1
|
Lee DH, Lee EC, Park SW, Lee JY, Kim KP, Oh JS. Prospero Homeobox 1 and Doublecortin Correlate with Neural Damage after Ischemic Stroke. J Korean Neurosurg Soc 2024; 67:333-344. [PMID: 37867430 PMCID: PMC11079562 DOI: 10.3340/jkns.2023.0154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/28/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023] Open
Abstract
OBJECTIVE Markers of neuroinflammation during ischemic stroke are well characterized, but additional markers of neural damage are lacking. The study identified associations of behavioral disorders after stroke with histologic neural damage and molecular biological change. METHODS Eight-week-old, 25 g male mice of the C57BL/6J strain were subjected to middle cerebral artery occlusion (MCAO) to induce ischemic stroke. The control group was a healthy wild type (WT), and the experimental group were designed as a low severity MCAO1 and a high severity MCAO2 based on post-stroke neurological scoring. All groups underwent behavioral tests, realtime polymerase chain reaction, triphenyltetrazolium chloride (TTC) staining and Hematoxylin and Eosin staining. One-way analysis of variance was used to analyze statistical significance between groups. RESULTS In TTC staining, MCAO1 showed 29.02% and MCAO2 showed 38.94% infarct volume (p<0.0001). The pro-inflammatory cytokine interleukin (IL)-1β was most highly expressed in MCAO2 (WT 0.44 vs. MCAO1 2.69 vs. MCAO2 5.02, p<0.0001). From the distance to target in the Barnes maze test, WT had a distance of 178 cm, MCAO1 had a distance of 276 cm, and MCAO2 had a distance of 1051 (p=0.0015). The latency to target was 13.3 seconds for WT, 27.9 seconds for MCAO1, and 87.9 seconds for MCAO2 (p=0.0007). Prospero homeobox 1 (Prox1) was most highly expressed in MCAO2 (p=0.0004). Doublecortin (Dcx) was most highly expressed in MCAO2 (p<0.0001). CONCLUSION The study demonstrated that histological damage to neural cells and changes in brain mRNA expression were associated with behavioral impairment after ischemic stroke. Prox1 and Dcx may be biomarkers of neural damage associated with long-term cognitive decline, and increased expression at the mRNA level was consistent with neural damage and long-term cognitive dysfunction.
Collapse
Affiliation(s)
- Dong-Hun Lee
- Department of Neurosurgery, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Eun Chae Lee
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sang-Won Park
- Department of Neurosurgery, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Ji young Lee
- Department of Neurosurgery, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kee-Pyo Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae Sang Oh
- Department of Neurosurgery, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
2
|
Noll JM, Sherafat AA, Ford GD, Ford BD. The case for neuregulin-1 as a clinical treatment for stroke. Front Cell Neurosci 2024; 18:1325630. [PMID: 38638304 PMCID: PMC11024452 DOI: 10.3389/fncel.2024.1325630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/01/2024] [Indexed: 04/20/2024] Open
Abstract
Ischemic stroke is the leading cause of serious long-term disability and the 5th leading cause of death in the United States. Revascularization of the occluded cerebral artery, either by thrombolysis or endovascular thrombectomy, is the only effective, clinically-approved stroke therapy. Several potentially neuroprotective agents, including glutamate antagonists, anti-inflammatory compounds and free radical scavenging agents were shown to be effective neuroprotectants in preclinical animal models of brain ischemia. However, these compounds did not demonstrate efficacy in clinical trials with human patients following stroke. Proposed reasons for the translational failure include an insufficient understanding on the cellular and molecular pathophysiology of ischemic stroke, lack of alignment between preclinical and clinical studies and inappropriate design of clinical trials based on the preclinical findings. Therefore, novel neuroprotective treatments must be developed based on a clearer understanding of the complex spatiotemporal mechanisms of ischemic stroke and with proper clinical trial design based on the preclinical findings from specific animal models of stroke. We and others have demonstrated the clinical potential for neuregulin-1 (NRG-1) in preclinical stroke studies. NRG-1 significantly reduced ischemia-induced neuronal death, neuroinflammation and oxidative stress in rodent stroke models with a therapeutic window of >13 h. Clinically, NRG-1 was shown to be safe in human patients and improved cardiac function in multisite phase II studies for heart failure. This review summarizes previous stroke clinical candidates and provides evidence that NRG-1 represents a novel, safe, neuroprotective strategy that has potential therapeutic value in treating individuals after acute ischemic stroke.
