1
|
Koutsogiannaki S, Limratana P, Bu W, Maisat W, McKinstry-Wu A, Han X, Ohto U, Eckenhoff RG, Soriano SG, Yuki K. Dexmedetomidine directly binds to and inhibits Toll-like receptor 4. Int Immunopharmacol 2024; 141:112975. [PMID: 39163686 PMCID: PMC11408083 DOI: 10.1016/j.intimp.2024.112975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND While a number of anesthetics has been shown potentially associated with neurotoxicity in the developing brain, dexmedetomidine, a drug that was rather recently introduced into the perioperative setting, is considered beneficial from neurological wellbeing. However, the underlying mechanism of how dexmedetomidine affects brain health remains to be determined. Based on our recent study, we hypothesized that dexmedetomidine would directly bind to and inhibit Toll-like receptor 4 (TLR4), a critical receptor largely expressed in microglia and responsible for neurological insult. METHODS We used TLR4 reporter assays to test if dexmedetomidine attenuates TLR4 activation. Furthermore, a direct binding of dexmedetomidine on TLR4 was tested using photoactivatable medetomidine. Lastly, the effect of dexmedetomidine on ketamine (anesthetic)-induced neurotoxicity was tested in rat pups (P7). RESULTS We showed that dexmedetomidine attenuated TLR4 activation using reporter assay (IC50 = 5.8 µg/mL). Photoactivatable dexmedetomidine delineated its direct binding sites on TLR4. We also showed that dexmedetomidine attenuated microglia activation both in vitro and in vivo. DISCUSSION We proposed a novel mechanism of dexmedetomidine-mediated neuroprotection.
Collapse
Affiliation(s)
- Sophia Koutsogiannaki
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA; Department of Immunology, Harvard Medical School, Boston, USA
| | - Panop Limratana
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA; Department of Immunology, Harvard Medical School, Boston, USA; Department of Anesthesiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Weiming Bu
- Department of Anesthesia and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Wiriya Maisat
- Department of Anesthesiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Andrew McKinstry-Wu
- Department of Anesthesia and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Xiaohui Han
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA
| | - Umeharu Ohto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Roderic G Eckenhoff
- Department of Anesthesia and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Sulpicio G Soriano
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, USA; Department of Anaesthesia, Harvard Medical School, Boston, USA; Department of Immunology, Harvard Medical School, Boston, USA.
| |
Collapse
|
2
|
Khayatan D, Razavi SM, Arab ZN, Khanahmadi M, Samanian A, Momtaz S, Sukhorukov VN, Jamialahmadi T, Abdolghaffari AH, Barreto GE, Sahebkar A. Protective Effects of Plant-Derived Compounds Against Traumatic Brain Injury. Mol Neurobiol 2024; 61:7732-7750. [PMID: 38427213 DOI: 10.1007/s12035-024-04030-w] [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: 09/26/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Inflammation in the nervous system is one of the key features of many neurodegenerative diseases. It is increasingly being identified as a critical pathophysiological primitive mechanism associated with chronic neurodegenerative diseases following traumatic brain injury (TBI). Phytochemicals have a wide range of clinical properties due to their antioxidant and anti-inflammatory effects. Currently, there are few drugs available for the treatment of neurodegenerative diseases other than symptomatic relief. Numerous studies have shown that plant-derived compounds, in particular polyphenols, protect against various neurodegenerative diseases and are safe for consumption. Polyphenols exert protective effects on TBI via restoration of nuclear factor kappa B (NF-κB), toll-like receptor-4 (TLR4), and Nod-like receptor family proteins (NLRPs) pathways. In addition, these phytochemicals and their derivatives upregulate the phosphatidylinositol-3-Kinase/Protein Kinase B (PI3K/AKT) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways, which have critical functions in modulating TBI symptoms. There is supporting evidence that medicinal plants and phytochemicals are protective in different TBI models, though future clinical trials are needed to clarify the precise mechanisms and functions of different polyphenolic compounds in TBI.
Collapse
Affiliation(s)
- Danial Khayatan
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Seyed Mehrad Razavi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zahra Najafi Arab
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Khanahmadi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirreza Samanian
- Department of Neurology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeideh Momtaz
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Vasily N Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, Moscow, 121609, Russia
- Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| | - Amirhossein Sahebkar
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Khayatan D, Razavi SM, Arab ZN, Niknejad AH, Nouri K, Momtaz S, Gumpricht E, Jamialahmadi T, Abdolghaffari AH, Barreto GE, Sahebkar A. Protective effects of curcumin against traumatic brain injury. Biomed Pharmacother 2022; 154:113621. [DOI: 10.1016/j.biopha.2022.113621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 02/06/2023] Open
|
4
|
Hu Y, Zhou H, Zhang H, Sui Y, Zhang Z, Zou Y, Li K, Zhao Y, Xie J, Zhang L. The neuroprotective effect of dexmedetomidine and its mechanism. Front Pharmacol 2022; 13:965661. [PMID: 36204225 PMCID: PMC9531148 DOI: 10.3389/fphar.2022.965661] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/16/2022] [Indexed: 11/28/2022] Open
Abstract
Dexmedetomidine (DEX) is a highly selective α2 receptor agonist that is routinely used in the clinic for sedation and anesthesia. Recently, an increasing number of studies have shown that DEX has a protective effect against brain injury caused by traumatic brain injury (TBI), subarachnoid hemorrhage (SAH), cerebral ischemia and ischemia–reperfusion (I/R), suggesting its potential as a neuroprotective agent. Here, we summarized the neuroprotective effects of DEX in several models of neurological damage and examined its mechanism based on the current literature. Ultimately, we found that the neuroprotective effect of DEX mainly involved inhibition of inflammatory reactions, reduction of apoptosis and autophagy, and protection of the blood–brain barrier and enhancement of stable cell structures in five way. Therefore, DEX can provide a crucial advantage in neurological recovery for patients with brain injury. The purpose of this study was to further clarify the neuroprotective mechanisms of DEX therefore suggesting its potential in the clinical management of the neurological injuries.
Collapse
Affiliation(s)
- Yijun Hu
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
- Graduate School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong Zhou
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Huanxin Zhang
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Yunlong Sui
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Zhen Zhang
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Yuntao Zou
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Kunquan Li
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Yunyi Zhao
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Jiangbo Xie
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Lunzhong Zhang
- Neurology Department, Weifang Hospital of Traditional Chinese Medicine, Weifang, China
- *Correspondence: Lunzhong Zhang,
| |
Collapse
|
5
|
Xu X, Zhi T, Hua L, Jiang K, Chen C. IRAK4 exacerbates traumatic brain injury via activation of TAK1 signaling pathway. Exp Neurol 2022; 351:114007. [PMID: 35149117 DOI: 10.1016/j.expneurol.2022.114007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 01/29/2022] [Accepted: 02/04/2022] [Indexed: 11/04/2022]
Abstract
Although multiple signaling pathways contributing to the pathophysiological process have been investigated, treatments for traumatic brain injury (TBI) against present targets have not acquired significant clinical progress. Interleukin-1 receptor-associated kinase 4 (IRAK4) is an important factor involved in regulating immunity and inflammation. However, the role of IRAK4 in TBI still remains largely unknown. Therefore, using a controlled cortical impact model (CCI), we investigated the function and molecular mechanism of IRAK4 in the context of TBI. IRAK4 was found to be activated in a time-dependent manner after TBI and mainly expressed in neurons. Inhibition of IRAK4 by siRNAs could significantly alleviates neuroinflammation, neuron apoptosis, brain edema, brain-blood barrier (BBB) dysfunction and improves neurological deficit in the context of CCI. Mechanistically, IRAK4 exacerbates CCI via activation of TAK1 signaling pathway. Interestingly, PF-0665083, an IRAK4 inhibitor, inhibits phosphorylation of IRAK4 and attenuates CCI-induced secondary injury. It could be conclude that IRAK4 plays a critical role in TBI-induced secondary injury and the underlining mechanism may be related to activation of TAK1 signaling pathway. PF-0665083 may serve as a potential treatment strategy to relieve TBI.
Collapse
Affiliation(s)
- Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Tongle Zhi
- Department of Neurosurgery, The First People's Hospital of Yancheng, the Fourth Affiliated Hospital of Nantong University, Yancheng 224006, Jiangsu Province, China
| | - Lingyang Hua
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200000, China
| | - Kuan Jiang
- Department of Neurosurgery, Yixing People's Hospital, Yixing 214200, Jiangsu Province, China
| | - Chen Chen
- Department of Cardiology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing 210029, Jiangsu Province, China.
| |
Collapse
|
6
|
Hou Q, Chen H, Liu Q, Yan X. FGF10 Attenuates Experimental Traumatic Brain Injury through TLR4/MyD88/NF-κB Pathway. Cells Tissues Organs 2021; 209:248-256. [PMID: 33440393 DOI: 10.1159/000511381] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/27/2020] [Indexed: 11/19/2022] Open
Abstract
Traumatic brain injury (TBI) can induce neuronal apoptosis and neuroinflammation, resulting in substantial neuronal damage and behavioral disorders. Fibroblast growth factors (FGFs) have been shown to be critical mediators in tissue repair. However, the role of FGF10 in experimental TBI remains unknown. In this study, mice with TBI were established via weight-loss model and validated by increase of modified neurological severity scores (mNSS) and brain water content. Secondly, FGF10 levels were elevated in mice after TBI, whereas intraventricular injection of Ad-FGF10 decreased mNSS score and brain water content, indicating the remittance of neurological deficit and cerebral edema in TBI mice. In addition, neuronal damage could also be ameliorated by stereotactic injection of Ad-FGF10. Overexpression of FGF10 increased protein expression of Bcl-2, while it decreased Bax and cleaved caspase-3/PARP, and improved neuronal apoptosis in TBI mice. In addition, Ad-FGF10 relieved neuroinflammation induced by TBI and significantly reduced the level of interleukin 1β/6, tumor necrosis factor α, and monocyte chemoattractant protein-1. Moreover, Ad-FGF10 injection decreased the protein expression level of Toll-like receptor 4 (TLR4), MyD88, and phosphorylation of NF-κB (p-NF-κB), suggesting the inactivation of the TLR4/MyD88/NF-κB pathway. In conclusion, overexpression of FGF10 could ameliorate neurological deficit, neuronal apoptosis, and neuroinflammation through inhibition of the TLR4/MyD88/NF-κB pathway, providing a potential therapeutic strategy for brain injury in the future.
