Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Psychiatry ; 13: 805851, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35530016

RESUMEN

Background: Radiomics is characterized by high-throughput extraction of texture features from medical images and the mining of information that can potentially be used to define neuroimaging markers in many neurological or psychiatric diseases. However, there have been few studies concerning MRI radiomics in post-traumatic stress disorder (PTSD). The study's aims were to appraise changes in microstructure of the medial prefrontal cortex (mPFC) in a PTSD animal model, specifically single-prolonged stress (SPS) rats, by using MRI texture analysis. The feasibility of using a radiomics approach to classify PTSD rats was examined. Methods: Morris water maze and elevated plus maze were used to assess behavioral changes in the rats. Two hundred and sixty two texture features were extracted from each region of interest in T2-weighted images. Stepwise discriminant analysis (SDA) and LASSO regression were used to perform feature selection and radiomics signature building to identify mPFC radiomics signatures consisting of optimal features, respectively. Receiver operating characteristic curve plots were used to evaluate the classification performance. Immunofluorescence techniques were used to examine the expression of glial fibrillary acidic protein (GFAP) and neuronal nuclei (NeuN) in the mPFC. Nuclear pycnosis was detected using 4',6-diamidino-2-phenylindole (DAPI) staining. Results: Behavioral results indicated decreased learning and spatial memory performance and increased anxiety-like behavior after SPS stimulation. SDA analysis showed that the general non-cross-validated and cross-validated discrimination accuracies were 86.5% and 80.4%. After LASSO dimensionality reduction, 10 classification models were established. For classifying PTSD rats between the control and each SPS group, these models achieved AUCs of 0.944, 0.950, 0.959, and 0.936. Among four SPS groups, the AUCs were 0.927, 0.943, 0.967, 0.916, 0.932, and 0.893, respectively. The number of GFAP-positive cells and intensity of GFAP-IR within the mPFC increased 1 day after SPS treatment, and then decreased. The intensity of NeuN-IR and number of NeuN-positive cells significantly decreased from 1 to 14 days after SPS stimulation. The brightness levels of DAPI-stained nuclei increased in SPS groups. Conclusion: Non-invasive MRI radiomics features present an efficient and sensitive way to detect microstructural changes in the mPFC after SPS stimulation, and they could potentially serve as a novel neuroimaging marker in PTSD diagnosis.

2.
Cell Mol Neurobiol ; 42(3): 723-737, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32930942

RESUMEN

Studies from postmortem and animal models have revealed altered synapse morphology and function in the brain of posttraumatic stress disorder (PTSD). And the effects of PTSD on dendrites and spines have been reported, however, the effection on axon include microtubule (MT) and synaptic vesicles of presynaptic elements remains unknown. Hippocampus is involved in abnormal memory in PTSD. In the present study, we used the single prolonged stress (SPS) model to mimic PTSD. Quantitative real-time polymerase chain reaction (RT-qPCR) and high-throughput sequencing (GSE153081) were utilized to analyze differentially expressed genes (DEGs) in the hippocampus of control and SPS rats. Immunofluorescence and western blotting were performed to examine change in axon-related proteins. Synaptic function was evaluated by measuring miniature excitatory postsynaptic currents (mEPSCs). RNA-sequencing analysis revealed 230 significantly DEGs between the control and SPS groups. Gene Ontology analysis revealed upregulation in axonemal assembly, MT formation, or movement, but downregulation in axon initial segment and synaptic vesicles fusion in the hippocampus of SPS rats. Increased expression in tau, ß-tubulin MAP1B, KIF9, CCDC40, DNAH12 and decreased expression in p-tau, stathmin suggested SPS induced axon extension. Increased protein expression in VAMP, STX1A, Munc18-1 and decreased expression in synaptotagmin-1 suggested SPS induced more SNARE complex formation but decreased ability in synaptic vesicle fusion to presynaptic active zone membrane in the hippocampus of SPS rats. Further, low mEPSC frequency in SPS rats indicated dysfunction in presynaptic membrane. These results suggest that axon extension and synaptic vesicles fusion abnormality are involved in dysfunction of PTSD.


Asunto(s)
Trastornos por Estrés Postraumático , Animales , Axones/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratas , Trastornos por Estrés Postraumático/genética
3.
J Comp Neurol ; 530(3): 592-606, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34387361

RESUMEN

Orexin neurons play an important role in stress-related mental disorders including post-traumatic stress disorder (PTSD). Anxiety- and depression-related symptoms commonly occur in combination with PTSD. However, the role of the orexin system in mediating alterations in these affective symptoms remains unclear. The medial prefrontal cortex (mPFC) is implicated in both cognitive and emotional processing. In the present study, we investigated anxiety- and depression-related behavioral changes using the elevated plus maze, the sucrose preference test, and the open field test in male rats with single prolonged stress (SPS) induced-PTSD. The expression of orexin-A in the hypothalamus and orexin receptors (OX1R and OX2R) in the mPFC was detected and quantified by immunohistochemistry, western blotting, and real-time polymerase chain reaction. We found that the SPS rats exhibited enhanced levels of anxiety, reduced exploratory activities, and anhedonia. Furthermore, SPS resulted in reductions in the expression of orexin-A in the hypothalamus and the increased the expression of OX1R in the mPFC. The intracerebroventricular administration of orexin-A alleviated behavioral changes in SPS rats and partly restored the increased levels of OX1R in the mPFC. These results suggest that the orexin system plays a role in the anxiety- and depression-related symptoms observed in PTSD.


Asunto(s)
Trastornos por Estrés Postraumático , Animales , Ansiedad , Depresión/etiología , Modelos Animales de Enfermedad , Humanos , Masculino , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Corteza Prefrontal/metabolismo , Ratas , Ratas Wistar , Trastornos por Estrés Postraumático/metabolismo
4.
J Mol Neurosci ; 71(12): 2487-2499, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33738762

RESUMEN

Apoptosis of hippocampal neurons is one of the mechanisms of hippocampal atrophy in posttraumatic stress disorder (PTSD), and it is also an important cause of memory impairment in PTSD patients. Endoplasmic reticulum stress (ERS) mediated by activated transcription factor 6α (ATF6α)/site 1 protease (S1P)/S2P is involved in cell apoptosis, but it is not clear whether it is involved in hippocampal neuron apoptosis caused by PTSD. A PTSD rat model was constructed by the single prolonged stress (SPS) method. The study was divided into three parts. Experiment 1 included the control group, SPS 1 d group, SPS 7 d group, and SPS 14 d group. Experiment 2 included the control group, SPS 7 d group, SPS 7 d + AEBSF group, and control + AEBSF group. (4-(2-Aminoethyl)benzenesulfonyl fluoride hydrochloride (AEBSF) is an ATF6α pathway inhibitor). Experiment 3 included the control group, SPS 4 d group, SPS 4 d + AEBSF group, and control + AEBSF group. The protein and mRNA expression levels of ATF6α, glucose-regulated protein (GRP78), S1P, S2P, C/EBP homologous protein (CHOP), and caspase-12 in the hippocampus of PTSD rats were detected by immunohistochemistry, Western blotting and qRT-PCR. Apoptosis of hippocampal neurons was detected by TUNEL staining. In experiment 1, the protein and mRNA expression of ATF6α and GRP78 increased gradually in the SPS 1 d group and the SPS 7 d group but decreased in the SPS 14 d group (P < 0.01). In experiment 2, compared with that in the control group, the protein and mRNA expression of ATF6α, GRP78, S1P, S2P, CHOP, and caspase-12 and the apoptosis rate were significantly increased in the SPS 7 d group (P < 0.01). However, the protein and mRNA expression of ATF6α, GRP78, S1P, S2P, CHOP, and caspase-12 and the apoptosis rate were significantly decreased after AEBSF pretreatment (P < 0.01). In experiment 3, compared with that in the control group, the protein and mRNA expression of ATF6α, GRP78, S1P, S2P, CHOP, and caspase-12 and the apoptosis rate were increased in the SPS 14 d group (P < 0.05). However, the protein and mRNA expression of ATF6α, GRP78, S1P, S2P, CHOP, and caspase-12 and the apoptosis rate were decreased after AEBSF pretreatment (P < 0.05). SPS induced apoptosis of hippocampal neurons by activating ERS mediated by ATF6α, suggesting that ERS-induced apoptosis is involved in the occurrence of PTSD.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Apoptosis , Hipocampo/metabolismo , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Factor de Transcripción Activador 6/genética , Animales , Caspasa 12/genética , Caspasa 12/metabolismo , Estrés del Retículo Endoplásmico , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Hipocampo/citología , Masculino , Memoria , Neuronas/metabolismo , Proproteína Convertasas/genética , Ratas , Ratas Wistar , Serina Endopeptidasas/genética , Transducción de Señal , Trastornos por Estrés Postraumático/genética , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo
5.
Neuroscience ; 432: 126-136, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32112915

RESUMEN

Post-traumatic stress disorder (PTSD) patients exhibit abnormal learning and memory. Axons from orexin neurons in the lateral hypothalamus innervate the hippocampus, modulating learning and memory via the orexin 1 and 2 receptors (OX1R and OX2R). However, the role of the orexin system in the learning and memory dysfunction observed in PTSD is unknown. This was investigated in the present study using PTSD animal model-single prolonged stress (SPS) rats. Spatial learning and memory in the rats were evaluated with the Morris water maze (MWM) test; changes in body weight and food intake were recorded to assess changes in appetite; and the expression of orexin-A and its receptors in the hypothalamus and hippocampus was examined and quantified by immunohistochemistry, western blotting and real-time PCR. The results showed that spatial memory was impaired and food intake was decreased in SPS rats; this was accompanied by downregulation of orexin-A in the hypothalamus and upregulation of OX1R and OX2R in the hippocampus and of OX1R in the hypothalamus. Intracerebroventricular administration of orexin-A improved spatial memory and enhanced appetite in SPS rats and partly reversed the increases in OX1R and OX2R levels in the hippocampus and hypothalamus. These results suggest that the orexin system plays a critical role in the memory and appetite dysfunction observed in PTSD.


Asunto(s)
Trastornos por Estrés Postraumático , Animales , Humanos , Trastornos de la Memoria/tratamiento farmacológico , Receptores de Orexina , Orexinas , Ratas , Ratas Wistar , Trastornos por Estrés Postraumático/tratamiento farmacológico
6.
J Affect Disord ; 268: 173-182, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32174475

RESUMEN

BACKGROUND: Ras and Rab interactor 1 (Rin1) is predominantly expressed in memory-related brain regions, and has been reported to play an important role in fear memory. Increased expression of Rin1 in an animal model of posttraumatic stress disorder (PTSD) has been associated with enhanced acquisition of fear memories, but the exact mechanism of Rin1 in memory regulation are not clear. METHODS: Here, we used Rin1-knockout rats to examine the effect of Rin1 on fear memories by fear conditional test and the molecular mechanisms that regulate these effects by immunofluorescence, western blotting and TUNEL. RESULTS: Our results show that Rin1-knockout rats have a deficit in formation and extinction of Auditory fear memories. Lack of Rin1 results in enhanced apoptosis in the hippocampus through a pathway related to the mitochondria rather than the endoplasmic reticulum-related pathway. Importantly, the lack of Rin1 induces a decrease in α-amino-3­hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) found in the cytoplasm, but not in those found in the membrane. Expression of CaMKII (which is important for insertion of cytoplasmic AMPAR into the membrane) and stargazin (which is important for immobilization of AMPAR in the membrane) was not changed. The lack of Rin1 also induced changes in AMPAR distribution, from diffuse spread in the cells to clusters around the edge of the cell. Additionally, clustered AMPAR distribution showed a high degree of overlap with actin distribution. CONCLUSION: These findings indicate that Rin1 affects not only apoptosis, but also the concentration and distribution pattern of AMPAR, which are important in the formation and extinction of fear memory.


Asunto(s)
Miedo , Receptores AMPA , Animales , Apoptosis/genética , Hipocampo/metabolismo , Memoria , Ratas , Receptores AMPA/genética , Receptores AMPA/metabolismo
7.
J Mol Neurosci ; 70(4): 576-589, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31933182

RESUMEN

Posttraumatic stress disorder (PTSD) is closely related to brain structures of the memory loop such as the hippocampus, amygdala, and medial prefrontal cortex (mPFC). The fear gene stathmin plays an important role in regulating fear memory. However, whether the fear gene stathmin is related to fear memory loop anomalies caused by PTSD is unclear. A single-prolonged stress (SPS) rat model of PTSD was constructed. Wistar rats were randomly divided into 5 groups: the control group, SPS 1-day group, SPS 4-day group, SPS 7-day group, and SPS 14-day group. Then, we measured the protein and mRNA expression of stathmin, p-stathmin (Ser16, Ser25, Ser38, and Ser63), ß-tubulin, and MAP-1B in the hippocampus, amygdala, and mPFC in the 5 groups by immunohistochemistry, Western blotting, and qRT-PCR. The expression of the stathmin protein in the hippocampus, mPFC, and amygdala of the rat memory loop decreased gradually in the SPS 1-day group, the SPS 4-day group, and the SPS 7-day group, in which it was the lowest, and then increased. The trend of the expression of stathmin mRNA in the three areas of the memory loop was consistent with the trend of the expression of the stathmin protein. The trend of the protein expression of p-stathmin (Ser25 and Ser38) was opposite of that of stathmin; it reached a peak on the 7th day, and then decreased in the hippocampus. The protein expression of p-stathmin (Ser63) showed the same trend in the mPFC. The protein and mRNA expression of ß-tubulin and MAP-1B was consistent with that of p-stathmin; it reached a peak on the 7th day, and then decreased in the rat hippocampus, mPFC, and amygdala. Stathmin in the memory loop, especially in the hippocampus, regulates microtubule structure through its phosphorylation at Ser25 and Ser38 and thereby participates in the mediation of fear memory abnormalities in PTSD.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Hipocampo/metabolismo , Estatmina/genética , Trastornos por Estrés Postraumático/metabolismo , Animales , Masculino , Memoria , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Corteza Prefrontal/metabolismo , Ratas , Ratas Wistar , Estatmina/metabolismo , Trastornos por Estrés Postraumático/genética , Trastornos por Estrés Postraumático/fisiopatología , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
8.
Synapse ; 74(6): e22146, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31869485

