rmTBI administration of CA significantly improved motor and cognitive function, and decreased Gfap and Iba1 immunoreactivities within white matter tracks. Taken together, these results show that rmTBI can cause cognitive impairments in the absence of overt neuronal pathologies, and post-injury treatment with CA can lessen some of these impairments.


Asunto(s)
Abietanos/farmacología , Antioxidantes/farmacología , Conmoción Encefálica , Recuperación de la Función/efectos de los fármacos , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
9.
J Neurotrauma ; 35(2): 362-374, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29088998

RESUMEN

Acetylcholine is an excitatory neurotransmitter in the central nervous system that plays a key role in cognitive function, including learning and memory. Previous studies have shown that experimental traumatic brain injury (TBI) reduces cholinergic neurotransmission, decreases evoked release of acetylcholine, and alters cholinergic receptor levels. Galantamine (U.S. Food and Drug Administration approved for the treatment of vascular dementia and Alzheimer's disease) has been shown to inhibit acetylcholinesterase activity and allosterically potentiate nicotinic receptor signaling. We investigated whether acute administration of galantamine can reduce TBI pathology and improve cognitive function tested days after the termination of the drug treatment. Post-injury administration of galantamine was found to decrease TBI-triggered blood-brain barrier (BBB) permeability (tested 24 h post-injury), attenuate the loss of both GABAergic and newborn neurons in the ipsilateral hippocampus, and improve hippocampal function (tested 10 days after termination of the drug treatment). Specifically, significant improvements in the Morris water maze, novel object recognition, and context-specific fear memory tasks were observed in injured animals treated with galantamine. Although messenger RNAs for both M1 (Nos2, TLR4, and IL-12ß) and M2 (Arg1, CCL17, and Mcr1) microglial phenotypes were elevated post-TBI, galantamine treatment did not alter microglial polarization tested 24 h and 6 days post-injury. Taken together, these findings support the further investigation of galantamine as a treatment for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Encéfalo/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Galantamina/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/patología , Cognición/efectos de los fármacos , Masculino , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función
10.
J Neurochem ; 139(1): 106-19, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27379837

RESUMEN

Prolonged metabolic suppression in the brain is a well-characterized secondary pathology of both experimental and clinical traumatic brain injury (TBI). AMP-activated kinase (AMPK) acts as a cellular energy sensor that, when activated, regulates various metabolic and catabolic pathways to decrease ATP consumption and increase ATP synthesis. As energy availability after TBI is suppressed, we questioned if increasing AMPK activity after TBI would improve cognitive outcome. TBI was delivered using the electromagnetic controlled cortical impact model on male Sprague-Dawley rats (275-300 g) and C57BL/6 mice (20-25 g). AMPK activity within the injured parietal cortex and ipsilateral hippocampus was inferred by western blots using phospho-specific antibodies. The consequences of acute manipulation of AMPK signaling on cognitive function were assessed using the Morris water maze task. We found that AMPK activity is decreased as a result of injury, as indicated by reduced AMPK phosphorylation and corresponding changes in the phosphorylation of its downstream targets: ribosomal protein S6 and Akt Substrate of 160 kDa (AS160). Increasing AMPK activity after injury using the drugs 5-amino-1-ß-d-ribofuranosyl-imidazole-4-carboxamide or metformin did not affect spatial learning, but significantly improved spatial memory. Taken together, our results suggest that decreased AMPK activity after TBI may contribute to the cellular energy crisis in the injured brain, and that AMPK activators may have therapeutic utility. Increased phosphorylation of Thr172 activates AMP-activated protein kinase (AMPK) under conditions of low cellular energy availability. This leads to inhibition of energy consuming, while activating energy generating, processes. Hill et al., present data to indicate that TBI decreases Thr172 phosphorylation and that its stimulation by pharmacological agents offers neuroprotection and improves memory. These results suggest that decreased AMPK phosphorylation after TBI incorrectly signals the injured brain that excess energy is available, thereby contributing to the cellular energy crisis and memory impairments.


Asunto(s)
Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Activadores de Enzimas/farmacología , Nootrópicos/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Lesiones Traumáticas del Encéfalo/psicología , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/prevención & control , Trastornos de la Memoria/psicología , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Lóbulo Parietal/patología , Fosforilación , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología
11.
Front Syst Neurosci ; 10: 36, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27199685

RESUMEN

Methionine is an essential proteinogenic amino acid that is obtained from the diet. In addition to its requirement for protein biosynthesis, methionine is metabolized to generate metabolites that play key roles in a number of cellular functions. Metabolism of methionine via the transmethylation pathway generates S-adenosylmethionine (SAM) that serves as the principal methyl (-CH3) donor for DNA and histone methyltransferases (MTs) to regulate epigenetic changes in gene expression. SAM is also required for methylation of other cellular proteins that serve various functions and phosphatidylcholine synthesis that participate in cellular signaling. Under conditions of oxidative stress, homocysteine (which is derived from SAM) enters the transsulfuration pathway to generate glutathione, an important cytoprotective molecule against oxidative damage. As both experimental and clinical studies have shown that traumatic brain injury (TBI) alters DNA and histone methylation and causes oxidative stress, we examined if TBI alters the plasma levels of methionine and its metabolites in human patients. Blood samples were collected from healthy volunteers (HV; n = 20) and patients with mild TBI (mTBI; GCS > 12; n = 20) or severe TBI (sTBI; GCS < 8; n = 20) within the first 24 h of injury. The levels of methionine and its metabolites in the plasma samples were analyzed by either liquid chromatography-mass spectrometry or gas chromatography-mass spectrometry (LC-MS or GC-MS). sTBI decreased the levels of methionine, SAM, betaine and 2-methylglycine as compared to HV, indicating a decrease in metabolism through the transmethylation cycle. In addition, precursors for the generation of glutathione, cysteine and glycine were also found to be decreased as were intermediate metabolites of the gamma-glutamyl cycle (gamma-glutamyl amino acids and 5-oxoproline). mTBI also decreased the levels of methionine, α-ketobutyrate, 2 hydroxybutyrate and glycine, albeit to lesser degrees than detected in the sTBI group. Taken together, these results suggest that decreased levels of methionine and its metabolic products are likely to alter cellular function in multiple organs at a systems level.

12.
J Neurosci ; 36(9): 2809-18, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26937017

RESUMEN

Traumatic brain injury (TBI) is a major human health concern that has the greatest impact on young men and women. The breakdown of the blood-brain barrier (BBB) is an important pathological consequence of TBI that initiates secondary processes, including infiltration of inflammatory cells, which can exacerbate brain inflammation and contribute to poor outcome. While the role of inflammation within the injured brain has been examined in some detail, the contribution of peripheral/systemic inflammation to TBI pathophysiology is largely unknown. Recent studies have implicated vagus nerve regulation of splenic cholinergic nicotinic acetylcholine receptor α7 (nAChRa7) signaling in the regulation of systemic inflammation. However, it is not known whether this mechanism plays a role in TBI-triggered inflammation and BBB breakdown. Following TBI, we observed that plasma TNF-α and IL-1ß levels, as well as BBB permeability, were significantly increased in nAChRa7 null mice (Chrna7(-/-)) relative to wild-type mice. The administration of exogenous IL-1ß and TNF-α to brain-injured animals worsened Evans Blue dye extravasation, suggesting that systemic inflammation contributes to TBI-triggered BBB permeability. Systemic administration of the nAChRa7 agonist PNU-282987 or the positive allosteric modulator PNU-120596 significantly attenuated TBI-triggered BBB compromise. Supporting a role for splenic nAChRa7 receptors, we demonstrate that splenic injection of the nicotinic receptor blocker α-bungarotoxin increased BBB permeability in brain-injured rats, while PNU-282987 injection decreased such permeability. These effects were not seen when α-bungarotoxin or PNU-282987 were administered to splenectomized, brain-injured rats. Together, these findings support the short-term use of nAChRa7-activating agents as a strategy to reduce TBI-triggered BBB permeability. SIGNIFICANCE STATEMENT: Breakdown of the blood-brain barrier (BBB) in response to traumatic brain injury (TBI) allows for the accumulation of circulating fluids and proinflammatory cells in the injured brain. These processes can exacerbate TBI pathology and outcome. While the role of inflammation in the injured tissue has been examined in some detail, the contribution of peripheral inflammation in BBB breakdown and ensuing pathology has not been well defined. We present experimental evidence to indicate that the stimulation of nicotinic acetylcholine α7 receptors (nAChRa7s) can reduce peripheral inflammation and BBB breakdown after TBI. These results suggest that activators of nAChRa7 may have therapeutic utility for the treatment of TBI.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Lesiones Encefálicas/sangre , Lesiones Encefálicas/patología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Análisis de Varianza , Animales , Lesiones Encefálicas/complicaciones , Modelos Animales de Enfermedad , Encefalitis/etiología , Ensayo de Inmunoadsorción Enzimática , Interleucina-1beta/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Permeabilidad , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/sangre , Receptor Nicotínico de Acetilcolina alfa 7/genética , Factor de von Willebrand/metabolismo
13.
J Neurotrauma ; 32(2): 139-48, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25027811

RESUMEN

Cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) signaling is required for short- and long-term memory. In contrast, enhanced PKA activity has been shown to impair working memory, a prefrontal cortex (PFC)-dependent, transient form of memory critical for cognition and goal-directed behaviors. Working memory can be impaired after traumatic brain injury (TBI) in the absence of overt damage to the PFC. The cellular and molecular mechanisms that contribute to this deficit are largely unknown. In the present study, we examined whether altered PKA signaling in the PFC as a result of TBI is a contributing mechanism. We measured PKA activity in medial PFC (mPFC) tissue homogenates prepared from sham and 14-day postinjury rats. PKA activity was measured both when animals were inactive and when actively engaged in a spatial working memory task. Our results demonstrate, for the first time, that PKA activity in the mPFC is actively suppressed in uninjured animals performing a working memory task. By comparison, both basal and working memory-related PKA activity was elevated in TBI animals. Inhibition of PKA activity by intra-mPFC administration of Rp-cAMPS into TBI animals had no influence on working memory performance 30 min postinfusion, but significantly improved working memory when tested 24 h later. This improvement was associated with reduced glutamic acid decarboxylase 67 messenger RNA levels. Taken together, these results suggest that TBI-associated working memory dysfunction may result, in part, from enhanced PKA activity, possibly leading to altered expression of plasticity-related genes in the mPFC.


Asunto(s)
Lesiones Encefálicas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Memoria a Corto Plazo/fisiología , Corteza Prefrontal/metabolismo , Animales , Lesiones Encefálicas/psicología , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
14.
Proc Natl Acad Sci U S A ; 108(10): 4200-5, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21368136

RESUMEN

Despite ubiquitous expression and a high level of metastasis-associated protein 1 (MTA1) coregulator, the physiological role of the MTA1 coactivator remains unknown. We found that MTA1 is a bona fide coactivator and stimulator of tyrosine hydroxylase (TH) transcription in neuronal cells and that MTA1-null mice had lower TH expression in the striatum and substantial nigra. MTA1 physically achieves these functions by interacting directly with DJ1 (Parkinson disease 7) and in turn recruits the DJ1/MTA1/RNA polymerase II complex to the bicoid binding element (BBE) in the TH promoter. Furthermore, we found that the MTA1/DJ1 complex is required for optimum stimulation of the TH expression by paired like homeodomain transcription factor (Pitx3) homeodomain transcription factor and that the MTA1/DJ1 complex is recruited to the TH gene chromatin via the direct interaction of MTA1 with Pitx3. These findings reveal a role for MTA1 as an upstream coactivator of TH and advance the notion of polygenic regulation of a disease-causing gene by coordinated interactions of three regulatory proteins.


Asunto(s)
Transcripción Genética/genética , Tirosina 3-Monooxigenasa/genética , Animales , Cuerpo Estriado/enzimología , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/fisiología , Ratones , Ratones Noqueados , Proteínas Represoras , Sustancia Negra/enzimología , Transactivadores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
15.
Learn Mem ; 14(8): 554-63, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17690339

RESUMEN

The prefrontal cortex is necessary for directing thought and planning action. Working memory, the active, transient maintenance of information in mind for subsequent monitoring and manipulation, lies at the core of many simple, as well as high-level, cognitive functions. Working memory has been shown to be compromised in a number of neurological and psychiatric conditions and may contribute to the behavioral and cognitive deficits associated with these disorders. It has been theorized that working memory depends upon reverberating circuits within the prefrontal cortex and other cortical areas. However, recent work indicates that intracellular signals and protein dephosphorylation are critical for working memory. The present article will review recent research into the involvement of the modulatory neurotransmitters and their receptors in working memory. The intracellular signaling pathways activated by these receptors and evidence that indicates a role for G(q)-initiated PI-PLC and calcium-dependent protein phosphatase calcineurin activity in working memory will be discussed. Additionally, the negative influence of calcium- and cAMP-dependent protein kinase (i.e., calcium/calmodulin-dependent protein kinase II (CaMKII), calcium/diacylglycerol-activated protein kinase C (PKC), and cAMP-dependent protein kinase A (PKA)) activities on working memory will be reviewed. The implications of these experimental findings on the observed inverted-U relationship between D(1) receptor stimulation and working memory, as well as age-associated working memory dysfunction, will be presented. Finally, we will discuss considerations for the development of clinical treatments for working memory disorders.


Asunto(s)
Trastornos de la Memoria/metabolismo , Memoria a Corto Plazo/fisiología , Transducción de Señal/fisiología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Humanos , Trastornos de la Memoria/terapia , Neurotransmisores/fisiología , Fosforilación
16.
J Neurotrauma ; 23(7): 1094-102, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16866622

RESUMEN

Traumatic brain injury (TBI)--induced dysfunction of the prefrontal cortex causes many high-level cognitive deficits, including working memory (WM) dysfunction. WM lies at the core of many high-level functions, yet the cellular and molecular mechanisms underlying its dysfunction are poorly understood. Lesion and pharmacological studies in rodents have implicated the medial prefrontal cortex (mPFC), which includes the prelimbic/infralimbic (PL/IL) cortices, in WM tasks. These studies have shown that optimal levels of catecholamine neurotransmission are critical for normalcy of WM function, suggesting that alterations in their synthesis may play a role in WM dysfunction. Using the cortical impact injury model of traumatic brain injury which reproducibly causes working memory deficits in rodents, we have measured the protein levels and activity of tyrosine hydroxylase (TH), the rate-limiting enzyme for catecholamine biosynthesis, and tissue dopamine (DA) and norepinephrine (NE) levels in microdissected PL/IL tissues. Our results show that TBI increases TH protein levels, its activity and tissue DA and NE content in the PL/IL. These findings suggest that altered catecholamine signaling within the PL/IL may contribute to impaired PFC function, and may have implications in the design and implementation of strategies to alleviate prefrontal dysfunction in brain injury patients.


Asunto(s)
Lesiones Encefálicas/metabolismo , Catecolaminas/biosíntesis , Corteza Prefrontal/metabolismo , Animales , Lesiones Encefálicas/patología , Masculino , Corteza Prefrontal/patología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
17.
J Neurosci ; 26(16): 4236-46, 2006 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-16624944

RESUMEN

Working memory (WM), the ability to transiently hold information in mind, is essential for high-level cognitive functions that are often impaired in brain-injured patients. The cellular and molecular mechanisms contributing to WM deficits, which can manifest in the absence of overt damage, in these patients are unknown. The function of the dorsolateral prefrontal cortex in humans and monkeys, and the medial prefrontal cortex (mPFC), in rodents is critical for WM. We demonstrate that controlled cortical impact injury of rats causes a long-lasting WM impairment that is associated with increased levels of the GABA-synthesizing enzyme glutamic acid decarboxylase 67 (GAD67) in the mPFC for up to 1 month after injury. A single administration of dopamine D1 antagonists at 14 d after injury is sufficient to decrease GAD67 levels and restore WM for at least 1 week. These findings indicate that inhibition of prefrontal neuronal activity contributes to WM deficits and that strategies to reduce GAD67 expression can offer prolonged WM improvement in brain-injured patients.


Asunto(s)
Lesiones Encefálicas/enzimología , Glutamato Descarboxilasa/biosíntesis , Trastornos de la Memoria/enzimología , Corteza Prefrontal/enzimología , Receptores de Dopamina D1/antagonistas & inhibidores , Animales , Lesiones Encefálicas/tratamiento farmacológico , Antagonistas de Dopamina/farmacología , Antagonistas de Dopamina/uso terapéutico , Antagonistas del GABA/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/fisiología , Isoenzimas/biosíntesis , Trastornos de la Memoria/tratamiento farmacológico , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D1/metabolismo
18.
Brain Res ; 1086(1): 142-51, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16626642

RESUMEN

High dopamine levels can contribute to neuronal dysfunction, impair plasticity and be toxic to neuronal cells in pathological conditions. The synthesis of dopamine is regulated by phosphorylation of the rate-limiting enzyme tyrosine hydroxylase (TH) under physiological conditions, with the phosphorylation of Ser31 and Ser40 directly increasing TH activity. Although a third phosphorylation site, Ser19, does not appear to directly regulate TH activity in physiological conditions, its role in pathological conditions is poorly understood. In this study, we examined the effects of serum deprivation (to mimic loss of retrogradely/anterogradely transported target-derived neurotrophic factors following axonal injury) and glutamate receptor stimulation (to mimic excitotoxicity) on TH phosphorylation and activity in a cell line and in mesencephalic primary culture cells. In addition, we also tested whether glial cell line-derived neurotrophic factor (GDNF) can alter these changes. We demonstrate that serum-deprivation resulted in a sustained increase in Ser19 phosphorylation beginning at 3 h and lasting up to 10 h without any detectable change in Ser31 or Ser40 phosphorylation within this time frame. This increase in Ser19 phosphorylation was associated with enhanced TH activity and was due, in part, to glutamate-receptor-mediated calcium influx and possibly calcium/calmodulin-dependent protein kinase II (CaMKII) activation. Interestingly in this serum-deprivation model, GDNF blocked the increase in Ser19 phosphorylation and TH activity at the 10-h time point following serum deprivation. Furthermore, GDNF also blocked the glutamate-mediated increase in Ser19 phosphorylation in rat primary mesencephalic neuronal cultures. Taken together, these findings suggest that GDNF may reduce dopamine synthesis in pathological conditions.


Asunto(s)
Medio de Cultivo Libre de Suero/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Neuronas/efectos de los fármacos , Serina/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Análisis de Varianza , Animales , Western Blotting/métodos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular Tumoral , Células Cultivadas , Interacciones Farmacológicas , Embrión de Mamíferos , Agonistas de Aminoácidos Excitadores/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Ácido Glutámico/farmacología , Humanos , Mesencéfalo/citología , N-Metilaspartato/farmacología , Neuroblastoma , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Factores de Tiempo
19.
Neurosci Lett ; 393(2-3): 108-12, 2006 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-16233958

RESUMEN

Stroke is the third leading cause of death and disability in the United States. As several biochemical mechanisms have been proposed to contribute to stroke pathophysiology, treatments acting on multiple targets may be desirable. Sulforaphane (SUL), a naturally occurring isothiocyanate present in cruciferous vegetables, has been shown to induce the expression of multiple NF-E2-related factor-2 (Nrf2) responsive genes. In the present study, we demonstrate that systemically administered SUL can enter the brain as determined by increased mRNA and protein levels of the Nrf2-responsive gene heme oxygenase 1 (HO-1). Delayed administration (15 min) of a single dose of SUL significantly decreased cerebral infarct volume following focal ischemia, suggesting a potential therapeutic value for this compound.


Asunto(s)
Infarto Encefálico/tratamiento farmacológico , Isquemia Encefálica/complicaciones , Fármacos Neuroprotectores/uso terapéutico , Tiocianatos/uso terapéutico , Análisis de Varianza , Animales , Northern Blotting , Infarto Encefálico/etiología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Inmunohistoquímica/métodos , Isotiocianatos , Masculino , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Fosfopiruvato Hidratasa/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Long-Evans , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sulfóxidos , Tiocianatos/química
20.
Neurochem Res ; 29(6): 1275-86, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15176484

RESUMEN

It has been estimated that 50% of human transcriptome, the collection of mRNA in a cell, is expressed in the brain, making it one of the most complex organs to understand in terms of genomic responses to injury. The availability of genome sequences for several organisms coupled with the increasing affordability of microarray technologies makes it feasible to monitor the mRNA levels of thousands of genes simultaneously. In this paper, we provide an overview of findings using both cDNA- and oligonucleotide-based microarray analyses after experimental traumatic brain injury (TBI). Specifically, the utility of this methodology as a means of cataloging the biochemical sequelae of brain trauma and elucidating novel genes or pathways for further study is discussed. Furthermore, we offer future directions for the continued evaluation of microarray results and discuss the usefulness of microarray techniques as a testing format for determining the efficacy of mechanism-based therapies.


Asunto(s)
Lesiones Encefálicas/genética , Lesiones Encefálicas/fisiopatología , Encéfalo/fisiopatología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Encéfalo/patología , ADN Complementario/genética , Regulación de la Expresión Génica/genética , Humanos , Oligodesoxirribonucleótidos , Reproducibilidad de los Resultados , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA