1
|
Lazzarin T, Tonon CR, Martins D, Fávero EL, Baumgratz TD, Pereira FWL, Pinheiro VR, Ballarin RS, Queiroz DAR, Azevedo PS, Polegato BF, Okoshi MP, Zornoff L, Rupp de Paiva SA, Minicucci MF. Post-Cardiac Arrest: Mechanisms, Management, and Future Perspectives. J Clin Med 2022; 12:259. [PMID: 36615059 PMCID: PMC9820907 DOI: 10.3390/jcm12010259] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Cardiac arrest is an important public health issue, with a survival rate of approximately 15 to 22%. A great proportion of these deaths occur after resuscitation due to post-cardiac arrest syndrome, which is characterized by the ischemia-reperfusion injury that affects the role body. Understanding physiopathology is mandatory to discover new treatment strategies and obtain better results. Besides improvements in cardiopulmonary resuscitation maneuvers, the great increase in survival rates observed in recent decades is due to new approaches to post-cardiac arrest care. In this review, we will discuss physiopathology, etiologies, and post-resuscitation care, emphasizing targeted temperature management, early coronary angiography, and rehabilitation.
Collapse
Affiliation(s)
- Taline Lazzarin
- Internal Medicine Department, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu 18607-741, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Olate-Briones A, Escalona E, Salazar C, Herrada MJ, Liu C, Herrada AA, Escobedo N. The meningeal lymphatic vasculature in neuroinflammation. FASEB J 2022; 36:e22276. [PMID: 35344212 DOI: 10.1096/fj.202101574rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 12/13/2022]
Abstract
The lymphatic vasculature is a unidirectional network of lymphatic endothelial cells, whose main role is to maintain fluid homeostasis along with the absorption of dietary fat in the gastrointestinal organs and management and coordination of immune cell trafficking into lymph nodes during homeostasis and under inflammatory conditions. In homeostatic conditions, immune cells, such as dendritic cells, macrophages, or T cells can enter into the lymphatic vasculature and move easily through the lymph reaching secondary lymph nodes where immune cell activation or peripheral tolerance can be modulated. However, under inflammatory conditions such as pathogen infection, increased permeabilization of lymphatic vessels allows faster immune cell migration into inflamed tissues following a chemokine gradient, facilitating pathogen clearance and the resolution of inflammation. Interestingly, since the re-discovery of lymphatic vasculature in the central nervous system, known as the meningeal lymphatic vasculature, the role of these lymphatics as a key player in several neurological disorders has been described, with emphasis on the neurodegenerative process. Alternatively, less has been discussed about meningeal lymphatics and its role in neuroinflammation. In this review, we discuss current knowledge about the anatomy and function of the meningeal lymphatic vasculature and specifically analyze its contribution to different neuroinflammatory processes, highlighting the potential therapeutic target of meningeal lymphatic vasculature in these pathological conditions.
Collapse
Affiliation(s)
- Alexandra Olate-Briones
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Emilia Escalona
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Celia Salazar
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | | | - Chaohong Liu
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andrés A Herrada
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Noelia Escobedo
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
3
|
Lin B, Chen R, Wang Q, Li Z, Yang S, Feng Y. Transcriptomic and Metabolomic Profiling Reveals the Protective Effect of Acanthopanax senticosus (Rupr. & Maxim.) Harms Combined With Gastrodia elata Blume on Cerebral Ischemia-Reperfusion Injury. Front Pharmacol 2021; 12:619076. [PMID: 33935709 PMCID: PMC8085551 DOI: 10.3389/fphar.2021.619076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
The effects of current treatment strategies used in ischemic stroke are weakened by cerebral ischemia-reperfusion (CIR) injury. Suitable treatment regimens targeting CIR injury are still lacking. Two herbs, namely, Acanthopanax senticosus (Rupr. & Maxim.) Harms (ASE) and Gastrodia elata Blume (GEB), have been used as traditional Chinese medicine and are indicated in the treatment of stroke and cerebrovascular diseases. However, there are no studies that report the effects of ASE combined with GEB in the treatment of CIR injury. In this study, we used the Zea Longa method to induce CIR injury in male Wistar rats. Results of the pharmacodynamic studies revealed that co-administration of ASE and GEB may improve neuronal injury and prevent neuronal apoptosis by reducing oxidative stress and inflammation, and also help prevent CIR injury. On the basis of our hypothesis, we combined the results from transcriptomic and metabonomic analyses and found that ASE and GEB could prevent CIR injury by targeting phenylalanine, pyrimidine, methionine, and sphingolipid metabolism. Therefore, our study provides the basis for the compatibility and efficacy of ASE and GEB.
Collapse
Affiliation(s)
- Bingfeng Lin
- Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Renhao Chen
- Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qi Wang
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| | - Zhifeng Li
- Jiangxi University of Traditional Chinese Medicine, Nanchang, China.,Nanchang Key Laboratory of Active Ingredients of Traditional Chinese Medicine and Natural Medicine, Nanchang, China
| | - ShiLin Yang
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| | - YuLin Feng
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| |
Collapse
|
4
|
Chen H, Li S, Xu W, Hong Y, Dou R, Shen H, Liu X, Wu T, He JC. Interleukin-17A promotes the differentiation of bone marrow mesenchymal stem cells into neuronal cells. Tissue Cell 2021; 69:101482. [PMID: 33418236 DOI: 10.1016/j.tice.2020.101482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 11/25/2022]
Abstract
Ischemia or hemorrhagic stroke is one of the leading causes of death and permanent disability in the worldwide population. As a consequence of the potential increasing in stroke, stem cell therapy is currently an area of intense focus. However, there are less data available regarding the promotion of healing efficacy after stroke. The present study aimed to investigate whether the cytokine interleukin-17A (IL-17A) could have a role in promoting the neuronal differentiation of mesenchymal stem cells (MSCs) and to investigate the associated molecular mechanism. Firstly, different concentration of IL-17A at range from 5-40 ng/mL was applied to stimulate bone marrow MSCs (BMSCs) during the course of neurogenic differentiation. Then reverse transcription-PCR, histological analyses and immunofluorescence assays were used to determine the optimum concentration of IL-17A in promoting the neuronal differentiation of BMSCs, which was 20 ng/mL. Mechanistically, Wnt signaling pathway was activated and Notch signaling pathway was suppressed. In addition, there were antergic effect of these two signaling pathways modulating the neurogenic differentiation of BMSCs induced by IL-17A. The present study demonstrated the potential role of IL-17A-based BMSCs strategy for promoting neuronal differentiation in vitro. However, the treatment efficacy could be considerably confirmed in animals with ischemia stroke. Therefore, a more sophisticated strategy that addresses the complicated treatment associated with stroke is needed.
Collapse
Affiliation(s)
- Hanlin Chen
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China
| | - Shasha Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China
| | - Wanting Xu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China
| | - Yongfeng Hong
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Shushan District, Hefei, Anhui, 230061, China
| | - Rengang Dou
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Shushan District, Hefei, Anhui, 230061, China
| | - Hongtao Shen
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Shushan District, Hefei, Anhui, 230061, China
| | - Xue Liu
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Shushan District, Hefei, Anhui, 230061, China
| | - Tingting Wu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China.
| | - Jia Cai He
- Stomatologic Hospital & College, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, 69 Meishan Road, Shushan District, Hefei, Anhui, 230001, China.
| |
Collapse
|
5
|
Diaz-Cañestro C, Reiner MF, Bonetti NR, Liberale L, Merlini M, Wüst P, Amstalden H, Briand-Schumacher S, Semerano A, Giacalone G, Sessa M, Beer JH, Akhmedov A, Lüscher TF, Camici GG. AP-1 (Activated Protein-1) Transcription Factor JunD Regulates Ischemia/Reperfusion Brain Damage via IL-1β (Interleukin-1β). Stroke 2019; 50:469-477. [PMID: 30626291 DOI: 10.1161/strokeaha.118.023739] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background and Purpose- Inflammation is a major pathogenic component of ischemia/reperfusion brain injury, and as such, interventions aimed at inhibiting inflammatory mediators promise to be effective strategies in stroke therapy. JunD-a member of the AP-1 (activated protein-1) family of transcription factors-was recently shown to regulate inflammation by targeting IL (interleukin)-1β synthesis and macrophage activation. The purpose of the present study was to assess the role of JunD in ischemia/reperfusion-induced brain injury. Methods- WT (wild type) mice randomly treated with either JunD or scramble (control) siRNA were subjected to 45 minutes of transient middle cerebral artery occlusion followed by 24 hours of reperfusion. Stroke size, neurological deficit, plasma/brain cytokines, and oxidative stress determined by 4-hydroxynonenal immunofluorescence staining were evaluated 24 hours after reperfusion. Additionally, the role of IL-1β was investigated by treating JunD siRNA mice with an anti-IL-1β monoclonal antibody on reperfusion. Finally, JunD expression was assessed in peripheral blood monocytes isolated from patients with acute ischemic stroke. Results- In vivo JunD knockdown resulted in increased stroke size, reduced neurological function, and increased systemic inflammation, as confirmed by higher neutrophil count and lymphopenia. Brain tissue IL-1β levels were augmented in JunD siRNA mice as compared with scramble siRNA, whereas no difference was detected in IL-6, TNF-α (tumor necrosis factor-α), and 4-hydroxynonenal levels. The deleterious effects of silencing of JunD were rescued by treating mice with an anti-IL-1β antibody. In addition, JunD expression was decreased in peripheral blood monocytes of patients with acute ischemic stroke at 6 and 24 hours after onset of stroke symptoms compared with sex- and age-matched healthy controls. Conclusions- JunD blunts ischemia/reperfusion-induced brain injury via suppression of IL-1β.