Collapse
Affiliation(s)
- Jessica M. Noll
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, Riverside, CA, United States
- Nanostring Technologies, Seattle, WA, United States
| | - Arya A. Sherafat
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, Riverside, CA, United States
| | - Gregory D. Ford
- Southern University-New Orleans, New Orleans, LA, United States
| | - Byron D. Ford
- Department of Anatomy, Howard University College of Medicine, Washington, DC, United States
| |
Collapse
|
3
|
Pereira JF, de Sousa Neves JC, Fonteles AA, Bezerra JR, Pires RC, da Silva ATA, Lima FAV, Neves KRT, Oriá RB, de Barros Viana GS, Tavares J, de Sousa Nascimento T, Oliveira AV, Parente ACB, Gomes JMP, de Andrade GM. Palmatine, a natural alkaloid, attenuates memory deficits and neuroinflammation in mice submitted to permanent focal cerebral ischemia. J Neuroimmunol 2023; 381:578131. [PMID: 37413943 DOI: 10.1016/j.jneuroim.2023.578131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/16/2023] [Accepted: 06/13/2023] [Indexed: 07/08/2023]
Abstract
Ischemic stroke is one of the major causes of human morbidity and mortality. The pathophysiology of ischemic stroke involves complex events, including oxidative stress and inflammation, that lead to neuronal loss and cognitive deficits. Palmatine (PAL) is a naturally occurring (Coptidis rhizome) isoquinoline alkaloid that belongs to the class of protoberberines and has a wide spectrum of pharmacological and biological effects. In the present study, we evaluated the impact of Palmatine on neuronal damage, memory deficits, and inflammatory response in mice submitted to permanent focal cerebral ischemia induced by middle cerebral artery (pMCAO) occlusion. The animals were treated with Palmatine (0.2, 2 and 20 mg/kg/day, orally) or vehicle (3% Tween + saline solution) 2 h after pMCAO once daily for 3 days. Cerebral ischemia was confirmed by evaluating the infarct area (TTC staining) and neurological deficit score 24 h after pMCAO. Treatment with palmatine (2 and 20 mg/kg) reduced infarct size and neurological deficits and prevented working and aversive memory deficits in ischemic mice. Palmatine, at a dose of 2 mg/kg, had a similar effect of reducing neuroinflammation 24 h after cerebral ischemia, decreasing TNF-, iNOS, COX-2, and NF- κB immunoreactivities and preventing the activation of microglia and astrocytes. Moreover, palmatine (2 mg/kg) reduced COX-2, iNOS, and IL-1β immunoreactivity 96 h after pMCAO. The neuroprotective properties of palmatine make it an excellent adjuvant treatment for strokes due to its inhibition of neuroinflammation.
Collapse
Affiliation(s)
- Juliana Fernandes Pereira
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil
| | - Juliana Catharina de Sousa Neves
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Analu Aragão Fonteles
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Jéssica Rabelo Bezerra
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Rayssa Costa Pires
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Ana Thais Araújo da Silva
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Francisco Arnaldo Viana Lima
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Kelly Rose Tavares Neves
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Reinaldo Barreto Oriá
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil
| | - Glauce Socorro de Barros Viana
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Juliete Tavares
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Tyciane de Sousa Nascimento
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Alfaete Vieira Oliveira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Ana Caroline Barros Parente
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Jessica Maria Pessoa Gomes
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Geanne Matos de Andrade
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil.
| |
Collapse
|
4
|
Fahmawi A, Khalifeh MS, Alzoubi KH, Rababa'h AM. The Effects of Acute and Chronic Sleep Deprivation on the Immune Profile in the Rat. Curr Mol Pharmacol 2023; 16:101-108. [PMID: 35297357 DOI: 10.2174/1874467215666220316104321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/11/2021] [Accepted: 12/29/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Acute and chronic sleep deprivation present many health-related problems in modern societies, mainly concerning the immune system. Immune factors, particularly the interleukins, regulate sleep and, therefore, may be altered by sleep deprivation (SD). OBJECTIVES We aimed to investigate the possible effects of acute and chronic sleep deprivation on selected cytokines, including interleukins (IL-1β, IL-9, IL-17, and IL-23) and tumor necrosis factor- alpha (TNF-α). METHODS The animals were grouped into acute sleep-deprived (SD; for 24 hours) and chronic sleep-deprived (8 hours a day for 10, 20, and 30-days). The SD was induced using the multipleplatforms model. The serum levels of cytokines were measured using commercially available ELISA. RESULTS The serum levels of IL-1β were significantly reduced after acute SD, whereas they were increased after 20-days of chronic SD. The IL-9 levels were reduced after acute SD, increased after 10-days of SD, and reduced again after 30-days of SD. Conversely, the levels of IL-23 were not changed after acute SD, reduced after 10 days of SD, and increased after 30-days of SD. Levels of TNF-α were not changed after acute SD, whereas they were increased after 20 and 30- days of SD. CONCLUSION In conclusion, both acute and chronic SD distinctly disturb the immune profile, which might result in the emergence of various pathologies presented during sleep deprivation.
