Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell Mol Life Sci ; 76(17): 3301-3310, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31073743

RESUMO

The channel kinase (chanzyme) transient receptor potential melastatin-like 7 (TRPM7) has a unique dual protein structure composed of an ion channel with an α-kinase domain on its C-terminus. In the nervous system, under physiological conditions, TRPM7 contributes to critical neurobiological processes ranging from synaptic transmission to cognitive functions. Following certain pathological triggers, TRPM7 mediates neurotoxicity, neuro-injuries, and neuronal death. Here, we summarize the current knowledge of TRPM7 functions in neuronal systems in health and disease. The molecular mechanisms by which this chanzyme might regulate synaptic and cognitive functions are discussed. We also discuss the lack of knowledge regarding the molecular mechanisms responsible for turning TRPM7 into "a vicious tool" that mediates neuronal death following certain pathological triggers. Some synthetic and natural pharmacological modulators of the TRPM7 channel and its α-kinase are reviewed. We suggest that based on current knowledge, we should reshape our thinking regarding the implications of TRPM7 in neurological and neurodegenerative disorders. Moreover, we propose a paradigm shift concerning the targeting of TRPM7 as a therapeutic approach for treating certain neurological diseases. We agree that TRPM7 overexpression or overactivation may mediate neurodegenerative processes following certain triggers. However, TRPM7 dysfunction and/or downregulation might also be among the pathological changes leading to neurodegeneration. Consequently, further investigations are required before we decide whether blocking or activating the chanzyme is the correct therapeutic approach to treat certain neurological and/or neurodegenerative diseases.


Assuntos
Sistema Nervoso/metabolismo , Doenças Neurodegenerativas/patologia , Canais de Cátion TRPM/metabolismo , Humanos , Magnésio/metabolismo , Doenças Neurodegenerativas/metabolismo , Plasticidade Neuronal , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Zinco/metabolismo
2.
J Neurosci ; 33(19): 8423-41, 2013 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-23658180

RESUMO

Profound synapse loss is one of the major pathological hallmarks associated with Alzheimer's disease (AD) and might underlie memory impairment. Our previous work demonstrated that the magnesium ion is a critical factor in controlling synapse density/plasticity. Here, we investigated whether elevation of brain magnesium by the use of a recently developed compound, magnesium-l-threonate (MgT), can ameliorate the AD-like pathologies and cognitive deficits in the APPswe/PS1dE9 mice, a transgenic (Tg) mouse model of AD. MgT treatment reduced Aß plaque and prevented synapse loss and memory decline in the Tg mice. Strikingly, MgT treatment was effective even when given to the mice at the end stage of their AD-like pathological progression. To explore how elevation of brain magnesium ameliorates the AD-like pathologies in the brains of Tg mice, we studied molecules critical for APP metabolism and signaling pathways implicated in synaptic plasticity/density. In the Tg mice, the NMDAR/CREB/BDNF signaling was downregulated, whereas calpain/calcineurin/Cdk5 neurodegenerative signaling and ß-secretase (BACE1) expression were upregulated. MgT treatment prevented the impairment of these signaling pathways, stabilized BACE1 expression, and reduced soluble APPß and ß-C-terminal fragments in the Tg mice. At the molecular level, elevation of extracellular magnesium prevented the high-Aß-induced reductions in synaptic NMDARs by preventing calcineurin overactivation in hippocampal slices. Correlation studies suggested that the protection of NMDAR signaling might underlie the stabilization of BACE1 expression. Our results suggest that elevation of brain magnesium exerts substantial synaptoprotective effects in a mouse model of AD and may have therapeutic potential for treating AD in humans.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Transtornos Cognitivos/etiologia , Magnésio/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Sinapses/patologia , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Butiratos/farmacologia , Butiratos/uso terapêutico , Transtornos Cognitivos/prevenção & controle , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Glutamato Descarboxilase/metabolismo , Humanos , Magnésio/uso terapêutico , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/líquido cefalorraquidiano , Presenilina-1/genética , Terminações Pré-Sinápticas/patologia , Terminações Pré-Sinápticas/ultraestrutura , Tempo de Reação/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/ultraestrutura , Distribuição Tecidual , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo
3.
STAR Protoc ; 5(2): 102967, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38492225