RESUMEN

Post-traumatic stress disorder (PTSD) is a long-lasting mental disorder and accompanied by worse fear extinction. Enhanced fear memory or poor fear extinction are typical features of PTSD. Dysfunction of the serotonergic neurotransmitter system is involved in numerous mental and behavioral disorders. Monoamine oxidase A (MAOA) is important in the metabolism of serotonin and play an important role in behavious. The aim of this study was to explore the change of MAOA and effect of MAOA on fear memory in PTSD. We used single prolonged stress (SPS) to create animal model of PTSD. A startle/fear box and elevated plus maze were used to observe fear memory and anxiety level, respectively. We examined the expression of MAOA and synaptic marker protein, as well as the immunological activity of MAOA in the infralimbic cortex (IL) area, which is a critical brain region involved in emotions, especially fear regulation. We found increased anxiety-like behavior, dysfunction in fear extinction, and increased MAOA in SPS rats. After treatment with moclobemide (a selective inhibitor of MAOA), SPS rats showed significantly improved fear memory and decreased anxiety-like behavior, which indicated that moclobemide could reverse fear extinction deficit and attenuate abnormally increased levels of anxiety caused by SPS in short term. On the contrary, decreased PSD-95 and SYN1 expression in the IL region were also reversed by moclobemide. These results suggest that increased MAOA play a negative role in fear extinction and levels of anxiety in PTSD, which may be involved in change in PSD-95 and SYN1.


Asunto(s)
Ansiolíticos/farmacología , Extinción Psicológica , Moclobemida/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Trastornos por Estrés Postraumático/tratamiento farmacológico , Sinapsis/efectos de los fármacos , Animales , Ansiolíticos/uso terapéutico , Homólogo 4 de la Proteína Discs Large/metabolismo , Miedo , Sistema Límbico/efectos de los fármacos , Sistema Límbico/metabolismo , Masculino , Aprendizaje por Laberinto , Moclobemida/uso terapéutico , Inhibidores de la Monoaminooxidasa/uso terapéutico , Proteínas Qa-SNARE/metabolismo , Ratas , Ratas Wistar , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/fisiopatología , Sinapsis/metabolismo
9.
Psychoneuroendocrinology ; 107: 169-178, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31132569

RESUMEN

BACKGROUND: Stressors activate a wide spectrum of interacting hormonal and neuronal systems resulting in behavioral and physiological responses, with consequences for the development of psychopathology. Several recent studies demonstrated that treatment with the glucocorticoid receptor (GR) antagonist RU486 during adulthood normalized effects of early life stress. We aimed to evaluate the potential of RU486 to reverse stress-induced changes in an animal model of adult stress. METHOD: We employed the single-prolonged stress (SPS) model as a multimodal stress exposure protocol in male rats. SPS rats and unstressed controls were treated with RU486 on days 8, 9, 10 after stress exposure and the effects of treatment were evaluated after another 4 days. We determined body weight gain, corticosterone levels, behavioral reactivity in anxiety tests, and brain gene expression of c-fos, corticosteroid receptors, drivers of the stress response and genes (epi-)genitally linked to PTSD. RESULTS: RU486 affected body weight gain, corticosterone levels and open field behavior only in SPS rats. RU486 had history-independent effects in reducing fear in the elevated plus maze and fear conditioning behavior. Gene expression analysis showed a diversity of in- and interdependent effects of stress and RU486. CONCLUSION: The effects of RU486 applied 1 week after stress and measured 4 days after treatment demonstrate that in the state of post-SPS the GR-dependence of homeostatic processes has changed. This suggests that GR-mediated processes are part of allostatic regulation after adult stress. The normalization of a number of SPS-effects after RU486 treatment reinforces the potential of targeting GR for treatment of stress-related psychopathologies.


Asunto(s)
Miedo/efectos de los fármacos , Mifepristona/farmacología , Estrés Psicológico/metabolismo , Animales , Encéfalo/metabolismo , Corticosterona/metabolismo , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Miedo/fisiología , Hipocampo/metabolismo , Masculino , Mifepristona/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico/genética
10.
J Affect Disord ; 246: 682-694, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30611912

RESUMEN

BACKGROUND: Early life and stressful experiences affect hippocampal and amygdala structure and function. They also increase the incidence of mental and nervous system disorders in adults. However, prospective studies have yet to show if early-life experiences affect the risk/severity of post-traumatic stress disorder (PTSD). METHODS: We applied neonatal isolation (NI) alone, single prolonged stress (SPS) alone and NI + SPS to rats. We evaluated anxiety-like behavior and spatial memory of behavior using open field, elevated plus maze, and Morris water maze tests. Then, we measured expression of glucocorticoid receptors (GRs) and synaptic-related proteins by immunofluorescence, immunohistochemistry and western blotting in the hippocampus and amygdala. RESULTS: NI + SPS exacerbated the increased anxiety levels and impaired spatial memory induced by NI alone or SPS alone. NI alone or SPS alone induced varying degrees of change in expression of GRs and synaptic proteins (synapsin I and postsynaptic density protein-95) in the hippocampus and amygdala. There were opposite changes in GR expression in the hippocampal dentate gyrus and basolateral amygdala. The degree of such change was exacerbated considerably by NI + SPS. In addition, neuroligin (NLG)-1 and NLG-2 were distributed in postsynaptic sites of excitatory and inhibitory synapses, respectively. NI, SPS, and NI + SPS altered the patterns of NLG-1 and NLG-2 colocalization as well as their intensity. NI + SPS strengthened the increased ratio of NLG-1/NLG-2 in the hippocampus, but decreased this ratio in the amygdala. CONCLUSIONS: NI and SPS together induced greater degrees of change in anxiety and spatial memory, as well as GR and synaptic protein levels, in the hippocampus and amygdala than the changes induced by NI alone or SPS alone.