Collapse
Affiliation(s)
- Alaa Fahmawi
- Department of Basic Medical Veterinary Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammad S Khalifeh
- Department of Basic Medical Veterinary Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, UAE
- Department of Clinical Pharmacy, College of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Abeer M Rababa'h
- Department of Clinical Pharmacy, College of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
5
|
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
|
6
|
del Zoppo GJ, Moskowitz MA, Nedergaard M. The Neurovascular Unit and Responses to Ischemia. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
7
|
Damous LL, Shiroma ME, Carvalho AETSD, Soares-Jr JM, Krieger JE, Baracat EC. Gene expression profile in experimental frozen-thawed ovarian grafts treated with scaffold-base delivery of adipose tissue-derived stem cells. Clinics (Sao Paulo) 2022; 77:100066. [PMID: 35777300 PMCID: PMC9253596 DOI: 10.1016/j.clinsp.2022.100066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
PURPOSE Gelfoam scaffold is a feasible and safe non-invasive technique for Adipose tissue-derived Stem Cell (ASC)-delivery in the treatment of frozen-thawed ovarian autografts. This study seeks to analyze the genes expression profile of rat frozen-thawed ovarian autografts treated with scaffold-based delivery of adipose tissue-derived stem cells. METHODS Eighteen adult Wistar rats were distributed into three groups: Control (frozen-thawed only); Group 1 (G1) and Group 2 (G2) (frozen-thawed ovaries treated with culture medium or ASC, respectively). Both treatments were performed immediately after autologous retroperitoneal transplant with scaffold-based delivery. The ovarian grafts were retrieved 30 days after transplantation. Quantitative gene expression (qPCR) for apoptosis, angiogenesis, and inflammatory cytokines (84 genes in each pathway) were evaluated by RT-PCR. Graft morphology (HE), apoptosis (cleaved-caspase-3), neoangiogenesis (VEGF), and cellular proliferation (Ki-67) were assessed. RESULTS In grafts treated with ASC, the apoptosis pathway showed the highest number of genes over-regulated - 49 genes - compared to inflammation cytokines and angiogenesis pathway - 36 and 23 genes respectively, compared to grafts treated with culture medium. Serpinb5 family was highlighted in the angiogenesis pathway and Cxcl6 in the inflammation cytokines pathway. In the apoptosis pathway, the most over-regulated gene was Capsase14. ASC treatment promoted the reduction of cleaved caspase-3 in the theca internal layer and increased cell proliferation by Ki-67 in the granulosa layer without altering VEGF. A mild inflammatory infiltrate was observed in both groups. CONCLUSION ASC therapy in rat frozen-thawed ovarian autografts promoted an abundance of genes involved with apoptosis and inflammatory cytokines without compromising the ovary graft morphology and viability for short time. Further studies are necessary to evaluate the repercussion of apoptosis and inflammation on the graft in the long term.
Collapse
Affiliation(s)
- Luciana Lamarão Damous
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Marcos Eiji Shiroma
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ana Elisa Teófilo Saturi de Carvalho
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (Incor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - José Maria Soares-Jr
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - José Eduardo Krieger
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (Incor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Edmund C Baracat
- Laboratório de Ginecologia Estrutural e Molecular (LIM-58), Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
8
|
Shah MA, Kang JB, Park DJ, Kim MO, Koh PO. Chlorogenic acid alleviates cerebral ischemia-induced neuroinflammation via attenuating nuclear factor kappa B activation. Neurosci Lett 2022; 773:136495. [PMID: 35108588 DOI: 10.1016/j.neulet.2022.136495] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
|
9
|
Van Zanden JE, 't Hart NA, Ottens PJ, Liu B, Rebolledo RA, Erasmus ME, Leuvenink HGD. Methylprednisolone Treatment in Brain Death-Induced Lung Inflammation-A Dose Comparative Study in Rats. Front Pharmacol 2021; 12:587003. [PMID: 33692687 PMCID: PMC7937885 DOI: 10.3389/fphar.2021.587003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/27/2021] [Indexed: 12/14/2022] Open
Abstract
Background: The process of brain death (BD) leads to a pro-inflammatory state of the donor lung, which deteriorates its quality. In an attempt to preserve lung quality, methylprednisolone is widely recommended in donor lung management. However, clinical treatment doses vary and the dose-effect relation of methylprednisolone on BD-induced lung inflammation remains unknown. The aim of this study was to investigate the effect of three different doses methylprednisolone on the BD-induced inflammatory response. Methods: BD was induced in rats by inflation of a Fogarty balloon catheter in the epidural space. After 60 min of BD, saline or methylprednisolone (low dose (5 mg/kg), intermediate dose (12.5 mg/kg) or high dose (22.5 mg/kg)) was administered intravenously. The lungs were procured and processed after 4 h of BD. Inflammatory gene expressions were analyzed by RT-qPCR and influx of neutrophils and macrophages were quantified with immunohistochemical staining. Results: Methylprednisolone treatment reduced neutrophil chemotaxis as demonstrated by lower IL-8-like CINC-1 and E-selectin levels, which was most evident in rats treated with intermediate and high doses methylprednisolone. Macrophage chemotaxis was attenuated in all methylprednisolone treated rats, as corroborated by lower MCP-1 levels compared to saline treated rats. Thereby, all doses methylprednisolone reduced TNF-α, IL-6 and IL-1β tissue levels. In addition, intermediate and high doses methylprednisolone induced a protective anti-inflammatory response, as reflected by upregulated IL-10 expression when compared to saline treated brain-dead rats. Conclusion: We showed that intermediate and high doses methylprednisolone share most potential to target BD-induced lung inflammation in rats. Considering possible side effects of high doses methylprednisolone, we conclude from this study that an intermediate dose of 12.5 mg/kg methylprednisolone is the optimal treatment dose for BD-induced lung inflammation in rats, which reduces the pro-inflammatory state and additionally promotes a protective, anti-inflammatory response.
