51
|
Zhang H, Sun X, Xie Y, Zan J, Tan W. Isosteviol Sodium Protects Against Permanent Cerebral Ischemia Injury in Mice via Inhibition of NF-κB–Mediated Inflammatory and Apoptotic Responses. J Stroke Cerebrovasc Dis 2017; 26:2603-2614. [DOI: 10.1016/j.jstrokecerebrovasdis.2017.06.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 06/01/2017] [Accepted: 06/09/2017] [Indexed: 01/01/2023] Open
|
52
|
Li D, Ni H, Rui Q, Gao R, Chen G. Deletion of Mst1 attenuates neuronal loss and improves neurological impairment in a rat model of traumatic brain injury. Brain Res 2017; 1688:15-21. [PMID: 29054447 DOI: 10.1016/j.brainres.2017.10.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/10/2017] [Accepted: 10/17/2017] [Indexed: 10/18/2022]
Abstract
Neuronal cell death following traumatic brain injury (TBI) is a considerable contributor to neurological deficits. In our work, we explored the functions of Mammalian STE20-like kinase-1 (Mst1), a apoptosis-promoting kinase and also a pivotal bridgebuilder of apoptotic signaling, in the etiopathogenesis of an experimental rat model of TBI. We found that the phosphorylation level of Mst1 in injured area was significantly increased after TBI. Furthermore, we discovered that inhibition of Mst1 phosphorylation can effectively reduce neuronal cell death by inhibiting the activation of caspase 3 and suppressing the damage of DNA during TBI. In addition, the decreased of Mst1 phosphorylation level, not only reduced brain edema and blood-brain barrier (BBB) damage in injured region but also weakened the impairment of neurologic behavior during TBI. In conclusion, our work demonstrates that Mst1 plays an important role in TBI-induced neuronal cell death, suggesting that Mst1 is expected to be a potential therapeutic target for TBI.
Collapse
Affiliation(s)
- Di Li
- Department of Neurosurgery and Translational Medicine Center, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Haibo Ni
- Department of Neurosurgery, The First People 's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Qin Rui
- Clinical Laboratory, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Rong Gao
- Department of Neurosurgery, The First People 's Hospital of Zhangjiagang, Soochow University, Suzhou, China.
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
53
|
Mizuma A, Yenari MA. Anti-Inflammatory Targets for the Treatment of Reperfusion Injury in Stroke. Front Neurol 2017; 8:467. [PMID: 28936196 PMCID: PMC5594066 DOI: 10.3389/fneur.2017.00467] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
While the mainstay of acute stroke treatment includes revascularization via recombinant tissue plasminogen activator or mechanical thrombectomy, only a minority of stroke patients are eligible for treatment, as delayed treatment can lead to worsened outcome. This worsened outcome at the experimental level has been attributed to an entity known as reperfusion injury (R/I). R/I is occurred when revascularization is delayed after critical brain and vascular injury has occurred, so that when oxygenated blood is restored, ischemic damage is increased, rather than decreased. R/I can increase lesion size and also worsen blood barrier breakdown and lead to brain edema and hemorrhage. A major mechanism underlying R/I is that of poststroke inflammation. The poststroke immune response consists of the aberrant activation of glial cell, infiltration of peripheral leukocytes, and the release of damage-associated molecular pattern (DAMP) molecules elaborated by ischemic cells of the brain. Inflammatory mediators involved in this response include cytokines, chemokines, adhesion molecules, and several immune molecule effectors such as matrix metalloproteinases-9, inducible nitric oxide synthase, nitric oxide, and reactive oxygen species. Several experimental studies over the years have characterized these molecules and have shown that their inhibition improves neurological outcome. Yet, numerous clinical studies failed to demonstrate any positive outcomes in stroke patients. However, many of these clinical trials were carried out before the routine use of revascularization therapies. In this review, we cover mechanisms of inflammation involved in R/I, therapeutic targets, and relevant experimental and clinical studies, which might stimulate renewed interest in designing clinical trials to specifically target R/I. We propose that by targeting anti-inflammatory targets in R/I as a combined therapy, it may be possible to further improve outcomes from pharmacological thrombolysis or mechanical thrombectomy.
Collapse
Affiliation(s)
- Atsushi Mizuma
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, CA, United States
| | - Midori A Yenari
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, CA, United States
| |
Collapse
|
54
|
Hyperglycemia in Stroke Impairs Polarization of Monocytes/Macrophages to a Protective Noninflammatory Cell Type. J Neurosci 2017; 36:9313-25. [PMID: 27605608 DOI: 10.1523/jneurosci.0473-16.2016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 07/14/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Hyperglycemia is common in patients with acute stroke, even in those without preexisting diabetes, and denotes a bad outcome. However, the mechanisms underlying the detrimental effects of hyperglycemia are largely unclear. In a mouse model of ischemic stroke, we found that hyperglycemia increased the infarct volume and decreased the number of protective noninflammatory monocytes/macrophages in the ischemic brain. Ablation of peripheral monocytes blocked the detrimental effect of hyperglycemia, suggesting that monocytes are required. In hyperglycemic mice, α-dicarbonyl glucose metabolites, the precursors for advanced glycation end products, were significantly elevated in plasma and ischemic brain tissue. The receptor of advanced glycation end products, AGER (previously known as RAGE), interfered with polarization of macrophages to a noninflammatory phenotype. When Ager was deleted, hyperglycemia did not aggravate ischemic brain damage any longer. Independently of AGER, methylglyoxal reduced the release of endothelial CSF-1 (M-CSF), which stimulates polarization of macrophages to a noninflammatory phenotype in the microenvironment of the ischemic brain. In summary, our study identified α-dicarbonyls and AGER as mediators by which hyperglycemia lowers the number of protective noninflammatory macrophages and consequently increases ischemic brain damage. Modulating the metabolism of α-dicarbonyls or blocking AGER may improve the treatment of stroke patients with hyperglycemia. SIGNIFICANCE STATEMENT Although glucose is the main energy substrate of the brain, hyperglycemia aggravates ischemic brain damage in acute stroke. So far, clinical trials have indicated that insulin treatment provides no solution to this common clinical problem. This study shows, in an experimental stroke model, that hyperglycemia interferes with the polarization of monocytes/macrophages to a protective cell type. Key players are α-dicarbonyls and the receptor for advanced glycation end products (AGER). Deletion of AGER normalized monocyte/macrophage polarization and reversed the detrimental effects of hyperglycemia, suggesting new avenues to treat stroke patients.
Collapse
|
55
|
Zhang YM, Zhang ZM, Guan QL, Liu YQ, Wu ZW, Li JT, Su Y, Yan CL, Luo YL, Qin J, Wang Q, Xie XD. Co-culture with lung cancer A549 cells promotes the proliferation and migration of mesenchymal stem cells derived from bone marrow. Exp Ther Med 2017; 14:2983-2991. [PMID: 28966680 PMCID: PMC5613203 DOI: 10.3892/etm.2017.4909] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 03/17/2017] [Indexed: 02/07/2023] Open
Abstract
The initiation and progression of various types of tumors, such as lung neoplasms, are driven by a population of cells with stem cell properties and their microenvironment. Bone marrow mesenchymal stem cells (BM-MSCs) in long-term in vitro culture may exhibit spontaneous changes in stem cell biological properties, including malignant transformations; however, the molecular mechanisms of this have not been fully elucidated. In the present study, a BM-MSC and lung cancer A549 cell co-culture system was utilized to investigate how the tumor microenvironment may spontaneously change the proliferation, migration and differentiation of BM-MSCs. It was demonstrated that the lung cancer A549 microenvironment is able to induce changes in the cell morphology, proliferation, karyotype, cytoskeleton and migration ability of BM-MSCs in vitro. Compared with the control group BM-MSCs, the expression of Ras, phosphorylated-extracellular regulated protein kinases, nuclear factor-κB, P62 and B-cell lymphoma 2 (Bcl-2) proteins in groups of co-cultured BM-MSCs increased significantly (P<0.05) and the expression of P53, Bcl-2 associated X protein and caspase-3 protein decreased significantly (P<0.05). The mechanisms responsible for the changes observed in BM-MSCs may be related to abnormal expression of related genes in the ERK signaling pathway.
Collapse
Affiliation(s)
- Yue-Mei Zhang
- Department of Ophthalmology, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhi-Ming Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Lanzhou, Gansu 730000, P.R. China
| | - Quan-Lin Guan
- Department of Ophthalmology, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yong-Qi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Lanzhou, Gansu 730000, P.R. China.,Key Laboratory of Dunhuang Medical and Transformation, Ministry of Education of The People's Republic of China, Lanzhou, Gansu 730000, P.R. China
| | - Zhi-Wei Wu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Lanzhou, Gansu 730000, P.R. China.,Department of Basic Medical Sciences, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, P.R. China
| | - Jin-Tian Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Lanzhou, Gansu 730000, P.R. China.,Key Laboratory of Dunhuang Medical and Transformation, Ministry of Education of The People's Republic of China, Lanzhou, Gansu 730000, P.R. China
| | - Yun Su
- Department of Ophthalmology, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Chun-Lu Yan
- Department of Ophthalmology, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Ya-Li Luo
- Department of Ophthalmology, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jie Qin
- Department of Ophthalmology, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Qian Wang
- Department of Ophthalmology, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiao-Dong Xie
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
56
|
Dogbevia GK, Töllner K, Körbelin J, Bröer S, Ridder DA, Grasshoff H, Brandt C, Wenzel J, Straub BK, Trepel M, Löscher W, Schwaninger M. Gene therapy decreases seizures in a model ofIncontinentia pigmenti. Ann Neurol 2017. [DOI: 10.1002/ana.24981] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Godwin K. Dogbevia
- Institute for Experimental and Clinical Pharmacology and Toxicology; University of Lübeck; Lübeck Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine and Center for Systems Neuroscience; Hannover Germany
| | - Jakob Körbelin
- University Medical Center Hamburg-Eppendorf, Hubertus Wald Cancer Center, Department of Oncology and Hematology; Hamburg Germany
| | - Sonja Bröer
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine and Center for Systems Neuroscience; Hannover Germany
| | - Dirk A. Ridder
- Institute of Pathology; University Medical Center Mainz; Mainz Germany
| | - Hanna Grasshoff
- Institute for Experimental and Clinical Pharmacology and Toxicology; University of Lübeck; Lübeck Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine and Center for Systems Neuroscience; Hannover Germany
| | - Jan Wenzel
- Institute for Experimental and Clinical Pharmacology and Toxicology; University of Lübeck; Lübeck Germany
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel; Lübeck Germany
| | - Beate K. Straub
- Institute of Pathology; University Medical Center Mainz; Mainz Germany
| | - Martin Trepel
- University Medical Center Hamburg-Eppendorf, Hubertus Wald Cancer Center, Department of Oncology and Hematology; Hamburg Germany
- Augsburg Medical Center, Department of Hematology and Oncology; Augsburg Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine and Center for Systems Neuroscience; Hannover Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology; University of Lübeck; Lübeck Germany
- DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel; Lübeck Germany
| |
Collapse
|
57
|
Time-lapse imaging of p65 and IκBα translocation kinetics following Ca 2+-induced neuronal injury reveals biphasic translocation kinetics in surviving neurons. Mol Cell Neurosci 2017; 80:148-158. [PMID: 28238890 DOI: 10.1016/j.mcn.2017.02.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 02/01/2017] [Accepted: 02/21/2017] [Indexed: 12/19/2022] Open
Abstract
The transcription factor nuclear factor-κB (NF-κB) regulates neuronal differentiation, plasticity and survival. It is well established that excitatory neurotransmitters such as glutamate control NF-κB activity. Glutamate receptor overactivation is also involved in ischemic- and seizure-induced neuronal injury and neurodegeneration. However, little is known at the single cell-level how NF-κB signaling relates to neuronal survival during excitotoxic injury. We found that silencing of p65/NF-κB delayed N-methyl-d-aspartate (NMDA)-induced excitotoxic injury in hippocampal neurons, suggesting a functional role of p65 in excitotoxicity. Time-lapse imaging of p65 and its inhibitor IκBα using GFP and Cerulean fusion proteins revealed specific patterns of excitotoxic NF-κB activation. Nuclear translocation of p65 began on average 8±3min following 15min of NMDA treatment and was observed in up to two thirds of hippocampal neurons. Nuclear translocation of IκBα preceded that of p65 suggesting independent translocation processes. In surviving neurons, the onset of p65 nuclear export correlated with mitochondrial membrane potential recovery. Dying neurons exhibited persistent nuclear accumulation of p65-eGFP until plasma membrane permeabilization. Our data demonstrate an important role for p65 activation kinetics in neuronal cell death decisions following excitotoxic injury.
Collapse
|
58
|
Lattke M, Reichel SN, Magnutzki A, Abaei A, Rasche V, Walther P, Calado DP, Ferger B, Wirth T, Baumann B. Transient IKK2 activation in astrocytes initiates selective non-cell-autonomous neurodegeneration. Mol Neurodegener 2017; 12:16. [PMID: 28193238 PMCID: PMC5307695 DOI: 10.1186/s13024-017-0157-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 02/09/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neuroinflammation is associated with a wide range of neurodegenerative disorders, however the specific contribution to individual disease pathogenesis and selective neuronal cell death is not well understood. Inflammatory cerebellar ataxias are neurodegenerative diseases occurring in various autoimmune/inflammatory conditions, e.g. paraneoplastic syndromes. However, how inflammatory insults can cause selective cerebellar neurodegeneration in the context of these diseases remains open, and appropriate animal models are lacking. A key regulator of neuroinflammatory processes is the NF-κB signalling pathway, which is activated by the IκB kinase 2 (IKK2) in response to various pathological conditions. Importantly, its activation is sufficient to initiate neuroinflammation on its own. METHODS To investigate the contribution of IKK/NF-κB-mediated neuroinflammation to neurodegeneration, we established conditional mouse models of cerebellar neuroinflammation, which depend either on the tetracycline-regulated expression of IKK2 in astrocytes or Cre-recombination based IKK2 activation in Bergmann glia. RESULTS We demonstrate that IKK2 activation for a limited time interval in astrocytes is sufficient to induce neuroinflammation, astrogliosis and loss of Purkinje neurons, resembling the pathogenesis of inflammatory cerebellar ataxias. We identified IKK2-driven irreversible dysfunction of Bergmann glia as critical pathogenic event resulting in Purkinje cell loss. This was independent of Lipocalin 2, an acute phase protein secreted by reactive astrocytes and well known to mediate neurotoxicity. Instead, downregulation of the glutamate transporters EAAT1 and EAAT2 and ultrastructural alterations suggest an excitotoxic mechanism of Purkinje cell degeneration. CONCLUSIONS Our results suggest a novel pathogenic mechanism how diverse inflammatory insults can cause inflammation/autoimmune-associated cerebellar ataxias. Disease-mediated elevation of danger signals like TLR ligands and inflammatory cytokines in the cerebellum activates IKK2/NF-κB signalling in astrocytes, which as a consequence triggers astrogliosis-like activation of Bergmann glia and subsequent non-cell-autonomous Purkinje cell degeneration. Notably, the identified hit and run mechanism indicates only an early window for therapeutic interventions.
Collapse
Affiliation(s)
- Michael Lattke
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT UK
| | - Stephanie N. Reichel
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Alexander Magnutzki
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Alireza Abaei
- Core Facility Small Animal MRI, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Volker Rasche
- Core Facility Small Animal MRI, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Dinis P. Calado
- Immunity and Cancer Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT UK
| | - Boris Ferger
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397 Biberach an der Riss, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
59
|
Sapkota A, Gaire BP, Cho KS, Jeon SJ, Kwon OW, Jang DS, Kim SY, Ryu JH, Choi JW. Eupatilin exerts neuroprotective effects in mice with transient focal cerebral ischemia by reducing microglial activation. PLoS One 2017; 12:e0171479. [PMID: 28178289 PMCID: PMC5298292 DOI: 10.1371/journal.pone.0171479] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/21/2017] [Indexed: 12/16/2022] Open
Abstract
Microglial activation and its-driven neuroinflammation are characteristic pathogenetic features of neurodiseases, including focal cerebral ischemia. The Artemisia asiatica (Asteraceae) extract and its active component, eupatilin, are well-known to reduce inflammatory responses. But the therapeutic potential of eupatilin against focal cerebral ischemia is not known, along with its anti-inflammatory activities on activated microglia. In this study, we investigated the neuroprotective effect of eupatilin on focal cerebral ischemia through its anti-inflammation, particularly on activated microglia, employing a transient middle cerebral artery occlusion/reperfusion (tMCAO), combined with lipopolysaccharide-stimulated BV2 microglia. Eupatilin exerted anti-inflammatory responses in activated BV2 microglia, in which it reduced secretion of well-known inflammatory markers, including nitrite, IL-6, TNF-α, and PGE2, in a concentration-dependent manner. These observed in vitro effects of eupatilin led to in vivo neuroprotection against focal cerebral ischemia. Oral administration of eupatilin (10 mg/kg) in a therapeutic paradigm significantly reduced brain infarction and improved neurological functions in tMCAO-challenged mice. The same benefit was also observed when eupatilin was given even within 5 hours after MCAO induction. In addition, the neuroprotective effects of a single administration of eupatilin (10 mg/kg) immediately after tMCAO challenge persisted up to 3 days after tMCAO. Eupatilin administration reduced the number of Iba1-immunopositive cells across ischemic brain and induced their morphological changes from amoeboid into ramified in the ischemic core, which was accompanied with reduced microglial proliferation in ischemic brain. Eupatilin suppressed NF-κB signaling activities in ischemic brain by reducing IKKα/β phosphorylation, IκBα phosphorylation, and IκBα degradation. Overall, these data indicate that eupatilin is a neuroprotective agent against focal cerebral ischemia through the reduction of microglial activation.
Collapse
Affiliation(s)
- Arjun Sapkota
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, Republic of Korea
| | - Bhakta Prasad Gaire
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, Republic of Korea
| | - Kyu Suk Cho
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Se Jin Jeon
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Oh Wook Kwon
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, Republic of Korea
| | - Dae Sik Jang
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, Republic of Korea
| | - Jong Hoon Ryu
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Ji Woong Choi
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, Republic of Korea
- * E-mail:
| |
Collapse
|
60
|
Müller K, Courtois G, Ursini MV, Schwaninger M. New Insight Into the Pathogenesis of Cerebral Small-Vessel Diseases. Stroke 2017; 48:520-527. [PMID: 28082670 DOI: 10.1161/strokeaha.116.012888] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Kristin Müller
- From the Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany (K.M., M.S.); INSERM U1038/BIG, CEA, Grenoble, France (G.C.); and Institute of Genetics and Biophysics, "Adriano Buzzati-Traverso", IGB-CNR, Naples, Italy (M.V.U.)
| | - Gilles Courtois
- From the Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany (K.M., M.S.); INSERM U1038/BIG, CEA, Grenoble, France (G.C.); and Institute of Genetics and Biophysics, "Adriano Buzzati-Traverso", IGB-CNR, Naples, Italy (M.V.U.)
| | - Matilde Valeria Ursini
- From the Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany (K.M., M.S.); INSERM U1038/BIG, CEA, Grenoble, France (G.C.); and Institute of Genetics and Biophysics, "Adriano Buzzati-Traverso", IGB-CNR, Naples, Italy (M.V.U.)
| | - Markus Schwaninger
- From the Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany (K.M., M.S.); INSERM U1038/BIG, CEA, Grenoble, France (G.C.); and Institute of Genetics and Biophysics, "Adriano Buzzati-Traverso", IGB-CNR, Naples, Italy (M.V.U.).