Asunto(s)
Plasticidad Neuronal/fisiología , Neuronas/fisiología , Receptores de Glucocorticoides/fisiología , Aislamiento Social/psicología , Estrés Psicológico/fisiopatología , Amígdala del Cerebelo/fisiopatología , Animales , Ansiedad/fisiopatología , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Masculino , Ratas , Ratas Wistar , Estrés Psicológico/psicología , Lóbulo Temporal/fisiopatología
11.
Mol Med Rep ; 17(1): 1099-1104, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115545

RESUMEN

The purpose of the present study was to investigate the role of endoplasmic reticulum (ER)­resident molecular chaperone proteins to identify whether these proteins were involved in post­traumatic stress disorder (PTSD). The present study detected changes of calreticulin (CRT), calnexin (CNX) and ERp57 in the amygdala of rats, which may with aim of providing a novel insight into the modulation effect of amygdala in PTSD. Single­prolonged stress (SPS) was applied to create the models of PTSD in rats. The expression levels of CRT, CNX and ERp57 were examined using immunohistochemistry or immunofluorescence, western blot analysis and reverse transcription­quantitative polymerase chain reaction (RT­qPCR). The results showed that SPS induced significant changes in CRT, CNX and ERp57 expression levels. Furthermore, the expression levels of CRT, CNX and ERp57 were significantly upregulated when compared to that in the control group after SPS exposure by western blot analysis (P<0.05). RT­qPCR analysis supported these results, indicating an upregulation of mRNA expression level. Taken together, the present findings suggest that SPS may induce changes to the expression of CRT, CNX and ERp57 in the amygdala of rats. The present study provides an insight into the effects of ER­resident molecular chaperones in the amygdala participating in PTSD, and provides the experimental basis and a mechanism for the pathophysiology of PTSD.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Chaperonas Moleculares/metabolismo , Estrés Psicológico , Animales , Biomarcadores , Peso Corporal , Conexinas/genética , Conexinas/metabolismo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Masculino , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Ratas
12.
Mol Med Rep ; 16(6): 8351-8358, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28983608

RESUMEN

The amygdalae are an important component of the human limbic system and exhibit a key role in emotional and behavioral reactions. Previous studies have demonstrated abnormal function and morphology in the amygdalae of post­traumatic stress disorder (PTSD)­like animal models, however the underlying molecular mechanisms remain elusive. The authors have previously demonstrated that PTSD induced increased apoptosis in the amygdala of PTSD­like animals. Cyclin D1 and cyclin­dependent kinase 4 (CDK4) are two important regulators of the cell cycle. The study explored the expression of cyclin D1 and CDK4 in the amygdala in PTSD. The single­prolonged stress (SPS) rat model was used as a PTSD­like model. Ultrastructural alterations of cells in the amygdala were observed using transmission electron microscopy (TEM). 4',6­Diamidino­2­phenylindole (DAPI) fluorescence was employed to detect nuclear pycnosis. Cyclin D1 and CDK4 expression in the amygdala cells was examined using immunofluorescence, Western blotting and reverse transcription­quantitative polymerase chain reaction. TEM revealed morphological alterations to the amygdala cells of the SPS rats. DAPI­stained nuclear brightness levels differed between the control and SPS groups. Expression of cyclin D1 and CDK4 in the amygdala increased gradually 1 day and 4 days following SPS stimulation, and peaked 7 days following SPS stimulation at the protein and mRNA levels, in comparison with the control rats. These findings suggest that SPS resulted in increased cyclin D1 and CDK4 expression, which may accelerate cell apoptosis. This may be associated with SPS­induced abnormal function and structure of the amygdala.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina/genética , Regulación de la Expresión Génica , Trastornos por Estrés Postraumático/genética , Amígdala del Cerebelo/patología , Amígdala del Cerebelo/ultraestructura , Animales , Biomarcadores , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Técnica del Anticuerpo Fluorescente , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/patología
13.
J Mol Neurosci ; 63(1): 28-35, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28752417

RESUMEN

In an animal model of post-traumatic stress disorder (PTSD), our previous studies showed mitochondrial stress-induced apoptosis in the hippocampus. Metformin, the most commonly prescribed anti-diabetic drug, exerts its effects through 5'-adenosine monophosphate-activated protein kinase (AMPK) activation. It was shown that a neuroprotective role was gradually established against stroke, spinal cord injury and Parkinson's disease. The aim of this study was to explore the role of the AMPK pathway in neuronal apoptosis in the hippocampus using a rat model of PTSD. The model PTSD rats received acute exposure to prolonged stress (single prolonged stress, SPS), followed by examination of the effects of genes and/or proteins related to the AMPK and oxidative stress pathways in the hippocampus with or without metformin preconditioning. The results indicated that the level of phosphorylated AMPK was markedly increased after SPS. Metformin protected the hippocampus as evidenced by abolishing down-regulation of the AMPK pathway and up-regulating expression of oxidative stress-related genes. These results indicated that metformin attenuated oxidative stress in the hippocampus in rats under SPS. AMPK pathway activation may be a novel therapeutic protocol for PTSD patients.


