51
|
Park WH, Kang S, Piao Y, Pak CJ, Oh MS, Kim J, Kang MS, Pak YK. Ethanol extract of Bupleurum falcatum and saikosaponins inhibit neuroinflammation via inhibition of NF-κB. JOURNAL OF ETHNOPHARMACOLOGY 2015; 174:37-44. [PMID: 26231448 DOI: 10.1016/j.jep.2015.07.039] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/16/2015] [Accepted: 07/27/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The root of Bupleurum falcatum L. (BF) has been used in traditional Korean and Chinese medicines for over 2000 years to treat infections, fever, and chronic liver diseases. Among the many active compounds in BF ethanol extract (BFE), saikosaponins exert pharmacological activities including anti-inflammatory effects. Activated microglial cells release a variety of pro-inflammatory substances, leading to neuronal cell death and neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. The aim of the present study was to investigate the mechanism of the anti-neuroinflammatory effects of BFE using lipopolysaccharide (LPS)-stimulated microglial cells and LPS-intraperitoneal injected C57BL/6 mice. MATERIALS AND METHODS Dried roots of BF were extracted with 70% ethanol (tenfold volume) on a stirring plate for 24h at room temperature to prepare BFE. Pure saikosaponins (SB3, SB4, and SD) were prepared by solvent extraction and column chromatography fractionation. BV2 murine microglial cells were treated with BFE or saikosaponins for 4h and stimulated with LPS. Generation of nitric oxide (NO), inflammatory cytokines, and reactive oxygen species (ROS) from activated microglial cells were monitored. The effects of BFE on NF-κB activation were determined using RT-PCR, reporter assay, and immunostaining. The in vivo effects of BFE were also assessed by immunohistochemical staining of tissue sections from LPS-injected mouse brains. RESULTS Treatment with BFE or saikosaponins dose-dependently attenuated LPS-induced production of NO, iNOS mRNA, and ROS by 30-50%. They reduced LPS-mediated increases in the mRNA levels of IL-6, IL-1β, and TNF-α by approximately 30-70% without affecting cell viability, and decreased LPS-mediated NF-κB activity via reducing p65/RELA mRNA, transcriptional activity, and nuclear localization of NF-κB. BFE also reduced LPS-induced activation of microglia and astrocytes in the hippocampus and substantia nigra of LPS-injected mice. CONCLUSION Our data suggest that BFE may be effective for reducing neuroinflammation-mediated neurodegeneration through suppressing NF-κB-mediated inflammatory pathways.
Collapse
Affiliation(s)
- Wook Ha Park
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Sora Kang
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Ying Piao
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Christine Jeehye Pak
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Myung Sook Oh
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Jinwoong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Min Seo Kang
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Youngmi Kim Pak
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea.
| |
Collapse
|
52
|
Yao N, Wu Y, Zhou Y, Ju L, Liu Y, Ju R, Duan D, Xu Q. Lesion of the locus coeruleus aggravates dopaminergic neuron degeneration by modulating microglial function in mouse models of Parkinson׳s disease. Brain Res 2015; 1625:255-74. [PMID: 26342895 DOI: 10.1016/j.brainres.2015.08.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/11/2015] [Accepted: 08/25/2015] [Indexed: 02/06/2023]
Abstract
The degeneration of noradrenergic neurons in the locus coeruleus (LC) commonly occurs in patients with Parkinson's disease (PD), which is characterized by a selective injury of dopaminergic neurons in the substantia nigra (SN). The pathological impact of the LC on the SN in the disease is unknown. In the present study, we used a noradrenergic toxin, N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4), to deplete noradrenaline (NA) derived from the LC to explore its influence on degeneration or injury of dopaminergic neurons in the SN in mouse model produced by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or lipopolysaccharide (LPS). Our results demonstrated that lesion of the LC could change microglial function in the brain, which led to enhanced or prolonged expression of pro-inflammatory cytokines, diminished neurotrophic factors, and weakened ability of anti-oxidation in the SN. The in vitro experiments further confirmed that NA could reduce the inflammatory reaction of microglia. The selective injury of dopaminergic neurons by inflammation, however, was due to the inflammation in different brain regions rather than the depletion of NA. Our results indicate that the lesion in the LC is an important factor in promoting dopaminergic neuron degeneration by impacting the function of microglia in the midbrain.
Collapse
Affiliation(s)
- Ning Yao
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Yanhong Wu
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing 100069, China; Beijing Children׳s Hospital Affiliated to Capital Medical University, Beijing 100045, China
| | - Yan Zhou
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Lili Ju
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Yujun Liu
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Rongkai Ju
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Deyi Duan
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing 100069, China
| | - Qunyuan Xu
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
53
|
Iyer A, Brown L, Whitehead JP, Prins JB, Fairlie DP. Nutrient and immune sensing are obligate pathways in metabolism, immunity, and disease. FASEB J 2015; 29:3612-25. [PMID: 26065858 DOI: 10.1096/fj.15-271155] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 06/02/2015] [Indexed: 12/13/2022]
Abstract
The growth and survival of multicellular organisms depend upon their abilities to acquire and metabolize nutrients, efficiently store and harness energy, and sense and fight infection. Systems for sensing and using nutrients have consequently coevolved alongside systems for sensing and responding to danger signals, including pathogens, and share many of the same cell signaling proteins and networks. Diets rich in carbohydrates and fats can overload these systems, leading to obesity, metabolic dysfunction, impaired immunity, and cardiovascular disease. Excessive nutrient intake promotes adiposity, typically altering adipocyte function and immune cell distribution, both of which trigger metabolic dysfunction. Here, we discuss novel mechanistic links between metabolism and immunity that underlie metabolic dysfunction in obesity. We aim to stimulate debate about how the endocrine and immune systems are connected through autocrine, paracrine, and neuroendocrine signaling in sophisticated networks that are only now beginning to be resolved. Understanding the expression and action of signaling proteins, together with modulating their receptors or pattern recognition using agonists or antagonists, will enable rational intervention in immunometabolism that may lead to novel treatments for obesity and metabolic dysfunction.
Collapse
Affiliation(s)
- Abishek Iyer
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - Lindsay Brown
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - Jonathan P Whitehead
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - Johannes B Prins
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - David P Fairlie
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| |
Collapse
|
54
|
Cardoso FL, Herz J, Fernandes A, Rocha J, Sepodes B, Brito MA, McGavern DB, Brites D. Systemic inflammation in early neonatal mice induces transient and lasting neurodegenerative effects. J Neuroinflammation 2015; 12:82. [PMID: 25924675 PMCID: PMC4440597 DOI: 10.1186/s12974-015-0299-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/10/2015] [Indexed: 12/11/2022] Open
Abstract
Background The inflammatory mediator lipopolysaccharide (LPS) has been shown to induce acute gliosis in neonatal mice. However, the progressive effects on the murine neurodevelopmental program over the week that follows systemic inflammation are not known. Thus, we investigated the effects of repeated LPS administration in the first postnatal week in mice, a condition mimicking sepsis in late preterm infants, on the developing central nervous system (CNS). Methods Systemic inflammation was induced by daily intraperitoneal administration (i.p.) of LPS (6 mg/kg) in newborn mice from postnatal day (PND) 4 to PND6. The effects on neurodevelopment were examined by staining the white matter and neurons with Luxol Fast Blue and Cresyl Violet, respectively. The inflammatory response was assessed by quantifying the expression/activity of matrix metalloproteinases (MMP), toll-like receptor (TLR)-4, high mobility group box (HMGB)-1, and autotaxin (ATX). In addition, B6 CX3CR1gfp/+ mice combined with cryo-immunofluorescence were used to determine the acute, delayed, and lasting effects on myelination, microglia, and astrocytes. Results LPS administration led to acute body and brain weight loss as well as overt structural changes in the brain such as cerebellar hypoplasia, neuronal loss/shrinkage, and delayed myelination. The impaired myelination was associated with alterations in the proliferation and differentiation of NG2 progenitor cells early after LPS administration, rather than with excessive phagocytosis by CNS myeloid cells. In addition to disruptions in brain architecture, a robust inflammatory response to LPS was observed. Quantification of inflammatory biomarkers revealed decreased expression of ATX with concurrent increases in HMGB1, TLR-4, and MMP-9 expression levels. Acute astrogliosis (GFAP+ cells) in the brain parenchyma and at the microvasculature interface together with parenchymal microgliosis (CX3CR1+ cells) were also observed. These changes preceded the migration/proliferation of CX3CR1+ cells around the vessels at later time points and the subsequent loss of GFAP+ astrocytes. Conclusion Collectively, our study has uncovered a complex innate inflammatory reaction and associated structural changes in the brains of neonatal mice challenged peripherally with LPS. These findings may explain some of the neurobehavioral abnormalities that develop following neonatal sepsis.
Collapse
Affiliation(s)
- Filipa L Cardoso
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Jasmin Herz
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892-1430, USA.
| | - Adelaide Fernandes
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal. .,Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - João Rocha
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Bruno Sepodes
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Maria A Brito
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal. .,Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Dorian B McGavern
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892-1430, USA.
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal. .,Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
55
|
Mezzapesa A, Orset C, Plawinski L, Doeuvre L, Martinez de Lizarrondo S, Chimienti G, Vivien D, Mansour A, Matà S, Pepe G, Anglés-Cano E. Plasminogen in cerebrospinal fluid originates from circulating blood. J Neuroinflammation 2014; 11:154. [PMID: 25220760 PMCID: PMC4173110 DOI: 10.1186/s12974-014-0154-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/14/2014] [Indexed: 01/20/2023] Open
Abstract
Background Plasminogen activation is a ubiquitous source of fibrinolytic and proteolytic activity. Besides its role in prevention of thrombosis, plasminogen is involved in inflammatory reactions in the central nervous system. Plasminogen has been detected in the cerebrospinal fluid (CSF) of patients with inflammatory diseases; however, its origin remains controversial, as the blood–CSF barrier may restrict its diffusion from blood. Methods We investigated the origin of plasminogen in CSF using Alexa Fluor 488–labelled rat plasminogen injected into rats with systemic inflammation and blood–CSF barrier dysfunction provoked by lipopolysaccharide (LPS). Near-infrared fluorescence imaging and immunohistochemistry fluorescence microscopy were used to identify plasminogen in brain structures, its concentration and functionality were determined by Western blotting and a chromogenic substrate assay, respectively. In parallel, plasminogen was investigated in CSF from patients with Guillain-Barré syndrome (n = 15), multiple sclerosis (n = 19) and noninflammatory neurological diseases (n = 8). Results Endogenous rat plasminogen was detected in higher amounts in the CSF and urine of LPS-treated animals as compared to controls. In LPS-primed rats, circulating Alexa Fluor 488–labelled rat plasminogen was abundantly localized in the choroid plexus, CSF and urine. Plasminogen in human CSF was higher in Guillain-Barré syndrome (median = 1.28 ng/μl (interquartile range (IQR) = 0.66 to 1.59)) as compared to multiple sclerosis (median = 0.3 ng/μl (IQR = 0.16 to 0.61)) and to noninflammatory neurological diseases (median = 0.27 ng/μl (IQR = 0.18 to 0.35)). Conclusions Our findings demonstrate that plasminogen is transported from circulating blood into the CSF of rats via the choroid plexus during inflammation. Our data suggest that a similar mechanism may explain the high CSF concentrations of plasminogen detected in patients with inflammation-derived CSF barrier impairment. Electronic supplementary material The online version of this article (doi:10.1186/s12974-014-0154-y) contains supplementary material, which is available to authorized users.
Collapse
|
56
|
Lee EJ, Han JE, Woo MS, Shin JA, Park EM, Kang JL, Moon PG, Baek MC, Son WS, Ko YT, Choi JW, Kim HS. Matrix metalloproteinase-8 plays a pivotal role in neuroinflammation by modulating TNF-α activation. THE JOURNAL OF IMMUNOLOGY 2014; 193:2384-93. [PMID: 25049354 DOI: 10.4049/jimmunol.1303240] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Matrix metalloproteinases (MMPs) play important roles in normal brain development and synaptic plasticity, although aberrant expression of MMPs leads to brain damage, including blood-brain barrier disruption, inflammation, demyelination, and neuronal cell death. In this article, we report that MMP-8 is upregulated in LPS-stimulated BV2 microglial cells and primary cultured microglia, and treatment of MMP-8 inhibitor (M8I) or MMP-8 short hairpin RNA suppresses proinflammatory molecules, particularly TNF-α secretion. Subsequent experiments showed that MMP-8 exhibits TNF-α-converting enzyme (TACE) activity by cleaving the prodomain of TNF-α (A(74)/Q(75), A(76)/V(77) residues) and, furthermore, that M8I inhibits TACE activity more efficiently than TAPI-0, a general TACE inhibitor. Biochemical analysis of the underlying anti-inflammatory mechanisms of M8I revealed that it inhibits MAPK phosphorylation, NF-κB/AP-1 activity, and reactive oxygen species production. Further support for the proinflammatory role of microglial MMP-8 was obtained from an in vivo animal model of neuroinflammatory disorder. MMP-8 is upregulated in septic conditions, particularly in microglia. Administration of M8I or MMP-8 short hairpin RNA significantly inhibits microglial activation and expression/secretion of TNF-α in brain tissue, serum, and cerebrospinal fluid of LPS-induced septic mice. These results demonstrate that MMP-8 critically mediates microglial activation by modulating TNF-α activity, which may explain neuroinflammation in septic mouse brain.
Collapse
Affiliation(s)
- Eun-Jung Lee
- Department of Molecular Medicine and Global Top 5 Research Program, Tissue Injury Defense Research Center, Ewha Womans University Medical School, Seoul 158-710, Republic of Korea
| | - Jeong Eun Han
- College of Pharmacy, Gachon University, Incheon 406-799, Republic of Korea
| | - Moon-Sook Woo
- Department of Molecular Medicine and Global Top 5 Research Program, Tissue Injury Defense Research Center, Ewha Womans University Medical School, Seoul 158-710, Republic of Korea
| | - Jin A Shin
- Department of Pharmacology, Ewha Womans University Medical School, Seoul 158-710, Republic of Korea
| | - Eun-Mi Park
- Department of Pharmacology, Ewha Womans University Medical School, Seoul 158-710, Republic of Korea
| | - Jihee Lee Kang
- Department of Physiology, Ewha Womans University Medical School, Seoul 158-710, Republic of Korea
| | - Pyong Gon Moon
- Department of Molecular Medicine, Kyongbuk National University, Daegu 700-842, Republic of Korea; and
| | - Moon-Chang Baek
- Department of Molecular Medicine, Kyongbuk National University, Daegu 700-842, Republic of Korea; and
| | - Woo-Sung Son
- College of Pharmacy, CHA University, Pocheon-si 487-010, Republic of Korea
| | - Young Tag Ko
- College of Pharmacy, Gachon University, Incheon 406-799, Republic of Korea
| | - Ji Woong Choi
- College of Pharmacy, Gachon University, Incheon 406-799, Republic of Korea;
| | - Hee-Sun Kim
- Department of Molecular Medicine and Global Top 5 Research Program, Tissue Injury Defense Research Center, Ewha Womans University Medical School, Seoul 158-710, Republic of Korea;
| |
Collapse
|
57
|
Shin JW, Cheong YJ, Koo YM, Kim S, Noh CK, Son YH, Kang C, Sohn NW. α-Asarone Ameliorates Memory Deficit in Lipopolysaccharide-Treated Mice via Suppression of Pro-Inflammatory Cytokines and Microglial Activation. Biomol Ther (Seoul) 2014; 22:17-26. [PMID: 24596617 PMCID: PMC3936426 DOI: 10.4062/biomolther.2013.102] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/07/2014] [Accepted: 01/10/2014] [Indexed: 12/31/2022] Open
Abstract
α-Asarone exhibits a number of pharmacological actions including neuroprotective, anti-oxidative, anticonvulsive, and cognitive enhancing action. The present study investigated the effects of α-asarone on pro-inflammatory cytokines mRNA, microglial activation, and neuronal damage in the hippocampus and on learning and memory deficits in systemic lipopolysaccharide (LPS)-treated C57BL/6 mice. Varying doses of α-asarone was orally administered (7.5, 15, or 30 mg/kg) once a day for 3 days before the LPS (3 mg/kg) injection. α-Asarone significantly reduced TNF-α and IL-1β mRNA at 4 and 24 hours after the LPS injection at dose of 30 mg/kg. At 24 hours after the LPS injection, the loss of CA1 neurons, the increase of TUNEL-labeled cells, and the up-regulation of BACE1 expression in the hippocampus were attenuated by 30 mg/kg of α-asarone treatment. α-Asarone significantly reduced Iba1 protein expression in the hippocampal tissue at a dose of 30 mg/kg. α-Asarone did not reduce the number of Iba1-expressing microglia on immunohistochemistry but the average cell size and percentage areas of Iba1-expressing microglia in the hippocampus were significantly decreased by 30 mg/kg of α-asarone treatment. In the Morris water maze test, α-asarone significantly prolonged the swimming time spent in the target and peri-target zones. α-Asarone also significantly increased the number of target heading and memory score in the Morris water maze. The results suggest that inhibition of pro-inflammatory cytokines and microglial activation in the hippocampus by α-asarone may be one of the mechanisms for the α-asarone-mediated ameliorating effect on memory deficits.
Collapse
Affiliation(s)
- Jung-Won Shin
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Republic of Korea
| | - Young-Jin Cheong
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Republic of Korea
| | - Yong-Mo Koo
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Republic of Korea
| | - Sooyong Kim
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Republic of Korea
| | - Chung-Ku Noh
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Republic of Korea
| | - Young-Ha Son
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Republic of Korea
| | - Chulhun Kang
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Republic of Korea
| | - Nak-Won Sohn
- Department of East-West Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 446-701, Republic of Korea
| |
Collapse
|
58
|
Glycyrrhizin alleviates neuroinflammation and memory deficit induced by systemic lipopolysaccharide treatment in mice. Molecules 2013; 18:15788-803. [PMID: 24352029 PMCID: PMC6269849 DOI: 10.3390/molecules181215788] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/09/2013] [Accepted: 12/10/2013] [Indexed: 12/18/2022] Open
Abstract
The present study investigated the effects of glycyrrhizin (GRZ) on neuroinflammation and memory deficit in systemic lipopolysaccharide (LPS)-treated C57BL/6 mice. Varying doses of GRZ was orally administered (10, 30, or 50 mg/kg) once a day for 3 days before the LPS (3 mg/kg) injection. At 24 h after the LPS injection, GRZ significantly reduced TNF-α and IL-1β mRNA at doses of 30 and 50 mg/kg. COX-2 and iNOS protein expressions were significantly reduced by GRZ at doses of 30 and 50 mg/kg. In the Morris water maze test, GRZ (30 mg/kg) significantly prolonged the swimming time spent in the target and peri-target zones. GRZ also significantly increased the target heading and memory score numbers. In the hippocampal tissue, GRZ significantly reduced the up-regulated Iba1 protein expression and the average cell size of Iba1-expressing microglia induced by LPS. The results indicate that GRZ ameliorated the memory deficit induced by systemic LPS treatment and the effect of GRZ was found to be mediated through the inhibition of pro-inflammatory mediators and microglial activation in the brain tissue. This study supports that GRZ may be a putative therapeutic drug on neurodegenerative diseases associated with cognitive deficits and neuroinflammation such as Alzheimer's disease.
Collapse
|
59
|
Choi MS, Kwak HJ, Kweon KJ, Hwang JM, Shin JW, Sohn NW. Effects of β-Asarone on Pro-Inflammatory Cytokines and Learning and Memory Impairment in Lipopolysaccharide-Treated Mice. ACTA ACUST UNITED AC 2013. [DOI: 10.6116/kjh.2013.28.6.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
60
|
Shin JW, Ma SH, Lee JW, Kim DK, Do K, Sohn NW. Ginsenoside Rg1 Attenuates Neuroinflammation Following Systemic Lipopolysaccharide Treatment in Mice. ACTA ACUST UNITED AC 2013. [DOI: 10.6116/kjh.2013.28.6.145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
61
|
Zhang Z, Hou L, Song JL, Song N, Sun YJ, Lin X, Wang XL, Zhang FZ, Ge YL. Pro-inflammatory cytokine-mediated ferroportin down-regulation contributes to the nigral iron accumulation in lipopolysaccharide-induced Parkinsonian models. Neuroscience 2013; 257:20-30. [PMID: 24183966 DOI: 10.1016/j.neuroscience.2013.09.037] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 09/10/2013] [Accepted: 09/19/2013] [Indexed: 12/21/2022]
Abstract
Pro-inflammatory cytokines induced by inflammation and iron accumulation in the substantia nigra (SN) have been implicated in the pathogenesis of Parkinson's disease (PD). In the present study, we aimed to investigate the relationship between inflammation and iron accumulation in a lipopolysaccharide (LPS)-induced Parkinsonian rat model. The activation of glial cells and elevated levels of pro-inflammatory cytokines were observed in the SN of LPS models, accompanied by iron deposits in the same region. Moreover, ferroportin (Fpn), the only channel for iron export, was down-regulated. SH-SY5Y dopaminergic cells were pre-incubated with conditioned media enriched in pro-inflammatory cytokines, and abnormal iron deposits and a drop of Fpn were observed. The expression of heme oxygenase-1 (HO-1) was also upregulated in vivo and in vitro. These results suggested that pro-inflammatory cytokines might induce Fpn downregulation, which leads to iron accumulation and dopaminergic neurons' degeneration in PD. HO-1 may also contribute to the iron accumulation in neurons, but its mechanism needs to be further investigated.
Collapse
Affiliation(s)
- Z Zhang
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, China
| | - L Hou
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, China.
| | - J-L Song
- Laboratory Department of Maternal and Child Health Hospital of Qingdao, China
| | - N Song
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Y-J Sun
- Laboratory Department of Qingdao Center Hospital, Qingdao, China
| | - X Lin
- Experiment Center of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, China
| | - X-L Wang
- Experiment Center of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, China
| | - F-Z Zhang
- Nutrition Department of Liaocheng People's Hospital, Shandong Province, China
| | - Y-L Ge
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, China
| |
Collapse
|
62
|
Kim KK, Jin SH, Lee BJ. Herpes virus entry mediator signaling in the brain is imperative in acute inflammation-induced anorexia and body weight loss. Endocrinol Metab (Seoul) 2013; 28:214-20. [PMID: 24396681 PMCID: PMC3811702 DOI: 10.3803/enm.2013.28.3.214] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 07/19/2013] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Reduced appetite and body weight loss are typical symptoms of inflammatory diseases. A number of inflammatory stimuli are responsible for the imbalance in energy homeostasis, leading to metabolic disorders. The herpes virus entry mediator (HVEM) protein plays an important role in the development of various inflammatory diseases, such as intestinal inflammation and diet-induced obesity. However, the role of HVEM in the brain is largely unknown. This study aims to investigate whether HVEM signaling in the brain is involved in inflammation-induced anorexia and body weight loss. METHODS Food intake and body weight were measured at 24 hours after intraperitoneal injection of lipopolysaccharide (LPS) or intracerebroventricular injection of recombinant mouse LIGHT (also called tumor necrosis factor receptor superfamily 14, TNFSF14), an HVEM ligand, into 8- to 10-week-old male C57BL/6 mice and mice lacking HVEM expression (HVEM-/-). We also assessed LPS-induced change in hypothalamic expression of HVEM using immunohistochemistry. RESULTS Administration of LPS significantly reduced food intake and body weight, and moreover, increased expression of HVEM in the hypothalamic arcuate nucleus. However, LPS induced only minor decreases in food intake and body weight in HVEM-/- mice. Administration of LIGHT into the brain was very effective at decreasing food intake and body weight in wild-type mice, but was less effective in HVEM-/- mice. CONCLUSION Activation of brain HVEM signaling is responsible for inflammation-induced anorexia and body weight loss.
Collapse
Affiliation(s)
- Kwang Kon Kim
- Department of Biological Sciences, University of Ulsan College of Natural Sciences, Ulsan, Korea
| | - Sung Ho Jin
- Department of Biological Sciences, University of Ulsan College of Natural Sciences, Ulsan, Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan College of Natural Sciences, Ulsan, Korea
| |
Collapse
|
63
|
Nigrostriatal damage after systemic rotenone and/or lipopolysaccharide and the effect of cannabis. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s00580-013-1788-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
64
|
Jeong HK, Ji K, Min K, Joe EH. Brain inflammation and microglia: facts and misconceptions. Exp Neurobiol 2013; 22:59-67. [PMID: 23833554 PMCID: PMC3699675 DOI: 10.5607/en.2013.22.2.59] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 06/17/2013] [Accepted: 06/17/2013] [Indexed: 11/19/2022] Open
Abstract
THE INFLAMMATION THAT ACCOMPANIES ACUTE INJURY HAS DUAL FUNCTIONS: bactericidal action and repair. Bactericidal functions protect damaged tissue from infection, and repair functions are initiated to aid in the recovery of damaged tissue. Brain injury is somewhat different from injuries in other tissues in two respects. First, many cases of brain injury are not accompanied by infection: there is no chance of pathogens to enter in ischemia or even in traumatic injury if the skull is intact. Second, neurons are rarely regenerated once damaged. This raises the question of whether bactericidal inflammation really occurs in the injured brain; if so, how is this type of inflammation controlled? Many brain inflammation studies have been conducted using cultured microglia (brain macrophages). Even where animal models have been used, the behavior of microglia and neurons has typically been analyzed at or after the time of neuronal death, a time window that excludes the inflammatory response, which begins immediately after the injury. Therefore, to understand the patterns and roles of brain inflammation in the injured brain, it is necessary to analyze the behavior of all cell types in the injured brain immediately after the onset of injury. Based on our experience with both in vitro and in vivo experimental models of brain inflammation, we concluded that not only microglia, but also astrocytes, blood inflammatory cells, and even neurons participate and/or regulate brain inflammation in the injured brain. Furthermore, brain inflammation played by these cells protects neurons and repairs damaged microenvironment but not induces neuronal damage.
Collapse
Affiliation(s)
- Hey-Kyeong Jeong
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 442-721, Korea. ; Department of Pharmacology, Ajou University School of Medicine, Suwon 442-721, Korea. ; Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 442-721, Korea. ; National Research Lab of Brain Inflammation, Ajou University School of Medicine, Suwon 442-721, Korea
| | | | | | | |
Collapse
|
65
|
Halder SK, Matsunaga H, Ishii KJ, Akira S, Miyake K, Ueda H. Retinal cell type-specific prevention of ischemia-induced damages by LPS-TLR4 signaling through microglia. J Neurochem 2013; 126:243-60. [DOI: 10.1111/jnc.12262] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 04/04/2013] [Accepted: 04/08/2013] [Indexed: 11/27/2022]
Affiliation(s)
- Sebok K. Halder
- Department of Molecular Pharmacology and Neuroscience; Nagasaki University Graduate School of Biomedical Sciences; Nagasaki Japan
| | - Hayato Matsunaga
- Department of Molecular Pharmacology and Neuroscience; Nagasaki University Graduate School of Biomedical Sciences; Nagasaki Japan
| | - Ken J. Ishii
- Laboratory of Vaccine Science; WPI Immunology Frontier Research Center; Osaka University, Osaka Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center; Department of Host Defense; Research Institute for Microbial Diseases; Osaka University; Osaka Japan
| | - Kensuke Miyake
- Division of Innate Immunity; The Institute of Medical Science; University of Tokyo; Tokyo Japan
| | - Hiroshi Ueda
- Department of Molecular Pharmacology and Neuroscience; Nagasaki University Graduate School of Biomedical Sciences; Nagasaki Japan
| |
Collapse
|
66
|
Jafri A, Belkadi A, Zaidi SIA, Getsy P, Wilson CG, Martin RJ. Lung inflammation induces IL-1β expression in hypoglossal neurons in rat brainstem. Respir Physiol Neurobiol 2013; 188:21-8. [PMID: 23648475 DOI: 10.1016/j.resp.2013.04.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/25/2013] [Accepted: 04/26/2013] [Indexed: 10/26/2022]
Abstract
Perinatal inflammation is associated with respiratory morbidity. Immune modulation of brainstem respiratory control centers may provide a link for this pathobiology. We exposed 11-day old rats to intratracheal lipopolysaccharide (LPS, 0.5 μg/g) to test the hypothesis that intrapulmonary inflammation increases expression of the proinflammatory cytokine IL-1β within respiratory-related brainstem regions. Intratracheal LPS resulted in a 32% increase in IL-1β protein expression in the medulla oblongata. In situ hybridization showed increased intensity of IL-1β mRNA but no change in neuronal numbers. Co-localization experiments showed that hypoglossal neurons express IL-1β mRNA and immunostaining showed a 43% increase in IL-1β protein-expressing cells after LPS exposure. LPS treatment also significantly increased microglial cell numbers though they did not express IL-1β mRNA. LPS-induced brainstem expression of neuronal IL-1β mRNA and protein may have implications for our understanding of the vulnerability of neonatal respiratory control in response to a peripheral proinflammatory stimulus.
Collapse
Affiliation(s)
- Anjum Jafri
- Department of Pediatrics, Division of Neonatology, Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | | | | | | | | | | |
Collapse
|
67
|
Effect of Cannabis sativa on oxidative stress and organ damage after systemic endotoxin administration in mice. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s00580-013-1745-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
68
|
Beynon AL, Brown MR, Wright R, Rees MI, Sheldon IM, Davies JS. Ghrelin inhibits LPS-induced release of IL-6 from mouse dopaminergic neurones. J Neuroinflammation 2013; 10:40. [PMID: 23509933 PMCID: PMC3614890 DOI: 10.1186/1742-2094-10-40] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 01/23/2013] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Ghrelin is an orexigenic stomach hormone that acts centrally to increase mid-brain dopamine neurone activity, amplify dopamine signaling and protect against neurotoxin-induced dopamine cell death in the mouse substantia nigra pars compacta (SNpc). In addition, ghrelin inhibits the lipopolysaccharide (LPS)-induced release of pro-inflammatory cytokines from peripheral macrophages, T-cells and from LPS stimulated microglia. Here we sought to determine whether ghrelin attenuates pro-inflammatory cytokine release from dopaminergic neurones. FINDINGS The dopaminergic SN4741 cell-line, which derives from the mouse substantia nigra (SN) and expresses the ghrelin-receptor (growth hormone secretagogue receptor (GHS-R)) and the ghrelin-O-acyl transferase (GOAT) enzyme, was used to determine the neuro-immunomodulatory action of ghrelin. We induced innate immune activation via LPS challenge (1 μg/ml) of SN4741 neurones that had been pre-cultured in the presence or absence of ghrelin (1, 10, 100 nM) for 4 h. After 24 h supernatants were collected for detection of IL-1 beta (IL-1β ), TNF alpha (TNF-α) and IL-6 cytokines via enzyme linked immunosorbent assay (ELISA) analysis. Nuclear translocation of the transcription factor nuclear factor kappa B (NF-κB) was analyzed by Western blotting, and to determine viability of treatments a cell viability assay and caspase-3 immunohistochemistry were performed.We provide evidence that while IL-1β and TNF-α were not detectable under any conditions, SN4741 neurones constitutively released IL-6 under basal conditions and treatment with LPS significantly increased IL-6 secretion. Pre-treatment of neurones with ghrelin attenuated LPS-mediated IL-6 release at 24 h, an affect that was inhibited by the GHS-R antagonist [D-Lys3]-GHRP-6. However, while ghrelin pre-treatment attenuated the LPS-mediated increase in NF-κB, there was no alteration in its nuclear translocation. Cell viability assay and caspase-3 immunocytochemistry demonstrated that the results were independent from activation of cytotoxic and/or apoptotic mechanisms in the neuronal population, respectively. CONCLUSION Our results provide evidence that the gut-hormone, ghrelin, attenuates IL-6 secretion to LPS challenge in mid-brain dopaminergic neurones. These data suggest that ghrelin may protect against dopaminergic SN nerve cell damage or death via modulation of the innate immune response.
Collapse
Affiliation(s)
- Amy L Beynon
- Molecular Neuroscience, Swansea University, Swansea, SA28PP, UK
| | | | | | | | | | | |
Collapse
|
69
|
Roy-Lacroix MÈ, Guérard M, Berthiaume M, Rola-Pleszczynski M, Crous-Tsanaclis AM, Pasquier JC. Time-dependent effect ofin uteroinflammation: a longitudinal study in rats. J Matern Fetal Neonatal Med 2013; 26:789-94. [DOI: 10.3109/14767058.2012.755164] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
70
|
Lee YH, Jeon SH, Kim SH, Kim C, Lee SJ, Koh D, Lim Y, Ha K, Shin SY. A new synthetic chalcone derivative, 2-hydroxy-3',5,5'-trimethoxychalcone (DK-139), suppresses the Toll-like receptor 4-mediated inflammatory response through inhibition of the Akt/NF-κB pathway in BV2 microglial cells. Exp Mol Med 2012; 44:369-77. [PMID: 22382990 PMCID: PMC3389075 DOI: 10.3858/emm.2012.44.6.042] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Microglial cells are the resident innate immune cells that sense pathogens and tissue injury in the central nervous system (CNS). Microglial activation is critical for neuroinflammatory responses. The synthetic compound 2-hydroxy-3',5,5'-trimethoxychalcone (DK-139) is a novel chalcone-derived compound. In this study, we investigated the effects of DK-139 on Toll-like receptor 4 (TLR4)-mediated inflammatory responses in BV2 microglial cells. DK-139 inhibited lipopolysaccharide (LPS)-induced TLR4 activity, as determined using a cell-based assay. DK-139 blocked LPS-induced phosphorylation of IκB and p65/RelA NF-κB, resulting in inhibition of the nuclear translocation and trans-acting activity of NF-κB in BV2 microglial cells. We also found that DK-139 reduced the expression of NF-κB target genes, such as those for COX-2, iNOS, and IL-1β, in LPS-stimulated BV2 microglial cells. Interestingly, DK-139 blocked LPS-induced Akt phosphorylation. Inhibition of Akt abrogated LPS-induced phosphorylation of p65/RelA, while overexpression of dominant-active p110CAAX enhanced p65/RelA phosphorylation as well as iNOS and COX2 expression. These results suggest that DK-139 exerts an anti-inflammatory effect on microglial cells by inhibiting the Akt/IκB kinase (IKK)/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Young Han Lee
- Department of Biomedical Science and Technology, Research Center for Transcription Control, SMART Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Lee JS, Song JH, Sohn NW, Shin JW. Inhibitory Effects of Ginsenoside Rb1 on Neuroinflammation Following Systemic Lipopolysaccharide Treatment in Mice. Phytother Res 2012; 27:1270-6. [DOI: 10.1002/ptr.4852] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 08/29/2012] [Accepted: 09/02/2012] [Indexed: 12/26/2022]
Affiliation(s)
- Joon-Suk Lee
- Department of Oriental Medical Science, Graduate School of East-west Medical Science; Kyung Hee University; Yongin; 446-701; South Korea
| | | | | | | |
Collapse
|
72
|
Lee YJ, Choi DY, Yun YP, Han SB, Kim HM, Lee K, Choi SH, Yang MP, Jeon HS, Jeong JH, Oh KW, Hong JT. Ethanol Extract of Magnolia officinalis
Prevents Lipopolysaccharide-Induced Memory Deficiency via Its Antineuroinflammatory and Antiamyloidogenic Effects. Phytother Res 2012; 27:438-47. [DOI: 10.1002/ptr.4740] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 04/25/2012] [Accepted: 04/26/2012] [Indexed: 12/13/2022]
Affiliation(s)
- Young-Jung Lee
- College of Pharmacy; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- Medical Research Center; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- CBITRC; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
| | - Dong-Young Choi
- College of Pharmacy; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- Medical Research Center; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- CBITRC; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
| | - Yeo-Pyo Yun
- College of Pharmacy; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- Medical Research Center; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- CBITRC; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
| | - Sang Bae Han
- College of Pharmacy; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- Medical Research Center; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- CBITRC; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
| | - Hwan Mook Kim
- College of Pharmacy; Gachon University of Medicine and Science; Incheon 406-799 Korea
| | - Kiho Lee
- College of Pharmacy; Korea University; Jochiwon Chungnam 339-700 Korea
| | - Seok Hwa Choi
- College of Veterinary Medicine; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
| | - Mhan-Pyo Yang
- College of Veterinary Medicine; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
| | - Hyun Soo Jeon
- Department of Obstetrics and Gynecology, School of Medicine; Konkuk University, Chungju Hospital; Chungju Korea
| | - Jea-Hwang Jeong
- Department of Biosciences and Biomedicine; Chungbuk Provincial College; Okcheongun Chungbuk 373-807 Korea
| | - Ki-Wan Oh
- College of Pharmacy; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
| | - Jin Tae Hong
- College of Pharmacy; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- Medical Research Center; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
- CBITRC; Chungbuk National University; 12 Gaesin-dong, Heungduk-gu Cheongju Chungbuk 361-763 Korea
| |
Collapse
|
73
|
Fan K, Wu X, Fan B, Li N, Lin Y, Yao Y, Ma J. Up-regulation of microglial cathepsin C expression and activity in lipopolysaccharide-induced neuroinflammation. J Neuroinflammation 2012; 9:96. [PMID: 22607609 PMCID: PMC3410810 DOI: 10.1186/1742-2094-9-96] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Accepted: 05/20/2012] [Indexed: 12/17/2022] Open
Abstract
Background Cathepsin C (Cat C) functions as a central coordinator for activation of many serine proteases in inflammatory cells. It has been recognized that Cat C is responsible for neutrophil recruitment and production of chemokines and cytokines in many inflammatory diseases. However, Cat C expression and its functional role in the brain under normal conditions or in neuroinflammatory processes remain unclear. Our previous study showed that Cat C promoted the progress of brain demyelination in cuprizone-treated mice. The present study further investigated the Cat C expression and activity in lipopolysaccharide (LPS)-induced neuroinflammation in vivo and in vitro. Methods C57BL/6 J mice were intraperitoneally injected with either 0.9% saline or lipopolysaccharide (LPS, 5 mg/kg). Immunohistochemistry (IHC) and in situ hybridization (ISH) were used to analyze microglial activation, TNF-α, IL-1β, IL-6, iNOS mRNAs expressions and cellular localization of Cat C in the brain. Nitrite assay was used to examine microglial activation in vitro; RT-PCR and ELISA were used to determine the expression and release of Cat C. Cat C activity was analyzed by cellular Cat C assay kit. Data were evaluated for statistical significance with paired t test. Results Cat C was predominantly expressed in hippocampal CA2 neurons in C57BL/6 J mice under normal conditions. Six hours after LPS injection, Cat C expression was detected in cerebral cortical neurons; whereas, twenty-four hours later, Cat C expression was captured in activated microglial cells throughout the entire brain. The duration of induced Cat C expression in neurons and in microglial cells was ten days and three days, respectively. In vitro, LPS, IL-1β and IL-6 treatments increased microglial Cat C expression in a dose-dependent manner and upregulated Cat C secretion and its activity. Conclusions Taken together, these data indicate that LPS and proinflammatory cytokines IL-1β, IL-6 induce the expression, release and upregulate enzymatic activity of Cat C in microglial cells. Further investigation is required to determine the functional role of Cat C in the progression of neuroinflammation, which may have implications for therapeutics for the prevention of neuroinflammation-involved neurological disorders in the future.
Collapse
Affiliation(s)
- Kai Fan
- Department of Anatomy, Dalian Medical University, No, 9, West Segment of South Lvshun Road, Dalian, Liaoning, 116044, China
| | | | | | | | | | | | | |
Collapse
|
74
|
Nasoohi S, Hemmati AA, Moradi F, Ahmadiani A. The γ-secretase blocker DAPT impairs recovery from lipopolysaccharide-induced inflammation in rat brain. Neuroscience 2012; 210:99-109. [PMID: 22445932 DOI: 10.1016/j.neuroscience.2012.02.051] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 02/28/2012] [Accepted: 02/29/2012] [Indexed: 11/15/2022]
Abstract
γ-Secretase is an important contributing enzyme in Alzheimer's disease and is therefore an important therapeutic target. However, the impact of γ-secretase inhibition is not well studied in acute neuroinflammation induced by systemic infection. In this study the influence of γ-secretase on the expression of some proinflammatory markers was assessed in the acute phase as well as the subsiding phase of neuroinflammation. Cerebral γ-secretase cleavage activity was measured by a fluorometric assay after lipopolysaccharide (LPS) intraperitoneal administration. Time profiles of TNF-α and COX-II expression were then determined to detect the time points relevant to the maximal inflammatory responses and the subsequent recovery phase. γ-Secretase activity coincident with TNF-α protein expression returned to its basal level till 8-12 h after systemic challenge with low dose LPS while COX-II over expression lasted for 48-72 h later. Pharmacological inhibition of γ-secretase with local or systemic administration of DAPT (N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester) was performed to indicate the results on the developmental and sinking phases of inflammatory responses in 6 and 72 h post LPS respectively. Our results demonstrate that both local and systemic modulation of γ-secretase hyper-activity with DAPT increase the duration of TNF-α, COX-II, and NFκB induction. We consistently found mild augmented apoptosis in animals treated with DAPT as determined by measuring cleaved caspase-3 expression and by TUNEL assay 72 h following LPS injection. These results suggest that γ-secretase modulation interferes with certain immune regulatory pathways which may restrict some inflammatory transcription factors such as NFκB.
Collapse
Affiliation(s)
- S Nasoohi
- Department of Pharmacology and Toxicology, School of Pharmacy and Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Golestan, PO Box 6287, Ahvaz, Iran
| | | | | | | |
Collapse
|
75
|
Abdel-Salam OM, Morsy SMY, Sleem AA. The effect of different antidepressant drugs on oxidative stress after lipopolysaccharide administration in mice. EXCLI JOURNAL 2011; 10:290-302. [PMID: 29033710 PMCID: PMC5611632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 12/06/2011] [Indexed: 10/25/2022]
Abstract
This study investigated the effect of the serotonin selective reuptake inhibitors (SSRIs) fluoxetine, sertraline, fluvoxamine and the tricyclic antidepressant (TCA) impiramine on oxidative stress in brain and liver induced by lipopolysaccharide administration in mice. Each drug was administered subcutaneously at doses of 10 or 20 mg/kg, for two days prior to intraperitoneal (i.p.) administration of lipopolysaccharide E (LPS: 200 µg/kg). Mice were euthanized 4 h after administration of the lipopolysaccharide. Lipid peroxidation (malondialdehyde; MDA), reduced glutathione (GSH) and nitric oxide (nitrite/nitrate) concentrations were measured in brain and liver. Results: The administration of lipopolysaccharide increased oxidative stress in brain and liver; it increased brain MDA by 36.1 and liver MDA by 159.8 %. GSH decreased by 34.1 % and 64.8 % and nitric oxide increased by 78.7 % and 103.8 % in brain and liver, respectively. In brain, MDA decreased after the administration of sertraline and by the lower dose of fluoxetine or fluvoxamine, but increased after the higher dose of imipramine. Reduced glutathione increased after sertraline, fluvoxamine and the lower dose of fluoxetine or imipramine. Nitric oxide decreased by sertraline, fluoxetine, fluvoxamine and by the lower dose of imipramine. In the liver, all drugs decreased MDA and increased GSH level. Nitric oxide is decreased by sertraline, fluvoxamine and by the lower dose of fluoxetine or imipramine. It is concluded that, during mild systemic inflammatory illness induced by peripheral bacterial endotoxin injection, the SSRIs fluoxetine, sertraline and fluvoxamine reduced, while the TCA impiramine increased oxidative stress induced in the brain. The SSRIs as well as imipramine reduced oxidative stress due to lipopolysaccharide in liver tissue.
Collapse
Affiliation(s)
- Omar M.E. Abdel-Salam
- Department of Toxicology and Narcotics, National Research Centre, Cairo,*To whom correspondence should be addressed: Omar M.E. Abdel-Salam, Department of Toxicology and Narcotics, National Research Centre, Tahrir St., Dokki, Cairo, Egypt; FAX: 202-33370931, E-mail:
| | | | - Amany A. Sleem
- Department of Pharmacology, National Research Centre, Cairo
| |
Collapse
|
76
|
Abdel-Salam OM, Mohammed NA, Sleem AA. The effects of trimetazidine on lipopolysaccharide-induced oxidative stress in mice. EXCLI JOURNAL 2011; 10:162-172. [PMID: 27857673 PMCID: PMC5109013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 10/03/2011] [Indexed: 11/28/2022]
Abstract
The effects of trimetazidine, a novel anti-ischemic agent, on the development of oxidative stress induced in mice with lipopolysaccharide endotoxin were investigated. The drug was administered orally once daily at doses of 1.8, 3.6 or 7.2 mg/kg for two days prior to intraperitoneal (i.p.) injection of lipopolysaccharide E (200 μg/kg) and at time of endotoxin administration. Mice were euthanized 4 h after administration of the lipopolysaccharide. Lipid peroxidation (malondialdehyde; MDA), reduced glutathione (GSH) and nitric oxide (nitrite/nitrate) concentrations were measured in brain and liver. The administration of lipopolysaccharide increased oxidative stress in both the brain and liver tissue. MDA increased by 33.9 and 107.1 %, GSH decreased by 23.9 and 84.3 % and nitric oxide increased 70.3 and 48.4 % in the brain and liver, respectively. Compared with the lipopolysaccharide control group, brain MDA decreased by 26.2 and 36.7 %, while GSH increased by 18.2 and 25.8 % after the administration of trimetazidine at 3.6 and 7.2 mg/kg, respectively. Brain nitric oxide decreased by 45.3, 50.8 and 57.0 % by trimetazidine at 1.8, 3.6 and 7.2 mg/kg, respectively. In the liver, MDA decreased by 18.7, 30.7 and 49.4 % and GSH increased by 150.3, 204.8 and 335.4 % following trimetazidine administration at 1.8, 3.6 and 7.2 mg/kg. Meanwhile, nitric oxide decreased by 17.3 % by 7.2 mg/kg of trimetazidine. These results indicate that administration of trimetazidine in the presence of mild systemic inflammatory response alleviates oxidative stress in the brain and liver.
Collapse
Affiliation(s)
- Omar M.E. Abdel-Salam
- Department of Toxicology and Narcotics, National Research Centre, Cairo,*To whom correspondence should be addressed: Omar M.E. Abdel-Salam, Department of Toxicology and Narcotics, National Research Centre, Dokki, Cairo/Egypt; FAX: 202-33370931, E-mail:
| | | | - Amany A. Sleem
- Department of Pharmacology, National Research Centre, Cairo
| |
Collapse
|
77
|
Abdel-Salam OME, Salem NA, Hussein JS. Effect of Aspartame on Oxidative Stress and Monoamine Neurotransmitter Levels in Lipopolysaccharide-Treated Mice. Neurotox Res 2011; 21:245-55. [DOI: 10.1007/s12640-011-9264-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 07/27/2011] [Accepted: 07/28/2011] [Indexed: 01/03/2023]
|