Collapse
Affiliation(s)
- Candela Diaz-Cañestro
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Martin F Reiner
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Department of Internal Medicine, Cantonal Hospital Baden, Switzerland (M.F.R., J.H.B.)
| | - Nicole R Bonetti
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Luca Liberale
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Italy (L.L.)
| | - Mario Merlini
- Gladstone Institute of Neurological Disease, University of California, San Francisco (M.M.)
| | - Patricia Wüst
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Heidi Amstalden
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Sylvie Briand-Schumacher
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Aurora Semerano
- Department of Neurology, San Raffaele Scientific Institute, Milan, Italy (A.S., G.G.)
| | - Giacomo Giacalone
- Department of Neurology, San Raffaele Scientific Institute, Milan, Italy (A.S., G.G.)
| | - Maria Sessa
- SC Neurologia, Dipartimento Interaziendale Neuroscienze Cremona-Mantova, Azienda Socio-Sanitaria Territoriale (ASST) di cremona, Cremona, Italy (M.S.)
| | - Jürg H Beer
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Department of Internal Medicine, Cantonal Hospital Baden, Switzerland (M.F.R., J.H.B.)
| | - Alexander Akhmedov
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Thomas F Lüscher
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom (T.F.L.)
| | - Giovanni G Camici
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Zurich Neuroscience Center, University of Zurich, Switzerland (G.G.C.)
| |
Collapse
|
6
|
Vinukonda G, Liao Y, Hu F, Ivanova L, Purohit D, Finkel DA, Giri P, Bapatla L, Shah S, Zia MT, Hussein K, Cairo MS, La Gamma EF. Human Cord Blood-Derived Unrestricted Somatic Stem Cell Infusion Improves Neurobehavioral Outcome in a Rabbit Model of Intraventricular Hemorrhage. Stem Cells Transl Med 2019; 8:1157-1169. [PMID: 31322326 PMCID: PMC6811700 DOI: 10.1002/sctm.19-0082] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022] Open
Abstract
Intraventricular hemorrhage (IVH) is a severe complication of preterm birth, which leads to hydrocephalus, cerebral palsy, and mental retardation. There are no available therapies to cure IVH, and standard treatment is supportive care. Unrestricted somatic stem cells (USSCs) from human cord blood have reparative effects in animal models of brain and spinal cord injuries. USSCs were administered to premature rabbit pups with IVH and their effects on white matter integrity and neurobehavioral performance were evaluated. USSCs were injected either via intracerebroventricular (ICV) or via intravenous (IV) routes in 3 days premature (term 32d) rabbit pups, 24 hours after glycerol‐induced IVH. The pups were sacrificed at postnatal days 3, 7, and 14 and effects were compared to glycerol‐treated but unaffected or nontreated control. Using in vivo live bioluminescence imaging and immunohistochemical analysis, injected cells were found in the injured parenchyma on day 3 when using the IV route compared to ICV where cells were found adjacent to the ventricle wall forming aggregates; we did not observe any adverse events from either route of administration. The injected USSCs were functionally associated with attenuated microglial infiltration, less apoptotic cell death, fewer reactive astrocytes, and diminished levels of key inflammatory cytokines (TNFα and IL1β). In addition, we observed better preservation of myelin fibers, increased myelin gene expression, and altered reactive astrocyte distribution in treated animals, and this was associated with improved locomotor function. Overall, our findings support the possibility that USSCs exert anti‐inflammatory effects in the injured brain mitigating many detrimental consequences associated with IVH. stem cells translational medicine2019;8:1157–1169
Collapse
Affiliation(s)
- Govindaiah Vinukonda
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,Cell Biology & Anatomy, New York Medical College, Valhalla, New York, USA
| | - Yanling Liao
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Furong Hu
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Larisa Ivanova
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Deepti Purohit
- The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Dina A Finkel
- The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Priyadarshani Giri
- The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | | | - Shetal Shah
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Muhammed T Zia
- The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Karen Hussein
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,Cell Biology & Anatomy, New York Medical College, Valhalla, New York, USA.,Department of Medicine, Pathology, Microbiology & Immunology, Cell Biology & Anatomy, New York Medical College, Valhalla, New York, USA
| | - Edmund F La Gamma
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA.,Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
7
|
Wan JJ, Wang PY, Zhang Y, Qin Z, Sun Y, Hu BH, Su DF, Xu DP, Liu X. Role of acute-phase protein ORM in a mice model of ischemic stroke. J Cell Physiol 2019; 234:20533-20545. [PMID: 31026065 DOI: 10.1002/jcp.28653] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/22/2019] [Accepted: 03/06/2019] [Indexed: 12/12/2022]
Abstract
The only Food and Drug Administration-approved treatment for acute ischemic stroke is tissue plasminogen activator, and the discovery of novel therapeutic targets is critical. Here, we found orosomucoid (ORM), an acute-phase protein mainly produced by the liver, might act as a treatment candidate for an ischemic stroke. The results showed that ORM2 is the dominant subtype in mice normal brain tissue. After middle cerebral artery occlusion (MCAO), the level of ORM2 is significantly increased in the ischemic penumbra compared with the contralateral normal brain tissue, whereas ORM1 knockout did not affect the infarct size. Exogenous ORM could significantly decrease infarct size and neurological deficit score. Inspiringly, the best administration time point was at 4.5 and 6 hr after MCAO. ORM could markedly decrease the Evans blue extravasation, and improve blood-brain barrier-associated proteins expression in the ischemic penumbra of MACO mice and oxygen-glucose deprivation (OGD)-treated bEnd3 cells. Meanwhile, ORM could significantly alleviate inflammation by inhibiting the production of interleukin 1β (IL-1β), IL-6, and tumor necrosis factor α (TNF-α), reduce oxidative stress by improving the balance of malondialdehyde (MDA) and superoxide dismutase (SOD), inhibit apoptosis by decreasing caspase-3 activity in ischemic penumbra of MCAO mice and OGD-treated bEnd.3 cells. Because of its protective role at multiple levels, ORM might be a promising therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Jing-Jing Wan
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.,Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Peng-Yuan Wang
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Yu Zhang
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Zhen Qin
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.,Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Yang Sun
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.,Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Bo-Han Hu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Ding-Feng Su
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Dong-Ping Xu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.,Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
8
|
Ludwig PE, Thankam FG, Patil AA, Chamczuk AJ, Agrawal DK. Brain injury and neural stem cells. Neural Regen Res 2018; 13:7-18. [PMID: 29451199 PMCID: PMC5840995 DOI: 10.4103/1673-5374.224361] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2017] [Indexed: 12/26/2022] Open
Abstract
Many therapies with potential for treatment of brain injury have been investigated. Few types of cells have spurred as much interest and excitement as stem cells over the past few decades. The multipotentiality and self-renewing characteristics of stem cells confer upon them the capability to regenerate lost tissue in ischemic or degenerative conditions as well as trauma. While stem cells have not yet proven to be clinically effective in many such conditions as was once hoped, they have demonstrated some effects that could be manipulated for clinical benefit. The various types of stem cells have similar characteristics, and largely differ in terms of origin; those that have differentiated to some extent may exhibit limited capability in differentiation potential. Stem cells can aid in decreasing lesion size and improving function following brain injury.
Collapse
Affiliation(s)
- Parker E. Ludwig
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| | - Finosh G. Thankam
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| | - Arun A. Patil
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
- Department of Neurosurgery, Creighton University School of Medicine, Omaha, NE, USA
| | - Andrea J. Chamczuk
- Department of Neurosurgery, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K. Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
9
|
Iglesias-Osma MC, Blanco EJ, Carretero-Hernández M, García-Barrado MJ, Catalano-Iniesta L, Carretero M, Sánchez-Robledo V, Carretero J. Endothelial immunocytochemical expression of pituitary IL-1β and its relation to ACTH-positive cells is regulated by corticosterone in the male rat. Cytokine 2017; 99:9-17. [PMID: 28675823 DOI: 10.1016/j.cyto.2017.06.020] [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: 01/09/2017] [Revised: 06/09/2017] [Accepted: 06/27/2017] [Indexed: 11/17/2022]
Abstract
Interleukin-1 beta (IL-1β) is a cytokine linking the neuroendocrine system and metabolic homeostasis. We have previously demonstrated the relevance of IL-1β for maintaining the pituitary ACTH-producing cells by immuno-blocking its effects in pituitary cultures. However, the morphological characteristics and the intimate relationship of the pituitary cells expressing IL-1β and ACTH remain unknown. For determining pituitary variations of immunoreactivity for IL-1β and its relation with ACTH-positive cells under stress situations, we performed an immunohistochemical analysis of the expression of IL-1β and ACTH in the pituitary gland of adult rats, in the absence or presence of corticosterone, by establishing different groups: untreated, sham-operated, and bilaterally adrenalectomized animals. In the rats subjected to surgery, the glucocorticoid was administered on the same day of the intervention and on the third day post-surgery. Interestingly, it was observed that IL-1β was located in the pituitary endothelial cells at the hypophyseal portal vessels, regardless of the treatment schedule. When comparing the pituitary immunoreactive surface to IL-1β expression without corticosterone, adrenalectomized animals displayed a significantly greater area than the sham-operated animals. Corticosterone significantly inhibited the effect of adrenalectomy depending on the time interval it was administered. By in situ hybridization, IL-1β mRNA expression was also correlated with immnunocytochemical expression of pituitary IL-1β. Our results demonstrate that IL-1β is a constitutive element in endothelial portal pituitary vessels and under stress experimental conditions IL-1β increases its expression and its relation with ACTH-positive cells, suggesting that IL-1β could participate in an autocrine-paracrine fashion thereby modulating the pituitary population of ACTH-positive cells.
Collapse
Affiliation(s)
- María Carmen Iglesias-Osma
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Spain; Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain
| | - Enrique J Blanco
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain; Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Spain
| | | | - María José García-Barrado
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Spain; Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain
| | - Leonardo Catalano-Iniesta
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Spain; Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain
| | - Manuel Carretero
- Faculty of Human and Social Sciences, Pontifical University of Salamanca, Spain
| | - Virginia Sánchez-Robledo
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Spain; Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain
| | - José Carretero
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), and Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Spain; Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Spain.
| |
Collapse
|
10
|
|
11
|
Ai J, Wan H, Shu M, Zhou H, Zhao T, Fu W, He Y. Guhong injection protects against focal cerebral ischemia-reperfusion injury via anti-inflammatory effects in rats. Arch Pharm Res 2016; 40:610-622. [PMID: 27624481 DOI: 10.1007/s12272-016-0835-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 09/07/2016] [Indexed: 11/30/2022]
Abstract
Guhong injection (GHI), composed of aceglutamide and safflower aqueous extract, has been used clinically for the treatment of cerebrovascular diseases such as cerebral embolism, hemorrhage and mental deterioration. In this paper, we reported the results of the first study on the anti-inflammatory effects of GHI in murine focal cerebral ischemia-reperfusion (I/R) injury. Adult male SD rats were randomly divided into six groups: Sham group, I/R group, GHI-L group (2.5 mL/kg), GHI-M group (5 mL/kg), GHI-H group (10 mL/kg) and Nimodipine group. I/R injury was induced by middle cerebral artery occlusion (MCAO) for 1.5 h followed by reperfusion for 24 h. Compared with I/R group, rats treated with GHI showed dose dependent reductions in neurological defect scores and cerebral infarct volume. GHI obviously down-regulated nitric oxide (NO), inducible NO synthase (iNOS), myeloperoxidase (MPO), interleukin-1β (IL-1β), TNF-α (tumor necrosis factor-α) and C reactive protein (CRP) levels in serum. Moreover, histological examination by H&E staining showed that clear cell outline, less vacuolated spaces and largely surviving neurons were observed in GHI-treated rats. The immunohistochemical staining revealed that GHI administration significantly diminished the positive expressions of intercellular cell adhesion molecule-1 (ICAM-1) and nuclear factor-κB p65 (NF-κB p65) in brain tissues. Western blot analysis for ICAM, NF-κB p65 and iNOS further solidified the above findings. All these results demonstrate that GHI exerts a strong and ameliorative effect on cerebral I/R injury in rats possibly through the inhibition of inflammation.
