Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Transl Med ; 22(1): 350, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38609979

RESUMO

BACKGROUND: Olfactory dysfunction occurs frequently in Parkinson's disease (PD). In this study, we aimed to explore the potential biomarkers and underlying molecular pathways of nicotine for the treatment of olfactory dysfunction in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced PD mice. METHODS: MPTP was introduced into C57BL/6 male mice to generate a PD model. Regarding in vivo experiments, we performed behavioral tests to estimate the protective effects of nicotine in MPTP-induced PD mice. RNA sequencing and traditional molecular methods were used to identify molecules, pathways, and biological processes in the olfactory bulb of PD mouse models. Then, in vitro experiments were conducted to evaluate whether nicotine can activate the prok2R/Akt/FoxO3a signaling pathway in both HEK293T cell lines and primary olfactory neurons treated with 1-methyl-4-phenylpyridinium (MPP+). Next, prok2R overexpression (prok2R+) and knockdown (prok2R-) were introduced with lentivirus, and the Akt/FoxO3a signaling pathway was further explored. Finally, the damaging effects of MPP+ were evaluated in prok2R overexpression (prok2R+) HEK293T cell lines. RESULTS: Nicotine intervention significantly alleviated olfactory and motor dysfunctions in mice with PD. The prok2R/Akt/FoxO3a signaling pathway was activated after nicotine treatment. Consequently, apoptosis of olfactory sensory neurons was significantly reduced. Furthermore, prok2R+ and prok2R- HEK293T cell lines exhibited upregulation and downregulation of the Akt/FoxO3a signaling pathway, respectively. Additionally, prok2R+ HEK293T cells were resistant to MPP+-induced apoptosis. CONCLUSIONS: This study showed the effectiveness and underlying mechanisms of nicotine in improving hyposmia in PD mice. These improvements were correlated with reduced apoptosis of olfactory sensory neurons via activated prok2R/Akt/FoxO3a axis. These results explained the potential protective functions of nicotine in PD patients.


Assuntos
Transtornos do Olfato , Doença de Parkinson , Humanos , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células HEK293 , Nicotina/farmacologia , Doença de Parkinson/complicações , Proteínas Proto-Oncogênicas c-akt , Transtornos do Olfato/complicações , Transtornos do Olfato/tratamento farmacológico
2.
Microbiol Spectr ; 11(6): e0023423, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37800955

RESUMO

IMPORTANCE: The gut and salivary microbiomes have been widely reported to be significantly associated with a number of neurological disorders. The stability of the microbiome in the oral cavity makes it a potentially ideal sample that can be conveniently obtained for the investigation of microbiome-based pathogenesis in diseases. In the present study, we used a single-molecule long-read sequencing technique to study the distribution of the salivary microbiota in patients with pituitary adenoma (PA) and healthy individuals, as well as among four clinical phenotypes of PA. We found that the diversity of salivary microbes was more abundant in PA patients than in healthy individuals. We also observed some unique genera in different PA phenotypes. The bioinformatics-based functional predictions identified potential links between microbes and different clinical phenotypes of PA. This study improves the existing understanding of the pathogenesis of PA and may provide diagnostic and therapeutic targets for PA.


Assuntos
Microbiota , Neoplasias Hipofisárias , Humanos , Saliva , Neoplasias Hipofisárias/diagnóstico , Neoplasias Hipofisárias/genética , RNA Ribossômico 16S/genética , Fenótipo
3.
Int Immunopharmacol ; 124(Pt A): 110911, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37696142

RESUMO

Neuroinflammation is believed to be a critical process involved in the pathophysiology of Alzheimer's disease (AD). In this study, we investigated the pharmacological ability of OAB-14, a small molecule compound derived from bexarotene, to reduce neuroinflammation and improve cognitive decline in an AD mouse model (in vivo) and its ability to regulate signaling pathways implicated in neuroinflammation in vitro. It was found that OAB-14 significantly improved the cognitive function of 11-month-old AD mice (APP/PS1 transgenic mice) in a dose-dependent manner. Simultaneously, OAB-14 dramatically inhibited the activation of microglia in the cerebral cortex and hippocampus of AD mice and dose-dependently downregulated the expression of nuclear factor kappa B (NF-κB) and NOD-like receptor protein 3 (NLRP3) in the cerebral cortex. At the cellular level, OAB-14 reversed the downregulation of M2 phenotypic markers, including mannose receptor C-type 1 (MRC1) and arginase 1 (ARG1), in lipopolysaccharide (LPS)- or amyloid-ß protein oligomer (oAß1-42)-activated BV2 microglial cells and partially restored their ability to clear Aß. However, these effects were suppressed when peroxisome proliferator-activated receptor-γ (PPAR-γ) was specifically inhibited by GW9662, a selective PPAR-γ antagonist. These results suggested that OAB-14 could regulate microglial polarization by regulating PPAR-γ signaling, thereby mitigating neuroinflammation and improving cognitive function in AD mice.

4.
Front Neurol ; 14: 1166598, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37409018

RESUMO

Introduction: Post-neurosurgical bacterial meningitis (PNBM) is a serious complication for patients who receive neurosurgical treatment, but the diagnosis is difficult given the complicated microenvironment orchestrated by sterile brain injury and pathogenic infection. In this study, we explored potential diagnostic biomarkers and immunological features using a proteomics platform. Methods: A total of 31 patients with aneurysmal subarachnoid hemorrhage (aSAH) who received neurosurgical treatment were recruited for this study. Among them, 15 were diagnosed with PNBM. The remaining 16 patients were categorized into the non-PNBM group. Proteomics analysis of the cerebrospinal fluid (CSF) was conducted on the Olink platform, which contained 92 immunity-related molecules. Results: We found that the expressions of 27 CSF proteins were significantly different between the PNBM and non-PNBM groups. Of those 27 proteins, 15 proteins were upregulated and 12 were downregulated in the CSF of the PNBM group. The receiver operating characteristic curve analysis indicated that three proteins (pleiotrophin, CD27, and angiopoietin 1) had high diagnostic accuracy for PNBM. Furthermore, we also performed bioinformatics analysis to explore potential pathways and the subcellular localization of the proteins. Conclusion: In summary, we found a cohort of immunity-related molecules that can serve as potential diagnostic biomarkers for PNBM in patients with aSAH. These molecules also provide an immunological profile of PNBM.

5.
Immun Inflamm Dis ; 11(6): e900, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37382264

RESUMO

BACKGROUND: Melatonin (MEL), an endogenous hormone, has been widely investigated in neurological diseases. Microglia (MG), a resident immunocyte localizing in central nervous system is reported to play important functions in the animal model of temporal lobe epilepsy (TLE). Some evidence showed that MEL influenced activation of MG, but the detailed model of action that MEL plays in remains uncertain. METHODS: In this study, we established a model of TLE in mice by stereotactic injection of kainic acid (KA). We treated the mice with MEL. Lipopolysaccharide, ROCK2-knockdown (ROCK-KD) and -overexpression (ROCK-OE) of lentivirus-treated cells were used in cell experiments to simulate an in vitro inflammatory model. RESULTS: The results of electrophysiological tests showed that MEL reduced frequency and severity of seizure. The results of behavioral tests indicated MEL improved cognition, learning, and memory ability. Histological evidences demonstrated a significant reduction of neuronal death in the hippocampus. In vivo study showed that MEL changed the polarization status of MG from a proinflammatory M1 phenotype to an anti-inflammatory M2 phenotype by inversely regulating the RhoA/ROCK signaling pathway. In cytological study, we found that MEL had a significant protective effect in LPS-treated BV-2 cells and ROCK-KD cells, while the protective effect of MEL was significantly attenuated in ROCK-OE cells. CONCLUSION: MEL played an antiepileptic role in the KA-induced TLE modeling mice both in behavioral and histological levels, and changed MG polarization status by regulating the RhoA/ROCK signaling pathway.


Assuntos
Epilepsia , Melatonina , Animais , Camundongos , Melatonina/farmacologia , Microglia , Transdução de Sinais , Lipopolissacarídeos/toxicidade
6.
J Neurooncol ; 163(1): 133-142, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37140882

RESUMO

PURPOSE: Bacteria have been observed in the tumor environment for decades and have been demonstrated to play important roles in the pathogenesis and development of several different tumors. So far there is a clear lack of specific studies relating to the presence of bacteria in pituitary neuroendocrine tumors (PitNETs). METHODS: In this study, we performed five region-based amplification and bacterial 16 S rRNA sequencing to identify the microbiome of PitNET tissues across four clinical phenotypes. Multiple filter procedures were performed to inhibit the risk of contamination with bacteria and bacterial DNA. Histological analysis was also conducted to validate the localization of bacteria in the intra-tumoral region. RESULTS: We identified common and diverse bacterial types across the four clinical phenotypes of PitNET. We also predicted the potential functions of these bacteria in tumor phenotypes and found that these functions were reported in certain previous mechanistic studies. Our data indicate that the pathogenesis and development of tumors may correlate with the behavior of intra-tumoral bacteria. Histological results, including lipopolysaccharide (LPS) staining and fluorescence in situ hybridization (FISH) for bacterial 16 S rRNA clearly demonstrated the localization of bacteria in the intra-tumoral region. Staining for Iba-1 suggested that the proportion of microglia was more abundant in FISH-positive regions than in FISH-negative regions. Furthermore, in FISH-positive regions, the microglia exhibited a longitudinally branched morphology that was different to the compact morphology observed in FISH-negative regions. CONCLUSION: In summary, we provide an evidence for the existence of intra-tumoral bacteria in PitNET.


Assuntos
Microbiota , Tumores Neuroendócrinos , Neoplasias Hipofisárias , Humanos , Hibridização in Situ Fluorescente , Neoplasias Hipofisárias/patologia , Hipófise/patologia
7.
Diagnostics (Basel) ; 13(2)2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36672998

RESUMO

Post-neurosurgical bacterial meningitis (PNBM) is one of the severe complications in patients receiving neurosurgical procedures. Recent studies have found microbe-related glycans play important roles in adhesion, invasion, and toxicity toward innate immunological reactions. In this study, we aimed to investigate the glycomic profile and its potential diagnostic efficacy in post-neurosurgical bacterial meningitis (PNBM) patients with hemorrhagic stroke. A total of 136 cerebrospinal fluid (CSF) samples were recruited and divided into a PNBM group and a non-PNBM group based on the clinical diagnostic criteria. A lectin biochip-based method was established for the detection of glycans in CSF. The clinicopathological data and biochemical parameters in CSF from all patients were analyzed. Two models for multivariate analysis investigating glycan changes in the CSF were conducted, aiming at determining the specific expression and diagnostic efficacy of lectin-probing glycans (LPGs) for PNBM. In univariate analysis, we found that 8 out of 11 LPGs were significantly correlated with PNBM. Model 1 multivariate analysis revealed that PNA (p = 0.034), Jacalin (p = 0.034) and LTL (p = 0.001) were differentially expressed in the CSF of PNBM patients compared with those of non-PNBM patients. Model 2 multivariate analysis further disclosed that LTL (p = 0.021) and CSF glucose (p < 0.001) had independent diagnostic efficacies in PNBM, with areas under the curve (AUC) of 0.703 and 0.922, respectively. In summary, this study provided a new insight into the subject of CSF glycomics concerning bacterial infection in patients with hemorrhagic stroke.

8.
Molecules ; 28(1)2023 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-36615583

RESUMO

Diabetes mellitus (DM) is a major risk factor for stroke and exacerbates white-matter damage in focal cerebral ischemia. Our previous study showed that the sigma-1 receptor agonist PRE084 ameliorates bilateral common-carotid-artery occlusion-induced brain damage in mice. However, whether this protective effect can extend to white matter remains unclear. In this study, C57BL/6 mice were treated with high-fat diets (HFDs) combined with streptozotocin (STZ) injection to mimic type 2 diabetes mellitus (T2DM). Focal cerebral ischemia in T2DM mice was established via injection of the vasoconstrictor peptide endothelin-1 (ET-1) into the hippocampus. Three different treatment plans were used in this study. In one plan, 1 mg/kg of PRE084 (intraperitoneally) was administered for 7 d before ET-1 injection; the mice were sacrificed 24 h after ET-1 injection. In another plan, PRE084 treatment was initiated 24 h after ET-1 injection and lasted for 7 d. In the third plan, PRE084 treatment was initiated 24 h after ET-1 injection and lasted for 21 d. The Y-maze, novel object recognition, and passive avoidance tests were used to assess neurobehavioral outcomes. We found no cognitive dysfunction or white-matter damage 24 h after ET-1 injection. However, 7 and 21 d after ET-1 injection, the mice showed significant cognitive impairment and white-matter damage. Only PRE084 treatment for 21 d could improve this white-matter injury; increase axon and myelin density; decrease demyelination; and increase the expressions of myelin regulator 2'-3'-cyclic nucleotide 3'-phosphodiesterase (CNpase) and myelin oligodendrocyte protein (MOG) (which was expressed by mature oligodendrocytes), the number of nerve/glial-antigen 2 (NG2)-positive cells, and the expression of platelet-derived growth factor receptor-alpha (PDGFRα), all of which were expressed by oligodendrocyte progenitor cells in mice with diabetes and focal cerebral ischemia. These results indicate that maybe there was more severe white-matter damage in the focal cerebral ischemia of the diabetic mice than in the mice with normal blood glucose levels. Long-term sigma-1 receptor activation may promote oligodendrogenesis and white-matter functional recovery in patients with stroke and with diabetes.


Assuntos
Isquemia Encefálica , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Acidente Vascular Cerebral , Substância Branca , Camundongos , Animais , Substância Branca/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Infarto Cerebral , Modelos Animais de Doenças
9.
J Physiol Sci ; 72(1): 15, 2022 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-35850611

RESUMO

It has been reported that wild-type p53-induced gene 1 (Wig1), which is downstream of p53, regulates the expression of mutant huntingtin protein (mHtt) in Huntington's disease (HD) patients and transgenic mouse brains. Intrastriatal injection of malonic acid in rats is often used as a model to study the pathological changes of Huntington's disease, and this model has the advantages of a fast preparation and low cost. Therefore, in this study, we used intrastriatal injections of 6 µM malonic acid in rats to evaluate the effect of tolfenamic acid on motor and cognitive deficits and the effect of 6 mg/kg and 32 mg/kg tolfenamic acid on p53 and its downstream targets, such as Wig1. The results showed that 32 mg/kg tolfenamic acid attenuated motor and spatial memory dysfunction, prevented Nox1-mediated reactive oxygen species (ROS) production, and downregulated the activity of p53 by increasing the phosphorylation level at the Ser378 site and decreasing the acetylation level at the Lys382 site. Tolfenamic acid reduced mouse double minute 2 (Mdm2), phosphatase and tensin homologue (Pten), P53-upregulated modulator of apoptosis (Puma) and Bcl2-associated X (Bax) at the mRNA level to inhibit apoptosis and downregulated sestrin 2 (Sesn2) and hypoxia inducible factor 1, alpha subunit (Hif-1α) mRNA levels to exert antioxidative stress effects. In addition, 32 mg/kg tolfenamic acid played a role in neuroprotection by decreasing the terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL)-positive cell numbers. However, there was no difference in the Wig mRNA level among all groups, and tolfenamic acid could not decrease the protein level of Wig1. In conclusion, tolfenamic acid inhibited the ROS-generating oxidase Nox1-regulated p53 activity and attenuated motor and spatial memory deficits in malonic acid-injected rats.


Assuntos
Doença de Huntington , Proteína Supressora de Tumor p53 , Animais , Apoptose , Doença de Huntington/genética , Doença de Huntington/patologia , Malonatos , Camundongos , Oxirredutases/metabolismo , Oxirredutases/farmacologia , RNA Mensageiro , Ratos , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/farmacologia
10.
Neurochem Int ; 154: 105298, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35134462

RESUMO

Disrupted mitochondrial fission/fusion balance is consistently involved in neurodegenerative diseases, including Alzheimer's disease. PTEN-induced putative kinase 1 (PINK1), a mitochondrial kinase, has been reported to prevent mitochondrial injury, oxidative stress, apoptosis, and inflammation. However, to the best of our knowledge, the contribution of PINK1 to Aß-induced mitochondrial fission/fusion has not been reported. In the present study, we showed that PINK1 deficiency promoted mitochondrial fission and fusion, aggravated mitochondrial dysfunction, and promoted neuroinflammatory cytokine factor production induced by intracerebroventricular (ICV) injection of Aß25-35 in rats. In vitro experiments have also showed that Aß25-35 caused more severe cell injury in PINK1-knockdown PC12 cells. These cells suffered more extensive death when exposed to proinflammatory cytokines. Lastly, we found that PINK1 overexpression significantly inhibited mitochondrial fusion, improved mitochondrial dysfunction, and reduced neuroinflammatory cytokine production induced by Aß25-35. The current study suggests the involvement of PINK1 in Aß25-35-mediated mitochondrial dynamics and that PINK1 may be a potential target for therapies aimed at enhancing neuroprotection to ameliorate Aß25-35-induced insults.


Assuntos
Doença de Alzheimer , Dinâmica Mitocondrial , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Doenças Neuroinflamatórias , Proteínas Quinases/metabolismo , Ratos
11.
Exp Neurol ; 347: 113867, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34582837

RESUMO

Blood-brain barrier (BBB) disruption is one of the most important pathological manifestations of ischemic stroke. Reducing BBB collapse is effective in alleviating brain parenchymal injury and cognitive dysfunction. Our previous study reported that Sigma-1 receptor (Sig-1R) activation in cerebral microvascular endothelial cells (CMECs) ameliorated BBB impairment, but the detailed mechanism remains unclear. In this study, we investigated Sig-1R activation as a BBB integrity promoter via many post ischemic stroke pathways. Sig-1R activation in BBB-associated astrocytes can increase glia-derived neurotrophic factor (GDNF) secretion in bilateral common carotid artery occlusion (BCCAO) mice. Upregulated GDNF activates its receptors in CMECs to promote BBB integrity, and activated Sig-1R in CMECs facilitates this process. In vitro experiments have found that Sig-1R activation in CMECs promotes the interaction between the GDNF α1 receptor and transduction rearrangement gene, increasing PI3K-AKT-junction protein signaling pathway expression. Sig-1R activation could be an effective therapeutic method for preventing BBB damage in ischemic stroke and other neurological conditions.


Assuntos
Barreira Hematoencefálica/patologia , Receptores sigma/metabolismo , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/patologia , Animais , Barreira Hematoencefálica/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
12.
Acta Pharmacol Sin ; 43(8): 1916-1927, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34893682

RESUMO

PTEN-induced putative kinase 1 (PINK1)/parkin pathway mediates mitophagy, which is a specialized form of autophagy. Evidence shows that PINK1 can exert protective effects against stress-induced neuronal cell death. In the present study we investigated the effects of PINK1 overexpression on tau hyperphosphorylation, mitochondrial dysfunction and oxidative stress in a specific rat model of tau hyperphosphorylation. We showed that intracerebroventricular (ICV) microinjection of forskolin (FSK, 80 µmol) induced tau hyperphosphorylation in the rat brain and resulted in significant spatial working memory impairments in Y-maze test, accompanied by synaptic dysfunction (reduced expression of synaptic proteins synaptophysin and postsynaptic density protein 95), and neuronal loss in the hippocampus. Adeno-associated virus (AAV)-mediated overexpression of PINK1 prevented ICV-FSK-induced cognition defect and pathological alterations in the hippocampus, whereas PINK1-knockout significantly exacerbated ICV-FSK-induced deteriorated effects. Furthermore, we revealed that AAV-PINK1-mediated overexpression of PINK1 alleviated ICV-FSK-induced tau hyperphosphorylation by restoring the activity of PI3K/Akt/GSK3ß signaling. PINK1 overexpression reversed the abnormal changes in mitochondrial dynamics, defective mitophagy, and decreased ATP levels in the hippocampus. Moreover, PINK1 overexpression activated Nrf2 signaling, thereby increasing the expression of antioxidant proteins and reducing oxidative damage. These results suggest that PINK1 deficiency exacerbates FSK-induced tau pathology, synaptic damage, mitochondrial dysfunction, and antioxidant system defects, which were reversed by PINK1 overexpression. Our data support a critical role of PINK1-mediated mitophagy in controlling mitochondrial quality, tau hyperphosphorylation, and oxidative stress in a rat model of Alzheimer's disease.


Assuntos
Doença de Alzheimer , Estresse Oxidativo , Proteínas Quinases , Proteínas tau , Doença de Alzheimer/metabolismo , Animais , Antioxidantes/metabolismo , Colforsina , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases/metabolismo , Ratos , Ubiquitina-Proteína Ligases/metabolismo , Proteínas tau/metabolismo
13.
ACS Chem Neurosci ; 12(21): 3985-3993, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34652916

RESUMO

In Alzheimer's disease (AD), damaged Aß clearance contributes to elevated levels of Aß that cause a series of cytotoxic cascade reactions. Thus, targeting Aß clearance has now been considered a valid therapeutic approach for AD. Cellular uptake and degradation are important mechanisms for Aß clearance, which are mainly performed by the endosomal-autophagic-lysosomal (EAL) pathway. Our previous study showed that OAB-14, a novel small molecule designed with bexarotene as the lead compound, treatment for 3 months significantly alleviated cognitive disorders and remarkably reduced the deposition of Aß without affecting its production in APP/PS1 transgenic mice. Here, we further revealed that enhancement of the EAL activity is one of the mechanisms that increases Aß clearance after OAB-14 administration for 3 months. OAB-14 facilitates receptor-mediated endocytosis and restores autophagy flux via the AMPK/mTOR pathway. Meanwhile, OAB-14 enhances the lysosomal activity, and reduced Aß accumulation in lysosomes was observed in OAB-14-treated AD mice. These results suggest that OAB-14 may promote Aß clearance in lysosomes by alleviating the EAL dysfunction in AD mice.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doença de Alzheimer/tratamento farmacológico , Precursor de Proteína beta-Amiloide/genética , Animais , Autofagia , Modelos Animais de Doenças , Lisossomos , Camundongos , Camundongos Transgênicos
14.
J Physiol Sci ; 70(1): 29, 2020 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-32517647

RESUMO

Tolfenamic acid, a nonsteroidal anti-inflammatory drug, alleviated learning and memory deficits and decreased the expression of specificity protein 1 (SP1)-mediated cyclin-dependent kinase-5 (CDK5), a major protein kinase that regulates hyperphosphorylated tau, in Alzheimer's disease (AD) transgenic mice. However, whether tolfenamic acid can regulate the major tau protein kinase, glycogen synthase kinase-3ß (GSK-3ß), or tau protein phosphatase, protein phosphatase 2A (PP2A), further inhibiting hyperphosphorylation of tau, remains unknown. To this end, tolfenamic acid was administered i.p. in a GSK-3ß overactivation postnatal rat model and orally in mice after intracerebroventricular (ICV) injection of okadaic acid (OA) to develop a PP2A inhibition model. We used four behavioural experiments to evaluate memory function in ICV-OA mice. In this study, tolfenamic acid attenuated memory dysfunction. Tolfenamic acid decreased the expression of hyperphosphorylated tau in the brain by inhibiting GSK-3ß activity, decreasing phosphorylated PP2A (Tyr307), and enhancing PP2A activity. Tolfenamic acid also increased wortmannin (WT) and GF-109203X (GFX) induced phosphorylation of GSK-3ß (Ser9) and prevented OA-induced downregulation of PP2A activity in PC12 cells. Altogether, these results show that tolfenamic acid not only decreased SP1/CDK5-mediated tau phosphorylation, but also inhibited GSK-3ß and PP2A-mediated tau hyperphosphorylation in AD models.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Proteína Fosfatase 2/antagonistas & inibidores , ortoaminobenzoatos/farmacologia , Proteínas tau/antagonistas & inibidores , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Modelos Animais de Doenças , Feminino , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Ratos , Ratos Wistar , Proteínas tau/genética , Proteínas tau/metabolismo
15.
Oxid Med Cell Longev ; 2020: 7950457, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32566101

RESUMO

Diabetic nephropathy (DN) is a chronic diabetic microvascular complication. Hyperactivity of the polyol pathway is involved in the pathogenesis of DN. Aldose reductase (AR), the rate-limiting enzyme of the polyol pathway, is expected to be an effective target in the treatment of DN. WJ-39 is a novel inhibitor of AR. The present study aimed at exploring the effects of WJ-39 in DN. DN was induced in rats by injecting 30 mg/kg streptozotocin (STZ). After 14 weeks, WJ-39 (10, 20, and 40 mg/kg) was intragastrically administered to the rats for 12 weeks. Treatment with WJ-39 significantly inhibited AR activation and ameliorated renal dysfunction and fibrosis in DN rats. WJ-39 reduced oxidative stress in the kidneys of DN rats by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. WJ-39 suppressed the activation of the nuclear factor-kappa B (NF-κB) pathway and the nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome to reduce the secretion of inflammatory factors. Rat mesangial cells (RMCs) were cultured under hyperglycemic conditions. WJ-39 abrogated the high glucose- (HG-) induced, excessive production of reactive oxygen species (ROS) and inflammatory factors. However, transfection with Nrf2 small interfering RNA abolished the effects of WJ-39. WJ-39 also blocked the transforming growth factor-ß1/Smad pathway to reduce the production of glomerular extracellular matrix proteins, ultimately reducing fibrogenesis in DN. Our results show that WJ-39 ameliorated renal injury in DN rats, and its effects on oxidative stress and inflammation were associated with the activation of Nrf2 signaling. Thus, WJ-39 and its mechanism of amelioration of renal lesions in DN rats by reducing renal inflammation, oxidative stress, and fibrosis injury could be an effective strategy for the treatment of DN.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Inibidores Enzimáticos/uso terapêutico , Rim/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais , Aldeído Redutase/metabolismo , Animais , Células Cultivadas , Nefropatias Diabéticas/fisiopatologia , Inibidores Enzimáticos/farmacologia , Fibrose , Glucose/toxicidade , Inflamação/patologia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Masculino , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/metabolismo , Células Mesangiais/patologia , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Ratos Sprague-Dawley , Proteínas Smad/metabolismo , Estreptozocina , Fator de Crescimento Transformador beta1/metabolismo
16.
Biomed Pharmacother ; 121: 109618, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31731189

RESUMO

Amyloid-ß (Aß) activating the pyroptotic cell pathway has been reported to act as a component in the progression of Alzheimer's disease (AD). As another major pathophysiological protein process in AD, the abnormal hyperphosphorylation of tau proteins exerts neurotoxic effects through a variety of mechanisms. However, data describing the relationship between hyperphosphorylated tau proteins and pyroptosis are very scarce. In this study, we used two hyperphosphorylated tau models, intracerebroventricular (ICV) forskolin (FSK, a PKA activator) rat model and ICV-streptozotocin (STZ) rat model; also, FSK and STZ treated PC12 cells as in vitro models to test the relationship between hyperphosphorylated tau proteins and pyroptosis. We found that FSK and STZ significantly increased the hyperphosphorylated tau level, pyroptosis-related protein in PC12 cell and rats' brain, and inhibited the activity of caspase-1 by caspase-1 inhibitor, caspase-1 siRNA, or incubated with Interleukin(IL)-1ß/IL-18 neutralizing antibody could notably alleviate the FSK and STZ induced PC12 cells damage and improve the cognitive disorder in ICV-FSK and ICV-STZ rats. Suppressed the level of hyperphosphorylated tau by LiCl also significantly decreased caspase-1 activity and the content of inflammatory cytokines in FSK or STZ treated PC12 cells. In summary, our results demonstrated that inflammasomes mediated pyroptosis at least one underlying pathogenic mechanism for the neurotoxicity induced by hyperphosphorylated tau in PC12 cells and dementia rats. IL-1ß and IL-18, the downstream of caspase-1, in turn increased hyperphosphorylated tau while spreading neuroinflammation.


Assuntos
Doença de Alzheimer/induzido quimicamente , Colforsina/farmacologia , Piroptose/fisiologia , Estreptozocina/farmacologia , Proteínas tau/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Caspase 1/fisiologia , Inflamassomos/fisiologia , Interleucina-18/fisiologia , Interleucina-1beta , Masculino , Células PC12 , Fosforilação , Ratos , Ratos Sprague-Dawley
17.
J Cell Mol Med ; 23(9): 6343-6354, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31318159

RESUMO

Protein kinase C (PKC) shows a neuronal protection effect in neurodegenerative diseases. In this study, we test whether berberine has a positive effect on the activity of PKC in quinolinic acid (QA)-induced neuronal cell death. We used intrastriatal injections of QA mice model to test the effect of berberine on motor and cognitive deficits, and the PKC signalling pathway. Treatment with 50 mg/kg b.w of berberine for 2 weeks significantly prevented QA-induced motor and cognitive impairment and related pathologic changes in the brain. QA inhibited the phosphorylation of PKC and its downstream molecules, GSK-3ß, ERK and CREB, enhanced the glutamate level and release of neuroinflammatory cytokines; these effects were attenuated by berberine. We used in vivo infusion of Go6983, a PKC inhibitor to disturb PKC activity in mice brain, and found that the effect of berberine to reverse motor and cognitive deficits was significantly reduced. Moreover, inhibition of PKC also blocked the anti-excitotoxicity effect of berberine, which is induced by glutamate in PC12 cells and BV2 cells, as well as anti-neuroinflammatory effect in LPS-stimulated BV2 cells. Above all, berberine showed neuroprotective effect against QA-induced acute neurotoxicity by activating PKC and its downstream molecules.


Assuntos
Berberina/farmacologia , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/tratamento farmacológico , Proteína Quinase C/metabolismo , Ácido Quinolínico/farmacologia , Animais , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Ácido Glutâmico/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Masculino , Camundongos , Doenças Neurodegenerativas/induzido quimicamente , Neurônios/efeitos dos fármacos , Síndromes Neurotóxicas/metabolismo , Transdução de Sinais/efeitos dos fármacos
18.
Oxid Med Cell Longev ; 2019: 4032428, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31049134

RESUMO

Tolfenamic acid is a nonsteroidal anti-inflammatory drug with neuroprotective properties, and it alleviates learning and memory deficits in the APP transgenic mouse model of Alzheimer's disease. However, whether tolfenamic acid can prevent motor and memory dysfunction in transgenic animal models of Huntington's disease (HD) remains unclear. To this end, tolfenamic acid was orally administered to transgenic R6/1 mice from 10 to 20 weeks of age, followed by several behavioral tests to evaluate motor and memory function. Tolfenamic acid improved motor coordination in R6/1 mice as tested by rotarod, grip strength, and locomotor behavior tests and attenuated memory dysfunction as analyzed using the novel object recognition test and passive avoidance test. Tolfenamic acid decreased the expression of mutant huntingtin in the striatum of 20-week-old R6/1 mice by inhibiting specificity protein 1 expression and enhancing autophagic function. Furthermore, tolfenamic acid exhibited antioxidant effects in both R6/1 mice and PC12 cell models. Collectively, these results suggest that tolfenamic acid has a good therapeutic effect on R6/1 mice, and may be a potentially useful agent in the treatment of HD.


Assuntos
Antioxidantes/farmacologia , Comportamento Animal/efeitos dos fármacos , Doença de Huntington , Transtornos da Memória , Desempenho Psicomotor/efeitos dos fármacos , ortoaminobenzoatos/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Doença de Huntington/prevenção & controle , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Transtornos da Memória/prevenção & controle , Camundongos , Camundongos Transgênicos , Mutação , Células PC12 , Ratos
19.
J Physiol Sci ; 69(3): 477-488, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30767122

RESUMO

Xanthoceraside, a novel triterpenoid saponin, has been found to attenuate learning and memory impairments in AD animal models. However, whether xanthoceraside has a positive effect on synaptic morphology remains unclear. Herein, we evaluated the effects of xanthoceraside on learning and memory impairments and the abnormalities of synaptic structure in APP/PS1 transgenic mice. The behavioral experiments demonstrated that xanthoceraside attenuated the imaginal memory and spatial learning impairments, and improved social interaction. Transmission electron microscopy and Golgi staining showed that xanthoceraside ameliorated synapse morphology abnormalities and dendritic spine density deficits, respectively. Western-blot analysis identified that xanthoceraside increased the expression of SYP and PSD95, activated BDNF/TrkB/MAPK/ERK and PI3K/Akt signaling pathways, meanwhile decreased the expression of RhoA, ROCK and Snk, increased the levels of SPAR, and activated the BDNF/TrkB/cofilin signaling pathway. Taken together, our study indicated that xanthoceraside improved cognitive function and protected both synaptic morphology and dendritic spine in APP/PS1 transgenic mice, which might be related in part to its activation in the BDNF/TrkB pathway.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Cognição/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/prevenção & controle , Memória/efeitos dos fármacos , Saponinas/farmacologia , Sinapses/efeitos dos fármacos , Triterpenos/farmacologia , Animais , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Modelos Animais de Doenças , Masculino , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais/efeitos dos fármacos , Sinapses/metabolismo
20.
Apoptosis ; 24(1-2): 157-167, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30387007

RESUMO

Reports have showed that Sigma-1 receptor (Sig-1R) activation can protect neurons against cerebral ischemia/reperfusion (I/R) injury in mice and alleviate endoplasmic reticulum (ER) stress in cultured cells, but little known is about the protective role of Sig-1R on ER stress induced by cerebral I/R. The purpose of this study was to determine whether Sig-1R exerts a protective effect against ER stress-mediated apoptosis in cerebral I/R using a 15-min bilateral common carotid artery occlusion (BCCAO) mouse model. At 72 h after reperfusion in BCCAO mice, we found that Sig-1R knockout (Sig-1R KO) significantly increased terminal dUTP nick-end labeling (TUNEL)-positive cells and nuclear structural damage in cortical neurons. Treatment with the Sig-1R agonist PRE084 once daily for three consecutive days reduced the number of TUNEL-positive cells and improved the ultrastructural damage of neurons in the cerebral cortex. These protective effects could be blocked by the Sig-1R antagonist BD1047. Then, we used BCCAO mice at 24 h after reperfusion to detect the expression of ER stress-mediated apoptotic pathway proteins. We found that expression of the pro-apoptotic proteins p-PERK, p-eIF2α, ATF, CHOP, p-IRE, p-JNK, Bim, PUMA, cleaved-caspase-12 and cleaved-caspase-3 was significantly increased and that expression of the anti-apoptotic protein Bcl-2 was significantly decreased in Sig-1R KO-BCCAO mice compared with BCCAO mice. Meanwhile, we found that treatment with PRE084 twice a day decreased pro-apoptotic protein expression and increased anti-apoptotic protein expression. The effects of PRE084 were blocked by the Sig-1R antagonist BD1047. These results suggest that Sig-1R activation inhibits ER stress-mediated apoptosis in BCCAO mice, indicating that Sig-1R may be a therapeutic target for neuroprotection particularly relevant to ER stress-induced apoptosis after cerebral I/R injury.


Assuntos
Apoptose/genética , Isquemia Encefálica , Estresse do Retículo Endoplasmático/fisiologia , Neuroproteção/genética , Receptores sigma/fisiologia , Traumatismo por Reperfusão , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/prevenção & controle , Citoproteção/genética , Estresse do Retículo Endoplasmático/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia , Substâncias Protetoras/metabolismo , Receptores sigma/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...