351
|
Sevoflurane attenuates systemic inflammation compared with propofol, but does not modulate neuro-inflammation: A laboratory rat study. Eur J Anaesthesiol 2018; 34:764-775. [PMID: 28759530 DOI: 10.1097/eja.0000000000000668] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Septic encephalopathy is believed to be a result of neuro-inflammation possibly triggered by endotoxins, such as lipopolysaccharides (LPS). Modulation of the immune system is a property of volatile anaesthetics. OBJECTIVE We aimed to investigate the systemic and cerebral inflammatory response in a LPS-induced sepsis model in rats. We compared two different sedation strategies, intravenous propofol and the volatile anaesthetic sevoflurane, with the hypothesis that the latter may attenuate neuro-inflammatory processes. DESIGN Laboratory rat study. SETTING Basic research laboratories at the University Hospital Zurich and University Zurich Irchel between August 2014 and June 2016. PATIENTS A total of 32 adult male Wistar rats. INTERVENTIONS After tracheotomy and mechanical ventilation, the anaesthetised rats were monitored before sepsis was induced by using intravenous LPS or phosphate-buffered saline as control. Rats were sedated with propofol (10 mg kg h) or sevoflurane (2 vol%) continuously for 12 h. MAIN OUTCOME MEASURES Systemic inflammatory markers such as cytokine-induced neutrophil chemo-attractant protein 1, monocyte chemo-tactic protein-1 and IL-6 were determined. The same cytokines were measured in brain tissue. Cellular response in the brain was assessed by defining neutrophil accumulation with myeloperoxidase and also activation of microglia with ionised calcium-binding adaptor molecule-1 and astrocytes with glial fibrillary acidic protein. Finally, brain injury was determined. RESULTS Animals were haemodynamically stable in both sedation groups treated with LPS. Blood cytokine peak values were lower in the sevoflurane-LPS compared with propofol-LPS animals. In brain tissue of LPS animals, chemoattractant protein-1 was the only significantly increased cytokine (P = 0.003), however with no significance between propofol and sevoflurane. After LPS challenge, cerebral accumulation of neutrophils was observed. Microglia activation was pronounced in the hippocampus of animals treated with LPS (P = 0.006). LPS induced prominent astrogliosis (P < 0.001). There was no significant difference in microglia or astrocyte activation or apoptosis in the brain between sevoflurane and propofol. CONCLUSION We have shown that systemic attenuation of inflammation by the volatile anaesthetic sevoflurane did not translate into attenuated neuro-inflammation in this LPS-induced inflammation model. TRIAL REGISTRATION Animal approval No. 134/2014, Veterinäramt Zürich.
Collapse
|
352
|
Ilievski V, Zuchowska PK, Green SJ, Toth PT, Ragozzino ME, Le K, Aljewari HW, O’Brien-Simpson NM, Reynolds EC, Watanabe K. Chronic oral application of a periodontal pathogen results in brain inflammation, neurodegeneration and amyloid beta production in wild type mice. PLoS One 2018; 13:e0204941. [PMID: 30281647 PMCID: PMC6169940 DOI: 10.1371/journal.pone.0204941] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/16/2018] [Indexed: 01/08/2023] Open
Abstract
Background The results from cross sectional and longitudinal studies show that periodontitis is closely associated with cognitive impairment (CI) and Alzhemer’s Disease (AD). Further, studies using animal model of periodontitis and human post-mortem brain tissues from subjects with AD strongly suggest that a gram-negative periodontal pathogen, Porphyromonas gingivalis (Pg) and/or its product gingipain is/are translocated to the brain. However, neuropathology resulting from Pg oral application is not known. In this work, we tested the hypothesis that repeated exposure of wild type C57BL/6 mice to orally administered Pg results in neuroinflammation, neurodegeneration, microgliosis, astrogliosis and formation of intra- and extracellular amyloid plaque and neurofibrillary tangles (NFTs) which are pathognomonic signs of AD. Methods Experimental chronic periodontitis was induced in ten wild type 8-week old C57BL/6 WT mice by repeated oral application (MWF/week) of Pg/gingipain for 22 weeks (experimental group). Another 10 wild type 8-week old C57BL/6 mice received vehicle alone (control group) MWF per week for 22 weeks. Brain tissues were collected and the presence of Pg/gingipain was determined by immunofluorescence (IF) microscopy, confocal microscopy, and quantitative PCR (qPCR). The hippocampi were examined for the signs of neuropathology related to AD: TNFα, IL1β, and IL6 expression (neuroinflammation), NeuN and Fluoro Jade C staining (neurodegeneration) and amyloid beta1-42 (Aβ42) production and phosphorylation of tau protein at Ser396 were assessed by IF and confocal microscopy. Further, gene expression of amyloid precursor protein (APP), beta-site APP cleaving enzyme 1 (BACE1), a disintegrin and metalloproteinase domain-containing protein10 (ADAM10) for α-secretase and presenilin1 (PSEN1) for ɣ-secretase, and NeuN (rbFox3) were determined by RT-qPCR. Microgliosis and astrogliosis were also determined by IF microscopy. Results Pg/gingipain was detected in the hippocampi of mice in the experimental group by immunohistochemistry, confocal microscopy, and qPCR confirming the translocation of orally applied Pg to the brain. Pg/gingipain was localized intra-nuclearly and peri-nuclearly in microglia (Iba1+), astrocytes (GFAP+), neurons (NeuN+) and was evident extracellularly. Significantly greater levels of expression of IL6, TNFα and IL1β were evident in experimental as compared to control group (p<0.01, p<0.00001, p<0.00001 respectively). In addition, microgliosis and astrogliosis were evident in the experimental but not in control group (p <0.01, p<0.0001 respectively). Neurodegeneration was evident in the experimental group based on a fewer number of intact neuronal cells assessed by NeuN positivity and rbFOX3 gene expression, and there was a greater number of degenerating neurons in the hippocampi of experimental mice assessed by Fluoro Jade C positivity. APP and BACE1 gene expression were increased in experimental group compared with control group (p<0.05, p<0.001 respectively). PSEN1 gene expression was higher in experimental than control group but the difference was not statistically significant (p = 0.07). ADAM10 gene expression was significantly decreased in experimental group compared with control group (p<0.01). Extracellular Aβ42 was detected in the parenchyma in the experimental but not in the control group (p< 0.00001). Finally, phospho-Tau (Ser396) protein was detected and NFTs were evident in experimental but not in the control group (p<0.00001). Conclusions This study is the first to show neurodegeneration and the formation of extracellular Aβ42 in young adult WT mice after repeated oral application of Pg. The neuropathological features observed in this study strongly suggest that low grade chronic periodontal pathogen infection can result in the development of neuropathology that is consistent with that of AD.
Collapse
Affiliation(s)
- Vladimir Ilievski
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Paulina K. Zuchowska
- Undergraduate Program, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Stefan J. Green
- Department of Biological Sciences University of Illinois at Chicago, Chicago, Illinois, Unites States of America
| | - Peter T. Toth
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Michael E. Ragozzino
- Department of Psychology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Khuong Le
- Undergraduate Program, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Haider W. Aljewari
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | | | - Eric C. Reynolds
- Melbourne Dental School, University of Melbourne, Melbourne, Victoria, Australia
| | - Keiko Watanabe
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
353
|
Resolving neuroinflammation, the therapeutic potential of the anti-malaria drug family of artemisinin. Pharmacol Res 2018; 136:172-180. [DOI: 10.1016/j.phrs.2018.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/01/2018] [Accepted: 09/04/2018] [Indexed: 12/15/2022]
|
354
|
Lee JY, Joo B, Nam JH, Nam HY, Lee W, Nam Y, Seo Y, Kang HJ, Cho HJ, Jang YP, Kim J, We YM, Koo JW, Hoe HS. An Aqueous Extract of Herbal Medicine ALWPs Enhances Cognitive Performance and Inhibits LPS-Induced Neuroinflammation via FAK/NF-κB Signaling Pathways. Front Aging Neurosci 2018; 10:269. [PMID: 30319390 PMCID: PMC6168635 DOI: 10.3389/fnagi.2018.00269] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that Liuwei Dihuang pills (LWPs) can positively affect learning, memory and neurogenesis. However, the underlying molecular mechanisms are not understood. In the present study, we developed ALWPs, a mixture of Antler and LWPs, and investigated whether ALWPs can affect neuroinflammatory responses. We found that ALWPs (500 mg/ml) inhibited lipopolysaccharide (LPS)-induced proinflammatory cytokine IL-1β mRNA levels in BV2 microglial cells but not primary astrocytes. ALWPs significantly reduced LPS-induced cell-surface levels of TLR4 to alter neuroinflammation. An examination of the molecular mechanisms by which ALWPs regulate the LPS-induced proinflammatory response revealed that ALWPs significantly downregulated LPS-induced levels of FAK phosphorylation, suggesting that ALWPs modulate FAK signaling to alter LPS-induced IL-1β levels. In addition, treatment with ALWPs followed by LPS resulted in decreased levels of the transcription factor NF-κB in the nucleus compared with LPS alone. Moreover, ALWPs significantly suppressed LPS-induced BV2 microglial cell migration. To examine whether ALWPs modulate learning and memory in vivo, wild-type C57BL/6J mice were orally administered ALWPs (200 mg/kg) or PBS daily for 3 days, intraperitoneally injected (i.p.) with LPS (250 μg/kg) or PBS, and assessed in Y maze and NOR tests. We observed that oral administration of ALWPs to LPS-injected wild-type C57BL/6J mice significantly rescued short- and long-term memory. More importantly, oral administration of ALWPs to LPS-injected wild-type C57BL/6J mice significantly reduced microglial activation in the hippocampus and cortex. Taken together, our results suggest that ALWPs can suppress neuroinflammation-associated cognitive deficits and that ALWPs have potential as a drug for neuroinflammation/neurodegeneration-related diseases, including Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Ju-Young Lee
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Bitna Joo
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, South Korea
| | - Jin Han Nam
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Hye Yeon Nam
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Wonil Lee
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Youngpyo Nam
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Yongtaek Seo
- Division of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, South Korea
| | - Hye-Jin Kang
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Hyun-Ji Cho
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Young Pyo Jang
- Division of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, South Korea
| | - Jeongyeon Kim
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Young-Man We
- College of Korean Medicine, Wonkwang University, Iksan, South Korea
- Oriental Medical Clinic Center, Hyoo Medical Clinic, Seoul, South Korea
| | - Ja Wook Koo
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, South Korea
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| |
Collapse
|
355
|
Blood-derived plasminogen drives brain inflammation and plaque deposition in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2018; 115:E9687-E9696. [PMID: 30254165 DOI: 10.1073/pnas.1811172115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Two of the most predominant features of the Alzheimer's disease (AD) brain are deposition of β-amyloid (Aβ) plaques and inflammation. The mechanism behind these pathologies remains unknown, but there is evidence to suggest that inflammation may predate the deposition of Aβ. Furthermore, immune activation is increasingly being recognized as a major contributor to the pathogenesis of the disease, and disorders involving systemic inflammation, such as infection, aging, obesity, atherosclerosis, diabetes, and depression are risk factors for the development of AD. Plasminogen (PLG) is primarily a blood protein synthesized in the liver, which when cleaved into its active form, plasmin (PL), plays roles in fibrinolysis, wound healing, cell signaling, and inflammatory regulation. Here we show that PL in the blood is a regulator of brain inflammatory action and AD pathology. Depletion of PLG in the plasma of an AD mouse model through antisense oligonucleotide technology dramatically improved AD pathology and decreased glial cell activation in the brain, whereas an increase in PL activity through α-2-antiplasmin (A2AP) antisense oligonucleotide treatment exacerbated the brain's immune response and plaque deposition. These studies suggest a crucial role for peripheral PL in mediating neuroimmune cell activation and AD progression and could provide a link to systemic inflammatory risk factors that are known to be associated with AD development.
Collapse
|
356
|
Paudel YN, Shaikh MF, Chakraborti A, Kumari Y, Aledo-Serrano Á, Aleksovska K, Alvim MKM, Othman I. HMGB1: A Common Biomarker and Potential Target for TBI, Neuroinflammation, Epilepsy, and Cognitive Dysfunction. Front Neurosci 2018; 12:628. [PMID: 30271319 PMCID: PMC6142787 DOI: 10.3389/fnins.2018.00628] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
High mobility group box protein 1 (HMGB1) is a ubiquitous nuclear protein released by glia and neurons upon inflammasome activation and activates receptor for advanced glycation end products (RAGE) and toll-like receptor (TLR) 4 on the target cells. HMGB1/TLR4 axis is a key initiator of neuroinflammation. In recent days, more attention has been paid to HMGB1 due to its contribution in traumatic brain injury (TBI), neuroinflammatory conditions, epileptogenesis, and cognitive impairments and has emerged as a novel target for those conditions. Nevertheless, HMGB1 has not been portrayed as a common prognostic biomarker for these HMGB1 mediated pathologies. The current review discusses the contribution of HMGB1/TLR4/RAGE signaling in several brain injury, neuroinflammation mediated disorders, epileptogenesis and cognitive dysfunctions and in the light of available evidence, argued the possibilities of HMGB1 as a common viable biomarker of the above mentioned neurological dysfunctions. Furthermore, the review also addresses the result of preclinical studies focused on HMGB1 targeted therapy by the HMGB1 antagonist in several ranges of HMGB1 mediated conditions and noted an encouraging result. These findings suggest HMGB1 as a potential candidate to be a common biomarker of TBI, neuroinflammation, epileptogenesis, and cognitive dysfunctions which can be used for early prediction and progression of those neurological diseases. Future study should explore toward the translational implication of HMGB1 which can open the windows of opportunities for the development of innovative therapeutics that could prevent several associated HMGB1 mediated pathologies discussed herein.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ayanabha Chakraborti
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yatinesh Kumari
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ángel Aledo-Serrano
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Katina Aleksovska
- Medical Faculty, Department of Neurology, "Saints Cyril and Methodius" University, Skopje, Macedonia
| | | | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
357
|
Enhancement of cognitive functions by rice bran extract in a neuroinflammatory mouse model via regulation of PPARγ. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
358
|
Gu SM, Lee HP, Ham YW, Son DJ, Kim HY, Oh KW, Han SB, Yun J, Hong JT. Piperlongumine Improves Lipopolysaccharide-Induced Amyloidogenesis by Suppressing NF-KappaB Pathway. Neuromolecular Med 2018; 20:312-327. [PMID: 29802525 PMCID: PMC6097046 DOI: 10.1007/s12017-018-8495-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/19/2018] [Indexed: 01/02/2023]
Abstract
Amyloidogenesis is known to cause Alzheimer's disease. Our previous studies have found that lipopolysaccharide (LPS) causes neuroinflammation and amyloidogenesis through activation of nuclear factor kappaB (NF-κB). Piperlongumine (PL) is an alkaloid amide found naturally in long pepper (Piper longum) isolates; it was reported to have inhibitory effects on NF-κB activity. We therefore investigated whether PL exhibits anti-inflammatory and anti-amyloidogenic effects by inhibiting NF-κB. A murine model of LPS-induced memory impairment was made via the intraperitoneal (i.p.) injection of LPS (0.25 mg/kg/day, i.p.). We then injected PL (1.5 or 3.0 mg/kg/day, i.p.) for 7 days in three groups of mice to observe effects on memory. We also conducted an in vitro study with astrocytes and microglial BV-2 cells, which were treated with LPS (1 µg/mL) or PL (0.5 or 1.0 or 2.5 µM). Results from our behavioral tests showed that PL inhibited LPS-induced memory. PL also prevented LPS-induced beta-amyloid (Aβ) accumulation and inhibited the activities of β- and γ-secretases. The expression of inflammatory proteins also was decreased in PL-treated mice, cultured BV-2, and primary astrocyte cells. These effects were associated with the inhibition of NF-κB activity. A docking model analysis and pull-down assay showed that PL binds to p50. Taken together, our findings suggest that PL diminishes LPS-induced amyloidogenesis and neuroinflammation by inhibiting NF-κB signaling; PL therefore demonstrates potential for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Sun Mi Gu
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Young Wan Ham
- Department of Chemistry, Utah Valley University, 800W University Pkwy, Orem, UT, 84058, USA
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Hoi Yeong Kim
- Department of Food Science and Technology, Korea National University of Transportation, 61 Daehak-ro, Jeungpyeong-eup, Jeungpyeong-gun, Chungbuk, 27909, Republic of Korea
| | - Ki Wan Oh
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Jaesuk Yun
- Department of Neuroimmunology, College of Pharmacy, Wonkwang University, 460 Iksan-daero, Iksan-si, Jeonbuk, 54538, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
359
|
McCluskey GE, Yates P, Villemagne VL, Rowe C, Szoeke CEI. Self-reported confusion is related to global and regional β-amyloid: data from the Women's healthy ageing project. Brain Imaging Behav 2018; 12:78-86. [PMID: 28108945 DOI: 10.1007/s11682-016-9668-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Disease-modifying treatments for Alzheimer's disease (AD) may require implementation during early stages of β-amyloid accumulation, well before patients have objective cognitive decline. In this study we aimed to assess the clinical value of subjective cognitive impairment (SCI) by examining the cross-sectional relationship between β-amyloid load and SCI. Cerebral β-amyloid and SCI was assessed in a cohort of 112 cognitively normal subjects. Subjective cognition was evaluated using specific questions on memory and cognition and the MAC-Q. Participants had cerebral β-amyloid load measured with 18F-Florbetaben Positron Emission Tomography (PET). No associations were found between measures of subjective memory impairment and cerebral β-amyloid. However, by self-reported confusion was predictive of a higher global β-amyloid burden (p = 0.002), after controlling for confounders. Regional analysis revealed significant associations of confusion with β-amyloid in the prefrontal region (p = 0.004), posterior cingulate and precuneus cortices (p = 0.004) and the lateral temporal lobes (p = 0.001) after controlling for confounders. An in vivo biomarker for AD pathology was associated with SCI by self-reported confusion on cross-sectional analysis. Whilst there has been a large body of research on SMC, our results indicate more research is needed to explore symptoms of confusion.
Collapse
Affiliation(s)
- Georgia E McCluskey
- Centre for Medical Research, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia.,Department of Medicine, University of Melbourne, Parkville, VIC, 3050, Australia
| | - Paul Yates
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, 3084, Australia
| | - Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, 3084, Australia
| | - Christopher Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, 3084, Australia.,Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3050, Australia
| | - Cassandra E I Szoeke
- Centre for Medical Research, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia. .,Department of Medicine, University of Melbourne, Parkville, VIC, 3050, Australia. .,Institute for Health and Ageing, Melbourne, 3000, Australia.
| |
Collapse
|
360
|
Pretorius E, Bester J, Page MJ, Kell DB. The Potential of LPS-Binding Protein to Reverse Amyloid Formation in Plasma Fibrin of Individuals With Alzheimer-Type Dementia. Front Aging Neurosci 2018; 10:257. [PMID: 30186156 PMCID: PMC6113936 DOI: 10.3389/fnagi.2018.00257] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 08/03/2018] [Indexed: 12/28/2022] Open
Abstract
Many studies indicate that there is a (mainly dormant) microbial component in the progressive development of Alzheimer-type dementias (ADs); and that in the case of Gram-negative organisms, a chief culprit might be the shedding of the highly inflammagenic lipopolysaccharide (LPS) from their cell walls. We have recently shown that a highly sensitive assay for the presence of free LPS [added to platelet poor plasma (PPP)] lies in its ability (in healthy individuals) to induce blood to clot into an amyloid form. This may be observed in a SEM or in a confocal microscope when suitable amyloid stains (such as thioflavin T) are added. This process could be inhibited by human lipopolysaccharide-binding protein (LBP). In the current paper, we show using scanning electron microscopy and confocal microscopy with amyloid markers, that PPP taken from individuals with AD exhibits considerable amyloid structure when clotting is initiated with thrombin but without added LPS. Furthermore, we could show that this amyloid structure may be reversed by the addition of very small amounts of LBP. This provides further evidence for a role of microbes and their inflammagenic cell wall products and that these products may be involved in pathological clotting in individuals with AD.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Janette Bester
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,School of Chemistry, The University of Manchester, Manchester, United Kingdom.,The Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
361
|
Effect of Edible Bird's Nest Extract on Lipopolysaccharide-Induced Impairment of Learning and Memory in Wistar Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:9318789. [PMID: 30186358 PMCID: PMC6112221 DOI: 10.1155/2018/9318789] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/20/2018] [Accepted: 07/30/2018] [Indexed: 01/09/2023]
Abstract
Cognitive disability is a common feature associated with a variety of neurological conditions including Alzheimer's Disease (AD), Parkinson's Disease (PD), brain injury, and stroke. Emerging evidence has demonstrated that neuroinflammation plays an important role in the development of cognitive impairment. Current available therapies are relatively ineffective in treating or preventing cognitive disabilities, thus representing an important, unfulfilled medical need. Hence, developing potential treatment is one of the major areas of research interest. Edible bird's nests (EBN) are nests formed by swiftlet's saliva containing sialic acid, which is believed to improve brain function. This present study was embarked upon to evaluate the learning and memory enhancing potential effect of EBN by using Morris water maze test in a Wistar rat model of LPS-induced neuroinflammation. LPS elicited cognitive impairment in the rats by significantly increasing the escape latency while decreasing the number of entries in the probe trial, which are coupled with increased production of proinflammatory cytokines (TNF-α, IL-1β, and IL-6) and oxidative markers (ROS and TBARS) in the hippocampus. Treatment with EBN (125 mg/kg, 250 mg/kg, and 500 mg/kg; p.o.) effectively reversed the effect of LPS on escape latency and probe trial and, in addition, inhibited the LPS-induced upregulation of proinflammatory cytokines and oxidative markers. These findings are suggestive that there is existence of neuroprotective effect contained inside the edible bird's nest.
Collapse
|
362
|
Bhat SA, Sood A, Shukla R, Hanif K. AT2R Activation Prevents Microglia Pro-inflammatory Activation in a NOX-Dependent Manner: Inhibition of PKC Activation and p47phox Phosphorylation by PP2A. Mol Neurobiol 2018; 56:3005-3023. [DOI: 10.1007/s12035-018-1272-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 07/19/2018] [Indexed: 12/22/2022]
|
363
|
Swanson A, Wolf T, Sitzmann A, Willette AA. Neuroinflammation in Alzheimer's disease: Pleiotropic roles for cytokines and neuronal pentraxins. Behav Brain Res 2018; 347:49-56. [PMID: 29462653 PMCID: PMC5988985 DOI: 10.1016/j.bbr.2018.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/30/2018] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is a potential factor speculated to underlie Alzheimer's disease (AD) etiopathogenesis and progression. The overwhelming focus in this area of research to date has been on the chronic upregulation of pro-inflammatory cytokines to understand how neuroinflammatory mechanisms contribute to neurodegeneration. Yet, it is important to understand the pleiotropic roles of these cytokines in modulating neuroinflammation in which they cannot be labeled as a strictly "good" or "bad" biomarker phenotype. As such, biomarkers with more precise functions are needed to better understand how neuroinflammation impacts the brain in AD. Neuronal pentraxins are a concentration- dependent group of pro- or anti- inflammatory cytokines. There is contradictory evidence of these pentraxins as being both neuroprotective and potentially detrimental in AD. Potential neuroprotective examples include their ability to predict AD-related outcomes such as cognition, memory function and synaptic refinement. This review will briefly outline the basis of AD and subsequently summarize findings for neuropathological mechanisms of neuroinflammation, roles for traditional pro-and anti-inflammatory cytokines, and data found thus far on the neuronal pentraxins.
Collapse
Affiliation(s)
- Ashley Swanson
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Tovah Wolf
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Alli Sitzmann
- Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States.
| | - Auriel A Willette
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States; Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States; Department of Biomedical Sciences, Iowa State University, 2008 Veterinary Medicine, Ames, IA 50011, United States; Department of Neurology, University of Iowa, 2007 Roy Carver Pavilion, 200 Hawkins Drive, Iowa City, IA 52242, United States.
| |
Collapse
|
364
|
He W, Tian X, Lv M, Wang H. Liraglutide Protects Neurite Outgrowth of Cortical Neurons Under Oxidative Stress though Activating the Wnt Pathway. J Stroke Cerebrovasc Dis 2018; 27:2696-2702. [PMID: 30042033 DOI: 10.1016/j.jstrokecerebrovasdis.2018.05.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/20/2018] [Accepted: 05/25/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Neurogenesis including neurite outgrowth is important for brain plasticity under physiological conditions and in brain repair after injury. Liraglutide has been found to have neuroprotective action in the risk of central nervous system disease. However, the effect and the potential mechanism of liraglutide-induced neurite outgrowth in primary cortical neurons under oxidative stress remain poorly documented. METHODS In the text, H2O2 was used to mimic ischemia injury in primary cortical neurons. The viability and apoptosis of cell was assessed by Cell Counting Kit-8 and Hoechst 33342. Immunofluorescence method was used to examine the effect of liraglutide on neurite outgrowth in cortical neuron under H2O2 condition. Then, the potential mechanisms involving the Wnt pathway were investigated. The expression of β-catenin, c-myc, and cyclin D1 was determined using quantitative real-time polymerase chain reaction and Western blot. RESULTS Liraglutide significantly increased the viability and alleviated the apoptosis rate of cortical neurons induced by H2O2. Next, liraglutide promoted neurite outgrowth, which could be partially inhibited by the Wnt pathway inhibitor Xav939. Besides, liraglutide induced an increase of β-catenin, c-myc, and cyclin D1 levels, which could also be blocked in the presence of Xav939. CONCLUSIONS These results illustrate that liraglutide exerts neurotrophin-like activity in cortical neurons under oxidative stress condition, partly through activating the Wnt pathway.
Collapse
Affiliation(s)
- Weiliang He
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, PR China
| | - Xiaochao Tian
- Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Mimi Lv
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, PR China
| | - Hebo Wang
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, PR China.
| |
Collapse
|
365
|
Rizzi C, Tiberi A, Giustizieri M, Marrone MC, Gobbo F, Carucci NM, Meli G, Arisi I, D'Onofrio M, Marinelli S, Capsoni S, Cattaneo A. NGF steers microglia toward a neuroprotective phenotype. Glia 2018; 66:1395-1416. [PMID: 29473218 PMCID: PMC6001573 DOI: 10.1002/glia.23312] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 01/22/2018] [Accepted: 01/31/2018] [Indexed: 12/21/2022]
Abstract
Microglia are the sentinels of the brain but a clear understanding of the factors that modulate their activation in physiological and pathological conditions is still lacking. Here we demonstrate that Nerve Growth Factor (NGF) acts on microglia by steering them toward a neuroprotective and anti-inflammatory phenotype. We show that microglial cells express functional NGF receptors in vitro and ex vivo. Our transcriptomic analysis reveals how, in primary microglia, NGF treatment leads to a modulation of motility, phagocytosis and degradation pathways. At the functional level, NGF induces an increase in membrane dynamics and macropinocytosis and, in vivo, it activates an outward rectifying current that appears to modulate glutamatergic neurotransmission in nearby neurons. Since microglia are supposed to be a major player in Aβ peptide clearance in the brain, we tested the effects of NGF on its phagocytosis. NGF was shown to promote TrkA-mediated engulfment of Aβ by microglia, and to enhance its degradation. Additionally, the proinflammatory activation induced by Aβ treatment is counteracted by the concomitant administration of NGF. Moreover, by acting specifically on microglia, NGF protects neurons from the Aβ-induced loss of dendritic spines and inhibition of long term potentiation. Finally, in an ex-vivo setup of acute brain slices, we observed a similar increase in Aβ engulfment by microglial cells under the influence of NGF. Our work substantiates a role for NGF in the regulation of microglial homeostatic activities and points toward this neurotrophin as a neuroprotective agent in Aβ accumulation pathologies, via its anti-inflammatory activity on microglia.
Collapse
Affiliation(s)
- Caterina Rizzi
- Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7Pisa56126Italy
| | - Alexia Tiberi
- Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7Pisa56126Italy
| | - Michela Giustizieri
- European Brain Research Institute‐Fondazione Rita Levi Montalcini, Viale Regina Elena 295Roma00161, Italy
| | - Maria Cristina Marrone
- European Brain Research Institute‐Fondazione Rita Levi Montalcini, Viale Regina Elena 295Roma00161, Italy
| | - Francesco Gobbo
- Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7Pisa56126Italy
| | - Nicola Maria Carucci
- Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7Pisa56126Italy
| | - Giovanni Meli
- European Brain Research Institute‐Fondazione Rita Levi Montalcini, Viale Regina Elena 295Roma00161, Italy
| | - Ivan Arisi
- European Brain Research Institute‐Fondazione Rita Levi Montalcini, Viale Regina Elena 295Roma00161, Italy
| | - Mara D'Onofrio
- European Brain Research Institute‐Fondazione Rita Levi Montalcini, Viale Regina Elena 295Roma00161, Italy
| | - Silvia Marinelli
- European Brain Research Institute‐Fondazione Rita Levi Montalcini, Viale Regina Elena 295Roma00161, Italy
| | - Simona Capsoni
- Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7Pisa56126Italy
- Section of Human Physiology, Department of Biomedical and Specialty Surgical SciencesUniversity of Ferrara, Via Fossato di Mortara 17‐19Ferrara44121Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7Pisa56126Italy
- European Brain Research Institute‐Fondazione Rita Levi Montalcini, Viale Regina Elena 295Roma00161, Italy
| |
Collapse
|
366
|
Chen T, Tian P, Huang Z, Zhao X, Wang H, Xia C, Wang L, Wei H. Engineered commensal bacteria prevent systemic inflammation-induced memory impairment and amyloidogenesis via producing GLP-1. Appl Microbiol Biotechnol 2018; 102:7565-7575. [PMID: 29955935 DOI: 10.1007/s00253-018-9155-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/30/2018] [Accepted: 06/04/2018] [Indexed: 12/31/2022]
Abstract
The anti-obesity drug GLP-1 has been proven to have an impact on central nervous system, while its extremely short half-life greatly limited its use. In this study, our group constructed two engineering strains MG1363-pMG36e-GLP-1 and VNP20009-pLIVE-GLP-1 to continuously express GLP-1, and supplementation of these strains, especially MG1363-pMG36e-GLP-1, had significantly restored the spatial learning and memory impairment of mice caused by LPS (p < 0.05), suppressed glia activation and Aβ accumulation, and downregulated inflammatory expressions of COX-2, TLR-4, TNF-a, and IL-1β. In addition, MG1363-pMG36e-GLP-1 had significantly blocked the translocation of NF-κB signal and inhibited the phosphorylation of redox-sensitive cytoplasmic signalings of MAPKs and PI3K/AKT. These data suggest that MG1363-pMG36e-GLP-1 could be used as a safe and effective nonabsorbed oral treatment for neuroinflammation-related diseases such as Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Tingtao Chen
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China.,Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China
| | - Puyuan Tian
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China.,Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China
| | - Zhixiang Huang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China
| | - Xiaoxiao Zhao
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China
| | - Huan Wang
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China
| | - Chaofei Xia
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China
| | - Le Wang
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China
| | - Hua Wei
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China. .,State Key Laboratory of Food Science and Technology, Nanchang, Jiangxi, 330031, People's Republic of China. .,State Key Laboratory of Food Science and Technology, Nanchang University 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China.
| |
Collapse
|
367
|
Early minor stimulation of microglial TLR2 and TLR4 receptors attenuates Alzheimer's disease-related cognitive deficit in rats: behavioral, molecular, and electrophysiological evidence. Neurobiol Aging 2018; 70:203-216. [PMID: 30031930 DOI: 10.1016/j.neurobiolaging.2018.06.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 05/26/2018] [Accepted: 06/18/2018] [Indexed: 11/20/2022]
Abstract
At early stages of Alzheimer's disease (AD), soluble amyloid beta (Aβ) accumulates in brain while microglia are in resting state. Microglia can recognize Aβ long after formation of plaques and release neurotoxic mediators. We examined impact of early minor activation of microglia by Toll-like receptors (TLRs) 2 and 4 agonists on Alzheimer's disease-related disturbed synaptic function and spatial memory in rats. Microglial BV-2 cells were treated by 0.1, 1, and 10 μg/mL of the TLRs ligands lipopolysaccharide, monophosphoryl lipid A (MPL), and Pam3Cys for 24 hours. Culture medium was then changed with media containing 1-μM Aβ. Tumour necrosis factor (TNF)-α and CCL3 levels were measured in the supernatant, 24 hours thereafter. One μg of TLRs ligands which was able to release low level of TNF-α and CCL3, was administered intracerebroventricularly (i.c.v) to adult male rats every 3 days for 24 days. At the half of the treatment period, Aβ1-42 was infused i.c.v (0.075 μg/hour) for 2 weeks. Finally, the following factors were measured: memory performance by Morris water maze, postsynaptic potentials of dentate gyrus following perforant pathway stimulation, hippocampal inflammatory cytokines interleukin 1 (IL-1)β and TNF-α, anti-inflammatory cytokines IL-10 and TGF-1β, microglia marker arginase 1, Aβ deposits, and the receptor involved in Aβ clearance, formyl peptide receptor 2 (FPR2). TLRs ligands caused dose-dependent release of TNF-α and CCL3 by BV-2 cells. Aβ-treated cells did not release TNF-α and CCL3, whereas those pretreated with MPL and Pam3Cys significantly released these cytokines in response to Aβ. Low-dose TLRs ligands improved the disturbance in spatial and working memory; restored the impaired long-term potentiation induced by Aβ; decreased TNF-α, and Aβ deposits; enhanced TGF-1β, IL-10, and arginase 1 in the hippocampus of Aβ-treated rats; and increased polarization of hippocampal microglia to the anti-inflammatory phenotype. The ligands increased formyl peptide receptor 2 in both BV-2 cells and hippocampus/cortex of Aβ-treated rats. Microglia can sense/clear soluble Aβ by early low-dose MPL and Pam3Cys and safeguard synaptic function and memory in rats.
Collapse
|
368
|
Gypenosides Attenuate Lipopolysaccharide-Induced Neuroinflammation and Memory Impairment in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:4183670. [PMID: 30018656 PMCID: PMC6029442 DOI: 10.1155/2018/4183670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/24/2018] [Indexed: 01/12/2023]
Abstract
Neuroinflammation is deliberated a major factor in various neurodegenerative diseases. Gypenosides (GPS) have pharmacological properties with multiple beneficial effects including anti-inflammatory, antioxidative, and protective properties. In the present study, whether GPS could improve cognitive dysfunction and chronic inflammation caused by injecting lipopolysaccharide (LPS) in the hippocampus was investigated. Effects of GPS on inflammatory factors in the hippocampus and the downstream mechanisms of these effects were also examined. Induction of LPS into the lateral ventricle caused inflammatory reactions and memory impairment on the rats. Every day treatment of GPS (25, 50, and 100 mg/kg) for 21 consecutive days attenuated spatial recognition, discrimination, and memory deficits. GPS treatment significantly decreased proinflammatory mediators such as interleukin-6 (IL-6), interleukin-1β (IL-1β), and nuclear factor-kappaB (NF-κB) levels in the brain. Furthermore, GPS reduced LPS-induced elevated levels of inducible nitric oxide synthase (iNOS) and toll-like receptor 4 (TLR4) mRNA and inhibition of brain-derived neurotrophic factor (BDNF) mRNA level. Collectively, these results showed that GPS may improve cognitive function and provide a potential therapy for memory impairment caused by neuroinflammation. Based on these, GPS may be effective in inhibiting the progress of neurodegenerative diseases by improving memory functions due to its anti-inflammatory activities and appropriate modulation of NF-κB/iNOS/TLR4/BDNF.
Collapse
|
369
|
Deoxyelephantopin ameliorates lipopolysaccharides (LPS)-induced memory impairments in rats: Evidence for its anti-neuroinflammatory properties. Life Sci 2018; 206:45-60. [PMID: 29792878 DOI: 10.1016/j.lfs.2018.05.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/19/2018] [Accepted: 05/20/2018] [Indexed: 02/07/2023]
Abstract
AIM Neuroinflammation is a critical pathogenic mechanism of most neurodegenerative disorders especially, Alzheimer's disease (AD). Lipopolysaccharides (LPS) are known to induce neuroinflammation which is evident from significant upsurge of pro-inflammatory mediators in in vitro BV-2 microglial cells and in vivo animal models. In present study, we investigated anti-neuroinflammatory properties of deoxyelephantopin (DET) isolated from Elephantopus scaber in LPS-induced neuroinflammatory rat model. MATERIALS AND METHODS In this study, DET (0.625. 1.25 and 2.5 mg/kg, i.p.) was administered in rats for 21 days and those animals were challenged with single injection of LPS (250 μg/kg, i.p.) for 7 days. Cognitive and behavioral assessment was carried out for 7 days followed by molecular assessment on brain hippocampus. Statistical significance was analyzed with one-way analysis of variance followed by Dunnett's test to compare the treatment groups with the control group. KEY FINDINGS DET ameliorated LPS-induced neuroinflammation by suppressing major pro-inflammatory mediators such as iNOS and COX-2. Furthermore, DET enhanced the anti-inflammatory cytokines and concomitantly suppressed the pro-inflammatory cytokines and chemokine production. DET treatment also reversed LPS-induced behavioral and memory deficits and attenuated LPS-induced elevation of the expression of AD markers. DET improved synaptic-functionality via enhancing the activity of pre- and post-synaptic markers, like PSD-95 and SYP. DET also prevented LPS-induced apoptotic neurodegeneration via inhibition of PARP-1, caspase-3 and cleaved caspase-3. SIGNIFICANCE Overall, our studies suggest DET can prevent neuroinflammation-associated memory impairment and neurodegeneration and it could be developed as a therapeutic agent for the treatment of neuroinflammation-mediated and neurodegenerative disorders, such as AD.
Collapse
|
370
|
Wang Y, Wang M, Fan K, Li T, Yan T, Wu B, Bi K, Jia Y. Protective effects of Alpinae Oxyphyllae Fructus extracts on lipopolysaccharide-induced animal model of Alzheimer's disease. JOURNAL OF ETHNOPHARMACOLOGY 2018; 217:98-106. [PMID: 29447949 DOI: 10.1016/j.jep.2018.02.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 02/09/2018] [Accepted: 02/10/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alpinae Oxyphyllae Fructus (AOF) with warming and tonifying the kidney and spleen, anti-salivation, anti-polyuria and anti-diarrhea functions is the dried ripe fruits of Alpinia oxyphylla Miq. (Zingiberaceae). As a traditional Chinese medicine, its application history is very long. AIMS OF THE STUDY The purpose of our study is to investigate the effects of different solvent extracts from AOF on lipopolysaccharide (LPS)-induced animal model of Alzheimer's disease (AD) to elucidate the traditional medical theories with modern pharmacological methods and provide a reference for further clarifying its active components and mechanisms. MATERIALS AND METHODS The method of stepwise screening was adopted in this paper. The animals were divided into 9 groups, including control (CT) group, model (MD) group, donepezil (DPZ) group, total extract (TT) group, petroleum ether extract (PE) group, chloroform extract (CF) group, ethyl acetate extract (EA) group, n-butanol extract (NB) group and water extract (WT) group. The anti-amnesic effects of different solvent extracts from AOF were measured in LPS-induced memory deficits mice by Y maze test and Morris water maze (MWM) test. Hematoxylin eosin (HE) staining was applied to observe pathological changes in hippocampus and cerebral cortex tissue of different groups. Biochemical indicators including ionized calcium-binding adaptor molecule 1 (IBA-1), interleukin beta 1 (IL-1β), Aβ1-42 and hyperphosphorylated tau proteins (p-tau) in hippocampus and cortex after treatment with LPS were measured according to the manufacturer's instructions of ELISA kits. HPLC was used to evaluate the major components of different extracts. RESULTS It was found that successive intragastric administration of AOF (360 mg/kg) extracts for 14 days showed different degrees of improvement on LPS-induced AD model as measured by Y-maze test, Morris water maze test, and Histopathological examination. Moreover, the results of ELISA suggested petroleum ether (PE) extracts were worth recommending for inhibiting the high level of IBA-1, IL-1β, Aβ1-42 and p-tau in hippocampus and cortex after treatment with LPS. CONCLUSIONS The present study demonstrated for the first time that AOF attenuated LPS-induced learning and memory impairment, which may be associated with its inhibitory effect on neuroinflammation, amyloids-β (Aβ) deposition and p-tau. This research provided a theoretical basis for elucidating the traditional theory of AOF, and was also the stepping stone to the next step.
Collapse
Affiliation(s)
- Yunlong Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Mengshi Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Kaiyue Fan
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Tongde Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Tingxu Yan
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Bo Wu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Ying Jia
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China.
| |
Collapse
|
371
|
Ekert JO, Gould RL, Reynolds G, Howard RJ. TNF alpha inhibitors in Alzheimer's disease: A systematic review. Int J Geriatr Psychiatry 2018. [PMID: 29516540 DOI: 10.1002/gps.4871] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVES The objective of this study was to evaluate the effect of tumour necrosis factor-alpha inhibitors (TNF-αI) on Alzheimer's disease-associated pathology. DESIGN A literature search of PubMed, Embase, PsychINFO, Web of Science, Scopus, and the Cochrane Library databases for human and animal studies that evaluated the use of TNF-αI was performed on 26 October 2016. RESULTS The main outcomes assessed were cognition and behaviour, reduction in brain tissue mass, presence of plaques and tangles, and synaptic function. Risk of bias was assessed regarding blinding, statistical model, outcome reporting, and other biases. Sixteen studies were included, 13 of which were animal studies and 3 of which were human. All animal studies found that treatment with TNF-αI leads to an improvement in cognition and behaviour. None of the studies measured change in brain tissue mass. The majority of studies documented a beneficial effect in other areas, including the presence of plaques and tangles and synaptic function. The amount of data from human studies was limited. Two out of 3 studies concluded that TNF-αI are beneficial in Alzheimer's disease patients, with one being an observational study and the latter being a small pilot study, with a high risk of bias. CONCLUSION It was concluded that a large-scale randomized controlled trial assessing the effectiveness of TNF-αI on humans is warranted.
Collapse
Affiliation(s)
- Justyna O Ekert
- Division of Psychiatry, University College London, London, UK
| | - Rebecca L Gould
- Division of Psychiatry, University College London, London, UK
| | - Gemma Reynolds
- Department of Psychology, Middlesex University, London, UK
| | - Robert J Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
372
|
Pistacia lentiscus oil attenuates memory dysfunction and decreases levels of biomarkers of oxidative stress induced by lipopolysaccharide in rats. Brain Res Bull 2018; 140:140-147. [PMID: 29715489 DOI: 10.1016/j.brainresbull.2018.04.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 04/13/2018] [Accepted: 04/26/2018] [Indexed: 11/21/2022]
Abstract
Pistacia lentiscus L. is a well-known medicinal plant that has been used for its antioxidant, anti-inflammatory, neuroprotective, and hepatoprotective effects. However, the neuroprotective effect of Pistacia lentiscus oil (PLo) of has not been reported. The present study was designed to examine the neuroprotective and hepatoprotective effects of PLo aigainst lipopolysaccharide (LPS)-induced memory impairment and oxidative damage in rats. Twenty-four adult male Wistar rats were equally divided into three groups. The first group was kept as a control. In the second group, LPS was given at the single dose of 1 mg/kg intraperitoneally (i.p.). In the third group, PLo (3.3 mL/kg; per orally (p.o.)) was administered daily for 15 days, and challenged with LPS (1 mg/kg; i.p. injection two h before behavioral test). Thereafter, memory was assessed using spatial object recognition test. Cholinesterase activity and oxidative stress response were estimated in brain tissues and liver. PLo attenuated LPS-induced memory impairment in spatial object recognition test (p < 0.05). LPS treatment caused significant oxidative damage via induction of lipid peroxidation and reductions antioxidant defense system potency in the brain tissue and liver. Moreover, LPS increased brain activity of acetylcholinesterase and butyrylcholinesterase activity in the liver. The present results suggest that the beneficial effects of PLo on memory impairment of LPS-treated rats may be due to its protective effects against oxidative stress damage presumably via its antioxidant property.
Collapse
|
373
|
Wang LM, Wu Q, Kirk RA, Horn KP, Ebada Salem AH, Hoffman JM, Yap JT, Sonnen JA, Towner RA, Bozza FA, Rodrigues RS, Morton KA. Lipopolysaccharide endotoxemia induces amyloid-β and p-tau formation in the rat brain. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2018; 8:86-99. [PMID: 29755842 PMCID: PMC5944824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/01/2018] [Indexed: 06/08/2023]
Abstract
Amyloid beta (Aβ) plaques are not specific to Alzheimer's disease and occur with aging and neurodegenerative disorders. Soluble brain Aβ may be neuroprotective and increases in response to neuroinflammation. Sepsis is associated with neurocognitive compromise. The objective was to determine, in a rat endotoxemia model of sepsis, whether neuroinflammation and soluble Aβ production are associated with Aβ plaque and hyperphosphorylated tau deposition in the brain. Male Sprague Dawley rats received a single intraperitoneal injection of 10 mg/kg of lipopolysaccharide endotoxin (LPS). Brain and blood levels of IL-1β, IL-6, and TNFα and cortical microglial density were measured in LPS-injected and control animals. Soluble brain Aβ and p-tau were compared and Aβ plaques were quantified and characterized. Brain uptake of [18F]flutemetamol was measured by phosphor imaging. LPS endotoxemia resulted in elevations of cytokines in blood and brain. Microglial density was increased in LPS-treated rats relative to controls. LPS resulted in increased soluble Aβ and in p-tau levels in whole brain. Progressive increases in morphologically-diffuse Aβ plaques occurred throughout the interval of observation (to 7-9 days post LPS). LPS endotoxemia resulted in increased [18F]flutemetamol in the cortex and increased cortex: white matter ratios of activity. In conclusion, LPS endotoxemia causes neuroinflammation, increased soluble Aβ and Aβ diffuse plaques in the brain. Aβ PET tracers may inform this neuropathology. Increased p-tau in the brain of LPS treated animals suggests that downstream consequences of Aβ plaque formation may occur. Further mechanistic and neurocognitive studies to understand the causes and consequences of LPS-induced neuropathology are warranted.
Collapse
Affiliation(s)
- Li-Ming Wang
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake City, UT, USA
| | - Qi Wu
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake City, UT, USA
| | - Ryan A Kirk
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake City, UT, USA
| | - Kevin P Horn
- Department of Radiology, University of WashingtonSeattle, WA, USA
| | - Ahmed H Ebada Salem
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake City, UT, USA
| | - John M Hoffman
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake City, UT, USA
| | - Jeffrey T Yap
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake City, UT, USA
| | - Joshua A Sonnen
- Department of Pathology, University of UtahSalt Lake City, UT, USA
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research FoundationOklahoma City, OK, USA
| | - Fernando A Bozza
- Critical Care Lab and Immunopharmacology Lab, Oswaldo Cruz FoundationRio de Janeiro, Brazil
| | - Rosana S Rodrigues
- Department of Radiology, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Kathryn A Morton
- Department of Radiology and Imaging Sciences, University of UtahSalt Lake City, UT, USA
| |
Collapse
|
374
|
Chronically raised C-reactive protein is inversely associated with cortical β-amyloid in older adults with subjective memory complaints. Exp Gerontol 2018; 108:226-230. [PMID: 29704641 DOI: 10.1016/j.exger.2018.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/15/2018] [Accepted: 04/18/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND Inflammation promotes amyloidogenesis in animals and markers of inflammation are associated with β-amyloid (Aβ) in humans. Hence, we sought to examine the cross-sectional associations between chronically elevated plasma C reactive protein (CRP) and cortical Aβ in 259 non-demented elderly individuals reporting subjective memory complaints from the Multidomain Alzheimer Preventive Trial (MAPT). METHODS Cortical-to-cerebellar standard uptake value ratios were obtained using [18F] florbetapir positron emission tomography (PET). CRP was measured in plasma using immunoturbidity. Chronically raised CRP was defined as having 2 consecutively high CRP readings (>3 mg/l ≤ 10 mg/l) between study baseline and the 1 year visit (visits were performed at baseline, 6 months, 1 year and then annually). Associations were explored using adjusted multiple linear regression. RESULTS Chronically raised CRP was found to be inversely associated with cortical Aβ (B-coefficient: -0.054, SE: 0.026, p = 0.040) and this association seemed to be specific to apolipoprotein E (Apo E) ε4 carriers (B-coefficient: -0.130, SE: 0.058, p = 0.027). CRP as an isolated reading measured closest to PET scan was also inversely associated with cortical Aβ when CRP was treated as a dichotomized variable (high CRP > 3 mg/l ≤ 10 mg/l, B-coefficient: -0.048, SE: 0.023, p = 0.043). CONCLUSIONS Our preliminary findings suggest that inflammation might be beneficial in the early stages of Alzheimer's disease as the immune systems attempts to combat Aβ pathology particularly in ApoE ε4 carriers. Investigating the temporal relationships between cerebral Aβ and a panel of inflammatory markers would provide further evidence as to whether chronic inflammation might modulate amyloidogenesis in vivo.
Collapse
|
375
|
Liu Y, Zhang Y, Zheng X, Fang T, Yang X, Luo X, Guo A, Newell KA, Huang XF, Yu Y. Galantamine improves cognition, hippocampal inflammation, and synaptic plasticity impairments induced by lipopolysaccharide in mice. J Neuroinflammation 2018; 15:112. [PMID: 29669582 PMCID: PMC5907415 DOI: 10.1186/s12974-018-1141-5] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/29/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Neuroinflammation plays an important role in the onset and progression of neurodegenerative diseases such as Alzheimer's disease. Lipopolysaccharide (LPS, endotoxin) levels are higher in the brains of Alzheimer's disease patients and are associated with neuroinflammation and cognitive decline, while neural cholinergic signaling controls inflammation. This study aimed to examine the efficacy of galantamine, a clinically approved cholinergic agent, in alleviating LPS-induced neuroinflammation and cognitive decline as well as the associated mechanism. METHODS Mice were treated with galantamine (4 mg/kg, intraperitoneal injection) for 14 days prior to LPS exposure (intracerebroventricular injection). Cognitive tests were performed, including the Morris water maze and step-through tests. mRNA expression of the microglial marker (CD11b), astrocytic marker (GFAP), and pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) were examined in the hippocampus by quantitative RT-PCR. The inflammatory signaling molecule, nuclear factor-kappa B (NF-κB p65), and synapse-associated proteins (synaptophysin, SYN, and postsynaptic density protein 95, PSD-95) were examined in the hippocampus by western blotting. Furthermore, NF-κB p65 levels in microglial cells and hippocampal neurons were examined in response to LPS and galantamine. RESULTS Galantamine treatment prevented LPS-induced deficits in spatial learning and memory as well as memory acquisition of the passive avoidance response. Galantamine decreased the expression of microglia and astrocyte markers (CD11b and GFAP), pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), and NF-κB p65 in the hippocampus of LPS-exposed mice. Furthermore, galantamine ameliorated LPS-induced loss of synapse-associated proteins (SYN and PSD-95) in the hippocampus. In the in vitro study, LPS increased NF-κB p65 levels in microglia (BV-2 cells); the supernatant of LPS-stimulated microglia (Mi-sup), but not LPS, decreased the viability of hippocampal neuronal cells (HT-22 cells) and increased NF-κB p65 levels as well as expression of pro-inflammatory cytokines (IL-1β, IL-6) in HT-22 cells. Importantly, galantamine reduced the inflammatory response not only in the BV-2 microglia cell line, but also in the HT-22 hippocampal neuronal cell line. CONCLUSIONS These findings indicate that galantamine could be a promising treatment to improve endotoxin-induced cognitive decline and neuroinflammation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia.
| | - Yuyun Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xian Zheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Tongyong Fang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xia Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xuan Luo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Anlei Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Kelly A Newell
- Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Xu-Feng Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Illawarra Health and Medical Research Institute, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
376
|
Cho MJ, Kim JH, Park CH, Lee AY, Shin YS, Lee JH, Park CG, Cho EJ. Comparison of the effect of three licorice varieties on cognitive improvement via an amelioration of neuroinflammation in lipopolysaccharide-induced mice. Nutr Res Pract 2018; 12:191-198. [PMID: 29854324 PMCID: PMC5974064 DOI: 10.4162/nrp.2018.12.3.191] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/08/2018] [Accepted: 02/08/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/OBJECTIVES Neuroinflammation plays critical role in neurodegenerative disorders, such as Alzheimer's disease (AD). We investigated the effect of three licorice varieties, Glycyrhiza uralensis, G. glabra, and Shinwongam (SW) on a mouse model of inflammation-induced memory and cognitive deficit. MATERIALS/METHODS C57BL/6 mice were injected with lipopolysaccharide (LPS; 2.5 mg/kg, intraperitoneally) and orally administrated G. uralensis, G. glabra, and SW extract (150 mg/kg/day). SW, a new species of licorice in Korea, was combined with G. uralensis and G. glabra. Behavioral tests, including the T-maze, novel object recognition and Morris water maze, were carried out to assess learning and memory. In addition, the expressions of inflammation-related proteins in brain tissue were measured by western blotting. RESULTS There was a significant decrease in spatial and objective recognition memory in LPS-induced cognitive impairment group, as measured by the T-maze and novel object recognition test; however, the administration of licorice ameliorated these deficits. In addition, licorice-treated groups exhibited improved learning and memory ability in the Morris water maze. Furthermore, LPS-injected mice had up-regulated pro-inflammatory proteins, such as inducible nitric oxide synthase (iNOS), cyclooxygenase-2, interleukin-6, via activation of toll like receptor 4 (TLR4) and nuclear factor-kappa B (NFκB) pathways in the brain. However, these were attenuated by following administration of the three licorice varieties. Interestingly, the SW-administered group showed greater inhibition of iNOS and TLR4 when compared with the other licorice varieties. Furthermore, there was a significant increase in the expression of brain-derived neurotrophic factor (BDNF) in the brain of LPS-induced cognitively impaired mice that were administered licorice, with the greatest effect following SW treatment. CONCLUSIONS The three licorice varieties ameliorated the inflammation-induced cognitive dysfunction by down-regulating inflammatory proteins and up-regulating BDNF. These results suggest that licorice, in particular SW, could be potential therapeutic agents against cognitive impairment.
Collapse
Affiliation(s)
- Min Ji Cho
- Department of Food Science and Nutrition, Pusan National University, 2 Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Ji Hyun Kim
- Department of Food Science and Nutrition, Pusan National University, 2 Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Chan Hum Park
- Department of Herbal Crop Research, NIHHS, RDA, Chungbuk 27709, Korea
| | - Ah Young Lee
- Department of Food Science and Nutrition, Pusan National University, 2 Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Yu Su Shin
- Department of Herbal Crop Research, NIHHS, RDA, Chungbuk 27709, Korea
| | - Jeong Hoon Lee
- Department of Herbal Crop Research, NIHHS, RDA, Chungbuk 27709, Korea
| | - Chun Geun Park
- Department of Herbal Crop Research, NIHHS, RDA, Chungbuk 27709, Korea
| | - Eun Ju Cho
- Department of Food Science and Nutrition, Pusan National University, 2 Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| |
Collapse
|
377
|
Huynh K, Martins RN, Meikle PJ. Lipidomic Profiles in Diabetes and Dementia. J Alzheimers Dis 2018; 59:433-444. [PMID: 28582856 DOI: 10.3233/jad-161215] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lipids are a diverse class of hydrophobic and amphiphilic molecules which make up the bulk of most biological systems and are essential for human life. The role of lipids in health and disease has been recognized for many decades, as evidenced by the early identification of cholesterol as an important risk factor of heart disease and the development and introduction of statins as a one of the most successful therapeutic interventions to date. While several studies have demonstrated an increased risk of dementia, including Alzheimer's disease (AD), in those with diabetes mellitus, the nature of this risk is not well understood. Recent developments in the field of lipidomics, driven primarily by technological advances in high pressure liquid chromatography and particularly mass spectrometry, have enabled the detailed characterization of the many hundreds of individual lipid species in mammalian systems and their association with disease states. Diabetes mellitus and AD have received particular attention due to their prominence in Western societies as a result of the ongoing obesity epidemic and the aging populations. In this review, we examine how these lipidomic studies are informing on the relationship between lipid metabolism with diabetes and AD and how this may inform on the common pathological pathways that link diabetes risk with dementia.
Collapse
Affiliation(s)
- Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Ralph N Martins
- School of Biomedical and Health Sciences, Edith Cowan University, Perth Western Australia, WA, Australia.,Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
378
|
Kokiko-Cochran ON, Godbout JP. The Inflammatory Continuum of Traumatic Brain Injury and Alzheimer's Disease. Front Immunol 2018; 9:672. [PMID: 29686672 PMCID: PMC5900037 DOI: 10.3389/fimmu.2018.00672] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/19/2018] [Indexed: 12/23/2022] Open
Abstract
The post-injury inflammatory response is a key mediator in long-term recovery from traumatic brain injury (TBI). Moreover, the immune response to TBI, mediated by microglia and macrophages, is influenced by existing brain pathology and by secondary immune challenges. For example, recent evidence shows that the presence of beta-amyloid and phosphorylated tau protein, two hallmark features of AD that increase during normal aging, substantially alter the macrophage response to TBI. Additional data demonstrate that post-injury microglia are “primed” and become hyper-reactive following a subsequent acute immune challenge thereby worsening recovery. These alterations may increase the incidence of neuropsychiatric complications after TBI and may also increase the frequency of neurodegenerative pathology. Therefore, the purpose of this review is to summarize experimental studies examining the relationship between TBI and development of AD-like pathology with an emphasis on the acute and chronic microglial and macrophage response following injury. Furthermore, studies will be highlighted that examine the degree to which beta-amyloid and tau accumulation as well as pre- and post-injury immune stressors influence outcome after TBI. Collectively, the studies described in this review suggest that the brain’s immune response to injury is a key mediator in recovery, and if compromised by previous, coincident, or subsequent immune stressors, post-injury pathology and behavioral recovery will be altered.
Collapse
Affiliation(s)
- Olga N Kokiko-Cochran
- Department of Neuroscience, Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jonathan P Godbout
- Department of Neuroscience, Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
379
|
Erickson MA, Banks WA. Neuroimmune Axes of the Blood-Brain Barriers and Blood-Brain Interfaces: Bases for Physiological Regulation, Disease States, and Pharmacological Interventions. Pharmacol Rev 2018; 70:278-314. [PMID: 29496890 PMCID: PMC5833009 DOI: 10.1124/pr.117.014647] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Central nervous system (CNS) barriers predominantly mediate the immune-privileged status of the brain, and are also important regulators of neuroimmune communication. It is increasingly appreciated that communication between the brain and immune system contributes to physiologic processes, adaptive responses, and disease states. In this review, we discuss the highly specialized features of brain barriers that regulate neuroimmune communication in health and disease. In section I, we discuss the concept of immune privilege, provide working definitions of brain barriers, and outline the historical work that contributed to the understanding of CNS barrier functions. In section II, we discuss the unique anatomic, cellular, and molecular characteristics of the vascular blood-brain barrier (BBB), blood-cerebrospinal fluid barrier, and tanycytic barriers that confer their functions as neuroimmune interfaces. In section III, we consider BBB-mediated neuroimmune functions and interactions categorized as five neuroimmune axes: disruption, responses to immune stimuli, uptake and transport of immunoactive substances, immune cell trafficking, and secretions of immunoactive substances. In section IV, we discuss neuroimmune functions of CNS barriers in physiologic and disease states, as well as pharmacological interventions for CNS diseases. Throughout this review, we highlight many recent advances that have contributed to the modern understanding of CNS barriers and their interface functions.
Collapse
Affiliation(s)
- Michelle A Erickson
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - William A Banks
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
380
|
Amiri S, Yousefi-Ahmadipour A, Hosseini MJ, Haj-Mirzaian A, Momeny M, Hosseini-Chegeni H, Mokhtari T, Kharrazi S, Hassanzadeh G, Amini SM, Jafarinejad S, Ghazi-Khansari M. Maternal exposure to silver nanoparticles are associated with behavioral abnormalities in adulthood: Role of mitochondria and innate immunity in developmental toxicity. Neurotoxicology 2018; 66:66-77. [PMID: 29550386 DOI: 10.1016/j.neuro.2018.03.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 03/06/2018] [Accepted: 03/10/2018] [Indexed: 01/06/2023]
Abstract
Silver nanoparticles (Ag-NPs) are currently used in a wide range of consumer products. Considering the small size of Ag-NPs, they are able to pass through variety of biological barriers and exert their effects. In this regard, the unique physicochemical properties of Ag-NPs along with its high application in the industry have raised concerns about their negative effects on human health. Therefore, it investigated whether prenatal exposure to low doses of Ag-NPs is able to induce any abnormality in the cognitive and behavioral performance of adult offspring. We gavaged pregnant NMRI mice with, 1) Deionized water as vehicle, 2) Ag-NPs 10 nm (0.26 mg/kg/day), 3) Ag-NPs 30 nm (0.26 mg/kg/day), and 4) AgNO3 (0.26 mg/kg/day) from gestational day (GD) 0 until delivery day. At the postnatal day (PD) 1, our results showed that high concentration of silver is present in the brain of pups. Further, we observed mitochondrial dysfunction and upregulation of the genes relevant to innate immune system in the brain. At PD 60, results revealed that prenatal exposure to Ag-NPs provoked severe cognitive and behavioral abnormalities in male offspring. In addition, we found that prenatal exposure to Ag-NPs was associated with abnormal mitochondrial function and significant up-regulation of the genes relevant to innate immunity in the brain. Although the Ag-NPs have been considered as safe compounds at low doses, our results indicate that prenatal exposure to low doses of Ag-NPs is able to induce behavioral and cognitive abnormalities in adulthood. Also, we found that these effects are at least partly associated with hippocampal mitochondrial dysfunction and the activation of sterile inflammation during early stages of life.
Collapse
Affiliation(s)
- Shayan Amiri
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliakbar Yousefi-Ahmadipour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Tissue Engineering and Applied Cell Sciences, Department of School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Arya Haj-Mirzaian
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Heshmat Hosseini-Chegeni
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Mokhtari
- Research Center of Nervous System Stem Cells, Department of Anatomy, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran; Department of Anatomy, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Sharmin Kharrazi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Amini
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jafarinejad
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Ghazi-Khansari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Iran Nanosafety Network (INSN) of Iran Nanotechnology Initiative Council (INIC), Tehran, Iran.
| |
Collapse
|
381
|
Frühauf-Perez PK, Temp FR, Pillat MM, Signor C, Wendel AL, Ulrich H, Mello CF, Rubin MA. Spermine protects from LPS-induced memory deficit via BDNF and TrkB activation. Neurobiol Learn Mem 2018; 149:135-143. [DOI: 10.1016/j.nlm.2018.02.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/18/2018] [Accepted: 02/14/2018] [Indexed: 12/18/2022]
|
382
|
Inflammasome Activation by Methamphetamine Potentiates Lipopolysaccharide Stimulation of IL-1β Production in Microglia. J Neuroimmune Pharmacol 2018; 13:237-253. [PMID: 29492824 DOI: 10.1007/s11481-018-9780-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/19/2018] [Indexed: 01/09/2023]
Abstract
Methamphetamine (Meth) is an addictive psychostimulant abused worldwide. Ample evidence indicate that chronic abuse of Meth induces neurotoxicity via microglia-associated neuroinflammation and the activated microglia present in both Meth-administered animals and human abusers. The development of anti-neuroinflammation as a therapeutic strategy against Meth dependence promotes research to identify inflammatory pathways that are specifically tied to Meth-induced neurotoxicity. Currently, the exact mechanisms for Meth-induced microglia activation are largely unknown. NLRP3 is a well-studied cytosolic pattern recognition receptor (PRR), which promotes the assembly of the inflammasome in response to the danger-associated molecular patterns (DAMPs). It is our hypothesis that Meth activates NLRP3 inflammasome in microglia and promotes the processing and release of interleukin (IL)-1β, resulting in neurotoxic activity. To test this hypothesis, we studied the effects of Meth on IL-1β maturation and release from rat cortical microglial cultures. Incubation of microglia with physiologically relevant concentrations of Meth after lipopolysaccharide (LPS) priming produced an enhancement on IL-1β maturation and release. Meth treatment potentiated aggregation of inflammasome adaptor apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), induced activation of the IL-1β converting enzyme caspase-1 and produced lysosomal and mitochondrial impairment. Blockade of capase-1 activity, lysosomal cathepsin B activity or mitochondrial ROS production by their specific inhibitors reversed the effects of Meth, demonstrating an involvement of inflammasome in Meth-induced microglia activation. Taken together, our results suggest that Meth triggers microglial inflammasome activation in a manner dependent on both mitochondrial and lysosomal danger-signaling pathways.
Collapse
|
383
|
Torres L, Robinson SA, Kim DG, Yan A, Cleland TA, Bynoe MS. Toxoplasma gondii alters NMDAR signaling and induces signs of Alzheimer's disease in wild-type, C57BL/6 mice. J Neuroinflammation 2018; 15:57. [PMID: 29471842 PMCID: PMC5824585 DOI: 10.1186/s12974-018-1086-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disease associated with cognitive decline and complete loss of basic functions. The ubiquitous apicomplexan parasite Toxoplasma gondii (T. gondii) infects up to one third of the world's population and is implicated in AD. METHODS We infected C57BL/6 wild-type male and female mice with 10 T. gondii ME49 cysts and assessed whether infection led to behavioral and anatomical effects using immunohistochemistry, immunofluorescence, Western blotting, cell culture assays, as well as an array of mouse behavior tests. RESULTS We show that T. gondii infection induced two major hallmarks of AD in the brains of C57BL/6 male and female mice: beta-amyloid (Aβ) immunoreactivity and hyperphosphorylated Tau. Infected mice showed significant neuronal death, loss of N-methyl-D-aspartate receptor (NMDAR) expression, and loss of olfactory sensory neurons. T. gondii infection also caused anxiety-like behavior, altered recognition of social novelty, altered spatial memory, and reduced olfactory sensitivity. This last finding was exclusive to male mice, as infected females showed intact olfactory sensitivity. CONCLUSIONS These results demonstrate that T. gondii can induce advanced signs of AD in wild-type mice and that it may induce AD in some individuals with underlying health problems.
Collapse
Affiliation(s)
- Luisa Torres
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Sudie-Ann Robinson
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Do-Geun Kim
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Angela Yan
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Thomas A Cleland
- Department of Psychology, Cornell University, Ithaca, NY, 14853, USA
| | - Margaret S Bynoe
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
384
|
Thymelaea lythroides extract attenuates microglial activation and depressive-like behavior in LPS-induced inflammation in adult male rats. Biomed Pharmacother 2018; 99:655-663. [PMID: 29710462 DOI: 10.1016/j.biopha.2018.01.125] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/13/2018] [Accepted: 01/24/2018] [Indexed: 12/29/2022] Open
Abstract
Thymelaea lythroides extract is widely used as a traditional folk medicine in Morocco, especially for the treatment of diabetes, rheumatism and Inflammatory disease. The aim of the study is to evaluate the possible effect of methanolic extract of Thymelaea lythroides in repressing the inflammatory responses and long-lasting depression-like behavior associated with neuroinflammation in adult rats after neonatal LPS exposure. Male rat pups were treated systemically with either LPS (250??g/kg) or vehicle (phosphate buffer saline) on postnatal day 14. Six hours later, the LPS groups were assigned to intraperitoneal (ip) injection of Minocycline (50?mg/kg) or Thymelaea lythroides (200?mg/kg). Thereafter, in adulthood (postnatal days 90-97), the spontaneous locomotor activity and depression-like behavior were assessed successively in open field and forced swim tests. The levels of proinflammatory cytokines, oxidative damage, and activation of microglia were determined in the hippocampus (HP) of male rats on (PND90-97). Our results showed that open field hypoactivity and increased immobility period in LPS-induced adult rats were normalized on treatment with Thymelaea lythroides and minocycline. Both treatments attenuate the overactivated microglial cells in the CA1 and CA3 of hippocampus (HP) and significantly reduced the oxidative-nitrosative stress markers and cytokine (TNF ?) production in the HP. Thymelaea lythroides seems to have similar neuroprotective effects to Minocycline, and such protection may be due to: reduction of oxidative stress, upregulation of inflammatory mediators production, antidepressant behavior which all are associated with neuroinflammation.
Collapse
|
385
|
Garranzo-Asensio M, San Segundo-Acosta P, Martínez-Useros J, Montero-Calle A, Fernández-Aceñero MJ, Häggmark-Månberg A, Pelaez-Garcia A, Villalba M, Rabano A, Nilsson P, Barderas R. Identification of prefrontal cortex protein alterations in Alzheimer's disease. Oncotarget 2018; 9:10847-10867. [PMID: 29541381 PMCID: PMC5834268 DOI: 10.18632/oncotarget.24303] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 01/13/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia in developed countries. A better understanding of the events taking place at the molecular level would help to identify novel protein alterations, which might be used in diagnosis or for treatment development. In this study, we have performed the high-throughput analysis of 706 molecules mostly implicated in cell-cell communication and cell signaling processes by using two antibody microarray platforms. We screened three AD pathological groups -each one containing four pooled samples- from Braak stages IV, V and VI, and three control groups from two healthy subjects, five frontotemporal and two vascular dementia patients onto Panorama and L-Series antibody microarrays to identify AD-specific alterations not common to other dementias. Forty altered proteins between control and AD groups were detected, and validated by i) meta-analysis of mRNA alterations, ii) WB, and iii) FISH and IHC using an AD-specific tissue microarray containing 44 samples from AD patients at different Braak stages, and frontotemporal and vascular dementia patients and healthy individuals as controls. We identified altered proteins in AD not common to other dementias like the E3 ubiquitin-protein ligase TOPORS, Layilin and MICB, and validated the association to AD of the previously controverted proteins DDIT3 and the E3 ubiquitin-protein ligase XIAP. These altered proteins constitute interesting targets for further immunological analyses using sera, plasma and CSF to identify AD blood- or cerebrospinal fluid-biomarkers and to perform functional analysis to determine their specific role in AD, and their usefulness as potential therapeutic targets of intervention.
Collapse
Affiliation(s)
- Maria Garranzo-Asensio
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
| | - Pablo San Segundo-Acosta
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
| | - Javier Martínez-Useros
- Translational Oncology Division, OncoHealth Institute, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
| | - Ana Montero-Calle
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
| | - María Jesús Fernández-Aceñero
- Servicio de Anatomía Patológica Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Departamento de Anatomía Patològica, Facultad de Medicina, Complutense University of Madrid, Madrid, Spain
| | - Anna Häggmark-Månberg
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH – Royal Institute of Technology, Stockholm, Sweden
| | | | - Mayte Villalba
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
| | - Alberto Rabano
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| | - Peter Nilsson
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Rodrigo Barderas
- Biochemistry and Molecular Biology Department I, Chemistry Faculty, Complutense University of Madrid, Madrid, Spain
- UFIEC, National Institute of Health Carlos III, Majadahonda, Madrid, Spain
| |
Collapse
|
386
|
Philippens IH, Ormel PR, Baarends G, Johansson M, Remarque EJ, Doverskog M. Acceleration of Amyloidosis by Inflammation in the Amyloid-Beta Marmoset Monkey Model of Alzheimer's Disease. J Alzheimers Dis 2018; 55:101-113. [PMID: 27662314 PMCID: PMC5115608 DOI: 10.3233/jad-160673] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background: The immune system is increasingly mentioned as a potential target for Alzheimer’s disease (AD) treatment. Objective: In the present pilot study, the effect of (neuro)inflammation on amyloidopathy was investigated in the marmoset monkey, which has potential as an AD animal model due to its natural cerebral amyloidosis similar to humans. Methods: Six adult/aged marmosets (Callithrix jacchus) were intracranial injected with amyloid-beta (Aβ) fibrils at three cortical locations in the right hemisphere. Additionally, in half of the monkeys, lipopolysaccharide (LPS) was co-injected with the Aβ fibrils and injected in the other hemisphere without Aβ fibrils. The other three monkeys received phosphate buffered saline instead of LPS, as a control for the inflammatory state. The effect of inflammation on amyloidopathy was also investigated in an additional monkey that suffered from chronic inflammatory wasting syndrome. Mirror histology sections were analyzed to assess amyloidopathy and immune reaction, and peripheral blood for AD biomarker expression. Results: All LPS-injected monkeys showed an early AD immune blood cell expression profile on CD95 and CD45RA. Two out of three monkeys injected with Aβ and LPS and the additional monkey, suffering from chronic inflammation, developed plaques. None of the controls, injected with Aβ only, developed any plaques. Conclusion: This study shows the importance of immune modulation on the susceptibility for amyloidosis, a hallmark of AD, which offers new perspectives for disease modifying approaches in AD.
Collapse
Affiliation(s)
| | - Paul R Ormel
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | - Guus Baarends
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | - Maja Johansson
- Umecrine Cognition AB, Karolinska Institute Science Park, Solna, Sweden
| | - Ed J Remarque
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | - Magnus Doverskog
- Umecrine Cognition AB, Karolinska Institute Science Park, Solna, Sweden
| |
Collapse
|
387
|
Yang CF, Lin SP, Chiang CP, Wu YH, H'ng WS, Chang CP, Chen YT, Wu JY. Loss of GPNMB Causes Autosomal-Recessive Amyloidosis Cutis Dyschromica in Humans. Am J Hum Genet 2018; 102:219-232. [PMID: 29336782 DOI: 10.1016/j.ajhg.2017.12.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 12/15/2017] [Indexed: 11/29/2022] Open
Abstract
Amyloidosis cutis dyschromica (ACD) is a distinct form of primary cutaneous amyloidosis characterized by generalized hyperpigmentation mottled with small hypopigmented macules on the trunks and limbs. Affected families and sporadic case subjects have been reported predominantly in East and Southeast Asian ethnicities; however, the genetic cause has not been elucidated. We report here that the compound heterozygosity or homozygosity of GPNMB truncating alleles is the cause of autosomal-recessive ACD. Six nonsense or frameshift mutations were identified in nine individuals diagnosed with ACD. Immunofluorescence analysis of skin biopsies showed that GPNMB is expressed in all epidermal cells, with the highest staining observed in melanocytes. GPNMB staining is significantly reduced in the lesional skin of affected individuals. Hyperpigmented lesions exhibited significantly increased amounts of DNA/keratin-positive amyloid deposits in the papillary dermis and infiltrating macrophages compared with hypo- or depigmented macules. Depigmentation of the lesions was attributable to loss of melanocytes. Intracytoplasmic fibrillary aggregates were observed in keratinocytes scattered in the lesional epidermis. Thus, our analysis indicates that loss of GPNMB, which has been implicated in melanosome formation, autophagy, phagocytosis, tissue repair, and negative regulation of inflammation, underlies autosomal-recessive ACD and provides insights into the etiology of amyloidosis and pigment dyschromia.
Collapse
Affiliation(s)
- Chi-Fan Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Shuan-Pei Lin
- Department of Medical Research, MacKay Memorial Hospital, Taipei 104, Taiwan; Department of Pediatrics, MacKay Memorial Hospital, Taipei 104, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | - Chien-Ping Chiang
- Departments of Dermatology, Tri-Service General Hospital, Taipei 114, Taiwan; Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan
| | - Yu-Hung Wu
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan; Department of Dermatology, MacKay Memorial Hospital, Taipei 104, Taiwan
| | - Weng Siong H'ng
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Chun-Ping Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.
| |
Collapse
|
388
|
Chamaa F, Bitar L, Darwish B, Saade NE, Abou-Kheir W. Intracerebroventricular injections of endotoxin (ET) reduces hippocampal neurogenesis. J Neuroimmunol 2018; 315:58-67. [DOI: 10.1016/j.jneuroim.2017.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/16/2017] [Accepted: 12/17/2017] [Indexed: 01/12/2023]
|
389
|
Hossain MS, Tajima A, Kotoura S, Katafuchi T. Oral ingestion of plasmalogens can attenuate the LPS-induced memory loss and microglial activation. Biochem Biophys Res Commun 2018; 496:1033-1039. [DOI: 10.1016/j.bbrc.2018.01.078] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 01/11/2018] [Indexed: 01/06/2023]
|
390
|
Chen PL, Wang WJ, Rao YQ, Li J, Cheng MJ. Serum containing Gengnianchun formula suppresses amyloid β‑induced inflammatory cytokines in BV‑2 microglial cells by inhibiting the NF‑κB and JNK signaling pathways. Mol Med Rep 2018; 17:5043-5048. [PMID: 29393440 PMCID: PMC5865966 DOI: 10.3892/mmr.2018.8524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 01/23/2017] [Indexed: 01/08/2023] Open
Abstract
As the resident macrophages of the brain's innate immune system, microglial cells are key modulators in the neurodegenerative disease Alzheimer's disease (AD). In particular, the activation and accumulation of microglial cells around amyloid plaques is considered to result in chronic neuroinflammation. Although the pathologic mechanism remains to be fully elucidated, inflammation has been shown to be critical in the pathogenesis of AD. The Gengnianchun (GNC) formula has long been used to treat perimenopausal syndrome clinically, and is particularly effective in improving learning ability and memory. Our previous study demonstrated that GNC formula had an anti-inflammatory effect and offered neuroprotection in animal experiments. In the present study, the anti-inflammatory properties of GNC and its underlying mechanism of action were examined in BV-2 microglial cells. Amyloid-β peptide (Aβ)-stimulated microglial cells were examined for the production of proinflammatory cytokines and the underlying signaling pathways. Compared with the normal control group, the protein expression levels of IL-1β and TNF-α were significantly increased following treatment with Aβ (P<0.01), but medicated rat serum containing GNC formula (MRS) could significantly attenuated the Aβ-induced secretion of these pro-inflammatory cytokines. It was identified by CCK-8 assay that the viability of the BV-2 cells was not reduced following treatment with various concentrations of MRS. The phosphorylation of factor-κB (NF-κB) and c-Jun N-terminal kinase (JNK) was markedly increased following treatment with Aβ, compared with the normal control group (P<0.01). However, treatment with MRS resulted in a significant reduction in the phosphorylation of NF-κB (P<0.05). These results suggested that MRS suppressed the Aβ-induced inflammatory response of microglial cells by inhibiting the NF-κB and JNK signaling pathways. These novel findings provide insights into the development of GNC formula as a therapeutic agent for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Pin-Li Chen
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Wen-Jun Wang
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Yan-Qiu Rao
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Jun Li
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Ming-Jun Cheng
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| |
Collapse
|
391
|
Pretorius E, Bester J, Kell DB. A Bacterial Component to Alzheimer's-Type Dementia Seen via a Systems Biology Approach that Links Iron Dysregulation and Inflammagen Shedding to Disease. J Alzheimers Dis 2018; 53:1237-56. [PMID: 27340854 PMCID: PMC5325058 DOI: 10.3233/jad-160318] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The progression of Alzheimer's disease (AD) is accompanied by a great many observable changes, both molecular and physiological. These include oxidative stress, neuroinflammation, and (more proximal to cognitive decline) the death of neuronal and other cells. A systems biology approach seeks to organize these observed variables into pathways that discriminate those that are highly involved (i.e., causative) from those that are more usefully recognized as bystander effects. We review the evidence that iron dysregulation is one of the central causative pathway elements here, as this can cause each of the above effects. In addition, we review the evidence that dormant, non-growing bacteria are a crucial feature of AD, that their growth in vivo is normally limited by a lack of free iron, and that it is this iron dysregulation that is an important factor in their resuscitation. Indeed, bacterial cells can be observed by ultrastructural microscopy in the blood of AD patients. A consequence of this is that the growing cells can shed highly inflammatory components such as lipopolysaccharides (LPS). These too are known to be able to induce (apoptotic and pyroptotic) neuronal cell death. There is also evidence that these systems interact with elements of vitamin D metabolism. This integrative systems approach has strong predictive power, indicating (as has indeed been shown) that both natural and pharmaceutical iron chelators might have useful protective roles in arresting cognitive decline, and that a further assessment of the role of microbes in AD development is more than highly warranted.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - Janette Bester
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - Douglas B Kell
- School of Chemistry, The University of Manchester, Manchester, Lancs, UK.,The Manchester Institute of Biotechnology, The University of Manchester, Manchester, Lancs, UK.,Centre for Synthetic Biology of Fine and Speciality Chemicals, The University of Manchester, Manchester, Lancs, UK
| |
Collapse
|
392
|
Lee KP, Choi NH, Kim HS, Ahn S, Park IS, Lee DW. Anti-neuroinflammatory effects of ethanolic extract of black chokeberry ( Aronia melanocapa L.) in lipopolysaccharide-stimulated BV2 cells and ICR mice. Nutr Res Pract 2018; 12:13-19. [PMID: 29399292 PMCID: PMC5792251 DOI: 10.4162/nrp.2018.12.1.13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 08/28/2017] [Accepted: 11/07/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND/OBJECTIVES One of the mechanisms considered to be prevalent in the development of Alzheimer's disease (AD) is hyper-stimulation of microglia. Black chokeberry (Aronia melanocapa L.) is widely used to treat diabetes and atherosclerosis, and is known to exert anti-oxidant and anti-inflammatory effects; however, its neuroprotective effects have not been elucidated thus far. MATERIALS/METHODS We undertook to assess the anti-inflammatory effect of the ethanolic extract of black chokeberry friut (BCE) in BV2 cells, and evaluate its neuroprotective effect in the lipopolysaccharide (LPS)-induced mouse model of AD. RESULTS Following stimulation of BV2 cells by LPS, exposure to BCE significantly reduced the generation of nitric oxide as well as mRNA levels of numerous inflammatory factors such as inducible nitric oxide synthase (iNOS), cyclooxygenase 2 (COX-2), interleukin 1 beta (IL-1β), and tumor necrosis factor alpha (TNF-α). In addition, AD was induced in a mouse model by intraperitoneal injection of LPS (250 µg/kg), subsequent to which we investigated the neuroprotective effects of BCE (50 mg/kg) on brain damage. We observed that BCE significantly reduced tissue damage in the hippocampus by downregulating iNOS, COX-2, and TNF-α levels. We further identified the quinic acids in BCE using liquid chromatography-mass spectrometry (LCMS). Furthermore, we confirmed the neuroprotective effect of BCE and quinic acid on amyloid beta-induced cell death in rat hippocampal primary neurons. CONCLUSIONS Our findings suggest that black chokeberry has protective effects against the development of AD.
Collapse
Affiliation(s)
- Kang Pa Lee
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| | - Nan Hee Choi
- Department of Biotechnology, College of Engineering, Daegu University, Gyeongsan 38453, Korea
| | - Hyun-Soo Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea
| | - Sanghyun Ahn
- Department of Anatomy, College of Korean Medicine, Semyung University, Jecheon 27136, Korea
| | - In-Sik Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju 38066, Korea
| | - Dea Won Lee
- Department of Bio-Science, College of Natural Science, Dongguk University, Dongdae-ro 123, Gyeongju, Gyeongbuk 38066, Korea
| |
Collapse
|
393
|
Chen M, Chang YY, Huang S, Xiao LH, Zhou W, Zhang LY, Li C, Zhou RP, Tang J, Lin L, Du ZY, Zhang K. Aromatic-Turmerone Attenuates LPS-Induced Neuroinflammation and Consequent Memory Impairment by Targeting TLR4-Dependent Signaling Pathway. Mol Nutr Food Res 2018; 62. [PMID: 28849618 DOI: 10.1002/mnfr.201700281] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 06/26/2017] [Indexed: 01/06/2023]
Abstract
SCOPE Curcuma longa (turmeric) is a folk medicine in South and Southeast Asia, which has been widely used to alleviate chronic inflammation. Aromatic-turmerone is one of the main components abundant in turmeric essential oil. However, little information is available from controlled studies regarding its biological activities and underlying molecular mechanisms against chronic inflammation in the brain. In the current study, we employed a classical LPS model to study the effect and mechanism of aromatic-turmerone on neuroinflammation. METHODS AND RESULTS The effects of aromatic-turmerone were studied in LPS-treated mice and BV2 cells. The cognitive function assays, protein analyses, and histological examination were performed. Oral administration of aromatic-turmerone could reverse LPS-induced memory disturbance and normalize glucose intake and metabolism in the brains of mice. Moreover, aromatic-turmerone significantly limited brain damage, through inhibiting the activation of microglia and generation of inflammatory cytokines. Further study in vitro revealed that aromatic-turmerone targeted Toll-like receptor 4 mediated downstream signaling, and lowered the release of inflammatory mediators. CONCLUSION These observations indicate that aromatic-turmerone is effective in preventing brain damage caused by neuroinflammation and may be useful in the treatment of neuronal inflammatory diseases.
Collapse
Affiliation(s)
- Min Chen
- Institute of Natural Medicinal Chemistry and Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yuan-Yuan Chang
- Institute of Natural Medicinal Chemistry and Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Shun Huang
- Institute of Natural Medicinal Chemistry and Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Li-Hang Xiao
- Institute of Natural Medicinal Chemistry and Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Wei Zhou
- Institute of Natural Medicinal Chemistry and Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Lan-Yue Zhang
- Institute of Natural Medicinal Chemistry and Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Chun Li
- Institute of Natural Medicinal Chemistry and Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Ren-Ping Zhou
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Jian Tang
- Infinitus (China) Company Ltd, Guangzhou, 510006, China
| | - Li Lin
- Allan Conney Biotechnology Company Ltd, Foshan, 528200, China
| | - Zhi-Yun Du
- Institute of Natural Medicinal Chemistry and Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Kun Zhang
- Institute of Natural Medicinal Chemistry and Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, 510006, China.,Wuyi University, Jiangmen, 529020, China
| |
Collapse
|
394
|
Anaeigoudari A, Norouzi F, Abareshi A, Beheshti F, Aaghaei A, Shafei MN, Gholamnezhad Z, Hosseini M. Protective effects of Nigella sativa on synaptic plasticity impairment induced by lipopolysaccharide. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2018; 9:27-33. [PMID: 29719661 PMCID: PMC5913558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 11/07/2017] [Indexed: 10/28/2022]
Abstract
In the present study the protective effect of Nigella sativa (N. sativa) on synaptic plasticity impairment induced by lipopolysaccharide (LPS) in rats was investigated. Fifty-eight rats were grouped and treated as follows: 1) control (saline), 2) LPS, 3) LPS-N. sativa, and 4) N. sativa. In a Morris water maze test, the escape latency and traveled path to find the platform as well as time spent and the traveled distance in target quadrant (Q1) were measured. Long term potentiation (LTP) from CA1 area of hippocampus followed by high frequency stimulation to Schafer collateral was studied and slope, slope 10-90% and amplitude of field excitatory field potential (fEPSP) were calculated. The escape latency and traveled path in LPS group were significantly higher than those in the control group while, in LPS-N. sativa group these parameters were significantly lower than those in LPS group. The rats in LPS group spent less time and traveled shorter distance in Q1 than the rats in the control group while, in LPS-N. sativa group the rats spent more time and traveled longer distance than the rats in LPS group. LPS significantly decreased slope, slope 10-90% and amplitude of fEPSP while, in LPS-N. sativa group these parameters increased compared to LPS group. The results indicated that the hydro-alcohol extract of N. sativa protected against synaptic plasticity and spatial learning and memory impairment induced by LPS in rats.
Collapse
Affiliation(s)
- Akbar Anaeigoudari
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran;
| | - Fatemeh Norouzi
- Department of Physiology, Esfarayan Faculty of Medical Sciences, Esfarayan, Iran;
| | - Azam Abareshi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran;
| | - Farimah Beheshti
- Department of Basic Sciences and Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; ,Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Azita Aaghaei
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran;
| | - Mohammad Naser Shafei
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran;
| | - Zahra Gholamnezhad
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran;
| | - Mahmoud Hosseini
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Correspondence: Mahmoud Hosseini. PhD Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
395
|
Hwang DS, Gu PS, Kim N, Jang YP, Oh MS. Effects of Rhei Undulati Rhizoma on lipopolysaccharide-induced neuroinflammation in vitro and in vivo. ENVIRONMENTAL TOXICOLOGY 2018; 33:23-31. [PMID: 28984087 DOI: 10.1002/tox.22463] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 06/07/2023]
Abstract
Neuroinflammation plays a critical role in the pathogenesis of degenerative brain diseases such as Alzheimer's disease and Parkinson's disease. Microglia are the major components of the brain immune system that regulate inflammatory processes. Activated microglia release pro-inflammatory factors and cytokines, resulting in neuronal cell death. We focused on inhibiting the activation of microglia from a stimulus as a strategy to search for neuroprotective drugs. Rhei Undulati Rhizoma (RUR) is traditionally used to treat various inflammatory disorders. In this study, we investigated whether RUR modulates inflammatory processes in lipopolysaccharide (LPS)-stimulated BV2 microglia cells and the mouse brain. RUR exerted anti-neuroinflammatory effects by inhibiting the production of nitric oxide and reactive oxygen species induced by LPS via the downregulation of transcription factors such as inducible nitric oxide synthase and cyclooxygenase-2 (COX-2) without causing cytotoxicity. RUR also regulated mitogen-activated protein kinase pathway by inhibiting phosphorylation of p38 and c-Jun N-terminal kinases and translocation of nuclear factor kappa B. Moreover, RUR attenuated LPS-induced glial activation and COX-2 expression in the substantia nigra and hippocampus of the mouse brain. These results indicate that RUR is a potential candidate to treat neurodegenerative diseases by regulating neuroinflammation.
Collapse
Affiliation(s)
- Deok-Sang Hwang
- Department of Oriental Gynecology, College of Oriental Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Pil Sung Gu
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Namkwon Kim
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Young Pyo Jang
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
396
|
Rey NL, Wesson DW, Brundin P. The olfactory bulb as the entry site for prion-like propagation in neurodegenerative diseases. Neurobiol Dis 2018; 109:226-248. [PMID: 28011307 PMCID: PMC5972535 DOI: 10.1016/j.nbd.2016.12.013] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/13/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023] Open
Abstract
Olfactory deficits are present in numerous neurodegenerative disorders and are accompanied by pathology in related brain regions. In several of these disorders, olfactory disturbances appear early and are considered as prodromal symptoms of the disease. In addition, pathological protein aggregates affect olfactory regions prior to other regions, suggesting that the olfactory system might be particularly vulnerable to neurodegenerative diseases. Exposed to the external environment, the olfactory epithelium and olfactory bulb allow pathogen and toxin penetration into the brain, a process that has been proposed to play a role in neurodegenerative diseases. Determining whether the olfactory bulb could be a starting point of pathology and of pathology spread is crucial to understanding how neurodegenerative diseases evolve. We argue that pathological changes following environmental insults contribute to the initiation of protein aggregation in the olfactory bulb, which then triggers the spread of the pathology within the brain by a templating mechanism in a prion-like manner. We review the evidence for the early involvement of olfactory structures in neurodegenerative diseases and the relationship between neuropathology and olfactory function. We discuss the vulnerability and putative underlying mechanisms by which pathology could be initiated in the olfactory bulb, from the entry of pathogens (promoted by increased permeability of the olfactory epithelium with aging or inflammation) to the sensitivity of the olfactory system to oxidative stress and inflammation. Finally, we review changes in protein expression and neural excitability triggered by pathogenic proteins that can promote pathogenesis in the olfactory bulb and beyond.
Collapse
Affiliation(s)
- Nolwen L Rey
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA.
| | - Daniel W Wesson
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
397
|
González-Reyes RE, Nava-Mesa MO, Vargas-Sánchez K, Ariza-Salamanca D, Mora-Muñoz L. Involvement of Astrocytes in Alzheimer's Disease from a Neuroinflammatory and Oxidative Stress Perspective. Front Mol Neurosci 2017; 10:427. [PMID: 29311817 PMCID: PMC5742194 DOI: 10.3389/fnmol.2017.00427] [Citation(s) in RCA: 352] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022] Open
Abstract
Alzheimer disease (AD) is a frequent and devastating neurodegenerative disease in humans, but still no curative treatment has been developed. Although many explicative theories have been proposed, precise pathophysiological mechanisms are unknown. Due to the importance of astrocytes in brain homeostasis they have become interesting targets for the study of AD. Changes in astrocyte function have been observed in brains from individuals with AD, as well as in AD in vitro and in vivo animal models. The presence of amyloid beta (Aβ) has been shown to disrupt gliotransmission, neurotransmitter uptake, and alter calcium signaling in astrocytes. Furthermore, astrocytes express apolipoprotein E and are involved in the production, degradation and removal of Aβ. As well, changes in astrocytes that precede other pathological characteristics observed in AD, point to an early contribution of astroglia in this disease. Astrocytes participate in the inflammatory/immune responses of the central nervous system. The presence of Aβ activates different cell receptors and intracellular signaling pathways, mainly the advanced glycation end products receptor/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway, responsible for the transcription of pro-inflammatory cytokines and chemokines in astrocytes. The release of these pro-inflammatory agents may induce cellular damage or even stimulate the production of Aβ in astrocytes. Additionally, Aβ induces the appearance of oxidative stress (OS) and production of reactive oxygen species and reactive nitrogen species in astrocytes, affecting among others, intracellular calcium levels, NADPH oxidase (NOX), NF-κB signaling, glutamate uptake (increasing the risk of excitotoxicity) and mitochondrial function. Excessive neuroinflammation and OS are observed in AD, and astrocytes seem to be involved in both. The Aβ/NF-κB interaction in astrocytes may play a central role in these inflammatory and OS changes present in AD. In this paper, we also discuss therapeutic measures highlighting the importance of astrocytes in AD pathology. Several new therapeutic approaches involving phenols (curcumin), phytoestrogens (genistein), neuroesteroids and other natural phytochemicals have been explored in astrocytes, obtaining some promising results regarding cognitive improvements and attenuation of neuroinflammation. Novel strategies comprising astrocytes and aimed to reduce OS in AD have also been proposed. These include estrogen receptor agonists (pelargonidin), Bambusae concretio Salicea, Monascin, and various antioxidatives such as resveratrol, tocotrienol, anthocyanins, and epicatechin, showing beneficial effects in AD models.
Collapse
Affiliation(s)
- Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Mauricio O Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, Bogotá, Colombia
| | - Daniel Ariza-Salamanca
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Laura Mora-Muñoz
- Grupo de Investigación en Neurociencias (NeURos), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
398
|
Marottoli FM, Katsumata Y, Koster KP, Thomas R, Fardo DW, Tai LM. Peripheral Inflammation, Apolipoprotein E4, and Amyloid-β Interact to Induce Cognitive and Cerebrovascular Dysfunction. ASN Neuro 2017; 9:1759091417719201. [PMID: 28707482 PMCID: PMC5521356 DOI: 10.1177/1759091417719201] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cerebrovascular dysfunction is rapidly reemerging as a major process of Alzheimer’s disease (AD). It is, therefore, crucial to delineate the roles of AD risk factors in cerebrovascular dysfunction. While apolipoprotein E4 (APOE4), Amyloid-β (Aβ), and peripheral inflammation independently induce cerebrovascular damage, their collective effects remain to be elucidated. The goal of this study was to determine the interactive effect of APOE4, Aβ, and chronic repeated peripheral inflammation on cerebrovascular and cognitive dysfunction in vivo. EFAD mice are a well-characterized mouse model that express human APOE3 (E3FAD) or APOE4 (E4FAD) and overproduce human Aβ42 via expression of 5 Familial Alzheimer’s disease (5xFAD) mutations. Here, we utilized EFAD carriers [5xFAD+/−/APOE+/+ (EFAD+)] and noncarriers [5xFAD−/−/APOE+/+ (EFAD−)] to compare the effects of peripheral inflammation in the presence or absence of human Aβ overproduction. Low-level, chronic repeated peripheral inflammation was induced in EFAD mice via systemic administration of lipopolysaccharide (LPS; 0.5 mg/kg/wk i.p.) from 4 to 6 months of age. In E4FAD+ mice, peripheral inflammation caused cognitive deficits and lowered post-synaptic protein levels. Importantly, cerebrovascular deficits were observed in LPS-challenged E4FAD+ mice, including cerebrovascular leakiness, lower vessel coverage, and cerebral amyloid angiopathy-like Aβ deposition. Thus, APOE4, Aβ, and peripheral inflammation interact to induce cerebrovascular damage and cognitive deficits.
Collapse
Affiliation(s)
- Felecia M Marottoli
- 1 Department of Anatomy and Cell Biology, University of Illinois at Chicago, IL, USA
| | - Yuriko Katsumata
- 2 Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Kevin P Koster
- 1 Department of Anatomy and Cell Biology, University of Illinois at Chicago, IL, USA
| | - Riya Thomas
- 1 Department of Anatomy and Cell Biology, University of Illinois at Chicago, IL, USA
| | - David W Fardo
- 2 Department of Biostatistics, University of Kentucky, Lexington, KY, USA.,3 Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Leon M Tai
- 1 Department of Anatomy and Cell Biology, University of Illinois at Chicago, IL, USA
| |
Collapse
|
399
|
Peralta Ramos JM, Bussi C, Gaviglio EA, Arroyo DS, Baez NS, Rodriguez-Galan MC, Iribarren P. Type I IFNs Are Required to Promote Central Nervous System Immune Surveillance through the Recruitment of Inflammatory Monocytes upon Systemic Inflammation. Front Immunol 2017; 8:1666. [PMID: 29255461 PMCID: PMC5722985 DOI: 10.3389/fimmu.2017.01666] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/14/2017] [Indexed: 01/09/2023] Open
Abstract
Brain-resident microglia and peripheral migratory leukocytes play essential roles in shaping the immune response in the central nervous system. These cells activate and migrate in response to chemokines produced during active immune responses and may contribute to the progression of neuroinflammation. Herein, we addressed the participation of type I–II interferons in the response displayed by microglia and inflammatory monocytes to comprehend the contribution of these cytokines in the establishment and development of a neuroinflammatory process. Following systemic lipopolysaccharide (LPS) challenge, we found glial reactivity and an active recruitment of CD45hi leukocytes close to CD31+ vascular endothelial cells in circumventricular organs. Isolated CD11b+ CD45hi Ly6Chi Ly6G−-primed inflammatory monocytes were able to induce T cell proliferation, unlike CD11b+ CD45lo microglia. Moreover, ex vivo re-stimulation with LPS exhibited an enhancement of T cell proliferative response promoted by inflammatory monocytes. These myeloid cells also proved to be recruited in a type I interferon-dependent fashion as opposed to neutrophils, unveiling a role of these cytokines in their trafficking. Together, our results compares the phenotypic and functional features between tissue-resident vs peripheral recruited cells in an inflamed microenvironment, identifying inflammatory monocytes as key sentinels in a LPS-induced murine model of neuroinflammation.
Collapse
Affiliation(s)
- Javier María Peralta Ramos
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Claudio Bussi
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Emilia Andrea Gaviglio
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Daniela Soledad Arroyo
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Natalia Soledad Baez
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Cecilia Rodriguez-Galan
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Pablo Iribarren
- Centro de Investigación en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
400
|
Das S, Mishra K, Ganju L, Singh S. Andrographolide - A promising therapeutic agent, negatively regulates glial cell derived neurodegeneration of prefrontal cortex, hippocampus and working memory impairment. J Neuroimmunol 2017; 313:161-175. [DOI: 10.1016/j.jneuroim.2017.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/24/2017] [Accepted: 11/06/2017] [Indexed: 11/28/2022]
|