1
|
Yao H, Tian J, Cheng S, Dou H, Zhu Y. The mechanism of hypoxia-inducible factor-1α enhancing the transcriptional activity of transferrin ferroportin 1 and regulating the Nrf2/HO-1 pathway in ferroptosis after cerebral ischemic injury. Neuroscience 2024; 559:26-38. [PMID: 39168172 DOI: 10.1016/j.neuroscience.2024.08.025] [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: 01/06/2024] [Revised: 07/05/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024]
Abstract
Cerebral ischemic/reperfusion (I/R) injury has high disability and morbidity. Hypoxia-inducible factor-1α (HIF-1α) may enhance the transcriptional activity of transferrin ferroportin 1 (FPN1) in regulating ferroptosis after cerebral ischemia injury (CII). In this study, cerebral I/R injury rat models were established and treated with pcDNA3.1-HIF-1α, pcDNA3.1-NC lentiviral plasmid, or ML385 (a specific Nrf2 inhibitor). Additionally, oxygen-glucose deprivation/reoxygenation (OGD/R) exposed PC12 cells were used as an in vitro model of cerebral ischemia and treated with pcDNA3.1-HIF-1α, si-FPN1, or ML385. The results elicited that cerebral I/R injury rats exhibited increased Longa scores, TUNEL and NeuN co-positive cells, Fe2+ concentration, ROS and HIF-1α levels, and MDA content, while reduced cell density and number, GSH content, and GPX4 protein level. Morphologically abnormal and disordered hippocampal neurons were also observed in CII rats. HIF-1α inhibited brain neuron ferroptosis and ameliorated I/R injury. HIF-1α alleviated OGD-induced PC12 cell ferroptosis. OGD/R decreased FPN1 protein level in PC12 cells, and HIF-1α enhanced FPN1 transcriptional activity. FPN1 knockdown reversed HIF-1α-mediated alleviation of OGD/R-induced ferroptosis. HIF-1α activated the Nrf2/HO-1 pathway by enhancing FPN1 expression and alleviating OGD/R-induced ferroptosis. Conjointly, HIF-1α enhanced the transcriptional activity of FPN1, activated the Nrf2/HO-1 pathway, and inhibited ferroptosis of brain neurons, thereby improving I/R injury in CII rats.
Collapse
Affiliation(s)
- Haiqian Yao
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Jianan Tian
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Shi Cheng
- Department of Orthopaedics, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Haitong Dou
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Yulan Zhu
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
2
|
Nakamura A, Sakai S, Taketomi Y, Tsuyama J, Miki Y, Hara Y, Arai N, Sugiura Y, Kawaji H, Murakami M, Shichita T. PLA2G2E-mediated lipid metabolism triggers brain-autonomous neural repair after ischemic stroke. Neuron 2023; 111:2995-3010.e9. [PMID: 37490917 DOI: 10.1016/j.neuron.2023.06.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 03/08/2023] [Accepted: 06/23/2023] [Indexed: 07/27/2023]
Abstract
The brain is generally resistant to regeneration after damage. The cerebral endogenous mechanisms triggering brain self-recovery have remained unclarified to date. We here discovered that the secreted phospholipase PLA2G2E from peri-infarct neurons generated dihomo-γ-linolenic acid (DGLA) as necessary for triggering brain-autonomous neural repair after ischemic brain injury. Pla2g2e deficiency diminished the expression of peptidyl arginine deiminase 4 (Padi4), a global transcriptional regulator in peri-infarct neurons. Single-cell RNA sequencing (scRNA-seq) and epigenetic analysis demonstrated that neuronal PADI4 had the potential for the transcriptional activation of genes associated with recovery processes after ischemic stroke through histone citrullination. Among various DGLA metabolites, we identified 15-hydroxy-eicosatrienoic acid (15-HETrE) as the cerebral metabolite that induced PADI4 in peri-infarct-surviving neurons. Administration of 15-HETrE enhanced functional recovery after ischemic stroke. Thus, our research clarifies the promising potential of brain-autonomous neural repair triggered by the specialized lipids that initiate self-recovery processes after brain injury.
Collapse
Affiliation(s)
- Akari Nakamura
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Department of Neuroinflammation and Repair, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; Core Research for Evolutionary Medical Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo 100-0004, Japan
| | - Seiichiro Sakai
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Department of Neuroinflammation and Repair, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; Core Research for Evolutionary Medical Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo 100-0004, Japan
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Science Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Jun Tsuyama
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Department of Neuroinflammation and Repair, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; Core Research for Evolutionary Medical Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo 100-0004, Japan
| | - Yoshimi Miki
- Laboratory of Microenvironmental and Metabolic Health Science Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yuichiro Hara
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Nobutaka Arai
- Laboratory for Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hideya Kawaji
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Takashi Shichita
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; Department of Neuroinflammation and Repair, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Core Research for Evolutionary Medical Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo 100-0004, Japan.
| |
Collapse
|
3
|
Gou X, Ying J, Yue Y, Qiu X, Hu P, Qu Y, Li J, Mu D. The Roles of High Mobility Group Box 1 in Cerebral Ischemic Injury. Front Cell Neurosci 2020; 14:600280. [PMID: 33384585 PMCID: PMC7770223 DOI: 10.3389/fncel.2020.600280] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that plays an important role in stabilizing nucleosomes and DNA repair. HMGB1 can be passively released from necrotic neurons or actively secreted by microglia, macrophages/monocytes, and neutrophils. Cerebral ischemia is a major cause of mortality and disability worldwide, and its outcome depends on the number of neurons dying due to hypoxia in the ischemic area. HMGB1 contributes to the pathogenesis of cerebral ischemia via mediating neuroinflammatory responses to cerebral ischemic injury. Extracellular HMGB1 regulates many neuroinflammatory events by interacting with its different cell surface receptors, such as receptors for advanced glycation end products, toll-like receptor (TLR)-2, and TLR-4. Additionally, HMGB1 can be redox-modified, thus exerting specific cellular functions in the ischemic brain and has different roles in the acute and late stages of cerebral ischemic injury. However, the role of HMGB1 in cerebral ischemia is complex and remains unclear. Herein, we summarize and review the research on HMGB1 in cerebral ischemia, focusing especially on the role of HMGB1 in hypoxic ischemia in the immature brain and in white matter ischemic injury. We also outline the possible mechanisms of HMGB1 in cerebral ischemia and the main strategies to inhibit HMGB1 pertaining to its potential as a novel critical molecular target in cerebral ischemic injury.
Collapse
Affiliation(s)
- Xiaoyun Gou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Yue
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Xia Qiu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Peng Hu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Jinhui Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Kolosowska N, Gotkiewicz M, Dhungana H, Giudice L, Giugno R, Box D, Huuskonen MT, Korhonen P, Scoyni F, Kanninen KM, Ylä-Herttuala S, Turunen TA, Turunen MP, Koistinaho J, Malm T. Intracerebral overexpression of miR-669c is protective in mouse ischemic stroke model by targeting MyD88 and inducing alternative microglial/macrophage activation. J Neuroinflammation 2020; 17:194. [PMID: 32560730 PMCID: PMC7304130 DOI: 10.1186/s12974-020-01870-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 06/08/2020] [Indexed: 12/30/2022] Open
Abstract
Background Ischemic stroke is a devastating disease without a cure. The available treatments for ischemic stroke, thrombolysis by tissue plasminogen activator, and thrombectomy are suitable only to a fraction of patients and thus novel therapeutic approaches are urgently needed. The neuroinflammatory responses elicited secondary to the ischemic attack further aggravate the stroke-induced neuronal damage. It has been demonstrated that these responses are regulated at the level of non-coding RNAs, especially miRNAs. Methods We utilized lentiviral vectors to overexpress miR-669c in BV2 microglial cells in order to modulate their polarization. To detect whether the modulation of microglial activation by miR-669c provides protection in a mouse model of transient focal ischemic stroke, miR-669c overexpression was driven by a lentiviral vector injected into the striatum prior to induction of ischemic stroke. Results Here, we demonstrate that miR-669c-3p, a member of chromosome 2 miRNA cluster (C2MC), is induced upon hypoxic and excitotoxic conditions in vitro and in two different in vivo models of stroke. Rather than directly regulating the neuronal survival in vitro, miR-669c is capable of attenuating the microglial proinflammatory activation in vitro and inducing the expression of microglial alternative activation markers arginase 1 (Arg1), chitinase-like 3 (Ym1), and peroxisome proliferator-activated receptor gamma (PPAR-γ). Intracerebral overexpression of miR-669c significantly decreased the ischemia-induced cell death and ameliorated the stroke-induced neurological deficits both at 1 and 3 days post injury (dpi). Albeit miR-669c overexpression failed to alter the overall Iba1 protein immunoreactivity, it significantly elevated Arg1 levels in the ischemic brain and increased colocalization of Arg1 and Iba1. Moreover, miR-669c overexpression under cerebral ischemia influenced several morphological characteristics of Iba1 positive cells. We further demonstrate the myeloid differentiation primary response gene 88 (MyD88) transcript as a direct target for miR-669c-3p in vitro and show reduced levels of MyD88 in miR-669c overexpressing ischemic brains in vivo. Conclusions Collectively, our data provide the evidence that miR-669c-3p is protective in a mouse model of ischemic stroke through enhancement of the alternative microglial/macrophage activation and inhibition of MyD88 signaling. Our results accentuate the importance of controlling miRNA-regulated responses for the therapeutic benefit in conditions of stroke and neuroinflammation.
Collapse
Affiliation(s)
- Natalia Kolosowska
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Maria Gotkiewicz
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Hiramani Dhungana
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Luca Giudice
- Department of Computer Science, University of Verona, Verona, Italy
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, Verona, Italy
| | - Daphne Box
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Mikko T Huuskonen
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Paula Korhonen
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Flavia Scoyni
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Katja M Kanninen
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Tiia A Turunen
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Mikko P Turunen
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Jari Koistinaho
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland.,Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tarja Malm
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
5
|
Famakin BM, Vemuganti R. Toll-Like Receptor 4 Signaling in Focal Cerebral Ischemia: a Focus on the Neurovascular Unit. Mol Neurobiol 2020; 57:2690-2701. [PMID: 32306272 DOI: 10.1007/s12035-020-01906-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/19/2020] [Indexed: 12/14/2022]
Abstract
A robust innate immune activation leads to downstream expression of inflammatory mediators that amplify tissue damage and consequently increase the morbidity after stroke. The Toll-like receptor 4 (TLR4) pathway is a major innate immune pathway activated acutely and chronically after stroke. Hence, understanding the intricacies of the temporal profile, specific control points, and cellular specificity of TLR4 activation is crucial for the development of any novel therapeutics targeting the endogenous innate immune response after focal cerebral ischemia. The goal of this review is to summarize the current findings related to TLR4 signaling after stroke with a specific focus on the components of the neurovascular unit such as astrocytes, neurons, endothelial cells, and pericytes. In addition, this review will examine the effects of focal cerebral ischemia on interaction of these neurovascular unit components.
Collapse
Affiliation(s)
| | - R Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- William S. Middleton VA Hospital, Madison, WI, USA
| |
Collapse
|
6
|
HMGB1 is a Potential Mediator of Astrocytic TLR4 Signaling Activation following Acute and Chronic Focal Cerebral Ischemia. Neurol Res Int 2020; 2020:3929438. [PMID: 32148958 PMCID: PMC7053497 DOI: 10.1155/2020/3929438] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/02/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022] Open
Abstract
Limited, and underutilized, therapeutic options for acute stroke require new approaches to treatment. One such potential approach involves better understanding of innate immune response to brain injury such as acute focal cerebral ischemia. This includes understanding the temporal profile, and specificity, of Toll-like receptor 4 (TLR4) signaling in brain cell types, such as astrocytes, following focal cerebral ischemia. This study evaluated TLR4 signaling, and downstream mediators, in astrocytes, during acute and chronic phases post transient middle cerebral artery occlusion (MCAO). We also determined whether high mobility group box 1 (HMGB1), an endogenous TLR4 ligand, was sufficient to induce TLR4 signaling activation in astrocytes in vivo and in vitro. We injected HMGB1 into normal cortex, in vivo, and stimulated cultured astrocytes with HMGB1, in vitro, and determined TLR4, and downstream mediator, expression by immunohistochemistry. We found that expression of TLR4, and downstream mediators, such as inducible nitric oxide synthase (iNOS), occurs in penumbral astrocytes in acute and chronic phases after focal cerebral ischemia, but was undetectable in cortical astrocytes in the contralateral hemisphere. In addition, cortical injection of recombinant HMGB1 led to a trend towards an almost 2-fold increase in TLR4 expression in astrocytes surrounding the injection site. Consistent with these results, in vitro stimulation of the DI TNC1 astrocyte cell line, with recombinant HMGB1, led to increased TLR4 and iNOS message levels. These findings suggest that HMGB1, an endogenous TLR4 ligand, is an important physiological ligand for TLR4 signaling activation, in penumbral astrocytes, following acute and chronic ischemia and HMGB1 amplifies TLR4 signaling in astrocytes.
Collapse
|
7
|
Xu M, Liu PP, Li H. Innate Immune Signaling and Its Role in Metabolic and Cardiovascular Diseases. Physiol Rev 2019; 99:893-948. [PMID: 30565509 DOI: 10.1152/physrev.00065.2017] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The innate immune system is an evolutionarily conserved system that senses and defends against infection and irritation. Innate immune signaling is a complex cascade that quickly recognizes infectious threats through multiple germline-encoded cell surface or cytoplasmic receptors and transmits signals for the deployment of proper countermeasures through adaptors, kinases, and transcription factors, resulting in the production of cytokines. As the first response of the innate immune system to pathogenic signals, inflammatory responses must be rapid and specific to establish a physical barrier against the spread of infection and must subsequently be terminated once the pathogens have been cleared. Long-lasting and low-grade chronic inflammation is a distinguishing feature of type 2 diabetes and cardiovascular diseases, which are currently major public health problems. Cardiometabolic stress-induced inflammatory responses activate innate immune signaling, which directly contributes to the development of cardiometabolic diseases. Additionally, although the innate immune elements are highly conserved in higher-order jawed vertebrates, lower-grade jawless vertebrates lack several transcription factors and inflammatory cytokine genes downstream of the Toll-like receptors (TLRs) and retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) pathways, suggesting that innate immune signaling components may additionally function in an immune-independent way. Notably, recent studies from our group and others have revealed that innate immune signaling can function as a vital regulator of cardiometabolic homeostasis independent of its immune function. Therefore, further investigation of innate immune signaling in cardiometabolic systems may facilitate the discovery of new strategies to manage the initiation and progression of cardiometabolic disorders, leading to better treatments for these diseases. In this review, we summarize the current progress in innate immune signaling studies and the regulatory function of innate immunity in cardiometabolic diseases. Notably, we highlight the immune-independent effects of innate immune signaling components on the development of cardiometabolic disorders.
Collapse
Affiliation(s)
- Meng Xu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Peter P Liu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| |
Collapse
|
8
|
Long J, He C, Dai H, Kang X, Zou J, Ye S, Yu Q. Effects of transection of cervical sympathetic trunk on cognitive function of traumatic brain injury rats. Neuropsychiatr Dis Treat 2019; 15:1121-1131. [PMID: 31118645 PMCID: PMC6506013 DOI: 10.2147/ndt.s199450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 03/28/2019] [Indexed: 01/04/2023] Open
Abstract
Objective: To observe the effects of transection of cervical sympathetic trunk (TCST) on the cognitive function of traumatic brain injury (TBI) rats and the potential mechanisms. Methods: A total of 288 adult male SD rats were divided into 3 groups using a random number table: TBI group (n=96), TBI + TCST group (n=96) and Sham group (n=96). The water maze test was performed before TBI (T0) and at day 1 (T1), day 2 (T2), day 3 (T3), 1 week (T4), 2 weeks (T5), 6 weeks (T6) and 12 weeks (T7) after TBI. The levels of α1-adrenergic receptors (α1-ARs), α2-adrenergic receptors (α2-ARs), toll-like receptor 4 (TLR-4) and P38 in hippocampi were detected by real-time PCR. Hippocampal P38 expression was assayed by Western blot. The expressions of interleukin-6 (IL-6), tumor necrosis factor (TNF-α) and brain-derived neurotrophic factor (BDNF) were examined by immunohistochemistry. Noradrenaline (NE) expression in plasma was evaluated by ELISA. The respiratory control ratio (RCR) of brain mitochondria was detected using a Clark oxygen electrode. Results: TCST effectively improved the cognitive function of TBI rats. TCST significantly inhibited sympathetic activity in the rats and effectively inhibited inflammatory responses. The expression of BDNF at T1-T6 in TBI+TCST group was higher than that in TBI group (P<0.05). Furthermore, P38 expression was inhibited more effectively in TBI+TCST group (P<0.05), than in TBI group (P<0.05), and the RCR of the brain was significantly higher in TBI+TCST group than in TBI group (P<0.05). Conclusions: TCST can enhance cognitive function in TBI rats by inhibiting sympathetic activity, reducing inflammatory responses and brain edema, upregulating BDNF and improving brain mitochondrial function.
Collapse
Affiliation(s)
- Juan Long
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Chunjing He
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Hong Dai
- Department of Neurology, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Xinguo Kang
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Jinfeng Zou
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Shengli Ye
- Department of Pain, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| | - Qian Yu
- Department of Neurology, Guizhou Province People's Hospital, Guiyang, People's Republic of China
| |
Collapse
|
9
|
Li M, Liu J, Bi Y, Chen J, Zhao L. Potential Medications or Compounds Acting on Toll-like Receptors in Cerebral Ischemia. Curr Neuropharmacol 2018; 16:160-175. [PMID: 28571545 PMCID: PMC5883378 DOI: 10.2174/1570159x15666170601125139] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/24/2017] [Accepted: 05/31/2017] [Indexed: 01/22/2023] Open
Abstract
Background: Toll-like receptors play an integral role in the process of inflammatory response after ischemic in-jury. The therapeutic potential acting on TLRs is worth of evaluations. The aim of this review was to introduce readers some potential medications or compounds which could alleviate the ischemic damage via TLRs. Methods: Research articles online on TLRs were reviewed. Categorizations were listed according to the follows, methods acting on TLRs directly, modulations of MyD88 or TRIF signaling pathway, and the ischemic tolerance induced by the pre-conditioning or postconditioning with TLR ligands or minor cerebral ischemia via acting on TLRs. Results: There are only a few studies concerning on direct effects. Anti-TLR4 or anti-TLR2 therapies may serve as promis-ing strategies in acute events. Approaches targeting on inhibiting NF-κB signaling pathway and enhancing interferon regu-latory factor dependent signaling have attracted great interests. Not only drugs but compounds extracted from traditional Chinese medicine have been used to identify their neuroprotective effects against cerebral ischemia. In addition, many re-searchers have reported the positive therapeutic effects of preconditioning with agonists of TLR2, 3, 4, 7 and 9. Several trails have also explored the potential of postconditioning, which provide a new idea in ischemic treatments. Considering all the evidence above, many drugs and new compounds may have great potential to reduce ischemic insults. Conclusion: This review will focus on promising therapies which exerting neuroprotective effects against ischemic injury by acting on TLRs.
Collapse
Affiliation(s)
- Man Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of Neurology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, China
| | - Ying Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jixiang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Ye X, Kong D, Wang J, Ishrat T, Shi H, Ding X, Cui G, Hua F. MyD88 contributes to neuroinflammatory responses induced by cerebral ischemia/reperfusion in mice. Biochem Biophys Res Commun 2016; 480:69-74. [PMID: 27717824 DOI: 10.1016/j.bbrc.2016.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/03/2016] [Indexed: 02/06/2023]
Abstract
Myeloid differentiation primary-response protein-88 (MyD88) is one of adaptor proteins mediating Toll-like receptors (TLRs) signaling. Activation of MyD88 results in the activation of nuclear factor kappa B (NFκB) and the increase of inflammatory responses. Evidences have demonstrated that TLRs signaling contributes to cerebral ischemia/reperfusion (I/R) injury. However, the role of MyD88 in this mechanism of action is disputed and needs to be clarified. In the present study, in a mouse model of cerebral I/R, we examined the activities of NFκB and interferon factor-3 (IRF3), and the inflammatory responses in ischemic brain tissue using ELISA, Western blots, and real-time PCR. Neurological function and cerebral infarct size were also evaluated 24 h after cerebral I/R. Our results showed that NFκB activity increased in ischemic brains, but IRF3 was not activated after cerebral I/R, in wild-type (WT) mice. MyD88 deficit inhibited the activation of NFκB, and the expression of interleukin-1β (IL-1β), IL-6, Beclin-1 (BECN1), pellino-1, and cyclooxygenase-2 (COX-2) increased by cerebral I/R compared with WT mice. Interestingly, the expression of interferon Beta 1 (INFB1) and vascular endothelial growth factor (VEGF) increased in MyD88 KO mice. Unexpectedly, although the neurological function improved in the MyD88 knockout (KO) mice, the deficit of MyD88 failed to reduce cerebral infarct size compared to WT mice. We concluded that MyD88-dependent signaling contributes to the inflammatory responses induced by cerebral I/R. MyD88 deficit may inhibit the increased inflammatory response and increase neuroprotective signaling.
Collapse
Affiliation(s)
- Xinchun Ye
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Delian Kong
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Jun Wang
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Tauheed Ishrat
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Hongjuan Shi
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Xiaohui Ding
- Department of Human Anatomy, Histology and Embryology, Shenyang Medical College, Shenyang, 110000, China
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA; Key Laboratory of Anesthesiology of Jiangsu Province, Xuzhou 221002, China.
| |
Collapse
|
11
|
Singh V, Roth S, Veltkamp R, Liesz A. HMGB1 as a Key Mediator of Immune Mechanisms in Ischemic Stroke. Antioxid Redox Signal 2016; 24:635-51. [PMID: 26493086 DOI: 10.1089/ars.2015.6397] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
SIGNIFICANCE Stroke is the leading cause of morbidity and mortality worldwide. Inflammatory cascades have a major impact on outcome and regeneration after ischemic stroke. High-mobility group box 1 (HMGB1) has come into the focus of experimental and clinical stroke research because it is released from necrotic brain tissue and its differential redox forms attract and activate immune cells after ischemic brain injury. HMGB1 is a potent inducer of inflammatory cascades, and thereby, secondary deterioration of neurological outcome. RECENT ADVANCES The role of HMGB1 in sterile inflammation is well established. Emerging evidence suggests that HMGB1 modulates neuroinflammation after experimental brain ischemia and that it may be a useful prognostic biomarker for stroke patients. CRITICAL ISSUES HMGB1 is instantly released from necrotic cells in the ischemic core and activates an early inflammatory response. In addition, brain-released HMGB1 can be redox modified in the circulation and activate peripheral immune cells. HMGB1 concentrations correlate with disease severity and outcome after brain injury. This is the first review depicting the crucial role of HMGB1 in the initiation and perpetuation of secondary immune alterations after experimental and clinical stroke. FUTURE DIRECTIONS HMGB1-dependent signaling pathways are on the verge and have the potential to become a central topic in experimental stroke research. Current and upcoming projects in this field will be paving the way for future translational approaches targeting the center of poststroke inflammation to improve stroke recovery and long-term outcome.
Collapse
Affiliation(s)
- Vikramjeet Singh
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| | - Stefan Roth
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| | - Roland Veltkamp
- 3 Division of Brain Sciences, Imperial College London , London, United Kingdom
| | - Arthur Liesz
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| |
Collapse
|
12
|
Liang KC, Patil A, Nakai K. Discovery of Intermediary Genes between Pathways Using Sparse Regression. PLoS One 2015; 10:e0137222. [PMID: 26348038 PMCID: PMC4562633 DOI: 10.1371/journal.pone.0137222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 08/14/2015] [Indexed: 01/18/2023] Open
Abstract
The use of pathways and gene interaction networks for the analysis of differential expression experiments has allowed us to highlight the differences in gene expression profiles between samples in a systems biology perspective. The usefulness and accuracy of pathway analysis critically depend on our understanding of how genes interact with one another. That knowledge is continuously improving due to advances in next generation sequencing technologies and in computational methods. While most approaches treat each of them as independent entities, pathways actually coordinate to perform essential functions in a cell. In this work, we propose a methodology based on a sparse regression approach to find genes that act as intermediary to and interact with two pathways. We model each gene in a pathway using a set of predictor genes, and a connection is formed between the pathway gene and a predictor gene if the sparse regression coefficient corresponding to the predictor gene is non-zero. A predictor gene is a shared neighbor gene of two pathways if it is connected to at least one gene in each pathway. We compare the sparse regression approach to Weighted Correlation Network Analysis and a correlation distance based approach using time-course RNA-Seq data for dendritic cell from wild type, MyD88-knockout, and TRIF-knockout mice, and a set of RNA-Seq data from 60 Caucasian individuals. For the sparse regression approach, we found overrepresented functions for shared neighbor genes between TLR-signaling pathway and antigen processing and presentation, apoptosis, and Jak-Stat pathways that are supported by prior research, and compares favorably to Weighted Correlation Network Analysis in cases where the gene association signals are weak.
Collapse
Affiliation(s)
- Kuo-ching Liang
- Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ashwini Patil
- Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Kenta Nakai
- Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
13
|
Deng Y, Yuan X, Guo XL, Zhu D, Pan YY, Liu HG. Efficacy of atorvastatin on hippocampal neuronal damage caused by chronic intermittent hypoxia: Involving TLR4 and its downstream signaling pathway. Respir Physiol Neurobiol 2015. [PMID: 26200444 DOI: 10.1016/j.resp.2015.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Hippocampal neuronal damage is critical for the initiation and progression of neurocognitive impairment accompanied obstructive sleep apnea syndrome (OSAS). Toll-like receptor 4 (TLR4) plays an important role in the development of several hippocampus-related neural disorders. Atorvastatin was reported beneficially regulates TLR4. Here, we examined the effects of atorvastatin on hippocampal injury caused by chronic intermittent hypoxia (CIH), the most characteristic pathophysiological change of OSAS. Mice were exposed to intermittent hypoxia with or without atorvastatin for 4 weeks. Cell damage, the expressions of TLR4 and its two downstream factors myeloid differentiation factor 88 (MYD88) and TIR-domain-containing adapter-inducing interferon-β (TRIF), inflammatory agents (tumor necrosis factor α and interleukin 1β), and the oxidative stress (superoxide dismutase and malondialdehyde) were determined. Atorvastatin decreased the neural injury and the elevation of TLR4, MyD88, TRIF, pro-inflammatory cytokines and oxidative stress caused by CIH. Our study suggests that atorvastatin may attenuate CIH induced hippocampal neuronal damage partially via TLR4 and its downstream signaling pathway.
Collapse
Affiliation(s)
- Yan Deng
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, No.1095 Jiefang Ave, Wuhan 430030, China
| | - Xiao Yuan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, No.1095 Jiefang Ave, Wuhan 430030, China
| | - Xue-ling Guo
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, No.1095 Jiefang Ave, Wuhan 430030, China
| | - Die Zhu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, No.1095 Jiefang Ave, Wuhan 430030, China
| | - Yue-ying Pan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, No.1095 Jiefang Ave, Wuhan 430030, China
| | - Hui-guo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, No.1095 Jiefang Ave, Wuhan 430030, China.
| |
Collapse
|
14
|
Larochelle A, Bellavance MA, Rivest S. Role of adaptor protein MyD88 in TLR-mediated preconditioning and neuroprotection after acute excitotoxicity. Brain Behav Immun 2015; 46:221-31. [PMID: 25733102 DOI: 10.1016/j.bbi.2015.02.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/03/2015] [Accepted: 02/20/2015] [Indexed: 12/18/2022] Open
Abstract
Excitotoxic cell death is a crucial mechanism through which neurodegeneration occurs in numerous pathologies of the central nervous system (CNS), such as Alzheimer's disease, stroke and spinal cord injury. Toll-like receptors (TLRs) are strongly expressed on microglial cells and are key regulators of the innate immune response to neuronal damage. However, it is still unclear whether their stimulation is protective or harmful in excitotoxic contexts. In this study, we demonstrate that systemic administration of lipopolysaccharide (LPS) or Pam3CSK4 24h prior to an intrastriatal injection of kainic acid (KA) significantly protected cortical neurons in the acute phase of injury. Protection could not be detected with the TLR3 ligand poly-IC. Histological analyses revealed that microglia of LPS and Pam3CSK4 pre-conditioned group were primed to react to injury and exhibited a stronger expression of Tnf and Tlr2 mRNA. We also found that mice deficient for MyD88, a critical adaptor protein for most TLR, were more vulnerable than WT mice to KA-induced excitotoxicity at early (12h and 24h) and late (10days) time points. Finally, bone-marrow chimeric mice revealed that MyD88 signaling in CNS resident cells, but not in cells of hematopoietic origin, mediates the protective effect. This study unravels the potential of TLR2 and TLR4 agonists to induce a protective state of preconditioning against KA-mediated excitotoxicity and further highlights the beneficial role of cerebral MyD88 signaling in this context.
Collapse
Affiliation(s)
- Antoine Larochelle
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada
| | - Marc-André Bellavance
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada.
| |
Collapse
|
15
|
Wang PF, Xiong XY, Chen J, Wang YC, Duan W, Yang QW. Function and mechanism of toll-like receptors in cerebral ischemic tolerance: from preconditioning to treatment. J Neuroinflammation 2015; 12:80. [PMID: 25928750 PMCID: PMC4422156 DOI: 10.1186/s12974-015-0301-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/13/2015] [Indexed: 01/13/2023] Open
Abstract
Increasing evidence suggests that toll-like receptors (TLRs) play an important role in cerebral ischemia-reperfusion injury. The endogenous ligands released from ischemic neurons activate the TLR signaling pathway, resulting in the production of a large number of inflammatory cytokines, thereby causing secondary inflammation damage following cerebral ischemia. However, the preconditioning for minor cerebral ischemia or the preconditioning with TLR ligands can reduce cerebral ischemic injury by regulating the TLR signaling pathway following ischemia in brain tissue (mainly, the inhibition of the TLR4/NF-κB signaling pathway and the enhancement of the interferon regulatory factor-dependent signaling), resulting in TLR ischemic tolerance. Additionally, recent studies found that postconditioning with TLR ligands after cerebral ischemia can also reduce ischemic damage through the regulation of the TLR signaling pathway, showing a significant therapeutic effect against cerebral ischemia. These studies suggest that the ischemic tolerance mediated by TLRs can serve as an important target for the prevention and treatment of cerebral ischemia. On the basis of describing the function and mechanism of TLRs in mediating cerebral ischemic damage, this review focuses on the mechanisms of cerebral ischemic tolerance induced by the preconditioning and postconditioning of TLRs and discusses the clinical application of TLRs for ischemic tolerance.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of Neurology, Xinqiao Hospital & the Second Affiliated Hospital, the Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China. .,Department of Neurology, Weihai municipal Hospital, Weihai, 264200, China.
| | - Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital & the Second Affiliated Hospital, the Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| | - Jing Chen
- Department of Neurology, Xinqiao Hospital & the Second Affiliated Hospital, the Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| | - Yan-Chun Wang
- Department of Neurology, Xinqiao Hospital & the Second Affiliated Hospital, the Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| | - Wei Duan
- Department of Neurology, Xinqiao Hospital & the Second Affiliated Hospital, the Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital & the Second Affiliated Hospital, the Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| |
Collapse
|
16
|
Intravenous immunoglobulin (IVIg) dampens neuronal toll-like receptor-mediated responses in ischemia. J Neuroinflammation 2015; 12:73. [PMID: 25886362 PMCID: PMC4409750 DOI: 10.1186/s12974-015-0294-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 04/02/2015] [Indexed: 11/14/2022] Open
Abstract
Background Ischemic stroke causes a high rate of deaths and permanent neurological damage in survivors. Ischemic stroke triggers the release of damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1), which activate toll-like receptors (TLRs) and receptor for advanced glycation endproducts (RAGE) in the affected area, leading to an exaggerated inflammatory response and cell death. Both TLRs and RAGE are transmembrane pattern recognition receptors (PRRs) that have been shown to contribute to ischemic stroke-induced brain injury. Intravenous immunoglobulin (IVIg) preparations obtained by fractionating human blood plasma are increasingly being used as an effective therapeutic agent in the treatment of several inflammatory diseases. Its use as a potential therapeutic agent for treatment of stroke has been proposed, but little is known about the direct neuroprotective mechanisms of IVIg. We therefore investigate whether IVIg exerts its beneficial effects on the outcome of neuronal injury by modulating HMGB1-induced TLR and RAGE expressions and activations. Methods Primary cortical neurons were subjected to glucose deprivation or oxygen and glucose deprivation conditions and treated with IVIg and recombinant HMGB1. C57/BL6J mice were subjected to middle cerebral artery occlusion, followed by reperfusion, and IVIg was administered intravenously 3 h after the start of reperfusion. Expression of TLRs, RAGE and downstream signalling proteins in neurons and brain tissues were evaluated by immunoblot. Results Treatment of cultured neurons with IVIg reduced simulated ischemia-induced TLR2, TLR4, TLR8 and RAGE expressions, pro-apoptotic caspase-3 cleavage and phosphorylation of the cell death-associated kinases such as c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK) as well as the p65 subunit of nuclear factor kappa B (NF-κB). These results were recapitulated in an in vivo model of stroke. IVIg treatment also upregulated the anti-apoptotic protein B-cell lymphoma 2 (Bcl-2) in cortical neurons under ischemic conditions. Finally, IVIg protected neurons against HMGB1-induced neuronal cell death by modulating TLR and RAGE expressions and signalling pathways. Conclusions Taken together, these results provide a rationale for the potential use of IVIg to target inappropriately activated components of the innate immune system following ischemic stroke.
Collapse
|
17
|
Fadakar K, Dadkhahfar S, Esmaeili A, Rezaei N. The role of Toll-like receptors (TLRs) in stroke. Rev Neurosci 2014; 25:699-712. [PMID: 24807166 DOI: 10.1515/revneuro-2013-0069] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 04/06/2014] [Indexed: 02/05/2023]
Abstract
Toll-like receptors (TLRs) recognizing the exogenous pathogen-associated molecular patterns (PAMPs) are part of the innate immune system that plays a role in various challenging interactions between the neurons and the immune system. Stroke as a major injury to the central nervous system (CNS) is one of the hot points of such cross-talk. The various roles of the different types of TLRs in stroke can be classified into three major categories: (1) the hazardous effect of TLRs with a focus on the part in poststroke neurodegeneration, (2) the beneficial effect of those types of TLRs that exert a neuroprotective effect following an ischemic insult, and (3) the role of TLRs in immunomodulation on one hand and the possible autoimmunity as a consequence of neuronal injury due to an ischemic attack on the other hand. However, the mentioned functions of TLRs, similar to many other parts of the immune system, might overlap in many aspects. The current review article, including both experimental and clinical studies, is an attempt to bring together the studies that have investigated the roles of TLRs in stroke while referring to the apparent controversies in this field, with pointing out the new ideas for further considerations.
Collapse
|
18
|
Zhang X, Ha T, Lu C, Lam F, Liu L, Schweitzer J, Kalbfleisch J, Kao RL, Williams DL, Li C. Poly (I:C) therapy decreases cerebral ischaemia/reperfusion injury via TLR3-mediated prevention of Fas/FADD interaction. J Cell Mol Med 2014; 19:555-65. [PMID: 25351293 PMCID: PMC4369813 DOI: 10.1111/jcmm.12456] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/05/2014] [Indexed: 12/30/2022] Open
Abstract
Toll-like receptor (TLR)-mediated signalling plays a role in cerebral ischaemia/reperfusion (I/R) injury. Modulation of TLRs has been reported to protect against cerebral I/R injury. This study examined whether modulation of TLR3 with poly (I:C) will induce protection against cerebral I/R injury. Mice were treated with or without Poly (I:C) (n = 8/group) 1 hr prior to cerebral ischaemia (60 min.) followed by reperfusion (24 hrs). Poly (I:C) pre-treatment significantly reduced the infarct volume by 57.2% compared with untreated I/R mice. Therapeutic administration of Poly (I:C), administered 30 min. after cerebral ischaemia, markedly decreased infarct volume by 34.9%. However, Poly (I:C)-induced protection was lost in TLR3 knockout mice. In poly (I:C)-treated mice, there was less neuronal damage in the hippocampus compared with untreated I/R mice. Poly (I:C) treatment induced IRF3 phosphorylation, but it inhibited NF-κB activation in the brain. Poly (I:C) also decreased I/R-induced apoptosis by attenuation of Fas/FasL-mediated apoptotic signalling. In addition, Poly (I:C) treatment decreased microglial cell caspase-3 activity. In vitro data showed that Poly (I:C) prevented hypoxia/reoxygenation (H/R)-induced interaction between Fas and FADD as well as caspase-3 and -8 activation in microglial cells. Importantly, Poly (I:C) treatment induced co-association between TLR3 and Fas. Our data suggest that Poly (I:C) decreases in cerebral I/R injury via TLR3 which associates with Fas, thereby preventing the interaction of Fas and FADD, as well as microglial cell caspase-3 and -8 activities. We conclude that TLR3 modulation by Poly (I:C) could be a potential approach for protection against ischaemic stroke.
Collapse
Affiliation(s)
- Xia Zhang
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Famakin BM. The Immune Response to Acute Focal Cerebral Ischemia and Associated Post-stroke Immunodepression: A Focused Review. Aging Dis 2014; 5:307-26. [PMID: 25276490 DOI: 10.14336/ad.2014.0500307] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022] Open
Abstract
It is currently well established that the immune system is activated in response to transient or focal cerebral ischemia. This acute immune activation occurs in response to damage, and injury, to components of the neurovascular unit and is mediated by the innate and adaptive arms of the immune response. The initial immune activation is rapid, occurs via the innate immune response and leads to inflammation. The inflammatory mediators produced during the innate immune response in turn lead to recruitment of inflammatory cells and the production of more inflammatory mediators that result in activation of the adaptive immune response. Under ideal conditions, this inflammation gives way to tissue repair and attempts at regeneration. However, for reasons that are just being understood, immunosuppression occurs following acute stroke leading to post-stroke immunodepression. This review focuses on the current state of knowledge regarding innate and adaptive immune activation in response to focal cerebral ischemia as well as the immunodepression that can occur following stroke. A better understanding of the intricate and complex events that take place following immune response activation, to acute cerebral ischemia, is imperative for the development of effective novel immunomodulatory therapies for the treatment of acute stroke.