| |
Collapse
|
61
|
Xie Z, Han P, Cui Z, Wang B, Zhong Z, Sun Y, Yang G, Sun Q, Bian L. Pretreatment of Mouse Neural Stem Cells with Carbon Monoxide-Releasing Molecule-2 Interferes with NF-κB p65 Signaling and Suppresses Iron Overload-Induced Apoptosis. Cell Mol Neurobiol 2016; 36:1343-1351. [PMID: 26961543 PMCID: PMC11482508 DOI: 10.1007/s10571-016-0333-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/22/2016] [Indexed: 12/27/2022]
Abstract
Neural stem cell (NSC) transplantation is a promising approach to repair the damaged brain after hemorrhagic stroke; however, it is largely limited by the poor survival of donor cells. Breakdown products of the hematoma and subsequent iron overload contribute to the impairment of survival of neural cells. There is little information regarding the mechanism involved in the death of grafted cells. Furthermore, therapeutic research targeted to improving the survival of grafted neural stem cells (NSCs) is strikingly lacking. Here, we showed that iron overload induced apoptosis of C17.2 cells, a cell line originally cloned from mouse NSCs and immortalized by v-myc. Pretreatment with carbon monoxide-releasing molecule-2 (CORM-2) markedly protected C17.2 cells against iron overload in a dose-dependent manner. Moreover, CORM-2 interfered with NF-κB signaling, including inhibition of nuclear translocation and down-regulation of NF-κB p65. TUNEL staining showed that preconditioning C17.2 cells with CORM-2 enhanced their resistance to apoptosis induced by iron overload, which was concomitant with down-regulation of the pro-apoptotic proteins (Bax and cleaved caspase-3) and up-regulation of the anti-apoptotic protein Bcl2. The protective effect of CORM-2 could be simulated by BAY11-7082, a special inhibitor of NF-κB p65. These results provide a novel and effective strategy to enhance the survival of NSCs after transplantation and, therefore, their efficacy in repairing brain injury due to hemorrhagic stroke.
Collapse
Affiliation(s)
- Zhengxing Xie
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Ping Han
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Zhenwen Cui
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Baofeng Wang
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Zhihong Zhong
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yuhao Sun
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Guoyuan Yang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Qingfang Sun
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
- Department of Neurosurgery, Luwan Branch of Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Liuguan Bian
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
62
|
Gushchina SV, Balashov VP, Magoulas KB. Methodological Approaches to In Vitro Evaluation of Transcription Activity of Nuclear Factor Kappa B (NF-κB) in Sensory Neurons. Bull Exp Biol Med 2016; 161:736-740. [PMID: 27704345 DOI: 10.1007/s10517-016-3497-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Indexed: 10/20/2022]
Abstract
Transcription activity of NF-κB in sensory neurons was analyzed in vitro using classical immunocytochemical methods and transgenic technologies. Activation of NF-κB in NIH3T3 cells and in murine sensory neurons after in vitro stimulation with TNF-α was demonstrated by the immunocytochemical method; however, the expression of the reporter NF-κB/LacZ transgene was detected only after addition of histon deacetylase inhibitor. Hence, formally contradictory conclusions from the results of immunocytochemical analysis and reporter transgene expression were in line with the hypothesis on epigenetic repression of NF-κB activity in sensory neurons mediated by histon deacetylases.
Collapse
Affiliation(s)
- S V Gushchina
- Department of Histology, Cytology, and Embryology, N. P. Ogarev Mordovia State University, Saransk, Republic of Mordovia, Russia. .,Molecular Neuroscience Group, Centre for Neuroscience and Trauma, Queen Mary University of London, London, UK.
| | - V P Balashov
- Department of Histology, Cytology, and Embryology, N. P. Ogarev Mordovia State University, Saransk, Republic of Mordovia, Russia
| | - K B Magoulas
- School of Science and Technology, Middlesex University, London, UK
| |
Collapse
|
63
|
Zhan J, Qin W, Zhang Y, Jiang J, Ma H, Li Q, Luo Y. Upregulation of neuronal zinc finger protein A20 expression is required for electroacupuncture to attenuate the cerebral inflammatory injury mediated by the nuclear factor-kB signaling pathway in cerebral ischemia/reperfusion rats. J Neuroinflammation 2016; 13:258. [PMID: 27716383 PMCID: PMC5048665 DOI: 10.1186/s12974-016-0731-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/23/2016] [Indexed: 02/16/2023] Open
Abstract
Background Zinc finger protein A20 (tumor necrosis factor alpha-induced protein 3) functions as a potent negative feedback inhibitor of the nuclear factor-kB (NF-kB) signaling. It exerts these effects by interrupting the activation of IkB kinase beta (IKKβ), the most critical kinase in upstream of NF-kB, and thereby controlling inflammatory homeostasis. We reported previously that electroacupuncture (EA) could effectively suppress IKKβ activation. However, the mechanism underlying these effects was unclear. Therefore, the current study further explored the effects of EA on A20 expression in rat brain and investigated the possible mechanism of A20 in anti-neuroinflammation mediated by EA using transient middle cerebral artery occlusion (MCAO) rats. Methods Rats were treated with EA at the “Baihui (GV20),” “Hegu (L14),” and “Taichong (Liv3)” acupoints once a day starting 2 h after focal cerebral ischemia. The spatiotemporal expression of A20, neurobehavioral scores, infarction volumes, cytokine levels, glial cell activation, and the NF-kB signaling were assessed at the indicated time points. A20 gene interference (overexpression and silencing) was used to investigate the role of A20 in mediating the neuroprotective effects of EA and in regulating the interaction between neuronal and glial cells by suppressing neuronal NF-kB signaling during cerebral ischemia/reperfusion-induced neuroinflammation. Results EA treatment increased A20 expression with an earlier peak and longer lasting upregulation. The upregulated A20 protein was predominantly located in neurons in the cortical zone of the ischemia/reperfusion. Furthermore, neuronal A20 cell counts were positively correlated with neurobehavioral scores but negatively correlated with infarct volume, the accumulation of pro-inflammatory cytokines, and glial cell activation. Moreover, the effects of EA on improving the neurological outcome and suppressing neuroinflammation in the brain were reversed by A20 silencing. Finally, A20 silencing also suppressed the ability of EA to inhibit neuronal NF-kB signaling pathway. Conclusions Ischemia/reperfusion cortical neurons in MCAO rats are the main cell types that express A20, and there is a correlation between A20 expression and the suppression of neuroinflammation and the resulting neuroprotective effects. EA upregulated neuronal A20 expression, which played an essential role in the anti-inflammatory effects of EA by suppressing the neuronal NF-kB signaling pathway in the brains of MCAO rats.
Collapse
Affiliation(s)
- Jian Zhan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Chongqing Key Laboratory of Neurology, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.,Department of Neurology, The Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou Province, 563000, China
| | - Wenyi Qin
- Department of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ying Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Chongqing Key Laboratory of Neurology, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Jing Jiang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Chongqing Key Laboratory of Neurology, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Hongmei Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Chongqing Key Laboratory of Neurology, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Qiongli Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Chongqing Key Laboratory of Neurology, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Yong Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China. .,Chongqing Key Laboratory of Neurology, 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
64
|
Zhao S, Yin J, Zhou L, Yan F, He Q, Huang L, Peng S, Jia J, Cheng J, Chen H, Tao W, Ji X, Xu Y, Yuan Z. Hippo/MST1 signaling mediates microglial activation following acute cerebral ischemia-reperfusion injury. Brain Behav Immun 2016; 55:236-248. [PMID: 26721416 DOI: 10.1016/j.bbi.2015.12.016] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 12/18/2015] [Accepted: 12/21/2015] [Indexed: 01/16/2023] Open
Abstract
Cerebral ischemia-reperfusion injury is a major public health concern that causes high rates of disability and mortality in adults. Microglial activation plays a crucial role in ischemic stroke-induced alteration of the immune microenvironment. However, the mechanism underlying the triggering of microglial activation by ischemic stroke remains to be elucidated. Previously, we demonstrated that the protein kinase Hippo/MST1 plays an important role in oxidative stress-induced cell death in mammalian primary neurons and that the protein kinase c-Abl phosphorylates MST1 at Y433, which increases MST1 kinase activity. Microglial activation has been implicated as a secondary detrimental cellular response that contributes to neuronal cell death in ischemic stroke. Here, we are the first, to our knowledge, to demonstrate that MST1 mediates stroke-induced microglial activation by directly phosphorylating IκBα at residues S32 and S36. We further demonstrate that Src kinase functions upstream of MST1-IκB signaling during microglial activation. Specific deletion of MST1 in microglia mitigates stroke-induced brain injury. Therefore, we propose that Src-MST1-IκB signaling plays a critical role in stroke-induced microglial activation. Together with our previous work demonstrating that MST1 is important for oxidative stress-induced neuronal cell death, our results indicate that MST1 could represent a potent therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Siqi Zhao
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jie Yin
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, Graduate School of the Chinese Academy of Sciences, Beijing 100049, China
| | - Lujun Zhou
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, Graduate School of the Chinese Academy of Sciences, Beijing 100049, China
| | - Feng Yan
- The Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Qing He
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, Graduate School of the Chinese Academy of Sciences, Beijing 100049, China
| | - Li Huang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shengyi Peng
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Junying Jia
- Core Facility Center, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jinbo Cheng
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hong Chen
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wufan Tao
- School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xunming Ji
- The Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yun Xu
- Department of Neurology and Radiology, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Zengqiang Yuan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
65
|
Li Z, Hua C, Pan X, Fu X, Wu W. Carvacrol Exerts Neuroprotective Effects Via Suppression of the Inflammatory Response in Middle Cerebral Artery Occlusion Rats. Inflammation 2016; 39:1566-72. [DOI: 10.1007/s10753-016-0392-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
66
|
Kawabori M, Yenari MA. Inflammatory responses in brain ischemia. Curr Med Chem 2016; 22:1258-77. [PMID: 25666795 DOI: 10.2174/0929867322666150209154036] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/02/2014] [Accepted: 02/02/2015] [Indexed: 12/20/2022]
Abstract
Brain infarction causes tissue death by ischemia due to occlusion of the cerebral vessels and recent work has shown that post stroke inflammation contributes significantly to the development of ischemic pathology. Because secondary damage by brain inflammation may have a longer therapeutic time window compared to the rescue of primary damage following arterial occlusion, controlling inflammation would be an obvious therapeutic target. A substantial amount of experimentall progress in this area has been made in recent years. However, it is difficult to elucidate the precise mechanisms of the inflammatory responses following ischemic stroke because inflammation is a complex series of interactions between inflammatory cells and molecules, all of which could be either detrimental or beneficial. We review recent advances in neuroinflammation and the modulation of inflammatory signaling pathways in brain ischemia. Potential targets for treatment of ischemic stroke will also be covered. The roles of the immune system and brain damage versus repair will help to clarify how immune modulation may treat stroke.
