Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 66(4): 2646-2662, 2023 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-36774555

RESUMEN

Enhancer of zeste homolog 2 (EZH2) mediates the trimethylation of histone 3 lysine 27 (H3K27) to promote gene silencing. Inhibition of EZH2 is a viable strategy for cancer treatment; however, only a small subset of hematological malignancies are sensitive to small-molecule EZH2 inhibitors. EZH2 inhibitors cause H3K27 acetylation in most solid tumors, leading to drug resistance. Bromodomain-containing protein 4 (BRD4) inhibitors were reported to enhance the sensitivity of solid tumors to EZH2 inhibitors. Thus, we designed and evaluated a series of dual EZH2-BRD4 inhibitors. ZLD-2, the most promising compound, exhibited potent inhibitory activity against EZH2 and BRD4. Compared to the EZH2 inhibitor GSK126, ZLD-2 displayed potent antiproliferation activity against breast, lung, bladder, and pancreatic cancer cells. In vivo, ZLD-2 exhibited antitumor activity in a BxPC-3 mouse xenograft model, whereas GSK126 promoted tumor growth. Thus, ZLD-2 may be a lead compound for treating solid tumors.


Asunto(s)
Antineoplásicos , Proteínas de Ciclo Celular , Diseño de Fármacos , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos , Neoplasias , Factores de Transcripción , Animales , Humanos , Ratones , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Histonas/metabolismo , Neoplasias/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología
2.
Eur Arch Psychiatry Clin Neurosci ; 270(2): 195-205, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29882089

RESUMEN

Although depressive symptoms including anhedonia (i.e., loss of pleasure) frequently accompany pain, little is known about the risk factors contributing to individual differences in pain-induced anhedonia. In this study, we examined if signaling of brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-receptor-kinase B (TrkB) contribute to individual differences in the development of neuropathic pain-induced anhedonia. Rats were randomly subjected to spared nerved ligation (SNI) or sham surgery. The SNI rats were divided into two groups based on the results of a sucrose preference test. Rats with anhedonia-like phenotype displayed lower tissue levels of BDNF in the medial prefrontal cortex (mPFC) compared with rats without anhedonia-like phenotype and sham-operated rats. In contrast, tissue levels of BDNF in the nucleus accumbens (NAc) of rats with an anhedonia-like phenotype were higher compared with those of rats without anhedonia-like phenotype and sham-operated rats. Furthermore, tissue levels of BDNF in the hippocampus, L2-5 spinal cord, muscle, and liver from both rats with or without anhedonia-like phenotype were lower compared with those of sham-operated rats. A single injection of 7,8-dihydroxyflavone (10 mg/kg; TrkB agonist), but not ANA-12 (0.5 mg/kg; TrkB antagonist), ameliorated reduced sucrose preference and reduced BDNF-TrkB signaling in the mPFC in the rats with anhedonia-like phenotype. These findings suggest that reduced BDNF-TrkB signaling in the mPFC might contribute to neuropathic pain-induced anhedonia, and that TrkB agonists could be potential therapeutic drugs for pain-induced anhedonia.


Asunto(s)
Anhedonia/fisiología , Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuralgia/metabolismo , Neuralgia/fisiopatología , Corteza Prefrontal/metabolismo , Receptor trkB/metabolismo , Transducción de Señal/fisiología , Anhedonia/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Neuralgia/etiología , Núcleo Accumbens/metabolismo , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/fisiopatología , Fenotipo , Ratas , Ratas Sprague-Dawley , Receptor trkB/agonistas , Receptor trkB/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sacarosa
3.
Psychopharmacology (Berl) ; 236(12): 3513-3523, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31321459

RESUMEN

RATIONALE: Basic and clinical studies have reported rapid and long-lasting antidepressant effects of ketamine. Although previous studies have proposed several mechanisms underlying the antidepressant effects of ketamine, these mechanisms have not been completely elucidated. OBJECTIVES: The present study evaluated the effects of systemically administered ketamine treatment in a lipopolysaccharide (LPS)-induced mouse model of depression. METHODS: Non-targeted metabolomics, western blotting, and behavioral tests (locomotion, tail suspension, and forced swimming tests) were performed. RESULT: Ketamine significantly attenuated the abnormally increased immobility time in a lipopolysaccharide (LPS)-induced mouse model of depression. Aminomalonic acid, glutaraldehyde, glycine, histidine, N-methyl-L-glutamic acid, and ribose levels in skeletal muscle were altered following ketamine administration. Furthermore, ketamine significantly decreased the LPS-induced increase in glycine receptor A1 (GlyA1) levels. However, the glycine receptor antagonist strychnine did not elicit any pharmacological effects on ketamine-induced alterations in behaviors or muscular GlyA1 levels. Exogenous glycine and L-serine significantly improved depression-like symptoms in LPS-induced mice. CONCLUSIONS: Our findings suggest that skeletal muscular glycine contributes to the antidepressant effects of ketamine in inflammation. Effective strategies for improving skeletal muscular glycine levels may be a novel approach to depression treatment.


Asunto(s)
Depresión/tratamiento farmacológico , Depresión/metabolismo , Glicina/metabolismo , Ketamina/uso terapéutico , Lipopolisacáridos/toxicidad , Músculo Esquelético/metabolismo , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Depresión/inducido químicamente , Modelos Animales de Enfermedad , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ketamina/farmacología , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos
4.
Neuroscience ; 409: 142-151, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30940563

RESUMEN

Stress is a non-specific, systemic, physiological response of the body to strong internal and external environmental stimuli. Accumulating evidence has suggested that stress, particularly chronic restraint stress (CRS), can reduce pain threshold and increase pain sensitivity. However, pathogenic and therapeutic mechanisms underlying CRS remain unclear. Here, we aimed to investigate roles of the brain-derived neurotrophic factor (BDNF)-mammalian target of rapamycin (mTOR) signaling pathway in CRS-induced abnormal pain sensitivity. CRS was successfully mimicked 7 days after model development, and paw withdrawal mechanical threshold (PWMT) and tail-flick latency (TFL) were evaluated. CRS significantly altered BDNF and mTOR phosphorylation in the anterior cingulate cortex and spinal cord but not in the hippocampus. On day 7, a single dose of 7,8-dihydroxyflavone, an activator of BDNF-tropomyosin receptor kinase B, was administered via intraperitoneal or intrathecal injection. Notably, only the intrathecal injection improved PWMT and TFL. Additionally, an intraperitoneal injection of rapamycin, an mTOR inhibitor, failed to induce any behavioral changes, whereas a single intrathecal injection of rapamycin improved abnormal CRS-induced PWMT and TFL. In conclusion, CRS can induce abnormal pain sensitivity, probably by altering the BDNF-mTOR signaling pathway in the spinal cord.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hiperalgesia/metabolismo , Transducción de Señal/fisiología , Médula Espinal/metabolismo , Estrés Psicológico/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Masculino , Ratones , Dimensión del Dolor , Umbral del Dolor/fisiología , Fosforilación , Ratas , Ratas Sprague-Dawley , Restricción Física
5.
Aging (Albany NY) ; 11(6): 1778-1790, 2019 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-30904902

RESUMEN

It is well known that the incidence of postoperative cognitive dysfunction (POCD) is high in elderly patients. The pathogenesis and therapeutic mechanisms of POCD, however, have not yet been completely elucidated. The effects of gut microbiota, particularly in terms of regulating brain function, have gradually attracted increasing attention. In this study, we investigated the potential role of gut microbiota in POCD in aged male mice and attempted to determine whether alterations in gut microbiota would be helpful in the diagnosis of POCD. POCD and non-POCD mice were classified by hierarchical cluster analysis of behavioral results. Additionally, α- and ß-diversity of gut microbiota showed a differential profile between the groups. In total, 24 gut bacteria were significantly altered in POCD mice compared with those in non-POCD mice, in which 13 gut bacteria were significantly correlated with escape latency in the Morris water maze test (MWMT). Remarkably, receiver operating characteristic curves revealed that the Dehalobacteriaceae family and Dehalobacterium genus are potentially important bacteria for the diagnosis of POCD. These findings indicate that alterations in the composition of gut microbiota are probably involved in the pathogenesis of POCD in aged mice. Novel therapeutic strategies regulating specific gut bacteria may be helpful for the prevention and treatment of POCD.