Collapse
Affiliation(s)
- Qinhan Hou
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| | - Hongmou Chen
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China,
| | - Quan Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| | - Xianlei Yan
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| |
Collapse
|
7
|
Ismael S, Ahmed HA, Adris T, Parveen K, Thakor P, Ishrat T. The NLRP3 inflammasome: a potential therapeutic target for traumatic brain injury. Neural Regen Res 2021; 16:49-57. [PMID: 32788447 PMCID: PMC7818859 DOI: 10.4103/1673-5374.286951] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although the precise mechanisms contributing to secondary brain injury following traumatic brain injury are complex and obscure, a number of studies have demonstrated that inflammatory responses are an obvious and early feature in the pathogenesis of traumatic brain injury. Inflammasomes are multiprotein complexes that prompt the stimulation of caspase-1 and subsequently induce the maturation and secretion of proinflammatory cytokines, such as interleukin-1β and interleukin-18. These cytokines play a pivotal role in facilitating innate immune responses and inflammation. Among various inflammasome complexes, the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is the best characterized, a crucial role for NLRP3 has been demonstrated in various brain diseases, including traumatic brain injury. Several recent studies have revealed the contribution of NLRP3 inflammasome in identifying cellular damage and stimulating inflammatory responses to aseptic tissue injury after traumatic brain injury. Even more important, blocking or inhibiting the activation of the NLRP3 inflammasome may have substantial potential to salvage tissue damage during traumatic brain injury. In this review, we summarize recently described mechanisms that are involved in the activation and regulation of the NLRP3 inflammasome. Moreover, we review the recent investigations on the contribution of the NLRP3 inflammasome in the pathophysiology of TBI, and current advances and challenges in potential NLRP3-targeted therapies. A significant contribution of NLRP3 inflammasome activation to traumatic brain injury implies that therapeutic approaches focused on targeting specific inflammasome components could significantly improve the traumatic brain injury outcomes.
Collapse
Affiliation(s)
- Saifudeen Ismael
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Heba A Ahmed
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tusita Adris
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Parth Thakor
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
8
|
Robb JL, Morrissey NA, Weightman Potter PG, Smithers HE, Beall C, Ellacott KLJ. Immunometabolic Changes in Glia - A Potential Role in the Pathophysiology of Obesity and Diabetes. Neuroscience 2020; 447:167-181. [PMID: 31765625 PMCID: PMC7567742 DOI: 10.1016/j.neuroscience.2019.10.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/15/2022]
Abstract
Chronic low-grade inflammation is a feature of the pathophysiology of obesity and diabetes in the CNS as well as peripheral tissues. Glial cells are critical mediators of the response to inflammation in the brain. Key features of glia include their metabolic flexibility, sensitivity to changes in the CNS microenvironment, and ability to rapidly adapt their function accordingly. They are specialised cells which cooperate to promote and preserve neuronal health, playing important roles in regulating the activity of neuronal networks across the brain during different life stages. Increasing evidence points to a role of glia, most notably astrocytes and microglia, in the systemic regulation of energy and glucose homeostasis in the course of normal physiological control and during disease. Inflammation is an energetically expensive process that requires adaptive changes in cellular metabolism and, in turn, metabolic intermediates can also have immunomodulatory actions. Such "immunometabolic" changes in peripheral immune cells have been implicated in contributing to disease pathology in obesity and diabetes. This review will discuss the evidence for a role of immunometabolic changes in glial cells in the systemic regulation of energy and glucose homeostasis, and how this changes in the context of obesity and diabetes.
Collapse
Affiliation(s)
- Josephine L Robb
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Nicole A Morrissey
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Paul G Weightman Potter
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Hannah E Smithers
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Craig Beall
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Kate L J Ellacott
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
9
|
Toll-Like Receptor 2 Attenuates Traumatic Brain Injury-Induced Neural Stem Cell Proliferation in Dentate Gyrus of Rats. Neural Plast 2020; 2020:9814978. [PMID: 32879625 PMCID: PMC7448220 DOI: 10.1155/2020/9814978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/10/2020] [Accepted: 07/24/2020] [Indexed: 01/06/2023] Open
Abstract
It was not clear how and whether neural stem cells (NSCs) responded to toll-like receptor 2 (TLR2) in the inflammatory environment after traumatic brain injury (TBI). The current study investigated the correlation of TLR2 and NSC proliferation in the dentate gyrus (DG) using the TBI model of rats. Immunofluorescence (IF) was used to observe the expression of BrdU, nestin, and TLR2 in the DG in morphology. Proliferating cells in the DG were labelled by thymidine analog 5-bromo-2-deoxyuridine (BrdU). Three-labelled BrdU, nestin, and DAPI was used for the identification of newly generated NSCs. Western blotting and real-time polymerase chain reaction (PCR) were used to observe the expression of TLR2 from the level of protein and mRNA. We observed that BrdU+/nestin+/DAPI+ cells accounted for 84.30% ± 6.54% among BrdU+ cells; BrdU+ and nestin+ cells in the DG were also TLR2+ cells. BrdU+ cells and the expression of TLR2 (both protein and mRNA levels) both elevated immediately at 6 hours (h), 24 h, 3 days (d), and 7 d posttrauma and peaked in 3 d. Results indicated that TLR2 was expressed on proliferating cells in the DG (NSCs possibly) and there was a potential correlation between increased TLR2 and proliferated NSCs after TBI. Taken together, these findings suggested that TLR2 was involved in endogenous neurogenesis in the DG after TBI.
Collapse
|
10
|
Sahab Negah S, Oliazadeh P, Jahanbazi Jahan-Abad A, Eshaghabadi A, Samini F, Ghasemi S, Asghari A, Gorji A. Transplantation of human meningioma stem cells loaded on a self-assembling peptide nanoscaffold containing IKVAV improves traumatic brain injury in rats. Acta Biomater 2019; 92:132-144. [PMID: 31075516 DOI: 10.1016/j.actbio.2019.05.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/02/2019] [Accepted: 05/06/2019] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) can result in permanent brain function impairment due to the poor regenerative ability of neural tissue. Tissue engineering has appeared as a promising approach to promote nerve regeneration and to ameliorate brain damage. The present study was designed to investigate the effect of transplantation of the human meningioma stem-like cells (hMgSCs) seeded in a promising three-dimensional scaffold (RADA4GGSIKVAV; R-GSIK) on the functional recovery of the brain and neuroinflammatory responses following TBI in rats. After induction of TBI, hMgSCs seeded in R-GSIK was transplanted within the injury site and its effect was compared to several control groups. Application of hMgSCs with R-GSIK improved functional recovery after TBI. A significant higher number of hMgSCs was observed in the brain when transplanted with R-GSIK scaffold compared to the control groups. Application of hMgSCs seeded in R-GSIK significantly decreased the lesion volume, reactive gliosis, and apoptosis at the injury site. Furthermore, treatment with hMgSCs seeded in R-GSIK significantly inhibited the expression of Toll-like receptor 4 and its downstream signaling molecules, including interleukin-1β and tumor necrosis factor. These data revealed the potential for hMgSCs seeded in R-GSIK to improve the functional recovery of the brain after TBI; possibly via amelioration of inflammatory responses. STATEMENT OF SIGNIFICANCE: Tissue engineered scaffolds that mimic the natural extracellular matrix of the brain may modulate stem cell fate and contribute to tissue repair following traumatic brain injury (TBI). Among several scaffolds, self-assembly peptide nanofiber scaffolds markedly promotes cellular behaviors, including cell survival and differentiation. We developed a novel three-dimensional scaffold (RADA16GGSIKVAV; R-GSIK). Transplantation of the human meningioma stem-like cells seeded in R-GSIK in an animal model of TBI significantly improved functional recovery of the brain, possibly via enhancement of stem cell survival as well as reduction of the lesion volume, inflammatory process, and reactive gliosis at the injury site. R-GSIK is a suitable microenvironment for human stem cells and could be a potential biomaterial for the reconstruction of the injured brain after TBI.
Collapse
|
11
|
Zheng D, Zang Y, Xu H, Wang Y, Cao X, Wang T, Pan M, Shi J, Li X. MicroRNA-214 promotes the calcification of human aortic valve interstitial cells through the acceleration of inflammatory reactions with activated MyD88/NF-κB signaling. Clin Res Cardiol 2018; 108:691-702. [PMID: 30519780 DOI: 10.1007/s00392-018-1398-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022]
Abstract
Calcific aortic valve disease (CAVD) is a complex active process involving in endothelial injury, lipid infiltration, chronic inflammation, matrix remodeling, cell differentiation, progressive bone formation, and new angiogenesis. The excess inflammatory responses induced by aortic valve interstitial cells (AVICs) are one of the common pathogeneses of this disease. Although many microRNAs (miRs) have been identified to play crucial roles in the calcification process of the aortic valve, numerous miRs are still waiting to be explored. In this study, we explored the functional role of miR-214 in the inflammatory reaction and calcification of human AVICs and its underlying molecular mechanism. Alizarin red staining was used to determine the number of calcified nodules. The protein levels of ICAM-1, IL-6, IL-8, and MCP-1 detected by enzyme-linked immunosorbent assay (ELISA) were used to assess the inflammatory reaction of AVICs; expression levels of RUNX2, Msx2, and BMP2 were used to evaluate AVICs osteoblast differentiation. Results showed that the expression levels of TLR4, MyD88, NF-κB, and miR-214 were up-regulated in the blood and aortic valve tissue samples of patients with CAVD when compared with normal individuals. Knockdown of miR-214 in AVICs inhibited the secretion of IL-6, IL-8, ICAM-1, and MCP-1, while this effect was repressed when lipopolysaccharide (LPS) was added to AVICs. LPS also enhanced the effects of miR-214 in promoting the secretion of pro-inflammatory factors. Besides, up-regulation of miR-214 promoted the protein expression of MyD88 and NF-κB but had no influence on TLR4, and miR-214 could directly combine with MyD88 protein. Up-regulation of MyD88 facilitated the secretion of pro-inflammatory factors and increased calcified nodules number and accelerated the expression of RUNX2, Msx2, and BMP2. Moreover, promotion of the expressions of pro-inflammatory factors and "osteoblast-like" cell markers induced by miR-214 overexpression was abolished when MyD88 was down-regulated in AVICs. In conclusion, this study revealed that miR-214 promoted calcification by facilitating inflammatory reaction through MyD88/NF-κB signaling pathway in AVICs.