Collapse
Affiliation(s)
- Bolanle M Famakin
- National Institutes of Health, National Institute of Neurological Diseases and Stroke, Stroke Branch, Branch, Bethesda, MD, 20892, USA
| |
Collapse
|
20
|
Heuss ND, Pierson MJ, Montaniel KRC, McPherson SW, Lehmann U, Hussong SA, Ferrington DA, Low WC, Gregerson DS. Retinal dendritic cell recruitment, but not function, was inhibited in MyD88 and TRIF deficient mice. J Neuroinflammation 2014; 11:143. [PMID: 25116321 PMCID: PMC4149240 DOI: 10.1186/s12974-014-0143-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 07/29/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Immune system cells are known to affect loss of neurons due to injury or disease. Recruitment of immune cells following retinal/CNS injury has been shown to affect the health and survival of neurons in several models. We detected close, physical contact between dendritic cells and retinal ganglion cells following an optic nerve crush, and sought to understand the underlying mechanisms. METHODS CD11c-DTR/GFP mice producing a chimeric protein of diphtheria toxin receptor (DTR) and GFP from a transgenic CD11c promoter were used in conjunction with mice deficient in MyD88 and/or TRIF. Retinal ganglion cell injury was induced by an optic nerve crush, and the resulting interactions of the GFPhi cells and retinal ganglion cells were examined. RESULTS Recruitment of GFPhi dendritic cells to the retina was significantly compromised in MyD88 and TRIF knockout mice. GFPhi dendritic cells played a significant role in clearing fluorescent-labeled retinal ganglion cells post-injury in the CD11c-DTR/GFP mice. In the TRIF and MyD88 deficient mice, the resting level of GFPhi dendritic cells was lower, and their influx was reduced following the optic nerve crush injury. The reduction in GFPhi dendritic cell numbers led to their replacement in the uptake of fluorescent-labeled debris by GFPlo microglia/macrophages. Depletion of GFPhi dendritic cells by treatment with diphtheria toxin also led to their displacement by GFPlo microglia/macrophages, which then assumed close contact with the injured neurons. CONCLUSIONS The contribution of recruited cells to the injury response was substantial, and regulated by MyD88 and TRIF. However, the presence of these adaptor proteins was not required for interaction with neurons, or the phagocytosis of debris. The data suggested a two-niche model in which resident microglia were maintained at a constant level post-optic nerve crush, while the injury-stimulated recruitment of dendritic cells and macrophages led to their transient appearance in numbers equivalent to or greater than the resident microglia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dale S Gregerson
- Department of Ophthalmology & Visual Neurosciences, University of Minnesota, Lions Research Bldg, Rm 314, 2001 6th St SE, Minneapolis 55455, MN, USA.
| |
Collapse
|
21
|
Famakin BM, Mou Y, Johnson K, Spatz M, Hallenbeck J. A new role for downstream Toll-like receptor signaling in mediating immediate early gene expression during focal cerebral ischemia. J Cereb Blood Flow Metab 2014; 34:258-67. [PMID: 24301291 PMCID: PMC3915199 DOI: 10.1038/jcbfm.2013.182] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/16/2013] [Accepted: 09/20/2013] [Indexed: 12/20/2022]
Abstract
To better understand the role of downstream Toll-like receptor (TLR) signaling during acute cerebral ischemia, we performed cDNA microarrays, on brain RNA, and cytokine arrays, on serum, from wild type (WT), MyD88-/- and TRIF-mutant mice, at baseline and following permanent middle cerebral artery occlusion (pMCAO). The acute stress response pathway was among the top pathways identified by Ingenuity Pathway Analysis of microarray data. We used real-time polymerase chain reaction to confirm the expression of four immediate early genes; EGR1, EGR2, ARC, Nurr77, in this pathway, and insulin degrading enzyme (IDE). Compared to WT, baseline immediate early gene expression was increased up to10-fold in MyD88-/- and TRIF-mutant mice. However, following pMCAO, immediate early gene expression remained unchanged, from this elevated baseline in these mice, but increased up to 12-fold in WT. Furthermore, expression of IDE, which also degrades β-amyloid, decreased significantly only in TRIF-mutant mice. Finally, sE-Selectin, sICAM, sVCAM-1, and MMP-9 levels were significantly decreased only in MyD88-/- compared with WT mice. We thus report a new role for downstream TLR signaling in immediate early gene expression during acute cerebral ischemia. We also show that the TRIF pathway regulates IDE expression; a major enzyme that clears β-amyloid from the brain.
Collapse
Affiliation(s)
- Bolanle M Famakin
- National Institute of Neurological Disorders and Stroke, Stroke Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Yongshan Mou
- National Institute of Neurological Disorders and Stroke, Stroke Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Kory Johnson
- National Institute of Neurological Disorders & Stroke, Section on Bioinformatics, Information Technology & Bioinformatics Program, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria Spatz
- National Institute of Neurological Disorders and Stroke, Stroke Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - John Hallenbeck
- National Institute of Neurological Disorders and Stroke, Stroke Branch, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
22
|
Toll-like receptor-3 activation increases the vulnerability of the neonatal brain to hypoxia-ischemia. J Neurosci 2013; 33:12041-51. [PMID: 23864690 DOI: 10.1523/jneurosci.0673-13.2013] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Susceptibility and progression of brain injury in the newborn is closely associated with an exacerbated innate immune response, but the underlying mechanisms are often unclear. Toll-like receptors (TLRs) are important innate immune sensors that may influence the vulnerability of the developing brain. In the current study, we provide novel data to show that activation of the viral innate immune receptor TLR-3 sensitizes the neonatal brain to subsequent hypoxic-ischemic (HI) damage. Poly inosinic:poly cytidylic acid (Poly I:C), a synthetic ligand for TLR-3, was administered to neonatal mice 14 h before cerebral HI. Activation of TLR-3 before HI increased infarct volume from 3.0 ± 0.5 to 15.4 ± 2.1 mm³ and augmented loss of myelin basic protein from 13.4 ± 6.0 to 70.6 ± 5.3%. The sensitizing effect of Poly I:C was specific for the TLR-3 pathway because mice deficient in the TLR-3 adaptor protein Toll/IL-1R domain-containing adaptor molecule-1 (TRIF) did not develop larger brain damage. The increased vulnerability was associated with a TRIF-dependent heightened inflammatory response, including proinflammatory cytokines, chemokines, and the apoptosis-associated mediator Fas, whereas there was a decrease in reparative M2-like CD11b⁺ microglia and phosphorylation of Akt. Because TLR-3 is activated via double-stranded RNA during most viral infections, the present study provides evidence that viral infections during pregnancy or in the neonate could have great impact on subsequent HI brain injury.
Collapse
|
23
|
Evidence for a gender-specific protective role of innate immune receptors in a model of perinatal brain injury. J Neurosci 2013; 33:11556-72. [PMID: 23843525 DOI: 10.1523/jneurosci.0535-13.2013] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hypoxia-ischemia is a common cause of neurological impairments in newborns, but little is known about how neuroinflammation contributes to the long-term outcome after a perinatal brain injury. In this study, we investigated the role of the fractalkine receptor chemokine CX3C motif receptor 1 (CX3CR1) and of toll-like receptor (TLR) signaling after a neonatal hypoxic-ischemic brain injury. Mice deficient in the TLR adaptor proteins Toll/interleukin-1 receptor-domain-containing adaptor protein inducing interferon β (TRIF) or myeloid differentiation factor-88 (MyD88) and CX3CR1 knock-out (KO) mice were subjected to hypoxia-ischemia at postnatal day 3. In situ hybridization was used to evaluate the expression of TLRs during brain development and after hypoxic-ischemic insults. Behavioral deficits, hippocampal damage, reactive microgliosis, and subplate injury were compared among the groups. Although MyD88 KO mice exhibited no differences from wild-type animals in long-term structural and functional outcomes, TRIF KO mice presented a worse outcome, as evidenced by increased hippocampal CA3 atrophy in males and by the development of learning and motor deficits in females. CX3CR1-deficient female mice showed a marked increase in brain damage and long-lasting learning deficits, whereas CX3CR1 KO male animals did not exhibit more brain injury than wild-type mice. These data reveal a novel, gender-specific protective role of TRIF and CX3CR1 signaling in a mouse model of neonatal hypoxic-ischemic brain injury. These findings suggest that future studies seeking immunomodulatory therapies for preterm infants should consider gender as a critical variable and should be cautious not to abrogate the protective role of neuroinflammation.
Collapse
|
24
|
Cui G, Ye X, Zuo T, Zhao H, Zhao Q, Chen W, Hua F. Chloroquine pretreatment inhibits toll-like receptor 3 signaling after stroke. Neurosci Lett 2013; 548:101-4. [DOI: 10.1016/j.neulet.2013.02.072] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/23/2013] [Accepted: 02/25/2013] [Indexed: 12/28/2022]
|
25
|
Wu D, Lee YCG, Liu HC, Yuan RY, Chiou HY, Hung CH, Hu CJ. Identification of TLR downstream pathways in stroke patients. Clin Biochem 2013; 46:1058-1064. [DOI: 10.1016/j.clinbiochem.2013.05.059] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 05/14/2013] [Indexed: 12/21/2022]
|
26
|
MyD88 is a critical regulator of hematopoietic cell-mediated neuroprotection seen after stroke. PLoS One 2013; 8:e57948. [PMID: 23483951 PMCID: PMC3587572 DOI: 10.1371/journal.pone.0057948] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 01/28/2013] [Indexed: 01/28/2023] Open
Abstract
Neuroinflammation is critical in the neural cell death seen in stroke. It has been shown that CNS and peripheral responses drive this neuroinflammatory response in the brain. The Toll-like receptors (TLRs) are important regulators of inflammation in response to both exogenous and endogenous stressors. Taking advantage of a downstream adapter molecule that controls the majority of TLR signalling, this study investigated the role of the TLR adaptor protein myeloid differentiation factor 88 (MyD88) in the control of CNS and peripheral inflammation. Reversible middle-cerebral artery occlusion was used as the model of stroke in vivo; in vitro primary cultured neurons and glia were subject to four hours of oxygen and glucose deprivation (OGD). Both in vitro and in vivo Myd88−/− animals or cells were compared with wild type (WT). We found that after stroke Myd88−/− animals have a larger infarct volume compared to WT animals. Interestingly, in vitro there was no difference between the survival of Myd88−/− and WT cells following OGD, suggesting that peripheral responses were influencing stroke outcome. We therefore generated bone marrow chimeras and found that Myd88−/− animals have a smaller stroke infarct than their radiation naive counterparts if their hematopoietic cells are WT. Furthermore, WT animals have a larger stroke than their radiation naive counterparts if the hematopoietic cells are Myd88−/−. We have demonstrated that MyD88-dependent signalling in the hematopoietic cell lineage reduces infarct size following stroke and that infiltrating cells to the site of neuroinflammation are neuroprotective following stroke.
Collapse
|
27
|
Shichita T, Sakaguchi R, Ito M, Kondo T, Yoshimura A. Peroxiredoxin triggers cerebral post-ischemic inflammation. Inflamm Regen 2013. [DOI: 10.2492/inflammregen.33.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
28
|
Shichita T, Ago T, Kamouchi M, Kitazono T, Yoshimura A, Ooboshi H. Novel therapeutic strategies targeting innate immune responses and early inflammation after stroke. J Neurochem 2012; 123 Suppl 2:29-38. [PMID: 23050640 DOI: 10.1111/j.1471-4159.2012.07941.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Post-ischemic inflammation is an essential step in the progression of ischemic stroke. This review focuses on the function of infiltrating immune cells, macrophages, and T cells, in ischemic brain injury. The brain is a sterile organ; however, the activation of Toll-like receptor (TLR) 2 and TLR4 is pivotal in the beginning of post-ischemic inflammation. Some endogenous TLR ligands are released from injured brain cells, including high mobility group box 1 and peroxiredoxin family proteins, and activate the infiltrating macrophages and induce the expression of inflammatory cytokines. Following this step, T cells also infiltrate into the ischemic brain and mediate post-ischemic inflammation in the delayed phase. Various cytokines from helper T cells and γδT cells function as neurotoxic (IL-23/IL-17, IFN-γ) or neuroprotective (IL-10, IL-4) mediators. Novel neuroprotective strategies should therefore be developed through more detailed understanding of this process and the regulation of post-ischemic inflammation.
Collapse
Affiliation(s)
- Takashi Shichita
- Department of Microbiology and Immunology, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Pharmacological inhibition of TLR4-NOX4 signal protects against neuronal death in transient focal ischemia. Sci Rep 2012. [PMID: 23193438 PMCID: PMC3508453 DOI: 10.1038/srep00896] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Recent data have shown that TLR4 performs a key role in cerebral ischemia-reperfusion injury which serves as the origin of the immunological inflammatory reactions. However, the therapeutic effects of pharmacological inhibitions of TLR4 and its immediate down-stream pathway remain to be uncovered. In the present study, on mice, intracerebroventricular injection of resatorvid (TLR4 signal inhibitor; 0.01 μg) significantly reduced infarct volume and improved neurological score after middle cerebral artery occlusion and reperfusion. The levels of phospho-p38, nuclear factor-kappa B, and matrix metalloproteinase 9 expressions were significantly suppressed in the resatorvid-treated group. In addition, NOX4 associates with TLR4 after cerebral ischemia-reperfusion seen in mice and human. Genetic and pharmacological inhibitions of TLR4 each reduced NOX4 expression, leading to suppression of oxidative/nitrative stress and of neuronal apoptosis. These data suggest that resatorvid has potential as a therapeutic agent for stroke since it inhibits TLR4-NOX4 signaling which may be the predominant causal pathway.
Collapse
|
30
|
Deng S, Yu K, Zhang B, Yao Y, Liu Y, He H, Zhang H, Cui M, Fu J, Lian Z, Li N. Effects of over-expression of TLR2 in transgenic goats on pathogen clearance and role of up-regulation of lysozyme secretion and infiltration of inflammatory cells. BMC Vet Res 2012; 8:196. [PMID: 23082910 PMCID: PMC3560201 DOI: 10.1186/1746-6148-8-196] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 10/17/2012] [Indexed: 01/07/2023] Open
Abstract
Background Toll-like receptor 2 (TLR2) is important to host recognition of invading gram-positive microbes. In goats, these microbes can cause serious mastitis, anthrax, tetanus, and other problems. Transgenic goats constitutively over-expressing TLR2 in many tissues serve as a suitable model for the study of the role of TLR2 over-expression in bacterial clearance. Results Capra hircus TLR2 over-expression vector (p3S-LoxP-TLR2) was used to generate transgenic goats by egg microinjection. The integration efficiency was 8.57%. Real-time PCR and immunohistochemical results confirmed that the goats over-expressing the TLR2 gene (Tg) expressed more TLR2 than wild-type goats (WT). Monocyte-macrophages from the bloodstreams of transgenic goats were stimulated with synthetic bacterial lipoprotein (Pam3CSK4) and by the promotion of interleukin-6 (IL-6) and IL-10 expression in vitro. The oxidative damage was significantly reduced, and lysozyme (LZM) secretion was found to be up-regulated. Ear tissue samples from transgenic goats that had been stimulated with Pam3CSK4 via hypodermic injection showed that transgenic individuals can undergo the inflammation response very quickly. Conclusions Over-expression of TLR2 was found to decrease radical damage to host cells through low-level production of NO and MDA and to promote the clearance of invasive bacteria by up-regulating lysozyme secretion and filtration of inflammatory cells to the infected site.
Collapse
Affiliation(s)
- Shoulong Deng
- National key Lab of Agro-Biotechnology, China Agricultural University, Beijing 100193, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Mallard C. Innate immune regulation by toll-like receptors in the brain. ISRN NEUROLOGY 2012; 2012:701950. [PMID: 23097717 PMCID: PMC3477747 DOI: 10.5402/2012/701950] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 09/04/2012] [Indexed: 01/29/2023]
Abstract
The innate immune system plays an important role in cerebral health and disease. In recent years the role of innate immune regulation by toll-like receptors in the brain has been highlighted. In this paper the expression of toll-like receptors and endogenous toll-like receptor ligands in the brain and their role in cerebral ischemia will be discussed. Further, the ability of systemic toll-like receptor ligands to induce cerebral inflammation will be reviewed. Finally, the capacity of toll-like receptors to both increase (sensitization) and decrease (preconditioning/tolerance) the vulnerability of the brain to damage will be disclosed. Studies investigating the role of toll-like receptors in the developing brain will be emphasized.
Collapse
Affiliation(s)
- Carina Mallard
- Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 40530 Gothenburg, Sweden
| |
Collapse
|
32
|
Famakin B, Mou Y, Spatz M, Lawal M, Hallenbeck J. Downstream Toll-like receptor signaling mediates adaptor-specific cytokine expression following focal cerebral ischemia. J Neuroinflammation 2012; 9:174. [PMID: 22799573 PMCID: PMC3419098 DOI: 10.1186/1742-2094-9-174] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 07/16/2012] [Indexed: 01/18/2023] Open
Abstract
Background Deletion of some Toll-like receptors (TLRs) affords protection against cerebral ischemia, but disruption of their known major downstream adaptors does not. To determine whether compensation in the production of downstream effectors by one pathway when the other is disrupted can explain these findings, we examined cytokine/chemokine expression and inflammatory infiltrates in wild-type (WT), MyD88−/− and TRIF-mutant mice following permanent middle cerebral artery occlusion (pMCAO). Methods Cytokine/chemokine expression was measured with a 25-plex bead array in the serum and brains of all three groups of mice at baseline (no surgery/naïve) and at 3 hours and 24 hours following pMCAO. Brain inflammatory and neutrophil infiltrates were examined 24 hours following pMCAO. Results IL-6, keratinocyte chemoattractant (KC), granulocyte colony-stimulating factor (G-CSF) and IL-10 were significantly decreased in MyD88−/− mice compared to WT mice following pMCAO. Significantly, decreased levels of the neutrophil chemoattractants KC and G-CSF corresponded with a trend toward fewer neutrophils in the brains of MyD88−/− mice. IP-10 was significantly decreased when either pathway was disrupted. MIP-1α was significantly decreased in TRIF-mutant mice, consistent with TRIF-dependent production. Finally, MyD88−/− mice showed elevations of a number of Th2 cytokines, such as IL-13, at baseline, which became significantly decreased following pMCAO. Conclusions Both MyD88 and TRIF mediate pathway-specific cytokine production following focal cerebral ischemia. Our results also suggest a compensatory Th2-type skew at baseline in MyD88−/− mice and a paradoxical switch to a Th1 phenotype following focal cerebral ischemia. Finally, the MyD88 pathway directs the expression of neutrophil chemoattractants following cerebral ischemia.
Collapse
Affiliation(s)
- Bolanle Famakin
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Building 10, Room 5B06, MSC 1401, Bethesda, MD 20892-1401, USA.
| | | | | | | | | |
Collapse
|
33
|
Shichita T, Sakaguchi R, Suzuki M, Yoshimura A. Post-ischemic inflammation in the brain. Front Immunol 2012; 3:132. [PMID: 22833743 PMCID: PMC3400935 DOI: 10.3389/fimmu.2012.00132] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Accepted: 05/08/2012] [Indexed: 12/25/2022] Open
Abstract
Post-ischemic inflammation is an essential step in the progression of brain ischemia-reperfusion injury. In this review, we focus on the post-ischemic inflammation triggered by infiltrating immune cells, macrophages, and T lymphocytes. Brain ischemia is a sterile organ, but injury-induced inflammation is mostly dependent on Toll-like receptor (TLR) 2 and TLR4. Some endogenous TLR ligands, high mobility group box 1 (HMGB1) and peroxiredoxin family proteins, in particular, are implicated in the activation and inflammatory cytokine expression in infiltrating macrophages. Following macrophage activation, T lymphocytes infiltrate the ischemic brain and regulate the delayed phase inflammation. IL-17-producing γδT lymphocytes induced by IL-23 from macrophages promote ischemic brain injury, whereas regulatory T lymphocytes suppress the function of inflammatory mediators. A deeper understanding of the inflammatory mechanisms of infiltrating immune cells may lead to the development of novel neuroprotective therapies.
Collapse
Affiliation(s)
- Takashi Shichita
- Department of Microbiology and Immunology, School of Medicine, Keio University Tokyo, Japan
| | | | | | | |
Collapse
|
34
|
Kim TH, Yoon SJ, Lee SM. Genipin attenuates sepsis by inhibiting Toll-like receptor signaling. Mol Med 2012; 18:455-65. [PMID: 22252713 DOI: 10.2119/molmed.2011.00308] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 01/10/2012] [Indexed: 01/27/2023] Open
Abstract
The pathogenesis of sepsis is characterized by overwhelming inflammatory responses that lead to tissue damage and organ failure. Toll-like receptor (TLR) signaling is crucial for induction of hyperinflammatory responses and tissue injury during sepsis. Genipin, an aglycon of geniposide, has antiinflammatory and antimicrobial activities. The purpose of this study was to test the hypothesis that genipin reduces multiple organ dysfunction and mortality during sepsis through inhibition of TLR signaling. Male ICR were subjected to sepsis by cecal ligation and puncture (CLP) or endotoxemia by lipopolysaccharide (LPS). Various doses of genipin (1, 2.5 and 5 mg/kg) or a vehicle were administered intravenously immediately after CLP or intraperitoneally after LPS treatment. In another set of survival tests, mice were treated with 2.5 mg/kg of genipin 0 and 24 h after CLP. Genipin was found to improve survival and to attenuate multiple organ dysfunction. Genipin attenuated production of proinflammatory cytokines and release of high-mobility group box 1 (HMGB1). Genipin prevented TLR2 and TLR4, myeloid differentiation factor 88 and the Toll/interleukin-1 receptor domain-containing adaptor protein, inducing interferon-β overexpression. Phosphorylation of mitogen-activated protein kinases and interferon regulatory factor 3 and translocation of nuclear factor (NF)-κB were prevented by genipin. Moreover, genipin attenuated increases in serum tumor necrosis factor-α and HMGB1 in LPS-induced endotoxemia. Pam3CSK4- and LPS-mediated production of nitrites and proinflammatory cytokines was suppressed by genipin in RAW264.7 cells. Genipin attenuated mortality and organ injuries during sepsis through interference with TLR signaling. Therefore, genipin might be useful as a potential therapeutic agent for treatment of sepsis.
Collapse
Affiliation(s)
- Tae-Hoon Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | | | | |
Collapse
|
35
|
Hanke ML, Kielian T. Toll-like receptors in health and disease in the brain: mechanisms and therapeutic potential. Clin Sci (Lond) 2011; 121:367-87. [PMID: 21745188 PMCID: PMC4231819 DOI: 10.1042/cs20110164] [Citation(s) in RCA: 384] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The discovery of mammalian TLRs (Toll-like receptors), first identified in 1997 based on their homology with Drosophila Toll, greatly altered our understanding of how the innate immune system recognizes and responds to diverse microbial pathogens. TLRs are evolutionarily conserved type I transmembrane proteins expressed in both immune and non-immune cells, and are typified by N-terminal leucine-rich repeats and a highly conserved C-terminal domain termed the TIR [Toll/interleukin (IL)-1 receptor] domain. Upon stimulation with their cognate ligands, TLR signalling elicits the production of cytokines, enzymes and other inflammatory mediators that can have an impact on several aspects of CNS (central nervous system) homoeostasis and pathology. For example, TLR signalling plays a crucial role in initiating host defence responses during CNS microbial infection. Furthermore, TLRs are targets for many adjuvants which help shape pathogen-specific adaptive immune responses in addition to triggering innate immunity. Our knowledge of TLR expression and function in the CNS has greatly expanded over the last decade, with new data revealing that TLRs also have an impact on non-infectious CNS diseases/injury. In particular, TLRs recognize a number of endogenous molecules liberated from damaged tissues and, as such, influence inflammatory responses during tissue injury and autoimmunity. In addition, recent studies have implicated TLR involvement during neurogenesis, and learning and memory in the absence of any underlying infectious aetiology. Owing to their presence and immune-regulatory role within the brain, TLRs represent an attractive therapeutic target for numerous CNS disorders and infectious diseases. However, it is clear that TLRs can exert either beneficial or detrimental effects in the CNS, which probably depend on the context of tissue homoeostasis or pathology. Therefore any potential therapeutic manipulation of TLRs will require an understanding of the signals governing specific CNS disorders to achieve tailored therapy.
Collapse
Affiliation(s)
- Mark L. Hanke
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|