RESUMO

Uncontrollability could lead to behavioral adjustment or even giving up when facing repeated failure. Here, we detail a protocol to study the behavioral transition from action to no-action induced by prolonged uncontrollable experiences in mice. We describe the behavioral devices, video analysis, and the exponential learning curve fitting for mathematical assessment. We perform further validation experiments evaluating locomotor, social, and anxiety-/depression-like behaviors. This approach helps study neural mechanisms underlying adaptive decision-making when facing repeated failure. For complete details on the use and execution of this protocol, please refer to Li et al.1.


Assuntos
Comportamento Animal , Animais , Camundongos , Comportamento Animal/fisiologia , Masculino , Tomada de Decisões/fisiologia , Camundongos Endogâmicos C57BL , Ansiedade/fisiopatologia
4.
Behav Pharmacol ; 24(4): 255-63, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23764903

RESUMO

Enhancement of pattern separation could be helpful in improving the quality of normal daily learning and in treating individuals with cognitive impairment and certain psychiatric disorders. Previously, we have shown that elevating brain magnesium, by a novel magnesium compound (magnesium-L-threonate; MgT), enhances extinction of fear memory without enhancing amygdala-dependent fear memory. Here, we investigated the effects of MgT treatment on contextual-fear memory and subsequent pattern separation. Sprague-Dawley male rats were treated with MgT for 4 weeks and memory was evaluated using a spatial-context fear conditioning task. The pattern separation ability of MgT-treated rats was assessed using a spatial-context-discrimination task. MgT treatment did not enhance the retention of contextual-fear memory. Interestingly, the ability to discriminate between two, more or less distinct, contexts was enhanced in MgT-treated rats. Our results suggest that elevation of brain magnesium might be helpful in enhancing spatial-context discrimination and/or pattern separation besides preventing aversive-event-induced overgeneralization of fear.


Assuntos
Condicionamento Psicológico/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Compostos de Magnésio/farmacologia , Percepção Espacial/efeitos dos fármacos , Análise de Variância , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Discriminação Psicológica/efeitos dos fármacos , Divórcio , Reação de Congelamento Cataléptica/efeitos dos fármacos , Magnésio/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Treonina/farmacologia
5.
Channels (Austin) ; 17(1): 2200874, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37040321

RESUMO

Transient receptor potential melastatin-like 7 (TRPM7) is a key player in various physiological and pathological processes. TRPM7 channel activity is regulated by different factors. The effects of cleavage of different domains on channel activity remain unknown. Here, we constructed several TRPM7 clones and explored the effects of truncating the mouse TRPM7 at different locations on the ion channel activity in two cell lines. We compared the clones' activity with the full-length TRPM7 and the native TRPM7 in transfected and untransfected cells. We also expressed fluorescently tagged truncated clones to examine their protein stability and membrane targeting. We found that truncating the kinase domain induced reduction in TRPM7 channel activity. Further truncations beyond the kinase (serine/threonine rich domain and/or coiled-coil domain) did not result in further reductions in channel activity. Two truncated clones lacking the TRP domain or the melastatin homology domain had a completely nonfunctional channel apparently due to disruption of protein stability. We identified the shortest structure of TRPM7 with measurable channel activity. We found that the truncated TRPM7 containing only S5 and S6 domains retained some channel activity. Adding the TRP domain to the S5-S6 resulted in a significant increase in channel activity. Finally, our analysis showed that TRPM7 outward currents are more sensitive to truncations than inward currents. Our data provide insights on the effects of truncating TRPM7 at different locations on the channel functions, highlighting the importance of different domains in impacting channel activity, protein stability, and/or membrane targeting.


Assuntos
Canais de Cátion TRPM , Canais de Potencial de Receptor Transitório , Camundongos , Animais , Proteínas Serina-Treonina Quinases/metabolismo , Canais de Cátion TRPM/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Transdução de Sinais , Linhagem Celular
6.
Sci Signal ; 16(793): eade6325, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37433006

RESUMO

Altered abundance or activity of the dual-function transient receptor potential melastatin-like 7 (TRPM7) protein is implicated in neurodegenerative disorders, including Alzheimer's disease (AD). Toxic aggregation of amyloid-ß (Aß) in neurons is implicated in AD pathology. Here, we found that the kinase activity of TRPM7 is important to stimulate the degradation of Aß. TRPM7 expression was decreased in hippocampal tissue samples from patients with AD and two mouse models of AD (APP/PS1 and 5XFAD). In cultures of hippocampal neurons from mice, overexpression of full-length TRPM7 or of its functional kinase domain M7CK prevented synapse loss induced by exogenous Aß. In contrast, this neuroprotection was not afforded by overexpression of either the functional ion channel portion alone or a TRPM7 mutant lacking kinase activity. M7CK overexpression in the hippocampus of young and old 5XFAD mice prevented and reversed, respectively, memory deficits, synapse loss, and Aß plaque accumulation. In both neurons and mice, M7CK interacted with and activated the metalloprotease MMP14 to promote Aß degradation. Thus, TRPM7 loss in patients with AD may contribute to the associated Aß pathology.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Canais de Cátion TRPM , Animais , Camundongos , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Cognição , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Canais de Cátion TRPM/genética
7.
Research (Wash D C) ; 6: 0120, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37223470

RESUMO

Studies have demonstrated that a functional network of meningeal lymphatic vessels exists in the brain. However, it is unknown whether lymphatic vessels could also extend deep into the brain parenchyma and whether the vessels could be regulated by stressful life events. We used tissue clearing techniques, immunostaining, light-sheet whole-brain imaging, confocal imaging in thick brain sections and flow cytometry to demonstrate the existence of lymphatic vessels deep in the brain parenchyma. Chronic unpredictable mild stress or chronic corticosterone treatment was used to examine the regulation of brain lymphatic vessels by stressful events. Western blotting and coimmunoprecipitation were used to provide mechanistic insights. We demonstrated the existence of lymphatic vessels deep in the brain parenchyma and characterized their features in the cortex, cerebellum, hippocampus, midbrain, and brainstem. Furthermore, we showed that deep brain lymphatic vessels can be regulated by stressful life events. Chronic stress reduced the length and areas of lymphatic vessels in the hippocampus and thalamus but increased the diameter of lymphatic vessels in the amygdala. No changes were observed in prefrontal cortex, lateral habenula, or dorsal raphe nucleus. Chronic corticosterone treatment reduced lymphatic endothelial cell markers in the hippocampus. Mechanistically, chronic stress might reduce hippocampal lymphatic vessels by down-regulating vascular endothelial growth factor C receptors and up-regulating vascular endothelial growth factor C neutralization mechanisms. Our results provide new insights into the characteristic features of deep brain lymphatic vessels, as well as their regulation by stressful life events.

8.
Neuron ; 111(17): 2727-2741.e7, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37352858

RESUMO

Persistence in the face of failure helps to overcome challenges. But the ability to adjust behavior or even give up when the task is uncontrollable has advantages. How the mammalian brain switches behavior when facing uncontrollability remains an open question. We generated two mouse models of behavioral transition from action to no-action during exposure to a prolonged experience with an uncontrollable outcome. The transition was not caused by pain desensitization or muscle fatigue and was not a depression-/learned-helplessness-like behavior. Noradrenergic neurons projecting to GABAergic neurons within the orbitofrontal cortex (OFC) are key regulators of this behavior. Fiber photometry, microdialysis, mini-two-photon microscopy, and tetrode/optrode in vivo recording in freely behaving mice revealed that the reduction of norepinephrine and downregulation of alpha 1 receptor in the OFC reduced the number and activity of GABAergic neurons necessary for driving action behavior resulting in behavioral transition. These findings define a circuit governing behavioral switch in response to prolonged uncontrollability.


Assuntos
Encéfalo , Desamparo Aprendido , Camundongos , Animais , Córtex Pré-Frontal/fisiologia , Mamíferos
9.
J Neurosci ; 31(42): 14871-81, 2011 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22016520

RESUMO

Anxiety disorders, such as phobias and posttraumatic stress disorder, are among the most common mental disorders. Cognitive therapy helps in treating these disorders; however, many cases relapse or resist the therapy, which justifies the search for cognitive enhancers that might augment the efficacy of cognitive therapy. Studies suggest that enhancement of plasticity in certain brain regions such as the prefrontal cortex (PFC) and/or hippocampus might enhance the efficacy of cognitive therapy. We found that elevation of brain magnesium, by a novel magnesium compound [magnesium-l-threonate (MgT)], enhances synaptic plasticity in the hippocampus and learning and memory in rats. Here, we show that MgT treatment enhances retention of the extinction of fear memory, without enhancing, impairing, or erasing the original fear memory. We then explored the molecular basis of the effects of MgT treatment on fear memory and extinction. In intact animals, elevation of brain magnesium increased NMDA receptors (NMDARs) signaling, BDNF expression, density of presynaptic puncta, and synaptic plasticity in the PFC but, interestingly, not in the basolateral amygdala. In vitro, elevation of extracellular magnesium concentration increased synaptic NMDAR current and plasticity in the infralimbic PFC, but not in the lateral amygdala, suggesting a difference in their sensitivity to elevation of brain magnesium. The current study suggests that elevation of brain magnesium might be a novel approach for enhancing synaptic plasticity in a regional-specific manner leading to enhancing the efficacy of extinction without enhancing or impairing fear memory formation.


Assuntos
Tonsila do Cerebelo/metabolismo , Condicionamento Clássico/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Potenciação de Longa Duração/fisiologia , Magnésio/metabolismo , Córtex Pré-Frontal/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Análise de Variância , Animais , Comportamento Animal , Biofísica , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína de Ligação a CREB/metabolismo , Condicionamento Clássico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Técnicas In Vitro , Potenciação de Longa Duração/efeitos dos fármacos , Compostos de Magnésio/farmacologia , Masculino , Técnicas de Patch-Clamp , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinaptofisina/metabolismo , Fatores de Tempo
10.
Cells ; 11(7)2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-35406741

RESUMO

Oxidative stress induced by brain ischemia upregulates transient receptor potential melastatin-like-7 (TRPM7) expression and currents, which could contribute to neurotoxicity and cell death. Accordingly, suppression of TRPM7 reduces neuronal death, tissue damage and motor deficits. However, the neuroprotective effects of TRPM7 suppression in different cell types have not been investigated. Here, we found that induction of ischemia resulted in loss of parvalbumin (PV) gamma-aminobutyric acid (GABAergic) neurons more than Ca2+/calmodulin-kinase II (CaMKII) glutamatergic neurons in the mouse cortex. Furthermore, brain ischemia increased TRPM7 expression in PV neurons more than that in CaMKII neurons. We generated two lines of conditional knockout mice of TRPM7 in GABAergic PV neurons (PV-TRPM7-/-) and in glutamatergic neurons (CaMKII-TRPM7-/-). Following exposure to brain ischemia, we found that deleting TRPM7 reduced the infarct volume in both lines of transgenic mice. However, the volume in PV-TRPM7-/- mice was more significantly lower than that in the control group. Neuronal survival of both GABAergic and glutamatergic neurons was increased in PV-TRPM7-/- mice; meanwhile, only glutamatergic neurons were protected in CaMKII-TRPM7-/-. At the behavioral level, only PV-TRPM7-/- mice exhibited significant reductions in neurological and motor deficits. Inflammatory mediators such as GFAP, Iba1 and TNF-α were suppressed in PV-TRPM7-/- more than in CaMKII-TRPM7-/-. Mechanistically, p53 and cleaved caspase-3 were reduced in both groups, but the reduction in PV-TRPM7-/- mice was more than that in CaMKII-TRPM7-/- following ischemia. Upstream from these signaling molecules, the Akt anti-oxidative stress signaling was activated only in PV-TRPM7-/- mice. Therefore, deleting TRPM7 in GABAergic PV neurons might have stronger neuroprotective effects against ischemia pathologies than doing so in glutamatergic neurons.


Assuntos
Isquemia Encefálica , Neuroproteção , Canais de Cátion TRPM , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Neurônios GABAérgicos/metabolismo , Deleção de Genes , Isquemia/metabolismo , Camundongos , Parvalbuminas/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo
11.
Hippocampus ; 20(1): 174-85, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19330847

RESUMO

Exposure to chronic stress alters the number and morphology of neurons and glia in the hippocampal formation; however, little is known about possible changes in vasculature. Here, we examined the effect of chronic social defeat stress on hippocampal vascular supply in rats. Recent reports document that antidepressant treatment can influence angiogenesis in the hippocampus; therefore, we also studied the effect of antidepressant drug treatment on hippocampal capillarization. Animals were subjected to 5 weeks of daily social defeat by an aggressive conspecific and received concomitant, daily, oral fluoxetine (10 mg/kg) treatment during the last 4 weeks. Rat endothelial cell antigen-1 (RECA-1)-labeling of capillaries and quantitative stereological techniques were used to evaluate the treatment effects on capillary number. Special attention was paid to analysis of the vascular supply of the subgranular zone, which is regarded as an important component of the neurogenic niche for adult hippocampal neurogenesis. Chronic stress significantly decreased the number of microvessels by 30% in all hippocampal subregions, whereas fluoxetine treatment had no influence on capillary number. Furthermore, chronic stress decreased the capillarization of the subgranular zone to a similar extent, indicating that chronic stress affects the vascular niche for adult hippocampal neurogenesis. However, fluoxetine treatment had no impact on capillarization in the subgranular zone. We also detected a decrease in hippocampal volume in the animals as a result of stress, which was mildly altered by fluoxetine treatment. These pronounced changes in vascular supply may explain why the hippocampus is more vulnerable to insults when chronic stress precedes or coincides with other harmful conditions. Reduced microvasculature may also contribute to hippocampal volume decrease in stress-related disorders.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Fluoxetina/farmacologia , Hipocampo/efeitos dos fármacos , Microvasos/efeitos dos fármacos , Estresse Psicológico/tratamento farmacológico , Animais , Capilares/efeitos dos fármacos , Capilares/patologia , Doença Crônica , Hipocampo/irrigação sanguínea , Hipocampo/patologia , Masculino , Microvasos/patologia , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar , Predomínio Social , Nicho de Células-Tronco/efeitos dos fármacos , Nicho de Células-Tronco/patologia , Estresse Psicológico/patologia , Fatores de Tempo
12.
Neuron ; 106(1): 37-65.e5, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32027825

RESUMO

The Cre-loxP system is invaluable for spatial and temporal control of gene knockout, knockin, and reporter expression in the mouse nervous system. However, we report varying probabilities of unexpected germline recombination in distinct Cre driver lines designed for nervous system-specific recombination. Selective maternal or paternal germline recombination is showcased with sample Cre lines. Collated data reveal germline recombination in over half of 64 commonly used Cre driver lines, in most cases with a parental sex bias related to Cre expression in sperm or oocytes. Slight differences among Cre driver lines utilizing common transcriptional control elements affect germline recombination rates. Specific target loci demonstrated differential recombination; thus, reporters are not reliable proxies for another locus of interest. Similar principles apply to other recombinase systems and other genetically targeted organisms. We hereby draw attention to the prevalence of germline recombination and provide guidelines to inform future research for the neuroscience and broader molecular genetics communities.


Assuntos
Marcação de Genes/métodos , Integrases/genética , Neurônios/metabolismo , Oócitos/metabolismo , Recombinação Genética/genética , Espermatozoides/metabolismo , Animais , Feminino , Genes Reporter , Células Germinativas , Masculino , Camundongos , Camundongos Transgênicos , Mosaicismo
13.
J Neurosci Res ; 87(11): 2551-60, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19360902

RESUMO

Escitalopram (ES-CIT) is a widely used, highly specific antidepressant. Until now there has been very little evidence on how this drug under pathological conditions affects an important feature within the pathophysiology of stress-related disorders such as depression: the endogenous neurotrophins. By using a well-characterized rat model in which chronic stress induces depressive-like behavior, the levels of neurotrophins brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) were determined in representative brain regions and serum using a highly sensitive improved fluorometric two-site ELISA system. There was a significant increase of BDNF in the left and right cortices after stress treatment (twofold increase) that was reversed by application of ES-CIT. An ES-CIT-dependent NGF reduction in stressed rats was detectable in the right cortex only (P = 0.027). The left hippocampus revealed significantly higher amounts of BDNF (2.5-fold increase) protein than the right hippocampus. These interhemispheric differences were unrelated to stress or ES-CIT treatment in all animals. BDNF and NGF of the frontal cortex, cerebellum, and serum did not change between the study groups. There was a negative correlation between body weight and serum BDNF, independent of stress or ES-CIT treatment. In conclusion, BDNF and NGF show substantial changes in this rodent model of chronic social stress, which is susceptible to antidepressant treatment with ES-CIT and therefore may constitute a neurobiological correlate for the disease.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Citalopram/farmacologia , Fator de Crescimento Neural/metabolismo , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/metabolismo , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/patologia , Animais , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/sangue , Doença Crônica/tratamento farmacológico , Dominação-Subordinação , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Lateralidade Funcional , Modelos Lineares , Masculino , Fator de Crescimento Neural/sangue , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar , Estresse Psicológico/sangue
14.
Front Neurosci ; 13: 790, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31427916

RESUMO

Stroke remains the leading cause of long-term disability with limited options available to aid in recovery. Significant effort has been made to try and minimize neuronal damage following stroke with use of neuroprotective agents, however, these treatments have yet to show clinical efficacy. Regenerative interventions have since become of huge interest as they provide the potential to restore damaged neural tissue without being limited by a narrow therapeutic window. Neurotrophins, such as brain-derived neurotrophic factor (BDNF), and their high affinity receptors are actively produced throughout the brain and are involved in regulating neuronal activity and normal day-to-day function. Furthermore, neurotrophins are known to play a significant role in both protection and recovery of function following neurodegenerative diseases such as stroke and traumatic brain injury (TBI). Unfortunately, exogenous administration of these neurotrophins is limited by a lack of blood-brain-barrier (BBB) permeability, poor half-life, and rapid degradation. Therefore, we have focused this review on approaches that provide a direct and sustained neurotrophic support using pharmacological therapies and mimetics, physical activity, and potential drug delivery systems, including discussion around advantages and limitations for use of each of these systems. Finally, we discuss future directions of biomaterial drug-delivery systems, including the incorporation of heparan sulfate (HS) in conjunction with neurotrophin-based interventions.