Collapse
Affiliation(s)
- Dongdong Zheng
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20, Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China
| | - Yue Zang
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20, Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China
| | - Haixia Xu
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20, Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China
| | - Yan Wang
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20, Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China
| | - Xiang Cao
- Department of Cardio-Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Teng Wang
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20, Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China
| | - Min Pan
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20, Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China
| | - Jiahai Shi
- Department of Cardio-Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xiaofei Li
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20, Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
12
|
Shi H, Hua X, Kong D, Stein D, Hua F. Role of Toll-like receptor mediated signaling in traumatic brain injury. Neuropharmacology 2018; 145:259-267. [PMID: 30075158 DOI: 10.1016/j.neuropharm.2018.07.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 07/04/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
The mechanisms underlying secondary brain damage following traumatic brain injury (TBI) remain unclear. A great many studies have demonstrated that inflammatory cascades contribute to brain damage through the activation of immune/inflammatory responses, including the increased release of cytokines and chemokines, and the recruitment of leukocytes. The cells and tissues damaged by primary mechanical injury release a number of endogenous factors acting as damage-associated molecular patterns (DAMPs), which initiate and perpetuate noninfectious inflammatory responses through transduction signaling pathways. Toll-like receptors (TLRs) are a transmembrane receptor family that can recognize the specific DAMPs released from damaged cells and recruit a set of adaptors leading to the activation of downstream kinases and nuclear factors which regulate the expression of inflammatory genes. The activation of inflammatory responses mediated by TLR signaling is closely associated with brain tissue damage and neurological dysfunction following TBI. TLRs and their downstream protein kinases may be potential targets for the treatment of TBI. Modulation of TLR-mediated signaling may attenuate brain damage and improve TBI outcome. In this review, we briefly discuss the role of TLR-mediated signaling in TBI and the new treatments targeting TLR signaling. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
Affiliation(s)
- Hongjuan Shi
- Department of Neurology, The Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Xiaodong Hua
- Augusta University/University of Georgia Medical Partnership, Athens, GA, 30606, USA; Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Delian Kong
- Department of Neurology, The Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Donald Stein
- Brain Research Laboratory, Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, 30032, USA
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Key Laboratory of Anesthesiology of Jiangsu Province, Xuzhou, 221002, China.
| |
Collapse
|
13
|
Wang Z, Zhang YH, Guo C, Gao HL, Zhong ML, Huang TT, Liu NN, Guo RF, Lan T, Zhang W, Wang ZY, Zhao P. Tetrathiomolybdate Treatment Leads to the Suppression of Inflammatory Responses through the TRAF6/NFκB Pathway in LPS-Stimulated BV-2 Microglia. Front Aging Neurosci 2018. [PMID: 29535623 PMCID: PMC5835334 DOI: 10.3389/fnagi.2018.00009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although the positive relationship between copper and Alzheimer's disease (AD) was reported by a lot of epidemiological data, the mechanism is not completely known. Copper is a redox metal and serves as a mediator of inflammation. Because the homeostasis of copper is altered in Aβ precursor protein (APP) and presenilin 1 (PS1) transgenic (Tg) mice, the using of copper chelators is a potential therapeutic strategy for AD. Here we report that a copper chelator, tetrathiomolybdate (TM), is a potential therapeutic drug of AD. We investigated whether TM treatment led to a decrease of pro-inflammatory cytokines in vivo and in vitro, and found that TM treatment reduced the expression of iNOS and TNF-α in APP/PS1 Tg mice through up-regulating superoxide dismutase 1 (SOD1) activity. In vitro, once stimulated, microglia secretes a variety of proinflammatory cytokines, so we utilized LPS-stimulated BV-2 cells as the inflammatory cell model to detect the anti-inflammatory effects of TM. Our results indicated that TM-pretreatment suppressed the ubiquitination of TRAF6 and the activation of NFκB without affecting the expression of TLR4 and Myd88 in vitro. By detecting the activity of SOD1 and the production of reactive oxygen species (ROS), we found that the anti-inflammatory effects of TM could be attributed to its ability to reduce the amount of intracellular bioavailable copper, and the production of ROS which is an activator of the TRAF6 auto-ubiquitination. Hence, our results revealed that TM-treatment could reduce the production of inflammatory cytokines by the suppression of ROS/TRAF6/AKT/NFκB signaling pathway.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Ya-Hong Zhang
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Chuang Guo
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Hui-Ling Gao
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Man-Li Zhong
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Ting-Ting Huang
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Na-Na Liu
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Rui-Fang Guo
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Tian Lan
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Wei Zhang
- Department of Hepatobiliary Surgery, General Hospital of Shenyang Military Area Command, Shenyang, China
| | - Zhan-You Wang
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Pu Zhao
- Department of Neurobiology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
14
|
Je EM, Kim SS, Yoo NJ, Lee SH. Mutational and Expressional Analyses of MYD88 Gene in Common Solid Cancers. TUMORI JOURNAL 2018; 98:663-9. [DOI: 10.1177/030089161209800518] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aims and background Myeloid differentiation primary response gene 88 (MYD88) is a protein involved in hematopoietic differentiation and innate immunity. Recent studies revealed MYD88 mutation in hematological malignancies and MYD88 overexpression in some solid cancers. The aim of this study was to see whether alterations of MYD88 protein expression and somatic mutation of MYD88 gene are features of common solid cancers. Methods We analyzed MYD88 mutation in 45 gastric, 45 colorectal, 45 breast, 45 hepatocellular, 45 prostate and 45 lung carcinomas by single-strand conformation polymorphism (SSCP). We also analyzed MYD88 protein expression in 60 gastric, 60 coloretal and 107 prostate carcinomas by immunohistochemistry. Results In the immunohistochemistry results, MYD88 protein was highly expressed in gastric (75%), colorectal (80%) and prostate (83%) cancers. However, MYD88 expression was significantly different among normal tissues (gastric: 58%, colon: 100%, prostate: 86%). MYD88 expression was significantly increased in gastric cancer cells compared with normal cells, whereas it was decreased in colorectal cancer cells compared with normal cells. There were no somatic mutations of the MYD88 gene in gastric, colorectal, breast, hepatocellular, prostate and lung carcinomas. Conclusions Our data indicate that MYD88 overexpression might be a feature of many solid cancers, but MYD88 expression in normal cells differs depending on the organs. The data suggest that a gain of MYD88 expression in gastric cancers might play a role in cancer pathogenesis by activating oncogenic functions of MYD88.
Collapse
Affiliation(s)
- Eun Mi Je
- Departments of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung Soo Kim
- Departments of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Nam Jin Yoo
- Departments of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sug Hyung Lee
- Departments of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
15
|
Dietary teasaponin ameliorates alteration of gut microbiota and cognitive decline in diet-induced obese mice. Sci Rep 2017; 7:12203. [PMID: 28939875 PMCID: PMC5610180 DOI: 10.1038/s41598-017-12156-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/30/2017] [Indexed: 12/14/2022] Open
Abstract
A high-fat (HF) diet alters gut microbiota and promotes obesity related inflammation and cognitive impairment. Teasaponin is the major active component of tea, and has been associated with anti-inflammatory effects and improved microbiota composition. However, the potential protective effects of teasaponin, against HF diet-induced obesity and its associated alteration of gut microbiota, inflammation and cognitive decline have not been studied. In this study, obesity was induced in C57BL/6 J male mice by feeding a HF diet for 8 weeks, followed by treatment with oral teasaponin (0.5%) mixed in HF diet for a further 6 weeks. Teasaponin treatment prevented the HF diet-induced recognition memory impairment and improved neuroinflammation, gliosis and brain-derived neurotrophic factor (BDNF) deficits in the hippocampus. Furthermore, teasaponin attenuated the HF diet-induced endotoxemia, pro-inflammatory macrophage accumulation in the colon and gut microbiota alterations. Teasaponin also improved glucose tolerance and reduced body weight gain in HF diet-induced obese mice. The behavioral and neurochemical improvements suggest that teasaponin could limit unfavorable gut microbiota alterations and cognitive decline in HF diet-induced obesity.
Collapse
|
16
|
Xie F, Min S, Chen J, Yang J, Wang X. Ulinastatin inhibited sepsis-induced spinal inflammation to alleviate peripheral neuromuscular dysfunction in an experimental rat model of neuromyopathy. J Neurochem 2017; 143:225-235. [PMID: 28796387 DOI: 10.1111/jnc.14145] [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: 06/09/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/30/2022]
Abstract
Sepsis initiates a neuroinflammatory cascade that contributes to spinal cord inflammation and behavioral impairment, and Toll-like receptor 4 (TLR4) is an important mediator of this cascade. In this study, we tested the hypothesis that ulinastatin (ULI) inhibits sepsis-induced spinal inflammation to alleviate peripheral neuromuscular dysfunction through the TLR4/myeloid differentiation factor 88 (MyD88)/NF-κB signaling pathway. Muscular function, spinal cord water content, and cytokine levels of spinal cord were tested in TLR4-inhibited rats subjected to cecal ligation and puncture (CLP). The normal rats were intrathecally injected with different concentrations of ULI or normal saline 60 min before CLP. At 24 h after CLP, the activation of microglia/macrophage was detected by immunofluorescence staining; and the cytokines were assayed by ELISA. The protein expression level of the TLR4 and its downstream effectors (MyD88 and NF-κB), the neuregulin-1, and the γ- and α7-nicotinic acetylcholine receptor was measured using western blotting. The protein expression of TLR4 in the spinal cord reached a maximum at 24 h post-CLP. Compared to the sham rats, the TLR4-inhibited rats showed attenuated functional impairment and cytokine release. ULI (5000 U/kg ) treatment pre-CLP significantly reduced the number of TLR4-positive microglia/macrophages as well as inflammatory mediator release in septic rats. Furthermore, the levels of TLR4, MyD88, and NF-κB and the expression level of γ-/α7-nicotinic acetylcholine receptors also decreased after ULI treatment. ULI administration may improve patient outcome by reducing the spinal inflammation through a mechanism involving the TLR4/MyD88/NF-κB signaling in sepsis.
Collapse
Affiliation(s)
- Fei Xie
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Su Min
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingyuan Chen
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Yang
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Wang
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Electroacupuncture Improved Hippocampal Neurogenesis following Traumatic Brain Injury in Mice through Inhibition of TLR4 Signaling Pathway. Stem Cells Int 2017; 2017:5841814. [PMID: 28848607 PMCID: PMC5564094 DOI: 10.1155/2017/5841814] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/02/2017] [Accepted: 07/09/2017] [Indexed: 11/18/2022] Open
Abstract
The protective role of electroacupuncture (EA) treatment in diverse neurological diseases such as ischemic stroke is well acknowledged. However, whether and how EA act on hippocampal neurogenesis following traumatic brain injury (TBI) remains poorly understood. This study aims to investigate the effect of EA on hippocampal neurogenesis and neurological functions, as well as its underlying association with toll-like receptor 4 (TLR4) signaling in TBI mice. BrdU/NeuN immunofluorescence was performed to label newborn neurons in the hippocampus after EA treatment. Water maze test and neurological severity score were used to evaluate neurological function posttrauma. The hippocampal level of TLR4 and downstream molecules and inflammatory cytokines were, respectively, detected by Western blot and enzyme-linked immunosorbent assay. EA enhanced hippocampal neurogenesis and inhibited TLR4 expression at 21, 28, and 35 days after TBI, but the beneficial effects of EA on posttraumatic neurogenesis and neurological functions were attenuated by lipopolysaccharide-induced TLR4 activation. In addition, EA exerted an inhibitory effect on both TLR4/Myd88/NF-κB and TLR4/TRIF/NF-κB pathways, as well as the inflammatory cytokine expression in the hippocampus following TBI. In conclusion, EA promoted hippocampal neurogenesis and neurological recovery through inhibition of TLR4 signaling pathway posttrauma, which may be a potential approach to improve the outcome of TBI.
Collapse
|
18
|
Protective Functions of PJ34, a Poly(ADP-ribose) Polymerase Inhibitor, Are Related to Down-Regulation of Calpain and Nuclear Factor-κB in a Mouse Model of Traumatic Brain Injury. World Neurosurg 2017. [PMID: 28642177 DOI: 10.1016/j.wneu.2017.06.076] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Poly(ADP-ribose) polymerase (PARP), calpain, and nuclear factor-κB (NF-κB) are reported to participate in inflammatory reactions in pathologic conditions and are involved in traumatic brain injury. The objective of this study was to investigate whether PARP participates in inflammation related to calpain and NF-κB in a mouse model of controlled cortical impact (CCI). METHODS PJ34 (10 mg/kg), a selective PARP inhibitor, was administered intraperitoneally 5 minutes and 8 hours after experimental CCI. We then performed a histopathologic analysis, and we measured calpain activity and protein levels in all animals. The cytosolic, mitochondria, and nuclear fractions were prepared and used to determine the levels of PARP, calpastatin, NF-κB p65, inhibitory-κB-α, tumor necrosis factor-α, interleukin-1β, intracellular adhesion molecule-1, inducible nitric oxide synthase, and cyclooxygenase-2. We then measured blood-brain barrier disruption using electron microscopy at 6 and 24 hours after CCI. RESULTS Treatment with PJ34 markedly reduced the extent of both cerebral contusion and edema, improved neurologic scores, and attenuated blood-brain barrier damage resulting from CCI. Our data showed that the cytosolic and nuclear fractions of calpain and NF-κB were up-regulated in the injured cortex and that these changes were reversed by PJ34. Moreover, PJ34 significantly enhanced the calpastatin and inhibitory-κB levels and decreased the levels of inflammatory mediators. CONCLUSIONS PARP inhibition by PJ34 suppresses the overactivation of calpain and the production of inflammatory factors that are caused by NF-κB activation and attenuates neuronal cell death in a mouse model of CCI.
Collapse
|
19
|
Mangiferin attenuates blast-induced traumatic brain injury via inhibiting NLRP3 inflammasome. Chem Biol Interact 2017; 271:15-23. [DOI: 10.1016/j.cbi.2017.04.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023]
|
20
|
Laskowitz DT, Wang H, Chen T, Lubkin DT, Cantillana V, Tu TM, Kernagis D, Zhou G, Macy G, Kolls BJ, Dawson HN. Neuroprotective pentapeptide CN-105 is associated with reduced sterile inflammation and improved functional outcomes in a traumatic brain injury murine model. Sci Rep 2017; 7:46461. [PMID: 28429734 PMCID: PMC5399447 DOI: 10.1038/srep46461] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 03/16/2017] [Indexed: 12/31/2022] Open
Abstract
At present, there are no proven pharmacological treatments demonstrated to improve long term functional outcomes following traumatic brain injury(TBI). In the setting of non-penetrating TBI, sterile brain inflammatory responses are associated with the development of cerebral edema, intracranial hypertension, and secondary neuronal injury. There is increasing evidence that endogenous apolipoprotein E(apoE) modifies the neuroinflammatory response through its role in downregulating glial activation, however, the intact apoE holoprotein does not cross the blood-brain barrier due to its size. To address this limitation, we developed a small 5 amino acid apoE mimetic peptide(CN-105) that mimics the polar face of the apoE helical domain involved in receptor interactions. The goal of this study was to investigate the therapeutic potential of CN-105 in a murine model of closed head injury. Treatment with CN-105 was associated with a durable improvement in functional outcomes as assessed by Rotarod and Morris Water Maze and a reduction in positive Fluoro-Jade B stained injured neurons and microglial activation. Administration of CN-105 was also associated with reduction in mRNA expression of a subset of inflammatory and immune-related genes.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA.,Aegis-CN LLC., Durham, NC, USA
| | - Haichen Wang
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tony Chen
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - David T Lubkin
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Viviana Cantillana
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tian Ming Tu
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
| | - Dawn Kernagis
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Guanen Zhou
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gary Macy
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bradley J Kolls
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hana N Dawson
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
21
|
Feng Y, Gao J, Cui Y, Li M, Li R, Cui C, Cui J. Neuroprotective Effects of Resatorvid Against Traumatic Brain Injury in Rat: Involvement of Neuronal Autophagy and TLR4 Signaling Pathway. Cell Mol Neurobiol 2017; 37:155-168. [PMID: 26961544 DOI: 10.1007/s10571-016-0356-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 02/24/2016] [Indexed: 12/23/2022]
Abstract
Accumulating evidence indicates that autophagy and inflammatory responses contributes to secondary brain injury after traumatic brain injury (TBI), and toll-like receptor 4 (TLR4) is considered to involvement of this cascade and plays an important role. The present study was designed to determine the hypothesis that administration of resatorvid (TAK-242), a TLR4 antagonist, might provide a neuroprotective effect by inhibit TLR4-mediated pathway in a TBI rat model. Rat subjected to controlled cortical impact injury were injected with TAK-242 (0.5 mg/kg, i.v. injected) 10 min prior to injury. The results demonstrated that TAK-242 treatment significantly attenuated TBI-induced neurons loss, brain edema, and neurobehavioral impairment in rats. Immunoblotting analysis showed that TAK-242 treatment reduced TBI-induced TLR4, Beclin 1, and LC3-II levels, and maintained p62 levels at 24 h. Double immunolabeling demonstrated that LC3 dots co-localized with the hippocampus pyramidal neurons, and TLR4 was localized with the hippocampus neurons and astrocytes. In addition, the expression of TLR4 downstream signaling molecules, including MyD88, TRIF, NF-κB, TNF-α, and IL-1β, was significantly downregulated in hippocampus tissue by Western blot analysis. In conclusion, our findings indicate that pre-injury treatment with TAK-242 could inhibit neuronal autophagy and neuroinflammation responses in the hippocampus in a rat model of TBI. The neuroprotective effects of TAK-242 may be related to modulation of the TLR4-MyD88/TRIF-NF-κB signaling pathway. Furthermore, the study also suggests that TAK-242, an attractive potential drug, may be a promising drug candidate for TBI.
Collapse
Affiliation(s)
- Yan Feng
- Department of Surgery, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, People's Republic of China
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Junling Gao
- School of Basic Medical Science, North China University of Science and Technology, Tangshan, People's Republic of China
| | - Ying Cui
- Department of Neurosurgery, Tangshan Workers' Hospital, Tangshan, 063000, Hebei, People's Republic of China
| | - Minghang Li
- School of Basic Medical Science, North China University of Science and Technology, Tangshan, People's Republic of China
| | - Ran Li
- School of Basic Medical Science, North China University of Science and Technology, Tangshan, People's Republic of China
| | - Changmeng Cui
- Department of Surgery, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, People's Republic of China
| | - Jianzhong Cui
- Department of Surgery, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050017, People's Republic of China.
- Department of Neurosurgery, Tangshan Workers' Hospital, Tangshan, 063000, Hebei, People's Republic of China.
| |
Collapse
|
22
|
Lv Y, Qian Y, Ou-Yang A, Fu L. Hydroxysafflor Yellow A Attenuates Neuron Damage by Suppressing the Lipopolysaccharide-Induced TLR4 Pathway in Activated Microglial Cells. Cell Mol Neurobiol 2016; 36:1241-1256. [PMID: 26754542 DOI: 10.1007/s10571-015-0322-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/22/2015] [Indexed: 10/22/2022]
Abstract
Microglia activation initiates a neurological deficit cascade that contributes to substantial neuronal damage and impairment following ischemia stroke. Toll-like receptor 4 (TLR4) has been demonstrated to play a critical role in this cascade. In the current study, we tested the hypothesis that hydroxysafflor yellow A (HSYA), an active ingredient extracted from Flos Carthami tinctorii, alleviated inflammatory damage, and mediated neurotrophic effects in neurons by inducing the TLR4 pathway in microglia. A non-contact Transwell co-culture system comprised microglia and neurons was treated with HSYA followed by a 1 mg/mL lipopolysaccharide (LPS) stimulation. The microglia were activated prior to neuronal apoptosis, which were induced by increasing TLR4 expression in the activated microglia. However, HSYA suppressed TLR4 expression in the activated microglia, resulting in less neuronal damage at the early stage of LPS stimulation. Western blot analysis and immunofluorescence indicated that dose-dependently HSYA down-regulated TLR4-induced downstream effectors myeloid differentiation factor 88 (MyD88), nuclear factor kappa b (NF-κB), and the mitogen-activated protein kinases (MAPK)-regulated proteins c-Jun NH2-terminal protein kinase (JNK), protein kinase (ERK) 1/2 (ERK1/2), p38 MAPK (p38), as well as the LPS-induced inflammatory cytokine release. However, HSYA up-regulated brain-derived neurotrophic factor (BDNF) expression. Our data suggest that HSYA could exert neurotrophic and anti-inflammatory functions in response to LPS stimulation by inhibiting TLR4 pathway-mediated signaling.
Collapse
Affiliation(s)
- Yanni Lv
- Pharmacy Department, The First Affiliated Hospital of Nanchang University, Yongwai Street 17, Nanchang, 330006, China.
| | - Yisong Qian
- Institute of Translational Medicine, Nanchang University, Nanchang, 330001, China
| | - Aijun Ou-Yang
- Pharmacy Department, The First Affiliated Hospital of Nanchang University, Yongwai Street 17, Nanchang, 330006, China
| | - Longsheng Fu
- Pharmacy Department, The First Affiliated Hospital of Nanchang University, Yongwai Street 17, Nanchang, 330006, China
| |
Collapse
|
23
|
Altered gene expression and possible immunodeficiency in cases of sudden infant death syndrome. Pediatr Res 2016; 80:77-84. [PMID: 26959483 DOI: 10.1038/pr.2016.45] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/23/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND A large number of studies have tried to uncover a genetic predisposition for sudden infant death syndrome (SIDS), but there is still uncertainty concerning the pathogenesis of these deaths. The purpose of this study was to investigate mRNA gene expression in SIDS cases and controls, in order to uncover genes that are differentially expressed in the two groups. METHODS Tissue from brain, heart, and liver from 15 SIDS cases and 15 controls were included in the study, and mRNA expression was determined using the Illumina whole genome gene expression DASL HT assay. RESULTS Seventeen genes showed significantly altered expression compared to controls, after correction for multiple testing. Three genes involved in the immune system were of particular interest, including the downregulation of MyD88 in tissue from SIDS brains, as well as the downregulation of the genes encoding CCL3 and UNC13 in the liver. CONCLUSION These findings indicate that there is an altered expression of genes involved in the inflammatory process in a proportion of SIDS cases, which further strengthen the hypothesis that impaired immune response play a role in this syndrome.
Collapse
|
24
|
Inhibition of myeloid differentiation factor 88(MyD88) by ST2825 provides neuroprotection after experimental traumatic brain injury in mice. Brain Res 2016; 1643:130-9. [PMID: 27155455 DOI: 10.1016/j.brainres.2016.05.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 04/11/2016] [Accepted: 05/03/2016] [Indexed: 11/24/2022]
Abstract
Myeloid differentiation factor 88(MyD88) is an endogenous adaptor protein that plays an important role in coordinating intracellular inflammatory responses induced by agonists of the Toll-like receptor and interleukin-1 receptor families. MyD88 has been reported to be essential for neuronal death in animal models and may represent a therapeutic target for pharmacologic inhibition following traumatic brain injury (TBI). The purpose of the current study was to investigate the neuroprotective effect of MyD88 specific inhibitor ST2825 in an experimental mouse model of TBI. Intracerebroventricular (ICV) injection of high concentration (20μg/μL) ST2825 (15min post TBI) attenuated the development of TBI in mice, markedly improved neurological function and reduced brain edema. Decreased neural apoptosis and increased neuronal survival were also observed. Biochemically, the high concentration of ST2825 significantly reduced the levels of MyD88, further decreased TAK1, p-TAK1, nuclear p65 and increased IκB-α. Additionally, ST2825 significantly reduced the levels of Iba-1 and inflammatory factors TNF-α and IL-1β. These data provide an experimental rationale for evaluation of MyD88 as a drug target and highlight the potential therapeutic implications of ST2825 in TBI.
Collapse
|
25
|
Tao X, Chen X, Mao X, Hou Z, Hao S, Tian R, Zhu Z, Sun M, Liu B. Protective effects of PARP inhibitor, PJ34, is related to down-regulation of calpain and NF-κB in a mouse model of TBI. Brain Inj 2016:1-11. [PMID: 27119554 DOI: 10.3109/02699052.2016.1160151] [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] [Indexed: 11/13/2022]
Abstract
BACKGROUND Poly(ADP-ribose) polymerase (PARP), calpain and nuclear factor-κB (NF-κB) are reported to participate in inflammatory reactions in pathological conditions and are involved in traumatic brain injury. The objective of this study was to investigate whether PARP participated in inflammation related to calpain and NF-κB in a mouse model of controlled cortical impact (CCI). MATERIALS AND METHODS PJ34 (10 mg kg-1), a selective PARP inhibitor, was administered intraperitoneally 5 minutes and 8 hours after experimental CCI. A neurobehavioural evaluation and a histopathological analysis were then performed and the contusion volume, calpain activity and protein levels were measured in all animals. RESULTS Treatment with PJ34 markedly reduced neurological deficits, decreased contusion volume and attenuated necrotic and apoptotic neuronal cell death 24 hours after CCI. The data showed that the cytosolic and nuclear fractions of calpain and NF-κB were up-regulated in the injured cortex and that these changes were reversed by PJ34. Moreover, PJ34 significantly enhanced the calpastatin and IκB levels and decreased the levels of inflammatory mediators. CONCLUSIONS PARP inhibition by PJ34 suppresses the over-activation of calpain and the production of inflammatory factors that are caused by NF-κB activation and it improves neurological functioning, decreases the contusion volume and attenuates neuronal cell death in a mouse model of CCI.
Collapse
Affiliation(s)
- Xiaogang Tao
- a Department of Neurosurgery, Beijing Tiantan Hospital , Capital Medical University , Beijing , PR China
| | - Xuetao Chen
- a Department of Neurosurgery, Beijing Tiantan Hospital , Capital Medical University , Beijing , PR China
| | - Xiang Mao
- a Department of Neurosurgery, Beijing Tiantan Hospital , Capital Medical University , Beijing , PR China
| | - Zonggang Hou
- a Department of Neurosurgery, Beijing Tiantan Hospital , Capital Medical University , Beijing , PR China
| | - Shuyu Hao
- a Department of Neurosurgery, Beijing Tiantan Hospital , Capital Medical University , Beijing , PR China
| | - Runfa Tian
- a Department of Neurosurgery, Beijing Tiantan Hospital , Capital Medical University , Beijing , PR China
| | - Zhendan Zhu
- b Department of Neurotrauma, General Hospital of the Armed Police Force , Beijing , PR China
| | - Ming Sun
- c Department of Neuropharmacology
| | - Baiyun Liu
- a Department of Neurosurgery, Beijing Tiantan Hospital , Capital Medical University , Beijing , PR China
- b Department of Neurotrauma, General Hospital of the Armed Police Force , Beijing , PR China
- d Department of Neurotrauma , Beijing Neurosurgical Institute, Capital Medical University , Beijing , PR China
- e Nerve Injury and Repair Center of Beijing Institute for Brain Disorders , Beijing , PR China
- f China National Clinical Research Center for Neurological Diseases , Beijing , PR China
- g Beijing Key Laboratory of Central Nervous System Injury , Beijing , PR China
| |
Collapse
|
26
|
Feng Y, Cui Y, Gao JL, Li MH, Li R, Jiang XH, Tian YX, Wang KJ, Cui CM, Cui JZ. Resveratrol attenuates neuronal autophagy and inflammatory injury by inhibiting the TLR4/NF-κB signaling pathway in experimental traumatic brain injury. Int J Mol Med 2016; 37:921-30. [PMID: 26936125 PMCID: PMC4790669 DOI: 10.3892/ijmm.2016.2495] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 02/08/2016] [Indexed: 12/18/2022] Open
Abstract
Previous research has demonstrated that traumatic brain injury (TBI) activates autophagy and a neuroinflammatory cascade that contributes to substantial neuronal damage and behavioral impairment, and Toll-like receptor 4 (TLR4) is an important mediator of this cascade. In the present study, we investigated the hypothesis that resveratrol (RV), a natural polyphenolic compound with potent multifaceted properties, alleviates brain damage mediated by TLR4 following TBI. Adult male Sprague Dawley rats, subjected to controlled cortical impact (CCI) injury, were intraperitoneally injected with RV (100 mg/kg, daily for 3 days) after the onset of TBI. The results demonstrated that RV significantly reduced brain edema, motor deficit, neuronal loss and improved spatial cognitive function. Double immunolabeling demonstrated that RV decreased microtubule-associated protein 1 light chain 3 (LC3), TLR4‑positive cells co-labeled with the hippocampal neurons, and RV also significantly reduced the number of TLR4‑positive neuron‑specific nuclear protein (NeuN) cells following TBI. Western blot analysis revealed that RV significantly reduced the protein expression of the autophagy marker proteins, LC3II and Beclin1, in the hippocampus compared with that in the TBI group. Furthermore, the levels of TLR4 and its known downstream signaling molecules, nuclear factor-κB (NF-κB), and the inflammatory cytokines, interleukin (IL)-1β and tumor necrosis factor (TNF)-α were also decreased after RV treatment. Our results suggest that RV reduces neuronal autophagy and inflammatory reactions in a rat model of TBI. Thus, we suggest that the neuroprotective effect of RV is associated with the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yan Feng
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Ying Cui
- Department of Neurosurgery,Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Jun-Ling Gao
- School of Basic Medical Science, Hebei United University, Tangshan, Hebei 063000, P.R. China
| | - Ming-Hang Li
- School of Basic Medical Science, Hebei United University, Tangshan, Hebei 063000, P.R. China
| | - Ran Li
- School of Basic Medical Science, Hebei United University, Tangshan, Hebei 063000, P.R. China
| | - Xiao-Hua Jiang
- School of Basic Medical Science, Hebei United University, Tangshan, Hebei 063000, P.R. China
| | - Yan-Xia Tian
- School of Basic Medical Science, Hebei United University, Tangshan, Hebei 063000, P.R. China
| | - Kai-Jie Wang
- Department of Neurosurgery,Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| | - Chang-Meng Cui
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Jian-Zhong Cui
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
27
|
Geng F, Ma Y, Xing T, Zhuang X, Zhu J, Yao L. Effects of Hyperbaric Oxygen Therapy on Inflammasome Signaling after Traumatic Brain Injury. Neuroimmunomodulation 2016; 23:122-9. [PMID: 27216735 DOI: 10.1159/000445689] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/18/2016] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Neuroinflammation plays an important role in secondary tissue damage after traumatic brain injury (TBI). Recently, the inflammasome-mediated inflammatory pathway has been observed in the inflammatory response of TBI. In this study, we investigated the influence of hyperbaric oxygen therapy (HBOT) on inflammasome activation after TBI. METHODS The experimental mice were randomly divided into 4 groups as follows: sham-operated normobaric air (21% O2 at one absolute atmosphere), HBOT only, TBI + normobaric air and TBI + HBOT. Following the evaluation of motor deficits and brain edema, the expression of inflammasome components and effectors was measured by qRT-PCR and Western blotting. Moreover, alterations in IL-1β, IL-18 and high-mobility group box 1 (HMGB1) were calculated by enzyme-linked immunosorbent assay at each time point after injury. RESULTS HBOT improved motor score and reduced brain edema. Furthermore, it suppressed protein expression of inflammasome components and reduced the levels of IL-1β and IL-18, accompanied by the reduction of HMGB1 in brain tissues and serum. CONCLUSION These results suggest that HBOT may alleviate the inflammatory response after TBI by inhibiting the activation of inflammasome signaling.
Collapse
Affiliation(s)
- Fengyang Geng
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, PR China
| | | | | | | | | | | |
Collapse
|
28
|
Yu Z, Li H, Yan HY, Yang YQ, Zhang DD, Huang LT, Xie GB, Liu M, Tohti M, Hang CH. Expression and Cell Distribution of SENP3 in Brain Tissue After Traumatic Brain Injury in Mice: A Pilot Study. Cell Mol Neurobiol 2015; 35:733-40. [PMID: 25772139 DOI: 10.1007/s10571-015-0169-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 02/26/2015] [Indexed: 11/25/2022]
Abstract
SUMO-specific proteases 3 (SENP3) is a member of the small ubiquitin-like modifier-specific protease family and deconjugates SUMO2/3 from protein substrates. To date, the expression and function of SENP3 in traumatic brain injury (TBI) are unclear. The present study examined dynamic changes in SENP3 expression in the cerebral cortex and in its cellular localization, using an acute TBI model in adult mice. SENP3 expression was examined at 3, 6, 12, 24 h, 3, and 5 days after TBI using Western Blot analysis and quantitative real-time PCR. Immunohistochemistry and immunofluorescence were examined to detect SENP3 localization. Western Blot indicated that SENP3 protein levels gradually increased from 3 h after TBI and peaked at 24 h. Quantitative real-time PCR demonstrated a gradual increase in SENP3 expression, which peaked 12 h after TBI and declined subsequently. Immunohistochemical staining demonstrated that SENP3-positive cells were observed in both the sham and 24 h post-TBI groups. However, robust expression of SENP3 was seldom observed in the sham group, while it was notably enhanced after TBI. Furthermore, immunofluorescence results revealed that the expression of SENP3 increased more significantly in neurons at day 1 after TBI compared with sham group and less significantly in astrocytes and microglia. Moreover, the SENP3-positive cells that were co-expressed with NeuN also expressed caspase-3, indicating a potential correlation between SENP3 and apoptosis after TBI. Collectively, our results showed obvious up-regulation of SENP3 expression in the brain after TBI, especially in the neurons. However, the full role of SENP3 and its therapeutic potential in TBI needs further investigation.
Collapse
Affiliation(s)
- Zhuang Yu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Amenta PS, Jallo JI, Tuma RF, Hooper DC, Elliott MB. Cannabinoid receptor type-2 stimulation, blockade, and deletion alter the vascular inflammatory responses to traumatic brain injury. J Neuroinflammation 2014; 11:191. [PMID: 25416141 PMCID: PMC4248435 DOI: 10.1186/s12974-014-0191-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/31/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Immunomodulatory therapies have been identified as interventions for secondary injury after traumatic brain injury (TBI). The cannabinoid receptor type-2 (CB2R) is proposed to play an important, endogenous role in regulating inflammation. The effects of CB2R stimulation, blockade, and deletion on the neurovascular inflammatory responses to TBI were assessed. METHODS Wild-type C57BL/6 or CB2R knockout mice were randomly assigned to controlled cortical impact (CCI) injury or to craniotomy control groups. The effects of treatment with synthetic, selective CB2R agonists (0-1966 and JWH-133), a selective CB2R antagonist, or vehicle solution administered to CCI groups were assessed at 1-day after injury. Changes in TNF-α, intracellular adhesion molecule (ICAM-1), inducible nitric oxide synthase (iNOS), macrophage/microglial ionized calcium-binding adaptor molecule, and blood-brain-barrier (BBB) permeability were assessed using ELISA, quantitative RT-PCR, immunohistochemistry, and fluorometric analysis of sodium fluorescein uptake. CB2R knockouts and wild-type mice with CCI injury were treated with a CB2R agonist or vehicle treatment. RESULTS TNF-α mRNA increased at 6 hours and 1 to 3 days after CCI; a CB2R antagonist and genetic knockout of the CB2R exacerbated TNF-α mRNA expression. Treatment with a CB2R agonist attenuated TNF-α protein levels indicating post-transcriptional mechanisms. Intracellular adhesion molecule (ICAM-1) mRNA was increased at 6 hours, and at 1 to 2 days after CCI, reduced in mice treated with a CB2R agonist, and increased in CB2R knockout mice with CCI. Sodium fluorescein uptake was increased in CB2R knockouts after CCI, with and without a CB2R agonist. iNOS mRNA expression peaked early (6 hours) and remained increased from 1 to 3 days after injury. Treatment with a CB2R agonist attenuated increases in iNOS mRNA expression, while genetic deletion of the CB2R resulted in substantial increases in iNOS expression. Double label immunohistochemistry confirmed that iNOS was expressed by macrophage/microglia in the injured cortex. CONCLUSION Findings demonstrate that the endogenous cannabinoid system and CB2R play an important role in regulating inflammation and neurovascular responses in the traumatically injured brain. CB2R stimulation with two agonists (0-1966 and JWH-133) dampened post-traumatic inflammation, while blockade or deletion of the CB2R worsened inflammation. Findings support previous evidence that modulating the CB2R alters infiltrating macrophages and activated resident microglia. Further investigation into the role of the CB2R on specific immune cell populations in the injured brain is warranted.
Collapse
Affiliation(s)
- Peter S Amenta
- Department of Neurological Surgery, Thomas Jefferson University Hospital, 1020 Locust Street, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Jack I Jallo
- Department of Neurological Surgery, Thomas Jefferson University Hospital, 1020 Locust Street, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Ronald F Tuma
- Department of Physiology, Temple University School of Medicine, 3500 N Broad St, Philadelphia, PA, 19140, USA.
| | - D Craig Hooper
- Department of Cancer Biology, Thomas Jefferson University Hospital, 1020 Locust Street, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Melanie B Elliott
- Department of Neurological Surgery, Thomas Jefferson University Hospital, 1020 Locust Street, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
30
|
Association between toll-like receptor 4 expression and neural stem cell proliferation in the hippocampus following traumatic brain injury in mice. Int J Mol Sci 2014; 15:12651-64. [PMID: 25036030 PMCID: PMC4139865 DOI: 10.3390/ijms150712651] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/03/2014] [Accepted: 07/04/2014] [Indexed: 12/20/2022] Open
Abstract
Whether or how neural stem cells (NSCs) respond to toll-like receptor 4 (TLR4) in an inflammatory environment caused by traumatic brain injury (TBI) has not been understood. In the present study, association between TLR4 expression and NSCs proliferation in the hippocampus was investigated in a mouse model of TBI using controlled cortical impact (CCI). Hippocampal proliferating cells were labeled with the thymidine analog 5-bromo-2-deoxyuridine (BrdU). In order to identify NSCs, the proliferating cells were further co-labeled with BrdU/sex determination region of Y chromosome related high mobility group box gene 2 (SOX2). Morphological observation on the expression of BrdU, SOX2, and TLR4 in the hippocampus was performed by inmmunofluorescence (IF). Relative quantification of TLR4 expression at the protein and mRNA level was performed using Western blotting and real-time polymerase chain reaction (PCR). It was observed that BrdU+/SOX2+ cells accounted for 95.80% ± 7.91% among BrdU+ cells; several BrdU+ cells and SOX2+ cells in the hippocampus were also TLR4-positive post injury, and that BrdU+ cell numbers, together with TLR4 expression at either protein or mRNA level, increased significantly in TBI mice over 1, 3, 7, 14, and 21 days survivals and changed in a similar temporal pattern with a peak at 3 day post-injury. These results indicate that hippocampal proliferating cells (suggestive of NSCs) expressed TLR4, and that there was a potential association between increased expression of TLR4 and the proliferation of NSCs post TBI. It is concluded that hippocampal TLR4 may play a potential role in endogenous neurogenesis after TBI.
Collapse
|
31
|
Zhu HT, Bian C, Yuan JC, Chu WH, Xiang X, Chen F, Wang CS, Feng H, Lin JK. Curcumin attenuates acute inflammatory injury by inhibiting the TLR4/MyD88/NF-κB signaling pathway in experimental traumatic brain injury. J Neuroinflammation 2014; 11:59. [PMID: 24669820 PMCID: PMC3986937 DOI: 10.1186/1742-2094-11-59] [Citation(s) in RCA: 262] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/17/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) initiates a neuroinflammatory cascade that contributes to substantial neuronal damage and behavioral impairment, and Toll-like receptor 4 (TLR4) is an important mediator of thiscascade. In the current study, we tested the hypothesis that curcumin, a phytochemical compound with potent anti-inflammatory properties that is extracted from the rhizome Curcuma longa, alleviates acute inflammatory injury mediated by TLR4 following TBI. METHODS Neurological function, brain water content and cytokine levels were tested in TLR4⁻/⁻ mice subjected to weight-drop contusion injury. Wild-type (WT) mice were injected intraperitoneally with different concentrations of curcumin or vehicle 15 minutes after TBI. At 24 hours post-injury, the activation of microglia/macrophages and TLR4 was detected by immunohistochemistry; neuronal apoptosis was measured by FJB and TUNEL staining; cytokines were assayed by ELISA; and TLR4, MyD88 and NF-κB levels were measured by Western blotting. In vitro, a co-culture system comprised of microglia and neurons was treated with curcumin following lipopolysaccharide (LPS) stimulation. TLR4 expression and morphological activation in microglia and morphological damage to neurons were detected by immunohistochemistry 24 hours post-stimulation. RESULTS The protein expression of TLR4 in pericontusional tissue reached a maximum at 24 hours post-TBI. Compared with WT mice, TLR4⁻/⁻ mice showed attenuated functional impairment, brain edema and cytokine release post-TBI. In addition to improvement in the above aspects, 100 mg/kg curcumin treatment post-TBI significantly reduced the number of TLR4-positive microglia/macrophages as well as inflammatory mediator release and neuronal apoptosis in WT mice. Furthermore, Western blot analysis indicated that the levels of TLR4 and its known downstream effectors (MyD88, and NF-κB) were also decreased after curcumin treatment. Similar outcomes were observed in the microglia and neuron co-culture following treatment with curcumin after LPS stimulation. LPS increased TLR4 immunoreactivity and morphological activation in microglia and increased neuronal apoptosis, whereas curcumin normalized this upregulation. The increased protein levels of TLR4, MyD88 and NF-κB in microglia were attenuated by curcumin treatment. CONCLUSIONS Our results suggest that post-injury, curcumin administration may improve patient outcome by reducing acute activation of microglia/macrophages and neuronal apoptosis through a mechanism involving the TLR4/MyD88/NF-κB signaling pathway in microglia/macrophages in TBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jiang-Kai Lin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 30 Gaotanyan Street, Chongqing 400038, China.
| |
Collapse
|
32
|
Phenoxybenzamine is neuroprotective in a rat model of severe traumatic brain injury. Int J Mol Sci 2014; 15:1402-17. [PMID: 24447929 PMCID: PMC3907876 DOI: 10.3390/ijms15011402] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/01/2014] [Accepted: 01/14/2014] [Indexed: 01/26/2023] Open
Abstract
Phenoxybenzamine (PBZ) is an FDA approved α-1 adrenergic receptor antagonist that is currently used to treat symptoms of pheochromocytoma. However, it has not been studied as a neuroprotective agent for traumatic brain injury (TBI). While screening neuroprotective candidates, we found that phenoxybenzamine reduced neuronal death in rat hippocampal slice cultures following exposure to oxygen glucose deprivation (OGD). Using this system, we found that phenoxybenzamine reduced neuronal death over a broad dose range (0.1 μM–1 mM) and provided efficacy when delivered up to 16 h post-OGD. We further tested phenoxybenzamine in the rat lateral fluid percussion model of TBI. When administered 8 h after TBI, phenoxybenzamine improved neurological severity scoring and foot fault assessments. At 25 days post injury, phenoxybenzamine treated TBI animals also showed a significant improvement in both learning and memory compared to saline treated controls. We further examined gene expression changes within the cortex following TBI. At 32 h post-TBI phenoxybenzamine treated animals had significantly lower expression of pro-inflammatory signaling proteins CCL2, IL1β, and MyD88, suggesting that phenoxybenzamine may exert a neuroprotective effect by reducing neuroinflammation after TBI. These data suggest that phenonxybenzamine may have application in the treatment of TBI.
Collapse
|
33
|
Rau TF, Kothiwal AS, Rova AR, Brooks DM, Rhoderick JF, Poulsen AJ, Hutchinson J, Poulsen DJ. Administration of low dose methamphetamine 12 h after a severe traumatic brain injury prevents neurological dysfunction and cognitive impairment in rats. Exp Neurol 2013; 253:31-40. [PMID: 24333768 DOI: 10.1016/j.expneurol.2013.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/20/2013] [Accepted: 12/02/2013] [Indexed: 10/25/2022]
Abstract
We recently published data that showed low dose of methamphetamine is neuroprotective when delivered 3 h after a severe traumatic brain injury (TBI). In the current study, we further characterized the neuroprotective potential of methamphetamine by determining the lowest effective dose, maximum therapeutic window, pharmacokinetic profile and gene expression changes associated with treatment. Graded doses of methamphetamine were administered to rats beginning 8 h after severe TBI. We assessed neuroprotection based on neurological severity scores, foot fault assessments, cognitive performance in the Morris water maze, and histopathology. We defined 0.250 mg/kg/h as the lowest effective dose and treatment at 12 h as the therapeutic window following severe TBI. We examined gene expression changes following TBI and methamphetamine treatment to further define the potential molecular mechanisms of neuroprotection and determined that methamphetamine significantly reduced the expression of key pro-inflammatory signals. Pharmacokinetic analysis revealed that a 24-hour intravenous infusion of methamphetamine at a dose of 0.500 mg/kg/h produced a plasma Cmax value of 25.9 ng/ml and a total exposure of 544 ng/ml over a 32 hour time frame. This represents almost half the 24-hour total exposure predicted for a daily oral dose of 25mg in a 70 kg adult human. Thus, we have demonstrated that methamphetamine is neuroprotective when delivered up to 12 h after injury at doses that are compatible with current FDA approved levels.
Collapse
Affiliation(s)
- Thomas F Rau
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Aakriti S Kothiwal
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Annela R Rova
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Diane M Brooks
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Joseph F Rhoderick
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Austin J Poulsen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Jim Hutchinson
- Montana Department of Justice Forensic Science Division, 2679 Palmer Street, Missoula, MT 59808, USA
| | - David J Poulsen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA.
| |
Collapse
|
34
|
Expression and cell distribution of receptor for advanced glycation end-products in the rat cortex following experimental subarachnoid hemorrhage. Brain Res 2013; 1543:315-23. [PMID: 24291745 DOI: 10.1016/j.brainres.2013.11.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 11/15/2013] [Accepted: 11/20/2013] [Indexed: 02/08/2023]
Abstract
Convincing evidence indicates that inflammation contributes to the adverse prognosis of subarachnoid hemorrhage (SAH). Some pro-inflammatory molecules such as high mobility group protein 1, S100 family of proteins, β-amyloid peptide, and macrophage antigen complex 1 have been involved in the damaging inflammation process following SAH. The receptor for advanced glycation end-products (RAGE) is a transmembrane receptor that senses these molecules and plays central role in inflammatory processes. This study aimed to determine the expression and cell distribution of RAGE in the brain cortex after SAH. Male Sprague-Dawley rats were randomly divided into sham group and SAH groups at 6 h, 12 h and on day 1, day 2 and day 3 (n=6 for each subgroup). SAH groups suffered experimental SAH by injection of 0.3 ml autologous blood into the prechiasmatic cistern. RAGE expression was measured by Western blot, real-time PCR, immunohistochemistry and immunofluorescence. Nuclear expression of p65 protein, the major subunit of nuclear factor kappa B, was also detected. Our data demonstrated that the expression levels of RAGE and nuclear p65 protein were both markedly increased after SAH. Moreover, there was a significant positive correlation between the expression of RAGE and that of p65 protein. Double immunofluorescence staining showed that RAGE was expressed by neuron and microglia rather than astrocyte after SAH. These results suggest that RAGE may be directly involved in the inflammatory response after SAH, and there might be important implications for further studies using specific RAGE antagonists to decrease inflammation-mediated brain injury following SAH.
Collapse
|
35
|
Liu HD, Li W, Chen ZR, Hu YC, Zhang DD, Shen W, Zhou ML, Zhu L, Hang CH. Expression of the NLRP3 inflammasome in cerebral cortex after traumatic brain injury in a rat model. Neurochem Res 2013; 38:2072-83. [PMID: 23892989 DOI: 10.1007/s11064-013-1115-z] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 07/11/2013] [Accepted: 07/17/2013] [Indexed: 12/15/2022]
Abstract
Inflammatory response plays an important role in the pathogenesis of secondary damage after traumatic brain injury (TBI). The inflammasome is a multiprotein complex involved in innate immunity and a number of studies have suggested that the inflammasome plays a critical role in a host inflammatory signaling. Nucleotide-binding domain, leucine-rich repeat, pyrin domain containing 3 (NLRP3) is a key component of the NLRP3-inflammasome, which also includes apoptotic speck-containing protein (ASC) with a cysteine protease (caspase)-activating recruitment domain and pro-caspase1. Activation of the NLRP3-inflammasome causes the processing and release of the interleukin 1 beta (IL-1β) and interleukin 18 (IL-18). Based on this, we hypothesized that the NLRP3-inflammasome could participate in the inflammatory response following TBI. However, the expression of NLRP3-inflammasome in cerebral cortex after TBI is not well known. Rats were randomly divided into control, sham and TBI groups (including 6 h, 1 day, 3 day and 7 day sub-group). TBI model was induced, and animals were sacrificed at each time point respectively. The expression of NLRP3-inflammasome was measured by quantitative real-time polymerase chain reaction, western blot and immunohistochemistry respectively. Immunofluorescent double labeling was performed to identify the cell types of NLRP3-inflammasome's expression. Moreover, enzyme linked immunosorbent assay was used to detect the alterations of IL-1β and IL-18 at each time point post-injury. The results showed that, TBI could induce assembly of NLRP3-inflammasome complex, increased expression of ASC, activation of caspase1, and processing of IL-1β and IL-18. These results suggested that NLRP3-inflammasome might play an important role in the inflammation induced by TBI and could be a target for TBI therapy.
Collapse
Affiliation(s)
- Huan-Dong Liu
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Southern Medical University (Guangzhou), 305 East Zhongshan Road, Nanjing, 210002, Jiangsu Province, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Li W, Ling HP, You WC, Liu HD, Sun Q, Zhou ML, Shen W, Zhao JB, Zhu L, Hang CH. Elevated Cerebral Cortical CD24 Levels in Patients and Mice with Traumatic Brain Injury: A Potential Negative Role in Nuclear Factor Kappa B/Inflammatory Factor Pathway. Mol Neurobiol 2013; 49:187-98. [DOI: 10.1007/s12035-013-8509-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 07/08/2013] [Indexed: 01/28/2023]
|
37
|
Mao SS, Hua R, Zhao XP, Qin X, Sun ZQ, Zhang Y, Wu YQ, Jia MX, Cao JL, Zhang YM. Exogenous administration of PACAP alleviates traumatic brain injury in rats through a mechanism involving the TLR4/MyD88/NF-κB pathway. J Neurotrauma 2013; 29:1941-59. [PMID: 22583372 DOI: 10.1089/neu.2011.2244] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is effective in reducing axonal damage associated with traumatic brain injury (TBI), and has immunomodulatory properties. Toll-like receptor 4 (TLR4) is an important mediator of the innate immune response. It significantly contributes to neuroinflammation induced by brain injury. However, it remains unknown whether exogenous PACAP can modulate TBI through the TLR4/adapter protein myeloid differentiation factor 88 (MyD88)/nuclear factor-κB (NF-κB) signaling pathway. In this study, we investigated the potential neuroprotective mechanisms of PACAP pretreatment in a weight-drop model of TBI. PACAP38 was microinjected intracerebroventricularly before TBI. Brain samples were extracted from the pericontusional area in the cortex and hippocampus. We found that TBI induced significant upregulation of TLR4, with peak expression occurring 24 h post-trauma, and that pretreatment with PACAP significantly improved motor and cognitive dysfunction, attenuated neuronal apoptosis, and decreased brain edema. Pretreatment with PACAP inhibited upregulation of TLR4 and its downstream signaling molecules MyD88, p-IκB, and NF-κB, and suppressed increases in the levels of the downstream inflammatory agents interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), in the brain tissue around the injured cortex and in the hippocampus. Administration of PACAP both in vitro and in vivo attenuated the ability of the TLR4 agonist lipopolysaccharide (LPS) to increase TLR4 protein levels. Therefore, PACAP exerts a neuroprotective effect in this rat model of TBI, by inhibiting a secondary inflammatory response mediated by the TLR4/MyD88/NF-κB signaling pathway in microglia and neurons, thereby reducing neuronal death and improving the outcome following TBI.
Collapse
Affiliation(s)
- Shan-Shan Mao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sun Q, Dai Y, Zhang X, Hu YC, Zhang D, Li W, Zhang XS, Zhu JH, Zhou ML, Hang CH. Expression and cell distribution of myeloid differentiation primary response protein 88 in the cerebral cortex following experimental subarachnoid hemorrhage in rats: a pilot study. Brain Res 2013; 1520:134-44. [PMID: 23684713 DOI: 10.1016/j.brainres.2013.05.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 04/18/2013] [Accepted: 05/02/2013] [Indexed: 11/24/2022]
Abstract
Subarachnoid hemorrhage (SAH) which is mostly caused by aneurysm rupture causes a lot of death every year. Convincing evidence can be made that inflammation contributes to the poor outcome caused by SAH. Toll like receptors (TLRs), nuclear factor-kappaB (NF-κB), Interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α) are involved in the damaging inflammation process after SAH. Myeloid differentiation primary response protein 88 (MyD88) is essential to deliver TLRs signals down to NF-κB and pro-inflammatory factors. The study aims to detect the expression level of MyD88 and know more about the role of MyD88 after SAH. Sprague Dawley (SD) rats were randomly divided into sham group and SAH groups at 2h, 6h, 12h and on day 1, day 2, day 3, day 5 and day 7. SAH groups suffered experimental subarachnoid hemorrhage by injection of 0.3 ml autoblood into the prechiasmatic cistern. MyD88 expression is measured by western blot analysis, real-time polymerase chain reaction (PCR), immunohistochemistry and immunofluorescence. The levels of TNF-α and IL-1β were measured by real-time PCR. Our results demonstrated MyD88 expression was increased after SAH, and peaked on day 1 and day 5, which showed a parallel time course to the up-regulation of IL-1β, there was a highly positive relationship between them. Immunohistochemistry and immunofluorescence results indicated up-regulated MyD88 was mainly located in neurons while over expressed MyD88 could also be found in astrocytes and microglia. These results might have important implications during the administration of specific MyD88 antagonists in order to prevent or reduce inflammatory response following SAH.
Collapse
Affiliation(s)
- Qing Sun
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hernandez-Ontiveros DG, Tajiri N, Acosta S, Giunta B, Tan J, Borlongan CV. Microglia activation as a biomarker for traumatic brain injury. Front Neurol 2013; 4:30. [PMID: 23531681 PMCID: PMC3607801 DOI: 10.3389/fneur.2013.00030] [Citation(s) in RCA: 201] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 03/10/2013] [Indexed: 12/24/2022] Open
Abstract
Traumatic brain injury (TBI) has become the signature wound of wars in Afghanistan and Iraq. Injury may result from a mechanical force, a rapid acceleration-deceleration movement, or a blast wave. A cascade of secondary cell death events ensues after the initial injury. In particular, multiple inflammatory responses accompany TBI. A series of inflammatory cytokines and chemokines spreads to normal brain areas juxtaposed to the core impacted tissue. Among the repertoire of immune cells involved, microglia is a key player in propagating inflammation to tissues neighboring the core site of injury. Neuroprotective drug trials in TBI have failed, likely due to their sole focus on abrogating neuronal cell death and ignoring the microglia response despite these inflammatory cells’ detrimental effects on the brain. Another relevant point to consider is the veracity of results of animal experiments due to deficiencies in experimental design, such as incomplete or inadequate method description, data misinterpretation, and reporting may introduce bias and give false-positive results. Thus, scientific publications should follow strict guidelines that include randomization, blinding, sample-size estimation, and accurate handling of all data (Landis et al., 2012). A prolonged state of inflammation after brain injury may linger for years and predispose patients to develop other neurological disorders, such as Alzheimer’s disease. TBI patients display progressive and long-lasting impairments in their physical, cognitive, behavioral, and social performance. Here, we discuss inflammatory mechanisms that accompany TBI in an effort to increase our understanding of the dynamic pathological condition as the disease evolves over time and begin to translate these findings for defining new and existing inflammation-based biomarkers and treatments for TBI.
Collapse
Affiliation(s)
- Diana G Hernandez-Ontiveros
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | | | | | | | | | | |
Collapse
|
40
|
Li W, Liu HD, You WC, Zhou ML, Ling HP, Shen W, Zhu L, Hang CH. Enhanced cortical expression of myeloid differentiation primary response protein 88 (Myd88) in patients with traumatic brain injury. J Surg Res 2013. [DOI: 10.1016/j.jss.2012.10.928] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Zhang D, Hu Y, Sun Q, Zhao J, Cong Z, Liu H, Zhou M, Li K, Hang C. Inhibition of transforming growth factor beta-activated kinase 1 confers neuroprotection after traumatic brain injury in rats. Neuroscience 2013; 238:209-17. [PMID: 23485590 DOI: 10.1016/j.neuroscience.2013.02.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 01/23/2013] [Accepted: 02/14/2013] [Indexed: 01/23/2023]
Abstract
The transforming growth factor beta-activated kinase 1 (TAK1), a member of the Mitogen-activated protein kinase kinase kinase family, is characterized as a key regulator in inflammatory and apoptosis signaling pathways. The aim of the present study was to evaluate the role of the TAK1 pathway in experimental traumatic brain injury (TBI) in rats. Adult male Sprague-Dawley rats were subjected to TBI using a modified Feeney's weight-drop model. The time course showed that a significant increase of TAK1 and p-TAK1 expression in the cortex after TBI. Moreover, TBI induced TAK1 redistribution both in neurons and astrocytes of the lesion boundary zone. The effects of specific inhibition of the TAK1 pathway by 5Z-7-oxozeaenol (OZ, intracerebroventricular injection at 10min post-trauma) on histopathological and behavioral outcomes in rats were assessed at 24h post injury. The number of TUNEL-positive stained cells was diminished and neuronal survival and neurological function were improved with OZ treatment. Biochemically, the high dose of OZ significantly reduced the levels of TAK1 and p-TAK1, further decreased nuclear factor-κB and activator protein 1 activities and the release of inflammatory cytokines. In addition, we found that both 10min and 3h post-trauma OZ therapies could markedly improve neurological function and neuronal survival after long-term survival. These results revealed that the TAK1 pathway is activated after experimental TBI and the inhibitor OZ affords significant neuro- protection and amelioration of neurobehavioral deficits after experimental TBI, suggesting a potential rationale for manipulating this pathway in clinical practice.
Collapse
Affiliation(s)
- D Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu Province, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ji J. Expression of myeloid-differentiation factor 88 after traumatic brain injury. J Surg Res 2013; 185:547-8. [PMID: 23394933 DOI: 10.1016/j.jss.2013.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 12/23/2012] [Accepted: 01/03/2013] [Indexed: 01/04/2023]
Affiliation(s)
- Jing Ji
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
43
|
Recombinant high-mobility group box 1 protein (HMGB-1) promotes myeloid differentiation primary response protein 88 (Myd88) upregulation in mouse primary cortical neurons. Neurol Sci 2012; 34:847-53. [PMID: 22710699 DOI: 10.1007/s10072-012-1131-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 06/05/2012] [Indexed: 01/03/2023]
Abstract
Myeloid differentiation primary response protein 88 (Myd88) is a vital factor for inflammation and immunity, and high-mobility group box 1 protein (HMGB-1) can be released from neurons after injury and may contribute to the initial stages of inflammatory response. Therefore, the current study was intended to investigate the expression of Myd88 in cultured neurons following recombinant HMGB-1 (rHMGB-1) addition and to clarify the potential role of Myd88 after neuron injury in vitro. The cultured neurons were randomly divided into six groups: control group and rHMGB-1 groups at hours 1, 6, 12, 24, and 48. The cultured neurons in rHMGB-1 groups were subjected to rHMGB-1 addition. The expression of Myd88 was assessed by quantitative real-time polymerase chain reaction (PCR), Western blotting and immunofluorescence, and nuclear factor kappa B (NF-κB) DNA-binding activity was detected by electrophoretic mobility shift assay, and the levels of tumor necrosis factor-α (TNF-α) and interleukin 1β (IL-1β) were measured by quantitative real-time PCR. The elevated mRNA and protein levels of Myd88, peaking at 24 h, were detected after rHMGB-1 addition. NF-κB, TNF-α, and IL-1β also ascended significantly after rHMGB-1 addition. Interestingly, Myd88 increasingly expressed in a parallel time course to the upregulation of NF-κB, TNF-α, and IL-1β. These findings indicated a possible role of Myd88 in the inflammatory response after neuron injury, and might provide an attractive therapeutic approach of targeting the Myd88 cascade to achieve better outcomes for patients with central nervous system injury.
Collapse
|
44
|
Seddik L, Bah TM, Aoues A, Slimani M, Benderdour M. Elucidation of mechanisms underlying the protective effects of olive leaf extract against lead-induced neurotoxicity in Wistar rats. J Toxicol Sci 2012; 36:797-809. [PMID: 22129743 DOI: 10.2131/jts.36.797] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Recently, we identified that olive leaf extract (OLE) prevents lead (Pb)-induced abnormalities in behavior and neurotransmitters production in chronic Pb exposure in rats. The aim of the present study was to provide additional evidence that OLE acts as an anti-apoptotic, anti-inflammatory, and antioxidant mediator in Pb exposed rats. 4-weeks old Wistar rats were exposed or not to 250 mg/l Pb for 13-weeks and then exposed to tap water containing or not 0.1% OLE for additional 2-weeks. Atomic absorption spectrophotometry showed significantly elevated Pb levels in the hippocampus and serum and reaches 5 and 42 µg/mg tissue, respectively. In the hippocampus, the examination of markers of apoptosis and inflammation revealed an increase in caspase-3 activity and DNA fragmentation as well as tumor necrosis factor alpha, interleukin-1 beta and prostaglandin E2 in Pb-exposed rats. In addition, our findings showed that Pb induced 4-hydroxynonenal production and inhibited antioxidant-related enzyme activity, such as glutathione-S-transferase as wells as energy metabolism-related enzyme activity, such as NADP-isocitrate dehydrogenase and glucose transporter. Upon examination of signaling pathways involved in apoptosis process, we found that Pb induced p38 mitogen activated protein kinase (MAPK) and Akt phosphorylation, but in contrast, inhibited that of ERK(1/2). Interestingly, OLE administration diminished tissue Pb deposition and prevented all Pb effects. In the frontal cortex, our data also showed that OLE-abolished Pb-induced caspase-3 activity and DNA fragmentation. Collectively, these data support the use of OLE by traditional medicine to counter Pb neurotoxicity.
Collapse
Affiliation(s)
- Leila Seddik
- Département de Biologie, Faculté des Sciences, Université d'Es-Senia, Oran, Algeria
| | | | | | | | | |
Collapse
|