Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.553
1.
Int J Mol Sci ; 25(6)2024 Mar 15.
Article En | MEDLINE | ID: mdl-38542310

Nonalcoholic fatty liver disease (NAFLD) exponentially affects the global healthcare burden, and it is currently gaining increasing interest in relation to its potential impact on central nervous system (CNS) diseases, especially concerning cognitive deterioration and dementias. Overall, scientific research nowadays extends to different levels, exploring NAFLD's putative proinflammatory mechanism of such dysmetabolic conditions, spreading out from the liver to a multisystemic involvement. The aim of this review is to analyze the most recent scientific literature on cognitive involvement in NAFLD, as well as understand its underlying potential background processes, i.e., neuroinflammation, the role of microbiota in the brain-liver-gut axis, hyperammonemia neurotoxicity, insulin resistance, free fatty acids, and vitamins.


Cognition Disorders , Cognitive Dysfunction , Insulin Resistance , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Liver/metabolism , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Cognition Disorders/metabolism
2.
Life Sci ; 339: 122413, 2024 Feb 15.
Article En | MEDLINE | ID: mdl-38219919

AIMS: The gut microbiota is increasingly recognised as a pivotal regulator of immune system homeostasis and brain health. Recent research has implicated the gut microbiota in age-related cognitive impairment and dementia. Agathobaculum butyriciproducens SR79 T (SR79), which was identified in the human gut, has been reported to be beneficial in addressing cognitive deficits and pathophysiologies in a mouse model of Alzheimer's disease. However, it remains unknown whether SR79 affects age-dependent cognitive impairment. MAIN METHOD: To explore the effects of SR79 on cognitive function during ageing, we administered SR79 to aged mice. Ageing-associated behavioural alterations were examined using the open field test (OFT), tail suspension test (TST), novel object recognition test (NORT), Y-maze alternation test (Y-maze), and Morris water maze test (MWM). We investigated the mechanisms of action in the gut and brain using molecular and histological analyses. KEY FINDINGS: Administration of SR79 improved age-related cognitive impairment without altering general locomotor activity or depressive behaviour in aged mice. Furthermore, SR79 increased mature dendritic spines in the pyramidal cells of layer III and phosphorylation of CaMKIIα in the cortex of aged mice. Age-related activation of astrocytes in the cortex of layers III-V of the aged brain was reduced following SR79 administration. Additionally, SR79 markedly increased IL-10 production and Foxp3 and Muc2 mRNA expression in the colons of aged mice. SIGNIFICANCE: These findings suggest that treatment with SR79 may be a beneficial microbial-based approach for enhancing cognitive function during ageing.


Clostridiales , Cognition Disorders , Cognitive Dysfunction , Mice , Humans , Animals , Aged , Cognition Disorders/metabolism , Brain/metabolism , Aging/metabolism
3.
J Neuroinflammation ; 20(1): 237, 2023 Oct 19.
Article En | MEDLINE | ID: mdl-37858199

INTRODUCTION: Glucose transporter 1 (GLUT1) is essential for glucose transport into the brain and is predominantly expressed in the cerebral microvasculature. Downregulation of GLUT1 precedes the development of cognitive impairment in neurodegenerative conditions. Surgical trauma induces blood-brain barrier (BBB) disruption, neuroinflammation, neuronal mitochondria dysfunction, and acute cognitive impairment. We hypothesized that surgery reduces the expression of GLUT1 in the BBB that in turn disrupts its integrity and contributes to metabolic dysregulation in the brain that culminates in postoperative cognitive impairment. METHODOLOGY: Using an abdominal surgery model in aged WT mice, we assessed the perioperative changes in cognitive performance, tight junction proteins expression, GLUT1 expression, and the associated metabolic effects in the hippocampus. Thereafter, we evaluated the effects of these parameters in aged mice with conditional overexpression of GLUT1, and then again in aged mice with conditional overexpression of GLUT1 with or without prior exposure to the GLUT1 inhibitor ST-31. RESULTS: We showed a significant decline in cognitive performance, along with GLUT1 reduction and diminished glucose metabolism, especially in the ATP level in the postoperative mice compared with controls. Overexpression of GLUT1 expression alleviated postoperative cognitive decline and improved metabolic profiles, especially in adenosine, but did not directly restore ATP generation to control levels. GLUT1 inhibition ameliorated the postoperative beneficial effects of GLUT1 overexpression. CONCLUSIONS: Surgery-induced GLUT1 reduction significantly contributes to postoperative cognitive deficits in aged mice by affecting glucose metabolism in the brain. It indicates the potential of targeting GLUT1 to ameliorate perioperative neurocognitive disorders.


Blood-Brain Barrier , Cognition Disorders , Animals , Mice , Adenosine Triphosphate/metabolism , Blood-Brain Barrier/metabolism , Cognition Disorders/etiology , Cognition Disorders/metabolism , Down-Regulation , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Microvessels/metabolism
4.
Int J Dev Neurosci ; 83(5): 442-455, 2023 Aug.
Article En | MEDLINE | ID: mdl-37269159

Physical or psychological stress experienced by a mother during gestation is often associated with serious behavioural and cognitive deficits in newborns. Investigations of protective agents, which could prevent the adverse outcomes of prenatal stress (PS), are warranted. Agmatine is a neurotransmitter putatively involved in the physiological response to stress, and exogenous administration of agmatine has been shown to produce a variety of neuroprotective effects. In this study, we aimed to assess whether prenatal agmatine exposure could ameliorate behavioural and cognitive deficits in female offspring born to prenatally stressed mice. Pregnant Swiss Webster (SW) mice were exposed to physical or psychological stress from the 11th to 17th days of gestation. Agmatine (37.5 mg/kg, i.p.) was administrated 30 min before the induction of stress for seven consecutive days. The pups were assessed using a variety of behavioural tests and molecular assays on postnatal days 40 to 47. Agmatine attenuated impairments in locomotor activity, anxiety-like behaviour, and drug-seeking behaviour associated with both physical and psychological PS. Furthermore, agmatine reduced PS-induced impairments in passive avoidance memory and learning. Neither PS nor agmatine treatment affected the mRNA expression level of hippocampal brain-derived neurotrophic factor (BDNF) or tyrosine hydroxylase (TH) in the ventral tegmental area (VTA). Taken together, our findings highlight the protective effects of prenatally administered agmatine on PS-mediated behavioural and cognitive deficits of the offspring. Future studies are needed to elucidate the underlying mechanisms, which could allow for more targeted prenatal treatments.


Agmatine , Cognition Disorders , Cognitive Dysfunction , Prenatal Exposure Delayed Effects , Pregnancy , Mice , Animals , Female , Humans , Agmatine/pharmacology , Agmatine/therapeutic use , Agmatine/metabolism , Cognitive Dysfunction/metabolism , Cognition Disorders/metabolism , Cognition/physiology , Stress, Psychological/psychology , Hippocampus/metabolism , Prenatal Exposure Delayed Effects/metabolism
5.
ACS Chem Neurosci ; 14(11): 2074-2088, 2023 06 07.
Article En | MEDLINE | ID: mdl-37236204

c-Jun N-terminal kinases (JNKs) are a family of protein kinases activated by a myriad of stimuli consequently modulating a vast range of biological processes. In human postmortem brain samples affected with Alzheimer's disease (AD), JNK overactivation has been described; however, its role in AD onset and progression is still under debate. One of the earliest affected areas in the pathology is the entorhinal cortex (EC). Noteworthy, the deterioration of the projection from EC to hippocampus (Hp) point toward the idea that the connection between EC and Hp is lost in AD. Thus, the main objective of the present work is to address if JNK3 overexpression in the EC could impact on the hippocampus, inducing cognitive deficits. Data obtained in the present work suggest that JNK3 overexpression in the EC influences the Hp leading to cognitive impairment. Moreover, proinflammatory cytokine expression and Tau immunoreactivity were increased both in the EC and in the Hp. Therefore, activation of inflammatory signaling and induction of Tau aberrant misfolding caused by JNK3 could be responsible for the observed cognitive impairment. Altogether, JNK3 overexpression in the EC may impact on the Hp inducing cognitive dysfunction and underlie the alterations observed in AD.


Alzheimer Disease , Cognition Disorders , Cognitive Dysfunction , Humans , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Hippocampus/metabolism , Alzheimer Disease/metabolism , Cognition Disorders/metabolism , Cognitive Dysfunction/metabolism , Cognition , tau Proteins/metabolism
6.
Int J Mol Sci ; 24(9)2023 Apr 25.
Article En | MEDLINE | ID: mdl-37175535