15.
Behav Brain Res ; 340: 81-86, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-27555537

RESUMO

Transient receptor potential melastatin-like 7 (TRPM7) has a unique dual protein structure. It is an ion channel that has biophysical characteristics enabling divalent cations transport and a kinase domain involved in molecular events starting from modulating signaling pathways to inducing chromatin remodeling. Over the past 15 years, significant progress in the molecular and functional characterization of TRPM7 has been made in peripheral tissue and/or cell lines. TRPM7 appears to be involved in a plethora of physiological and pathological processes including embryonic development, organogenesis, cell proliferation and survival, and cell death following certain triggers. In the post-mitotic neuronal cells, however, the functional role of TRPM7 remains unclear. Majority of the progress in this area of research has focused on the potential role of TRPM7 in mediating neuronal death following ischemia-like and neuronal injuries-like conditions. Here, we summarize major progress on the biological roles of the TRPM7 during development and in mitotic systems (cell lines). Then, we address the recent developments made in neuronal systems. Besides its role in neuronal death, we emphasize on direct and indirect evidences that could link TRPM7 to fundamental neurobiological processes such as synaptic transmission, synapse remodeling, plasticity, cognitive functions as well as to some mental disorders. Therefore, we propose that an equivalent effort is demanded to systematically characterize the role of TRPM7 in healthy neural system before presenting it as a potential molecular target to treat neurodegenerative disorders or to prevent neuronal death following ischemia and/or neuronal injuries.


Assuntos
Encéfalo/metabolismo , Neurônios/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Humanos
17.
Cell Rep ; 23(12): 3480-3491, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29924992

RESUMO

The TRPM7 chanzyme contributes to several biological and pathological processes in different tissues. However, its role in the CNS under physiological conditions remains unclear. Here, we show that TRPM7 knockdown in hippocampal neurons reduces structural synapse density. The synapse density is rescued by the α-kinase domain in the C terminus but not by the ion channel region of TRPM7 or by increasing extracellular concentrations of Mg2+ or Zn2+. Early postnatal conditional knockout of TRPM7 in mice impairs learning and memory and reduces synapse density and plasticity. TRPM7 knockdown in the hippocampus of adult rats also impairs learning and memory and reduces synapse density and synaptic plasticity. In knockout mice, restoring expression of the α-kinase domain in the brain rescues synapse density/plasticity and memory, probably by interacting with and phosphorylating cofilin. These results suggest that brain TRPM7 is important for having normal synaptic and cognitive functions under physiological, non-pathological conditions.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Memória , Canais de Cátion TRPM/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Animais , Animais Recém-Nascidos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Potenciação de Longa Duração , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal/fisiologia , Fosforilação , Domínios Proteicos , Ratos Sprague-Dawley , Sinapses/metabolismo , Canais de Cátion TRPM/química
18.
Neuropsychopharmacology ; 32(7): 1490-503, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17164819

RESUMO

Profound neuroplastic changes have been demonstrated in various limbic structures after chronic stress exposure and antidepressant treatment in animal models of mood disorders. Here, we examined in rats the effect of chronic social stress and concomitant antidepressant treatment on cell proliferation in the medial prefrontal cortex (mPFC). We also examined possible hemispheric differences. Animals were subjected to 5 weeks of daily social defeat by an aggressive conspecific and received concomitant, daily, oral fluoxetine (10 mg/kg) during the last 4 weeks. Bromodeoxyuridine (BrdU) labeling and quantitative stereological techniques were used to evaluate the treatment effects on proliferation and survival of newborn cells in limbic structures such as the mPFC and the hippocampal dentate gyrus, in comparison with nonlimbic structures such as the primary motor cortex and the subventricular zone. Phenotypic analysis showed that neurogenesis dominated the dentate gyrus, whereas in the mPFC most newborn cells were glia, with smaller numbers of endothelial cells. Chronic stress significantly suppressed cytogenesis in the mPFC and neurogenesis in the dentate gyrus, but had minor effect in nonlimbic structures. Fluoxetine treatment counteracted the inhibitory effect of stress. Hemispheric comparison revealed that the rate of cytogenesis was significantly higher in the left mPFC of control animals, whereas stress inverted this asymmetry, yielding a significantly higher incidence of newborn cells in the right mPFC. Fluoxetine treatment abolished hemispheric asymmetry in both control and stressed animals. These pronounced changes in gliogenesis after chronic stress exposure may relate to the abnormalities of glial cell numbers reported in the frontolimbic areas of depressed patients.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fluoxetina/farmacologia , Córtex Pré-Frontal/fisiopatologia , Comportamento Social , Estresse Psicológico/fisiopatologia , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Bromodesoxiuridina , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Doença Crônica , Giro Denteado/efeitos dos fármacos , Giro Denteado/fisiopatologia , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/fisiopatologia , Lateralidade Funcional/efeitos dos fármacos , Lateralidade Funcional/fisiologia , Masculino , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Tamanho do Órgão/fisiologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Ratos , Ratos Wistar , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Estresse Psicológico/tratamento farmacológico
19.
Eur Neuropsychopharmacol ; 17(6-7): 417-29, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17182223