Collapse
Affiliation(s)
- Jinchao Ai
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Haitong Wan
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Mingchun Shu
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Huifen Zhou
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Tao Zhao
- Buchang Pharmaceutical Co., Ltd., Xi'an, 710075, China
| | - Wei Fu
- Buchang Pharmaceutical Co., Ltd., Xi'an, 710075, China
| | - Yu He
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
12
|
Zhao B, Zou CJ, Zhou P. Delayed administration IL-1β neutralizing antibody improves cognitive function after transient global ischemia in rats. Behav Brain Res 2016; 303:53-60. [DOI: 10.1016/j.bbr.2016.01.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/06/2016] [Accepted: 01/10/2016] [Indexed: 12/23/2022]
|
13
|
Ma Y, Bu J, Dang H, Sha J, Jing Y, Shan-jiang AI, Li H, Zhu Y. Inhibition of adenosine monophosphate-activated protein kinase reduces glial cell-mediated inflammation and induces the expression of Cx43 in astroglias after cerebral ischemia. Brain Res 2015; 1605:1-11. [DOI: 10.1016/j.brainres.2014.11.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/25/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
|
14
|
PET Demonstrates Functional Recovery after Treatment by Danhong Injection in a Rat Model of Cerebral Ischemic-Reperfusion Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:430757. [PMID: 24707308 PMCID: PMC3953511 DOI: 10.1155/2014/430757] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 01/13/2014] [Accepted: 01/17/2014] [Indexed: 01/07/2023]
Abstract
This study aimed to investigate neuroprotection of Danhong injection (DHI) in a rat model of cerebral ischemia using 18F-fluorodeoxyglucose positron emission tomography (18F-FDG-PET). Method. Rats were divided into 5 groups: sham group, ischemia-reperfusion untreated (IRU) group, DHI-1 group (DHI 1 mL/kg/d), DHI-2 group (DHI 2 mL/kg/d), and DHI-4 group (DHI 4 mL/kg/d). AII the treated groups were intraperitoneally injected with DHI daily for 14 days. The therapeutic effects in terms of cerebral infarct volume, neurological function, and cerebral glucose metabolism were evaluated. Expression of TNF-α and IL-1β was detected with enzyme-linked immunosorbent assay (ELISA). Levels of mature neuronal marker (NeuN), glial marker (GFAP), vascular density factor (vWF), and glucose transporter 1 (GLUT1) were assessed by immunohistochemistry. Results. Compared with the IRU group, rats treated with DHI showed dose dependent reductions in cerebral infarct volume and levels of proinflammatory cytokines, improvement of neurological function, and recovery of cerebral glucose metabolism. Meanwhile, the significantly increased numbers of neurons, gliocytes, and vessels and the recovery of glucose utilization were found in the peri-infarct region after DHI treatment using immunohistochemical analysis. Conclusion. This study demonstrated the metabolic recovery after DHI treatment by micro-PET imaging with 18F-FDG and the neuroprotective effects of DHI in a rat model of cerebral ischemic-reperfusion injury.
Collapse
|
15
|
Noraini O, Jahwarhar NA, Sabri MY, Emikpe BO, Tanko PN, Latifah MH, Jamil S. The effect of heat stress on clinicopathological changes and immunolocalization of antigens in experimental Streptococcus agalactiae infection in Red hybrid tilapia (Oreochromis spp.). Vet World 2013. [DOI: 10.14202/vetworld.2013.997-1003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
16
|
Taşçi A, Okay O, Gezici AR, Ergün R, Ergüngör F. Prognostic value of interleukin-1 beta levels after acute brain injury. Neurol Res 2013; 25:871-4. [PMID: 14669533 DOI: 10.1179/016164103771953998] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Traumatic injury to central nervous system results in the production of inflammatory cytokines via intrinsic mechanisms by neurons, astrocytes and microglia, and extrinsic mechanisms by infiltrating macrophages, lymphocytes and other leukocytes. Interleukin-1 beta is the key mediator of the acute inflammatory host response. While this response is necessary for resolution of the pathologic event, the toxic nature of many of its products can cause significant tissue damage. We analyzed serum interleukin-1 beta levels by enzyme-linked immunosorbent assay in 48 patients with solitary head injury who were transported to our clinic immediately after trauma. We categorized the patients according to their initial Glasgow coma scores in three groups, and compared their serum interleukin-1 beta values both with their Glasgow coma initial and outcome scores. This study helped to provide quantitative data to estimate clinical impressions and prognosis after head injury.
Collapse
Affiliation(s)
- Alptekin Taşçi
- Department of Neurosurgery, Ankara Numune Hospital, Ankara, Turkey
| | | | | | | | | |
Collapse
|
17
|
Sildenafil enhances neurogenesis and oligodendrogenesis in ischemic brain of middle-aged mouse. PLoS One 2012; 7:e48141. [PMID: 23118941 PMCID: PMC3485244 DOI: 10.1371/journal.pone.0048141] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 09/19/2012] [Indexed: 11/19/2022] Open
Abstract
Adult neural stem cells give rise to neurons, oligodendrocytes and astrocytes. Aging reduces neural stem cells. Using an inducible nestin-CreERT2/R26R-yellow fluorescent protein (YFP) mouse, we investigated the effect of Sildenafil, a phosphodiesterase type 5 (PDE5) inhibitor, on nestin lineage neural stem cells and their progeny in the ischemic brain of the middle-aged mouse. We showed that focal cerebral ischemia induced nestin lineage neural stem cells in the subventricular zone (SVZ) of the lateral ventricles and nestin expressing NeuN positive neurons and adenomatous polyposis coli (APC) positive mature oligodendrocytes in the ischemic striatum and corpus callosum in the aged mouse. Treatment of the ischemic middle-aged mouse with Sildenafil increased nestin expressing neural stem cells, mature neurons, and oligodendrocytes by 33, 75, and 30%, respectively, in the ischemic brain. These data indicate that Sildenafil amplifies nestin expressing neural stem cells and their neuronal and oligodendrocyte progeny in the ischemic brain of the middle-aged mouse.
Collapse
|
18
|
Liu Y, Biarnés Costa M, Gerhardinger C. IL-1β is upregulated in the diabetic retina and retinal vessels: cell-specific effect of high glucose and IL-1β autostimulation. PLoS One 2012; 7:e36949. [PMID: 22615852 PMCID: PMC3353989 DOI: 10.1371/journal.pone.0036949] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 04/16/2012] [Indexed: 12/12/2022] Open
Abstract
Many molecular and cellular abnormalities detected in the diabetic retina support a role for IL-1β-driven neuroinflammation in the pathogenesis of diabetic retinopathy. IL-1β is well known for its role in the induction and, through autostimulation, amplification of neuroinflammation. Upregulation of IL-1β has been consistently detected in the diabetic retina; however, the mechanisms and cellular source of IL-1β overexpression are poorly understood. The aim of this study was to investigate the effect of high glucose and IL-1β itself on IL-1β expression in microglial, macroglial (astrocytes and Müller cells) and retinal vascular endothelial cells; and to study the effect of diabetes on the expression of IL-1β in isolated retinal vessels and on the temporal pattern of IL-1β upregulation and glial reactivity in the retina of streptozotocin-diabetic rats. IL-1β was quantified by RealTime RT-PCR and ELISA, glial fibrillar acidic protein, α2-macroglobulin, and ceruloplasmin by immunoblotting. We found that high glucose induced a 3-fold increase of IL-1β expression in retinal endothelial cells but not in macroglia and microglia. IL-1β induced its own synthesis in endothelial and macroglial cells but not in microglia. In retinal endothelial cells, the high glucose-induced IL-1β overexpression was prevented by calphostin C, a protein kinase C inhibitor. The retinal vessels of diabetic rats showed increased IL-1β expression as compared to non-diabetic rats. Retinal expression of IL-1β increased early after the induction of diabetes, continued to increase with progression of the disease, and was temporally associated with upregulation of markers of glial activation. These findings point to hyperglycemia as the trigger and to the endothelium as the origin of the initial retinal upregulation of IL-1β in diabetes; and to IL-1β itself, via autostimulation in endothelial and macroglial cells, as the mechanism of sustained IL-1β overexpression. Interrupting the vicious circle triggered by IL-1β autostimulation could limit the progression of diabetic retinopathy.
Collapse
Affiliation(s)
- Yang Liu
- Schepens Eye Research Institute and the Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Montserrat Biarnés Costa
- Schepens Eye Research Institute and the Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Chiara Gerhardinger
- Schepens Eye Research Institute and the Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
19
|
Tang NY, Liu CH, Hsieh CT, Hsieh CL. The Anti-Inflammatory Effect of Paeoniflorin on Cerebral Infarction Induced by Ischemia-Reperfusion Injury in Sprague-Dawley Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 38:51-64. [PMID: 20128044 DOI: 10.1142/s0192415x10007786] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Paeoniflorin, a component in Paeonia lactiflora Pall, inhibits nuclear factor-κB expression in chronic hypoperfusion rat and has anti-inflammatory properties. Therefore, the aim of the present study was to investigate the effect of paeoniflorin on cerebral infarct, and the involvement of anti-inflammation. We established an animal model of cerebral infarct by occluding both the common carotid arteries and the right middle cerebral artery for 90 min, followed by reperfusion of 24 hours. The ratios of cerebral infarction area to total brain area, and neuro-deficit score were used as an index to observe the effects of paeoniflorin on cerebral infarct. ED1 (mouse anti rat CD68), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), intercellular adhesion molecular-1 (ICAM-1), myeloperoxidase (MPO) immunostaining and apoptotic cells in the cerebral infarction region also were studied. The results indicated that both pre-treatment and post-treatment with paeoniflorin reduced the ratio of cerebral infarction area; pre-treatment with paeoniflorin also reduced the neurological deficit score. The counts of ED1, IL-1β, TNF-α, ICAM-1 of microvessels and MPO immunoreactive cells and apoptotic cells were increased in the cerebral infarction region; however, these increases were reduced by Paeoniflorin pre-treatment. In conclusion, Paeoniflorin reduced cerebral infarct and neurological deficit in ischemia-reperfusion injured rats, suggesting that paeoniflorin may have a similar effect in humans and might be a suitable treatment for stroke. Paeoniflorin reduced cerebral infarct, at least in part, involves the anti-inflammatory properties.
Collapse
Affiliation(s)
- Nou-Ying Tang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chung-Hsiang Liu
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | - Ching-Tou Hsieh
- Department of Internal Medicine, Jen-Ai Hospital, Taichung, Taiwan
| | - Ching-Liang Hsieh
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Acupuncture Research Center, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
20
|
Frentzou GA, Bradford C, Harkness KA, Haddock G, Woodroofe MN, Cross AK. IL-1β down-regulates ADAMTS-13 mRNA expression in cells of the central nervous system. J Mol Neurosci 2011; 46:343-51. [PMID: 21732076 DOI: 10.1007/s12031-011-9591-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 06/22/2011] [Indexed: 01/06/2023]
Abstract
ADAMTS-13 is the Von Willebrand factor (vWF) cleaving protease, responsible for the cleavage and down-regulation of the pro-thrombotic properties of ultra large VWF multimers. It is expressed predominantly by the hepatic stellate cells of the liver, but is also found to be expressed in other tissues, including brain. Reduced ADAMTS-13 is associated with a variety of thrombotic microangiopathies. Since the cellular origin and regulation of ADAMTS-13 expression in the brain is unknown, we aimed to investigate this in four different central nervous system (CNS)-derived cell lines, SHSY-5Y (human neuroblastoma), U373 (human astroglioma), CHME-3 (human foetal microglia) and hCMEC/D3 (adult human brain endothelial cells). All cell lines expressed ADAMTS-13 mRNA constitutively with neuroblastoma cells showing the highest expression. Interleukin (IL)-1β down-regulated ADAMTS-13 mRNA expression in astroglioma cells and microglial cells whereas TNF and IL-6 treatment showed no significant differences in ADAMTS-13 mRNA expression in any cell line tested. ADAMTS-13 protein expression was reduced in a dose-dependent manner only in astroglioma cells following stimulation by IL-1β. The ability of IL-1β to significantly reduce ADAMTS-13 mRNA expression in human microglia and astroglioma cells suggests a role in the haemostasis of the local microenvironment under inflammatory conditions. This is the first report of ADAMTS-13 expression in cells of the CNS; however, its function remains to be determined.
Collapse
Affiliation(s)
- G Alkistis Frentzou
- Biomedical Research Centre, Faculty of Health and Wellbeing, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK.
| | | | | | | | | | | |
Collapse
|
21
|
Yao J, Xu Y, Ji F, Wang C, Zhang Y, Ni J, Wang R. Protective effects of MLIF analogs on cerebral ischemia-reperfusion injury in rats. Peptides 2011; 32:1047-54. [PMID: 21402115 DOI: 10.1016/j.peptides.2011.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 03/03/2011] [Accepted: 03/03/2011] [Indexed: 11/18/2022]
Abstract
The monocyte locomotion inhibitory factor (MLIF) is an anti-inflammatory oligopeptide produced by Entamoeba histolytica. Among its different effects, it inhibits locomotion of human monocytes, hence its original name. The carboxyl-terminal end group Cys-Asn-Ser is the pharmacophore of anti-inflammatory peptide Met-Gln-Cys-Asn-Ser. In this study, the N-terminal of Cys-Asn-Ser was modified. With the aim to enhance the antioxidant ability and penetrability of Cys-Asn-Ser, we designed and synthesized two tetrapeptides Tyr-Cys-Asn-Ser and His-Cys-Asn-Ser. The neuroprotective effects of Tyr-Cys-Asn-Ser and His-Cys-Asn-Ser on focal ischemia reperfusion were investigated, and their pharmacological activities compared with Cys-Asn-Ser were studied. In order to study the mechanism of neuroprotective effect of these peptides, the level of oxidative stress markers malondialdehyde (MDA) and superoxide dismutase (SOD) and pro-inflammatory factors interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and myeloperoxidase (MPO) were detected in brain tissue homogenate.
Collapse
Affiliation(s)
- Jia Yao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, 222 Tian Shui South Road, Lanzhou 730000, PR China
| | | | | | | | | | | | | |
Collapse
|
22
|
Lee JS, Choi HS, Kang SW, Chung JH, Park HK, Ban JY, Kwon OY, Hong HP, Ko YG. Therapeutic effect of Korean red ginseng on inflammatory cytokines in rats with focal cerebral ischemia/reperfusion injury. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2011; 39:83-94. [PMID: 21213400 DOI: 10.1142/s0192415x1100866x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study aims to identify the therapeutic effect of Korean red ginseng (KRG) on the expression of inflammatory cytokines in rats with focal cerebral ischemia/reperfusion injury. Adult male Sprague-Dawley rats were subjected to transient middle cerebral artery occlusion (tMCAO) for two hours. They were fed KRG extract (100 mg/kg/day per orally) or saline after reperfusion. Tests for neurological deficits, using the modified neurologic severity score and the corner turn test, were performed before the ischemic event, and one, three, and seven days after tMCAO. Serum levels of cytokines were measured three and seven days after the operation, using enzyme-linked immunosorbent assays. The infarct volume was assessed after seven days by staining brain tissue with 2% 2, 3, 5-triphenyltetrazolium chloride. Oral administration of KRG significantly reduced the infarct volumes and rapidly improved neurological deficits. Serum levels of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and IL-6 were higher in tMCAO-operated rats than in the sham-operated rats. These changes were attenuated by daily KRG intake for seven days. Serum IL-10 levels were significantly increased in KRG-fed rats, as compared to sham-operated and saline-fed rats. Our results suggested that KRG provides neuroprotection for rats with focal cerebral ischemia/reperfusion injury. This neuroprotection may be due to raised IL-10 expression and a reduction in the serum levels of TNF-α, IL-1β, and IL-6.
Collapse
Affiliation(s)
- Jong Seok Lee
- Department of Emergency Medicine, School of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Zhang RL, Chopp M, Roberts C, Jia L, Wei M, Lu M, Wang X, Pourabdollah S, Zhang ZG. Ascl1 lineage cells contribute to ischemia-induced neurogenesis and oligodendrogenesis. J Cereb Blood Flow Metab 2011; 31:614-25. [PMID: 20736965 PMCID: PMC3049516 DOI: 10.1038/jcbfm.2010.134] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/06/2010] [Accepted: 07/08/2010] [Indexed: 11/08/2022]
Abstract
Neural and oligodendrocyte progenitor cells in the adult brain express Ascl1 (also known as Mash1), a basic helix-loop-helix transcription factor. We examined the progeny and fate of this progenitor population in adult male Ascl1-CreER(TM);R26R-stop-yellow fluorescent protein mice subjected to right middle cerebral occlusion over 60 days after stroke using inducible Cre recombination to label Ascl1-expressing cells at poststroke days 2 to 6 in vivo. Seven days after stroke, a substantial increase in Ascl1 lineage cells was detected in the ipsilateral subventricular zone (SVZ), striatum, and corpus callosum. These cells exhibited proliferating progenitor cell phenotypes (Sox2(+), BrdU(+), and Ki67(+)). Although Ascl1 lineage cells in the ipsilateral SVZ gradually decreased during 14 to 60 days after stroke, Ascl1 lineage cells in the ischemic striatum revealed a remarkable increase during this period. Thirty and sixty days after stroke, Ascl1 lineage cells in the ischemic striatum gave rise to GABAergic neurons and mature oligodendrocytes. In contrast, none of the Ascl1 lineage cells in the contralateral striatum exhibited neuronal and oligodendrocyte phenotypes. Moreover, Ascl1 lineage cells in the corpus callosum were only fated to become mature oligodendrocytes. Our data suggest that Ascl1 lineage cells contribute to stroke-induced neurogenesis and oligodendrogenesis in the adult ischemic brain.
Collapse
Affiliation(s)
- Rui Lan Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Cindi Roberts
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Longfei Jia
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Min Wei
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Mei Lu
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Xinli Wang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | | | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
24
|
Ye XH, Wu Y, Guo PP, Wang J, Yuan SY, Shang Y, Yao SL. Lipoxin A4 analogue protects brain and reduces inflammation in a rat model of focal cerebral ischemia reperfusion. Brain Res 2010; 1323:174-83. [DOI: 10.1016/j.brainres.2010.01.079] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2009] [Revised: 01/26/2010] [Accepted: 01/28/2010] [Indexed: 12/30/2022]
|
25
|
Identification of distinct cellular pools of interleukin-1β during the evolution of the neuroinflammatory response induced by transient middle cerebral artery occlusion in the brain of rat. Brain Res 2010; 1313:259-69. [DOI: 10.1016/j.brainres.2009.12.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 12/01/2009] [Accepted: 12/04/2009] [Indexed: 11/23/2022]
|
26
|
Oruckaptan HH, Ozisik P, Atilla P, Tuncel M, Kilinc K, Geyik PO, Basaran N, Yüksel E, Ozcan OE. Systemic Administration of Interleukin-10 Attenuates Early Ischemic Response Following Spinal Cord Ischemia Reperfusion Injury in Rats. J Surg Res 2009; 155:345-56. [DOI: 10.1016/j.jss.2008.07.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 06/15/2008] [Accepted: 07/11/2008] [Indexed: 10/21/2022]
|
27
|
Amantea D, Nappi G, Bernardi G, Bagetta G, Corasaniti MT. Post-ischemic brain damage: pathophysiology and role of inflammatory mediators. FEBS J 2009; 276:13-26. [PMID: 19087196 DOI: 10.1111/j.1742-4658.2008.06766.x] [Citation(s) in RCA: 334] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neuroinflammatory mediators play a crucial role in the pathophysiology of brain ischemia, exerting either deleterious effects on the progression of tissue damage or beneficial roles during recovery and repair. Within hours after the ischemic insult, increased levels of cytokines and chemokines enhance the expression of adhesion molecules on cerebral endothelial cells, facilitating the adhesion and transendothelial migration of circulating neutrophils and monocytes. These cells may accumulate in the capillaries, further impairing cerebral blood flow, or extravasate into the brain parenchyma. Infiltrating leukocytes, as well as resident brain cells, including neurons and glia, may release pro-inflammatory mediators, such as cytokines, chemokines and oxygen/nitrogen free radicals that contribute to the evolution of tissue damage. Moreover, recent studies have highlighted the involvement of matrix metalloproteinases in the propagation and regulation of neuroinflammatory responses to ischemic brain injury. These enzymes cleave protein components of the extracellular matrix such as collagen, proteoglycan and laminin, but also process a number of cell-surface and soluble proteins, including receptors and cytokines such as interleukin-1beta. The present work reviewed the role of neuroinflammatory mediators in the pathophysiology of ischemic brain damage and their potential exploitation as drug targets for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Diana Amantea
- Department of Pharmacobiology, University of Calabria, Rende (CS), Italy.
| | | | | | | | | |
Collapse
|
28
|
Vemula S, Roder KE, Yang T, Bhat GJ, Thekkumkara TJ, Abbruscato TJ. A functional role for sodium-dependent glucose transport across the blood-brain barrier during oxygen glucose deprivation. J Pharmacol Exp Ther 2009; 328:487-95. [PMID: 18981287 PMCID: PMC2630371 DOI: 10.1124/jpet.108.146589] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 10/30/2008] [Indexed: 11/22/2022] Open
Abstract
In the current study, we determined the functional significance of sodium-dependent/-independent glucose transporters at the neurovasculature during oxygen glucose deprivation (OGD). Confluent brain endothelial cells cocultured with astrocytes were exposed to varying degrees of in vitro stroke conditions. Glucose transporter (GLUT) 1 and sodium glucose cotransporter (SGLT) activity were investigated by luminal membrane uptake and transport studies using [(3)H]D-glucose and also by [(14)C]alpha-methyl D-glucopyranoside (AMG), a specific, nonmetabolized substrate of SGLT. In vivo middle cerebral artery occlusion experiments were tested to determine whether blood-brain barrier (BBB) SGLT activity was induced during ischemia. Increases in luminal D-glucose and AMG uptake and transport were observed with in vitro stroke conditions. Specific inhibitor experiments suggest a combined role for both SGLT and GLUT1 at the BBB during OGD. A time-dependent increase in the uptake of AMG was also seen in mice exposed to permanent focal ischemia, and this increase was sensitive to the SGLT inhibitor, phlorizin. Infarct and edema ratio during ischemia were significantly decreased by the inhibition of this transporter. These results show that both GLUT1 and SGLT play a role at the BBB in the blood-to-brain transport of glucose during ischemic conditions, and inhibition of SGLT during stroke has the potential to improve stroke outcome. Pharmacological modulation of this novel BBB transporter could prove to be a brain vascular target in stroke.
Collapse
Affiliation(s)
- Sharanya Vemula
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79016, USA
| | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Fogal B, Hewett SJ. Interleukin-1beta: a bridge between inflammation and excitotoxicity? J Neurochem 2008; 106:1-23. [PMID: 18315560 DOI: 10.1111/j.1471-4159.2008.05315.x] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Interleukin-1 (IL-1) is a proinflammatory cytokine released by many cell types that acts in both an autocrine and/or paracrine fashion. While IL-1 is best described as an important mediator of the peripheral immune response during infection and inflammation, increasing evidence implicates IL-1 signaling in the pathogenesis of several neurological disorders. The biochemical pathway(s) by which this cytokine contributes to brain injury remain(s) largely unidentified. Herein, we review the evidence that demonstrates the contribution of IL-1beta to the pathogenesis of both acute and chronic neurological disorders. Further, we highlight data that leads us to propose IL-1beta as the missing mechanistic link between a potential beneficial inflammatory response and detrimental glutamate excitotoxicity.
Collapse
Affiliation(s)
- Birgit Fogal
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
31
|
Nakka VP, Gusain A, Mehta SL, Raghubir R. Molecular mechanisms of apoptosis in cerebral ischemia: multiple neuroprotective opportunities. Mol Neurobiol 2008; 37:7-38. [PMID: 18066503 DOI: 10.1007/s12035-007-8013-9] [Citation(s) in RCA: 216] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Accepted: 11/05/2007] [Indexed: 12/18/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury triggers multiple and distinct but overlapping cell signaling pathways, which may lead to cell survival or cell damage. There is overwhelming evidence to suggest that besides necrosis, apoptosis do contributes significantly to the cell death subsequent to I/R injury. Both extrinsic and intrinsic apoptotic pathways play a vital role, and upon initiation, these pathways recruit downstream apoptotic molecules to execute cell death. Caspases and Bcl-2 family members appear to be crucial in regulating multiple apoptotic cell death pathways initiated during I/R. Similarly, inhibitor of apoptosis family of proteins (IAPs), mitogen-activated protein kinases, and newly identified apoptogenic molecules, like second mitochondrial-activated factor/direct IAP-binding protein with low pI (Smac/Diablo), omi/high-temperature requirement serine protease A2 (Omi/HtrA2), X-linked mammalian inhibitor of apoptosis protein-associated factor 1, and apoptosis-inducing factor, have emerged as potent regulators of cellular apoptotic/antiapoptotic machinery. All instances of cell survival/death mechanisms triggered during I/R are multifaceted and interlinked, which ultimately decide the fate of brain cells. Moreover, apoptotic cross-talk between major subcellular organelles suggests that therapeutic strategies should be optimally directed at multiple targets/mechanisms for better therapeutic outcome. Based on the current knowledge, this review briefly focuses I/R injury-induced multiple mechanisms of apoptosis, involving key apoptotic regulators and their emerging roles in orchestrating cell death programme. In addition, we have also highlighted the role of autophagy in modulating cell survival/death during cerebral ischemia. Furthermore, an attempt has been made to provide an encouraging outlook on emerging therapeutic approaches for cerebral ischemia.
Collapse
Affiliation(s)
- Venkata Prasuja Nakka
- Division of Pharmacology, Central Drug Research Institute, Chatter Manzil Palace, POB-173, Lucknow, 226001, India
| | | | | | | |
Collapse
|
32
|
Fogal B, Li J, Lobner D, McCullough LD, Hewett SJ. System x(c)- activity and astrocytes are necessary for interleukin-1 beta-mediated hypoxic neuronal injury. J Neurosci 2007; 27:10094-105. [PMID: 17881516 PMCID: PMC6672668 DOI: 10.1523/jneurosci.2459-07.2007] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The purpose of this study was to elucidate the cellular/biochemical pathway(s) by which interleukin-1beta (IL-1beta) contributes to the pathogenesis of hypoxic-ischemic brain damage. In vivo, IL-1 receptor type I (IL-1RI)-deficient mice showed smaller infarcts and less neurological deficits than wild-type animals after a 90 min reversible middle cerebral artery occlusion. In vitro, IL-1beta mediated an enhancement of hypoxic neuronal injury in murine cortical cultures that was lacking in cultures derived from IL-1RI null mutant animals and was blocked by the IL-1 receptor antagonist or an IL-1RI blocking antibody. This IL-1beta-mediated potentiation of hypoxic neuronal injury was associated with an increase in both cellular cystine uptake ([cystine]i) and extracellular glutamate levels ([glutamate]e) and was prevented by either ionotropic glutamate receptor antagonism or removal of L-cystine, suggesting a role for the cystine/glutamate antiporter (System x(c)-). Indeed, dual System x(c)-/metabotropic glutamate receptor subunit 1 (mGluR1) antagonism but not selective mGluR1 antagonism prevented neuronal injury. Additionally, cultures derived from mGluR1-deficient mice exhibited the same potentiation in injury after treatment with IL-1beta as wild-type cultures, an effect prevented by System x(c)-/mGluR1 antagonism. Finally, assessment of System x(c)- function and kinetics in IL-1beta-treated cultures revealed an increase in velocity of cystine transport (Vmax), in the absence of a change in affinity (Km). Neither the enhancement in [cystine]i, [glutamate]e, or neuronal injury were observed in chimeric cultures consisting of IL-1RI(+/+) neurons plated on top of IL-1RI(-/-) astrocytes, highlighting the importance of astrocyte-mediated alterations in System x(c)- as a novel contributor to the development and progression of hypoxic neuronal injury.
Collapse
Affiliation(s)
| | - Jun Li
- Neurology, University of Connecticut Health Center, Farmington, Connecticut 06030, and
| | - Doug Lobner
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53233
| | - Louise D. McCullough
- Neurology, University of Connecticut Health Center, Farmington, Connecticut 06030, and
| | | |
Collapse
|
33
|
Kim SJ, Jeong HJ, Lee KM, Moon PD, Yun JM, Cho KH, Moon BS, Lee HJ, Hong SH, Kim HM, Um JY. The effect of SHJKS on cytokines production and NF-kappaB activation in the peripheral blood mononuclear cells of patients with cerebral infarction. Immunopharmacol Immunotoxicol 2007; 28:557-70. [PMID: 17190734 DOI: 10.1080/08923970601066930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The Korean genuine medicine "Seonghyangjeongkisan" (SHJKS) has long been used for various cerebrovascular diseases. However, very little scientific investigation has been carried out. Cytokines involved in the regulation of inflammatory reactions and immune responses may play a role in the pathogenesis of cerebral infarction (CI). The aim of the present study is to elucidate how SHJKS modulates the inflammatory reaction in lipopolysaccaride (LPS) plus phytohaemagglutinin (PHA)-stimulated peripheral mononuclear cells (PBMCs) from CI patients. The amount of tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, and IL-8 in PBMC culture supernatant was significantly increased in the LPS plus PHA treated cells compared to unstimulated cells. SHJKS inhibited the TNF-alpha, IL-1beta, IL-6, and IL-8 production in dose dependent manner. Maximal inhibition rate of the TNF-alpha, IL-1beta, IL-6, and IL-8 by SHJGS (1.0 mg/ml) was 68.01 +/- 0.28% (P < 0.01), 52.11 +/- 0.56 % (P < 0.01), 53.42 +/- 0.46 % (P < 0.01), and 46.70 +/- 0.37% (P < 0.05), respectively. In addition, we show that SHJKS suppressed nuclear factor (NF)-kappaB activation induced by LPS plus PHA, leading to suppression of IkappaB-alpha phosphorylation and degradation. These results suggest that SHJKS might have regulatory effects on LPS plus PHA-induced cytokine production and NF-kappaB activation, which might explain its beneficial effect in the treatment of CI.