Asunto(s)
Antioxidantes/farmacología , Hipocampo/metabolismo , Hipoglucemiantes/farmacología , Metformina/farmacología , Estrés Oxidativo , Trastornos por Estrés Postraumático/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Antioxidantes/uso terapéutico , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Masculino , Metformina/uso terapéutico , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Quinasas/metabolismo , Ratas , Ratas Wistar , Trastornos por Estrés Postraumático/tratamiento farmacológico
14.
Front Behav Neurosci ; 11: 62, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28491025

RESUMEN

The molecular mechanism of fear memory is poorly understood. Therefore, the pathogenesis of post-traumatic stress disorder (PTSD), whose symptom presentation can enhance fear memory, remains largely unclear. Recent studies with knockout animals have reported that Rin1 and stathmin regulate fear memory. Rin1 inhibits acquisition and promotes memory extinction, whereas stathmin regulates innate and basal fear. The aim of our study was to examine changes in the expression of Rin1 and stathmin in different animal models of stress, particluarly traumatic stress. We used three animal traumatic stresses: single prolonged stress (SPS, which is a rodent model of PTSD), an immobilization-stress (IM) and a Loud sound stress (LSS), to examine the change and uniqueness in Rin1/stathmin expression. Behavioral tests of SPS rats demonstrated increased anxiety and contextual fear-conditioning. They showed decreased long-term potentiation (LTP), as well as decreased stathmin and increased Rin1 expression in the hippocampus and the amygdala. Expression of the stathmin effector, tubulin, and downstream molecules Rin1, Rab5, and Abl, appeared to increase. Rin1 and EphA4 were endogenously coexpressed in primary neurons after SPS stimulation. IM rats exhibited increased anxiety behavior and enhanced fear-conditioning to contextual and auditory stimuli. Similar changes in expression of Rin1/stathmin were observed in IM rats whereas no changes were observed in rats exposed to a loud sound. These data suggest that changes in expression of the Rin1 and stathmin genes may be involved in rodents with SPS and IM stresses, which provide valuable insight into fear memories under abnormal conditions, particularly in PTSD.

15.
Apoptosis ; 22(6): 753-768, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28391375

RESUMEN

Post-traumatic stress disorder (PTSD) is characterized with abnormal learning and memory. Impairments in learning and memory are closely associated with apoptosis in the medial prefrontal cortex (mPFC). We previously examined the endoplasmic reticulum (ER) stress was involved in the apoptosis in the mPFC of PTSD. The PERK pathway plays the important role in the ER stress-induced apoptosis. The aim of the present study was to explore the role of PERK pathway in neuronal apoptosis in the mPFC of rat models of PTSD. We used the single prolonged stress (SPS) to mimic PTSD in rats and studied the effects of the PERK pathway in mPFC. Learning and memory behavior were examined by Morris water maze and novel object recognition tests. Apoptosis in mPFC was detected by TUNEL staining. Our results showed decreased learning memory and increased apoptosis of mPFC neurons in rats exposed to SPS. SPS exposure upregulate mRNA expressions of PERK, p-PERK, eIF2α, p-eIF2α, nuclear ATF4 and C/EBP-homologous protein (CHOP) in mPFC neurons, but the protein levels of these molecules showed difference in magnitude and time course. GSK2606414 (an antagonist of PERK) treatment successfully reversed the above changes. These results suggested that the PERK pathway mediated SPS-induced neural apoptosis in the mPFC. These findings will be helpful in understanding mPFC-related pathogenesis of PTSD.


Asunto(s)
Neuronas/enzimología , Neuronas/patología , Corteza Prefrontal/fisiopatología , Transducción de Señal , Estrés Fisiológico , eIF-2 Quinasa/metabolismo , Factor de Transcripción Activador 4/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Caspasa 12/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Indoles/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
16.
J Mol Neurosci ; 62(1): 43-54, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28341893

RESUMEN

Autophagy, or type II programmed cell death, plays a crucial role in many nervous system diseases. However, few studies have examined the role of autophagy in post-traumatic stress disorder (PTSD), and the mechanisms underlying PTSD are poorly understood. The objective of this research was to explore the expression of three important autophagy-related proteins, Beclin-1, microtubule-associated protein 1 light chain 3 (LC3), and p62/SQSTM1 (p62), in the medial prefrontal cortex (mPFC) of an animal model of PTSD to identify changes in autophagic activity during PTSD pathogenesis. PTSD was induced in rats by exposure to a single-prolonged stress (SPS). The Morris water maze was used to assess cognitive changes in rats from the SPS and control groups. Transmission electron microscopy (TEM) was employed to observe mPFC morphological changes. Immunohistochemistry, immunofluorescence, and Western blotting techniques were used to detect expression of Beclin-1, LC3, and p62 in the mPFC. The Morris water maze test results showed that the escape latency time was increased and that the percent time in the target quadrant was decreased in the SPS group compared with that in the control group. Numerous visible autolysosomes in mPFC neurons were observed using TEM after SPS stimulation. Compared with that in the control group, the expression of Beclin-1 and the LC3-II/I ratio significantly decreased at 1 day, then increased and peaked at 7 days, and slightly decreased at 14 days after SPS stimulation, whereas the converse was found for p62 expression. In conclusion, dysregulation of autophagic activity in the mPFC may play a crucial role in PTSD pathogenesis.


