1
|
Chen L, Yue Z, Liu Z, Liu H, Zhang J, Zhang F, Hu T, Fu J. The impact of Nrf2 knockout on the neuroprotective effects of dexmedetomidine in a mice model of cognitive impairment. Behav Brain Res 2024; 469:115006. [PMID: 38692357 DOI: 10.1016/j.bbr.2024.115006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/08/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024]
Abstract
The nuclear factor erythroid 2-related factor 2 (Nrf2) signalling pathway represents a crucial intrinsic protective system against oxidative stress and inflammation and plays a significant role in various neurological disorders. However, the effect of Nrf2 signalling on the regulation of cognitive impairment remains unknown. Dexmedetomidine (DEX) has neuroprotective effects and can ameliorate lipopolysaccharide (LPS)-induced cognitive dysfunction. Our objective was to observe whether Nrf2 knockout influences the efficacy of DEX in improving cognitive impairment and to attempt to understand its underlying mechanisms. An LPS-induced cognitive dysfunction model in wild-type and Nrf2 knockout mice (Institute of Cancer Research background; male; 8-12 weeks) was used to observe the impact of DEX on cognitive dysfunction. LPS was intraperitoneally injected, followed by novel object recognition and morris water maze experiments 24 h later. Hippocampal tissues were collected for histopathological and molecular analyses. Our research findings suggest that DEX enhances the expression of NQO1, HO-1, PSD95, and SYP proteins in hippocampal tissue, inhibits microglial proliferation, reduces pro-inflammatory cytokines IL-1β and TNF-ɑ, increases anti-inflammatory cytokine IL-10, and improves dendritic spine density, thereby alleviating cognitive dysfunction induced by LPS. However, the knockout of the Nrf2 gene negated the aforementioned effects of DEX. In conclusion, DEX alleviates cognitive deficits induced by LPS through mechanisms of anti-oxidative stress and anti-inflammation, as well as by increasing synaptic protein expression and dendritic spine density. However, the knockout of the Nrf2 gene reversed the effects of DEX. The Nrf2 signaling pathway plays a crucial role in the mitigation of LPS-induced cognitive impairment by DEX.
Collapse
Affiliation(s)
- Liang Chen
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Department of Anesthesiology, Shijiazhuang Fourth Hospital, Shijiazhuang, China
| | - Zhifeng Yue
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ziyu Liu
- Department of Human Anatomy, Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huaqin Liu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jin Zhang
- Department of Anesthesiology, Shijiazhuang Fourth Hospital, Shijiazhuang, China
| | - Fengjiao Zhang
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tao Hu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianfeng Fu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
2
|
Nibuya M, Kezuka D, Kanno Y, Wakamatsu S, Suzuki E. Behavioral stress and antidepressant treatments altered hippocampal expression of Nogo signal-related proteins in rats. J Psychiatr Res 2024; 170:207-216. [PMID: 38157668 DOI: 10.1016/j.jpsychires.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/26/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Some immune molecules including neurite outgrowth inhibitor (Nogo) ligands and their receptor(Nogo receptor-1: NgR1)are expressed at the neuronal synaptic sites. Paired immunoglobulin-like receptor B (PirB) is another Nogo receptor that also binds to major histocompatibility complex I and β-amyloid and suppresses dendritic immune cell functions and neuronal plasticity in the central nervous system. Augmenting structural and functional neural plasticity by manipulating the Nogo signaling pathway is a novel promising strategy for treating brain ischemia and degenerative processes such as Alzheimer's disease. In recent decades psychiatric research using experimental animals has focused on the attenuation of neural plasticity by stress loadings and on the enhanced resilience by psychopharmacological treatments. In the present study, we examined possible expressional alterations in Nogo signal-related proteins in the rat hippocampus after behavioral stress loadings and antidepressant treatments. To validate the effectiveness of the procedures, previously reported increase in brain-derived neurotrophic factor (BDNF) by ECS or ketamine administration and decrease of BDNF by stress loadings are also shown in the present study. Significant increases in hippocampal NgR1 and PirB expression were observed following chronic variable stress, and a significant increase in NgR1 expression was observed under a single prolonged stress paradigm. These results indicate a possible contribution of enhanced Nogo signaling to the attenuation of neural plasticity in response to stressful experiences. Additionally, the suppression of hippocampal NgR1 expression using electroconvulsive seizure treatment and administration of subanesthetic dose of ketamine supported the increased neural plasticity induced by the antidepressant treatments.
Collapse
Affiliation(s)
- Masashi Nibuya
- Division of Psychiatry, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino, Sendai City, Miyagi, 983-8536, Japan.
| | - Dai Kezuka
- Division of Psychiatry, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino, Sendai City, Miyagi, 983-8536, Japan
| | - Yoshihiko Kanno
- Division of Psychiatry, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino, Sendai City, Miyagi, 983-8536, Japan
| | - Shunosuke Wakamatsu
- Division of Psychiatry, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino, Sendai City, Miyagi, 983-8536, Japan
| | - Eiji Suzuki
- Division of Psychiatry, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino, Sendai City, Miyagi, 983-8536, Japan
| |
Collapse
|
3
|
Zhao P, Zhang J, Kuai J, Li L, Li X, Feng N, Du H, Li C, Wang Q, Deng B. TAT-PEP Alleviated Cognitive Impairment by Alleviating Neuronal Mitochondria Damage and Apoptosis After Cerebral Ischemic Reperfusion Injury. Mol Neurobiol 2023; 60:5655-5671. [PMID: 37335462 PMCID: PMC10471703 DOI: 10.1007/s12035-023-03404-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/27/2023] [Indexed: 06/21/2023]
Abstract
Paired immunoglobulin-like receptor B (PirB) was identified as a myelin-associated inhibitory protein (MAIP) receptor that plays a critical role in axonal regeneration, synaptic plasticity and neuronal survival after stroke. In our previous study, a transactivator of transcription-PirB extracellular peptide (TAT-PEP) was generated that can block the interactions between MAIs and PirB. We found that TAT-PEP treatment improved axonal regeneration, CST projection and long-term neurobehavioural recovery after stroke through its effects on PirB-mediated downstream signalling. However, the effect of TAT-PEP on the recovery of cognitive function and the survival of neurons also needs to be investigated. In this study, we investigated whether pirb RNAi could alleviate neuronal injury by inhibiting the expression of PirB following exposure to oxygen-glucose deprivation (OGD) in vitro. In addition, TAT-PEP treatment attenuated the volume of the brain infarct and promoted the recovery of neurobehavioural function and cognitive function. This study also found that TAT-PEP exerts neuroprotection by reducing neuronal degeneration and apoptosis after ischemia-reperfusion injury. In addition, TAT-PEP improved neuron survival and reduced lactate dehydrogenase (LDH) release in vitro. Results also showed that TAT-PEP reduced malondialdehyde (MDA) levels, increased superoxide dismutase (SOD) activity and reduced reactive oxygen species (ROS) accumulation in OGD-injured neurons. The possible mechanism was that TAT-PEP could contribute to the damage of neuronal mitochondria and affect the expression of cleaved caspase 3, Bax and Bcl-2. Our results suggest that PirB overexpression in neurons after ischaemic-reperfusion injury induces neuronal mitochondrial damage, oxidative stress and apoptosis. This study also suggests that TAT-PEP may be a potent neuroprotectant with therapeutic potential for stroke by reducing neuronal oxidative stress, mitochondrial damage, degeneration and apoptosis in ischemic stroke.
Collapse
Affiliation(s)
- Pin Zhao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
- Department of Anesthesiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710018, People's Republic of China
| | - Jiapo Zhang
- Department of Emergency Medicine, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, 361102, Fujian, China
| | - JianKe Kuai
- Department of Anesthesiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710018, People's Republic of China
| | - Liya Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, Liaoning, China
| | - Xuying Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Namin Feng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hailiang Du
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Chen Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Bin Deng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
4
|
Nouraeinejad A. The functional and structural changes in the hippocampus of COVID-19 patients. Acta Neurol Belg 2023; 123:1247-1256. [PMID: 37226033 PMCID: PMC10208918 DOI: 10.1007/s13760-023-02291-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
Since the hippocampus is predominantly susceptible to injuries caused by COVID-19, there are increasing data indicating the likelihood of post-infection memory loss and quickening neurodegenerative disorders, such as Alzheimer's disease. This is due to the fact that the hippocampus has imperative functions in spatial and episodic memory as well as learning. COVID-19 activates microglia in the hippocampus and induces a CNS cytokine storm, leading to loss of hippocampal neurogenesis. The functional and structural changes in the hippocampus of COVID-19 patients can explain neuronal degeneration and reduced neurogenesis in the human hippocampus. This will open a window to explain memory and cognitive dysfunctions in "long COVID" through the resultant loss of hippocampal neurogenesis.
Collapse
Affiliation(s)
- Ali Nouraeinejad
- Faculty of Brain Sciences, Institute of Ophthalmology, University College London (UCL), London, UK.
| |
Collapse
|
5
|
Martin M, Pusceddu MM, Teichenné J, Negra T, Connolly A, Escoté X, Torrell Galceran H, Cereto Massagué A, Samarra Mestre I, Del Pino Rius A, Romero-Gimenez J, Egea C, Alcaide-Hidalgo JM, Del Bas JM. Preventive Treatment with Astaxanthin Microencapsulated with Spirulina Powder, Administered in a Dose Range Equivalent to Human Consumption, Prevents LPS-Induced Cognitive Impairment in Rats. Nutrients 2023; 15:2854. [PMID: 37447181 DOI: 10.3390/nu15132854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Cognitive alterations are a common feature associated with many neurodegenerative diseases and are considered a major health concern worldwide. Cognitive alterations are triggered by microglia activation and oxidative/inflammatory processes in specific areas of the central nervous system. Consumption of bioactive compounds with antioxidative and anti-inflammatory effects, such as astaxanthin and spirulina, can help in preventing the development of these pathologies. In this study, we have investigated the potential beneficial neuroprotective effects of a low dose of astaxanthin (ASX) microencapsulated within spirulina (ASXSP) in female rats to prevent the cognitive deficits associated with the administration of LPS. Alterations in memory processing were evaluated in the Y-Maze and Morris Water Maze (MWM) paradigms. Changes in microglia activation and in gut microbiota content were also investigated. Our results demonstrate that LPS modified long-term memory in the MWM and increased microglia activation in the hippocampus and prefrontal cortex. Preventive treatment with ASXSP ameliorated LPS-cognitive alterations and microglia activation in both brain regions. Moreover, ASXSP was able to partially revert LPS-induced gut dysbiosis. Our results demonstrate the neuroprotective benefits of ASX when microencapsulated with spirulina acting through different mechanisms, including antioxidant, anti-inflammatory and, probably, prebiotic actions.
Collapse
Affiliation(s)
- Miquel Martin
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Matteo M Pusceddu
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Joan Teichenné
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | | | | | - Xavier Escoté
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Helena Torrell Galceran
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Adrià Cereto Massagué
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Iris Samarra Mestre
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Antoni Del Pino Rius
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Jordi Romero-Gimenez
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Cristina Egea
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | | | - Josep Maria Del Bas
- Eurecat-Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain
| |
Collapse
|
6
|
Li L, Mou Y, Zhai Q, Yan C, Zhang X, Du M, Li Y, Wang Q, Xiao Z. PirB negatively regulates the inflammatory activation of astrocytes in a mouse model of sleep deprivation. Neuropharmacology 2023; 235:109571. [PMID: 37146940 DOI: 10.1016/j.neuropharm.2023.109571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/16/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Reactive astrocytes play a potential regulatory role in sleep deprivation (SD). Paired immunoglobulin-like receptor B (PirB) is expressed in reactive astrocytes, suggesting that PirB may participate in regulating the inflammatory response of astrocytes. We used lentiviral and adeno-associated viral approaches to interfere with the expression of PirB in vivo and in vitro. C57BL/6 mice were sleep deprived for 7 days and neurological function was measured via behavioral tests. We found that overexpressed PirB in SD mice could decrease the number of neurotoxic reactive astrocytes, alleviate cognitive deficits, and promote reactive astrocytes tended to be neuroprotective state. IL-1α, TNFα, and C1q were used to induce neurotoxic reactive astrocytes in vitro. Overexpression of PirB relieved the toxicity of neurotoxic astrocytes. Silencing PirB expression had the opposite effect and exacerbated the transition of reactive astrocytes to a neurotoxic state in vitro. Moreover, PirB-impaired astrocytes demonstrated STAT3 hyperphosphorylation which could be reversed by stattic (p-STAT3 inhibitor). Furthermore, Golgi-Cox staining confirmed that dendrite morphology defects and synapse-related protein were significantly increased in PirB-overexpressed SD mice. Our data demonstrated that SD induced neurotoxic reactive astrocytes and contributed to neuroinflammation and cognitive deficits. PirB performs a negative regulatory role in neurotoxic reactive astrocytes via the STAT3 signaling pathway in SD.
Collapse
Affiliation(s)
- Liya Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Yan Mou
- The Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Qian Zhai
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Chaoying Yan
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xin Zhang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Mengyu Du
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Zhaoyang Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning, China.
| |
Collapse
|
7
|
Li X, Zhai Q, Gou X, Quan M, Li Y, Zhang X, Deng B, Tian Y, Wang Q, Hou L. Involvement of Paired Immunoglobulin-Like Receptor B in Cognitive Dysfunction Through Hippocampal-Dependent Synaptic Plasticity Impairments in Mice Subjected to Chronic Sleep Restriction. Mol Neurobiol 2023; 60:1132-1149. [PMID: 36417104 PMCID: PMC9899186 DOI: 10.1007/s12035-022-03127-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022]
Abstract
Sleep loss is often associated with cognitive dysfunction. Alterations in the structure and function of synapses in the hippocampus are thought to underlie memory storage. Paired immunoglobulin-like receptor B (PirB) plays a negative role in various neurological diseases by inhibiting axon regeneration and synaptic plasticity. However, the contributions of PirB to the mechanisms underlying the changes in synaptic plasticity after sleep loss that ultimately promote deficits in cognitive function have not been well elucidated. Here, we showed that chronic sleep restriction (CSR) mice displayed cognitive impairment and synaptic deficits accompanied by upregulation of PirB expression in the hippocampus. Mechanistically, PirB caused the dysregulation of actin through the RhoA/ROCK2/LIMK1/cofilin signalling pathway, leading to abnormal structural and functional plasticity, which in turn resulted in cognitive dysfunction. PirB knockdown alleviated synaptic deficits and cognitive impairment after CSR by inhibiting the RhoA/ROCK2/LIMK1/cofilin signalling pathway. Moreover, we found that fasudil, a widely used ROCK2 inhibitor, could mimic the beneficial effect of PirB knockdown and ameliorate synaptic deficits and cognitive impairment, further demonstrating that PirB induced cognitive dysfunction after CSR via the RhoA/ROCK2/LIMK1/cofilin signalling pathway. Our study sheds new light on the role of PirB as an important mediator in modulating the dysfunction of synaptic plasticity and cognitive function via the RhoA/ROCK2/LIMK1/cofilin signalling pathway, which indicated that hippocampal PirB is a promising therapeutic target for counteracting cognitive impairment after CSR. This illustration depicts the signalling pathway by PirB in mediating cognitive impairment and synaptic deficits in CSR mice. In the hippocampus of CSR mice, the expression level of PirB was significantly increased. In addition, CSR increases RhoA and ROCK2 levels and reduces levels of both LIMK1 and cofilin phosphorylation. PirB knockdown reverses cognitive impairment and synaptic plasticity disorders caused by CSR through the RhoA/ROCK2/LIMK1/cofilin signalling pathway.
Collapse
Affiliation(s)
- Xuying Li
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102 Fujian China ,Department of Anesthesiology, Affiliated Haikou Hospital, Xiangya Medical College of Central South University, Haikou, 570000 Hainan China
| | - Qian Zhai
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, 710021 Shaanxi China
| | - Minxue Quan
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102 Fujian China
| | - Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi China
| | - Xiaohua Zhang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, 710021 Shaanxi China
| | - Bin Deng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi China
| | - Yi Tian
- Department of Anesthesiology, Affiliated Haikou Hospital, Xiangya Medical College of Central South University, Haikou, 570000 Hainan China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 Shaanxi China
| | - Lichao Hou
- Department of Anesthesiology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102 Fujian China
| |
Collapse
|
8
|
Pechacek KM, Vonder Haar C. Chronic lipopolysaccharide impairs motivation when delivered to the ventricles, but not when delivered peripherally in male rats. Physiol Behav 2022; 257:113998. [PMID: 36257462 PMCID: PMC11305111 DOI: 10.1016/j.physbeh.2022.113998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/18/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
Abstract
Increased neuroinflammation relative to controls is observed in major depression. Moreover, depressive disorders are significantly elevated in conditions which increase neuroinflammation (e.g., brain injury, Parkinson's disease, Alzheimer's disease). To better understand the relationship between neuroinflammation and depression, additional research is needed. The current set of studies made use of the progressive ratio (PR) task in male rats, a stable measure of motivation which can be evaluated daily and thus is ideally suited for examining a potential role for chronic neuroinflammation in depressive-like behavior. Lipopolysaccharide (LPS) was used to induce an inflammatory response. Experiment 1 confirmed prior acute LPS administration experiments for sensitivity of the PR task, with a large effect at 2 mg/kg, a partial effect at 1 mg/kg, and no effect at 0.5 mg/kg. Experiment 2 evaluated a dose-response of continuous s.c. LPS infusion but found no significant elevation in brain cytokines after 14 days at any doses of 0.1, 0.5, 1, or 2 mg/kg/week. Experiment 3 assessed motivation during continuous s.c. infusion of a large 5 mg/kg/week LPS dose and found no significant impairments in motivation, but transient decreases in rates of lever pressing (i.e., only motoric deficits). Experiment 4 measured motivation during continuous ICV infusion of 10.5 μg/kg/week LPS and found significantly decreased motivation without changes to rates of lever pressing (i.e., only motivational deficits). Together these results suggest that the PR task is efficient for evaluating models of chronic inflammation, and that the adaptive response to chronic LPS exposure, even when delivered centrally, may necessitate alternative strategies for generating long-term neuroinflammation.
Collapse
Affiliation(s)
- Kristen M Pechacek
- Injury and Recovery Laboratory, Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Cole Vonder Haar
- Injury and Recovery Laboratory, Department of Neuroscience, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
9
|
Im H, Ju IG, Kim JH, Lee S, Oh MS. Trichosanthis Semen and Zingiberis Rhizoma Mixture Ameliorates Lipopolysaccharide-Induced Memory Dysfunction by Inhibiting Neuroinflammation. Int J Mol Sci 2022; 23:ijms232214015. [PMID: 36430493 PMCID: PMC9692726 DOI: 10.3390/ijms232214015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/02/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Neuroinflammation, a key pathological contributor to various neurodegenerative diseases, is mediated by microglial activation and subsequent secretion of inflammatory cytokines via the mitogen-activated protein kinase (MAPK) signaling pathway. Moreover, neuroinflammation leads to synaptic loss and memory impairment. This study investigated the inhibitory effects of PNP001, a mixture of Trichosanthis Semen and Zingiberis Rhizoma in a ratio of 3:1, on neuroinflammation and neurological deficits induced by lipopolysaccharide (LPS). For the in vitro study, PNP001 was administered in LPS-stimulated BV2 microglial cells, and reduced the pro-inflammatory mediators, such as nitric oxide, inducible nitric oxide synthase, and cyclooxygenase-2 by downregulating MAPK signaling. For the in vivo study, ICR mice were orally administered PNP001 for 18 consecutive days, and concurrently treated with LPS (1 mg/kg, i.p.) for 10 days, beginning on the 4th day of PNP001 administration. The remarkably decreased number of activated microglial cells and increased expression of pre- and post-synaptic proteins were observed more in the hippocampus of the PNP001 administered groups than in the LPS-treated group. Furthermore, daily PNP001 administration significantly attenuated long-term memory decline compared with the LPS-treated group. Our study demonstrated that PNP001 inhibits LPS-induced neuroinflammation and its associated memory dysfunction by alleviating microglial activation and synaptic loss.
Collapse
Affiliation(s)
- Hyeri Im
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - In Gyoung Ju
- Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul 02447, Korea
| | - Jin Hee Kim
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Seungmin Lee
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Myung Sook Oh
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul 02447, Korea
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Correspondence: ; Tel.: +82-2-961-9436; Fax: +82-2-963-9436
| |
Collapse
|
10
|
De Luca SN, Brassington K, Chan SMH, Dobric A, Mou K, Seow HJ, Vlahos R. Ebselen prevents cigarette smoke-induced cognitive dysfunction in mice by preserving hippocampal synaptophysin expression. J Neuroinflammation 2022; 19:72. [PMID: 35351173 PMCID: PMC8966248 DOI: 10.1186/s12974-022-02432-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/13/2022] [Indexed: 11/26/2022] Open
Abstract
Background Cigarette smoking (CS) is the leading cause of chronic obstructive pulmonary disease (COPD). The “spill-over” of pulmonary inflammation into the systemic circulation may damage the brain, leading to cognitive dysfunction. Cessation of CS can improve pulmonary and neurocognitive outcomes, however, its benefit on the neuroinflammatory profile remains uncertain. Here, we investigate how CS exposure impairs neurocognition and whether this can be reversed with CS cessation or an antioxidant treatment. Methods Male BALB/c mice were exposed to CS (9 cigarettes/day for 8 weeks) followed by 4 weeks of CS cessation. Another cohort of CS-exposed mice were co-administrated with a glutathione peroxidase mimetic, ebselen (10 mg/kg) or vehicle (5% CM-cellulose). We assessed pulmonary inflammation, spatial and working memory, and the hippocampal microglial, oxidative and synaptic profiles. Results CS exposure increased lung inflammation which was reduced following CS cessation. CS caused spatial and working memory impairments which were attributed to hippocampal microglial activation and suppression of synaptophysin. CS cessation did not improve memory deficits or alter microglial activation. Ebselen completely prevented the CS-induced working and spatial memory impairments, which was associated with restored synaptophysin expression without altering microglial activation. Conclusion We were able to model the CS-induced memory impairment and microglial activation seen in human COPD. The preventative effects of ebselen on memory impairment is likely to be dependent on a preserved synaptogenic profile. Cessation alone also appears to be insufficient in correcting the memory impairment, suggesting the importance of incorporating antioxidant therapy to help maximising the benefit of cessation.
Collapse
|
11
|
Widmann CN, Wieberneit M, Bieler L, Bernsen S, Gräfenkämper R, Brosseron F, Schmeel C, Tacik P, Skowasch D, Radbruch A, Heneka MT. Longitudinal Neurocognitive and Pulmonological Profile of Long COVID-19: Protocol for the COVIMMUNE-Clin Study. JMIR Res Protoc 2021; 10:e30259. [PMID: 34559059 PMCID: PMC8589042 DOI: 10.2196/30259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/12/2021] [Accepted: 09/08/2021] [Indexed: 01/10/2023] Open
Abstract
Background There is a dearth of information about “brain fog,” characterized by concentration, word-finding, or memory problems, which has been listed in the new World Health Organization provisional classification “U09.9 Post-COVID-19 Condition.” Moreover, the extent to which these symptoms may be associated with neurological, pulmonary, or psychiatric difficulties is unclear. Objective This ongoing cohort study aims to carefully assess neurocognitive function in the context of the neurological, psychiatric, and pulmonary sequelae of SARS-CoV-2 infection among patients with asymptomatic/mild and severe cases of COVID-19 after remission, including actively recruited healthy controls. Methods A total of 150 participants will be included in this pilot study. The cohort will comprise patients who tested positive for SARS-CoV-2 infection with either an asymptomatic course or a mild course defined as no symptoms except for olfactory and taste dysfunction (n=50), patients who tested positive for SARS-CoV-2 infection with a severe disease course (n=50), and a healthy control group (n=50) with similar age and sex distribution based on frequency matching. A comprehensive neuropsychological assessment will be performed comprising nuanced aspects of complex attention, including language, executive function, verbal and visual learning, and memory. Psychiatric, personality, social and lifestyle factors, sleep, and fatigue will be evaluated. Brain magnetic resonance imaging, neurological and physical assessment, and pulmonological and lung function examinations (including body plethysmography, diffusion capacity, clinical assessments, and questionnaires) will also be performed. Three visits are planned with comprehensive testing at the baseline and 12-month visits, along with brief neurological and neuropsychological examinations at the 6-month assessment. Blood-based biomarkers of neurodegeneration will be quantified at baseline and 12-month follow-up. Results At the time of submission, the study had begun recruitment through telephone and in-person screenings. The first patient was enrolled in the study at the beginning of April 2021. Interim data analysis of baseline information is expected to be complete by December 2021 and study completion is expected at the end of December 2022. Preliminary group comparisons indicate worse word list learning, short- and long-delayed verbal recall, and verbal recognition in both patient cohorts compared with those of the healthy control group, adjusted for age and sex. Initial volumetric comparisons show smaller grey matter, frontal, and temporal brain volumes in both patient groups compared with those of healthy controls. These results are quite robust but are neither final nor placed in the needed context intended at study completion. Conclusions To the best of our knowledge, this is the first study to include objective and comprehensive longitudinal analyses of neurocognitive sequelae of COVID-19 in an extreme group comparison stratified by disease severity with healthy controls actively recruited during the pandemic. Results from this study will contribute to the nascent literature on the prolonged effects of COVID-19 on neurocognitive performance via our coassessment of neuroradiological, neurological, pulmonary, psychiatric, and lifestyle factors. Trial Registration International Clinical Trials Registry Platform DRKS00023806; https://trialsearch.who.int/Trial2.aspx?TrialID=DRKS00023806 International Registered Report Identifier (IRRID) DERR1-10.2196/30259
Collapse
Affiliation(s)
- Catherine N Widmann
- Section Neuropsychology, Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical Center, Bonn, Germany.,German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Michelle Wieberneit
- Section Neuropsychology, Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Luzie Bieler
- Section Neuropsychology, Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Sarah Bernsen
- German Center for Neurodegenerative Diseases, Bonn, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Robin Gräfenkämper
- Section Neuropsychology, Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical Center, Bonn, Germany.,Department of Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | | | - Carsten Schmeel
- Department of Neuroradiology, University of Bonn Medical Center, Bonn, Germany
| | - Pawel Tacik
- German Center for Neurodegenerative Diseases, Bonn, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Dirk Skowasch
- Department of Cardiology, Pneumology and Angiology, Internal Medicine II, University of Bonn Medical Center, Bonn, Germany
| | - Alexander Radbruch
- Department of Neuroradiology, University of Bonn Medical Center, Bonn, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases, Bonn, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
12
|
Zhang Z, Wang Z, Ling Z, Li Y, Pan J, Gao Q, Zhang J, Yan L, Zhang Z, Li J, Xiao F. A screened PirB antagonist peptide antagonizes Aβ 42-mediated inhibition of neurite outgrowth in vitro. Appl Microbiol Biotechnol 2021; 105:4649-4662. [PMID: 34059940 DOI: 10.1007/s00253-021-11363-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/07/2021] [Accepted: 05/20/2021] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is a type of progressive neurodegenerative disease, and amyloid β-protein 42 (Aβ42) serves an important role in the pathological process of development of AD. Paired immunoglobulin-like receptor B (PirB) is a functional receptor for myelin inhibitors of neuron regeneration in the CNS, and it has also been identified to function as a high-affinity receptor for Aβ. Here, we used a phage display to identify a specific PirB antagonist peptide 11(PAP11, PFRLQLS), which could reverse Aβ42-induced neurotoxicity and promote neurite outgrowth in vitro. Immunofluorescence analysis showed that PAP11 colocalized with PirB on the membrane of cortical neurons. Horseradish peroxidase-streptavidin-biotin assay further proved that PAP11 directly binds to PirB and the dissociation constant (Kd) was 0.128μM. PAP11 functionally antagonized the neurite outgrowth inhibitory effect induced by Aβ42 in cortical neurons, and the underlying mechanism was associated with a PirB-ROCK2/CRMP2 signaling pathway. The novel PirB antagonist peptide PAP11 may be a promising candidate therapeutic agent for the treatment of AD and other neurodegenerative diseases. KEY POINTS: • PAP11 was the first PirB antagonist peptide screened by phage display technology. • PAP11 could protect neurons by blocking the binding of Aβ42 and PirB. • PAP11 reverse inhibitory effect of neurite outgrowth through ROCK2/CRMP2 pathway.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zijian Wang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Zhipeng Ling
- Department of Microbial and Biochemical Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yu Li
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Junping Pan
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
| | - Qin Gao
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Jichun Zhang
- Department of Physiology, School of medicine, Jinan University, Guangzhou, China
| | - Li Yan
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhidong Zhang
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou, China.
| | - Junliang Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China.
| | - Fei Xiao
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
13
|
Mora S, Martín-González E, Prados-Pardo Á, Flores P, Moreno M. Increased Compulsivity in Adulthood after Early Adolescence Immune Activation: Preclinical Evidence. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:4684. [PMID: 33924858 PMCID: PMC8125663 DOI: 10.3390/ijerph18094684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 12/17/2022]
Abstract
Immune activation during early developmental stages has been proposed as a contributing factor in the pathogenesis of neuropsychiatric conditions such as obsessive-compulsive disorder, attention-deficit/hyperactivity disorder, and autism in both human and animal studies. However, its relationship with the vulnerability to inhibitory control deficit, which is a shared feature among those conditions, remains unclear. The present work studied whether postnatal immune activation during early adolescence, combined with exposure to early-life adverse events, could lead to adult vulnerability to impulsive and/or compulsive behaviors. Male Wistar rats were exposed to lipopolysaccharide (LPS) in early adolescence at postnatal day 26 (PND26). During peripuberal period, half of the animals were exposed to a mild stress protocol. In adulthood, behavioral assessment was performed with the aid of the sustained attentional 5-choice serial reaction time (5-CSRT) task, schedule-induced polydipsia (SIP), and open-field locomotor activity and novelty reactivity. Rats exposed to LPS showed more compulsive responses than their control counterparts on 5-CSRT task, although no differences were observed in SIP or locomotor responses. Our study contributes to the knowledge of the relationship between immune activation and inhibitory control deficit. Future studies should aim to disentangle how, and to what extent, immune activation impacts behavior, and to understand the role of early life mild stress.
Collapse
Affiliation(s)
- Santiago Mora
- Department of Psychology and Health Research Centre, University of Almería, Carretera de Sacramento s/n, 04120 Almería, Spain; (E.M.-G.); (Á.P.-P.); (P.F.)
| | | | | | | | - Margarita Moreno
- Department of Psychology and Health Research Centre, University of Almería, Carretera de Sacramento s/n, 04120 Almería, Spain; (E.M.-G.); (Á.P.-P.); (P.F.)
| |
Collapse
|
14
|
Presynaptic L-Type Ca 2+ Channels Increase Glutamate Release Probability and Excitatory Strength in the Hippocampus during Chronic Neuroinflammation. J Neurosci 2020; 40:6825-6841. [PMID: 32747440 DOI: 10.1523/jneurosci.2981-19.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 06/18/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation is involved in the pathogenesis of several neurologic disorders, including epilepsy. Both changes in the input/output functions of synaptic circuits and cell Ca2+ dysregulation participate in neuroinflammation, but their impact on neuron function in epilepsy is still poorly understood. Lipopolysaccharide (LPS), a toxic byproduct of bacterial lysis, has been extensively used to stimulate inflammatory responses both in vivo and in vitro LPS stimulates Toll-like receptor 4, an important mediator of the brain innate immune response that contributes to neuroinflammation processes. Although we report that Toll-like receptor 4 is expressed in both excitatory and inhibitory mouse hippocampal neurons (both sexes), its chronic stimulation by LPS induces a selective increase in the excitatory synaptic strength, characterized by enhanced synchronous and asynchronous glutamate release mechanisms. This effect is accompanied by a change in short-term plasticity with decreased facilitation, decreased post-tetanic potentiation, and increased depression. Quantal analysis demonstrated that the effects of LPS on excitatory transmission are attributable to an increase in the probability of release associated with an overall increased expression of L-type voltage-gated Ca2+ channels that, at presynaptic terminals, abnormally contributes to evoked glutamate release. Overall, these changes contribute to the excitatory/inhibitory imbalance that scales up neuronal network activity under inflammatory conditions. These results provide new molecular clues for treating hyperexcitability of hippocampal circuits associated with neuroinflammation in epilepsy and other neurologic disorders.SIGNIFICANCE STATEMENT Neuroinflammation is thought to have a pathogenetic role in epilepsy, a disorder characterized by an imbalance between excitation/inhibition. Fine adjustment of network excitability and regulation of synaptic strength are both implicated in the homeostatic maintenance of physiological levels of neuronal activity. Here, we focused on the effects of chronic neuroinflammation induced by lipopolysaccharides on hippocampal glutamatergic and GABAergic synaptic transmission. Our results show that, on chronic stimulation with lipopolysaccharides, glutamatergic, but not GABAergic, neurons exhibit an enhanced synaptic strength and changes in short-term plasticity because of an increased glutamate release that results from an anomalous contribution of L-type Ca2+ channels to neurotransmitter release.
Collapse
|
15
|
Mora S, Martín-González E, Flores P, Moreno M. Neuropsychiatric consequences of childhood group A streptococcal infection: A systematic review of preclinical models. Brain Behav Immun 2020; 86:53-62. [PMID: 30818033 DOI: 10.1016/j.bbi.2019.02.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 12/21/2018] [Accepted: 02/22/2019] [Indexed: 12/14/2022] Open
Abstract
In recent years, clinical studies have shown strong epidemiological evidence of an increased risk of developing neuropsychiatric disorders after childhood exposure to streptococcal infection, including the Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal infection (PANDAS). New preclinical studies on group A streptococcus (GAS) exposure investigate how to disentangle the influences of immune activation to induce long-term neurobehavioral effects associated with neuropsychiatric disorders such as obsessive-compulsive disorder, schizophrenia or autism. The present systematic review collects neurobehavioral evidence regarding the use of GAS exposure in animal models to study the vulnerability to different neuropsychiatric disorders, improving our understanding of its possible causes and consequences, and compares its contribution with other preclinical models of immune activation in a variety of paradigms. Specifically, we reviewed the effects of postnatal GAS exposure, in comparison with post- and prenatal exposure to Lipopolysaccharide (LPS) and Polyinosinic:polycytidylic acid (Poly I:C), on the long-term effects concerning psychomotor, cognition and socioemotional outcomes in rodents. GAS exposure in animal models has revealed different behavioral alterations such as reduced locomotion and motor coordination, a deficit in sensorimotor gating, learning, working memory, altered social behavior, and increased anxiety and stereotyped behavior. Most of the results found are in accordance with other immune activation models -LPS and Poly I:C-, with some discrepancies. The systematic review of the literature supports the preclinical model of GAS exposure as a valid model for studying the neurobehavioral consequences of streptococcal infections. Future studies on streptococcal infection could contribute increasing our knowledge on preventive actions or treatments for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Santiago Mora
- Department of Psychology, University of Almería, Campus de Excelencia Internacional Agroalimentario CeiA3, Almería, Spain
| | - Elena Martín-González
- Department of Psychology, University of Almería, Campus de Excelencia Internacional Agroalimentario CeiA3, Almería, Spain
| | - Pilar Flores
- Department of Psychology, University of Almería, Campus de Excelencia Internacional Agroalimentario CeiA3, Almería, Spain
| | - Margarita Moreno
- Department of Psychology, University of Almería, Campus de Excelencia Internacional Agroalimentario CeiA3, Almería, Spain.
| |
Collapse
|
16
|
Ji MH, Zhang L, Mao MJ, Zhang H, Yang JJ, Qiu LL. Overinhibition mediated by parvalbumin interneurons might contribute to depression-like behavior and working memory impairment induced by lipopolysaccharide challenge. Behav Brain Res 2020; 383:112509. [PMID: 31987933 DOI: 10.1016/j.bbr.2020.112509] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 12/13/2022]
Abstract
Systemic inflammation induces cognitive impairments via unclear mechanisms. Accumulating evidence has demonstrated that a subset of neurons that express parvalbumin (PV) play a critical role in regulation of cognitive and emotional behavior. Thus, the aim of the present study was to test whether disruption of PV interneuron mediates systemic inflammation-induced depression-like behavior and working memory impairment by lipopolysaccharide (LPS) challenge. Here we showed that LPS induces depression-like behavior and working memory impairment, coinciding with increased PV expression, enhanced GABAergic transmission, and impaired long-term potentiation (LTP) in the hippocampus. Notably, systemic administration of NMDA (N-methyl-D-aspartate) receptor (NMDAR) antagonist ketamine was able to interfere with PV expression and reverse depression-like behavior and working memory impairment, which is probably mediated by reversing impaired LTP. In addition, flumazenil, a competitive antagonist acting at the benzodiazepine binding site of the GABAA receptor, also ameliorated these abnormal behaviors. Collectively, our study added growing evidence to the limited studies that overinhibition mediated by PV interneurons might play a critical role in LPS-induced depression-like behavior and working memory impairment.
Collapse
Affiliation(s)
- Mu-Huo Ji
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, China
| | - Ling Zhang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Ming-Jie Mao
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Hui Zhang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Jiao-Jiao Yang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Li-Li Qiu
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
17
|
Jahangiri Z, Gholamnezhad Z, Hosseini M. The effects of exercise on hippocampal inflammatory cytokine levels, brain oxidative stress markers and memory impairments induced by lipopolysaccharide in rats. Metab Brain Dis 2019; 34:1157-1169. [PMID: 30937699 DOI: 10.1007/s11011-019-00410-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/25/2019] [Indexed: 02/02/2023]
Abstract
The exercise effects on behavioral tests, hippocampal and cortical oxidative stress, and hippocampal inflammatory cytokines of lipopolysaccharide (LPS) administered rats were investigated. The rats were divided into four groups (N = 8): (1) control; (2) moderate training (MT, 15 m/min, 30 min/day, 9 weeks); (3) LPS (1 mg/kg LPS) and (4) LPS + MT (1 mg/kg LPS; 15 m/min, 30 min/day, 9 weeks). LPS was injected 2 h before the behavioral experiments during the last week of training. Finally, the rats' brain were removed for biochemical assessments. LPS increased escape latency and traveled distance to reach the platform in Morris water maze (MWM) test (P < 0.05-P < 0.001). In the passive avoidance (PA) test, LPS decreased the latency to enter the dark compartment and the time spent in the light compartment and increased the time spent in the dark compartment (P < 0.01-P < 0.001), while MT improved the rats performances in MWM and PA tests (P < 0.01-P < 0.001). Additionally, LPS increased tumor necrosis factor α (TNF-α), interleukin 1 beta (IL-1β) and C-reactive protein levels in the hippocampal tissues, malondialdehyde (MDA) and nitric oxide metabolite in hippocampal and cortical tissues, and decreased thiol contents and catalase (CAT) and superoxide dismutase (SOD) activity in hippocampal and cortical tissues compared to the control group (P < 0.01-P < 0.001); while moderate training decreased the levels of TNF-α, IL-1β and MDA; increased thiol contents, and SOD and CAT activity in the LPS + MT compared to the LPS group (P < 0.001). These results indicated that moderate training improved LPS-induced learning and memory impairments by attenuating the hippocampal cytokine levels and brain oxidative damage.
Collapse
Affiliation(s)
- Zahra Jahangiri
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IR, 9177948564, Iran
| | - Zahra Gholamnezhad
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, IR, 9177948564, Iran.
| | - Mahmoud Hosseini
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Avdic U, Ahl M, Öberg M, Ekdahl CT. Immune Profile in Blood Following Non-convulsive Epileptic Seizures in Rats. Front Neurol 2019; 10:701. [PMID: 31333561 PMCID: PMC6615316 DOI: 10.3389/fneur.2019.00701] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
Non-convulsive status epilepticus (NCSE) is a prolonged epileptic seizure with subtle symptoms that may delay clinical diagnosis. Emerging experimental evidence shows brain pathology and epilepsy development following NCSE. New diagnostic/prognostic tools are therefore needed for earlier and better stratification of treatment. Here we examined whether NCSE initiates a peripheral immune response in blood serum from rats that experienced electrically-induced NCSE. ELISA analysis showed an acute transient increase in serum protein levels including interleukin-6 6 h post-NCSE, similar to the immune reaction in the brain. At 4 weeks post-NCSE, when 75% of rats subjected to NCSE had also developed spontaneous seizures, several immune proteins were altered. In particular, markers associated with microglia, macrophages and antigen presenting cells, such as CD68, MHCII, and galectin-3, were increased and the T-cell marker CD4 was decreased in serum compared to both non-stimulated controls and NCSE rats without spontaneous seizures, without correlation to interictal epileptiform activity. Analyses of serum following intracerebral injection of lipopolysaccharide (LPS) showed an acute increase in interleukin-6, but at 4 weeks unaltered levels of MHCII and galectin-3, an increase in CD8 and CD11b and a decrease in CD68. None of the increased serum protein levels after NCSE or LPS could be confirmed in spleen tissue. Our data identifies the possibility to detect peripheral changes in serum protein levels following NCSE, which may be related to the development of subsequent spontaneous seizures.
Collapse
Affiliation(s)
- Una Avdic
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Lund University, Lund, Sweden.,Department of Clinical Sciences, Epilepsy Center, Lund University, Lund, Sweden
| | - Matilda Ahl
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Lund University, Lund, Sweden.,Department of Clinical Sciences, Epilepsy Center, Lund University, Lund, Sweden
| | - Maria Öberg
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Lund University, Lund, Sweden.,Department of Clinical Sciences, Epilepsy Center, Lund University, Lund, Sweden
| | - Christine T Ekdahl
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Lund University, Lund, Sweden.,Department of Clinical Sciences, Epilepsy Center, Lund University, Lund, Sweden
| |
Collapse
|
19
|
Wang X, Qi Y, Zhou X, Zhang G, Fu C. Alteration of scaffold: Possible role of MACF1 in Alzheimer's disease pathogenesis. Med Hypotheses 2019; 130:109259. [PMID: 31383338 DOI: 10.1016/j.mehy.2019.109259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/27/2019] [Accepted: 06/04/2019] [Indexed: 02/09/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, with the sign of sensory or motor function loss, memory decline, and dementia. Histopathological study shows AD neuron has irregular cytoskeleton and aberrant synapse. Amyloid-β (Aβ) is believed as the trigger of AD, however, the detailed pathogenesis is not fully elucidated. Microtubule-actin crosslinking factor 1 (MACF1) is a unique giant molecule which can bind to all three types of cytoskeleton fibers, different linkers/adaptors, as well as various functional proteins. MACF1 is a critical scaffold for orchestrating the complex 3D structure, and is essential for correct synaptic function. MACF1's binding ability to microtubule depends on Glycogen synthase kinase 3 Bate (GSK3β) mediated phosphorylation. While GSK3β can be regulated by the binding of Aβ and the receptor Paired immunoglobulin-like receptor B (PirB), possibly via Protein phosphatase 2A (PP2A). So based on literature search and logic analysis, we propose a hypothesis: Aβ binds to its receptor PirB, and triggers cytosol PP2A, which might activate GSK3β. GSK3β might further phosphorylates microtubule-binding domain (MTBD) of MACF1, causes the separation of microtubule and MACF1. Thus MACF1 might lose the control of the whole cytoskeleton system, synapse might change and AD might develop. That is Aβ-PirB-PP2A-GSK3β-MACF1 axis might give rise to AD. We hope our hypothesis might provide new clue and evidence to AD pathogenesis.
Collapse
Affiliation(s)
- Xiaolong Wang
- School of Basic Medical Sciences & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an 710021, China.
| | - Yangyang Qi
- School of Clinical Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Xin Zhou
- School of Clinical Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Geyang Zhang
- School of Clinical Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Caiyu Fu
- School of Clinical Medicine, Xi'an Medical University, Xi'an 710021, China
| |
Collapse
|
20
|
Sheppard O, Coleman MP, Durrant CS. Lipopolysaccharide-induced neuroinflammation induces presynaptic disruption through a direct action on brain tissue involving microglia-derived interleukin 1 beta. J Neuroinflammation 2019; 16:106. [PMID: 31103036 PMCID: PMC6525970 DOI: 10.1186/s12974-019-1490-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/29/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Systemic inflammation has been linked to synapse loss and cognitive decline in human patients and animal models. A role for microglial release of pro-inflammatory cytokines has been proposed based on in vivo and primary culture studies. However, mechanisms are hard to study in vivo as specific microglial ablation is challenging and the extracellular fluid cannot be sampled without invasive methods. Primary cultures have different limitations as the intricate multicellular architecture in the brain is not fully reproduced. It is essential to confirm proposed brain-specific mechanisms of inflammatory synapse loss directly in brain tissue. Organotypic hippocampal slice cultures (OHSCs) retain much of the in vivo neuronal architecture, synaptic connections and diversity of cell types whilst providing convenient access to manipulate and sample the culture medium and observe cellular reactions. METHODS OHSCs were generated from P6-P9 C57BL/6 mice. Inflammation was induced via addition of lipopolysaccharide (LPS), and cultures were analysed for changes in synaptic proteins, gene expression and protein secretion. Microglia were selectively depleted using clodronate, and the effect of IL1β was assessed using a specific neutralising monoclonal antibody. RESULTS LPS treatment induced loss of the presynaptic protein synaptophysin without altering PSD95 or Aβ protein levels. Depletion of microglia prior to LPS application prevented the loss of synaptophysin, whilst microglia depletion after the inflammatory insult was partially effective, although less so than pre-emptive treatment, indicating a time-critical window in which microglia can induce synaptic damage. IL1β protein and mRNA were increased after LPS addition, with these effects also prevented by microglia depletion. Direct application of IL1β to OHSCs resulted in synaptophysin loss whilst pre-treatment with IL1β neutralising antibody prior to LPS addition prevented a significant loss of synaptophysin but may also impact basal synaptic levels. CONCLUSIONS The loss of synaptophysin in this system confirms LPS can act directly within brain tissue to disrupt synapses, and we show that microglia are the relevant cellular target when all major CNS cell types are present. By overcoming limitations of primary culture and in vivo work, our study strengthens the evidence for a key role of microglia-derived IL1β in synaptic dysfunction after inflammatory insult.
Collapse
Affiliation(s)
- Olivia Sheppard
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Michael P Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK.,Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Claire S Durrant
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK. .,Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
21
|
Tyebji S, Seizova S, Hannan AJ, Tonkin CJ. Toxoplasmosis: A pathway to neuropsychiatric disorders. Neurosci Biobehav Rev 2018; 96:72-92. [PMID: 30476506 DOI: 10.1016/j.neubiorev.2018.11.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/23/2018] [Accepted: 11/22/2018] [Indexed: 12/24/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite that resides, in a latent form, in the human central nervous system. Infection with Toxoplasma drastically alters the behaviour of rodents and is associated with the incidence of specific neuropsychiatric conditions in humans. But the question remains: how does this pervasive human pathogen alter behaviour of the mammalian host? This fundamental question is receiving increasing attention as it has far reaching public health implications for a parasite that is very common in human populations. Our current understanding centres on neuronal changes that are elicited directly by this intracellular parasite versus indirect changes that occur due to activation of the immune system within the CNS, or a combination of both. In this review, we explore the interactions between Toxoplasma and its host, the proposed mechanisms and consequences on neuronal function and mental health, and discuss Toxoplasma infection as a public health issue.
Collapse
Affiliation(s)
- Shiraz Tyebji
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, 3052, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, Victoria, Australia.
| | - Simona Seizova
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, 3052, Australia.
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, Victoria, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, 3052, Victoria, Australia.
| | - Christopher J Tonkin
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, 3052, Australia.
| |
Collapse
|
22
|
Zhang F, Zhang JG, Yang W, Xu P, Xiao YL, Zhang HT. 6-Gingerol attenuates LPS-induced neuroinflammation and cognitive impairment partially via suppressing astrocyte overactivation. Biomed Pharmacother 2018; 107:1523-1529. [PMID: 30257370 DOI: 10.1016/j.biopha.2018.08.136] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/24/2018] [Accepted: 08/25/2018] [Indexed: 12/22/2022] Open
Abstract
6-Gingerol, the major component of gingerols extracted from Zingiber officinale, has been shown to exhibit anti-inflammatory and antioxidant bioactivities. Since neuroinflammation plays an important role in neurodegenerative diseases, such as Alzheimer's disease (AD), and astrocytes have been considered important in the process of neurodegeneration, it was of interest to know whether 6-gingerol reduced astrocytes activation or even attenuated cognitive impairment. Here we examined the neuroprotective effects of 6-gingerol in lipopolysaccharide (LPS)-induced disorder models both in vitro and in vivo. C6 astroglioma cells treated with LPS were found to release excessive pro-inflammatory cytokines, including TNF-α and IL-6, and also increase intercellular ROS, NO, and iNOS (i.e. NOS2). All these were blocked by 6-gingerol in a concentration-dependent manner. The spatial learning and memory of rats challenged with LPS (10 μg, i.c.v.) in the absence or presence of 6-gingerol were evaluated using the Morris water-maze (MWM) test. 6-Gingerol attenuated LPS-induced imapirement of MWM learning and memory in a dose-dependent manner. Besides, 6-gingerol inhibited LPS-induced increases in levels of GFAP and TNF-α in the rat brain. The results suggest that 6-gingerol suppresses astrocyte overactivation, through which it contributes to improvement of cognitive ability.
Collapse
Affiliation(s)
- Feng Zhang
- Institute of Pharmacology, Taishan Medical University, Taian, Shandong, 271016, China
| | - Ji-Guo Zhang
- Institute of Pharmacology, Taishan Medical University, Taian, Shandong, 271016, China
| | - Wei Yang
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Pu Xu
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yu-Liang Xiao
- Institute of Pharmacology, Taishan Medical University, Taian, Shandong, 271016, China
| | - Han-Ting Zhang
- Institute of Pharmacology, Taishan Medical University, Taian, Shandong, 271016, China; Departments of Behavioral Medicine & Psychiatry, Physiology & Pharmacology, and Neuroscience, The Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA.
| |
Collapse
|
23
|
Anthocyanins Improve Hippocampus-Dependent Memory Function and Prevent Neurodegeneration via JNK/Akt/GSK3β Signaling in LPS-Treated Adult Mice. Mol Neurobiol 2018; 56:671-687. [PMID: 29779175 DOI: 10.1007/s12035-018-1101-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/29/2018] [Indexed: 10/16/2022]
Abstract
Microglia plays a critical role in the brain and protects neuronal cells from toxins. However, over-activation of microglia leads to deleterious effects. Lipopolysaccharide (LPS) has been reported to affect neuronal cells via activation of microglia as well as directly to initiate neuroinflammation. In the present study, we evaluated the anti-inflammatory and anti-oxidative effect of anthocyanins against LPS-induced neurotoxicity in an animal model and in cell cultures. Intraperitoneal injections of LPS (250 μg/kg/day for 1 week) induce ROS production and promote neuroinflammation and neurodegeneration which ultimately leads to memory impairment. However, anthocyanins treatment at a dose of 24 mg/kg/day for 2 weeks (1 week before and 1 week co-treated with LPS) prevented ROS production, inhibited neuroinflammation and neurodegeneration, and improved memory functions in LPS-treated mice. Both histological and immunoblot analysis indicated that anthocyanins reversed the activation of JNK, prevented neuroinflammation by lowering the levels of inflammatory markers (p-NF-kB, TNF-α, and IL-1β), and reduced neuronal apoptosis by reducing the expression of Bax, cytochrome c, cleaved caspase-3, and cleaved PARP-1, while increasing the level of survival proteins p-Akt, p-GSK3β, and anti-apoptotic Bcl-2 protein. Anthocyanins treatment increased the levels of memory-related pre- and post-synaptic proteins and improved the hippocampus-dependent memory in the LPS-treated mice. Overall, this data suggested that consumption of naturally derived anti-oxidant agent such as anthocyanins ameliorated several pathological events in the LPS-treated animal model and we believe that anthocyanins would be a safe therapeutic agent for slowing the inflammation-induced neurodegeneration in the brain against several diseases such as Alzheimer's disease and Parkinson's disease.
Collapse
|
24
|
Norouzi F, Hosseini M, Abareshi A, Beheshti F, Khazaei M, Shafei MN, Soukhtanloo M, Gholamnezhad Z, Anaeigoudari A. Memory enhancing effect of Nigella Sativa hydro-alcoholic extract on lipopolysaccharide-induced memory impairment in rats. Drug Chem Toxicol 2018; 42:270-279. [PMID: 29589766 DOI: 10.1080/01480545.2018.1447578] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In this study, the effects of Nigella Sativa (NS) hydro-alcoholic extract on lipopolysaccharide (LPS)-induced learning and memory impairments, hippocampal cytokine levels, and brain tissues oxidative damage were investigated in rats. The rats were grouped and treated: (1) control (saline), (2) LPS (1 mg/kg i.p.), and (3-5) 100, 200, or 400 mg/kg NS hydro-alcoholic extract 30 min before LPS injection. The treatment was started since 6 days before the behavioral experiments and continued during the behavioral tests (LPS injection 2 h before each behavioral experiment). Finally, the brains were removed for biochemical assessments. In Morris water maze (MWM) test, LPS increased the escape latency and traveled path compared to control group, whereas all doses of NS hydro-alcoholic extract decreased them compared to LPS group. In passive avoidance (PA) test, the latency to enter the dark compartment in LPS group was shorter than control group while in all treated groups it was longer than LPS group. LPS increased tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), malondialdehyde (MDA), and nitric oxide (NO) metabolites, and decreased thiol content, superoxide dismutase (SOD), and catalase (CAT) in the hippocampal tissues compared to control group while NS hydro-alcoholic extract decreased MDA and NO metabolites and increased thiol content, SOD, and CAT compared to LPS group. Findings of the current study indicated that the hydro-alcoholic extract of NS improved the LPS-induced learning and memory impairments induced by LPS in rats by improving hippocampal cytokine levels and brain tissues oxidative damage.
Collapse
Affiliation(s)
- Fatemeh Norouzi
- a Division of Neurocognitive Sciences , Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Mahmoud Hosseini
- a Division of Neurocognitive Sciences , Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Azam Abareshi
- b Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Farimah Beheshti
- c Department of Basic Science and Neuroscience Research Center , Torbat Heydariyeh University of Medical Sciences , Torbat Heydariyeh , Iran
| | - Majid Khazaei
- d Department of Physiology, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Mohammad Naser Shafei
- a Division of Neurocognitive Sciences , Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Mohammad Soukhtanloo
- e Department of Biochemistry, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Zahra Gholamnezhad
- f Neurogenic Inflammation Research Center Mashhad University of Medical Sciences , Mashhad , Iran
| | - Akbar Anaeigoudari
- g Department of Physiology, School of Medicine , Jiroft University of Medical Sciences , Jiroft , Iran
| |
Collapse
|
25
|
Jha SK, Jha NK, Kumar D, Sharma R, Shrivastava A, Ambasta RK, Kumar P. Stress-Induced Synaptic Dysfunction and Neurotransmitter Release in Alzheimer's Disease: Can Neurotransmitters and Neuromodulators be Potential Therapeutic Targets? J Alzheimers Dis 2018; 57:1017-1039. [PMID: 27662312 DOI: 10.3233/jad-160623] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The communication between neurons at synaptic junctions is an intriguing process that monitors the transmission of various electro-chemical signals in the central nervous system. Albeit any aberration in the mechanisms associated with transmission of these signals leads to loss of synaptic contacts in both the neocortex and hippocampus thereby causing insidious cognitive decline and memory dysfunction. Compelling evidence suggests that soluble amyloid-β (Aβ) and hyperphosphorylated tau serve as toxins in the dysfunction of synaptic plasticity and aberrant neurotransmitter (NT) release at synapses consequently causing a cognitive decline in Alzheimer's disease (AD). Further, an imbalance between excitatory and inhibitory neurotransmission systems induced by impaired redox signaling and altered mitochondrial integrity is also amenable for such abnormalities. Defective NT release at the synaptic junction causes several detrimental effects associated with altered activity of synaptic proteins, transcription factors, Ca2+ homeostasis, and other molecules critical for neuronal plasticity. These detrimental effects further disrupt the normal homeostasis of neuronal cells and thereby causing synaptic loss. Moreover, the precise mechanistic role played by impaired NTs and neuromodulators (NMs) and altered redox signaling in synaptic dysfunction remains mysterious, and their possible interlink still needs to be investigated. Therefore, this review elucidates the intricate role played by both defective NTs/NMs and altered redox signaling in synaptopathy. Further, the involvement of numerous pharmacological approaches to compensate neurotransmission imbalance has also been discussed, which may be considered as a potential therapeutic approach in synaptopathy associated with AD.
Collapse
|
26
|
Chaves Filho AJM, Lima CNC, Vasconcelos SMM, de Lucena DF, Maes M, Macedo D. IDO chronic immune activation and tryptophan metabolic pathway: A potential pathophysiological link between depression and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:234-249. [PMID: 28595944 DOI: 10.1016/j.pnpbp.2017.04.035] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/03/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
Obesity and depression are among the most pressing health problems in the contemporary world. Obesity and depression share a bidirectional relationship, whereby each condition increases the risk of the other. By inference, shared pathways may underpin the comorbidity between obesity and depression. Activation of cell-mediated immunity (CMI) is a key factor in the pathophysiology of depression. CMI cytokines, including IFN-γ, TNFα and IL-1β, induce the catabolism of tryptophan (TRY) by stimulating indoleamine 2,3-dioxygenase (IDO) resulting in the synthesis of kynurenine (KYN) and other tryptophan catabolites (TRYCATs). In the CNS, TRYCATs have been related to oxidative damage, inflammation, mitochondrial dysfunction, cytotoxicity, excitotoxicity, neurotoxicity and lowered neuroplasticity. The pathophysiology of obesity is also associated with a state of aberrant inflammation that activates aryl hydrocarbon receptor (AHR), a pathway involved in the detection of intracellular or environmental changes as well as with increases in the production of TRYCATs, being KYN an agonists of AHR. Both AHR and TRYCATS are involved in obesity and related metabolic disorders. These changes in the TRYCAT pathway may contribute to the onset of neuropsychiatric symptoms in obesity. This paper reviews the role of immune activation, IDO stimulation and increased TRYCAT production in the pathophysiology of depression and obesity. Here we suggest that increased synthesis of detrimental TRYCATs is implicated in comorbid obesity and depression and is a new drug target to treat both diseases.
Collapse
Affiliation(s)
- Adriano José Maia Chaves Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Camila Nayane Carvalho Lima
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Silvânia Maria Mendes Vasconcelos
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - David Freitas de Lucena
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Michael Maes
- Impact Strategic Research Center, Deakin University, Geelong, Australia; Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Londrina, Brazil
| | - Danielle Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
27
|
Chronic N-acetylcysteine treatment alleviates acute lipopolysaccharide-induced working memory deficit through upregulating caveolin-1 and synaptophysin in mice. Psychopharmacology (Berl) 2018; 235:179-191. [PMID: 29058042 DOI: 10.1007/s00213-017-4762-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 09/27/2017] [Indexed: 12/30/2022]
Abstract
RATIONALE Working memory (WM) is a dynamic encoding process and an active representation of information over a short time. The ability to guide forthcoming behavior would be disrupted if WM was impaired by various factors including inflammation, stress, free radicals, and disease states such as schizophrenia. However, the mechanism underlying acute working memory impairment remains to be defined. OBJECTIVES In this study, we tested the hypothesis that decreased caveolin-1 (Cav-1) and synaptophysin (SYP) accounted for the WM impairment challenged with acute intraperitoneally lipopolysaccharide (LPS), which mimicked neuroinflammation. Delayed alternation T-maze task (DAT) was used to assess working memory of adult male C57BL/6 mice, and western blot and immunostaining were used to detect protein expression and distribution in medial prefrontal cortex (mPFC) and hippocampus. RESULTS Our results showed that LPS dose-dependently induced working memory deficit accompanied by the decrease of Cav-1 and SYP in mPFC but not hippocampus. In addition, LPS significantly decreased protein level of Cav-1 and SYP in neurons by activating microglia cells. More important, 2-week N-acetylcysteine (NAC) treatment dose-dependently inhibited LPS-induced working memory deficit by improving the ability to use Lose-shift but not Win-shift strategy and significantly inhibited LPS-induced downregulation of Cav-1 and SYP in mPFC. CONCLUSIONS Taken together, our findings demonstrate that chronic NAC treatment alleviates acute LPS-induced working memory deficit through upregulating Cav-1 and SYP in mice.
Collapse
|
28
|
Frost GR, Li YM. The role of astrocytes in amyloid production and Alzheimer's disease. Open Biol 2017; 7:170228. [PMID: 29237809 PMCID: PMC5746550 DOI: 10.1098/rsob.170228] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/16/2017] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is marked by the presence of extracellular amyloid beta (Aβ) plaques, intracellular neurofibrillary tangles (NFTs) and gliosis, activated glial cells, in the brain. It is thought that Aβ plaques trigger NFT formation, neuronal cell death, neuroinflammation and gliosis and, ultimately, cognitive impairment. There are increased numbers of reactive astrocytes in AD, which surround amyloid plaques and secrete proinflammatory factors and can phagocytize and break down Aβ. It was thought that neuronal cells were the major source of Aβ. However, mounting evidence suggests that astrocytes may play an additional role in AD by secreting significant quantities of Aβ and contributing to overall amyloid burden in the brain. Astrocytes are the most numerous cell type in the brain, and therefore even minor quantities of amyloid secretion from individual astrocytes could prove to be substantial when taken across the whole brain. Reactive astrocytes have increased levels of the three necessary components for Aβ production: amyloid precursor protein, β-secretase (BACE1) and γ-secretase. The identification of environmental factors, such as neuroinflammation, that promote astrocytic Aβ production, could redefine how we think about developing therapeutics for AD.
Collapse
Affiliation(s)
- Georgia R Frost
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Programs of Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Programs of Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
- Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| |
Collapse
|
29
|
Lei Y, Chen CJ, Yan XX, Li Z, Deng XH. Early-life lipopolysaccharide exposure potentiates forebrain expression of NLRP3 inflammasome proteins and anxiety-like behavior in adolescent rats. Brain Res 2017; 1671:43-54. [DOI: 10.1016/j.brainres.2017.06.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 06/08/2017] [Accepted: 06/12/2017] [Indexed: 12/14/2022]
|
30
|
Mi YJ, Chen H, Guo N, Sun MY, Zhao ZH, Gao XC, Wang XL, Zhang RS, Zhou JB, Gou XC. Inhibition of PirB Activity by TAT-PEP Improves Mouse Motor Ability and Cognitive Behavior. Front Aging Neurosci 2017; 9:199. [PMID: 28676756 PMCID: PMC5476690 DOI: 10.3389/fnagi.2017.00199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 06/02/2017] [Indexed: 01/07/2023] Open
Abstract
Paired immunoglobulin-like receptor B (PirB), a functional receptor for myelin-associated inhibitory proteins, plays an important role in axon regeneration in injured brains. However, its role in normal brain function with age has not been previously investigated. Therefore in this study, we examined the expression level of PirB in the cerebral cortex, hippocampus and cerebellum of mice at 1 month, 3 months and 18 months of age. The results showed that the expression of PirB increased with age. We further demonstrated that overexpression of PirB inhibited neurite outgrowth in PC12 cells, and this inhibitory activity of PirB could be reversed by TAT-PEP, which is a recombinant soluble PirB ectodomain fused with TAT domain for blood-brain barrier penetration. In vivo study, intraperitoneal administration of TAT-PEP was capable of enhancing motor capacity and spatial learning and memory in mice, which appeared to be mediated through regulation of brain-derived neurotrophic factor (BDNF) secretion. Our study suggests that PirB is associated with aging and TAT-PEP may be a promising therapeutic agent for modulation of age-related motor and cognitive dysfunctions.
Collapse
Affiliation(s)
- Ya-Jing Mi
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Hai Chen
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China.,Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical UniversityXi'an, China
| | - Na Guo
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Meng-Yi Sun
- Department of Neurosurgery, School of Medicine, Yale UniversityNew Haven, CT, United States
| | - Zhao-Hua Zhao
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Xing-Chun Gao
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Xiao-Long Wang
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Rui-San Zhang
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China
| | - Jiang-Bing Zhou
- Department of Neurosurgery, School of Medicine, Yale UniversityNew Haven, CT, United States
| | - Xing-Chun Gou
- Institute of Basic and Translational Medicine, and School of Basic Medical Sciences, and Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical UniversityXi'an, China.,Department of Neurosurgery, School of Medicine, Yale UniversityNew Haven, CT, United States
| |
Collapse
|
31
|
Zhao ZH, Deng B, Xu H, Zhang JF, Mi YJ, Meng XZ, Gou XC, Xu LX. PirB Overexpression Exacerbates Neuronal Apoptosis by Inhibiting TrkB and mTOR Phosphorylation After Oxygen and Glucose Deprivation Injury. Cell Mol Neurobiol 2017; 37:707-715. [PMID: 27443384 DOI: 10.1007/s10571-016-0406-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 07/08/2016] [Indexed: 11/24/2022]
Abstract
Previous studies have proven that paired immunoglobulin-like receptor B (PirB) plays a crucial suppressant role in neurite outgrowth and neuronal plasticity after central nervous system injury. However, the role of PirB in neuronal survival after cerebral ischemic injury and its mechanisms remains unclear. In the present study, the role of PirB is investigated in the survival and apoptosis of cerebral cortical neurons in cultured primary after oxygen and glucose deprivation (OGD)-induced injury. The results have shown that rebarbative PirB exacerbates early neuron apoptosis and survival. PirB gene silencing remarkably decreases early apoptosis and promotes neuronal survival after OGD. The expression of bcl-2 markedly increased and the expression of bax significantly decreased in PirB RNAi-treated neurons, as compared with the control- and control RNAi-treated ones. Further, phosphorylated TrkB and mTOR levels are significantly downregulated in the damaged neurons. However, the PirB silencing markedly upregulates phosphorylated TrkB and mTOR levels in the neurons after the OGD. Taken together, the overexpression of PirB inhibits the neuronal survival through increased neuron apoptosis. Importantly, the inhibition of the phosphorylation of TrkB and mTOR may be one of its mechanisms.
Collapse
Affiliation(s)
- Zhao-Hua Zhao
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Bin Deng
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Xu
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Jun-Feng Zhang
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Ya-Jing Mi
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China
| | - Xiang-Zhong Meng
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xing-Chun Gou
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
- Institute of Basic and Translational Medicine & School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China.
| | - Li-Xian Xu
- Department of Anesthesiology, College of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
32
|
Bombeiro AL, Thomé R, Oliveira Nunes SL, Monteiro Moreira B, Verinaud L, de Oliveira ALR. MHC-I and PirB Upregulation in the Central and Peripheral Nervous System following Sciatic Nerve Injury. PLoS One 2016; 11:e0161463. [PMID: 27551751 PMCID: PMC4995013 DOI: 10.1371/journal.pone.0161463] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 08/05/2016] [Indexed: 12/12/2022] Open
Abstract
Major histocompatibility complex class one (MHC-I) antigen-presenting molecules participate in central nervous system (CNS) synaptic plasticity, as does the paired immunoglobulin-like receptor B (PirB), an MHC-I ligand that can inhibit immune-cells and bind to myelin axon growth inhibitors. Based on the dual roles of both molecules in the immune and nervous systems, we evaluated their expression in the central and peripheral nervous system (PNS) following sciatic nerve injury in mice. Increased PirB and MHC-I protein and gene expression is present in the spinal cord one week after nerve transection, PirB being mostly expressed in the neuropile region. In the crushed nerve, MHC-I protein levels increased 2 weeks after lesion (wal) and progressively decreased over the next eight weeks. The same kinetics were observed for infiltrating cytotoxic T lymphocytes (CTLs) but not for PirB expression, which continuously increased. Both MHC-I and PirB were found in macrophages and Schwann cells but rarely in axons. Interestingly, at 8 wal, PirB was mainly restricted to the myelin sheath. Our findings reinforce the participation of MHC-I and PirB in CNS plasticity events. In contrast, opposing expression levels of these molecules were found in the PNS, so that MHC-I and PirB seem to be mostly implicated in antigen presentation to CTLs and axon myelination, respectively.
Collapse
Affiliation(s)
- André Luis Bombeiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Rodolfo Thomé
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Sérgio Luiz Oliveira Nunes
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Bárbara Monteiro Moreira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Liana Verinaud
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
| | - Alexandre Leite Rodrigues de Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato, 255, CEP: 13083–865, Campinas, SP, Brazil
- * E-mail:
| |
Collapse
|
33
|
Song X, Zhou B, Zhang P, Lei D, Wang Y, Yao G, Hayashi T, Xia M, Tashiro SI, Onodera S, Ikejima T. Protective Effect of Silibinin on Learning and Memory Impairment in LPS-Treated Rats via ROS–BDNF–TrkB Pathway. Neurochem Res 2016; 41:1662-72. [DOI: 10.1007/s11064-016-1881-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 02/26/2016] [Accepted: 02/29/2016] [Indexed: 12/15/2022]
|
34
|
Anaeigoudari A, Shafei MN, Soukhtanloo M, Sadeghnia HR, Reisi P, Nosratabadi R, Behradnia S, Hosseini M. The effects of L-arginine on spatial memory and synaptic plasticity impairments induced by lipopolysaccharide. Adv Biomed Res 2015; 4:202. [PMID: 26601090 PMCID: PMC4620614 DOI: 10.4103/2277-9175.166138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/06/2015] [Indexed: 01/07/2023] Open
Abstract
Background: An important role of nitric oxide (NO) in neuroinflammation has been suggested. It is also suggested that NO has a critical role in learning and memory. Neuro-inflammation induced by lipopolysaccharide (LPS) has been reported that deteriorates learning and memory. The effect of L-arginine (LA) as a precursor of NO on LPS-induced spatial learning and memory and neuronal plasticity impairment was evaluated. Materials and Methods: The animals were grouped into: (1) Control, (2) LPS, (3) LA-LPS, and (4) LA. The rats received intraperitoneally LPS (1 mg/kg) 2 h before experiments and LA (200 mg/kg) 30 min before LPS. The animals were examined in Morris water maze (MWM). Long-term potentiation (LTP) from CA1 area of the hippocampus was also assessed by 100 Hz stimulation in the ipsilateral Schaffer collateral pathway. Results: In MWM, time latency and traveled path were higher in LPS group than the control group (P < 0.001) whereas in LA-LPS group they were shorter than LPS group (P < 0.001). The amplitude and slope of field excitatory postsynaptic potential (fEPSP) decreased in LPS group compared to control group (P < 0.05 and P < 0.01) whereas, there was not any significant difference in these parameters between LPS and LA-LPS groups. Conclusion: Administration of LPS impaired spatial memory and synaptic plasticity. Although LA ameliorated deleterious effects of LPS on learning of spatial tasks, it could not restore LPS-induced LTP impairment.
Collapse
Affiliation(s)
- Akbar Anaeigoudari
- Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Naser Shafei
- Neurocognitive Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Sadeghnia
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Nosratabadi
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sepehr Behradnia
- Neurogenic Inflammation Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Neurogenic Inflammation Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
35
|
Liu J, Wang Y, Fu W. Axon regeneration impediment: the role of paired immunoglobulin-like receptor B. Neural Regen Res 2015; 10:1338-42. [PMID: 26487866 PMCID: PMC4590251 DOI: 10.4103/1673-5374.162771] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Regenerative capacity is weak after central nervous system injury because of the absence of an enhancing microenvironment and presence of an inhibitory microenvironment for neuronal and axonal repair. In addition to the Nogo receptor (NgR), the paired immunoglobulin-like receptor B (PirB) is a recently discovered coreceptor of Nogo, myelin-associated glycoprotein, and myelin oligodendrocyte glycoprotein. Concurrent blocking of NgR and PirB almost completely eliminates the inhibitory effect of myelin-associated inhibitory molecules on axonal regeneration. PirB participates in a key pathological process of the nervous system, specifically axonal regeneration inhibition. PirB is an inhibitory receptor similar to NgR, but their effects are not identical. This study summarizes the structure, distribution, relationship with common nervous system diseases, and known mechanisms of PirB, and concludes that PirB is also distributed in cells of the immune and hematopoietic systems. Further investigations are needed to determine if immunomodulation and blood cell migration involve inhibition of axonal regeneration.
Collapse
Affiliation(s)
- Jing Liu
- Neonatal Intensive Care Center, BAYI Children's Hospital, Beijing Military General Hospital of Chinese PLA, Beijing, China
- Correspondence to: Jing Liu,
| | - Yan Wang
- Neonatal Intensive Care Center, BAYI Children's Hospital, Beijing Military General Hospital of Chinese PLA, Beijing, China
- Graduate School, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wei Fu
- Neonatal Intensive Care Center, BAYI Children's Hospital, Beijing Military General Hospital of Chinese PLA, Beijing, China
- Graduate School, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
36
|
Nazem A, Sankowski R, Bacher M, Al-Abed Y. Rodent models of neuroinflammation for Alzheimer's disease. J Neuroinflammation 2015; 12:74. [PMID: 25890375 PMCID: PMC4404276 DOI: 10.1186/s12974-015-0291-y] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 03/27/2015] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease remains incurable, and the failures of current disease-modifying strategies for Alzheimer's disease could be attributed to a lack of in vivo models that recapitulate the underlying etiology of late-onset Alzheimer's disease. The etiology of late-onset Alzheimer's disease is not based on mutations related to amyloid-β (Aβ) or tau production which are currently the basis of in vivo models of Alzheimer's disease. It has recently been suggested that mechanisms like chronic neuroinflammation may occur prior to amyloid-β and tau pathologies in late-onset Alzheimer's disease. The aim of this study is to analyze the characteristics of rodent models of neuroinflammation in late-onset Alzheimer's disease. Our search criteria were based on characteristics of an idealistic disease model that should recapitulate causes, symptoms, and lesions in a chronological order similar to the actual disease. Therefore, a model based on the inflammation hypothesis of late-onset Alzheimer's disease should include the following features: (i) primary chronic neuroinflammation, (ii) manifestations of memory and cognitive impairment, and (iii) late development of tau and Aβ pathologies. The following models fit the pre-defined criteria: lipopolysaccharide- and PolyI:C-induced models of immune challenge; streptozotocin-, okadaic acid-, and colchicine neurotoxin-induced neuroinflammation models, as well as interleukin-1β, anti-nerve growth factor and p25 transgenic models. Among these models, streptozotocin, PolyI:C-induced, and p25 neuroinflammation models are compatible with the inflammation hypothesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Amir Nazem
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institute for Medical Research, 350 Community drive, Manhasset, NY, 11030, USA.
| | - Roman Sankowski
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institute for Medical Research, 350 Community drive, Manhasset, NY, 11030, USA.
| | - Michael Bacher
- Institute of Immunology, Philipps University Marburg, Hans-Meerwein-Str., 35043, Marburg, Germany.
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, 350 Community drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
37
|
Xu YQ, Sun ZQ, Wang YT, Xiao F, Chen MW. Function of Nogo-A/Nogo-A receptor in Alzheimer's disease. CNS Neurosci Ther 2015; 21:479-85. [PMID: 25732725 DOI: 10.1111/cns.12387] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 01/01/2015] [Accepted: 01/02/2015] [Indexed: 12/11/2022] Open
Abstract
Nogo-A is a protein inhibiting axonal regeneration, which is considered a major obstacle to nerve regeneration after injury in mammals. Rapid progress has been achieved in new physiopathological function of Nogo-A in Alzheimer's disease in the past decade. Recent research shows that through binding to Nogo-A receptor, Nogo-A plays an important role in Alzheimer's disease (AD) pathogenesis. Particularly, Nogo-A/Nogo-A receptors modulate the generation of amyloid β-protein (Aβ), which is thought to be a major cause of AD. This review describes the recent development of Nogo-A, Nogo-A receptor, and downstream signaling involved in AD and pharmacological basis of therapeutic drugs. We concluded the Nogo-A/Nogo-A receptor provide new insight into potential mechanisms and promising therapy strategies in AD.
Collapse
Affiliation(s)
- Ying-Qi Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Zhong-Qing Sun
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Yi-Tao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Fei Xiao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.,Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Mei-Wan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
38
|
Zhang XY, Cao JB, Zhang LM, Li YF, Mi WD. Deferoxamine attenuates lipopolysaccharide-induced neuroinflammation and memory impairment in mice. J Neuroinflammation 2015; 12:20. [PMID: 25644393 PMCID: PMC4323121 DOI: 10.1186/s12974-015-0238-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/06/2015] [Indexed: 12/11/2022] Open
Abstract
Background Neuroinflammation often results in enduring cognitive impairment and is a risk factor for postoperative cognitive dysfunction. There are currently no effective treatments for infection-induced cognitive impairment. Previous studies have shown that the iron chelator deferoxamine (DFO) can increase the resistance of neurons to injury and disease by stimulating adaptive cellular stress responses. However, the impact of DFO on the cognitive sequelae of neuroinflammation is unknown. Methods A mouse model of lipopolysaccharide (LPS)-induced cognitive impairment was established to evaluate the neuroprotective effects of DFO against LPS-induced memory deficits and neuroinflammation. Adult C57BL/6 mice were treated with 0.5 μg of DFO 3 days prior to intracerebroventricular microinjection of 2 μg of LPS. Cognitive function was assessed using a Morris water maze from post-injection days 1 to 3. Animal behavioral tests, as well as pathological and biochemical assays were performed to evaluate the LPS-induced hippocampal damage and the neuroprotective effect of DFO. Results Treatment of mice with LPS resulted in deficits in cognitive performance in the Morris water maze without changing locomotor activity, which were ameliorated by pretreatment with DFO. DFO prevented LPS-induced microglial activation and elevations of IL-1β and TNF-α levels in the hippocampus. Moreover, DFO attenuated elevated expression of caspase-3, modulated GSK3β activity, and prevented LPS-induced increases of MDA and SOD levels in the hippocampus. DFO also significantly blocked LPS-induced iron accumulation and altered expression of proteins related to iron metabolism in the hippocampus. Conclusions Our results suggest that DFO may possess a neuroprotective effect against LPS-induced neuroinflammation and cognitive deficits via mechanisms involving maintenance of less brain iron, prevention of neuroinflammation, and alleviation of oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Xiao-Ying Zhang
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jiang-Bei Cao
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Li-Ming Zhang
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Yun-Feng Li
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Wei-Dong Mi
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
39
|
Deng X, Li M, Ai W, He L, Lu D, Patrylo PR, Cai H, Luo X, Li Z, Yan X. Lipolysaccharide-Induced Neuroinflammation Is Associated with Alzheimer-Like Amyloidogenic Axonal Pathology and Dendritic Degeneration in Rats. ADVANCES IN ALZHEIMER'S DISEASE 2014; 3:78-93. [PMID: 25360394 PMCID: PMC4211261 DOI: 10.4236/aad.2014.32009] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic neuroinflammation is thought to play an etiological role in Alzheimer's disease (AD), which is characterized pathologically by amyloid and tau formation, as well as neuritic dystrophy and synaptic degeneration. The causal relationship between these pathological events is a topic of ongoing research and discussion. Recent data from transgenic AD models point to a tight spatiotemporal link between neuritic and amyloid pathology, with the obligatory enzyme for β-amyloid (Aβ) production, namely β-secretase-1 (BACE1), is overexpressed in axon terminals undergoing dystrophic change. However, the axonal pathology inherent with BACE1 elevation seen in transgenic AD mice may be secondary to increased soluble Aβ in these genetically modified animals. Here we explored the occurrence of the AD-like axonal and dendritic pathology in adult rat brain affected by LPS-induced chronic neuroinflammation. Unilateral intracerebral LPS injection induced prominent inflammatory response in glial cells in the ipsilateral cortex and hippocampal formation. BACE1 protein levels were elevated the ipsilateral hippocampal lysates in the LPS treated animals relative to controls. BACE1 immunoreactive dystrophic axons appeared in the LPS-treated ipsilateral cortex and hippocampal formation, colocalizing with increased β-amyloid precursor protein and Aβ antibody (4G8) immunolabeling. Quantitative Golgi studies revealed reduction of dendritic branching points and spine density on cortical layer III and hippocampal CA3 pyramidal neurons in the LPS-treated ipsilateral cerebrum. These findings suggest that Alzheimer-like amyloidogenic axonal pathology and dendritic degeneration occur in wildtype mammalian brain in partnership with neuroinflammation following LPS injection.
Collapse
Affiliation(s)
- Xiaohua Deng
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Meili Li
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Weiming Ai
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
- Department of Nursing in Internal Medicine, School of Nursing, Xiangtan Vocational and Technical College, Xiangtan, China
| | - Lixin He
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
- Department of Anatomy and Physiology, School of Nursing, Xiangtan Vocational and technical College, Xiangtan, China
| | - Dahua Lu
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Peter R. Patrylo
- Departments of Physiology, Anatomy and Center for Integrated Research in Cognitive and Neural Sciences, Southern Illinois University Carbondale, Carbondale, USA
| | - Huaibin Cai
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, USA
| | - Xuegang Luo
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Zhiyuan Li
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medical Science, Changsha, China
| |
Collapse
|
40
|
Parlog A, Harsan LA, Zagrebelsky M, Weller M, von Elverfeldt D, Mawrin C, Korte M, Dunay IR. Chronic murine toxoplasmosis is defined by subtle changes in neuronal connectivity. Dis Model Mech 2014; 7:459-69. [PMID: 24524910 PMCID: PMC3974456 DOI: 10.1242/dmm.014183] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recent studies correlate chronic Toxoplasma gondii (T. gondii) infection with behavioral changes in rodents; additionally, seropositivity in humans is reported to be associated with behavioral and neuropsychiatric diseases. In this study we investigated whether the described behavioral changes in a murine model of chronic toxoplasmosis are associated with changes in synaptic plasticity and brain neuronal circuitry. In mice chronically infected with T. gondii, magnetic resonance imaging (MRI) data analysis displayed the presence of heterogeneous lesions scattered throughout all brain areas. However, a higher density of lesions was observed within specific regions such as the somatosensory cortex (SSC). Further histopathological examination of these brain areas indicated the presence of activated resident glia and recruited immune cells accompanied by limited alterations of neuronal viability. In vivo diffusion-tensor MRI analysis of neuronal fiber density within the infected regions revealed connectivity abnormalities in the SSC. Altered fiber density was confirmed by morphological analysis of individual, pyramidal and granule neurons, showing a reduction in dendritic arbor and spine density within the SSC, as well as in the hippocampus. Evaluation of synapse efficacy revealed diminished levels of two key synaptic proteins, PSD95 and synaptophysin, within the same brain areas, indicating deficits in functionality of the synaptic neurotransmission in infected mice. Our results demonstrate that persistent T. gondii infection in a murine model results in synaptic deficits within brain structures leading to disturbances in the morphology of noninfected neurons and modified brain connectivity, suggesting a potential explanation for the behavioral and neuropsychiatric alterations.
Collapse
Affiliation(s)
- Alexandru Parlog
- Institute of Medical Microbiology, Otto-von-Guericke University, 39120-Magdeburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Gou Z, Mi Y, Jiang F, Deng B, Yang J, Gou X. PirB is a novel potential therapeutic target for enhancing axonal regeneration and synaptic plasticity following CNS injury in mammals. J Drug Target 2014; 22:365-71. [PMID: 24405091 DOI: 10.3109/1061186x.2013.878939] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A major barrier to axonal regeneration in mammals is the unfavorable extracellular environment that develops following injury to the central nervous system (CNS). In particular, three myelin-associated inhibitory proteins (MAIs) - Nogo, myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMgp) - are known to inhibit axonal regeneration and functional recovery. These MAIs share a common receptor, glycosylphosphatidylinositol-anchored Nogo receptor (NgR). However, paired immunoglobulin-like receptor B (PirB) - which was originally identified as a receptor for class I major histocompatibility complex (MHCI) in the immune system - is also expressed in neurones and plays a similarly inhibitory role in axonal regeneration and synaptic plasticity following CNS injury through its association with MAIs. Importantly, suppression of PirB activity through antibody antagonism or genetic means can partially relieve the inhibition of neurite outgrowth in vitro and in vivo. In this review, we present the molecular features, expression patterns and known signaling pathways of PirB, and we specifically focus on putative roles for PirB in the CNS and its potential as a target of molecular therapies for enhancing axonal regeneration and synaptic plasticity following CNS injury.
Collapse
Affiliation(s)
- Zhaoyu Gou
- College of Life Science, China West Normal University , Nanchong , China and
| | | | | | | | | | | |
Collapse
|
42
|
Zhu B, Wang ZG, Ding J, Liu N, Wang DAM, Ding LC, Yang C. Chronic lipopolysaccharide exposure induces cognitive dysfunction without affecting BDNF expression in the rat hippocampus. Exp Ther Med 2014; 7:750-754. [PMID: 24520281 PMCID: PMC3919865 DOI: 10.3892/etm.2014.1479] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 01/02/2014] [Indexed: 11/12/2022] Open
Abstract
Previous studies have shown that lipopolysaccharide (LPS) has the potential to cause cognitive dysfunction. However, the underlying pathogenesis has yet to be fully elucidated. Increasing attention is being focused on infection in the central nervous system. Therefore, the present study aimed to investigate the behavioral performance of rats receiving intraperitoneal injections of LPS and to determine the expression levels of amyloid-β (Aβ), brain-derived neurotrophic factor (BDNF) and pro-inflammatory cytokines in the hippocampus. In total, 30 male Wistar rats were randomly divided into 3 groups (each n=10): Control and 3 and 7 day LPS administration groups. The rats were intraperitoneally injected with saline or LPS for 3 or 7 days. Following this, rats performed the Morris water maze test, in which the latency to the platform and proportion of time spent in the target quadrant were recorded. Rats were then sacrificed and the hippocampi were harvested for determination of interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), Aβ and BDNF expression levels. LPS administration for 3 and 7 days significantly increased the latency to the platform and decreased the proportion of time spent in the target quadrant compared with those in the control group, (P<0.05). Administration of LPS for 3 and 7 days induced statistically significant increases in the expression levels of IL-1β, IL-6 and TNF-α in the hippocampus, compared with those in the control group (P<0.05). Additionally, the administration of LPS for 7 days induced a statistically significant increase in the expression level of Aβ in the hippocampus, compared with that in the control group (P<0.05). However, the administration of LPS did not elicit a statistically significant change in the expression level of BDNF in the hippocampus, compared with that in the control group (P>0.05). The results indicate that LPS induces cognitive dysfunction, which is associated with increased expression levels of pro-inflammatory cytokines and Aβ, but does not affect the expression of BDNF in the hippocampus.
Collapse
Affiliation(s)
- Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Zhi-Gang Wang
- Department of Respiratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Jie Ding
- Department of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Ning Liu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - DA-Ming Wang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Liang-Cai Ding
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Chun Yang
- Department of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
43
|
Lee B, Sur B, Park J, Kim SH, Kwon S, Yeom M, Shim I, Lee H, Hahm DH. Ginsenoside rg3 alleviates lipopolysaccharide-induced learning and memory impairments by anti-inflammatory activity in rats. Biomol Ther (Seoul) 2013; 21:381-90. [PMID: 24244826 PMCID: PMC3825202 DOI: 10.4062/biomolther.2013.053] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 09/12/2013] [Accepted: 09/23/2013] [Indexed: 11/05/2022] Open
Abstract
The purpose of this study was to examine whether ginsenoside Rg3 (GRg3) could improve learning and memory impairments and inflammatory reactions induced by injecting lipopolysaccharide (LPS) into the brains of rats. The effects of GRg3 on proinflammatory mediators in the hippocampus and the underlying mechanisms of these effects were also investigated. Injection of LPS into the lateral ventricle caused chronic inflammation and produced deficits in learning in a memory-impairment animal model. Daily administration of GRg3 (10, 20, and 50 mg/kg, i.p.) for 21 consecutive days markedly improved the LPS-induced learning and memory disabilities demonstrated on the step-through passive avoidance test and Morris water maze test. GRg3 administration significantly decreased expression of pro-inflammatory mediators such as tumor necrosis factor-α, interleukin-1β, and cyclooxygenase-2 in the hippocampus, as assessed by reverse transcription-polymerase chain reaction analysis and immunohistochemistry. Together, these findings suggest that GRg3 significantly attenuated LPS-induced cognitive impairment by inhibiting the expression of pro-inflammatory mediators in the rat brain. These results suggest that GRg3 may be effective for preventing or slowing the development of neurological disorders, including Alzheimer's disease, by improving cognitive and memory functions due to its anti-inflammatory activity in the brain.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chugh D, Nilsson P, Afjei SA, Bakochi A, Ekdahl CT. Brain inflammation induces post-synaptic changes during early synapse formation in adult-born hippocampal neurons. Exp Neurol 2013; 250:176-88. [PMID: 24047952 DOI: 10.1016/j.expneurol.2013.09.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/16/2013] [Accepted: 09/09/2013] [Indexed: 10/26/2022]
Abstract
An inflammatory reaction in the brain is primarily characterized by activation of parenchymal microglial cells. Microglia regulate several aspects of adult neurogenesis, i.e. the continuous production of new neurons in the adult brain. Hippocampal neurogenesis is thought to be important for memory formation, but its role in brain diseases is not clear. We have previously shown that brain inflammation modulates the functional integration of newly formed hippocampal neurons. Here, we explored whether there is a defined time period during synaptic development when new neurons are susceptible to brain inflammation. Newly formed hippocampal neurons, born in an intact environment in the adult mouse brain, were exposed to lipopolysaccharide (LPS)-induced inflammation during either early or late phases of excitatory and inhibitory synaptogenesis. We used intra-hippocampal injections of GFP-retroviral vector (RV-GFP) to label the new neurons and ipsilateral LPS injection at either 1 or 4weeks post-RV-GFP injection. A single intra-hippocampal LPS injection induced an inflammatory response for at least 3weeks, including an acute transient pro-inflammatory cytokine release as well as a sub-acute and sustained change in microglial morphology. The general cytoarchitecture of the hippocampal dentate gyrus, including granule cell layer (GCL) volume, and astrocytic glial fibrillary acidic protein expression was not different compared to vehicle controls, and no Fluoro-Jade-positive cell death was observed. New neurons encountering this inflammatory environment exhibited no changes in their gross morphology. However, when inflammation occurred during early stages of synapse formation, we found a region-specific increase in the number of thin dendritic spines and post-synaptic density-95 (PSD-95) cluster formation on spines, suggesting an enhanced excitatory synaptic connectivity in the newborn neurons. No changes were observed in the expression of N-cadherin, an adhesion molecule primarily associated with excitatory synapses. At the inhibitory synapses, alterations due to inflammation were also evident during early but not later stages of synaptic development. Gephyrin, an inhibitory scaffolding protein, was down-regulated in the somatic region, while the adhesion molecules neuroligin-2 (NL-2) and neurofascin were increased in the somatic region and/or on the dendrites. The GABAA receptor-α2 subunit (GABAAR-α2) was increased, while pre/peri-synaptic GABA clustering remained unaltered. The disproportional changes in post-synaptic adhesion molecules and GABAA receptor compared to scaffolding protein expression at the inhibitory synapses during brain inflammation are likely to cause an imbalance in GABAergic transmission. These changes were specific for the newborn neurons and were not observed when estimating the overall expression of gephyrin, NL-2, and GABAAR-α2 in the hippocampal GCL. The expression of interleukin-1-type 1 receptor (IL-1R1) on preferentially the somatic region of new neurons, often in close apposition to NL-2 clusters, may indicate a direct interaction between brain inflammation and synaptic proteins on newborn neurons. In summary, this study provides evidence that adult-born hippocampal neurons alter their inhibitory and excitatory synaptic integration when encountering an LPS-induced brain inflammation during the initial stages of synapse formation. Changes at this critical developmental period are likely to interfere with the physiological functions of new neurons within the hippocampus.
Collapse
Affiliation(s)
- Deepti Chugh
- Inflammation and Stem Cell Therapy Group, Wallenberg Neuroscience Center, Division of Clinical Neurophysiology, Lund University, SE-221 84 Lund, Sweden; Epilepsy Center, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| | | | | | | | | |
Collapse
|
45
|
Fan H, Guo Y, Liang X, Yuan Y, Qi X, Wang M, Ma J, Zhou H. Hydrogen sulfide protects against amyloid beta-peptide induced neuronal injury via attenuating inflammatory responses in a rat model. J Biomed Res 2013; 27:296-304. [PMID: 23885269 PMCID: PMC3721038 DOI: 10.7555/jbr.27.20120100] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 10/25/2012] [Accepted: 04/24/2013] [Indexed: 11/18/2022] Open
Abstract
Neuroinflammation has been recognized to play a critical role in the pathogenesis of Alzheimer's disease (AD), which is pathologically characterized by the accumulation of senile plaques containing activated microglia and amyloid β-peptides (Aβ). In the present study, we examined the neuroprotective effects of hydrogen sulfide (H2S) on neuroinflammation in rats with Aβ1-40 hippocampal injection. We found that Aβ-induced rats exhibited a disorder of pyramidal cell layer arrangement, and a decrease of mean pyramidal cell number in the CA1 hippocampal region compared with those in sham operated rats. NaHS (a donor of H2S, 5.6 mg/kg/d, i.p.) treatment for 3 weeks rescued neuronal cell death significantly. Moreover, we found that H2S dramatically suppressed the release of TNF-α, IL-1β and IL-6 in the hippocampus. Consistently, both immunohistochemistry and Western blotting assays showed that H2S inhibited the upregulation of COX-2 and the activation of NF-κB in the hippocampus. In conclusion, our data indicate that H2S suppresses neuroinflammation via inhibition of the NF-κB activation pathway in the Aβ-induced rat model and has potential value for AD therapy.
Collapse
Affiliation(s)
- Hao Fan
- Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | | | | | | | | | | | | | | |
Collapse
|