Asunto(s)
Envejecimiento/metabolismo , Disfunción Cognitiva/inducido químicamente , Microbioma Gastrointestinal , Complicaciones Cognitivas Postoperatorias/inducido químicamente , Anestesia/efectos adversos , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos
6.
Transl Psychiatry ; 9(1): 57, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30705252

RESUMEN

Patients with chronic neuropathic pain frequently suffer from symptoms of anhedonia, which is a core symptom of depression. Accumulating studies suggest that gut microbiota may play a role in depression via gut-microbiota-brain axis. However, it is unknown whether gut microbiota plays a role in neuropathic pain-associated anhedonia. Here, we used a rat model of spared nerve injury (SNI). Hierarchical cluster analysis of sucrose preference test (SPT) results was used to classify the SNI rats with or without anhedonia-like phenotype. The 16S ribosomal RNA sequencing analysis showed abnormal composition of gut microbiota in the anhedonia susceptible compared to sham-operated rats and resilient rats. Furthermore, antibiotics-treated mice showed pain as well as depression-like and anhedonia-like phenotypes, suggesting a role of gut microbiota in these abnormal behaviors. Transplantation of fecal microbiota from anhedonia susceptible rats into antibiotics-treated pseudo-germ-free mice significantly exaggerated pain and depression-like phenotypes, including anhedonia. In contrast, transplantation of fecal microbiota from resilient rats into antibiotics-treated pseudo-germ-free mice significantly improved pain and depression-like phenotypes, including anhedonia. In conclusion, this study suggests that abnormal composition of gut microbiota may contribute to anhedonia susceptibility post SNI surgery, and that gut microbiota also plays a role in the pain as well as depression-like phenotypes. Interestingly, fecal microbiota transplantation from SNI rats with or without anhedonia can alter pain, depression-like and anhedonia-like phenotypes in the pseudo-germ-free mice. Therefore, it is likely that gut microbiota plays a key role in the pain as well as depression-like phenotypes including anhedonia in rodents with neuropathic pain.


Asunto(s)
Anhedonia/fisiología , Depresión/microbiología , Microbioma Gastrointestinal , Neuralgia/microbiología , Neuralgia/psicología , Animales , Conducta Animal , Depresión/complicaciones , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Neuralgia/complicaciones , Fenotipo , Ratas Sprague-Dawley , Nervio Ciático/lesiones
7.
CNS Neurosci Ther ; 25(6): 685-696, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30680947

RESUMEN

AIMS: Anesthesia and surgery can cause delirium-like symptoms postoperatively. Increasing evidence suggests that gut microbiota is a physiological regulator of the brain. Herein, we investigated whether gut microbiota plays a role in postoperative delirium (POD). METHODS: Mice were separated into non-POD and POD phenotypes after abdominal surgery by applying hierarchical clustering analysis to behavioral tests. Fecal samples were collected, and 16S ribosomal RNA gene sequencing was performed to detect differences in gut microbiota composition among sham, non-POD, and POD mice. Fecal bacteria from non-POD and POD mice were transplanted into antibiotics-induced pseudo-germ-free mice to investigate the effects on behaviors. RESULTS: α-diversity and ß-diversity indicated differences in gut microbiota composition between the non-POD and POD mice. At the phylum level, the non-POD mice had significantly higher levels of Tenericutes, which were not detected in the POD mice. At the class level, levels of Gammaproteobacteria were higher in the POD mice, whereas the non-POD mice had significantly higher levels of Mollicutes, which were not detected in the POD mice. A total of 20 gut bacteria differed significantly between the POD and non-POD mice. Interestingly, the pseudo-germ-free mice showed abnormal behaviors prior to transplant. The pseudo-germ-free mice that received fecal bacteria transplants from non-POD mice but not from POD mice showed improvements in behaviors. CONCLUSIONS: Abnormal gut microbiota composition after abdominal surgery may contribute to the development of POD. A therapeutic strategy that targets gut microbiota could provide a novel alterative for POD treatment.


