1
|
Gust KA, Amar SK, Gut CP, Styles RM, Karna RR, James RA, Holtzapple DM, Stricker JL, McInturf SM, Phillips EA, Honnold C, Luo X, Mumy KL, Mattie DR, Chappell MA, Mayo ML. Multi-disciplinary investigation identifies increased potency of ethyl-parathion inhaled within a soil-dust matrix to cause acetylcholinesterase-dependent molecular impacts. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 114:104615. [PMID: 39710123 DOI: 10.1016/j.etap.2024.104615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
Neurotoxicity investigations of inhaled organophosphorus pesticide (OP), ethyl-parathion (EP), were conducted in Sprague Dawley rats comparing exposures to EP volatilized at 0, 1, 10, and 20 mg/m3 versus EP incorporated into soil dust (5 mg/m3) at 0, 0.0095, 0.09, and 0.185 mg/mg3. All exposures were sublethal, caused no respiratory effects, and no effects on balance and coordination behavior. Both volatilized and dust-incorporated EP exposures significantly decreased acetylcholinesterase (AChE) activity in plasma and hippocampus tissue. Correspondingly, plasma and hippocampal dopamine levels spiked in these exposures suggesting compensatory cholinergic / dopaminergic signal balancing. The EP exposures significantly increased expression of pro-inflammatory genes, including MAPK-14, IL6, IL1β, and TNF-α, while global RNA-seq results identified significant enrichment of inflammation, oxidative stress, and apoptosis pathways. Remarkably, dust-incorporated EP impacted similar molecular endpoints as volatilized EP but at concentrations two orders of magnitude lower highlighting potentially increased potency of EP incorporated into soil dust.
Collapse
Affiliation(s)
- Kurt A Gust
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States.
| | - Saroj K Amar
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States; Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, United States
| | - Chet P Gut
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Renee M Styles
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Ranju R Karna
- Credere Associates LLC, Westbrook, ME located at US Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - R Arden James
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - David M Holtzapple
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Joshua L Stricker
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Shawn M McInturf
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Elizabeth A Phillips
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Cary Honnold
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Xiao Luo
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Karen L Mumy
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - David R Mattie
- Air Force Research Laboratory/711 HPW, Wright-Patterson Air Force Base, OH, United States
| | - Mark A Chappell
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Michael L Mayo
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| |
Collapse
|
2
|
Singh P, Chaudhary M, Kazmi JS, Kuschner CE, Volpe BT, Chaudhuri TD, Becker LB. Vagus nerve stimulation: A targeted approach for reducing tissue-specific ischemic reperfusion injury. Biomed Pharmacother 2025; 184:117898. [PMID: 39923406 DOI: 10.1016/j.biopha.2025.117898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
Vagus Nerve Stimulation (VNS), a neuromodulation technique of applying controlled electrical impulses to the vagus nerve, has now emerged as a potential therapeutic approach for ischemia-reperfusion insults. It provides a pivotal link in improving functional outcomes for the central nervous system and multiple target organs affected by ischemia-reperfusion injury (I/RI). Reduced blood flow during ischemia and subsequent resumption of blood supply during reperfusion to the tissue compromises cellular health because of the combination of mitochondrial dysfunction, oxidative stress, cytokine release, inflammation, apoptosis, intracellular calcium overload, and endoplasmic reticulum stress, which ultimately leads to cell death and irreversible tissue damage. Furthermore, inflammation and apoptosis also play critical roles in the acute progression of ischemic injury pathology. Emerging evidence indicates that VNS in I/RI may act in an anti-inflammatory capacity, reducing oxidative stress and apoptosis, while also improving endothelial and mitochondrial function leading to reduced infarct sizes and cytoprotection in skeletal muscle, gastrointestinal tract, liver, kidney, lung, heart, and brain tissue. In this review, we attempt to shed light on the mechanistic links between tissue-specific damage following I/RI and the therapeutic approach of VNS in attenuating damage, considering both direct and remote I/RI scenarios. Thus, we want to advance the understanding of VNS that could further warrant its clinical implementation, especially as a treatment for I/RI.
Collapse
Affiliation(s)
- Parmeshar Singh
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Department of Emergency Medicine, Northwell Health, NY, USA
| | - Manju Chaudhary
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Jacob S Kazmi
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Cyrus E Kuschner
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Department of Emergency Medicine, Northwell Health, NY, USA
| | - Bruce T Volpe
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Timir D Chaudhuri
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA; Department of Emergency Medicine, Northwell Health, NY, USA; Department of Emergency Medicine, Kindai University Faculty of Medicine, Osaka, Japan.
| |
Collapse
|
3
|
Su AC, Zhang LY, Zhang JG, Hu YY, Liu XY, Li SC, Xian XH, Li WB, Zhang M. The Regulation of Autophagy by p38 MAPK-PPARγ Signaling During the Brain Ischemic Tolerance Induced by Cerebral Ischemic Preconditioning. DNA Cell Biol 2022; 41:838-849. [PMID: 35944278 DOI: 10.1089/dna.2022.0087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Several studies indicated that autophagy activation participates in brain ischemic tolerance (BIT) induced by cerebral ischemic preconditioning (CIP). However, the mechanism of autophagy activation during the process still remains unclear. The present study aimed to evaluate the role of p38 MAPK-peroxisome proliferator-activated receptor γ (PPARγ) signaling cascade in autophagy during the CIP-induced BIT. The results shown that, initially, autophagy activation was observed after CIP in the model of global cerebral ischemia in rats, as was indicated by the upregulation of Beclin 1 expression, an increase in LC3-II/LC3-I ratio, the enhanced LC3 immunofluorescence, and a rise in the number of autophagosomes in the neurons of the hippocampal CA1 area. Besides, the inhibitor of autophagy 3-methyladenine obliterated the neuroprotection induced by CIP. Furthermore, the upregulation of p-p38 MAPK and PPARγ expressions was earlier than autophagy activation after CIP. In addition, pretreatment with SB203580 (the inhibitor of p38 MAPK) reversed CIP-induced PPARγ upregulation, autophagy activation, and neuroprotection. Pretreatment with GW9662 (the inhibitor of PPARγ) reversed autophagy activation and neuroprotection, while it had no effect on p-p38 MAPK upregulation induced by CIP. These data suggested that the p38 MAPK-PPARγ signaling pathway participates in autophagy activation during the induction of BIT by CIP.
Collapse
Affiliation(s)
- A-Chou Su
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Ling-Yan Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xi-Yun Liu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Shi-Chao Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Hebei Medical University, Shijiazhuang, People's Republic of China
| |
Collapse
|
4
|
Nuclear Receptors in Myocardial and Cerebral Ischemia-Mechanisms of Action and Therapeutic Strategies. Int J Mol Sci 2021; 22:ijms222212326. [PMID: 34830207 PMCID: PMC8617737 DOI: 10.3390/ijms222212326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Nearly 18 million people died from cardiovascular diseases in 2019, of these 85% were due to heart attack and stroke. The available therapies although efficacious, have narrow therapeutic window and long list of contraindications. Therefore, there is still an urgent need to find novel molecular targets that could protect the brain and heart against ischemia without evoking major side effects. Nuclear receptors are one of the promising targets for anti-ischemic drugs. Modulation of estrogen receptors (ERs) and peroxisome proliferator-activated receptors (PPARs) by their ligands is known to exert neuro-, and cardioprotective effects through anti-apoptotic, anti-inflammatory or anti-oxidant action. Recently, it has been shown that the expression of aryl hydrocarbon receptor (AhR) is strongly increased after brain or heart ischemia and evokes an activation of apoptosis or inflammation in injury site. We hypothesize that activation of ERs and PPARs and inhibition of AhR signaling pathways could be a promising strategy to protect the heart and the brain against ischemia. In this Review, we will discuss currently available knowledge on the mechanisms of action of ERs, PPARs and AhR in experimental models of stroke and myocardial infarction and future perspectives to use them as novel targets in cardiovascular diseases.
Collapse
|
5
|
Comparison and Analysis on the Existing Single-Herbal Strategies against Viral Myocarditis. Genet Res (Camb) 2021; 2021:9952620. [PMID: 34456633 PMCID: PMC8371739 DOI: 10.1155/2021/9952620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/31/2021] [Indexed: 02/08/2023] Open
Abstract
Purpose Herbal medicine is one of crucial symbols of Chinese national medicine. Investigation on molecular responses of different herbal strategies against viral myocarditis is immeasurably conducive to targeting drug development in the current international absence of miracle treatment. Methods Literature retrieval platforms were applied in the collection of existing empirical evidences for viral myocarditis-related single-herbal strategies. SwissTargetPrediction, Metascape, and Discovery Studio coordinating with multidatabases investigated underlying target genes, interactive proteins, and docking molecules in turn. Results Six single-herbal medicines consisting of Huangqi (Hedysarum Multijugum Maxim), Yuganzi (Phyllanthi Fructus), Kushen (Sophorae Flavescentis Radix), Jianghuang (Curcumaelongae Rhizoma), Chaihu (Radix Bupleuri), and Jixueteng (Spatholobus Suberectus Dunn) meet the requirement. There were 11 overlapped and 73 unique natural components detected in these herbs. SLC6A2, SLC6A4, NOS2, PPARA, PPARG, ACHE, CYP2C19, CYP51A1, and CHRM2 were equally targeted by six herbs and identified as viral myocarditis-associated symbols. MCODE algorithm exposed the hub role of SRC and EGFR in strategies without Jianghuang. Subsequently, we learned intermolecular interactions of herbal components and their targeting heart-tissue-specific CHRM2, FABP3, TNNC1, TNNI3, TNNT2, and SCN5A and cardiac-myocytes-specific IL6, MMP1, and PLAT coupled with viral myocarditis. Ten interactive characteristics such as π-alkyl and van der Waals were modeled in which ARG111, LYS253, ILE114, and VAL11 on cardiac troponin (TNNC1-TNNI3-TNNT2) and ARG208, ASN106, and ALA258 on MMP1 fulfilled potential communicating anchor with ellagic acid, 5α, 9α-dihydroxymatrine, and leachianone g via hydrogen bond and hydrophobic interaction, respectively. Conclusions The comprehensive outcomes uncover differences and linkages between six herbs against viral myocarditis through component and target analysis, fostering development of drugs.
Collapse
|
6
|
A selective peroxisome proliferator-activated receptor-γ agonist benefited propionic acid induced autism-like behavioral phenotypes in rats by attenuation of neuroinflammation and oxidative stress. Chem Biol Interact 2019; 311:108758. [DOI: 10.1016/j.cbi.2019.108758] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/10/2019] [Accepted: 07/18/2019] [Indexed: 01/08/2023]
|
7
|
Pioglitazone, a PPARγ agonist, reduces cisplatin-evoked neuropathic pain by protecting against oxidative stress. Pain 2019; 160:688-701. [PMID: 30507781 DOI: 10.1097/j.pain.0000000000001448] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Painful peripheral neuropathy is a dose-limiting side effect of cisplatin treatment. Using a murine model of cisplatin-induced hyperalgesia, we determined whether a PPARγ synthetic agonist, pioglitazone, attenuated the development of neuropathic pain and identified underlying mechanisms. Cisplatin produced mechanical and cold hyperalgesia and decreased electrical thresholds of Aδ and C fibers, which were attenuated by coadministration of pioglitazone (10 mg/kg, intraperitoneally [i.p.]) with cisplatin. Antihyperalgesic effects of pioglitazone were blocked by the PPARγ antagonist T0070907 (10 mg/kg, i.p.). We hypothesized that the ability of pioglitazone to reduce the accumulation of reactive oxygen species (ROS) in dorsal root ganglion (DRG) neurons contributed to its antihyperalgesic activity. Effects of cisplatin and pioglitazone on somatosensory neurons were studied on dissociated mouse DRG neurons after 24 hours in vitro. Incubation of DRG neurons with cisplatin (13 µM) for 24 hours increased the occurrence of depolarization-evoked calcium transients, and these were normalized by coincubation with pioglitazone (10 µM). Oxidative stress in DRG neurons was considered a significant contributor to cisplatin-evoked hyperalgesia because a ROS scavenger attenuated hyperalgesia and normalized the evoked calcium responses when cotreated with cisplatin. Pioglitazone increased the expression and activity of ROS-reducing enzymes in DRG and normalized cisplatin-evoked changes in oxidative stress and labeling of mitochondria with the dye MitoTracker Deep Red, indicating that the antihyperalgesic effects of pioglitazone were attributed to its antioxidant properties in DRG neurons. These data demonstrate clear benefits of broadening the use of the antidiabetic drug pioglitazone, or other PPARγ agonists, to minimize the development of cisplatin-induced painful neuropathy.
Collapse
|
8
|
Song YJ, Dai CX, Li M, Cui MM, Ding X, Zhao XF, Wang CL, Li ZL, Guo MY, Fu YY, Wen XR, Qi DS, Wang YL. The potential role of HO-1 in regulating the MLK3-MKK7-JNK3 module scaffolded by JIP1 during cerebral ischemia/reperfusion in rats. Behav Brain Res 2019; 359:528-535. [PMID: 30412737 DOI: 10.1016/j.bbr.2018.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 11/03/2018] [Accepted: 11/05/2018] [Indexed: 01/01/2023]
Abstract
Heme oxygenase (HO-1), which may be induced by Cobaltic protoporphyrin IX chloride (CoPPIX) or Rosiglitazone (Ros), is a neuroprotective agent that effectively reduces ischemic stroke. Previous studies have shown that the neuroprotective mechanisms of HO-1 are related to JNK signaling. The expression of HO-1 protects cells from death through the JNK signaling pathway. This study aimed to ascertain whether the neuroprotective effect of HO-1 depends on the assembly of the MLK3-MKK7-JNK3 signaling module scaffolded by JIP1 and further influences the JNK signal transmission through HO-1. Prior to the ischemia-reperfusion experiment, CoPPIX was injected through the lateral ventricle for 5 consecutive days or Ros was administered via intraperitoneal administration in the week prior to transient ischemia. Our results demonstrated that HO-1 could inhibit the assembly of the MLK3-MKK7-JNK3 signaling module scaffolded by JIP1 and could ultimately diminish the phosphorylation of JNK3. Furthermore, the inhibition of JNK3 phosphorylation downregulated the level of p-c-Jun and elevated neuronal cell death in the CA1 of the hippocampus. Taken together, these findings suggested that HO-1 could ameliorate brain injury by regulating the MLK3-MKK7-JNK3 signaling module, which was scaffolded by JIP1 and JNK signaling during cerebral ischemia/reperfusion.
Collapse
Affiliation(s)
- Yuan-Jian Song
- Jiangsu Key Laboratory of Brain Disease Bioinformatics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China; Department of Genetics, Research Facility Center for Morphology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Chun-Xiao Dai
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Man Li
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Miao-Miao Cui
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Xin Ding
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Xiao-Fang Zhao
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Cai-Lin Wang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Zhen-Ling Li
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Meng-Yuan Guo
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Yan-Yan Fu
- Jiangsu Key Laboratory of Brain Disease Bioinformatics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China; Department of Genetics, Research Facility Center for Morphology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Xiang-Ru Wen
- Jiangsu Key Laboratory of Brain Disease Bioinformatics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China; School of Basic Education Science, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
| | - Da-Shi Qi
- Jiangsu Key Laboratory of Brain Disease Bioinformatics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China; Department of Genetics, Research Facility Center for Morphology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
| | - Yu-Lan Wang
- Jiangsu Key Laboratory of Brain Disease Bioinformatics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China; Department of Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
| |
Collapse
|
9
|
Hu Y, Xu Y. Relationship between interleukin‐6 and brain ischemia. IBRAIN 2019. [DOI: 10.1002/j.2769-2795.2019.tb00039.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Yue Hu
- Department of AnesthesiologyThe First People's Hospital of Shuangliu DistrictChengduSichuanChina
| | - Yang Xu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
10
|
Fumagalli M, Lombardi M, Gressens P, Verderio C. How to reprogram microglia toward beneficial functions. Glia 2018; 66:2531-2549. [PMID: 30195261 PMCID: PMC6585737 DOI: 10.1002/glia.23484] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
Microglia, brain cells of nonneural origin, orchestrate the inflammatory response to diverse insults, including hypoxia/ischemia or maternal/fetal infection in the perinatal brain. Experimental studies have demonstrated the capacity of microglia to recognize pathogens or damaged cells activating a cytotoxic response that can exacerbate brain damage. However, microglia display an enormous plasticity in their responses to injury and may also promote resolution stages of inflammation and tissue regeneration. Despite the critical role of microglia in brain pathologies, the cellular mechanisms that govern the diverse phenotypes of microglia are just beginning to be defined. Here we review emerging strategies to drive microglia toward beneficial functions, selectively reporting the studies which provide insights into molecular mechanisms underlying the phenotypic switch. A variety of approaches have been proposed which rely on microglia treatment with pharmacological agents, cytokines, lipid messengers, or microRNAs, as well on nutritional approaches or therapies with immunomodulatory cells. Analysis of the molecular mechanisms relevant for microglia reprogramming toward pro‐regenerative functions points to a central role of energy metabolism in shaping microglial functions. Manipulation of metabolic pathways may thus provide new therapeutic opportunities to prevent the deleterious effects of inflammatory microglia and to control excessive inflammation in brain disorders.
Collapse
Affiliation(s)
- Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9 -20133, Milan, Italy
| | | | - Pierre Gressens
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, 1141 Paris, France.,Centre for the Developing Brain, Department of Perinatal Health and Imaging, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, United Kingdom
| | - Claudia Verderio
- IRCCS Humanitas, via Manzoni 56, 20089, Rozzano, Italy.,CNR Institute of Neuroscience, via Vanvitelli 32, 20129 Milan, Italy
| |
Collapse
|
11
|
Pei Z, Deng S, Xie D, Lv M, Guo W, Liu D, Zheng Z, Long X. Protective role of fenofibrate in sepsis-induced acute kidney injury in BALB/c mice. RSC Adv 2018; 8:28510-28517. [PMID: 35542461 PMCID: PMC9083917 DOI: 10.1039/c8ra00488a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 08/04/2018] [Indexed: 01/12/2023] Open
Abstract
Acute kidney injury (AKI) is a severe complication of sepsis, which largely contributes to the associated high mortality rate. Fenofibrate, a peroxisome proliferator activated receptor α (PPARα) agonist, has received considerable attention because of its effects related to renal damage-related energy metabolism and inflammation. The present study investigated the effects of fenofibrate on sepsis-associated AKI in BALB/c mice subjected to caecal ligation and puncture (CLP). Eight-week-old male BALB/c mice were divided into four groups: control group, fenofibrate group, caecal ligation and puncture (CLP) group, and fenofibrate + CLP group. CLP was performed after mice were gavaged with fenofibrate for 2 weeks. After 48 hours, we measured the histopathological alterations of the kidney tissue and plasma levels of serum creatinine (CRE), neutrophil gelatinase-associated lipocalin (NGAL), reactive oxygen species (ROS), ATP, and ADP. We evaluated PPARα and P53 protein levels as well as interleukin (IL)-1β, IL-6, and tumour necrosis factor-α mRNA levels. Our results showed that administering fenofibrate significantly reduced kidney histological alterations caused by CLP. Fenofibrate inhibited the plasma levels of ROS, CRE, NGAL, and increased the ATP/ADP ratio. Fenofibrate significantly inhibited elevations in P53, IL-1β, IL-6, and tumour necrosis factor-α expression. The results suggest that fenofibrate administration effectively modulates energy metabolism and may be a novel approach to treat sepsis-induced renal damage.
Collapse
Affiliation(s)
- Zuowei Pei
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University No. 6 Jiefang Street Dalian China
| | - Shuling Deng
- Department of Intensive Care Units, Affiliated Zhongshan Hospital of Dalian University No. 6 Jiefang Street Dalian China +86-0411-62893373 +86-0411-62893373
| | - Dengmei Xie
- Department of Clinical Pharmacy, Affiliated Zhongshan Hospital of Dalian University No. 6 Jiefang Street Dalian China
| | - Mingyi Lv
- Department of Intensive Care Units, Affiliated Zhongshan Hospital of Dalian University No. 6 Jiefang Street Dalian China +86-0411-62893373 +86-0411-62893373
| | - Wenyan Guo
- Department of Intensive Care Units, Affiliated Zhongshan Hospital of Dalian University No. 6 Jiefang Street Dalian China +86-0411-62893373 +86-0411-62893373
| | - Duping Liu
- Department of Intensive Care Units, Affiliated Zhongshan Hospital of Dalian University No. 6 Jiefang Street Dalian China +86-0411-62893373 +86-0411-62893373
| | - Zhenzhen Zheng
- Department of Intensive Care Units, Affiliated Zhongshan Hospital of Dalian University No. 6 Jiefang Street Dalian China +86-0411-62893373 +86-0411-62893373
| | - Xiaofeng Long
- Department of Intensive Care Units, Affiliated Zhongshan Hospital of Dalian University No. 6 Jiefang Street Dalian China +86-0411-62893373 +86-0411-62893373
| |
Collapse
|
12
|
Cell death after traumatic brain injury: Detrimental role of anoikis in healing. Clin Chim Acta 2018; 482:149-154. [DOI: 10.1016/j.cca.2018.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 12/19/2022]
|
13
|
Luo Y, Kuang S, Li H, Ran D, Yang J. cAMP/PKA-CREB-BDNF signaling pathway in hippocampus mediates cyclooxygenase 2-induced learning/memory deficits of rats subjected to chronic unpredictable mild stress. Oncotarget 2018; 8:35558-35572. [PMID: 28415673 PMCID: PMC5482598 DOI: 10.18632/oncotarget.16009] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/02/2017] [Indexed: 12/11/2022] Open
Abstract
To investigate the mechanism of cyclooxygenase 2 (COX2) in learning and memory impairments in rats subjected to chronic unpredictable mild stress (CUMS), meloxicam was used intragastrically to inhibit the activity of cyclooxygenase 2. Moreover, cyclooxygenase 2 over-expressing or RNA interfere lentivirus was injected intraventricularly to increase or decrease the enzyme's expression, respectively. The body weights and sucrose consumption were used to analyze depressive behaviors, while the Morris water maze and step-down-type passive avoidance tests were carried out to evaluate the learning-memory functions. The levels of inflammatory cytokines were measured to estimate inflammation and the contents of cyclic adenosine monophosphate (cAMP) were used to measure the levels of the second messenger. Changes in cyclooxygenase 2 mRNA levels were analyzed using reverse transcription polymerase chain reaction. Moreover, the expression of cyclooxygenase 2, brain-derived neurotrophic factor (BDNF), prostaglandins receptor 3 (EP3), protein kinase A (PKA), cAMP response element binding protein (CREB), and phosphorylated CREB were estimated using immunohistochemical staining or western blotting. The results showed that CUMS led to significant depressive-like behaviors and learning and memory dysfunctions. Also, the cAMP levels decreased significantly, while levels of inflammatory cytokines and prostaglandins E2 increased significantly. The expressions of PKA, BDNF, phosphorylated CREB/CREB declined and cyclooxygenase 2 was increased. Meloxicam and cyclooxygenase 2 RNA interfere lentivirus reversed the changes caused by CUMS while cyclooxygenase 2-overexpressing lentivirus worsened these abnormalities. The findings also showed that CUMS increased cyclooxygenase 2 expression, which can cause learning and memory impairments, mainly through activating the hippocampal neuronal cAMP/PKA-CREB-BDNF signaling pathways.
Collapse
Affiliation(s)
- Ying Luo
- Department of Pharmacology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| | - Shengnan Kuang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China.,Department of Pharmacy, People's Hospital of Rongchang, Chongqing, China
| | - Huan Li
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Dongzhi Ran
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Junqing Yang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China
| |
Collapse
|
14
|
Asai Y, Tanaka H, Nadai M, Katoh M. Effect of status epilepticus on expression of brain UDP-glucuronosyltransferase 1a in rats. Biopharm Drug Dispos 2017; 39:75-82. [PMID: 29131354 DOI: 10.1002/bdd.2114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 10/26/2017] [Accepted: 10/29/2017] [Indexed: 12/17/2022]
Abstract
Status epilepticus (SE) involves severe epileptic seizures that cause oxidative stress in the brain. Oxidative stress is known to influence uridine 5'-diposphate-glucuronosyltransferase (UGT) 1A expression. The present study aimed at elucidating the effect of SE on Ugt1a1, Ugt1a6 and Ugt1a7 expression in the rat brain. Kainic acid was used to create an animal model of SE. Sprague-Dawley rats were treated intraperitoneally with 10 mg/kg kainic acid. Ugt1a1 and Ugt1a7 mRNA levels were increased by SE in the cortex and hippocampus (Ugt1a1: 4.0- and 5.3-fold, respectively; Ugt1a7: 2.8- and 2.5-fold, respectively). Moreover, the induction degree of heme oxygenase-1 mRNA, an oxidative stress marker, was high in these regions, suggesting that oxidative stress could be involved in Ugt1a1 and Ugt1a7 induction. Ugt1a6 was elevated by 1.8-fold in the cortex in both SE and non-response (non-epileptic seizure response) rats, implying that Ugt1a6 induction may be independent from SE. An intraperitoneal single administration of 25 mg/kg diazepam (DZP) for the treatment of SE could attenuate heme oxygenase-1 induction in the cortex, whereas Ugt1a1 was decreased in the hippocampus, but not in the cortex, suggesting that there likely exists an alternative mechanism for Ugt1a1 reduction by DZP treatment. Continuous 14-day administration of DZP inhibited Ugt1a1 induction in the cortex, but did not have an effect on Ugt1a7 induction. This study indicated that SE altered the expression of brain Ugt1a1 and Ugt1a7, which could alter glucuronidation in the brain.
Collapse
Affiliation(s)
- Yuki Asai
- Pharmaceutics, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, 468-8503, Japan
| | - Hatsuna Tanaka
- Pharmaceutics, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, 468-8503, Japan
| | - Masayuki Nadai
- Pharmaceutics, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, 468-8503, Japan
| | - Miki Katoh
- Pharmaceutics, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, 468-8503, Japan
| |
Collapse
|
15
|
Roles of Peroxisome Proliferator-Activated Receptor Gamma on Brain and Peripheral Inflammation. Cell Mol Neurobiol 2017; 38:121-132. [PMID: 28975471 DOI: 10.1007/s10571-017-0554-5] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/23/2017] [Indexed: 02/08/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) has been implicated in the pathology of numerous diseases involving diabetes, stroke, cancer, or obesity. It is expressed in diverse cell types, including vessels, immune and glial cells, and neurons. PPARγ plays crucial roles in the regulation of cellular differentiation, lipid metabolism, or glucose homeostasis. PPARγ ligands also exert effects on attenuating degenerative processes in the brain, as well as in peripheral systems, and it has been associated with the control of anti-inflammatory mechanisms, oxidative stress, neuronal death, neurogenesis, differentiation, and angiogenesis. This review will highlight key advances in the understanding of the PPARγ-related mechanisms responsible for neuroprotection after brain injuries, both ischemia and traumatic brain injury, and it will also cover the natural and synthetic agonist for PPARγ, angiotensin receptor blockers, and PPARγ antagonists, used in experimental and clinical research. A better understanding of the pleiotropic mechanisms and applications of these drugs to improve the recovery and to repair the acute and chronic induced neuroinflammation after brain injuries will pave the way for more effective therapeutic strategies after brain deficits.
Collapse
|
16
|
Luo Y, Kuang S, Xue L, Yang J. The mechanism of 5-lipoxygenase in the impairment of learning and memory in rats subjected to chronic unpredictable mild stress. Physiol Behav 2016; 167:145-153. [PMID: 27640130 DOI: 10.1016/j.physbeh.2016.09.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 07/20/2016] [Accepted: 09/13/2016] [Indexed: 12/23/2022]
Abstract
OBJECTIVES To examine the mechanism of 5-lipoxygenase (5-LO) in the learning and memory dysfunction in rats subjected to chronic unpredictable mild stress (CUMS). METHODS Eighty rats were divided into eight groups: the 0.5% sodium carboxymethyl cellulose solution (NaCMC)-treated group, empty vector (LV-Mock)-treated group, CUMS+NaCMC-treated group, CUMS+sertraline-treated group, CUMS+caffeic acid (10mg/kg)-treated group, CUMS+caffeic acid (30mg/kg)-treated group, CUMS+LV-Mock-treated group, and CUMS+5-LO-silencers lentiviral vectors (LV-si-5-LO)-treated group, n=10. Sucrose preference tests were performed to assess depression-like behavior. The Morris water maze and step-down tests were used to evaluate learning and memory performance. The levels of inflammatory cytokines, malondialdehyde, and the activity of superoxide dismutase (SOD) were detected to estimate inflammation and oxidative stress. Changes in 5-LO mRNA and protein were detected using reverse transcription polymerase chain reaction and Western blotting. The expression of synaptophysin, postsynaptic density-95 (PSD-95), and brain-derived neurotrophic factor (BDNF) in the hippocampus were measured using immunohistochemical staining. RESULTS Treatment with caffeic acid or LV-si-5-LO increased sucrose consumption, decreased escape latency and increased the number of platform crosses in the Morris water maze test, and decreased the number of errors and prolonged the latency in the step-down test. We observed a decreased expression of 5-LO, and levels of malondialdehyde, leukotriene-B4, tumor necrosis factor-α, and interleukin-6, while the protein levels of synaptophysin, PSD-95, BDNF, and the activity of SOD were increased in the hippocampus of the CUMS-treated rats. CONCLUSIONS CUMS-induced impairment in learning and memory could be triggered by an inflammatory response in the rat hippocampus, which results in oxidative stress injury and impacts the synaptic plasticity of hippocampal neurons. Inhibition of the activity or expression of 5-LO could suppress hippocampal inflammation, enhance synaptic plasticity, and improve learning and memory function in depressed rats.
Collapse
Affiliation(s)
- Ying Luo
- Department of Pharmacology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China.
| | - Shengnan Kuang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China.
| | - Lai Xue
- Department of Pharmacology, Chongqing Medical University, Chongqing, China.
| | - Junqing Yang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China.
| |
Collapse
|
17
|
Liu J, Yang L, Tian H, Ma Q. Cathepsin D is involved in the oxygen and glucose deprivation/reperfusion-induced apoptosis of astrocytes. Int J Mol Med 2016; 38:1257-63. [PMID: 27573911 DOI: 10.3892/ijmm.2016.2709] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 07/12/2016] [Indexed: 01/18/2023] Open
Abstract
The lysosome and its associated protein cathe-psin D (Cat D) play critical roles in the pathological process of secondary damage following ischemia/reperfusion (I/R) injury. However, the roles of Cat D in I/R-exposed astrocytesremain unclear. In this study, we determined the roles of Cat D in the oxygen-glucose deprivation/reperfusion (OGD/R)-induced apoptosis of astrocytes as well as the underlying mechanisms. We found that OGD/R markedly increased cell apoptosis and the production of inflammatory cytokines, namely IL-6, tumor necrosis factor (TNF)-α and FasL in a reperfusion time‑dependent manner and their elevation peaked at 24 h after reperfusion. Moreover, the cytosolic Cat D level and Cat D activity was significantly upregulated in response to OGD/R exposure. Furthermore, OGD/R exposure gradually disrupted the innate acidic conditions of the lysosome. Exogenous TNF-α and FasL administration elevated cytosolic Cat D levels and cell apoptosis whereas TNFR1 and Fas inhibition significantly reversed these effects induced by OGD/R. Cat D overexpression enhanced cell apoptosis and the levels of apoptogenic proteins, including Bax and caspase-3, whereas Cat D siRNA transfection had an inhibitory effect on cell apoptosis and the expression of proapoptotic proteins. In addition, we observed that Cat D upregulation disrupted mitochondrial membrane potential and induced the production of reactive oxygen species. In conclusion, OGD/R injury induced the production of TNF-α, IL-6 and FasL which promoted lysosomal dysfunction and Cat D leakage into the cytoplasm. This eventually resulted in caspase‑dependent apoptosis, mitochondrial membrane potential loss and oxidative stress in astrocytes.
Collapse
Affiliation(s)
- Jianlin Liu
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lin Yang
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Hongyan Tian
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qiang Ma
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
18
|
Song GJ, Nam Y, Jo M, Jung M, Koo JY, Cho W, Koh M, Park SB, Suk K. A novel small-molecule agonist of PPAR-γ potentiates an anti-inflammatory M2 glial phenotype. Neuropharmacology 2016; 109:159-169. [PMID: 27288982 DOI: 10.1016/j.neuropharm.2016.06.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 01/01/2023]
Abstract
Neuroinflammation is a key process for many neurodegenerative diseases. Activated microglia and astrocytes play an essential role in neuroinflammation by producing nitric oxide (NO), inflammatory cytokines, chemokines, and neurotoxins. Therefore, targeting glia-mediated neuroinflammation using small-molecules is a potential therapeutic strategy. In this study, we performed a phenotypic screen using microglia cell-based assay to identify a hit compound containing N-carbamoylated urethane moiety (SNU-BP), which inhibits lipopolysaccharide (LPS)-induced NO production in microglia. SNU-BP inhibited pro-inflammatory cytokines and inducible nitric oxide synthase in LPS-stimulated microglia, and potentiated interleukin-4-induced arginase-1 expression. PPAR-γ was identified as a molecular target of SNU-BP. The PPAR response element reporter assay revealed that SNU-BP specifically activated PPAR-γ, but not PPAR-δ or -α, confirming that PPAR-γ is the target protein of SNU-BP. The anti-inflammatory effect of SNU-BP was attenuated by genetic and pharmacological inhibition of PPAR-γ. In addition, SNU-BP induced an anti-inflammatory phenotype in astrocytes as well, by inhibiting pro-inflammatory NO and TNF-α, while increasing anti-inflammatory genes, such as arginase-1 and Ym-1. Finally, SNU-BP exhibited an anti-inflammatory effect in the LPS-injected mouse brain, demonstrating a protective potential for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Gyun Jee Song
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Youngpyo Nam
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Myungjin Jo
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Myungsu Jung
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Ja Young Koo
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Wansang Cho
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Minseob Koh
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Seung Bum Park
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea.
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
19
|
Chang CZ, Wu SC, Kwan AL. Glycyrrhizin Attenuates Proinflammatory Cytokines through a Peroxisome Proliferator-Activated Receptor-γ-Dependent Mechanism and Experimental Vasospasm in a Rat Model. J Vasc Res 2015; 52:12-21. [PMID: 25896311 DOI: 10.1159/000381099] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/15/2015] [Indexed: 11/19/2022] Open
Abstract
The peroxisome proliferator-activated receptor (PPAR) is downregulated in the cortex of experimental subarachnoid hemorrhage (SAH) animals. This study is to examine the effect of glycyrrhizin on the alternation of PPARs and proinflammatory cytokines in a rodent SAH model. CSF cytokines were evaluated by RT-PCR. Basilar arteries (BAs) were harvested to examine PPARs (RT-PCR and Western blot), and a morphological examination was conducted. Deformed endothelium and tortuous elastic lamina were observed in the BAs of the SAH groups, but they were absent in the glycyrrhizin groups or the healthy controls. The PPAR-γ and -δ protein levels were reduced in the SAH groups (p < 0.01). Glycyrrhizin significantly increased the expressed PPAR-γ protein and mRNA (preconditioning) and PPAR-δ mRNA (both treatment and preconditioning), which corresponded to the reduced IL-1β and TNF-α levels. The administration of a PPAR-γ inhibitor, BADGE, halted the reduction of IL-1β and TNF-α in the glycyrrhizin groups. Conclusively, glycyrrhizin exerts anti-inflammatory effects on SAH-induced vasospasm and attenuates the expression of PPARs, especially PPAR-γ, which corresponds to the severity of SAH-related inflammation. These findings also offer credit to the antivasospastic effect of glycyrrhizin and its vasculoprotective effect in animals subjected to SAH.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Basilar Artery/metabolism
- Cytokines/biosynthesis
- Cytokines/cerebrospinal fluid
- Cytokines/genetics
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Endothelium, Vascular/pathology
- Gene Expression Regulation/drug effects
- Glycyrrhizic Acid/pharmacology
- Glycyrrhizic Acid/therapeutic use
- Inflammation
- Infusion Pumps
- Male
- PPAR delta/biosynthesis
- PPAR delta/genetics
- PPAR gamma/antagonists & inhibitors
- PPAR gamma/biosynthesis
- PPAR gamma/genetics
- PPAR gamma/physiology
- Phytotherapy
- Premedication
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Single-Blind Method
- Subarachnoid Hemorrhage/complications
- Subarachnoid Hemorrhage/drug therapy
- Subarachnoid Hemorrhage/genetics
- Vasospasm, Intracranial/etiology
- Vasospasm, Intracranial/physiopathology
- Vasospasm, Intracranial/prevention & control
Collapse
Affiliation(s)
- Chih-Zen Chang
- Department of Surgery, Faculty of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | | | | |
Collapse
|