Asunto(s)
Beclina-1/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Corteza Prefrontal/metabolismo , Proteína Sequestosoma-1/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Animales , Autofagia , Beclina-1/genética , Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Aprendizaje por Laberinto , Proteínas Asociadas a Microtúbulos/genética , Neuronas/metabolismo , Neuronas/ultraestructura , Corteza Prefrontal/citología , Ratas , Ratas Wistar , Proteína Sequestosoma-1/genética
17.
J Affect Disord ; 213: 1-8, 2017 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-28167453

RESUMEN

BACKGROUND: Post-traumatic stress disorder (PTSD) can be categorised as a disorder of dysregulated fear processing. In the formation and development of PTSD, whether fear/anxious-related memory is regulated by ß-arrestin-2, and happened along the signal transduction pathways remains unknown. METHOD: We used single prolonged stress (SPS) as the animal model of PTSD. Next, elevated plus maze tests (EPM) was performed to examine fear/anxious memory- related behaviors. Then, we detected ß-arrestin-2, PDE-4, and signal transduction pathways with immunofluorescence, co-immunoprecipitation, immunohistochemistry, Elisa, western blot, RT-PCR, and real-time PCR. RESULTS: Our data indicated that SPS caused fear/anxious memory-related behaviors enhancement. The low expression of ß-arrestin-2, PDE-4 and their complex on SPS 7d, and high expression of signal transduction pathways on SPS7d in basolateral amygdala (BLA). CONCLUSIONS: That indicating that ß-arrestin-2 is critical for the formation of abnormal fear/anxious memory in PTSD; and fear/anxious memory occured through signal transduction pathways. Finally, these results suggest that ß-arrestin-2, PDE-4 and signal transduction pathways may be by influencing the fear/anxious memory thereby involved in the formation and development of PTSD.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Miedo/fisiología , Memoria/fisiología , Trastornos por Estrés Postraumático/metabolismo , Arrestina beta 2/metabolismo , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Animales , Ansiedad/metabolismo , Conducta Animal/fisiología , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Trastornos por Estrés Postraumático/psicología
18.
J Mol Neurosci ; 61(4): 531-541, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28210980

RESUMEN

The goal of this study was to further elucidate the molecular mechanisms of post-traumatic stress disorder (PTSD) pathogenesis and to provide experimental evidence for new drug targets for effective PTSD treatment. Expression changes of IRE1α, ASK1, and other downstream molecules of the IRE1α-ASK1 endoplasmic reticulum stress (ERS) signaling pathway were investigated. JNK, P38, CHOP, Bcl-2, and Bax were analyzed at both protein and mRNA levels of dorsal raphe nucleus (DRN) neurons of PTSD rats. The rat PTSD model was established via the single-prolonged stress (SPS) method. Animals were randomly divided into five groups: a normal control group, a 1-day SPS group, a 4-days SPS group, a 7-day SPS group, and a 14-day SPS group. Spatial memory and learning ability of rats were evaluated subsequent to SPS using the Morris water maze test. Changes of IRE1α expression in the control and SPS groups were detected via immunohistochemistry (IHC). Protein and mRNA expressions of IRE1α, ASK1, JNK, P38, CHOP, Bcl-2, and Bax in the control and SPS groups were detected via Western blot and RT-PCR, respectively. The Morris water maze test revealed significantly longer average escape latencies in all SPS groups compared to the control group. In the spatial probe test, the percentage of time spent in the target quadrant was significantly lower in the SPS groups compared to control. IHC revealed increased positive expression of IRE1α subsequent to SPS challenge, reaching maximal levels on days four and seven (P < 0.01), while significantly decreasing on day 14 (P < 0.01). Western blot and RT-PCR revealed that protein and mRNA expressions of IRE1α, ASK1, JNK, CHOP, and P38 were significantly increased compared to control, peaking on days one, four, and seven post-SPS before returning to previous levels. Compared to control, expressions of Bcl-2 and Bax presented an initial increasing tendency followed by a decrease. A peak of Bcl-2 expression appeared early on day one following SPS, then decreased to a steady level. Bax expression in the SPS groups remained constant during early stages after SPS (days one to three) compared to control; however, expression significantly increased on day four and maintained a high level. In summary, 1) SPS challenge significantly activated the IRE1α-ASK1-JNK and IRE1α-ASK1-P38 apoptosis-signaling pathways in DRN neurons of PTSD rats. This resulted in a cascade of downstream reactions and ultimately apoptosis of DRN neurons. 2) Increased expression of apoptosis-associated molecules Bcl-2 and Bax in DRN neurons following SPS challenge was revealed as a central mechanism, inducing apoptosis of DRN neurons in PTSD rats.