Asunto(s)
Abdomen/cirugía , Delirio/microbiología , Microbioma Gastrointestinal , Complicaciones Posoperatorias/microbiología , Animales , Biodiversidad , Trasplante de Microbiota Fecal , Vida Libre de Gérmenes , Masculino , Ratones Endogámicos C57BL , Distribución Aleatoria
8.
Front Pharmacol ; 10: 1702, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32116688

RESUMEN

Patients are more likely to suffer from central nervous system (CNS) complications after anesthesia and surgery. However, postoperative depression (POD) has not yet received sufficient attentions, and its pathogenesis and therapeutic strategies remain poorly understood. We here aimed to investigate whether brain derived neurotrophic factor (BDNF)-tropomyosin-related kinase B (TrkB) signaling plays an important role in POD. BDNF-TrkB signaling was altered in brain and peripheral tissues, including medial prefrontal cortex (mPFC), hippocampus, liver, and muscle, among control, POD susceptible, and resilient groups. Additionally, we demonstrated that 7,8-dihydroxyflavone (7,8-DHF), a TrkB agonist, could exert its pharmacologic property to alleviate POD-like symptoms. More importantly, ketamine, a non-competitive N-methyl-D-aspartic acid (NMDA) receptor antagonist, also has significant antidepressant effects in POD model, associating with the improving effects on levels of BDNF-TrkB signaling in brain and peripheral tissues. Interestingly, the beneficial effects of ketamine on POD-like symptoms are fully attenuated by a TrkB antagonist. These findings suggest that abnormal expressions of BDNF-TrkB signaling in brain and peripheral tissues are implicated in the pathogenesis of POD, and that therapeutic agents targeting BDNF-TrkB, particularly ketamine, could favor the beneficial effects for POD.

9.
Pharmacol Biochem Behav ; 176: 93-100, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30528936

RESUMEN

Ketamine, an N-methyl-d-aspartic acid receptor (NMDAR) antagonist, elicits rapid-acting and sustained antidepressant effects in treatment-resistant depressed patients. Accumulating evidence suggests that gut microbiota via the gut-brain axis play a role in the pathogenesis of depression, thereby contributing to the antidepressant actions of certain compounds. Here we investigated the role of gut microbiota in the antidepressant effects of ketamine in lipopolysaccharide (LPS)-induced inflammation model of depression. Ketamine (10 mg/kg) significantly attenuated the increased immobility time in forced swimming test (FST), which was associated with the improvements in α-diversity, consisting of Shannon, Simpson and Chao 1 indices. In addition to α-diversity, ß-diversity, such as principal coordinates analysis (PCoA), and linear discriminant analysis (LDA) coupled with effect size measurements (LEfSe), showed a differential profile after ketamine treatment. Furthermore, a total of 30 bacteria were significantly altered in the LPS + saline treated mice and LPS + ketamine treated mice. Interestingly, two bacteria, including the phylum Actinobacteria and the class Coriobacteriia were significantly correlated with the immobility time of FST. Importantly, the receiver operating characteristic (ROC) curves demonstrated that the phylum Actinobacteria and the class Coriobacteriia were potential biomarker for the antidepressant effects of ketamine in an inflammation model. These findings suggest that antidepressant effects of ketamine might be related to the regulation of abnormal composition of gut microbiota, and that the phylum Actinobacteria and the class Coriobacteriia might be potential biomarkers for ketamine's antidepressant efficacy.


Asunto(s)
Actinobacteria/fisiología , Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Depresión/microbiología , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/microbiología , Ketamina/farmacología , Actinobacteria/efectos de los fármacos , Animales , Antidepresivos/administración & dosificación , Conducta Animal/efectos de los fármacos , Biomarcadores , Depresión/inducido químicamente , Trastorno Depresivo/inducido químicamente , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Ketamina/administración & dosificación , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Curva ROC , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
10.
Curr Med Sci ; 38(6): 962-967, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30536056

RESUMEN

A series of factors can be involved in the perioperative period to cause an increase in cancer-related mortality. Unfortunately, volatile anesthesia might aggravate the deleterious effects. In this article, we review the association of diverse volatile anesthetic agents with immune system and cancer cell biology, and examine the effects on angeogenesis and postoperative metastasis or recurrence. Isoflurane, haloflurane and enflurane enhance immunosuppression and upregulate hypoxia-inducible-factor 1 and matrix metalloproteinases, leading to the cancer malignant progression, whereas roles of desflurane and sevoflurane are still unclear. As the effects of volatile anesthetics on tumor immunity have been known, it will be beneficial for using selective drugs into anesthesia and operation in cancer patients.


Asunto(s)
Anestésicos por Inhalación/efectos adversos , Anestésicos por Inhalación/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Progresión de la Enfermedad , Humanos , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/patología , Recurrencia Local de Neoplasia/inducido químicamente , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología
11.
Psychopharmacology (Berl) ; 235(11): 3351-3358, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30259075

RESUMEN

Skeletal muscle consumes two thirds of the body's energy and may play a role in stress-related disorders. Evidence suggests that the peroxisome proliferator-activated receptor γ coactivator α (PGC-1α)-fibronectin type III domain-containing 5 (FNDC5)-brain-derived neurotrophic factor (BDNF) signaling pathway in skeletal muscle plays a key role in conferring the beneficial effects of exercise. In this study, we aimed to determine whether this pathway contributes to the resilience or susceptibility of mice subjected to chronic social defeat stress (CSDS). BDNF-tropomyosin receptor kinase B (TrkB) and proBDNF-p75NTR signaling in the medial prefrontal cortex and nucleus accumbens of susceptible but not resilient mice were significantly altered compared with the controls. Furthermore, the levels of PGC-1α, FNDC5, and BDNF, as well as the p-TrkB/TrkB ratio in the skeletal muscle of susceptible but not resilient mice, were significantly lower than those of the controls, but the levels of proBDNF and p75NTR in the skeletal muscle of susceptible mice were significantly higher than those of the controls. Moreover, there were significant positive associations between social interaction test data and the expression of PGC-1α, FNDC5, and BDNF or the p-TrkB/TrkB ratio in skeletal muscle. These results suggest that the downregulation of the PGC-1α-FNDC5-BDNF signaling pathway in skeletal muscle contributes to resilience vs. susceptibility to CSDS. Therefore, alterations in this pathway in skeletal muscle may play a crucial role in mediating stress-related disorders.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Fibronectinas/metabolismo , Relaciones Interpersonales , Músculo Esquelético/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Estrés Psicológico/metabolismo , Animales , Enfermedad Crónica , Regulación hacia Abajo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/metabolismo , Condicionamiento Físico Animal/fisiología , Resiliencia Psicológica , Transducción de Señal/fisiología , Estrés Psicológico/psicología
12.
Neuroscience ; 388: 357-366, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30086367

RESUMEN

Peripheral immune activation could cause neuroinflammation, leading to a series of central nervous system (CNS) disorders, such as spatial learning and memory dysfunction. However, its pathogenic mechanism and therapeutic strategies are not yet determined. The present study aimed to investigate the therapeutic effects of sulforaphane (SFN) on lipopolysaccharide (LPS)-induced spatial learning and memory dysfunction, and tried to elucidate its relationship with the role of hippocampal brain-derived neurotrophic factor (BDNF)-mammalian target of rapamycin (mTOR) signaling pathway. Intraperitoneal injection of LPS for consecutive 7 days to mice caused abnormal behaviors in Morris water maze test (MWMT), while systemic administration of SFN notably reversed the abnormal behaviors. In addition, hippocampal levels of inflammatory cytokines, synaptic proteins, BDNF-tropomyosin receptor kinase B (TrkB) and mTOR signaling pathways were altered in the processes of LPS-induced cognitive dysfunction and SFN's therapeutic effects. Furthermore, we found that ANA-12 (a TrkB inhibitor) or rapamycin (a mTOR inhibitor) could block the beneficial effects of SFN on LPS-induced cognitive dysfunction, and that hippocampal levels of synaptic proteins, BDNF-TrkB and mTOR signaling pathways were also notably changed. In conclusion, the results of the present study suggest that SFN could elicit improving effects on LPS-induced spatial learning and memory dysfunction, which is likely related to the regulation of hippocampal BDNF-mTOR signaling pathway.