RESUMO

Using a model of depression in which chronic social stress induces depressive-like symptoms, we investigated effects of the selective serotonin-reuptake inhibitor (SSRI) citalopram on gene expression in the dorsal raphe nucleus of male rats. Expression of tryptophan hydroxylase (TPH) protein was found to be upregulated by the stress and normalized by citalopram, while mRNAs for genes TPH 1 and 2 were differentially affected. Citalopram had no effect on serotonin transporter mRNA but reduced serotonin-1A autoreceptor mRNA in stressed animals. The SSRI prevented the stress-induced upregulation of mRNA for CREB binding protein, synaptic vesicle glycoprotein 2b and the glial N-myc downstream-regulated gene 2, but increased mRNA for neuron-specific enolase (NSE) in both stressed and unstressed animals having no effect on stress-induced upregulation of NSE protein. These findings demonstrate that in the dorsal raphe nucleus of chronically stressed rats, citalopram normalizes TPH expression and blocks stress effects on distinct genes related to neurotransmitter release and neuroplasticity.


Assuntos
Citalopram/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Núcleos da Rafe/fisiologia , Serotonina/efeitos adversos , Estresse Fisiológico/genética , Estresse Fisiológico/fisiopatologia , Triptofano Hidroxilase/genética , Animais , Masculino , Modelos Animais , Reação em Cadeia da Polimerase , RNA/genética , RNA/isolamento & purificação , Núcleos da Rafe/efeitos dos fármacos , Ratos , Ratos Wistar , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Comportamento Social , Estresse Fisiológico/induzido quimicamente
20.
Front Mol Neurosci ; 10: 454, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29375311

RESUMO

Although studies provide insights into the neurobiology of stress and depression, the exact molecular mechanisms underlying their pathologies remain largely unknown. Long non-coding RNA (lncRNA) has been implicated in brain functions and behavior. A potential link between lncRNA and psychiatric disorders has been proposed. However, it remains undetermined whether IncRNA regulation, in the brain, contributes to stress or depression pathologies. In this study, we used a valid animal model of depression-like symptoms; namely learned helplessness, RNA-seq, Gene Ontology and co-expression network analyses to profile the expression pattern of lncRNA and mRNA in the hippocampus of mice. We identified 6346 differentially expressed transcripts. Among them, 340 lncRNAs and 3559 protein coding mRNAs were differentially expressed in helpless mice in comparison with control and/or non-helpless mice (inescapable stress resilient mice). Gene Ontology and pathway enrichment analyses indicated that induction of helplessness altered expression of mRNAs enriched in fundamental biological functions implicated in stress/depression neurobiology such as synaptic, metabolic, cell survival and proliferation, developmental and chromatin modification functions. To explore the possible regulatory roles of the altered lncRNAs, we constructed co-expression networks composed of the lncRNAs and mRNAs. Among our differentially expressed lncRNAs, 17% showed significant correlation with genes. Functional co-expression analysis linked the identified lncRNAs to several cellular mechanisms implicated in stress/depression neurobiology. Importantly, 57% of the identified regulatory lncRNAs significantly correlated with 18 different synapse-related functions. Thus, the current study identifies for the first time distinct groups of lncRNAs regulated by induction of learned helplessness in the mouse brain. Our results suggest that lncRNA-directed regulatory mechanisms might contribute to stress-induced pathologies; in particular, to inescapable stress-induced synaptic modifications.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa