301
|
Lee SW, Gajavelli S, Spurlock MS, Andreoni C, de Rivero Vaccari JP, Bullock MR, Keane RW, Dietrich WD. Microglial Inflammasome Activation in Penetrating Ballistic-Like Brain Injury. J Neurotrauma 2018; 35:1681-1693. [PMID: 29439605 PMCID: PMC6016174 DOI: 10.1089/neu.2017.5530] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Penetrating traumatic brain injury (PTBI) is a significant cause of death and disability in the United States. Inflammasomes are one of the key regulators of the interleukin (IL)-1β mediated inflammatory responses after traumatic brain injury. However, the contribution of inflammasome signaling after PTBI has not been determined. In this study, adult male Sprague-Dawley rats were subjected to sham procedures or penetrating ballistic-like brain injury (PBBI) and sacrificed at various time-points. Tissues were assessed by immunoblot analysis for expression of IL-1β, IL-18, and components of the inflammasome: apoptosis-associated speck-like protein containing a caspase-activation and recruitment domain (ASC), caspase-1, X-linked inhibitor of apoptosis protein (XIAP), nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3), and gasdermin-D (GSDMD). Specific cell types expressing inflammasome proteins also were evaluated immunohistochemically and assessed quantitatively. After PBBI, expression of IL-1β, IL-18, caspase-1, ASC, XIAP, and NLRP3 peaked around 48 h. Brain protein lysates from PTBI animals showed pyroptosome formation evidenced by ASC laddering, and also contained increased expression of GSDMD at 48 h after injury. ASC-positive immunoreactive neurons within the perilesional cortex were observed at 24 h. At 48 h, ASC expression was concentrated in morphologically activated cortical microglia. This expression of ASC in activated microglia persisted until 12 weeks following PBBI. This is the first report of inflammasome activation after PBBI. Our results demonstrate cell-specific patterns of inflammasome activation and pyroptosis predominantly in microglia, suggesting a sustained pro-inflammatory state following PBBI, thus offering a therapeutic target for this type of brain injury.
Collapse
Affiliation(s)
- Stephanie W. Lee
- Department of Neurological Surgery, the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Shyam Gajavelli
- Department of Neurological Surgery, the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Markus S. Spurlock
- Department of Neurological Surgery, the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Cody Andreoni
- Department of Neurological Surgery, the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - M. Ross Bullock
- Department of Neurological Surgery, the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Robert W. Keane
- Department of Neurological Surgery, the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
| | - W. Dalton Dietrich
- Department of Neurological Surgery, the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
302
|
Magalhães DM, Pereira N, Rombo DM, Beltrão-Cavacas C, Sebastião AM, Valente CA. Ex vivo model of epilepsy in organotypic slices-a new tool for drug screening. J Neuroinflammation 2018; 15:203. [PMID: 29996878 PMCID: PMC6042335 DOI: 10.1186/s12974-018-1225-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 06/14/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Epilepsy is a prevalent neurological disorder worldwide. It is characterized by an enduring predisposition to generate seizures and its development is accompanied by alterations in many cellular processes. Organotypic slice cultures represent a multicellular environment with the potential to assess biological mechanisms, and they are used as a starting point for refining molecules for in vivo studies. Here, we investigated organotypic slice cultures as a model of epilepsy. METHODS We assessed, by electrophysiological recordings, the spontaneous activity of organotypic slices maintained under different culture protocols. Moreover, we evaluated, through molecular-based approaches, neurogenesis, neuronal death, gliosis, expression of proinflammatory cytokines, and activation of NLRP3 inflammasome (nucleotide-binding, leucine-rich repeat, pyrin domain) as biomarkers of neuroinflammation. RESULTS We demonstrated that organotypic slices, maintained under a serum deprivation culture protocol, develop epileptic-like activity. Furthermore, throughout a comparative study with slices that do not depict any epileptiform activity, slices with epileptiform activity were found to display significant differences in terms of inflammation-related features, such as (1) increased neuronal death, with higher incidence in CA1 pyramidal neurons of the hippocampus; (2) activation of astrocytes and microglia, assessed through western blot and immunohistochemistry; (3) upregulation of proinflammatory cytokines, specifically interleukin-1β (IL-1β), interleukin-6, and tumor necrosis factor α, revealed by qPCR; and (4) enhanced expression of NLRP3, assessed by western blot, together with increased NLRP3 activation, showed by IL-1β quantification. CONCLUSIONS Thus, organotypic slice cultures gradually deprived of serum mimic the epileptic-like activity, as well as the inflammatory events associated with in vivo epilepsy. This system can be considered a new tool to explore the interplay between neuroinflammation and epilepsy and to screen potential drug candidates, within the inflammatory cascades, to reduce/halt epileptogenesis.
Collapse
Affiliation(s)
- Daniela M Magalhães
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Noémia Pereira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Diogo M Rombo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia Beltrão-Cavacas
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia A Valente
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
303
|
Morris G, Walker AJ, Berk M, Maes M, Puri BK. Cell Death Pathways: a Novel Therapeutic Approach for Neuroscientists. Mol Neurobiol 2018; 55:5767-5786. [PMID: 29052145 PMCID: PMC5994217 DOI: 10.1007/s12035-017-0793-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 09/26/2017] [Indexed: 02/08/2023]
Abstract
In the first part, the following mechanisms involved in different forms of cell death are considered, with a view to identifying potential therapeutic targets: tumour necrosis factor receptors (TNFRs) and their engagement by tumour necrosis factor-alpha (TNF-α); poly [ADP-ribose] polymerase (PARP)-1 cleavage; the apoptosis signalling kinase (ASK)-c-Jun N-terminal kinase (JNK) axis; lysosomal permeability; activation of programmed necrotic cell death; oxidative stress, caspase-3 inhibition and parthanatos; activation of inflammasomes by reactive oxygen species and the development of pyroptosis; oxidative stress, calcium dyshomeostasis and iron in the development of lysosomal-mediated necrosis and lysosomal membrane permeability; and oxidative stress, lipid peroxidation, iron dyshomeostasis and ferroptosis. In the second part, there is a consideration of the role of lethal and sub-lethal activation of these pathways in the pathogenesis and pathophysiology of neurodegenerative and neuroprogressive disorders, with particular reference to the TNF-α-TNFR signalling axis; dysregulation of ASK-1-JNK signalling; prolonged or chronic PARP-1 activation; the role of pyroptosis and chronic inflammasome activation; and the roles of lysosomal permeabilisation, necroptosis and ferroptosis. Finally, it is suggested that, in addition to targeting oxidative stress and inflammatory processes generally, neuropsychiatric disorders may respond to therapeutic targeting of TNF-α, PARP-1, the Nod-like receptor NLRP3 inflammasome and the necrosomal molecular switch receptor-interacting protein kinase-3, since their widespread activation can drive and/or exacerbate peripheral inflammation and neuroinflammation even in the absence of cell death. To this end, the use is proposed of a combination of the tetracycline derivative minocycline and N-acetylcysteine as adjunctive treatment for a range of neuropsychiatric disorders.
Collapse
Affiliation(s)
- G Morris
- , Bryn Road Seaside 87, Llanelli, Wales, , SA15 2LW, UK
- School of Medicine, Deakin University, Geelong, 3220, Australia
| | - A J Walker
- School of Medicine, Deakin University, Geelong, 3220, Australia
| | - M Berk
- The Centre for Molecular and Medical Research, School of Medicine, Deakin University, P.O. Box 291, Geelong, 3220, Australia
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, 60430-040, Brazil
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, P.O. Box 291, Geelong, 3220, Australia
- Orygen Youth Health Research Centre and the Centre of Youth Mental Health, The Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Parkville, 3052, Australia
| | - M Maes
- School of Medicine, Deakin University, Geelong, 3220, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - B K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, W12 0HS, UK.
| |
Collapse
|
304
|
Targeting the IL33-NLRP3 axis improves therapy for experimental cerebral malaria. Proc Natl Acad Sci U S A 2018; 115:7404-7409. [PMID: 29954866 DOI: 10.1073/pnas.1801737115] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cerebral malaria (CM) is a serious neurological complication caused by Plasmodium falciparum infection. Currently, the only treatment for CM is the provision of antimalarial drugs; however, such treatment by itself often fails to prevent death or development of neurological sequelae. To identify potential improved treatments for CM, we performed a nonbiased whole-brain transcriptomic time-course analysis of antimalarial drug chemotherapy of murine experimental CM (ECM). Bioinformatics analyses revealed IL33 as a critical regulator of neuroinflammation and cerebral pathology that is down-regulated in the brain during fatal ECM and in the acute period following treatment of ECM. Consistent with this, administration of IL33 alongside antimalarial drugs significantly improved the treatment success of established ECM. Mechanistically, IL33 treatment reduced inflammasome activation and IL1β production in microglia and intracerebral monocytes in the acute recovery period following treatment of ECM. Moreover, treatment with the NLRP3-inflammasome inhibitor MCC950 alongside antimalarial drugs phenocopied the protective effect of IL33 therapy in improving the recovery from established ECM. We further showed that IL1β release from macrophages was stimulated by hemozoin and antimalarial drugs and that this was inhibited by MCC950. Our results therefore demonstrate that manipulation of the IL33-NLRP3 axis may be an effective therapy to suppress neuroinflammation and improve the efficacy of antimalarial drug treatment of CM.
Collapse
|
305
|
Zhu J, Hu Z, Han X, Wang D, Jiang Q, Ding J, Xiao M, Wang C, Lu M, Hu G. Dopamine D2 receptor restricts astrocytic NLRP3 inflammasome activation via enhancing the interaction of β-arrestin2 and NLRP3. Cell Death Differ 2018; 25:2037-2049. [PMID: 29786071 DOI: 10.1038/s41418-018-0127-2] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/08/2018] [Accepted: 05/02/2018] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are involved in the neuroinflammation of neurodegenerative diseases, such as Parkinson's disease (PD). Among the numerous inflammatory cytokines, interleukin-1β (IL-1β) produced by astrocytic Nod-like receptor protein (NLRP) inflammasome is crucial in the pathogenesis of PD. β-arrestin2-mediated dopamine D2 receptor (Drd2) signal transduction has been regarded as a potential anti-inflammatory target. Our previous study revealed that astrocytic Drd2 suppresses neuroinflammation in the central nervous system. However, the role of Drd2 in astrocytic NLRP3 inflammasome activation and subsequent IL-1β production remains unclear. In the present study, we used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced PD mouse model to investigate whether Drd2 could suppress astrocytic NLRP3 inflammasome activation. We showed that Drd2 agonist inhibited NLRP3 inflammasome activation, evidenced by decreased caspase-1 expression and reduced IL-1β release in the midbrain of wild type mice. The anti-inflammasome effect of Drd2 was abolished in β-arrestin2 knockout and β-arrestin2 small interfering RNA-injected mice, suggesting a critical role of β-arrestin2 in Drd2-regulated NLRP3 inflammasome activation. We also found that Drd2 agonists suppressed the upregulation of caspase-1 and IL-1β expression in primary cultured mouse astrocytes in response to the activation of NLRP3 inflammasome induced by lipopolysaccharide plus adenosine triphosphate. Furthermore, we demonstrated that β-arrestin2 mediated the inhibitory effect of Drd2 on NLRP3 inflammasome activation via interacting with NLRP3 and interfering the inflammasome assembly. Collectively, our study illustrates that astrocytic Drd2 inhibits NLRP3 inflammasome activation through a β-arrestin2-dependent mechanism, and provides a new strategy for treatment of PD.
Collapse
Affiliation(s)
- Jialei Zhu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Zhaoli Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Xiaojuan Han
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Dongshuo Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Qingling Jiang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Cong Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China.
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China. .,Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
306
|
韦 倩, 冯 洁, 何 春, 华 子. [Role of caspase-1 activation in bilirubin-induced injury in cultured primary rat hippocampal neurons]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:567-571. [PMID: 29891453 PMCID: PMC6743908 DOI: 10.3969/j.issn.1673-4254.2018.05.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To investigate the role of caspase-1 activation in bilirubin-induced neuronal injury and the protective effect of VX-765 against bilirubin-induced neurotoxicity in cultured primary rat hippocampal neurons. METHODS Cultured primary rat hippocampal neurons were exposed to DMSO (control group), 50 µmol/L bilirubin, or 50 µmol/L bilirubin 1 h after 50 µmol/L VX-765 treatment. The expressions of NLRP3 and caspase-1 in the neurons were detected by Western blotting, and the relative cell survival and death rates were assessed with a modified MTT assay, lactate dehydrogenase assay and Typan blue staining. Interleukin-18 (IL-18) concentration in the culture supernatant was measured using enzyme-linked immunosorbent assay (ELISA). RESULTS In cultured primary rat hippocampal neurons, bilirubin exposure for 3 and 6 h caused significant increases in the expressions of NLRP3 and activated caspase-1 compared with those in the control group (P<0.05). Pretreatment of the cells with VX-765 obviously suppressed bilirubin-induced activation of caspase-1 (P<0.05). The relative survival rate of the neurons was (84.02∓2.31)% in VX-765 intervention group, significantly higher than that in bilirubin group (P<0.05) but lower than that in the control group (P<0.05); LDH release rate in VX-765 intervention group was (10.78∓1.58)%, significantly lower than that in bilirubin group (P<0.05) but higher than that in the control group (P<0.05). The cell death rate in VX-765 intervention group was (5.58∓1.23)%, significantly lower than that in bilirubin group (P<0.05) but higher than that in the control group (P<0.05). CONCLUSION In cultured primary rat hippocampal neurons, caspase-1 activation plays a role in bilirubin-induced neurotoxicity, and VX-765 treatment provides protection against bilirubin-induced neuronal injury by inhibiting caspase-1 activation.
Collapse
Affiliation(s)
- 倩 韦
- />重庆医科大学附属儿童医院新生儿科//儿童发育疾病研究教育部重点实验室//儿童发育重大疾病国家国际科技合作基地//认知发育与学习记忆障碍转化医学重庆市重点实验室, 重庆 400014Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400014, China
| | - 洁 冯
- />重庆医科大学附属儿童医院新生儿科//儿童发育疾病研究教育部重点实验室//儿童发育重大疾病国家国际科技合作基地//认知发育与学习记忆障碍转化医学重庆市重点实验室, 重庆 400014Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400014, China
| | - 春梅 何
- />重庆医科大学附属儿童医院新生儿科//儿童发育疾病研究教育部重点实验室//儿童发育重大疾病国家国际科技合作基地//认知发育与学习记忆障碍转化医学重庆市重点实验室, 重庆 400014Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400014, China
| | - 子瑜 华
- />重庆医科大学附属儿童医院新生儿科//儿童发育疾病研究教育部重点实验室//儿童发育重大疾病国家国际科技合作基地//认知发育与学习记忆障碍转化医学重庆市重点实验室, 重庆 400014Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400014, China
| |
Collapse
|
307
|
Mirzaei F, Khazaei M, Komaki A, Amiri I, Jalili C. Virgin coconut oil (VCO) by normalizing NLRP3 inflammasome showed potential neuroprotective effects in Amyloid-β induced toxicity and high-fat diet fed rat. Food Chem Toxicol 2018; 118:68-83. [PMID: 29729307 DOI: 10.1016/j.fct.2018.04.064] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 01/05/2023]
Abstract
Both dyslipidemia and Alzheimer disease (AD) are associated with aging. In this study, the effects of virgin coconut oil (VCO) on inflammasome and oxidative stress in Alzheimer's model (receiving Amyloid-β (Aβ)) and high-fat diet (HFD) model were determined. A total of 120 male Wistar rats, were divided into 12 groups (n = 10), including; healthy control, sham surgery, sham surgery receiving normal saline, HFD, HFD + 8% VCO, HFD + 10% VCO, Aβ received rats, Aβ + 8%VCO, Aβ + 10%VCO, HFD + Aβ, HFD + Aβ+8%VCO, and HFD + Aβ + 10%VCO. Following memory and learning tests, blood sample prepared from the heart and hippocampus of rats in each group was kept at -70 °C for genes expression, oxidative stress, and biochemical tests. Aβ and HFD significantly impaired memory and learning by activating of both NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome and oxidative stress (p<0.05), while treatment with both 8 and 10% VCO normalized inflammasome genes expression and oxidative stress (p<0.05). The Congo Red, Cresyl Violet staining and immunohistochemistry (IHC) test revealed that VCO improved hippocampus histological changes, reduced Aβ plaques and phosphorylated Tau. High-fat diet has exacerbated the effects of Aβ, while VCO showed potential neuroprotective effect.
Collapse
Affiliation(s)
- Fatemeh Mirzaei
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Amiri
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Cyrus Jalili
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
308
|
PM2.5 exposure aggravates oligomeric amyloid beta-induced neuronal injury and promotes NLRP3 inflammasome activation in an in vitro model of Alzheimer's disease. J Neuroinflammation 2018; 15:132. [PMID: 29720213 PMCID: PMC5932821 DOI: 10.1186/s12974-018-1178-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/25/2018] [Indexed: 12/20/2022] Open
Abstract
Background Numerous studies suggested that PM2.5 exposure was associated with increased risk of Alzheimer’s disease (AD). But the precise mechanisms by which PM2.5 contributed to AD pathogenesis have not been clarified. Methods In the presence or absence of neurons, oligomeric amyloid beta (oAβ)-primed microglia were stimulated with PM2.5. Firstly, we determined the effects of PM2.5 exposure on neuronal injury and inflammation in neurons-microglia co-cultures. Then, we examined whether NLRP3 inflammasome activation was involved in PM2.5-induced inflammation. After that, we investigated whether PM2.5 exposure increased ROS level in oAβ-stimulated microglia. At last, we examined whether ROS and NLRP3 inflammasome activation was required for PM2.5-induced neuronal injury in neurons-microglia co-cultures. Results In the present study, we showed that PM2.5 exposure aggravated oAβ-induced neuronal injury and inflammation in neurons-microglia co-cultures via increasing IL-1β production. Further, PM2.5-induced IL-1β production in oAβ-stimulated microglia was possibly dependent on NLRP3 inflammasome activation. Meanwhile, PM2.5 exposure increased ROS level in oAβ-stimulated microglia. ROS was required for PM2.5-induced IL-1β production and NLRP3 inflammasome activation in oAβ-stimulated microglia. More importantly, ROS and NLRP3 inflammasome activation was required for PM2.5-induced neuronal injury in neurons-microglia co-cultures. Conclusions For the first time, these results suggested that the effects of PM2.5 under AD context were possibly mediated by NLRP3 inflammasome activation, which was triggered by ROS. Taken together, these findings have deepened our understanding on the role of PM2.5 in AD pathogenesis.
Collapse
|
309
|
Wang Q, Dong X, Li N, Wang Y, Guan X, Lin Y, Kang J, Zhang X, Zhang Y, Li X, Xu T. JSH-23 prevents depressive-like behaviors in mice subjected to chronic mild stress: Effects on inflammation and antioxidant defense in the hippocampus. Pharmacol Biochem Behav 2018; 169:59-66. [PMID: 29684396 DOI: 10.1016/j.pbb.2018.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/17/2018] [Accepted: 04/19/2018] [Indexed: 12/26/2022]
Abstract
Nuclear factor-kappa B (NF-κB), which is reported to play an important role in the pathogenesis of depression, also has a central role in the genesis and progression of inflammation. Here, we have targeted the nuclear translocation of NF-κB using 4-methyl-N1-(3-phenyl-propyl)-benzene-1,2-diamine (JSH-23) to elucidate its role in depression. We investigated the antidepressant-like effects of JSH-23 in the chronic mild stress (CMS) mouse model, which is a valid, reasonably reliable, and useful model of depression. The antidepressant-like effects of JSH-23 were evaluated using the sucrose preference test (SPT) and the forced swimming test (FST). We also assessed inflammatory markers [interleukin (IL)-6 and tumor necrosis factor-α (TNF-α)] and components of antioxidant defense [superoxide dismutase (SOD) and nuclear factor erythroid-2-related factor 2 (Nrf 2)] in the hippocampus. Fluoxetine, a classical antidepressant, was used in this study as a positive control. Administration of JSH-23 significantly prevented the decreased sucrose preference in the SPT and prevented the increased immobility time in the FST caused by CMS, but had no effect on locomotor activity. Expression of NF-κB p65 protein in the hippocampus was decreased, and elevated levels of IL-6 and TNF-α were reduced, after JSH-23 administration. In addition to its anti-inflammatory effect, JSH-23 treatment increased the expression of SOD and Nrf 2 in the hippocampus, suggesting that it strengthens antioxidant defense. The current study demonstrated that inhibiting the NF-κB signaling cascade using JSH-23 prevented depressive-like behaviors by decreasing inflammation and improving antioxidant defense in the hippocampus. We concluded that NF-κB activation plays an important role in the pathophysiology of depression and that targeting NF-κB signaling may provide a novel and effective therapy for depression. Additional preclinical studies and clinical trials are, however, needed to further elucidate the effects of this therapeutic strategy.
Collapse
Affiliation(s)
- Qi Wang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaomei Dong
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Nannan Li
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yan Wang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaofeng Guan
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yiwei Lin
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jiguang Kang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xia Zhang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yuchen Zhang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaobai Li
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Tianchao Xu
- Department of Medical Psychiatry, General Hospital of Shenyang Military Command, Shenyang, Liaoning Province, China.
| |
Collapse
|
310
|
Rea IM, Gibson DS, McGilligan V, McNerlan SE, Alexander HD, Ross OA. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front Immunol 2018; 9:586. [PMID: 29686666 PMCID: PMC5900450 DOI: 10.3389/fimmu.2018.00586] [Citation(s) in RCA: 722] [Impact Index Per Article: 120.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 03/08/2018] [Indexed: 12/11/2022] Open
Abstract
Cytokine dysregulation is believed to play a key role in the remodeling of the immune system at older age, with evidence pointing to an inability to fine-control systemic inflammation, which seems to be a marker of unsuccessful aging. This reshaping of cytokine expression pattern, with a progressive tendency toward a pro-inflammatory phenotype has been called "inflamm-aging." Despite research there is no clear understanding about the causes of "inflamm-aging" that underpin most major age-related diseases, including atherosclerosis, diabetes, Alzheimer's disease, rheumatoid arthritis, cancer, and aging itself. While inflammation is part of the normal repair response for healing, and essential in keeping us safe from bacterial and viral infections and noxious environmental agents, not all inflammation is good. When inflammation becomes prolonged and persists, it can become damaging and destructive. Several common molecular pathways have been identified that are associated with both aging and low-grade inflammation. The age-related change in redox balance, the increase in age-related senescent cells, the senescence-associated secretory phenotype (SASP) and the decline in effective autophagy that can trigger the inflammasome, suggest that it may be possible to delay age-related diseases and aging itself by suppressing pro-inflammatory molecular mechanisms or improving the timely resolution of inflammation. Conversely there may be learning from molecular or genetic pathways from long-lived cohorts who exemplify good quality aging. Here, we will discuss some of the current ideas and highlight molecular pathways that appear to contribute to the immune imbalance and the cytokine dysregulation, which is associated with "inflammageing" or parainflammation. Evidence of these findings will be drawn from research in cardiovascular disease, cancer, neurological inflammation and rheumatoid arthritis.
Collapse
Affiliation(s)
- Irene Maeve Rea
- School of Medicine, Dentistry and Biomedical Science, Queens University Belfast, Belfast, United Kingdom
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
- Care of Elderly Medicine, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - David S. Gibson
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
| | - Victoria McGilligan
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
| | - Susan E. McNerlan
- Regional Genetics Service, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - H. Denis Alexander
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, United States
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
311
|
Pranzatelli MR. Advances in Biomarker-Guided Therapy for Pediatric- and Adult-Onset Neuroinflammatory Disorders: Targeting Chemokines/Cytokines. Front Immunol 2018; 9:557. [PMID: 29670611 PMCID: PMC5893838 DOI: 10.3389/fimmu.2018.00557] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/05/2018] [Indexed: 12/26/2022] Open
Abstract
The concept and recognized components of “neuroinflammation” are expanding at the intersection of neurobiology and immunobiology. Chemokines (CKs), no longer merely necessary for immune cell trafficking and positioning, have multiple physiologic, developmental, and modulatory functionalities in the central nervous system (CNS) through neuron–glia interactions and other mechanisms affecting neurotransmission. They issue the “help me” cry of neurons and astrocytes in response to CNS injury, engaging invading lymphoid cells (T cells and B cells) and myeloid cells (dendritic cells, monocytes, and neutrophils) (adaptive immunity), as well as microglia and macrophages (innate immunity), in a cascade of events, some beneficial (reparative), others destructive (excitotoxic). Human cerebrospinal fluid (CSF) studies have been instrumental in revealing soluble immunobiomarkers involved in immune dysregulation, their dichotomous effects, and the cells—often subtype specific—that produce them. CKs/cytokines continue to be attractive targets for the pharmaceutical industry with varying therapeutic success. This review summarizes the developing armamentarium, complexities of not compromising surveillance/physiologic functions, and insights on applicable strategies for neuroinflammatory disorders. The main approach has been using a designer monoclonal antibody to bind directly to the chemo/cytokine. Another approach is soluble receptors to bind the chemo/cytokine molecule (receptor ligand). Recombinant fusion proteins combine a key component of the receptor with IgG1. An additional approach is small molecule antagonists (protein therapeutics, binding proteins, and protein antagonists). CK neutralizing molecules (“neutraligands”) that are not receptor antagonists, high-affinity neuroligands (“decoy molecules”), as well as neutralizing “nanobodies” (single-domain camelid antibody fragment) are being developed. Simultaneous, more precise targeting of more than one cytokine is possible using bispecific agents (fusion antibodies). It is also possible to inhibit part of a signaling cascade to spare protective cytokine effects. “Fusokines” (fusion of two cytokines or a cytokine and CK) allow greater synergistic bioactivity than individual cytokines. Another promising approach is experimental targeting of the NLRP3 inflammasome, amply expressed in the CNS and a key contributor to neuroinflammation. Serendipitous discovery is not to be discounted. Filling in knowledge gaps between pediatric- and adult-onset neuroinflammation by systematic collection of CSF data on CKs/cytokines in temporal and clinical contexts and incorporating immunobiomarkers in clinical trials is a challenge hereby set forth for clinicians and researchers.
Collapse
Affiliation(s)
- Michael R Pranzatelli
- National Pediatric Neuroinflammation Organization, Inc., Orlando, FL, United States.,College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
312
|
Kaur G, Bagam P, Pinkston R, Singh DP, Batra S. Cigarette smoke-induced inflammation: NLRP10-mediated mechanisms. Toxicology 2018; 398-399:52-67. [PMID: 29501574 DOI: 10.1016/j.tox.2018.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 02/07/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive, life-threatening disease that causes irreversible lung damage. Cigarette smoking is the chief etiologic factor for the commencement of this condition. Despite constant efforts to develop therapeutic interventions and to ascertain the molecular mechanism leading to the pathophysiology of this disease, much remains unknown. However, pattern recognition receptors (PRRs), i.e., Toll-like-receptors (TLRs) and NOD-like receptors (NLRs) are believed to play important roles in COPD and could serve as effective therapeutic targets. Although the role of TLRs in COPD has been well studied, the importance of NLRs has not yet been explored in detail. The NLR family member NLRP10 (aka NOD8, PAN5, PYNOD) is the only member of this family of proteins that lacks the leucine rich repeat (LRR) domain responsible for detection of pathogen and danger-associated molecular patterns (PAMPs/DAMPs). Therefore, instead of functioning as a PRR, NLRP10 may have a broader regulatory role. To elucidate the role of NLRP10 in secondhand smoke (SHS)-induced inflammation, we exposed C57Bl/6 (WT) and Nlrp10-deficient mice (Nlrp10-/-) on the C57Bl/6 background to filtered air- or SHS- for 6 weeks (acute exposure) and assessed the resulting molecular events. Leukocyte recruitment in SHS-exposed Nlrp10-/- mice was found to be significantly lower compared to SHS-exposed WT mice. In addition, we observed an important role for NLRP10 in SHS-mediated caspase-1 activation, cytokine/chemokine production (IL-1β, IL-18, MCP-1 and IL-17A), and induction of NF-κB and MAPKs in the lungs of C57Bl/6 mice. The reduced influx of CD4+IL-17A+ and CD8+IL-17A+ cells into the lungs of SHS-exposed Nlrp10-/- mice and impaired differentiation of Nlrp10-/- Th0 cells into Th17 cells (ex vivo) provide insight into the mechanistic details underlying NLRP10-dependent IL-17 production. We further substantiated our in vivo findings by challenging human alveolar type II epithelial cells (A549) transfected with scrambled- or Nlrp10-siRNA with cigarette smoke extract (CSE). We observed an important role of NLRP10 in cytokine and chemokine production as well as expression of NF-κB and MAPKs in CSE-exposed A549 cells. Furthermore, replenishment of A549 cell culture with recombinant IL-17A (rIL-17A) during NLRP10 knockdown rescued CSE-induced inflammatory responses. To identify upstream mediators of NLRP10 regulation we investigated epigenetic markers within the Nlrp10 promoter following cigarette smoke exposure and observed significant changes in active as well as repressive gene markers on histone 3 and histone 4 using both in vivo and in vitro study models. Further, alterations in the respective histone acetyl- and methyltransferases (PCAF, SET1, ESET, SUV20H1) correlated well with the observed histone modifications. Overall, our findings suggest a novel role of epigenetically regulated NLRP10 in Th17/IL-17 signaling during CS exposure.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States
| | - Prathyusha Bagam
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States
| | - Rakeysha Pinkston
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States
| | - Dhirendra P Singh
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States
| | - Sanjay Batra
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States.
| |
Collapse
|
313
|
Mugisho OO, Green CR, Kho DT, Zhang J, Graham ES, Acosta ML, Rupenthal ID. The inflammasome pathway is amplified and perpetuated in an autocrine manner through connexin43 hemichannel mediated ATP release. Biochim Biophys Acta Gen Subj 2018; 1862:385-393. [DOI: 10.1016/j.bbagen.2017.11.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/30/2017] [Accepted: 11/16/2017] [Indexed: 12/20/2022]
|
314
|
Awad F, Assrawi E, Louvrier C, Jumeau C, Georgin-Lavialle S, Grateau G, Amselem S, Giurgea I, Karabina SA. Inflammasome biology, molecular pathology and therapeutic implications. Pharmacol Ther 2018; 187:133-149. [PMID: 29466702 DOI: 10.1016/j.pharmthera.2018.02.011] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inflammasomes are intracellular multiprotein signaling complexes, mainly present in myeloid cells. They commonly assemble around a cytoplasmic receptor of the nucleotide-binding leucine-rich repeat containing receptor (NLR) family, although other cytoplasmic receptors like pyrin have been shown to form inflammasomes. The nucleation of the multiprotein scaffolding platform occurs upon detection of a microbial, a danger or a homeostasis pattern by the receptor that will, most commonly, associate with the adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD) through homotypic domain interactions resulting in recruitment of procaspase-1. This will lead to the autoproteolytic activation of caspase-1, which regulates the secretion of proinflammatory IL1β and IL18 cytokines and pyroptosis, a caspase-1-mediated form of cell death. Pyroptosis occurs through cleavage of Gasdermin D, a membrane pore forming protein. Recently, non-canonical inflammasomes have been described, which directly sense intracellular pathogens through caspase-4 and -5 in humans, leading to pyroptosis. Inflammasomes are important in host defense; however, a deregulated activity is associated with a number of inflammatory, immune and metabolic disorders. Furthermore, mutations in inflammasome receptor coding genes are causal for an increasing number of rare autoinflammatory diseases. Biotherapies targeting the products of inflammasome activation as well as molecules that directly or indirectly inhibit inflammasome nucleation and activation are promising therapeutic areas. This review discusses recent advances in inflammasome biology, the molecular pathology of several inflammasomes, and current therapeutic approaches in autoinflammatory diseases and in selected common multifactorial inflammasome-mediated disorders.
Collapse
Affiliation(s)
- Fawaz Awad
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Département de Génétique médicale, Hôpital Trousseau, Paris, F-75012, France
| | - Eman Assrawi
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Département de Génétique médicale, Hôpital Trousseau, Paris, F-75012, France
| | - Camille Louvrier
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Département de Génétique médicale, Hôpital Trousseau, Paris, F-75012, France
| | - Claire Jumeau
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Département de Génétique médicale, Hôpital Trousseau, Paris, F-75012, France
| | - Sophie Georgin-Lavialle
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Tenon, Service de Médecine interne, Paris, F-75012, France
| | - Gilles Grateau
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Hôpital Tenon, Service de Médecine interne, Paris, F-75012, France
| | - Serge Amselem
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Département de Génétique médicale, Hôpital Trousseau, Paris, F-75012, France.
| | - Irina Giurgea
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Département de Génétique médicale, Hôpital Trousseau, Paris, F-75012, France.
| | - Sonia-Athina Karabina
- Sorbonne Université, INSERM, UMR_S 933, Assistance Publique Hôpitaux de Paris, Département de Génétique médicale, Hôpital Trousseau, Paris, F-75012, France.
| |
Collapse
|
315
|
Slowik A, Lammerding L, Hoffmann S, Beyer C. Brain inflammasomes in stroke and depressive disorders: Regulation by oestrogen. J Neuroendocrinol 2018; 30. [PMID: 28477436 DOI: 10.1111/jne.12482] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/02/2017] [Accepted: 05/02/2017] [Indexed: 12/28/2022]
Abstract
Neuroinflammation is a devastating pathophysiological process that results in brain damage and neuronal death. Pathogens, cell fragments and cellular dysfunction trigger inflammatory responses. Irrespective of the cause, inflammasomes are key intracellular multiprotein signalling platforms that sense neuropathological conditions. The activation of inflammasomes leads to the auto-proteolytic cleavage of caspase-1, resulting in the proteolysis of the pro-inflammatory cytokines interleukin (IL)1β and IL18 into their bioactive forms. It also initiates pyroptosis, a type of cell death. The two cytokines contribute to the pathogenesis in acute and chronic brain diseases and also play a central role in human aging and psychiatric disorders. Sex steroids, in particular oestrogens, are well-described neuroprotective agents in the central nervous system. Oestrogens improve the functional outcome after ischaemia and traumatic brain injury, reduce neuronal death in Parkinson's and Alzheimer's disease, as well as in amyotrophic lateral sclerosis, attenuate glutamate excitotoxicity and the formation of radical oxygen species, and lessen the spread of oedema after damage. Moreover, oestrogens alleviate menopause-related depressive symptoms and have a positive influence on depressive disorders probably by influencing growth factor production and serotonergic brain circuits. Recent evidence also suggests that inflammasome signalling affects anxiety- and depressive-like behaviour and that oestrogen ameliorates depression-like behaviour through the suppression of inflammasomes. In the present review, we highlight the most recent findings demonstrating that oestrogens selectively suppress the activation of the neuroinflammatory cascade in the brain in acute and chronic brain disease models. Furthermore, we aim to describe putative regulatory signalling pathways involved in the control of inflammasomes. Finally, we consider that psychiatric disorders such as depression also contain an inflammatory component that could be modulated by oestrogen.
Collapse
Affiliation(s)
- A Slowik
- Medical Faculty, Institute of Neuroanatomy, RWTH Aachen, Aachen, Germany
| | - L Lammerding
- Medical Faculty, Institute of Neuroanatomy, RWTH Aachen, Aachen, Germany
| | - S Hoffmann
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, Aachen, Germany
- JARA - Translational Brain Medicine, Aachen, Germany
| | - C Beyer
- Medical Faculty, Institute of Neuroanatomy, RWTH Aachen, Aachen, Germany
- JARA - Translational Brain Medicine, Aachen, Germany
| |
Collapse
|
316
|
Affiliation(s)
- Giovanna Traina
- Department of Pharmaceutical Sciences, University of Perugia, Via S. Costanzo, 06126 Perugia, Italy. Tel.: ; Fax: ; E-mail:
| |
Collapse
|
317
|
Zou P, Liu X, Li G, Wang Y. Resveratrol pretreatment attenuates traumatic brain injury in rats by suppressing NLRP3 inflammasome activation via SIRT1. Mol Med Rep 2017; 17:3212-3217. [PMID: 29257276 DOI: 10.3892/mmr.2017.8241] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 11/21/2017] [Indexed: 11/05/2022] Open
Abstract
The inflammatory response in the cerebral cortex serves an important role in the progression of secondary injury following traumatic brain injury (TBI). The NLR family pyrin domain containing 3 (NLRP3) inflammasome is necessary for initiating inflammation and is involved in various central nervous system disorders. The aim of the present study was to investigate the neuroprotective effect of resveratrol and elucidate the underlying mechanisms of resveratrol associated regulation of the NLRP3 inflammasome in TBI. The results demonstrated that the activation of NLRP3, caspase‑1 and sirtuin 1 (SIRT1), enhanced the production of inflammatory cytokines and reactive oxygen species (ROS) following TBI. Administration of resveratrol alleviated the degree of TBI, as evidenced by the reduced neuron‑specific enolase (NSE) and brain water content (WBC). Resveratrol pretreatment also inhibited the activation of NLRP3 and caspase‑1, and reduced the production of inflammatory cytokines and ROS. In addition, resveratrol further promoted SIRT1 activation. Furthermore, the suppressing effect of resveratrol on the NLRP3 inflammasome and ROS was blocked by the SIRT1 inhibitor, sirtinol. The results revealed that the activation of the NLRP3 inflammasome and the subsequent inflammatory responses in the cerebral cortex were involved in the process of TBI. Resveratrol may attenuate the inflammatory response and relieve TBI by reducing ROS production and inhibiting NLRP3 activation. The effect of resveratrol on NLRP3 inflammasome and ROS may also be SIRT1 dependent.
Collapse
Affiliation(s)
- Peng Zou
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaoxiao Liu
- Department of Cardiology, Xi'an Medical University Affiliated Northern Hospital, Xi'an, Shaanxi 710032, P.R. China
| | - Gang Li
- Department of Neurosurgery, Jiangxian People's Hospital, Yuncheng, Shanxi 043600, P.R. China
| | - Yangang Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
318
|
Early life experience contributes to the developmental programming of depressive-like behaviour, neuroinflammation and oxidative stress. J Psychiatr Res 2017; 95:196-207. [PMID: 28886447 DOI: 10.1016/j.jpsychires.2017.08.020] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/04/2017] [Accepted: 08/24/2017] [Indexed: 12/25/2022]
Abstract
This study used an animal model of depression induced by maternal care deprivation (MCD) to investigate whether depressive behaviour, neuroinflammation and oxidative stress were underlying factors in developmental programming after early life stress. At postnatal days (PND) 20, 30, 40, and 60, individual subsets of animals were evaluated in behavioural tests and then euthanized to assess cytokine levels and oxidative stress parameters in the prefrontal cortex (PFC), hippocampus and serum. The results showed that MCD did not induce behavioural changes at PND 30 and 40. However, at PND 20 and 60, the rats displayed a depressive-like behaviour in the forced swimming test, without changes in locomotor spontaneous activity. In the brain and serum, the levels of pro-inflammatory cytokines (interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumour necrosis factor-α (TNF-α)) were increased, and the anti-inflammatory cytokine (interleukin-10) level was reduced throughout developmental programming (PND 20, 30, 40 and 60). Protein carbonyl levels increased in the brain at PND 30, 40 and 60. Superoxide dismutase (SOD) activity was decreased during all developmental programming phases evaluated in the brain. Catalase (CAT) activity was decreased at PND 20, 40 and 60 in the brain. Our results revealed that "critical episodes" in early life stressful events are able to induce behavioural alterations that persist into adulthood and can stimulate inflammation and oxidative damage in both central and peripheral systems, which are required for distinct patterns of resilience against psychiatric disorders later in life.
Collapse
|
319
|
Dabo S, Maillard P, Collados Rodriguez M, Hansen MD, Mazouz S, Bigot DJ, Tible M, Janvier G, Helynck O, Cassonnet P, Jacob Y, Bellalou J, Gatignol A, Patel RC, Hugon J, Munier-Lehmann H, Meurs EF. Inhibition of the inflammatory response to stress by targeting interaction between PKR and its cellular activator PACT. Sci Rep 2017; 7:16129. [PMID: 29170442 PMCID: PMC5701060 DOI: 10.1038/s41598-017-16089-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023] Open
Abstract
PKR is a cellular kinase involved in the regulation of the integrative stress response (ISR) and pro-inflammatory pathways. Two N-terminal dsRNA Binding Domains (DRBD) are required for activation of PKR, by interaction with either dsRNA or PACT, another cellular DRBD-containing protein. A role for PKR and PACT in inflammatory processes linked to neurodegenerative diseases has been proposed and raised interest for pharmacological PKR inhibitors. However, the role of PKR in inflammation is subject to controversy. We identified the flavonoid luteolin as an inhibitor of the PKR/PACT interaction at the level of their DRBDs using high-throughput screening of chemical libraries by homogeneous time-resolved fluorescence. This was further validated using NanoLuc-Based Protein Complementation Assay. Luteolin inhibits PKR phosphorylation, the ISR and the induction of pro-inflammatory cytokines in human THP1 macrophages submitted to oxidative stress and toll-like receptor (TLR) agonist. Similarly, luteolin inhibits induction of pro-inflammatory cytokines in murine microglial macrophages. In contrast, luteolin increased activation of the inflammasome, in a PKR-independent manner. Collectively, these data delineate the importance of PKR in the inflammation process to the ISR and induction of pro-inflammatory cytokines. Pharmacological inhibitors of PKR should be used in combination with drugs targeting directly the inflammasome.
Collapse
Affiliation(s)
- Stephanie Dabo
- Unité Hepacivirus and Innate Immunity, Institut Pasteur, 75015, Paris, France.,CNRS, UMR 3569, Paris, France
| | - Patrick Maillard
- Unité Hepacivirus and Innate Immunity, Institut Pasteur, 75015, Paris, France.,CNRS, UMR 3569, Paris, France
| | - Milagros Collados Rodriguez
- Unité Hepacivirus and Innate Immunity, Institut Pasteur, 75015, Paris, France.,CNRS, UMR 3569, Paris, France
| | - Marianne Doré Hansen
- Unité Hepacivirus and Innate Immunity, Institut Pasteur, 75015, Paris, France.,CNRS, UMR 3569, Paris, France.,Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7006, Trondheim, Norway
| | - Sabrina Mazouz
- Unité Hepacivirus and Innate Immunity, Institut Pasteur, 75015, Paris, France.,CNRS, UMR 3569, Paris, France
| | - Donna-Joe Bigot
- Unité Hepacivirus and Innate Immunity, Institut Pasteur, 75015, Paris, France.,CNRS, UMR 3569, Paris, France
| | - Marion Tible
- Center of Cognitive Neurology, Lariboisière Hospital AP-HP University Paris Diderot, Paris, France.,Inserm, U942, Paris, France
| | - Geneviève Janvier
- Unité Hepacivirus and Innate Immunity, Institut Pasteur, 75015, Paris, France.,CNRS, UMR 3569, Paris, France
| | - Olivier Helynck
- Unité de Chimie et Biocatalyse, Institut Pasteur, 75015, Paris, France.,CNRS, UMR3523, Paris, France
| | - Patricia Cassonnet
- CNRS, UMR 3569, Paris, France.,Unité de Génétique Moléculaire des Virus à ARN, Institut Pasteur, Université Paris Diderot, Paris, France
| | - Yves Jacob
- CNRS, UMR 3569, Paris, France.,Unité de Génétique Moléculaire des Virus à ARN, Institut Pasteur, Université Paris Diderot, Paris, France
| | - Jacques Bellalou
- Plate-forme des protéines recombinantes, Institut Pasteur, 75015, CNRS UMR 3528, Paris, France
| | - Anne Gatignol
- Virus-Cell Interactions Laboratory, Lady Davis Institute for Medical Research, Department of Medicine, department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Rekha C Patel
- University of South Carolina, Department of Biological Sciences, Columbia, South Carolina, 29208, USA
| | - Jacques Hugon
- Center of Cognitive Neurology, Lariboisière Hospital AP-HP University Paris Diderot, Paris, France.,Inserm, U942, Paris, France
| | - Hélène Munier-Lehmann
- Unité de Chimie et Biocatalyse, Institut Pasteur, 75015, Paris, France.,CNRS, UMR3523, Paris, France
| | - Eliane F Meurs
- Unité Hepacivirus and Innate Immunity, Institut Pasteur, 75015, Paris, France. .,CNRS, UMR 3569, Paris, France.
| |
Collapse
|
320
|
Bowyer JF, Tranter KM, Sarkar S, George NI, Hanig JP, Kelly KA, Michalovicz LT, Miller DB, O'Callaghan JP. Corticosterone and exogenous glucose alter blood glucose levels, neurotoxicity, and vascular toxicity produced by methamphetamine. J Neurochem 2017; 143:198-213. [PMID: 28792619 DOI: 10.1111/jnc.14143] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/01/2017] [Accepted: 08/04/2017] [Indexed: 12/29/2022]
Abstract
Our previous studies have raised the possibility that altered blood glucose levels may influence and/or be predictive of methamphetamine (METH) neurotoxicity. This study evaluated the effects of exogenous glucose and corticosterone (CORT) pretreatment alone or in combination with METH on blood glucose levels and the neural and vascular toxicity produced. METH exposure consisted of four sequential injections of 5, 7.5, 10, and 10 mg/kg (2 h between injections) D-METH. The three groups given METH in combination with saline, glucose (METH+Glucose), or CORT (METH+CORT) had significantly higher glucose levels compared to the corresponding treatment groups without METH except at 3 h after the last injection. At this last time point, the METH and METH+Glucose groups had lower levels than the non-METH groups, while the METH+CORT group did not. CORT alone or glucose alone did not significantly increase blood glucose. Mortality rates for the METH+CORT (40%) and METH+Glucose (44%) groups were substantially higher than the METH (< 10%) group. Additionally, METH+CORT significantly increased neurodegeneration above the other three METH treatment groups (≈ 2.5-fold in the parietal cortex). Thus, maintaining elevated levels of glucose during METH exposure increases lethality and may exacerbate neurodegeneration. Neuroinflammation, specifically microglial activation, was associated with degenerating neurons in the parietal cortex and thalamus after METH exposure. The activated microglia in the parietal cortex were surrounding vasculature in most cases and the extent of microglial activation was exacerbated by CORT pretreatment. Our findings show that acute CORT exposure and elevated blood glucose levels can exacerbate METH-induced vascular damage, neuroinflammation, neurodegeneration and lethality. Cover Image for this issue: doi. 10.1111/jnc.13819.
Collapse
Affiliation(s)
- John F Bowyer
- Division of Neurotoxicology, National Center for Toxicology/FDA, Jefferson, Arkansas, USA
| | - Karen M Tranter
- Division of Neurotoxicology, National Center for Toxicology/FDA, Jefferson, Arkansas, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicology/FDA, Jefferson, Arkansas, USA
| | - Nysia I George
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research/FDA, Jefferson, Arkansas, USA
| | - Joseph P Hanig
- Center for Drug Evaluation and Research/FDA Silver Spring, Silver Spring, Maryland, USA
| | - Kimberly A Kelly
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health Morgantown, Morgantown, West Virginia, USA
| | - Lindsay T Michalovicz
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health Morgantown, Morgantown, West Virginia, USA
| | - Diane B Miller
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health Morgantown, Morgantown, West Virginia, USA
| | - James P O'Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health Morgantown, Morgantown, West Virginia, USA
| |
Collapse
|
321
|
Kempuraj D, Thangavel R, Selvakumar GP, Zaheer S, Ahmed ME, Raikwar SP, Zahoor H, Saeed D, Natteru PA, Iyer S, Zaheer A. Brain and Peripheral Atypical Inflammatory Mediators Potentiate Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2017; 11:216. [PMID: 28790893 PMCID: PMC5522882 DOI: 10.3389/fncel.2017.00216] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/05/2017] [Indexed: 12/18/2022] Open
Abstract
Neuroinflammatory response is primarily a protective mechanism in the brain. However, excessive and chronic inflammatory responses can lead to deleterious effects involving immune cells, brain cells and signaling molecules. Neuroinflammation induces and accelerates pathogenesis of Parkinson’s disease (PD), Alzheimer’s disease (AD) and Multiple sclerosis (MS). Neuroinflammatory pathways are indicated as novel therapeutic targets for these diseases. Mast cells are immune cells of hematopoietic origin that regulate inflammation and upon activation release many proinflammatory mediators in systemic and central nervous system (CNS) inflammatory conditions. In addition, inflammatory mediators released from activated glial cells induce neurodegeneration in the brain. Systemic inflammation-derived proinflammatory cytokines/chemokines and other factors cause a breach in the blood brain-barrier (BBB) thereby allowing for the entry of immune/inflammatory cells including mast cell progenitors, mast cells and proinflammatory cytokines and chemokines into the brain. These peripheral-derived factors and intrinsically generated cytokines/chemokines, α-synuclein, corticotropin-releasing hormone (CRH), substance P (SP), beta amyloid 1–42 (Aβ1–42) peptide and amyloid precursor proteins can activate glial cells, T-cells and mast cells in the brain can induce additional release of inflammatory and neurotoxic molecules contributing to chronic neuroinflammation and neuronal death. The glia maturation factor (GMF), a proinflammatory protein discovered in our laboratory released from glia, activates mast cells to release inflammatory cytokines and chemokines. Chronic increase in the proinflammatory mediators induces neurotoxic Aβ and plaque formation in AD brains and neurodegeneration in PD brains. Glial cells, mast cells and T-cells can reactivate each other in neuroinflammatory conditions in the brain and augment neuroinflammation. Further, inflammatory mediators from the brain can also enter into the peripheral system through defective BBB, recruit immune cells into the brain, and exacerbate neuroinflammation. We suggest that mast cell-associated inflammatory mediators from systemic inflammation and brain could augment neuroinflammation and neurodegeneration in the brain. This review article addresses the role of some atypical inflammatory mediators that are associated with mast cell inflammation and their activation of glial cells to induce neurodegeneration.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Govindhasamy P Selvakumar
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Mohammad E Ahmed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Haris Zahoor
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Daniyal Saeed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Prashant A Natteru
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Shankar Iyer
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Asgar Zaheer
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| |
Collapse
|
322
|
Scholz H, Eder C. Lysophosphatidylcholine activates caspase-1 in microglia via a novel pathway involving two inflammasomes. J Neuroimmunol 2017; 310:107-110. [PMID: 28778433 DOI: 10.1016/j.jneuroim.2017.07.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/06/2017] [Accepted: 07/08/2017] [Indexed: 12/30/2022]
Abstract
Inflammasomes regulate microglial caspase-1 activation and subsequent neuroinflammatory processes in brain pathology. In the present study, we have identified inflammasomes causing caspase-1 activation following stimulation of microglia with lysophosphatidylcholine (LPC), a proinflammatory lipid generated under pathological conditions in the brain. LPC-induced caspase-1 activation in microglia was found to depend on LPS prestimulation, inflammasome NLRP3 and adaptor molecule ASC. Furthermore, knockdown of inflammasome NLRC4 inhibited LPC-stimulated caspase-1 activity in microglia, suggesting the requirement of two inflammasomes for optimal caspase-1 activity.
Collapse
Affiliation(s)
- Holger Scholz
- Charité - Universitätsmedizin Berlin, Institute of Physiology, 10117 Berlin, Germany
| | - Claudia Eder
- Charité - Universitätsmedizin Berlin, Institute of Physiology, 10117 Berlin, Germany.
| |
Collapse
|
323
|
Effect of paricalcitol on pancreatic oxidative stress, inflammatory markers, and glycemic status in diabetic rats. Ir J Med Sci 2017; 187:75-84. [PMID: 28551720 DOI: 10.1007/s11845-017-1635-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 05/18/2017] [Indexed: 01/09/2023]
Abstract
OBJECTIVES This study is designed to explore the effect of paricalcitol (vitamin D receptor agonist) on pancreatic oxidative stress, inflammatory markers, and adiponectin and glycemic status in diabetic rats. MATERIALS AND METHODS Forty Sprague-Dawley male rats aged 10-12 weeks (150-250 g) were used in this study. Type 2 diabetes was developed by providing 4 weeks of high-fat-diet feeding before one shot of streptozotocin injection (40 mg/kg i.p.). Four study groups were designed as normal control rats, diabetic control vehicle-treated, diabetic paricalcitol-treated (0.8 μg/kg), and diabetic glibenclamide-treated (0.6 mg/kg) groups with 10 animals in each. After treatment of diabetic rats for 3 months, pancreatic inflammatory and oxidative stress markers, plasma adiponectin, glycemic status parameters, and histopathological pancreatic islet changes were evaluated. RESULTS Paricalcitol and glibenclamide treatment significantly (P < 0.05) decreased plasma glucose, insulin resistance, and pancreatic malondialdehyde and tumor necrosis factor-α levels. Moreover, they significantly (P < 0.05) increased plasma fasting insulin, C-peptide, adiponectin, pancreatic IL-2, catalase, superoxide dismutase, glutathione peroxidase, and reduced glutathione when contrasted with diabetic control rats. Furthermore, they prevented extensive histopathological damage in the pancreas of diabetic rats. CONCLUSIONS Paricalcitol reduced pancreatic oxidative stress and inflammatory markers, and improved glycemic status in diabetic rats.
Collapse
|