Collapse
Affiliation(s)
| | - Midori A Yenari
- Dept. of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA.
| |
Collapse
|
67
|
Transcriptional Control of Synaptic Plasticity by Transcription Factor NF-κB. Neural Plast 2016; 2016:7027949. [PMID: 26881128 PMCID: PMC4736603 DOI: 10.1155/2016/7027949] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 11/04/2015] [Indexed: 01/09/2023] Open
Abstract
Activation of nuclear factor kappa B (NF-κB) transcription factors is required for the induction of synaptic plasticity and memory formation. All components of this signaling pathway are localized at synapses, and transcriptionally active NF-κB dimers move to the nucleus to translate synaptic signals into altered gene expression. Neuron-specific inhibition results in altered connectivity of excitatory and inhibitory synapses and functionally in selective learning deficits. Recent research on transgenic mice with impaired or hyperactivated NF-κB gave important insights into plasticity-related target gene expression that is regulated by NF-κB. In this minireview, we update the available data on the role of this transcription factor for learning and memory formation and comment on cross-sectional activation of NF-κB in the aged and diseased brain that may directly or indirectly affect κB-dependent transcription of synaptic genes.
Collapse
|
68
|
Lammerding L, Slowik A, Johann S, Beyer C, Zendedel A. Poststroke Inflammasome Expression and Regulation in the Peri-Infarct Area by Gonadal Steroids after Transient Focal Ischemia in the Rat Brain. Neuroendocrinology 2016; 103:460-75. [PMID: 26337121 DOI: 10.1159/000439435] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 08/16/2015] [Indexed: 11/19/2022]
Abstract
CNS ischemia results in locally confined and rapid tissue damage accompanied by a loss of neurons and their circuits. Early and time-delayed inflammatory responses are critical variables determining the extent of neural disintegration and regeneration. Inflammasomes are vital effectors in innate immunity. Their activation in brain-intrinsic immune cells contributes to ischemia-related brain damage. The steroids 17β-estradiol (E2) and progesterone (P) are neuroprotective and anti-inflammatory. Using a transient focal rat ischemic model, we evaluated the time response of different inflammasomes in the peri-infarct zone from the early to late phases after poststroke ischemia. We show that the different inflammasome complexes reveal a specific time-oriented sequential expression pattern with a maximum at approximately 24 h after the infarct. Within the limits of antibody availability, immunofluorescence labeling demonstrated that microglia and neurons are major sources of the locally activated inflammasomes NOD-like receptor protein-3 (NLRP3) and associated speck-like protein (ASC), respectively. E2 and P given for 24 h immediately after ischemia onset reduced hypoxia-induced mRNA expression of the inflammasomes NLRC4, AIM2 and ASC, and decreased the protein levels of ASC and NLRP3. In addition, mRNA protein levels of the cytokines interleukin-1β (IL1β), IL18 and TNFα were reduced by the steroids. The findings provide for the first time a detailed flow chart of hypoxia-driven inflammasome regulation in the peri-infarct cerebral cortex. Further, we demonstrate that E2 and P alleviate the expression of certain inflammasome components, sometimes in a hormone-specific way. Besides directly regulating other cellular neuroprotective pathways, the control of inflammasomes by these steroids might contribute to its neuroprotective potency.
Collapse
|
69
|
Anrather J, Iadecola C, Hallenbeck J. Inflammation and Immune Response. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
70
|
Plasma L5 levels are elevated in ischemic stroke patients and enhance platelet aggregation. Blood 2015; 127:1336-45. [PMID: 26679863 DOI: 10.1182/blood-2015-05-646117] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 12/02/2015] [Indexed: 12/30/2022] Open
Abstract
L5, the most electronegative and atherogenic subfraction of low-density lipoprotein (LDL), induces platelet activation. We hypothesized that plasma L5 levels are increased in acute ischemic stroke patients and examined whether lectin-like oxidized LDL receptor-1 (LOX-1), the receptor for L5 on endothelial cells and platelets, plays a critical role in stroke. Because amyloid β (Aβ) stimulates platelet aggregation, we studied whether L5 and Aβ function synergistically to induce prothrombotic pathways leading to stroke. Levels of plasma L5, serum Aβ, and platelet LOX-1 expression were significantly higher in acute ischemic stroke patients than in controls without metabolic syndrome (P < .01). In mice subjected to focal cerebral ischemia, L5 treatment resulted in larger infarction volumes than did phosphate-buffered saline treatment. Deficiency or neutralizing of LOX-1 reduced infarct volume up to threefold after focal cerebral ischemia in mice, illustrating the importance of LOX-1 in stroke injury. In human platelets, L5 but not L1 (the least electronegative LDL subfraction) induced Aβ release via IκB kinase 2 (IKK2). Furthermore, L5+Aβ synergistically induced glycoprotein IIb/IIIa receptor activation; phosphorylation of IKK2, IκBα, p65, and c-Jun N-terminal kinase 1; and platelet aggregation. These effects were blocked by inhibiting IKK2, LOX-1, or nuclear factor-κB (NF-κB). Injecting L5+Aβ shortened tail-bleeding time by 50% (n = 12; P < .05 vs L1-injected mice), which was prevented by the IKK2 inhibitor. Our findings suggest that, through LOX-1, atherogenic L5 potentiates Aβ-mediated platelet activation, platelet aggregation, and hemostasis via IKK2/NF-κB signaling. L5 elevation may be a risk factor for cerebral atherothrombosis, and downregulating LOX-1 and inhibiting IKK2 may be novel antithrombotic strategies.
Collapse
|
71
|
Kaltschmidt B, Kaltschmidt C. NF-KappaB in Long-Term Memory and Structural Plasticity in the Adult Mammalian Brain. Front Mol Neurosci 2015; 8:69. [PMID: 26635522 PMCID: PMC4656838 DOI: 10.3389/fnmol.2015.00069] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/30/2015] [Indexed: 11/13/2022] Open
Abstract
The transcription factor nuclear factor kappaB (NF-κB) is a well-known regulator of inflammation, stress, and immune responses as well as cell survival. In the nervous system, NF-κB is one of the crucial components in the molecular switch that converts short- to long-term memory-a process that requires de novo gene expression. Here, the researches published on NF-κB and downstream target genes in mammals will be reviewed, which are necessary for structural plasticity and long-term memory, both under normal and pathological conditions in the brain. Genetic evidence has revealed that NF-κB regulates neuroprotection, neuronal transmission, and long-term memory. In addition, after genetic ablation of all NF-κB subunits, a severe defect in hippocampal adult neurogenesis was observed during aging. Proliferation of neural precursors is increased; however, axon outgrowth, synaptogenesis, and tissue homeostasis of the dentate gyrus are hampered. In this process, the NF-κB target gene PKAcat and other downstream target genes such as Igf2 are critically involved. Therefore, NF-κB activity seems to be crucial in regulating structural plasticity and replenishment of granule cells within the hippocampus throughout the life. In addition to the function of NF-κB in neurons, we will discuss on a neuroinflammatory role of the transcription factor in glia. Finally, a model for NF-κB homeostasis on the molecular level is presented, in order to explain seemingly the contradictory, the friend or foe, role of NF-κB in the nervous system.
Collapse
|
72
|
Rolova T, Dhungana H, Korhonen P, Valonen P, Kolosowska N, Konttinen H, Kanninen K, Tanila H, Malm T, Koistinaho J. Deletion of Nuclear Factor kappa B p50 Subunit Decreases Inflammatory Response and Mildly Protects Neurons from Transient Forebrain Ischemia-induced Damage. Aging Dis 2015; 7:450-65. [PMID: 27493832 DOI: 10.14336/ad.2015.1123] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 11/23/2015] [Indexed: 12/16/2022] Open
Abstract
Transient forebrain ischemia induces delayed death of the hippocampal pyramidal neurons, particularly in the CA2 and medial CA1 area. Early pharmacological inhibition of inflammatory response can ameliorate neuronal death, but it also inhibits processes leading to tissue regeneration. Therefore, research efforts are now directed to modulation of post-ischemic inflammation, with the aim to promote beneficial effects of inflammation and limit adverse effects. Transcription factor NF-κB plays a key role in the inflammation and cell survival/apoptosis pathways. In the brain, NF-κB is predominantly found in the form of a heterodimer of p65 (RelA) and p50 subunit, where p65 has a transactivation domain while p50 is chiefly involved in DNA binding. In this study, we subjected middle-aged Nfkb1 knockout mice (lacking p50 subunit) and wild-type controls of both sexs to 17 min of transient forebrain ischemia and assessed mouse performance in a panel of behavioral tests after two weeks of post-operative recovery. We found that ischemia failed to induce clear memory and motor deficits, but affected spontaneous locomotion in genotype- and sex-specific way. We also show that both the lack of the NF-κB p50 subunit and female sex independently protected CA2 hippocampal neurons from ischemia-induced cell death. Additionally, the NF-κB p50 subunit deficiency significantly reduced ischemia-induced microgliosis, astrogliosis, and neurogenesis. Lower levels of hippocampal microgliosis significantly correlated with faster spatial learning. We conclude that NF-κB regulates the outcome of transient forebrain ischemia in middle-aged subjects in a sex-specific way, having an impact not only on neuronal death but also specific inflammatory responses and neurogenesis.
Collapse
Affiliation(s)
- Taisia Rolova
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Hiramani Dhungana
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Paula Korhonen
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Piia Valonen
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Natalia Kolosowska
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Henna Konttinen
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Katja Kanninen
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Heikki Tanila
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland; 2Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Tarja Malm
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Jari Koistinaho
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| |
Collapse
|
73
|
Protective features of peripheral monocytes/macrophages in stroke. Biochim Biophys Acta Mol Basis Dis 2015; 1862:329-38. [PMID: 26584587 DOI: 10.1016/j.bbadis.2015.11.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 11/02/2015] [Accepted: 11/04/2015] [Indexed: 12/13/2022]
Abstract
Hematogenous recruitment of monocytes and macrophages has traditionally been viewed as a harmful process causing exacerbation of brain injury after stroke. However, emerging findings suggest equally important protective features. Inflammatory monocytes are rapidly recruited to ischemic brain via a CCR2-dependent pathway and undergo secondary differentiation in the target tissue towards non-inflammatory macrophages, mediating neuroprotection and repair of the ischemic neurovascular unit. In contrast, independent recruitment of non-inflammatory monocytes via CX3CR1 does not occur. Thus, protective features of hematogenous macrophages mainly depend on initial CCR2-dependent cell recruitment. Under therapeutic considerations, specific modulation of monocyte-derived macrophages will therefore be more appropriate than non-selectively blocking their hematogenous recruitment. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
|
74
|
Kraut B, Maier HJ, Kókai E, Fiedler K, Boettger T, Illing A, Kostin S, Walther P, Braun T, Wirth T. Cardiac-Specific Activation of IKK2 Leads to Defects in Heart Development and Embryonic Lethality. PLoS One 2015; 10:e0141591. [PMID: 26539991 PMCID: PMC4634958 DOI: 10.1371/journal.pone.0141591] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/09/2015] [Indexed: 11/24/2022] Open
Abstract
The transcription factor NF-κB has been associated with a range of pathological conditions of the heart, mainly based on its function as a master regulator of inflammation and pro-survival factor. Here, we addressed the question what effects activation of NF-κB can have during murine heart development. We expressed a constitutively active (CA) mutant of IKK2, the kinase activating canonical NF-κB signaling, specifically in cardiomyocytes under the control of the α-myosin heavy chain promoter. Expression of IKK2-CA resulted in embryonic lethality around E13. Embryos showed defects in compact zone formation and the contractile apparatus, and overall were characterized by widespread inflammation with infiltration of myeloid cells. Gene expression analysis suggested an interferon type I signature, with increased expression of interferon regulatory factors. While apoptosis of cardiomyocytes was only increased at later stages, their proliferation was decreased early on, providing an explanation for the disturbed compact zone formation. Mechanistically, this could be explained by activation of the JAK/STAT axis and increased expression of the cell cycle inhibitor p21. A rescue experiment with an IκBα superrepressor demonstrated that the phenotype was dependent on NF-κB. We conclude that activation of NF-κB is detrimental during normal heart development due to excessive activation of pro-inflammatory pathways.
Collapse
Affiliation(s)
- Bärbel Kraut
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Harald J. Maier
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Enikö Kókai
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Katja Fiedler
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Thomas Boettger
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Annett Illing
- Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Sawa Kostin
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Paul Walther
- Core Facility Electron Microscopy, University of Ulm, Ulm, Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
75
|
Qi J, Han X, Liu HT, Chen T, Zhang JL, Yang P, Bo SH, Lu XT, Zhang J. 17-Dimethylaminoethylamino-17-demethoxygeldanamycin attenuates inflammatory responses in experimental stroke. Biol Pharm Bull 2015; 37:1713-8. [PMID: 25366476 DOI: 10.1248/bpb.b14-00208] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heat shock protein 90 (HSP90) is a ubiquitous molecular chaperone involved in the proper conformation of many proteins. HSP90 inhibitors (17-dimethyl aminoethylamino-17-demethoxygeldanamycin hydrochloride [17-DMAG]) bind to and inactivate HSP90, suppressing some key signaling pathways involved in the inflammatory process. Since considerable evidence suggests that inflammation accounts for the progression of cerebral ischemic injury, we investigated whether 17-DMAG can modulate inflammatory responses in middle cerebral artery occluded (MCAO) mice. Male C57/BL6 mice were pretreated with 17-DMAG or vehicle for 7 d before being subjected to transient occlusion of middle cerebral artery and reperfusion. Mice were evaluated at 24 h after MCAO for neurological deficit scoring. Moreover, the mechanism of the anti-inflammatory effect of 17-DMAG was investigated with a focus on nuclear factor kappa B (NF-κB) pathway. 17-DMAG significantly reduced cerebral infarction and improved neurological outcome. 17-DMAG suppressed activation of microglia and decreased phosphorylation of inhibitory (I)κB and subsequent nuclear translocation of p65, which eventually downregulated expression of NF-κB-regulated genes. These results suggest that 17-DMAG has a promising therapeutic effect in ischemic stroke treatment through an anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Jia Qi
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Iwamoto T, Ouchi Y. Emerging evidence of insulin-like growth factor 2 as a memory enhancer: a unique animal model of cognitive dysfunction with impaired adult neurogenesis. Rev Neurosci 2015; 25:559-74. [PMID: 24778346 DOI: 10.1515/revneuro-2014-0010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/28/2014] [Indexed: 12/29/2022]
Abstract
In the current aging society, cognitive dysfunction is one of the most serious issues that should be urgently resolved. It also affects a wide range of age groups harboring neurological and psychiatric disorders, such as Alzheimer's disease and schizophrenia. Although the molecular mechanism of memory impairment still remains to be determined, neuronal loss and dysfunction has been revealed to mainly attribute to its pathology. The discovery of neural stem cells in the adult brain that are proliferating and able to generate functional neurons has given rise to the idea that neuronal loss could be rescued by manipulating endogenous neural progenitor and stem cells. To this end, we must characterize them in detail and their developmental programming must be better understood. A growing body of evidence has indicated that insulin-like peptides are involved in learning and memory and maintenance of neural progenitor and stem cells, and clinical trials of insulin as a memory enhancer have begun. In contrast to the expectation of insulin and IGF1, the roles of IGF2 in cognitive ability have been poorly understood. However, recent evidence demonstrated in rodents suggests that IGF2 may play a pivotal role in adult neurogenesis and cognitive function. Here, we would like to review the rapidly growing world of IGF2 in cognitive neuroscience and introduce the evidence that its deficit is indeed involved in the impairment of the hippocampal neurogenesis and cognitive dysfunction in the model mouse of 22q11.2 deletion syndrome, which deletes Dgcr8, a critical gene for microRNA processing.
Collapse
|
77
|
Lanzillotta A, Porrini V, Bellucci A, Benarese M, Branca C, Parrella E, Spano PF, Pizzi M. NF-κB in Innate Neuroprotection and Age-Related Neurodegenerative Diseases. Front Neurol 2015; 6:98. [PMID: 26042083 PMCID: PMC4438602 DOI: 10.3389/fneur.2015.00098] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/21/2015] [Indexed: 12/20/2022] Open
Abstract
NF-κB factors are cardinal transcriptional regulators of inflammation and apoptosis, involved in the brain programing of systemic aging and in brain damage. The composition of NF-κB active dimers and epigenetic mechanisms modulating histone acetylation, finely condition neuronal resilience to brain insults. In stroke models, the activation of NF-κB/c-Rel promotes neuroprotective effects by transcription of specific anti-apoptotic genes. Conversely, aberrant activation of NF-κB/RelA showing reduced level of total acetylation, but site-specific acetylation on lysine 310, triggers the expression of pro-apoptotic genes. Constitutive knockout of c-Rel shatters the resilience of substantia nigra (SN) dopaminergic (DA) neurons to aging and induces a parkinsonian like pathology in mice. c-rel(-/-) mice show increased level of aberrantly acetylated RelA in the basal ganglia, neuroinflammation, accumulation of alpha-synuclein, and iron. Moreover, they develop motor deficits responsive to l-DOPA treatment and associated with loss of DA neurons in the SN. Here, we discuss the effect of unbalanced activation of RelA and c-Rel during aging and propose novel challenges for the development of therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Annamaria Lanzillotta
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Vanessa Porrini
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
- IRCCS, San Camillo Hospital, Venice, Italy
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Marina Benarese
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Caterina Branca
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Edoardo Parrella
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
| | - Pier Franco Spano
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
- IRCCS, San Camillo Hospital, Venice, Italy
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, National Institute of Neuroscience, University of Brescia, Brescia, Italy
- IRCCS, San Camillo Hospital, Venice, Italy
| |
Collapse
|
78
|
Offermanns S, Schwaninger M. Nutritional or pharmacological activation of HCA(2) ameliorates neuroinflammation. Trends Mol Med 2015; 21:245-55. [PMID: 25766751 DOI: 10.1016/j.molmed.2015.02.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 10/23/2022]
Abstract
Neuroinflammation is a pathology common to many neurological diseases, including multiple sclerosis (MS) and stroke. However, therapeutic attempts to modulate neuroinflammation have proved difficult. Neuroinflammatory cells express HCA2, a receptor for the endogenous neuroprotective ketone body β-hydroxybutyrate (BHB) as well as for the drugs dimethyl fumarate (DMF) and nicotinic acid, which have established efficacy in the treatment of MS and experimental stroke, respectively. This review summarizes the evidence that HCA2 is involved in the therapeutic effects of DMF, nicotinic acid, and ketone bodies in reducing neuroinflammation. Furthermore, we discuss the mechanisms underlying the beneficial effects of HCA2 activation in neuroinflammatory diseases and the therapeutic potential of recently developed synthetic ligands of HCA2.
Collapse
Affiliation(s)
- Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany; Medical Faculty, J.W. Goethe University, Frankfurt, Germany.
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany; DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany.
| |
Collapse
|
79
|
Habib P, Beyer C. Regulation of brain microglia by female gonadal steroids. J Steroid Biochem Mol Biol 2015; 146:3-14. [PMID: 24607811 DOI: 10.1016/j.jsbmb.2014.02.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 02/24/2014] [Indexed: 12/31/2022]
Abstract
Microglial cells are the primary mediators of the CNS immune defense system and crucial for shaping inflammatory responses. They represent a highly dynamic cell population which is constantly moving and surveying their environment. Acute brain damage causes a local attraction and activation of this immune cell type which involves neuron-to-glia and glia-to-glia interactions. The prevailing view attributes microglia a "negative" role such as defense and debris elimination. More topical studies also suggest a protective and "positive" regulatory function. Estrogens and progestins exert anti-inflammatory and neuroprotective effects in the CNS in acute and chronic brain diseases. Recent work revealed that microglial cells express subsets of classical and non-classical estrogen and progesterone receptors in a highly dynamic way. In this review article, we would like to stress the importance of microglia for the spreading of neural damage during hypoxia, their susceptibility to functional modulation by sex steroids, the potency of sex hormones to switch microglia from a pro-inflammatory M1 to neuroprotective M2 phenotype, and the regulation of pro- and anti-inflammatory properties including the inflammasome. We will further discuss the possibility that the neuroprotective action of sex steroids in the brain involves an early and direct modulation of local microglia cell function. This article is part of a Special Issue entitled 'Sex steroids and brain disorders'.
Collapse
Affiliation(s)
- Pardes Habib
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany.
| |
Collapse
|
80
|
CIIA negatively regulates neuronal cell death induced by oxygen–glucose deprivation and reoxygenation. Mol Cell Biochem 2014; 397:139-46. [DOI: 10.1007/s11010-014-2181-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 07/30/2014] [Indexed: 10/24/2022]
|
81
|
Blank T, Prinz M. NF-κB signaling regulates myelination in the CNS. Front Mol Neurosci 2014; 7:47. [PMID: 24904273 PMCID: PMC4033361 DOI: 10.3389/fnmol.2014.00047] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/05/2014] [Indexed: 01/12/2023] Open
Abstract
Besides myelination of neuronal axons by oligodendrocytes to facilitate propagation of action potentials, oligodendrocytes also support axon survival and function. A key transcription factor involved in these processes is nuclear factor-κB (NF-κB), a hetero or homodimer of the Rel family of proteins, including p65, c-Rel, RelB, p50, and p52. Under unstimulated, NF-κB remains inactive in the cytoplasm through interaction with NF-κB inhibitors (IκBs). Upon activation of NF-κB the cytoplasmic IκBs gets degradated, allowing the translocation of NF-κB into the nucleus where the dimer binds to the κB consensus DNA sequence and regulates gene transcription. In this review we describe how oligodendrocytes are, directly or indirectly via neighboring cells, regulated by NF-κB signaling with consequences for innate and adaptive immunity and for regulation of cell apoptosis and survival.
Collapse
Affiliation(s)
- Thomas Blank
- Institute of Neuropathology, University of Freiburg Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg Freiburg, Germany ; BIOSS Centre for Biological Signalling Studies, University of Freiburg Freiburg, Germany
| |
Collapse
|
82
|
Rahman M, Muhammad S, Khan MA, Chen H, Ridder DA, Müller-Fielitz H, Pokorná B, Vollbrandt T, Stölting I, Nadrowitz R, Okun JG, Offermanns S, Schwaninger M. The β-hydroxybutyrate receptor HCA2 activates a neuroprotective subset of macrophages. Nat Commun 2014; 5:3944. [PMID: 24845831 DOI: 10.1038/ncomms4944] [Citation(s) in RCA: 324] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/22/2014] [Indexed: 12/19/2022] Open
Abstract
The ketone body β-hydroxybutyrate (BHB) is an endogenous factor protecting against stroke and neurodegenerative diseases, but its mode of action is unclear. Here we show in a stroke model that the hydroxy-carboxylic acid receptor 2 (HCA2, GPR109A) is required for the neuroprotective effect of BHB and a ketogenic diet, as this effect is lost in Hca2(-/-) mice. We further demonstrate that nicotinic acid, a clinically used HCA2 agonist, reduces infarct size via a HCA2-mediated mechanism, and that noninflammatory Ly-6C(Lo) monocytes and/or macrophages infiltrating the ischemic brain also express HCA2. Using cell ablation and chimeric mice, we demonstrate that HCA2 on monocytes and/or macrophages is required for the protective effect of nicotinic acid. The activation of HCA2 induces a neuroprotective phenotype of monocytes and/or macrophages that depends on PGD2 production by COX1 and the haematopoietic PGD2 synthase. Our data suggest that HCA2 activation by dietary or pharmacological means instructs Ly-6C(Lo) monocytes and/or macrophages to deliver a neuroprotective signal to the brain.
Collapse
Affiliation(s)
- Mahbubur Rahman
- 1] Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany [2]
| | - Sajjad Muhammad
- 1] Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany [2] [3]
| | - Mahtab A Khan
- 1] Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany [2]
| | - Hui Chen
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Dirk A Ridder
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Helge Müller-Fielitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Barbora Pokorná
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Tillman Vollbrandt
- Institute for Systemic Inflammation Research, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Ines Stölting
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Roger Nadrowitz
- Institute of Radiotherapy and Nuclear Medicine, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Jürgen G Okun
- Department of Pediatrics, University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Stefan Offermanns
- 1] Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany [2] Medical Faculty, Goethe University, Frankfurt, Germany
| | - Markus Schwaninger
- 1] Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany [2] DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, 23562 Lübeck, Germany
| |
Collapse
|
83
|
Lee JE, Lim MS, Park JH, Park CH, Koh HC. Nuclear NF-κB contributes to chlorpyrifos-induced apoptosis through p53 signaling in human neural precursor cells. Neurotoxicology 2014; 42:58-70. [DOI: 10.1016/j.neuro.2014.04.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 03/25/2014] [Accepted: 04/02/2014] [Indexed: 01/01/2023]
|
84
|
Egea J, Romero A, Parada E, León R, Dal-Cim T, López M. Neuroprotective effect of dimebon against ischemic neuronal damage. Neuroscience 2014; 267:11-21. [DOI: 10.1016/j.neuroscience.2014.02.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 02/19/2014] [Indexed: 02/06/2023]
|
85
|
Wang CP, Zhang LZ, Li GC, Shi YW, Li JL, Zhang XC, Wang ZW, Ding F, Liang XM. Mulberroside a protects against ischemic impairment in primary culture of rat cortical neurons after oxygen-glucose deprivation followed by reperfusion. J Neurosci Res 2014; 92:944-54. [DOI: 10.1002/jnr.23374] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/27/2013] [Accepted: 01/27/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Cai-Ping Wang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Lu-Zhong Zhang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Gui-Cai Li
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Yun-wei Shi
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Jian-Long Li
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Xiao-Chuan Zhang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Zhi-Wei Wang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
- Department of Pharmacology; University of California; Irvine California
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
| | - Xin-Miao Liang
- Jiangsu Key Laboratory of Neuroregeneration; Nantong University; Nantong China
- Dalian Institute of Chemical Physics; Chinese Academy of Sciences; Dalian China
| |
Collapse
|
86
|
Heinonen AM, Rahman M, Dogbevia G, Jakobi H, Wölfl S, Sprengel R, Schwaninger M. Neuroprotection by rAAV-mediated gene transfer of bone morphogenic protein 7. BMC Neurosci 2014; 15:38. [PMID: 24618040 PMCID: PMC3975265 DOI: 10.1186/1471-2202-15-38] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 03/05/2014] [Indexed: 01/15/2023] Open
Abstract
Background Bone morphogenic proteins (BMPs) promote the survival of neurons, suggesting a therapeutic application of BMPs in the treatment of acute and chronic neurodegenerative disorders. However, the application of recombinant BMPs in vivo is limited by their short half-life. To provide a continuous supply for functionally active BMPs, we expressed BMP7, BMP2 and the BMP inhibitor Noggin under the control of rAAV vectors in vivo. For visual control of rAAV-mediated BMP (v-BMP) expression we fused the secreted morphogenic polypeptides and the fluorescent reporter protein Venus via the ‘ribosomal skip’ promoting 2A peptide-bridge. Results In primary cortical neurons, the rAAV-expressed morphogenic polypeptides were efficiently released from the 2A-Venus fusion precursors, were secreted, correctly processed and functionally active as shown by their effects on Smad phosphorylation in HeLa cells and in primary neurons, by the protection of v-BMP7-transduced primary cortical neurons against oxidative stress, and by the activation of BMP responsive GFP in v-BMP2 transduced reporter mice. In the stroke model of middle cerebral artery occlusion rAAV-transduced v-BMP7 reduced the infarct size in mice. Conclusion Polycistronic rAAV vectors encoding secreted polypeptides and 2A-linked reporter proteins are potential novel therapeutic tools for the treatment of neurological and neurodegenerative diseases. Using this technique we documented that rAAV delivery of BMP7 reduced ischemic cell death in mice.
Collapse
Affiliation(s)
| | | | | | | | | | - Rolf Sprengel
- Max Planck Institute for Medical Research, Jahnstrasse 29, Heidelberg D-69120, Germany.
| | | |
Collapse
|
87
|
|
88
|
Maqbool A, Lattke M, Wirth T, Baumann B. Sustained, neuron-specific IKK/NF-κB activation generates a selective neuroinflammatory response promoting local neurodegeneration with aging. Mol Neurodegener 2013; 8:40. [PMID: 24119288 PMCID: PMC3827934 DOI: 10.1186/1750-1326-8-40] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 10/09/2013] [Indexed: 12/17/2022] Open
Abstract
Background Increasing evidence indicates that neuroinflammation is a critical factor contributing to the progression of various neurodegenerative diseases. The IKK/NF-κB signalling system is a central regulator of inflammation, but it also affects neuronal survival and differentiation. A complex interplay between different CNS resident cells and infiltrating immune cells, which produce and respond to various inflammatory mediators, determines whether neuroinflammation is beneficial or detrimental. The IKK/NF-κB system is involved in both production of and responses to these mediators, although the precise contribution depends on the cell type as well as the cellular context, and is only partially understood. Here we investigated the specific contribution of neuronal IKK/NF-κB signalling on the regulation of neuroinflammatory processes and its consequences. To address this issue, we established and analysed a conditional gain-of-function mouse model that expresses a constitutively active allele of IKK2 in principal forebrain neurons (IKK2nCA). Proinflammatory gene and growth factor expression, histopathology, microgliosis, astrogliosis, immune cell infiltration and spatial learning were assessed at different timepoints after persistent canonical IKK2/NF-κB activation. Results In contrast to other cell types and organ systems, chronic IKK2/NF-κB signalling in forebrain neurons of adult IKK2nCA animals did not cause a full-blown inflammatory response including infiltration of immune cells. Instead, we found a selective inflammatory response in the dentate gyrus characterized by astrogliosis, microgliosis and Tnf-α upregulation. Furthermore, downregulation of the neurotrophic factor Bdnf correlated with a selective and progressive atrophy of the dentate gyrus and a decline in hippocampus-dependent spatial learning. Neuronal degeneration was associated with increased Fluoro-jade staining, but lacked activation of apoptosis. Remarkably, neuronal loss could be partially reversed when chronic IKK2/NF-κB signalling was turned off and Bdnf expression was restored. Conclusion Our results demonstrate that persistent IKK2/NF-κB signalling in forebrain neurons does not induce overall neuroinflammation, but elicits a selective inflammatory response in the dentate gyrus accompanied by decreased neuronal survival and impaired learning and memory. Our findings further suggest that chronic activation of neuronal IKK2/NF-κB signalling, possibly as a consequence of neuroinflammatory conditions, is able to induce apoptosis-independent neurodegeneration via paracrine suppression of Bdnf synthesis.
Collapse
Affiliation(s)
- Ayesha Maqbool
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, Ulm 89081 Germany.
| | | | | | | |
Collapse
|
89
|
Purkayastha S, Cai D. Neuroinflammatory basis of metabolic syndrome. Mol Metab 2013; 2:356-63. [PMID: 24327952 DOI: 10.1016/j.molmet.2013.09.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 09/23/2013] [Accepted: 09/25/2013] [Indexed: 01/07/2023] Open
Abstract
Inflammatory reaction is a fundamental defense mechanism against threat towards normal integrity and physiology. On the other hand, chronic diseases such as obesity, type 2 diabetes, hypertension and atherosclerosis, have been causally linked to chronic, low-grade inflammation in various metabolic tissues. Recent cross-disciplinary research has led to identification of hypothalamic inflammatory changes that are triggered by overnutrition, orchestrated by hypothalamic immune system, and sustained through metabolic syndrome-associated pathophysiology. While continuing research is actively trying to underpin the identity and mechanisms of these inflammatory stimuli and actions involved in metabolic syndrome disorders and related diseases, proinflammatory IκB kinase-β (IKKβ), the downstream nuclear transcription factor NF-κB and some related molecules in the hypothalamus were discovered to be pathogenically significant. This article is to summarize recent progresses in the field of neuroendocrine research addressing the central integrative role of neuroinflammation in metabolic syndrome components ranging from obesity, glucose intolerance to cardiovascular dysfunctions.
Collapse
Affiliation(s)
- Sudarshana Purkayastha
- Department of Molecular Pharmacology, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
90
|
Zamani M, Soleimani M, Golab F, Mohamadzadeh F, Mehdizadeh M, Katebi M. NeuroProtective effects of adenosine receptor agonist coadministration with ascorbic acid on CA1 hippocampus in a mouse model of ischemia reperfusion injury. Metab Brain Dis 2013; 28:367-74. [PMID: 23640013 DOI: 10.1007/s11011-013-9408-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 04/10/2013] [Indexed: 12/20/2022]
Abstract
Ischemic brain injury is a leading cause of sever neurological and neurobehavioral deficits and death. The hippocampus plays vital roles in learning and memory processes and it is impaired by ischemic insults. Cerebral ischemia/reperfusion leads to Oxidative stress damage impairing the hippocampus. Here we tested whether ascorbic acid and adenosine receptor played a neuroprotective role in a mouse brain ischemia model induced by common carotid arteries occlusion. Adult male mice were randomly assigned into nine experimental groups. The animals were subjected to ischemia by the ligation of common carotid arteries for 15 min. Drugs were injected intrapritoneally once daily for 7 days. Behavioral tests performed at day 14 and then mice were killed at day 21 and their brains were fixed for microscopic studies and some samples were prepared for western blot analysis. Western blot analysis utilized to evaluate the expression of apoptosis-related proteinsin the hippocampus. Short-term memory was assessed by shuttle-box test. Our findings revealed that administration of vitamin C and N6-cyclopentyladenosine (CPA) significantly attenuated ischemia-induced brain injury. Vitamin C and CPA administration increased the expression of anti-apoptotic protein Bcl-2 and decreased the expression of pro-apoptotic protein Bax in the ischemic mice. Ischemia caused short-term memory loss that was improved by vitamin c and CPA treatment. Our results demonstrate that treatment with vitamin C and adenosine receptor agonist attenuated cerebral ischemia/reperfusion-induced brain injury as a potential neuroprotective agent.
Collapse
Affiliation(s)
- M Zamani
- Cellular and Molecular Research Center and Department of Anatomy, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
91
|
Miyazaki D, Mihara S, Inata K, Sasaki SI, Tominaga T, Yakura K, Ishida W, Fukushima A, Inoue Y. Pharmacologic Inhibition of IκB Kinase Activates Immediate Hypersensitivity Reactions in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:96-107. [DOI: 10.1016/j.ajpath.2013.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 02/24/2013] [Accepted: 04/01/2013] [Indexed: 11/26/2022]
|
92
|
Butein inhibits NF-κB activation and reduces infiltration of inflammatory cells and apoptosis after spinal cord injury in rats. Neurosci Lett 2013; 542:87-91. [DOI: 10.1016/j.neulet.2013.03.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 03/02/2013] [Accepted: 03/05/2013] [Indexed: 11/22/2022]
|
93
|
Mc Guire C, Rahman M, Schwaninger M, Beyaert R, van Loo G. The ubiquitin editing enzyme A20 (TNFAIP3) is upregulated during permanent middle cerebral artery occlusion but does not influence disease outcome. Cell Death Dis 2013; 4:e531. [PMID: 23470538 PMCID: PMC3613833 DOI: 10.1038/cddis.2013.55] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
94
|
Abstract
Inflammation is a biological response mounted by the immune system against dangerous assaults that threaten the integrity and normal physiology of an organism. During the past decades, cross-disciplinary research from immunology and endocrinology has much broadened this knowledge by demonstrating that chronic conditions of nutritional excess constitute an independent category of inflammatory activators, and the resulting chronic and low-grade inflammation is an important characteristic of overnutrition-induced diseases. A large body of research has demonstrated that these diseases are pathogenically associated with the local, negative actions of inflammation in peripheral tissues predominantly including the liver, muscle, and fat. In this research background, more recent research has advanced to a new level, with the important discoveries showing that overnutrition-induced inflammation occurs in the brain and thus plays a broad and leadership role in overnutrition-induced diseases. While much more research establishments are expected in this emerging and quickly expanding research, the appreciated understandings have been mainly based on proinflammatory IKKβ/NF-κB pathway and related molecules in the hypothalamus. In this chapter, the author focuses on describing IKKβ/NF-κB-induced neural inflammation in the context of overnutrition-induced metabolic inflammation and especially the central roles of this neural inflammation in the development of a spectrum of overnutrition-related diseases.
Collapse
Affiliation(s)
- Dongsheng Cai
- Department of Molecular Pharmacology, Institute of Aging, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
95
|
Peluffo H, Gonzalez P, Acarin L, Arís A, Beyaert R, Villaverde A, Gonzalez B. Overexpression of the nuclear factor kappaB inhibitor A20 is neurotoxic after an excitotoxic injury to the immature rat brain. Neurol Res 2012; 35:308-19. [PMID: 23336395 DOI: 10.1179/1743132812y.0000000139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
BACKGROUND The zinc finger protein A20 is an ubiquitinating/deubiquitinating enzyme essential for the termination of inflammatory reactions through the inhibition of nuclear factor kappaB (NF-kappaB) signaling. Moreover, it also shows anti-apoptotic activities in some cell types and proapoptotic/pronecrotic effects in others. Although it is known that the regulation of inflammatory and cell death processes are critical in proper brain functioning and that A20 mRNA is expressed in the CNS, its role in the brain under physiological and pathological conditions is still unknown. METHODS In the present study, we have evaluated the effects of A20 overexpression in mixed cortical cultures in basal conditions: the in vivo pattern of endogenous A20 expression in the control and N-methyl-d-aspartate (NMDA) excitotoxically damaged postnatal day 9 immature rat brain, and the post-injury effects of A20 overexpression in the same lesion model. RESULTS Our results show that overexpression of A20 in mixed cortical cultures induced significant neuronal death by decreasing neuronal cell counts by 45 ± 9%. in vivo analysis of endogenous A20 expression showed widespread expression in gray matter, mainly in neuronal cells. However, after NMDA-induced excitotoxicity, neuronal A20 was downregulated in the neurodegenerating cortex and striatum at 10-24 hours post-lesion, and it was re-expressed at longer survival times in reactive astrocytes located mainly in the lesion border. When A20 was overexpressed in vivo 2 hours after the excitotoxic damage, the lesion volume at 3 days post-lesion showed a significant increase (20.8 ± 7.0%). No A20-induced changes were observed in the astroglial response to injury. CONCLUSIONS A20 is found in neuronal cells in normal conditions and is also expressed in astrocytes after brain damage, and its overexpression is neurotoxic for cortical neurons in basal mixed neuron-glia culture conditions and exacerbates postnatal brain excitotoxic damage.
Collapse
Affiliation(s)
- Hugo Peluffo
- Insitut Pasteur de Montevideo, Mataojo 2020, Montevideo 11400, Uruguay.
| | | | | | | | | | | | | |
Collapse
|
96
|
Brähler S, Ising C, Hagmann H, Rasmus M, Hoehne M, Kurschat C, Kisner T, Goebel H, Shankland S, Addicks K, Thaiss F, Schermer B, Pasparakis M, Benzing T, Brinkkoetter PT. Intrinsic proinflammatory signaling in podocytes contributes to podocyte damage and prolonged proteinuria. Am J Physiol Renal Physiol 2012; 303:F1473-85. [DOI: 10.1152/ajprenal.00031.2012] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Inflammation conveys the development of glomerular injury and is a major cause of progressive kidney disease. NF-κB signaling is among the most important regulators of proinflammatory signaling. Its role in podocytes, the epithelial cells at the kidney filtration barrier, is poorly understood. Here, we inhibited NF-κB signaling in podocytes by specific ablation of the NF-κB essential modulator (NEMO, IKKγ). Podocyte-specific NEMO-deficient mice (NEMOpko) were viable and did not show proteinuria or overt changes in kidney morphology. After induction of glomerulonephritis, both NEMOpkoand control mice developed significant proteinuria. However, NEMOpkomice recovered much faster, showing rapid remission of proteinuria and restoration of podocyte morphology. Interestingly, quantification of infiltrating macrophages, T-lymphocytes, and granulocytes at day 7 revealed no significant difference between wild-type and NEMOpko. To further investigate the underlying mechanisms, we created a stable NEMO knockdown mouse podocyte cell line. Again, no overt changes in morphology were observed. Translocation of NF-κB to the nucleus after stimulation with TNFα or IL-1 was sufficiently inhibited. Moreover, secretion of proinflammatory chemokines from podocytes after stimulation with TNFα or IL-1 was significantly reduced in NEMO-deficient podocytes and in glomerular samples obtained at day 7 after induction of nephrotoxic nephritis. Collectively, these results show that proinflammatory activity of NF-κB in podocytes aggravates proteinuria in experimental glomerulonephritis in mice. Based on these data, it may be speculated that immunosuppressive drugs may not only target professional immune cells but also podocytes directly to convey their beneficial effects in various types of glomerulonephritis.
Collapse
Affiliation(s)
- Sebastian Brähler
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Christina Ising
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Henning Hagmann
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Melanie Rasmus
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Martin Hoehne
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Christine Kurschat
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Tuelay Kisner
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Heike Goebel
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Stuart Shankland
- Division of Nephrology, University of Washington, Seattle, Washington
| | - Klaus Addicks
- Institute of Anatomy, University of Cologne, Cologne, Germany
| | - Friedrich Thaiss
- Department of Internal Medicine III, University Hospital, Hamburg-Eppendorf, Hamburg, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, Cologne, Germany; and
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Paul Thomas Brinkkoetter
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
97
|
Jiang Y, Zhu J, Wu L, Xu G, Dai J, Liu X. Tetracycline inhibits local inflammation induced by cerebral ischemia via modulating autophagy. PLoS One 2012; 7:e48672. [PMID: 23144925 PMCID: PMC3492486 DOI: 10.1371/journal.pone.0048672] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 09/28/2012] [Indexed: 11/24/2022] Open
Abstract
Background Tetracycline exerts neuroprotection via suppressing the local inflammation induced by cerebral ischemia. However, the underlying mechanism is not completely clear. Methodology/Principal Findings The mRNA and protein expressions of tumor necrosis factor α and interleukin 6 and the number of activated microglia were measured to detect the inflammatory process in the ischemic hemisphere. The key proteins of nuclear factor kappa B pathway and the binding activity of nuclear factor kappa B were also measured. Two key components of autophagy, Beclin 1 and LC3, were detected by western blotting. Pretreatment with tetracycline inhibited the mRNA and protein expressions of tumor necrosis factor α and interleukin 6 and decreased the numbers of activated and phagocytotic microglia. Tetracycline down regulated the total and phosphorylated expressions of IKK, IκB and p65 (P<0.05). The autophagy inhibitor, 3-methyladenine, inhibited inflammation and activation of nuclear factor kappa B pathway. The levels of Beclin 1 and LC3 were decreased by 3-methyladenine and tetracycline. Conclusions/Significance Our data suggested that pretreatment of tetracycline may inhibit autophagy in the ischemic stroke brain and then suppress the inflammatory process via inhibiting the activation of nuclear factor kappa B pathway.
Collapse
Affiliation(s)
- Yongjun Jiang
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
| | - Juehua Zhu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
| | - Li Wu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
| | - Jianwu Dai
- Key laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, PR China
- * E-mail: (XL); (JD)
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Jiangsu Province, China
- * E-mail: (XL); (JD)
| |
Collapse
|
98
|
Li H, Huang J, Du W, Jia C, Yao H, Wang Y. TRPC6 inhibited NMDA receptor activities and protected neurons from ischemic excitotoxicity. J Neurochem 2012; 123:1010-8. [DOI: 10.1111/jnc.12045] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 09/26/2012] [Accepted: 09/27/2012] [Indexed: 11/27/2022]
Affiliation(s)
- Hongyu Li
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
- The Graduate School, Chinese Academy of Science; Shanghai China
| | - Junbo Huang
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
- The Graduate School, Chinese Academy of Science; Shanghai China
| | - Wanlu Du
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
| | - Caixia Jia
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
| | - Hailan Yao
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
| | - Yizheng Wang
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
| |
Collapse
|
99
|
Ridder DA, Schwaninger M. TAK1 inhibition for treatment of cerebral ischemia. Exp Neurol 2012; 239:68-72. [PMID: 23022457 DOI: 10.1016/j.expneurol.2012.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 09/20/2012] [Indexed: 11/18/2022]
Abstract
TGFβ-activated kinase 1 (TAK1), a MAP3 kinase, is involved in at least five signaling cascades that modulate ischemic brain damage. Inhibition of TAK1 may therefore be an efficient way to interfere with multiple mechanisms in ischemic stroke. Indeed, a recent publication in Experimental Neurology confirmed that TAK1 inhibition by 5Z-7-oxozeaenol is neuroprotective. The beneficial effect of 5Z-7-oxozeaenol was associated with a reduced activation of Jun kinase that leads to inflammation and apoptosis. Recently, other TAK1 inhibitors were developed suggesting that TAK1 may prove as an efficient therapeutic target for neurodegenerative diseases if safety issues are not limiting.
Collapse
Affiliation(s)
- Dirk A Ridder
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany
| | | |
Collapse
|
100
|
Kanngiesser M, Häussler A, Myrczek T, Küsener N, Lim HY, Geisslinger G, Niederberger E, Tegeder I. Inhibitor kappa B kinase beta dependent cytokine upregulation in nociceptive neurons contributes to nociceptive hypersensitivity after sciatic nerve injury. THE JOURNAL OF PAIN 2012; 13:485-97. [PMID: 22564672 DOI: 10.1016/j.jpain.2012.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 02/04/2012] [Accepted: 02/21/2012] [Indexed: 10/28/2022]
Abstract
UNLABELLED Inhibitor kappa B kinase (IKK)-mediated nuclear factor-kappa B (NF-κB) activation is a major pathway for transcriptional control of various pro-inflammatory factors. We here assessed whether activation of this pathway specifically in primary nociceptive neurons of the dorsal root ganglia (DRG) contributes to the development of nociceptive hypersensitivity. Mice carrying a cre-loxP-mediated deletion of inhibitor kappa B kinase beta (IKKβ) in DRG neurons were protected from nerve injury-evoked allodynia and hyperalgesia. This effect was mimicked by systemic treatment with an IKKβ inhibitor but was not observed upon specific inhibition of IKKβ in the spinal cord, suggesting a specific role of IKKβ in the peripheral neurons. The deletion of IKKβ in DRG neurons did not affect constitutive neuronal NF-κB activity, but reduced nerve injury-evoked NF-κB stimulation in the DRG and was associated with reduced upregulation of interleukin-16, monocyte chemoattractant protein-1/chemokine (CC motif) ligand 2 (MCP-1/CCL2), and tumor necrosis factor alpha (TNFα) in the DRG. These cytokines evoked a rapid rise of intracellular calcium in subsets of primary DRG neurons. The results suggest that IKKβ-mediated NF-κB stimulation in injured primary sensory neurons promotes cytokine and chemokine production and contributes thereby to the development of chronic pain. PERSPECTIVE Inhibitors of IKK that do not pass the blood-brain barrier and act only in the periphery might be useful for reduction of the pro-inflammatory response in peripheral DRG neurons and reduce thereby nerve injury-evoked pain without affecting neuroprotective effects of NF-κB in the central nervous system.
Collapse
Affiliation(s)
- Maike Kanngiesser
- Pharmazentrum Frankfurt/ZAFES, Clinical Pharmacology, Goethe-University, Frankfurt, Germany
| | | | | | | | | | | | | | | |
Collapse
|