Parkinson's disease with cognitive impairment (PD-CI) results in several clinical outcomes for which specific treatment is lacking. Although the pathogenesis of PD-CI has not yet been fully elucidated, it is related to neuronal plasticity decline in the hippocampus region. The dopaminergic projections from the substantia nigra to the hippocampus are critical in regulating hippocampal plasticity. Recently, aerobic exercise has been recognized as an effective therapeutic strategy for enhancing plasticity through the secretion of various muscle factors. The exact role of FNDC5-an upregulated, newly identified myokine produced after exercise-in mediating hippocampal plasticity and regional dopaminergic projections in PD-CI remains unclear. In this study, the effect of treadmill exercise on hippocampal synaptic plasticity was evaluated in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced chronic PD models. The results showed that treadmill exercise substantially alleviated the motor dysfunction, cognition disorder, and dopaminergic neuron degeneration induced by MPTP. Here, we discovered that the quadriceps, serum, and brain FNDC5 levels were lower in PD mice and that intervention with treadmill exercise restored FNDC5 levels. Moreover, treadmill exercise enhanced the synaptic plasticity of hippocampal pyramidal neurons via increased dopamine levels and BDNF in the PD mice. The direct protective effect of FNDC5 is achieved by promoting the secretion of BDNF in the hippocampal neurons via binding the integrin αVß5 receptor, thereby improving synaptic plasticity. Regarding the indirect protection effect, FNDC5 promotes the dopaminergic connection from the substantia nigra to the hippocampus by mediating the interaction between the integrin αVß5 of the hippocampal neurons and the CD90 molecules on the membrane of dopaminergic terminals. Our findings demonstrated that treadmill exercise could effectively alleviate cognitive disorders via the activation of the FNDC5-BDNF pathway and enhance the dopaminergic synaptic connection from SNpc to the hippocampus in the MPTP-induced chronic PD model.


Cognition Disorders , Parkinson Disease , Mice , Animals , Parkinson Disease/metabolism , Integrin alphaV/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Substantia Nigra/metabolism , Cognition Disorders/metabolism , Dopamine/metabolism , Transcription Factors/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Dopaminergic Neurons/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Fibronectins/metabolism
7.
CNS Neurosci Ther ; 29(4): 1142-1153, 2023 04.
Article En | MEDLINE | ID: mdl-36740277

INTRODUCTION: The relationship between oscillatory activity in hippocampus and cognitive impairment in traumatic brain injury (TBI) remains unclear. Although TBI decreases gamma oscillations and 40 Hz light flicker improves TBI prognosis, the effects and mechanism of rhythmic flicker on TBI remain unclear. AIMS: In this study, we aimed to explore whether light flicker could reverse cognitive deficits, and further explore its potential mechanisms in TBI mouse model. METHODS: The Morris water maze test (MWM), step-down test (SDT), and novel object recognition test (NOR) were applied to evaluate the cognitive ability. The local field potential (LFP) recording was applied to measure low gamma reduction of CA1 in hippocampus after TBI. And electrophysiological experiments were applied to explore effects of the gamma frequency entrainment on long-term potentiation (LTP), postsynaptic transmission, and intrinsic excitability of CA1 pyramidal cells (PCs) in TBI mice. Immunofluorescence staining and western blotting were applied to explore the effects of 40 Hz light flicker on the expression of PSD95 in hippocampus of TBI mice. RESULTS: We found that 40 Hz light flicker restored low gamma reduction of CA1 in hippocampus after TBI. And 40 Hz, but not random or 80 Hz light flicker, reversed cognitive impairment after TBI in behavioral tests. Moreover, 40 Hz light flicker improved N-methyl-D-aspartate (NMDA) receptor-dependent LTP (LTPNMDAR ) and L-type voltage-gated calcium channel-dependent LTP (LTPL-VGCC ) after TBI treatment. And gamma frequency entrainment decreased excitatory postsynaptic currents (EPSCs) of CA1 PCs in TBI mice. Our results have illustrated that 40 Hz light flicker could decrease intrinsic excitability of PCs after TBI treatment in mice. Furthermore, 40 Hz light flicker decreased the expression of PSD95 in hippocampus of TBI mice. CONCLUSION: These results demonstrated that 40 Hz light flicker rescues cognitive impairment by decreasing postsynaptic transmission in PCs after TBI treatment in mice.


Brain Injuries, Traumatic , Cognition Disorders , Cognitive Dysfunction , Mice , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Long-Term Potentiation/physiology , Hippocampus/metabolism , Cognition Disorders/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
8.
Neurosci Lett ; 797: 137071, 2023 02 16.
Article En | MEDLINE | ID: mdl-36642239

Early-life social isolation induces emotional and cognitive dysregulation, such as increased aggression and anxiety, and decreases neuron excitability in the medial prefrontal cortex (mPFC). The noradrenergic system in the mPFC regulates emotion and cognitive function via α1 or α2A adrenergic receptors, depending on noradrenaline levels. However, social isolation-induced changes in the mPFC noradrenergic system have not been reported. Here, male Wistar rats received post-weaning social isolation for nine consecutive weeks and were administered behavioral tests (novel object recognition, elevated plus maze, aggression, and forced swimming, sequentially). Protein expression levels in the mPFC noradrenergic system (α1 and α2A adrenergic receptors, tyrosine hydroxylase, and dopamine-ß-hydroxylase used as indices of noradrenaline synthesis and release) were examined through western blotting. Social isolation caused cognitive dysfunction, anxiety-like behavior, and aggression, but not behavioral despair. Socially-isolated rats exhibited increased protein levels of the α1 adrenergic receptor, tyrosine hydroxylase, and dopamine-ß-hydroxylase in the mPFC; there was no significant difference between the groups in the α2A adrenergic receptor expression levels. Preferential activation of the α1 adrenergic receptor caused by high noradrenaline concentration in the mPFC may be involved in social isolation-induced emotional and cognitive regulation impairments. Targeting the α1 adrenergic receptor signaling pathway is a potential therapeutic strategy for psychiatric disorders with symptomatic features such as emotional and cognitive dysregulation.


Affective Symptoms , Cognition Disorders , Dopamine , Norepinephrine , Prefrontal Cortex , Receptors, Adrenergic, alpha-1 , Social Isolation , Animals , Male , Rats , Anxiety , Cognition , Dopamine/metabolism , Norepinephrine/metabolism , Prefrontal Cortex/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Adrenergic/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Signal Transduction , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation , Weaning , Cognition Disorders/metabolism , Affective Symptoms/metabolism
9.
CNS Neurosci Ther ; 29(5): 1229-1242, 2023 05.
Article En | MEDLINE | ID: mdl-36694341

AIMS: This study was designed to investigate the role of NR2B and the contribution of DNA methylation to NR2B expression in the pathogenesis of PND. METHODS: Eighteen-month-old C57BL/6J mice were subjected to experimental laparotomy under 1.4% isoflurane anesthesia. Hippocampus-dependent learning and memory were evaluated by using the Barnes maze and contextual fear conditioning tests. The protein and mRNA expression levels of NR2B were evaluated by western blotting and qRT-PCR respectively, and the methylation of the NR2B gene was examined by using targeted bisulfite sequencing. Long-term synaptic plasticity (LTP) was measured by electrophysiology. RESULTS: Mice that underwent laparotomy exhibited hippocampus-dependent cognitive deficits accompanied by decreased NR2B expressions and LTP deficiency. The overexpression of NR2B in the dorsal hippocampus could improve learning and memory in mice subjected to laparotomy. In particular, the decreased NR2B expressions induced by laparotomy was attributed to the NR2B gene hypermethylation. Preoperative administration of S-adenosylmethionine (SAM) could hypomethylate the NR2B gene, upregulate NR2B expression and improve LTP, exerting a dose-dependent therapeutic effect against PND. Moreover, inhibiting NR2B abrogated the benefits of SAM pretreatment. CONCLUSIONS: Laparotomy cause hippocampus-dependent cognitive decline by hypermethylating the NR2B gene, allowing us to understand the pathogenesis of PND in an epigenetic landscape.


Cognition Disorders , DNA Methylation , Receptors, N-Methyl-D-Aspartate , Animals , Mice , Cognition Disorders/metabolism , DNA/metabolism , Hippocampus/metabolism , Learning , Mice, Inbred C57BL , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Laparotomy
10.
Acta Neurobiol Exp (Wars) ; 82(2): 207-212, 2022.
Article En | MEDLINE | ID: mdl-35833820

The current study aimed to investigate the effects of LACC1 on cognitive disorder due to stroke, as well as its underlying mechanism. LACC1 promoted inflammation and aggravated cognitive impairment in a mouse model of stroke. In an in vitro model of stroke, inhibition of LACC1 reduced inflammation and ROS­induced oxidative stress by activating AMP­activated protein kinase (AMPK) expression and suppressing NLPR3 expression. Furthermore, our studies revealed that inhibition of AMPK activity reduced the effects of si­LACC1 on cognitive disorder in mice after stroke via the AMPK/NLPR3 pathway. AMPK activation also reduced the effects of LACC1 on inflammation and ROS­induced oxidative stress via the NLPR3 pathway in the in vitro model that we evaluated. Our study suggests that LACC1­aggravated inflammation causes cognitive impairment after stroke via the AMPK/NLRP3 pathway, which may provide a new therapeutic target for stroke and other neurological diseases and their associated complications. In sum, we identified an important role and regulatory mechanism for LACC1 in maintaining stroke­induced cognitive disorder via the AMPK/NLRP3 pathway.


AMP-Activated Protein Kinases , Cognition Disorders , Inflammation , Intracellular Signaling Peptides and Proteins , NLR Family, Pyrin Domain-Containing 3 Protein , Stroke , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cognition/physiology , Cognition Disorders/etiology , Cognition Disorders/genetics , Cognition Disorders/metabolism , Inflammation/genetics , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/genetics , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Signal Transduction , Stroke/complications , Stroke/genetics , Stroke/metabolism
11.
Nutrients ; 14(3)2022 Jan 26.
Article En | MEDLINE | ID: mdl-35276898

A lower body mass is associated with the progression of Alzheimer's disease (AD) and the risk of mortality in patients with AD; however, evidence of genetic determinants of decreased body mass in cognitively impaired older adults is limited. We therefore investigated the genetic effect of APOE-ε4 on body composition in older adults with mild cognitive impairment (MCI) and early-to-moderate-stage AD. A total of 1631 outpatients (aged 65-89 years) with MCI and early-to-moderate-stage AD were evaluated for the association between body composition and APOE-ε4 status. After adjusting for covariates, including cognitive function evaluated with the Mini-Mental State Examination, the presence of the APOE-ε4 was associated with lower weight (ß = -1.116 ± 0.468 kg per presence, p = 0.017), fat mass (ß = -1.196 ± 0.401 kg per presence, p = 0.003), and percentage of body fat (ß = -1.700 ± 0.539% per presence, p = 0.002) in women but not in men. Additionally, the impact of APOE-ε4 on measures of body composition in women was more remarkable in MCI than in AD patients. The presence of the APOE-ε4 allele was associated with lower fat mass, particularly in women with MCI, independent of cognitive decline.


Alzheimer Disease , Cognition Disorders , Cognitive Dysfunction , Age Factors , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/psychology , Apolipoprotein E4/genetics , Cognition , Cognition Disorders/metabolism , Cognition Disorders/psychology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/psychology , Female , Humans , Male
12.
Int J Mol Sci ; 23(4)2022 Feb 15.
Article En | MEDLINE | ID: mdl-35216241

Treatment of negative symptoms and cognitive disorders in patients with schizophrenia is still a serious clinical problem. The aim of our study was to compare the efficacy of chronic administration of the atypical antipsychotic drug aripiprazole (7-{4-[4-(2,3-dichlorophenyl)-1-piperazinyl] butoxy}-3,4-dihydro-2(1H)-quinolinone; ARI) and the well-known antioxidant N-acetylcysteine (NAC) both in alleviating schizophrenia-like social and cognitive deficits and in reducing the decreases in the levels of the brain-derived neurotrophic factor (BDNF) in the prefrontal cortex (PFC) and hippocampus (HIP) of adult Sprague-Dawley rats, that have been induced by chronic administration of the model compound L-buthionine-(S, R)-sulfoximine (BSO) during the early postnatal development (p5-p16). ARI was administered at doses of 0.1 and 0.3 mg/kg while NAC at doses of 10 and 30 mg/kg, alone or in combination. Administration of higher doses of ARI or NAC alone, or co-treatment with lower, ineffective doses of these drugs significantly improved social and cognitive performance as assessed in behavioral tests. Both doses of NAC and 0.3 mg/kg of ARI increased the expression of BDNF mRNA in the PFC, while all doses of these drugs and their combinations enhanced the levels of BDNF protein in this brain structure. In the HIP, only 0,3 mg/kg ARI increased the levels of both BDNF mRNA and its protein. These data show that in the rat BSO-induced neurodevelopmental model of schizophrenia, ARI and NAC differently modulated BDNF levels in the PFC and HIP.


Acetylcysteine/pharmacology , Aripiprazole/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Cognition/drug effects , Hippocampus/drug effects , Prefrontal Cortex/drug effects , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Disease Models, Animal , Female , Hippocampus/metabolism , Prefrontal Cortex/metabolism , Pregnancy , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley , Schizophrenia/metabolism , Social Behavior
13.
Anal Biochem ; 636: 114437, 2022 01 01.
Article En | MEDLINE | ID: mdl-34715068

Hepatic encephalopathy and depression share a number of clinical features, such as cognitive impairment and psychomotor retardation, and are highly prevalent in patients with chronic liver disease. Both conditions signify a poor prognosis, carry an increased mortality and are major determinants of reduced health related quality of life. The pathophysiology of hepatic encephalopathy is complex. Whilst cerebral accumulation of ammonia is well-recognised as being central to the development of hepatic encephalopathy, systemic inflammation, which acts in synergy with hyperammonaemia, is emerging as a key driver in its development. The pro-inflammatory state is also widely documented in depression, and peripheral to brain communication occurs resulting in central inflammation, behavioural changes and depressive symptoms. Gut dysbiosis, with a similar reduction in beneficial bacteria, increase in pathogens and decreased bacterial diversity, has been observed in both hepatic encephalopathy and depression, and it may be that the resultant increased bacterial translocation causes their shared inflammatory pathophysiology. Whilst the literature on a positive association between hepatic encephalopathy and depression in cirrhosis remains to be substantiated, there is evolving evidence that treatment with psychobiotics may be of dual benefit, improving cognition and mood in cirrhosis.


Cognition Disorders , Depression , Hepatic Encephalopathy , Hyperammonemia , Liver Cirrhosis , Brain/metabolism , Brain/physiopathology , Chronic Disease , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Depression/etiology , Depression/metabolism , Depression/physiopathology , Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/metabolism , Hepatic Encephalopathy/physiopathology , Humans , Hyperammonemia/etiology , Hyperammonemia/metabolism , Hyperammonemia/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Liver Cirrhosis/complications , Liver Cirrhosis/metabolism , Liver Cirrhosis/physiopathology , Quality of Life
14.
BMC Neurosci ; 22(1): 78, 2021 12 15.
Article En | MEDLINE | ID: mdl-34911449

BACKGROUND: Sepsis is considered to be a high-risk factor for cognitive impairment in the brain. The purpose of our study is to explore whether sepsis causes cognitive impairment and try to evaluate the underlying mechanisms and intervention measures. METHODS: Here, we used cecum ligation and puncture (CLP) to simulate sepsis. Open field, Novel Objective Recognition, and Morris Water Maze Test were used to detect cognitive function, long-term potentiation was used to assess of synaptic plasticity, and molecular biological technics were used to assess synaptic proteins, ELISA kits were used to detect inflammatory factors. Metformin was injected into the lateral ventricle of SD rats, and we evaluated whether metformin alleviated CLP-mediated cognitive impairment using behavioral, electrophysiological and molecular biological technology experiments. RESULTS: Here we report hippocampal-dependent cognitive deficits and synaptic dysfunction induced by the CLP, accompanied by a significant increase in inflammatory factors. At the same time, metformin was able to improve cognitive impairment induced by CLP in adult male rats. CONCLUSION: These findings highlight a novel pathogenic mechanism of sepsis-related cognitive impairment through activation of inflammatory factors, and these are blocked by metformin to attenuate sepsis-induced neuronal injury and cognitive impairment.


Cognition/drug effects , Cognitive Dysfunction/drug therapy , Metformin/pharmacology , Sepsis/complications , Animals , Brain/drug effects , Brain/metabolism , Cecum/drug effects , Cecum/injuries , Cecum/metabolism , Cecum/pathology , Cognition/physiology , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Cognitive Dysfunction/complications , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Ligation/adverse effects , Male , Neurons/drug effects , Neurons/metabolism , Rats, Sprague-Dawley , Sepsis/drug therapy , Sepsis/metabolism
15.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article En | MEDLINE | ID: mdl-34884506

Due to their potent antibacterial properties, silver nanoparticles (AgNPs) are widely used in industry and medicine. However, they can cross the brain-blood barrier, posing a risk to the brain and its functions. In our previous study, we demonstrated that oral administration of bovine serum albumin (BSA)-coated AgNPs caused an impairment in spatial memory in a dose-independent manner. In this study, we evaluated the effects of AgNPs coating material on cognition, spatial memory functioning, and neurotransmitter levels in rat hippocampus. AgNPs coated with BSA (AgNPs(BSA)), polyethylene glycol (AgNPs(PEG)), or citrate (AgNPs(Cit)) or silver ions (Ag+) were orally administered at a dose of 0.5 mg/kg b.w. to male Wistar rats for a period of 28 days, while the control (Ctrl) rats received 0.2 mL of water. The acquisition and maintenance of spatial memory related to place avoidance were assessed using the active allothetic place avoidance task, in which rats from AgNPs(BSA), AgNPs(PEG), and Ag+ groups performed worse than the Ctrl rats. In the retrieval test assessing long-term memory, only rats from AgNPs(Cit) and Ctrl groups showed memory maintenance. The analysis of neurotransmitter levels indicated that the ratio between serotonin and dopamine concentration was disturbed in the AgNPs(BSA) rats. Furthermore, treatment with AgNPs or Ag+ resulted in the induction of peripheral inflammation, which was reflected by the alterations in the levels of serum inflammatory mediators. In conclusion, depending on the coating material used for their stabilization, AgNPs induced changes in memory functioning and concentration of neurotransmitters.


Cognition Disorders/pathology , Hippocampus/pathology , Metal Nanoparticles/toxicity , Polyethylene Glycols/toxicity , Serum Albumin, Bovine/toxicity , Silver/chemistry , Animals , Citrates/chemistry , Citrates/toxicity , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Cytokines/metabolism , Hippocampus/drug effects , Male , Metal Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Rats , Rats, Wistar , Serum Albumin, Bovine/chemistry
16.
Int J Mol Sci ; 22(21)2021 Nov 04.
Article En | MEDLINE | ID: mdl-34769380

Despite the well-accepted role of the two main neuropathological markers (ß-amyloid and tau) in the progression of Alzheimer's disease, the interaction and specific contribution of each of them is not fully elucidated. To address this question, in the present study, an adeno-associated virus (AAV9) carrying the mutant P301L form of human tau, was injected into the dorsal hippocampi of APP/PS1 transgenic mice or wild type mice (WT). Three months after injections, memory tasks, biochemical and immunohistochemical analysis were performed. We found that the overexpression of hTauP301L accelerates memory deficits in APP/PS1 mice, but it did not affect memory function of WT mice. Likewise, biochemical assays showed that only in the case of APP/PS1-hTauP301L injected mice, an important accumulation of tau was observed in the insoluble urea fraction. Similarly, electron microscopy images revealed that numerous clusters of tau immunoparticles appear at the dendrites of APP/PS1 injected mice and not in WT animals, suggesting that the presence of amyloid is necessary to induce tau aggregation. Interestingly, these tau immunoparticles accumulate in dendritic mitochondria in the APP/PS1 mice, whereas most of mitochondria in WT injected mice remain free of tau immunoparticles. Taken together, it seems that amyloid induces tau aggregation and accumulation in the dendritic mitochondria and subsequently may alter synapse function, thus, contributing to accelerate cognitive decline in APP/PS1 mice.


Alzheimer Disease/complications , Amyloid beta-Peptides/adverse effects , Cognition Disorders/pathology , Disease Models, Animal , Mitochondria/pathology , tau Proteins/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , Cognition Disorders/etiology , Cognition Disorders/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Phosphorylation , Presenilin-1/physiology , Synapses , tau Proteins/genetics
17.
Sci Rep ; 11(1): 20057, 2021 10 08.
Article En | MEDLINE | ID: mdl-34625629

Brain-derived neurotrophic factor (BDNF) is reported to be involved in cognitive decline in patients with schizophrenia (SZ). Previous studies have found that cognitive deficits remain stable during the chronic disease phase in SZ, but the findings were inconsistent. The role of BDNF in cognitive deficits at different stage of illness remains unclear. This study aimed to examine the effect of BDNF polymorphisms on cognitive deficits in drug-naïve first-episode (DNFE) patients and chronic patients with SZ. 262 DNFE patients, 844 chronic patients, and 1043 healthy controls were recruited to compare 4 polymorphisms in BDNF gene and cognitive function. We found that there was no significant difference in genotype and allele frequencies between SZ patients and controls. However, they were closely related to cognitive functioning. BDNF rs2030324 polymorphism played a strong role in language performance only in DNFE patients with SZ. The language index of DNFE patients with rs2030324 TT and TC genotypes was worse than that of chronic patients, but there was no significant difference in CC genotypes between DNFE and chronic patients. Rs6265 had no significant effect on cognitive functioning in patients and controls. Our result suggests BDNF gene polymorphisms were related to different domains of cognitive function at the different stage of SZ, especially language in DNFE patients.


Brain-Derived Neurotrophic Factor/genetics , Cognition Disorders/pathology , Polymorphism, Genetic , Schizophrenia/complications , Adolescent , Adult , Aged , Case-Control Studies , Cognition Disorders/etiology , Cognition Disorders/metabolism , Female , Follow-Up Studies , Genotype , Humans , Male , Middle Aged , Pharmaceutical Preparations , Prognosis , Young Adult
18.
Nat Metab ; 3(8): 1058-1070, 2021 08.
Article En | MEDLINE | ID: mdl-34417591

Identifying secreted mediators that drive the cognitive benefits of exercise holds great promise for the treatment of cognitive decline in ageing or Alzheimer's disease (AD). Here, we show that irisin, the cleaved and circulating form of the exercise-induced membrane protein FNDC5, is sufficient to confer the benefits of exercise on cognitive function. Genetic deletion of Fndc5/irisin (global Fndc5 knock-out (KO) mice; F5KO) impairs cognitive function in exercise, ageing and AD. Diminished pattern separation in F5KO mice can be rescued by delivering irisin directly into the dentate gyrus, suggesting that irisin is the active moiety. In F5KO mice, adult-born neurons in the dentate gyrus are morphologically, transcriptionally and functionally abnormal. Importantly, elevation of circulating irisin levels by peripheral delivery of irisin via adeno-associated viral overexpression in the liver results in enrichment of central irisin and is sufficient to improve both the cognitive deficit and neuropathology in AD mouse models. Irisin is a crucial regulator of the cognitive benefits of exercise and is a potential therapeutic agent for treating cognitive disorders including AD.


Cognition , Fibronectins/metabolism , Hormones/metabolism , Physical Conditioning, Animal , Animals , Behavior, Animal , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/psychology , Disease Models, Animal , Fibronectins/genetics , Gene Deletion , Gene Expression , Mice , Mice, Knockout , Phenotype
19.
Cells ; 10(7)2021 07 20.
Article En | MEDLINE | ID: mdl-34359999

Much progress has been made toward deciphering RHO GTPase functions, and many studies have convincingly demonstrated that altered signal transduction through RHO GTPases is a recurring theme in the progression of human malignancies. It seems that 20 canonical RHO GTPases are likely regulated by three GDIs, 85 GEFs, and 66 GAPs, and eventually interact with >70 downstream effectors. A recurring theme is the challenge in understanding the molecular determinants of the specificity of these four classes of interacting proteins that, irrespective of their functions, bind to common sites on the surface of RHO GTPases. Identified and structurally verified hotspots as functional determinants specific to RHO GTPase regulation by GDIs, GEFs, and GAPs as well as signaling through effectors are presented, and challenges and future perspectives are discussed.


GTPase-Activating Proteins/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics , rhoA GTP-Binding Protein/genetics , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cognition Disorders/genetics , Cognition Disorders/metabolism , Cognition Disorders/pathology , Communicable Diseases/genetics , Communicable Diseases/metabolism , Communicable Diseases/pathology , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Humans , Multigene Family , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , rhoA GTP-Binding Protein/metabolism
20.
Mol Neurobiol ; 58(11): 5954-5970, 2021 Nov.
Article En | MEDLINE | ID: mdl-34435330

The influence of long-term tacrolimus treatment on cognitive function remains to be elucidated. Using a murine model of chronic tacrolimus neurotoxicity, we evaluated the effects of tacrolimus on cognitive function, synaptic balance, its regulating protein (Klotho), and oxidative stress in the hippocampus. Compared to vehicle-treated mice, tacrolimus-treated mice showed significantly decreased hippocampal-dependent spatial learning and memory function. Furthermore, tacrolimus caused synaptic imbalance, as demonstrated by decreased excitatory synapses and increased inhibitory synapses, and downregulated Klotho in a dose-dependent manner; the downregulation of Klotho was localized to excitatory hippocampal synapses. Moreover, tacrolimus increased oxidative stress and was associated with activation of the PI3K/AKT pathway in the hippocampus. These results indicate that tacrolimus impairs cognitive function via synaptic imbalance, and that these processes are associated with Klotho downregulation at synapses through tacrolimus-induced oxidative stress in the hippocampus.


Cognition Disorders/chemically induced , Hippocampus/physiopathology , Immunosuppressive Agents/toxicity , Klotho Proteins/physiology , Nerve Tissue Proteins/physiology , Synapses/drug effects , Tacrolimus/toxicity , Animals , Cognition Disorders/metabolism , Dendrites/metabolism , Down-Regulation/drug effects , Hippocampus/pathology , Immunosuppressive Agents/pharmacology , Klotho Proteins/biosynthesis , Klotho Proteins/genetics , Male , Maze Learning , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Open Field Test , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Random Allocation , Signal Transduction , Spatial Learning , Spatial Memory , Synapses/physiology , Tacrolimus/pharmacology
...