Collapse
Affiliation(s)
- Su-Jin Kim
- Department of Pharmacology, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mehta SL, Manhas N, Raghubir R. Molecular targets in cerebral ischemia for developing novel therapeutics. ACTA ACUST UNITED AC 2007; 54:34-66. [PMID: 17222914 DOI: 10.1016/j.brainresrev.2006.11.003] [Citation(s) in RCA: 540] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 11/09/2006] [Accepted: 11/10/2006] [Indexed: 11/20/2022]
Abstract
Cerebral ischemia (stroke) triggers a complex series of biochemical and molecular mechanisms that impairs the neurologic functions through breakdown of cellular integrity mediated by excitotoxic glutamatergic signalling, ionic imbalance, free-radical reactions, etc. These intricate processes lead to activation of signalling mechanisms involving calcium/calmodulin-dependent kinases (CaMKs) and mitogen-activated protein kinases (MAPKs) such as extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK). The distribution of these transducers bring them in contact with appropriate molecular targets leading to altered gene expression, e.g. ERK and JNK mediated early gene induction, responsible for activation of cell survival/damaging mechanisms. Moreover, inflammatory reactions initiated at the neurovascular interface and alterations in the dynamic communication between the endothelial cells, astrocytes and neurons are thought to substantially contribute to the pathogenesis of the disease. The damaging mechanisms may proceed through rapid nonspecific cell lysis (necrosis) or by active form of cell demise (apoptosis or necroptosis), depending upon the severity and duration of the ischemic insult. A systematic understanding of these molecular mechanisms with prospect of modulating the chain of events leading to cellular survival/damage may help to generate the potential strategies for neuroprotection. This review briefly covers the current status on the molecular mechanisms of stroke pathophysiology with an endeavour to identify potential molecular targets such as targeting postsynaptic density-95 (PSD-95)/N-methyl-d-aspartate (NMDA) receptor interaction, certain key proteins involved in oxidative stress, CaMKs and MAPKs (ERK, p38 and JNK) signalling, inflammation (cytokines, adhesion molecules, etc.) and cell death pathways (caspases, Bcl-2 family proteins, poly (ADP-ribose) polymerase-1 (PARP-1), apoptosis-inducing factor (AIF), inhibitors of apoptosis proteins (IAPs), heat shock protein 70 (HSP70), receptor interacting protein (RIP), etc., besides targeting directly the genes itself. However, selecting promising targets from various signalling cascades, for drug discovery and development is very challenging, nevertheless such novel approaches may lead to the emergence of new avenues for therapeutic intervention in cerebral ischemia.
Collapse
Affiliation(s)
- Suresh L Mehta
- Division of Pharmacology, Central Drug Research Institute, Chatter Manzil Palace, POB-173, Lucknow-226001, India
| | | | | |
Collapse
|
35
|
Chiappetta O, Gliozzi M, Siviglia E, Amantea D, Morrone LA, Berliocchi L, Bagetta G, Corasaniti MT. Evidence to Implicate Early Modulation of Interleukin‐1β Expression in the Neuroprotection Afforded by 17β‐Estradiol in Male Rats Undergone Transient Middle Cerebral Artery Occlusion. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 82:357-72. [PMID: 17678971 DOI: 10.1016/s0074-7742(07)82019-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Neuroprotection exerted by 17beta-estradiol (17beta-E(2)) has been widely investigated in animal models of acute cerebral ischemia. Estrogens interact with intracellular receptors (ERalpha and ERbeta) to modulate the transcription of target genes, including those implicated in neuronal survival. Neuroprotection may also occur via interaction with ER-like membrane receptors mediating rapid, non-genomic, actions or via receptor-independent mechanisms. There is also evidence that blockade of inflammatory factors may represent an important mechanism involved in estrogenic neuroprotection. Here we investigate whether reduced brain damage by acute pharmacological treatment with 17beta-E(2) in male rats subjected to transient (2h) middle cerebral artery occlusion (tMCAo) involves modulation of interleukin-1beta (IL-1beta), a proinflammatory cytokine strongly implicated in the pathophysiology of ischemic stroke. Administration of 17beta-E(2) (0.2mg/kg, i.p., 1h before tMCAo) results in significant reduction of brain infarct volume, and this is reverted by the ER antagonist ICI 182,780 (0.25mg/kg, i.p.) administered 1h before 17beta-E(2). Two hours MCAo followed by 2-h reperfusion results in a significant, threefold increase of IL-1beta levels in the cortical tissue ipsilateral to the ischemic damage. Interestingly, a pretreatment with a neuroprotective dose of 17beta-E(2) attenuates the cytokine elevation and this appears to occur through ER activation. In addition, neuroprotection by 17beta-E(2) is accompanied by reduced cytochrome c translocation both in the striatum and in the cortex as revealed by Western blotting 3h after reperfusion. In conclusion, we report the original observation that neuroprotection exerted by 17beta-E(2) in a rat model of transient focal brain ischemia is accompanied by reduced cytochrome c translocation to the cytosol and involves early modulation of IL-1beta production.
Collapse
Affiliation(s)
- Olga Chiappetta
- Department of Pharmacobiology, University of Calabria, Via P. Bucci 87036 Arcavacata di Rende (CS), Italy
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Amantea D, Russo R, Gliozzi M, Fratto V, Berliocchi L, Bagetta G, Bernardi G, Corasaniti MT. Early Upregulation of Matrix Metalloproteinases Following Reperfusion Triggers Neuroinflammatory Mediators in Brain Ischemia in Rat. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 82:149-69. [PMID: 17678960 DOI: 10.1016/s0074-7742(07)82008-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Abnormal expression of matrix metalloproteinases (MMPs) has been implicated in the pathophysiology of neuroinflammatory processes that accompany most central nervous system disease. In particular, early upregulation of the gelatinases MMP-2 and MMP-9 has been shown to contribute to disruption of the blood-brain barrier and to death of neurons in ischemic stroke. In situ zymography reveals a significant increase in gelatinolytic MMPs activity in the ischemic brain hemisphere after 2-h middle cerebral artery occlusion (MCAo) followed by 2-h reperfusion in rat. Accordingly, gel zymography demonstrates that expression and activity of MMP-2 and MMP-9 are enhanced in cortex and striatum ipsilateral to the ischemic insult. The latter effect appears to be instrumental for development of delayed brain damage since administration of a broad spectrum, highly specific MMPs inhibitor, GM6001, but not by its negative control, results in a significant (50%) reduction in ischemic brain volume. Increased gelatinase activity in the ischemic cortex coincides with elevation (166% vs sham) of mature interleukin-1beta (IL-1beta) after 2-h reperfusion and this does not appear to implicate a caspase-1-dependent processing of pro(31kDa)-IL-1beta to yield mature (17kDa) IL-1beta. More importantly, when administered at a neuroprotective dose GM6001 abolishes the early IL-1beta increase in the ischemic cortex and reduces the cleavage of the cytokine proform supporting the deduction that MMPs may initiate IL-1beta processing. In conclusion, development of tissue damage that follows transient ischemia implicates a crucial interplay between MMPs and mediators of neuroinflammation (e.g., IL-1beta), and this further underscores the therapeutic potential of MMPs inhibitors in the treatment of stroke.
Collapse
Affiliation(s)
- Diana Amantea
- Department of Pharmacobiology, UCHAD Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University of Calabria, 87036 Rende, Italy
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Konsman JP, Drukarch B, Van Dam AM. (Peri)vascular production and action of pro-inflammatory cytokines in brain pathology. Clin Sci (Lond) 2006; 112:1-25. [PMID: 17132137 DOI: 10.1042/cs20060043] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In response to tissue injury or infection, the peripheral tissue macrophage induces an inflammatory response through the release of IL-1β (interleukin-1β) and TNFα (tumour necrosis factor α). These cytokines stimulate macrophages and endothelial cells to express chemokines and adhesion molecules that attract leucocytes into the peripheral site of injury or infection. The aims of the present review are to (i) discuss the relevance of brain (peri)vascular cells and compartments to bacterial meningitis, HIV-1-associated dementia, multiple sclerosis, ischaemic and traumatic brain injury, and Alzheimer's disease, and (ii) to provide an overview of the production and action of pro-inflammatory cytokines by (peri)vascular cells in these pathologies of the CNS (central nervous system). The brain (peri)vascular compartments are highly relevant to pathologies affecting the CNS, as infections are almost exclusively blood-borne. Insults disrupt blood and energy flow to neurons, and active brain-to-blood transport mechanisms, which are the bottleneck in the clearance of unwanted molecules from the brain. Perivascular macrophages are the most reactive cell type and produce IL-1β and TNFα after infection or injury to the CNS. The main cellular target for IL-1β and TNFα produced in the brain (peri)vascular compartment is the endothelium, where these cytokines induce the expression of adhesion molecules and promote leucocyte infiltration. Whether this and other effects of IL-1 and TNF in the brain (peri)vascular compartments are detrimental or beneficial in neuropathology remains to be shown and requires a clear understanding of the role of these cytokines in both damaging and repair processes in the CNS.
Collapse
Affiliation(s)
- Jan P Konsman
- Laboratory of Integrative Neurobiology, CNRS FRE 2723/INRA UR 1244/University Bordeaux2, Institut François Magendie, Bordeaux, France
| | | | | |
Collapse
|
38
|
Mitsios N, Gaffney J, Kumar P, Krupinski J, Kumar S, Slevin M. Pathophysiology of Acute Ischaemic Stroke: An Analysis of Common Signalling Mechanisms and Identification of New Molecular Targets. Pathobiology 2006; 73:159-75. [PMID: 17119345 DOI: 10.1159/000096017] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Accepted: 07/17/2006] [Indexed: 12/18/2022] Open
Abstract
Stroke continues to be a major cause of death and disability. The currently available therapies have proven to be highly unsatisfactory (except thrombolysis) and attempts are being made to identify and characterize signalling proteins which could be exploited to design novel therapeutic modalities. The pathophysiology of stroke is a complex process. Delaying interventions from the first hours to days or even weeks following blood vessel occlusion may lead to worsening or impairment of recovery in later stages. The objective of this review is to critically evaluate the major mechanisms underlying stroke pathophysiology, especially the role of cell signalling in excitotoxicity, inflammation, apoptosis, neuroprotection and angiogenesis, and highlight potential novel targets for drug discovery.
Collapse
Affiliation(s)
- N Mitsios
- Department of Biological Sciences, Manchester Metropolitan University, Manchester, UK
| | | | | | | | | | | |
Collapse
|
39
|
Huang J, Upadhyay UM, Tamargo RJ. Inflammation in stroke and focal cerebral ischemia. ACTA ACUST UNITED AC 2006; 66:232-45. [PMID: 16935624 DOI: 10.1016/j.surneu.2005.12.028] [Citation(s) in RCA: 496] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Accepted: 12/26/2005] [Indexed: 10/24/2022]
Abstract
BACKGROUND A growing number of recent investigations have established a critical role for leukocytes in propagating tissue damage after ischemia and reperfusion in stroke. Experimental data obtained from animal models of middle cerebral artery occlusion implicate inflammatory cell adhesion molecules, chemokines, and cytokines in the pathogenesis of this ischemic damage. METHODS Data from recent animal and human studies were reviewed to demonstrate that inflammatory events occurring at the blood-endothelium interface of the cerebral capillaries underlie the resultant ischemic tissue damage. RESULTS After arterial occlusion, the up-regulated expression of cytokines including IL-1, and IL-6 act upon the vascular endothelium to increase the expression of intercellular adhesion molecule-1, P-selectin, and E-selectin, which promote leukocyte adherence and accumulation. Integrins then serve to structurally modify the basal lamina and extracellular matrix. These inflammatory signals then promote leukocyte transmigration across the endothelium and mediate inflammatory cascades leading to further cerebral infarction. CONCLUSIONS Inflammatory interactions that occur at the blood-endothelium interface, involving cytokines, adhesion molecules, chemokines and leukocytes, are critical to the pathogenesis of tissue damage in cerebral infarction. Exploring these pathophysiological mechanisms underlying ischemic tissue damage may direct rational drug design in the therapeutic treatment of stroke.
Collapse
Affiliation(s)
- Judy Huang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
40
|
Lai AY, Todd KG. Microglia in cerebral ischemia: molecular actions and interactions. Can J Physiol Pharmacol 2006; 84:49-59. [PMID: 16845890 DOI: 10.1139/y05-143] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The precise role of microglia in stroke and cerebral ischemia has been the subject of debate for a number of years. Microglia are capable of synthesizing numerous soluble and membrane-bound biomolecules, some known to be neuroprotective, some neurotoxic, whereas others have less definitive bioactivities. The molecular mechanisms through which microglia activate these molecules have thus become an important area of ischemia research. Here we provide a survey review that summarizes the key actions of microglial factors in cerebral ischemia including complement proteins, chemokines, pro-inflammatory cytokines, neurotrophic factors, hormones, and proteinases, as well several important messenger molecules that play a part in how these factors respond to extracellular signals during ischemic injuries. We also provide some new perspectives on how microglial intracellular signaling may contribute to the seemingly contradictory roles of several microglial effector molecules.
Collapse
Affiliation(s)
- Aaron Y Lai
- Neurochemical Research Unit, Department of Psychiatry and Centre for Neuroscience, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
41
|
Hsieh CL, Cheng CY, Tsai TH, Lin IH, Liu CH, Chiang SY, Lin JG, Lao CJ, Tang NY. Paeonol reduced cerebral infarction involving the superoxide anion and microglia activation in ischemia-reperfusion injured rats. JOURNAL OF ETHNOPHARMACOLOGY 2006; 106:208-15. [PMID: 16458462 DOI: 10.1016/j.jep.2005.12.027] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Revised: 12/15/2005] [Accepted: 12/15/2005] [Indexed: 05/06/2023]
Abstract
Both Moutan cortex of Paeonia suffruticosa Andrews (MC) and the root of Paeonia lactiflora Pall (PL) are important Traditional Chinese herbs used commonly to treat inflammatory and pyretic disorders. Paeonol, a common component of MC causes anti-platelet aggregation and scavenges free radicals. Therefore, the aim of the present study is to investigate the effects of Paeonol on cerebral infarct. A total of 60 male Sprague-Dawley (SD) rats were studied. An animal model of cerebral infarct was established by occluding both common carotid arteries and the right middle cerebral artery for 90 min, followed by a 24 h period of reperfusion. The percentage of cerebral infarction area to total brain area in each piece of brain tissue, and neuro-deficit score were measured. Superoxide anion was determined by the number of lucigenin-chemiluminescence (CL) counts. ED1 (mouse anti rat CD68) and interleukin-1beta (IL-1beta) immunostaining in the cerebral infarction region were also investigated for activation of microglia. The results indicated that Paeonol 15 and 20 mg/kg pretreatment and 20 mg posttreatment reduced the cerebral infarction area; Paeonol 15 and 20 mg/kg pretreatment reduced the neuro-deficit score. In addition, Paeonol 20 mg/kg pretreatment reduced the lucigenin-CL counts at 2 h period of reperfusion. The number of ED1 and IL-1beta immunoreactive cells also reduced in the cerebral infarction region; there were no significant changes in blood sugar levels. The results show that Paeonol reduced cerebral infarct and neuro-deficit in rat, suggesting Paeonol might play a similar role in reducing cerebral infarction in humans. Paeonol suppresses and scavenges superoxide anion, and inhibit microglia activation and IL-1beta in ischemia-reperfusion injured rats.
Collapse
Affiliation(s)
- Ching-Liang Hsieh
- Department of Chinese Medicine, China Medical University Hospital, Taichung City, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gupta R, Jovin TG, Krieger DW. Therapeutic hypothermia for stroke: do new outfits change an old friend? Expert Rev Neurother 2006; 5:235-46. [PMID: 15853493 DOI: 10.1586/14737175.5.2.235] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Clinically significant neuroprotection for the brain continues to be an elusive quest. All attempts at developing effective pharmacologic agents have failed in clinical trials. Hypothermia has been thought to confer protection after brain injury for many years, but has recently regained interest as a neuroprotectant for focal ischemic stroke in the basic science and clinical literature. The failure to develop safe and efficacious pharmacologic agents along with promising clinical data on the efficacy of hypothermia for cardiac arrest patients have raised a great interest in hypothermia as a neuroprotectant for ischemic stroke. As a clinically meaningful neuroprotectant for stroke, hypothermia confers several theoretical advantages over pharmacologic agents. A major problem with neuroprotectant therapy is instituting therapy within a narrow time window. This obstacle may be easier for hypothermia to overcome as emergency medical service personnel can theoretically initiate it in the field. Additionally, pharmacologic agents are usually restricted to one aspect of the pathophysiologic cascade triggered by focal ischemia, whereas hypothermia acts on several of these pathways simultaneously. The recent advances and future directions in the utilization of hypothermia as a potential therapy for focal ischemic stroke are reviewed.
Collapse
Affiliation(s)
- Rishi Gupta
- Department of Neurology, Stroke Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, USA
| | | | | |
Collapse
|
43
|
Kim SJ, Jeong HJ, Moon PD, Lee KM, Moon BS, Myung NY, An NH, Hong SH, Um JY, Kim HM. Effect of Danchunwhangagam on LPS or DFX-induced cytokine production in peripheral mononuclear cells of cerebral infarction patients. Immunopharmacol Immunotoxicol 2006; 27:683-96. [PMID: 16435585 DOI: 10.1080/08923970500418950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The Danchunwhangagam (DCWGG) has long been used for various cerebrovascular diseases. However, little scientific investigation has been carried out. Cytokines involved in the regulation of inflammatory reactions and immune responses may play a role in the pathogenesis of cerebral infarction (CI). The aim of the present study is to investigate the effect of DCWGG on the production of proinflammatory cytokines in peripheral blood mononuclear cells (PBMCs) from CI patients. The amount of tumor necrosis factor (TNF)-alpha, interleukin (IL)-1alpha, IL-1beta, IL-6, and IL-8 in PBMC culture supernatant was significantly increased in the lipopolysaccaride (LPS)- or desferrioxamine (DFX)-treated cells compared with unstimulated cells. We showed that DCWGG inhibited the production of TNF-alpha, IL-1alpha, IL-1beta IL-6, and IL-8 induced by LPS in a dose-dependent manner. Also, DCWGG inhibited TNF-alpha, IL-1alpha, IL-beta, IL-6, and IL-8 production-induced DFX in dose-dependent manner. These results suggest that DCWGG might have regulatory effects on LPS- or DFX-induced cytokine production, which might explain its beneficial effect in the treatment of CI.
Collapse
Affiliation(s)
- Su-Jin Kim
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lai AY, Todd KG. Hypoxia-activated microglial mediators of neuronal survival are differentially regulated by tetracyclines. Glia 2006; 53:809-16. [PMID: 16541436 DOI: 10.1002/glia.20335] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The tetracycline derivatives minocycline (MINO) and doxycycline (DOXY) have been shown to be neuroprotective in in vivo and in vitro models of stroke. This neuroprotection is thought to be due to the suppression of microglial activation. However, the specific molecular parameters in microglia of the tetracyclines' effect are not understood. We subjected cultured rat microglial and neuronal cells to in vitro hypoxia and examined the effects of MINO and DOXY pre-treatments. Our data showed that MINO and DOXY protect against hypoxia-induced neuronal death by a mechanism dependent on regulation of microglial factors, but likely unrelated to regulation of microglial proliferation/viability. Both MINO and DOXY suppressed the hypoxic activation of ED-1, a marker for microglial activation. Morphological analyses of hypoxic microglia using the microglial marker Iba1 revealed that treatment with MINO and DOXY caused a higher percentage of microglia to remain in a non-activated state. MINO suppressed the hypoxic upregulation of pro-inflammatory agents nitric oxide (NO), interleukin-1 beta (IL-1beta), and tumor necrosis factor alpha (TNF-alpha), while DOXY down-regulated only NO and IL-1beta. In contrast, the hypoxic activation of pro-survival/neuroprotective microglial proteins, such as brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF), were unaffected by tetracycline treatments. Taken together, these results suggest that MINO and DOXY may provide neuroprotection against stroke by selectively down-regulating microglial toxic factors while maintaining functional pro-survival factors.
Collapse
Affiliation(s)
- Aaron Y Lai
- Neurochemical Research Unit, Department of Psychiatry and Centre for Neuroscience, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
45
|
Kim SJ, Jeong HJ, Moon PD, Lee KM, Lee MG, Moon BS, An NH, Hong SH, Na HJ, Jeon H, Kim HM, Um JY. The Anti-inflammatory Effect of Gigukjiwhangwhangami through the Inhibition of Nuclear Factor-.KAPPA.B Activation in the Peripheral Blood Mononuclear Cells of Patients with Cerebral Infarction. Biol Pharm Bull 2006; 29:2251-5. [PMID: 17077523 DOI: 10.1248/bpb.29.2251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Korean genuine medicine "Gigukjiwhangwhangami (GJWGM)" has long been used for various cerebrovascular diseases. However, the exact mechanism that accounts for the anti-inflammatory effect of GJWGM is not completely understood. The aim of the present study is to elucidate how GJWGM modulates the inflammatory reaction in lipopolysaccaride (LPS)-stimulated peripheral mononuclear cells from patients with cerebral infarction. Production of cytokine was measured by the ELISA and RT-PCR method. The level of nuclear factor-kappaB (NF-kappaB)/Rel A protein and NF-kappaB DNA binding activity were determined by the Western blot analysis and TF-EIA method. We showed that GJWGM inhibited the production of tumor necrosis factor (TNF)-alpha, interleukin (IL)-1alpha, IL-6, and IL-8 induced by LPS in dose dependent manner (p<0.05). Maximal inhibition rate of TNF-alpha, IL-1beta, IL-6 and IL-8 production by GJWGM was about 54.34%, 41.37%, 44.04%, and 54.46%, respectively. GJWGM inhibited the TNF-alpha and IL-8 mRNA expression. In addition, we showed that the inhibitory mechanism of GJWGM is through the suppression of NF-kappaB pathway. Our study suggests that an important molecular mechanism by GJWGM reduce inflammation, which may explain its beneficial effect in the regulation of inflammatory reactions.
Collapse
Affiliation(s)
- Su-Jin Kim
- College of Oriental Medicine, Institute of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kis B, Chen L, Ueta Y, Busija DW. Autocrine peptide mediators of cerebral endothelial cells and their role in the regulation of blood-brain barrier. Peptides 2006; 27:211-22. [PMID: 16137789 DOI: 10.1016/j.peptides.2005.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Revised: 07/21/2005] [Accepted: 07/21/2005] [Indexed: 01/08/2023]
Abstract
A unique feature of cerebral endothelial cells (CECs) is the formation of the blood-brain barrier (BBB), which contributes to the stability of the brain microenvironment. CECs are capable of producing several substances mediating endothelium-dependent vasorelaxation or vasoconstriction, regulating BBB permeability, and participating in the regulation of cell-cell interactions during inflammatory and immunological processes. The chemical nature of these mediators produced by CECs ranges from gaseous anorganic molecules (e.g. nitric oxide) through lipid mediators (e.g. prostaglandins) to peptides. Peptide mediators are a large and diverse family of bioactive molecules which can elicit multiple effects on cerebral endothelial functions. In this review, we summarize current knowledge of peptide mediators produced by CECs, such as adrenomedullin, angiotensin, endothelin and several others and their role in the regulation of BBB functions.
Collapse
Affiliation(s)
- Bela Kis
- Department of Physiology and Pharmacology, Wake Forest University Health Sciences, Medical Center Blvd., Winston-Salem, NC 27157, USA.
| | | | | | | |
Collapse
|
47
|
Lao CJ, Lin JG, Kuo JS, Chao PDL, Cheng CY, Tang NY, Hsieh CL. Microglia, apoptosis and interleukin-1beta expression in the effect of sophora japonica l. on cerebral infarct induced by ischemia-reperfusion in rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2005; 33:425-38. [PMID: 16047560 DOI: 10.1142/s0192415x0500303x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sophora Japonica L. (SJ) is a traditional Chinese herb used to cool blood, stop bleeding and to treat hemorrhoids with bleeding. Although several recent studies found that both SJ and Ginkgo biloba have the same components of quercetin and rutin, only Ginkgo biloba has been widely used to treat cerebrovascular disorders and dementia in humans. This study investigated the effect of SJ on cerebral infarct in rats. A total of 66 Sprague-Dawley (SD) rats were studied. Focal cerebral infarct was established by occluding the bilateral common carotid arteries and the right middle cerebral artery for 90 minutes. After 24 hours of reperfusion, the neurological status was evaluated. The rats were then killed, and brain tissue was stained with 2,3,5-triphenyl-tetrazolium chloride. The grading scale of neurological deficit and the ratio of cerebral infarction area were used as an index to evaluate the effect of SJ on cerebral infarct. In addition, the number of ED1 and interleukin-1beta immunostaining positive cells, and apoptotic cells were measured in the cerebral infarction zone. The results indicated that pre-treatment with 100 or 200 mg/kg SJ and post-treatment with 200 mg/kg SJ significantly reduced the grade of neurological deficit and the ratio of cerebral infarction area. In addition, pre-treatment with 200 mg/kg SJ also significantly reduced ED1 and interleukin-1beta immunostaining positive cells, and apoptotic cells in ischemia-reperfusion cerebral infarct rats. This study demonstrated that SJ could reduce the cerebral infarction area and neurological deficit induced by ischemia-reperfusion in rats, suggesting its potential as a treatment for cerebral infarct in humans. This effect of SJ involves its suppressive action of microglia, interleukin-1beta and apoptosis.
Collapse
Affiliation(s)
- Chih-Jui Lao
- Graduate Institute of Chinese Medical Science, College of Chinese Medicine, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
48
|
Clausen BH, Lambertsen KL, Meldgaard M, Finsen B. A quantitative in situ hybridization and polymerase chain reaction study of microglial-macrophage expression of interleukin-1beta mRNA following permanent middle cerebral artery occlusion in mice. Neuroscience 2005; 132:879-92. [PMID: 15857694 DOI: 10.1016/j.neuroscience.2005.01.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2004] [Revised: 12/22/2004] [Accepted: 01/05/2005] [Indexed: 12/28/2022]
Abstract
Interleukin-1beta (IL-1beta) is known to play a central role in ischemia-induced brain damage in rodents. In comparison to the rat, however, the available data on the cellular synthesis of IL-1beta mRNA and protein in the mouse are very limited. Here, we report on the time profile, the topography and the quantitative, cellular expression of IL-1beta mRNA in mice subjected to permanent occlusion of the distal middle cerebral artery (MCA). The in situ hybridization analysis showed that IL-1beta mRNA was expressed during the first post-surgical hour in a small number of high-expressing macrophage-like cells, located in cortical layers I and II of the future infarct. At 2 h, a significant number of faintly labeled IL-1beta mRNA-expressing cells had appeared in the developing peri-infarct, and the number remained constant at 4 h and 6 h, when the hybridization signal began to distribute to the cellular processes. Quantitative PCR performed on whole hemispheres showed a significant 20-fold increase in the relative level of IL-1beta mRNA at 12 h and a highly significant 42-fold increase at 24 h, at which time single IL-1beta mRNA-expressing cells were supplemented by aggregates and perivascular infiltrates of intensely labeled IL-1beta mRNA-expressing cells. Immunohistochemistry and double immunohistochemical stainings in addition to combined in situ hybridization, confirmed that the intensely labeled IL-1beta mRNA-expressing and IL-1beta protein synthesizing cells predominantly were glial fibrillary acidic protein-immunonegative, macrophage associated antigen-1-immunopositive microglia-macrophages. By day 5 there was a dramatic decline in the relative level of IL-1beta mRNA in the ischemic hemisphere. In summary, the data provide evidence that permanent occlusion of the distal MCA in mice results in expression of IL-1beta mRNA and IL-1beta synthesis in spatially and temporally segregated subpopulations of microglia and macrophages.
Collapse
Affiliation(s)
- B H Clausen
- Medical Biotechnology Center, University of Southern Denmark, Odense, Denmark
| | | | | | | |
Collapse
|
49
|
Qin X, Hurn PD, Littleton-Kearney MT. Estrogen restores postischemic sensitivity to the thromboxane mimetic U46619 in rat pial artery. J Cereb Blood Flow Metab 2005; 25:1041-6. [PMID: 15758945 DOI: 10.1038/sj.jcbfm.9600105] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The objectives of the study were to (1) characterize the dose-response relationship to the TXA2 analog, U46619 (0.01, 0.1, and 1 micromol/L) after global cerebral ischemia, (2) determine whether chronic 17beta-estradiol (E2) replacement alters this relationship, and (3) determine if E2's mechanisms are transduced through cognate estrogen receptors. Rats were assigned to five groups (n=6): placebo-implanted ovariectomized (OVX) females, OVX plus chronic E2 (CE), OVX plus acute E2 (AE), OVX plus chronic E2 plus the estrogen receptor inhibitor ICI 182,780 (CEI), and OVX plus acute E2 plus ICI 182,780 (AEI). Rats were anesthetized, intubated, cannulated (femoral artery and vein), fitted with a closed cranial window, and subjected to 15-min reversible forebrain ischemia (4-vessel occlusion, 4-VO) and 60 mins of reperfusion. Arterial blood gases, intrawindow pressure, and temperature were controlled. Vessel diameter was measured before and 5 mins after superfusion of each concentration of U46619. Compared with preischemic responses, contractile response to U46619 was depressed at all concentrations after ischemia in the OVX group. In the chronic E2 and acute E2 groups, contractile response to 1 micromol/L of U46619 was normalized to near baseline values. However, in the CEI and the AEI groups, postischemic vasoconstriction was similar to that observed in the OVX rats. We conclude that E2 targets the cerebral microvasculature to preserve postischemic pial artery reactivity and that the effect is receptor mediated. Restoration of normal constriction to vascular agonists may be an important mechanism by which E2 protects the vasculature and diminishes tissue damage after ischemia.
Collapse
Affiliation(s)
- Xinyue Qin
- Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, USA
| | | | | |
Collapse
|
50
|
Panickar KS, Norenberg MD. Astrocytes in cerebral ischemic injury: morphological and general considerations. Glia 2005; 50:287-298. [PMID: 15846806 DOI: 10.1002/glia.20181] [Citation(s) in RCA: 232] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Asrocytic responses constitute one of the earliest and most prominent changes in the CNS following ischemic injury. Astrocytes are known to carry out critical functions such as maintenance of ionic homeostasis, prevention of excitotoxicity, scavenging free radicals, provision of nutrients and growth factors, promotion of neovascularization, and support of synaptogenesis and neurogenesis that potentially may influence the outcome of ischemic injury. This article reviews ischemia-associated alterations in astrocytes and their potential significance. Interactions with neurons, microglia, and endothelial cells are also considered. This article highlights the critical role of astrocytes in the CNS response to ischemic injury.
Collapse
Affiliation(s)
- Kiran S Panickar
- Department of Pathology, University of Miami School of Medicine, Miami, Florida
| | - Michael D Norenberg
- Department of Pathology, University of Miami School of Medicine, Miami, Florida
- Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, Florida
- Veterans Affairs Medical Center, Miami, Florida
| |
Collapse
|