Asunto(s)
Núcleo Dorsal del Rafe/metabolismo , Endorribonucleasas/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Respuesta de Proteína Desplegada , Animales , Apoptosis , Núcleo Dorsal del Rafe/citología , Endorribonucleasas/genética , Aprendizaje , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , Masculino , Complejos Multienzimáticos/genética , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Memoria Espacial , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
J Mol Neurosci ; 59(4): 471-82, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27112439

RESUMEN

Previous studies revealed that patients with post-traumatic stress disorder (PTSD) have a smaller than normal medial prefrontal cortex (mPFC), and PTSD rats [single prolonged stress, (SPS)] have an increased mPFC neuron apoptosis, which are related to the severity of PTSD symptoms. Three signalling pathways [protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor 6 (ATF6), and inositol-requiring enzyme 1 (IRE1)] in the endoplasmic reticulum (ER) play a critical role in resisting apoptosis. The aim of this study was to investigate whether the three branches of ER signalling are involved in SPS-induced mPFC neuron apoptosis. We used transmission electron microscopy (TEM) to detect morphological changes in ER and fluorescence spectrophotometry to detect the concentration of intracellular calcium in mPFC. We used molecular biological techniques to detect the expression levels of three branch signalling pathways of ER: phosphorylated PERK (p-PERK)/phosphorylated eukaryotic translation initiation factor 2A (p-eIF2a), ATF6a/X-box binding protein 1 (XBP1), and IRE1a. In addition, the ER molecular chaperone 78-kDa glucose-regulated protein (GRP78) and the ER-related apoptosis factors caspase family and Bax also were examined. Apoptosis neurons were detected by terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling. The results showed that the concentration of calcium in mPFC was increased in SPS rats. Using TEM, we found that mPFC neurons in SPS rats showed an expanded ER and chromatin margination. The increased expressions of p-PERK/p-eIF2a, ATF6a/XBP1, and IRE1 in response to SPS were also observed, although the degrees of increase were different. In addition, the protein and mRNA expression of GRP78 was increased in SPS rats; the upregulation of ER-related apoptosis factors and apoptosis neurons after SPS stimulation was observed. These results suggested that the three signalling pathways of unfolded protein response were involved in PTSD-induced, ER-dependent apoptosis in mPFC.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Corteza Prefrontal/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Trastornos por Estrés Postraumático/metabolismo , eIF-2 Quinasa/metabolismo , Factor de Transcripción Activador 6/genética , Animales , Apoptosis , Calcio/metabolismo , Retículo Endoplásmico/ultraestructura , Proteínas de Choque Térmico/metabolismo , Proteínas de la Membrana/genética , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Wistar , Transducción de Señal , eIF-2 Quinasa/genética
20.
Artículo en Inglés | MEDLINE | ID: mdl-27059130

RESUMEN

Our previous studies have shown evidence of endoplasmic reticulum (ER) stress-induced apoptosis in the hippocampus and mPFC in an animal model of post- traumatic stress disorder (PTSD). Inositol-requiring enzyme 1α (IRE1α) and its downstream molecule X-box binding protein 1 (XBP1) play key roles in the ER-related apoptosis pathway. Dysregulation of the locus coeruleus (LC) has been reported to contribute to cognitive and/or arousal impairments associated with PTSD. The aim of the present study was to explore the role of IRE1α pathway in neuronal apoptosis in the LC of rat models of PTSD. We used an acute exposure to prolonged stress (single prolonged stress, SPS) to model PTSD in rats and examined the effects related to the IRE1α pathway. Neuronal apoptosis in LC was detected by transmission electron microscopy and TUNEL staining. The results showed that the level of LC neuronal apoptosis was markedly increased after SPS. SPS exposure triggered IRE1α pathway, as evidenced by the increased activity of IRE1α, specific splicing of XBP1, and up-regulated expression of binding immunoglobulin protein/78kDa glucose-regulated protein (BiP/GRP78), and C/EBP-homologous protein (CHOP). Treatment with STF-083010, an IRE1α RNase-specific inhibitor, successfully attenuated the above changes. These results indicate that excessive activation of the ER stress-associated IRE1α pathway is involved in LC neuronal apoptosis induced by SPS exposure; this may be a crucial mechanism of the pathogenesis of PTSD.


Asunto(s)
Apoptosis/fisiología , Estrés del Retículo Endoplásmico/fisiología , Endorribonucleasas/metabolismo , Locus Coeruleus/metabolismo , Complejos Multienzimáticos/metabolismo , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas de Choque Térmico/metabolismo , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/patología , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Distribución Aleatoria , Ratas Wistar , Transducción de Señal , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/patología , Sulfonamidas/farmacología , Tiofenos/farmacología , Factor de Transcripción CHOP/metabolismo , Proteína 1 de Unión a la X-Box/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...