Asunto(s)
Inflamación/tratamiento farmacológico , Isotiocianatos/farmacología , Discapacidades para el Aprendizaje/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Nootrópicos/farmacología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Inflamación/complicaciones , Inflamación/psicología , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/inmunología , Lipopolisacáridos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/etiología , Trastornos de la Memoria/inmunología , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Sulfóxidos , Serina-Treonina Quinasas TOR/metabolismo
13.
Front Pharmacol ; 9: 887, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30135655

RESUMEN

Patients with chronic neuropathic pain frequently suffer from symptoms of anhedonia (loss of pleasure), which is a core clinical manifestation of depression. Accumulating studies have shown the beneficial effects of the natural compound sulforaphane (SFN), an activator of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), on depression-like phenotype through a potent anti-inflammatory effect. However, it is unknown whether SFN confers beneficial effects in neuropathic pain-associated anhedonia. Spared nerve injury (SNI) is classical rodent model of chronic neuropathic pain. We here used a rat model of SNI. Hierarchical cluster analysis of sucrose preference test (SPT) results was used to classify the SNI rats with or without an anhedonia phenotype. Nrf2 protein expression was significantly decreased in the medial prefrontal cortex (mPFC), hippocampus, spinal cord, and skeletal muscle, but not in the nucleus accumbens, in anhedonia-susceptible rats compared with sham or anhedonia-resistant rats. The expression of Kelch-like erythroid cell-derived protein with CNC homology (ECH)-associated protein 1 (Keap1), a partner of Nrf2, in mPFC, hippocampus, and muscle of anhedonia-susceptible rats was also significantly lower than that in sham or anhedonia-resilient rats. Subsequent SFN administration after SNI surgery exerted therapeutic effects on reduced mechanical withdrawal threshold (MWT) scores, but not on sucrose preference, through the normalization of Keap1-Nrf2 signaling in the spinal cords of anhedonia-susceptible rats. Interestingly, treatment with SFN 30 min prior to SNI surgery significantly attenuated reduced MWT scores and sucrose preference, and restored tissue Keap1 and Nrf2 levels. In conclusion, this study suggests that decreased Keap1-Nrf2 signaling in mPFC, hippocampus, and muscle may contribute to anhedonia susceptibility post-SNI surgery, and that SFN exerts beneficial effects in SNI rats by normalization of decreased Keap1-Nrf2 signaling.

14.
Aging (Albany NY) ; 10(6): 1257-1267, 2018 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-29886457

RESUMEN

Alzheimer's disease is characterized by cognitive dysfunction and aging is an important predisposing factor; however, the pathological and therapeutic mechanisms are not fully understood. Recently, the role of gut microbiota in Alzheimer's disease has received increasing attention. The cognitive function in senescence-accelerated mouse prone 8 (SAMP8) mice was significantly decreased and the Chao 1 and Shannon indices, principal coordinates analysis, and principal component analysis results were notably abnormal compared with that of those in senescence-accelerated mouse resistant 1 (SAMR1) mice. Moreover, 27 gut bacteria at six phylogenetic levels differed between SAMP8 and SAMR1 mice. In a separate study, we transplanted fecal bacteria from SAMP8 or SAMR1 mice into pseudo germ-free mice. Interestingly, the pseudo germ-free mice had significantly lower cognitive function prior to transplant. Pseudo germ-free mice that received fecal bacteria transplants from SAMR1 mice but not from SAMP8 mice showed improvements in behavior and in α-diversity and ß-diversity indices. In total, 14 bacteria at six phylogenetic levels were significantly altered by the gut microbiota transplant. These results suggest that cognitive dysfunction in SAMP8 mice is associated with abnormal composition of the gut microbiota. Thus, improving abnormal gut microbiota may provide an alternative treatment for cognitive dysfunction and Alzheimer's disease.


Asunto(s)
Envejecimiento , Disfunción Cognitiva/etiología , Microbioma Gastrointestinal/fisiología , Animales , Antibacterianos , Conducta Animal , Masculino , Memoria , Ratones , Ratones Endogámicos , Aprendizaje Espacial
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA