1
|
Chen E, Zhang ZQ, Xu AC, Huang F, He YX, Yu XC, He GX. INFLUENCES OF HEAT STRESS ON GLUTAMATE TRANSMISSION-DEPENDENT EXPRESSION LEVELS OF IL-1β and IL-18 IN BV-2 MICROGLIAL CELLS. Shock 2024; 62:146-152. [PMID: 38668801 DOI: 10.1097/shk.0000000000002374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
ABSTRACT Objective: This study aimed to explore the impact of heat stress (HS) on glutamate transmission-dependent expression levels of interleukin-1β (IL-1β) and IL-18 in BV-2 microglial cells. Methods: BV-2 microglial cells were cultured in vitro , with cells maintained at 37°C serving as the control. The HS group experienced incubation at 40°C for 1 h, followed by further culturing at 37°C for 6 or 12 h. The experimental group was preincubated with glutamate, the glutamate antagonist riluzole, or the mGluR5 agonist, 2-chloro-5-hydroxyphenylglycine (CHPG), before HS. Glutamate content in BV-2 culture supernatant was assessed using colorimetric assay. Moreover, mRNA expression levels of EAAT3 and/or mGluR5 in BV-2 cells were determined via quantitative polymerase chain reaction. Interleukins (IL-1β and IL-18) in cell culture supernatant were measured using enzyme-linked immunosorbent assay. Western blot analysis was employed to assess protein levels of IL-1β and IL-18 in BV-2 cells. Results: HS induced a significant release of glutamate and increased the expression levels of mGluR5 and EAAT3 in BV-2 cells. It also triggered the expression levels and release of proinflammatory factors, such as IL-1β and IL-18, synergizing with the effects of glutamate treatment. Preincubation with both riluzole and CHPG significantly reduced HS-induced glutamate release and mitigated the increased expression levels and release of IL-1β and IL-18 induced by HS. Conclusion: The findings confirmed that microglia could be involved in HS primarily through glutamate metabolisms, influencing the expression levels and release of IL-1β and IL-18.
Collapse
Affiliation(s)
- Er Chen
- Department of Intensive Care Unit, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | - An-Cong Xu
- Department of Intensive Care Unit, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fan Huang
- Wenzhou Medical University, Wenzhou, China
| | | | - Xi-Chong Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Guo-Xin He
- Department of Intensive Care Unit, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
2
|
Wang L, Ye B, Liu Y, Li J, Li C, Wen M, Li H. Xuebijing Injection Attenuates Heat Stroke-Induced Brain Injury through Oxidative Stress Blockage and Parthanatos Modulation via PARP-1/AIF Signaling. ACS OMEGA 2023; 8:33392-33402. [PMID: 37744847 PMCID: PMC10515343 DOI: 10.1021/acsomega.3c03084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/19/2023] [Indexed: 09/26/2023]
Abstract
Heat stroke (HS) is a potentially fatal acute condition caused by an interplay of complex events including inflammation, endothelial injury, and coagulation abnormalities that make its pharmacological treatment a challenging problem. The traditional Chinese medicine Xuebijing injection (XBJ) has been shown to reduce inflammatory responses and prevent organ injuries in HS-induced mice. However, the underlying mechanism of XBJ in HS-induced brain injury remains unclear. In this study, HS-induced rat models and cell models were established to elucidate the effects and underlying mechanisms of XBJ injection on HS-induced brain injury in vivo and in vitro. The results revealed that XBJ injection improved the survival outcome of HS rats and attenuated HS-induced brain injury in a concentration-dependent manner. Subsequently, the reduction in viability and proliferation of neurons induced by HS were reversed by XBJ treatment, while the HS-induced increased ROS levels and neuron death were also inhibited by XBJ injection. Mechanistically, HS activated PARP-1/AIF signaling in vitro and in vivo, inducing the translocation of AIF from the cytoplasm to the nucleus, leading to PARP-1-dependent cell death of neurons. Additionally, we compared XBJ injection effects in young and old age rats. Results showed that XBJ also provided protective effects in HS-induced brain injury in aging rats; however, the treatment efficacy of XBJ injection at the same concentration was more significant in the young age rats. In conclusion, XBJ injection attenuates HS-induced brain injury by inhibiting oxidative stress and Parthanatos via the PARP-1/AIF signaling, which might provide a novel therapeutic strategy for HS treatment.
Collapse
Affiliation(s)
- Lin Wang
- Department
of Emergency, The First Affiliated Hospital
of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Boxin Ye
- The
First Clinical Medical School, Guangzhou
University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Yongrui Liu
- The
First Clinical Medical School, Guangzhou
University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Jun Li
- The
First Clinical Medical School, Guangzhou
University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Chunhe Li
- Department
of Critical Care Medicine, The First Affiliated
Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Minyong Wen
- Department
of Critical Care Medicine, The First Affiliated
Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Hongbo Li
- Department
of Critical Care Medicine, The First Affiliated
Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| |
Collapse
|
3
|
Li F, Deng J, He Q, Zhong Y. ZBP1 and heatstroke. Front Immunol 2023; 14:1091766. [PMID: 36845119 PMCID: PMC9950778 DOI: 10.3389/fimmu.2023.1091766] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/19/2023] [Indexed: 02/12/2023] Open
Abstract
Heatstroke, which is associated with circulatory failure and multiple organ dysfunction, is a heat stress-induced life-threatening condition characterized by a raised core body temperature and central nervous system dysfunction. As global warming continues to worsen, heatstroke is expected to become the leading cause of death globally. Despite the severity of this condition, the detailed mechanisms that underlie the pathogenesis of heatstroke still remain largely unknown. Z-DNA-binding protein 1 (ZBP1), also referred to as DNA-dependent activator of IFN-regulatory factors (DAI) and DLM-1, was initially identified as a tumor-associated and interferon (IFN)-inducible protein, but has recently been reported to be a Z-nucleic acid sensor that regulates cell death and inflammation; however, its biological function is not yet fully understood. In the present study, a brief review of the main regulators is presented, in which the Z-nucleic acid sensor ZBP1 was identified to be a significant factor in regulating the pathological characteristics of heatstroke through ZBP1-dependent signaling. Thus, the lethal mechanism of heatstroke is revealed, in addition to a second function of ZBP1 other than as a nucleic acid sensor.
Collapse
Affiliation(s)
- Fanglin Li
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China,Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Jiayi Deng
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiuli He
- Department of Nephrology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China,*Correspondence: Qiuli He, ; Yanjun Zhong,
| | - Yanjun Zhong
- Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Qiuli He, ; Yanjun Zhong,
| |
Collapse
|
4
|
Sun Y, Koyama Y, Shimada S. Inflammation From Peripheral Organs to the Brain: How Does Systemic Inflammation Cause Neuroinflammation? Front Aging Neurosci 2022; 14:903455. [PMID: 35783147 PMCID: PMC9244793 DOI: 10.3389/fnagi.2022.903455] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
As inflammation in the brain contributes to several neurological and psychiatric diseases, the cause of neuroinflammation is being widely studied. The causes of neuroinflammation can be roughly divided into the following domains: viral infection, autoimmune disease, inflammation from peripheral organs, mental stress, metabolic disorders, and lifestyle. In particular, the effects of neuroinflammation caused by inflammation of peripheral organs have yet unclear mechanisms. Many diseases, such as gastrointestinal inflammation, chronic obstructive pulmonary disease, rheumatoid arthritis, dermatitis, chronic fatigue syndrome, or myalgic encephalomyelitis (CFS/ME), trigger neuroinflammation through several pathways. The mechanisms of action for peripheral inflammation-induced neuroinflammation include disruption of the blood-brain barrier, activation of glial cells associated with systemic immune activation, and effects on autonomic nerves via the organ-brain axis. In this review, we consider previous studies on the relationship between systemic inflammation and neuroinflammation, focusing on the brain regions susceptible to inflammation.
Collapse
Affiliation(s)
- Yuanjie Sun
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
- *Correspondence: Yoshihisa Koyama, ; orcid.org/0000-0003-3965-0716
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| |
Collapse
|
5
|
Bouchama A, Abuyassin B, Lehe C, Laitano O, Jay O, O'Connor FG, Leon LR. Classic and exertional heatstroke. Nat Rev Dis Primers 2022; 8:8. [PMID: 35115565 DOI: 10.1038/s41572-021-00334-6] [Citation(s) in RCA: 140] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2021] [Indexed: 12/28/2022]
Abstract
In the past two decades, record-breaking heatwaves have caused an increasing number of heat-related deaths, including heatstroke, globally. Heatstroke is a heat illness characterized by the rapid rise of core body temperature above 40 °C and central nervous system dysfunction. It is categorized as classic when it results from passive exposure to extreme environmental heat and as exertional when it develops during strenuous exercise. Classic heatstroke occurs in epidemic form and contributes to 9-37% of heat-related fatalities during heatwaves. Exertional heatstroke sporadically affects predominantly young and healthy individuals. Under intensive care, mortality reaches 26.5% and 63.2% in exertional and classic heatstroke, respectively. Pathological studies disclose endothelial cell injury, inflammation, widespread thrombosis and bleeding in most organs. Survivors of heatstroke may experience long-term neurological and cardiovascular complications with a persistent risk of death. No specific therapy other than rapid cooling is available. Physiological and morphological factors contribute to the susceptibility to heatstroke. Future research should identify genetic factors that further describe individual heat illness risk and form the basis of precision-based public health response. Prioritizing research towards fundamental mechanism and diagnostic biomarker discovery is crucial for the design of specific management approaches.
Collapse
Affiliation(s)
- Abderrezak Bouchama
- King Abdullah International Medical Research Center, Experimental Medicine Department, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia.
| | - Bisher Abuyassin
- King Abdullah International Medical Research Center, Experimental Medicine Department, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
| | - Cynthia Lehe
- King Abdullah International Medical Research Center, Experimental Medicine Department, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
| | - Orlando Laitano
- Department of Nutrition & Integrative Physiology, College of Health and Human Sciences, Florida State University, Tallahassee, FL, USA
| | - Ollie Jay
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Francis G O'Connor
- Military and Emergency Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Lisa R Leon
- Thermal and Mountain Medicine Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| |
Collapse
|
6
|
Eskandari M, Mellati AA. Liver X Receptor as a Possible Drug Target for Blood-Brain Barrier Integrity. Adv Pharm Bull 2021; 12:466-475. [PMID: 35935038 PMCID: PMC9348539 DOI: 10.34172/apb.2022.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/13/2021] [Indexed: 12/04/2022] Open
Abstract
Purpose: blood-brain barrier (BBB) is made of specialized cells that are responsible for the selective passage of substances directed to the brain. The integrated BBB is essential for precise controlling of the different substances passage as well as protecting the brain from various damages. In this article, we attempted to explain the role of liver X receptor (LXR) in maintaining BBB integrity as a possible drug target.
Methods: In this study, various databases, including PubMed, Google Scholar, and Scopus were searched using the following keywords: blood-brain barrier, BBB, liver X receptor, and LXR until July, 2020. Additionally, contents close to the subject of our study were surveyed.
Results: LXR is a receptor the roles of which in various diseases have been investigated. LXR can affect maintaining BBB by affecting various ways such as ATP-binding cassette transporter A1 (ABCA1), matrix metalloproteinase-9 (MMP9), insulin-like growth factor 1 (IGF1), nuclear factor-kappa B (NF-κB) signaling, mitogen-activated protein kinase (MAPK), tight junction molecules, both signal transducer and activator of transcription 1 (STAT1), Wnt/β-catenin Signaling, transforming growth factor beta (TGF-β) signaling, and expressions of Smad 2/3 and Snail.
Conclusion: LXR could possibly be used either as a target for drug delivery to brain tissue or as a target for maintaining the BBB integrity in different diseases; thereby the drug will be conducted to tissues, other than the brain. If it is verified that only LXRα is necessary for protecting BBB, some specific LXRα ligands must be found and then used in medication.
Collapse
Affiliation(s)
- Mahsa Eskandari
- Medical school, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Awsat Mellati
- Zanjan Metabolic Disease Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
7
|
Wang L, Zhao J, Zhu B, Shen J, Ye Z, Peng Q, Zhang Y. Microglia polarization in heat-induced early neural injury. Arch Med Sci 2021; 20:1307-1313. [PMID: 39439680 PMCID: PMC11493046 DOI: 10.5114/aoms/116550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/12/2020] [Indexed: 10/25/2024] Open
Abstract
Introduction In the occurrence and development of heat stroke (HS), factors such as hyperthermia, ischemia and hypoxia are essential to the central nervous system (CNS) inflammatory response, but the main mechanism underlying CNS inflammation remains unclear. The aim of the study was to observe the polarization of microglia in response to heat-induced early nerve injury and to explore its possible mechanism of action. Material and methods To establish a heatstroke animal model in Beagle dogs, 18 Beagle dogs were divided into control (group A) and experimental groups (group B, group C and group D) according to a random numbers table. The animals in the experimental groups were placed on an electric blanket of an animal body temperature maintaining apparatus. The temperature was set at 40 ±0.5°C, and the rectal temperature was monitored every 5 min until the target body temperature was reached. Once the target temperature was reached, the dogs were transferred to an environment of 26 ±0.5°C and 60 ±0.5% humidity. Western blot analysis was used to detect the expression of microglia-specific markers CD45, iNOS, arginase, and CD206 in normal and heat-damaged brain tissues at different time points (1 h, 6 h, 24 h). The expression of CD45 and arginase was further determined by co-localization with immunofluorescence. Results CD45 and iNOS protein expression was detected in group A. The two protein markers in group B were significantly higher than those in group A (p < 0.05), and the protein markers in group C were still higher than those in group A (p < 0.05). There was no statistically significant difference among the animals in group A (p > 0.05). Arginase and CD206 protein expression was also detected in group A. Levels of the two protein markers in group B were higher than those in group A (p < 0.05), and the protein marker levels in group C were even higher than those in group A (p < 0.05). Further analysis of the two groups of protein markers in group D showed significantly higher levels than those in group A (p < 0.001). Immunofluorescence co-localization of CD45 and arginase showed significantly increased fluorescence density at 6 h and 24 h after thermal injury (p < 0.001). Conclusions After heat-induced disease, microglia were found to be active in the brain tissues of dogs. The microglia activated in the early 1-6 h of CNS injury were mainly the M1 type, which were then converted to the M2 type after 6 h. The 24 h M2 type was dominant. The relationship between M1/M2 polarization trends and early brain injury in heat-induced disease may be a key to understanding CNS injury in heat-induced disease.
Collapse
Affiliation(s)
- Lei Wang
- Department of Emergency Center, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Jing Zhao
- Department of Dermatology, Affiliated Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Baofeng Zhu
- Department of Emergency Center, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Junhua Shen
- Department of Emergency Center, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Zi Ye
- Department of Neurosurgery, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Qiang Peng
- Department of Emergency Center, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Yi Zhang
- Department of Neurosurgery, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
8
|
Li P, Wang G, Zhang XL, He GL, Luo X, Yang J, Luo Z, Shen TT, Yang XS. MicroRNA-155 Promotes Heat Stress-Induced Inflammation via Targeting Liver X Receptor α in Microglia. Front Cell Neurosci 2019; 13:12. [PMID: 30778287 PMCID: PMC6369214 DOI: 10.3389/fncel.2019.00012] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background: The neuroinflammatory responses of microglial cells play an important role in the process of brain dysfunction caused by heat stroke. MicroRNAs are reportedly involved in a complex signaling network and have been identified as neuroinflammatory regulators. In this study, we determined the biological roles of microRNA-155 in the inflammatory responses in heat-stressed microglia and explored the underlying mechanisms. Methods: MicroRNA-155 mimic and inhibitor were used to separately upregulate or downregulate microRNA-155 expression. The activation state of BV-2 microglial cells (BV-2 cells) was assessed via immunoreactions using the microglial marker CD11b and CD68. Levels of induced interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were measured using real-time reverse transcription polymerase chain reaction (RT-PCR) and enzyme linked immunosorbent assays (ELISAs). The activation of nuclear factor kappa B (NF-κB) signaling proteins was evaluated by Western blotting for inhibitory kappa B alpha (IκBα) and NF-κB p65 phosphorylation and indirect immunofluorescence analysis using a p65 phosphorylation antibody. A luciferase reporter assay was used to verify liver X receptor α (LXRα) as a target gene of microRNA-155. Results: Heat stress significantly induced IL-1β, IL-6, and TNF-α release and increased the expression of CD11b and CD68. In addition, IκBα and NF-κB p65 phosphorylation were dramatically increased by heat stress, and microRNA-155 expression was also elevated. High expression of microRNA-155 in heat-stressed microglial cells was inversely correlated with LXRα expression. We then determined the role of microRNA-155 in the heat stress-induced inflammatory responses. The results revealed that by targeting LXRα, microRNA-155 enhanced NF-κB signaling activation and facilitated immune inflammation in heat stress-treated BV-2 cells. Conclusion: MicroRNA-155 promotes heat stress-induced inflammatory responses in microglia. The underlying mechanisms may include facilitating inflammatory factors expression by increasing NF-κB pathway activation via targeting LXRα.
Collapse
Affiliation(s)
- Ping Li
- Laboratory of Extreme Environmental Medicine, Department of Tropical Medicine, Army Medical University, Chongqing, China
| | - Gong Wang
- Laboratory of Extreme Environmental Medicine, Department of Tropical Medicine, Army Medical University, Chongqing, China.,Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiao-Liang Zhang
- Laboratory of Extreme Environmental Medicine, Department of Tropical Medicine, Army Medical University, Chongqing, China.,Department of Cardiology, Kunming General Hospital of Chengdu Military Command, Yunnan, China
| | - Gen-Lin He
- Laboratory of Extreme Environmental Medicine, Department of Tropical Medicine, Army Medical University, Chongqing, China
| | - Xue Luo
- Laboratory of Extreme Environmental Medicine, Department of Tropical Medicine, Army Medical University, Chongqing, China
| | - Ju Yang
- Laboratory of Extreme Environmental Medicine, Department of Tropical Medicine, Army Medical University, Chongqing, China
| | - Zhen Luo
- Laboratory of Extreme Environmental Medicine, Department of Tropical Medicine, Army Medical University, Chongqing, China
| | - Ting-Ting Shen
- Laboratory of Extreme Environmental Medicine, Department of Tropical Medicine, Army Medical University, Chongqing, China
| | - Xue-Sen Yang
- Laboratory of Extreme Environmental Medicine, Department of Tropical Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
9
|
Shen R, Lin S, He L, Zhu X, Zhou Z, Chen S, Wang Y, Ding J. Association of Two Polymorphisms in CCL2 With Parkinson's Disease: A Case-Control Study. Front Neurol 2019; 10:35. [PMID: 30761072 PMCID: PMC6362632 DOI: 10.3389/fneur.2019.00035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 01/11/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Parkinson's disease (PD) is the most common neurodegenerative movement disorder that is known to be related to neuro-inflammation. Chemokines participate in this process usually through upregulation of expression levels, which are closely related to the polymorphisms in their genes. Recent studies have further revealed the association between these polymorphisms and the risk of PD in multiple populations, but not the Chinese Han population. Methods:The promoter region of CCL2 was sequenced in 411 PD patients and 422 gender-age matched control from a Chinese Han population using PCR-RFLP method. Their genotype frequencies were analyzed statistically. Dual-luciferase reporter assays were conducted in neuroblastoma cells to assess the promoter transcriptional activity of the rs1024611 variants (T>C) and the GRCh38.p12chr17:34252593 G>C alleles in CCL2. Results:We found that the frequency of the CCL2 genotype of rs1024611 was significantly different between the PD and control groups (p = 0.021), while the C allele was associated with a significantly increased risk in the PD group (p = 0.004). Moreover, C allele of this newly identified alteration in CCL2 (GRCh38.p12chr17:34252593 G>C) was also found to be associated with an increased risk of PD (P genotype = 0.006, P allele = 0.006). Dual-luciferase reporter assay results indicated that rs1024611 C allele and GRCh38.p12chr17:.34252593 C allele increased the transcriptional activity of the CCL2 promoter. Conclusions: We, for the first time, report a risk polymorphism (rs1024611) and a new locus (GRCh38.p12chr17:.34252593 G>C) on CCL2, both of which are suggested as risk factors for PD in a Chinese Han population.
Collapse
Affiliation(s)
- Ruinan Shen
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suzhen Lin
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu He
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Zhu
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhekun Zhou
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shengdi Chen
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianqing Ding
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Dexmedetomidine Protects Against Multi-Organ Dysfunction Induced by Heatstroke via Sustaining The Intestinal Integrity. Shock 2018; 48:260-269. [PMID: 28709158 DOI: 10.1097/shk.0000000000000826] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Previous studies have indicated that gut-derived endotoxin played a pivotal role for aggravating systemic inflammatory response to multi-organ dysfunction under heatstroke. Dexmedetomidine (DEX) could protect against inflammation and multi-organ injury in various scenarios. The aim of this study was to explore the protective effect of DEX on heatstroke and the mechanism involved. Male C57BL/6 mice were placed in a controlled climate chamber (40 ± 1°C) until the maximum core temperature (Tc, Max) of 42.7°C, the received criterion of heatstroke, was attained, DEX (25 μg/kg) or 0.9% saline was injected intraperitoneally immediately. The results showed that DEX could significantly attenuate multi-organ injury induced by heatstroke, simultaneously decrease levels of serum inflammatory cytokines through inhibiting the intestinal nuclear factor-κB activation. Furthermore, to assess the effects of DEX on intestine mucosal barrier under heatstroke, the levels of plasma endotoxin, FD4, and D-lactate were detected and the expression of tight junction proteins occludin and ZO-1 was analyzed by western blot and immunohistochemistry. Meanwhile, transmission electron microscopy was employed to confirm the ultrastructure of intestine. Interestingly, we found that DEX decreased the intestinal permeability and sustained the integrity of intestinal barrier. Finally, to evaluate the anti-apoptosis effect of DEX, the pro-apoptotic protein Bax and anti-apoptotic protein Bcl-2 were analyzed by western blot, and terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling (TUNEL) staining was conducted. The results showed that DEX decreased TUNEL-positive cells induced by heatstroke in a Bax/Bcl-2-related manner. Taken together, our results indicate that DEX could protect against inflammation and multi-organ injury induced by heatstroke via sustaining the intestinal integrity.
Collapse
|
11
|
Lin YF, Liu TT, Hu CH, Chen CC, Wang JY. Expressions of chemokines and their receptors in the brain after heat stroke-induced cortical damage. J Neuroimmunol 2018; 318:15-20. [PMID: 29395321 DOI: 10.1016/j.jneuroim.2018.01.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 01/06/2018] [Accepted: 01/22/2018] [Indexed: 01/11/2023]
Abstract
Despite growing evidence that cytokines and chemokines are expressed in humans and rats after heat stress, the cellular mechanisms underlying the effects on the brain after heatstroke (HS) are not fully understood. In this study, we observed time course changes of chemokines in rat brain tissues and elucidated what kinds of cortical cells were affected after HS. Male SD rats were anesthetized and randomly separated into two groups as follows: (a) normothermic sham and (b) HS rats. Rats were sacrificed at different time points (0, 1, 3, 6, and 12h after heat exposure, n=5 in each group) to the end of the experiment in order to extract the mRNA/proteins of cortical tissues. Cerebrospinal fluid (CSF) of sham and HS rats was also collected before sacrifice. In the HS group, an elevated body temperature (Tco>40°C) and abnormality of cortical cells (e.g., pyknotic nuclei) were observed. When compared to the sham group, expression levels of either mRNAs or proteins of chemokines and their receptors (including CXCL1, MIP2, MCP1, CXCR1, CXCR2, and CCR2) peaked at different time points after heat exposure. We also found that CXCR2 was expressed in the cortex of rat brain and was colocalized with neurons and microglia after HS. Hence, MCP1, MIP2, and CXCR2 might play important roles in the brain after HS, possibly indicating a new direction for treating HS.
Collapse
Affiliation(s)
- Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Tsung-Ta Liu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Center for General Education, University of Kang Ning, Taipei, Taiwan, ROC
| | - Chou-Hui Hu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Chun-Chi Chen
- Department of Internal Medicine, Division of Nephrology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
12
|
Abstract
Heatstroke is an acute medical emergency that is always fatal if left untreated. The diagnosis of heatstroke should be considered in any hyperthermic patient with altered mental status during a heat wave or following vigorous muscle exertion. Heat can damage the structure and function of essential macromolecules, including proteins, membrane lipids, and nucleic acids, and thereby lead to multiple-organ failure, culminating in death. The cytotoxic effect of heat is a function of degree and duration of hyperthermia; thus, an early diagnosis and prompt initiation of cooling are paramount to halt progression to tissue damage and death.
Collapse
Affiliation(s)
- Saeed Al Mahri
- Intensive Care Unit and Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abderrezak Bouchama
- Intensive Care Unit and Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.
| |
Collapse
|
13
|
Transcriptome Sequencing Reveals Astrocytes as a Therapeutic Target in Heat-Stroke. Neurosci Bull 2017; 33:627-640. [PMID: 28699024 DOI: 10.1007/s12264-017-0156-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/24/2017] [Indexed: 10/19/2022] Open
Abstract
Heat-stroke is a serious form of hyperthermia with high mortality, and can induce severe central nervous system disorders. The neurovascular unit (NVU), which consists of vascular cells, glial cells, and neurons, controls blood-brain barrier (BBB) permeability and cerebral blood flow, and maintains the proper functioning of neuronal circuits. However, the detailed function of each BBB component in heat-stroke remains unknown. In order to interpret alterations caused by heat stress, we performed transcriptome comparison of neuron and astrocyte primary cultures after heat treatment. Differentially-expressed genes were then selected and underwent Gene Ontology annotation and Kyoto Encyclopedia of Genes and Genomes pathway analysis. Gene-act networks were also constructed, and the expression of pivotal genes was validated by quantitative PCR, as well as single-cell qPCR in heat-stroke rats. Our work provides valuable information on the transcriptional changes in NVU cells after heat stress, reveals the diverse regulatory mechanisms of two of these cellular components, and shows that a cell-type-specific approach may be a promising therapeutic strategy for heat-stroke treatments.
Collapse
|
14
|
Pearce SC, Sanz Fernandez MV, Torrison J, Wilson ME, Baumgard LH, Gabler NK. Dietary organic zinc attenuates heat stress-induced changes in pig intestinal integrity and metabolism. J Anim Sci 2016; 93:4702-13. [PMID: 26523563 DOI: 10.2527/jas.2015-9018] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dietary zinc (inorganic and organic or zinc AA complex forms) is essential for normal intestinal barrier function and regeneration of intestinal epithelium. Given that heat stress (HS) exposure can negatively affect intestinal integrity and caloric intake, possible nutritional mitigation strategies are needed to improve health, performance, and well-being. Therefore, our objective was to evaluate 2 dietary zinc sources and reduced caloric intake on intestinal integrity in growing pigs subjected to 12 h of HS. A total of 36 pigs were fed 1 of 2 diets: 1) a control diet (CON; 120 mg/kg of zinc from zinc sulfate) or 2) 60 mg/kg from zinc sulfate and 60 mg/kg from zinc AA complex (ZnAA). After 17 d, the CON pigs were then exposed to thermal neutral (TN) conditions with ad libitum intake (TN-CON), HS (37°C) with ad libitum intake (HS-CON), or pair-fed to HS intake under TN conditions (PFTN); the ZnAA pigs were exposed to only HS (HS-ZnAA). All pigs were sacrificed after 12 h of environmental exposure, and blood and tissue bioenergetics stress markers and ex vivo ileum and colon integrity were assessed. Compared with TN-CON, HS significantly ( < 0.05) increased rectal temperatures and respiration rates. Ileum villus and crypt morphology was reduced by both pair-feeding and HS. Both PFTN and HS-CON pigs also had reduced ileum integrity (dextran flux and transepithelial resistance) compared with the TN-CON pigs. However, ZnAA tended to mitigate the HS-induced changes in ileum integrity. Ileum mucin 2 protein abundance was increased due to HS and pair-feeding. Colonic integrity did not differ due to HS or PFTN treatments. Compared with the HS-CON, HS-ZnAA pigs tended to have reduced blood endotoxin concentrations. In conclusion, HS and reduced feed intake compromised intestinal integrity in pigs, and zinc AA complex source mitigates some of these negative effects.
Collapse
|
15
|
Audet GN, Dineen SM, Quinn CM, Leon LR. Altered hypothalamic inflammatory gene expression correlates with heat stroke severity in a conscious rodent model. Brain Res 2016; 1637:81-90. [DOI: 10.1016/j.brainres.2016.01.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/03/2015] [Accepted: 01/26/2016] [Indexed: 12/31/2022]
|
16
|
Abstract
Heat stroke is a life-threatening condition clinically diagnosed as a severe elevation in body temperature with central nervous system dysfunction that often includes combativeness, delirium, seizures, and coma. Classic heat stroke primarily occurs in immunocompromised individuals during annual heat waves. Exertional heat stroke is observed in young fit individuals performing strenuous physical activity in hot or temperature environments. Long-term consequences of heat stroke are thought to be due to a systemic inflammatory response syndrome. This article provides a comprehensive review of recent advances in the identification of risk factors that predispose to heat stroke, the role of endotoxin and cytokines in mediation of multi-organ damage, the incidence of hypothermia and fever during heat stroke recovery, clinical biomarkers of organ damage severity, and protective cooling strategies. Risk factors include environmental factors, medications, drug use, compromised health status, and genetic conditions. The role of endotoxin and cytokines is discussed in the framework of research conducted over 30 years ago that requires reassessment to more clearly identify the role of these factors in the systemic inflammatory response syndrome. We challenge the notion that hypothalamic damage is responsible for thermoregulatory disturbances during heat stroke recovery and highlight recent advances in our understanding of the regulated nature of these responses. The need for more sensitive clinical biomarkers of organ damage is examined. Conventional and emerging cooling methods are discussed with reference to protection against peripheral organ damage and selective brain cooling.
Collapse
Affiliation(s)
- Lisa R Leon
- US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Abderrezak Bouchama
- King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, Experimental Medicine Department-King Abdulaziz Medical City-Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Audet GN, Quinn CM, Leon LR. Point-of-care cardiac troponin test accurately predicts heat stroke severity in rats. Am J Physiol Regul Integr Comp Physiol 2015; 309:R1264-72. [PMID: 26290107 DOI: 10.1152/ajpregu.00286.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/14/2015] [Indexed: 12/28/2022]
Abstract
Heat stroke (HS) remains a significant public health concern. Despite the substantial threat posed by HS, there is still no field or clinical test of HS severity. We suggested previously that circulating cardiac troponin (cTnI) could serve as a robust biomarker of HS severity after heating. In the present study, we hypothesized that (cTnI) point-of-care test (ctPOC) could be used to predict severity and organ damage at the onset of HS. Conscious male Fischer 344 rats (n = 16) continuously monitored for heart rate (HR), blood pressure (BP), and core temperature (Tc) (radiotelemetry) were heated to maximum Tc (Tc,Max) of 41.9 ± 0.1°C and recovered undisturbed for 24 h at an ambient temperature of 20°C. Blood samples were taken at Tc,Max and 24 h after heat via submandibular bleed and analyzed on ctPOC test. POC cTnI band intensity was ranked using a simple four-point scale via two blinded observers and compared with cTnI levels measured by a clinical blood analyzer. Blood was also analyzed for biomarkers of systemic organ damage. HS severity, as previously defined using HR, BP, and recovery Tc profile during heat exposure, correlated strongly with cTnI (R(2) = 0.69) at Tc,Max. POC cTnI band intensity ranking accurately predicted cTnI levels (R(2) = 0.64) and HS severity (R(2) = 0.83). Five markers of systemic organ damage also correlated with ctPOC score (albumin, alanine aminotransferase, blood urea nitrogen, cholesterol, and total bilirubin; R(2) > 0.4). This suggests that cTnI POC tests can accurately determine HS severity and could serve as simple, portable, cost-effective HS field tests.
Collapse
Affiliation(s)
- Gerald N Audet
- Thermal and Mountain Medicine Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | - Carrie M Quinn
- Thermal and Mountain Medicine Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | - Lisa R Leon
- Thermal and Mountain Medicine Division, United States Army Research Institute of Environmental Medicine, Natick, Massachusetts
| |
Collapse
|
18
|
Wen YT, Liu TT, Lin YF, Chen CC, Kung WM, Huang CC, Lin TJ, Wang YH, Wei L. Heatstroke Effect on Brain Heme Oxygenase-1 in Rats. Int J Med Sci 2015; 12:737-41. [PMID: 26392811 PMCID: PMC4571551 DOI: 10.7150/ijms.12517] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/09/2015] [Indexed: 02/05/2023] Open
Abstract
Exposure to high environmental temperature leading to increased core body temperature above 40°C and central nervous system abnormalities such as convulsions, delirium, or coma is defined as heat stroke. Studies in humans and animals indicate that the heat shock responses of the host contribute to multiple organ injury and death during heat stroke. Heme oxygenase-1 (HO-1)-a stress-responsive enzyme that catabolizes heme into iron, carbon monoxide, and biliverdin-has an important role in the neuroprotective mechanism against ischemic stroke. Here, we investigated the role of endogenous HO-1 in heat-induced brain damage in rats. RT-PCR results revealed that levels of HO-1 mRNA peaked at 0 h after heat exposure and immunoblot analysis revealed that the maximal protein expression occurred at 1 h post-heat exposure. Subsequently, we detected the HO-1 expression in the cortical brain cells and revealed the neuronal cell morphology. In conclusion, HO-1 is a potent protective molecule against heat-induced brain damage. Manipulation of HO-1 may provide a potential therapeutic approach for heat-related diseases.
Collapse
Affiliation(s)
- Ya-Ting Wen
- 1. Department of Neurosurgery, Taipei Medical University-Wan Fang Hospital, Taipei 11696, Taiwan
| | - Tsung-Ta Liu
- 2. Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan
| | - Yuh-Feng Lin
- 3. Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chun-Chi Chen
- 4. Division of Nephrology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Woon-Man Kung
- 5. Department of Neurosurgery, Lo-Hsu Foundation, Lotung Poh-Ai Hospital, Luodong, Yilan 265, Taiwan
- 6. Department of Exercise and Health Promotion, College of Education, Chinese Culture University, Taipei 11114, Taiwan
| | - Chi-Chang Huang
- 7. Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 33301, Taiwan
| | - Tien-Jen Lin
- 1. Department of Neurosurgery, Taipei Medical University-Wan Fang Hospital, Taipei 11696, Taiwan
- 8. Graduate Institute of Injury Prevention and Control, College of Public Health and Nutrition, Taipei Medical University, Taipei 11031, Taiwan
| | - Yuan-Hung Wang
- 3. Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- 9. Division of General Surgery, Department of Urology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- ✉ Corresponding authors: Dr. Li Wei: Department of Neurosurgery, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan. Tel.: +886-2-29307930 (ext. 6942). E-Mail: . Dr. Yuan-Hung Wang: Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. Tel.: +886-2-22490088 (ext. 8891). E-Mail:
| | - Li Wei
- 1. Department of Neurosurgery, Taipei Medical University-Wan Fang Hospital, Taipei 11696, Taiwan
- 10. The PhD Program of Translational Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- ✉ Corresponding authors: Dr. Li Wei: Department of Neurosurgery, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan. Tel.: +886-2-29307930 (ext. 6942). E-Mail: . Dr. Yuan-Hung Wang: Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. Tel.: +886-2-22490088 (ext. 8891). E-Mail:
| |
Collapse
|