Collapse
Affiliation(s)
- Judith E Van Zanden
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Nils A 't Hart
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Petra J Ottens
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Bo Liu
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Rolando A Rebolledo
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Institute for Medical and Biological Engineering, Schools of Engineering, Biological Sciences and Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Michiel E Erasmus
- Department of cardiothoracic surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Henri G D Leuvenink
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
10
|
A scalable, fully automated approach for regional quantification of immunohistochemical staining of astrocytes in the rat brain. J Neurosci Methods 2020; 348:108994. [PMID: 33176173 DOI: 10.1016/j.jneumeth.2020.108994] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/26/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Astrocytes play a critical role in CNS functions by providing physiological support to surrounding cells. These cells present a particularly unique challenge for in vitro immunohistochemical quantification due reactive gliosis after insult or injury, which is characterized by the extension of long processes. NEW METHOD We present an optimized QuPath protocol that is scalable, fully automated, and capable of being applied to images generated by whole slide scanning technology using this open-source software. RESULTS We induced mechanical injury in the rat brain and stained astrocytes using glial fibrillary acidic protein (GFAP) and 3,3-diaminobenzidine (DAB) chromogen detection. Slides were scanned using a whole slide scanner, Vectra Polaris. Using QuPath, we summarize and contrast three ways of quantifying astrocytes in uninjured (contralateral) and injured (ipsilateral) hemispheres: optical density, positive pixels and positive proportion. COMPARISON WITH EXISTING METHODS Robust quantification of DAB stained astrocytes remains elusive. Previous methodologies have relied on software that is not compatible with whole slide scanner images. Use of such software can compromise the data integrity within the image and is limited by issues with scalability and lack of automation. Previous methods using manual histopathological scoring are also limited by the ability to quantify large numbers of astrocytes. Given these limitations, we were unable to directly compare our method with those using other software or manual histopathology. CONCLUSIONS Based on an analysis of our method, we conclude that positive proportion may be the most effective way to quantify astrocytic responses using GFAP and DAB immunohistochemistry in the brain.
Collapse
|
11
|
Amruta N, Rahman AA, Pinteaux E, Bix G. Neuroinflammation and fibrosis in stroke: The good, the bad and the ugly. J Neuroimmunol 2020; 346:577318. [PMID: 32682140 PMCID: PMC7794086 DOI: 10.1016/j.jneuroim.2020.577318] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 02/08/2023]
Abstract
Stroke is the leading cause of death and the main cause of disability in surviving patients. The detrimental interaction between immune cells, glial cells, and matrix components in stroke pathology results in persistent inflammation that progresses to fibrosis. A substantial effort is being directed toward understanding the exact neuroinflammatory events that take place as a result of stroke. The initiation of a potent cytokine response, along with immune cell activation and infiltration in the ischemic core, has massive acute deleterious effects, generally exacerbated by comorbid inflammatory conditions. There is secondary neuroinflammation that promotes further injury, resulting in cell death, but conversely plays a beneficial role, by promoting recovery. This highlights the need for a better understanding of the neuroinflammatory and fibrotic processes, as well as the need to identify new mechanisms and potential modulators. In this review, we summarize several aspects of stroke-induced inflammation, fibrosis, and include a discussion of cytokine inhibitors/inducers, immune cells, and fibro-inflammation signaling inhibitors in order to identify new pharmacological means of intervention.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Abir A Rahman
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom.
| | - Gregory Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
12
|
Nor Arfuzir NN, Agarwal R, Iezhitsa I, Agarwal P, Ismail NM. Magnesium acetyltaurate protects against endothelin-1 induced RGC loss by reducing neuroinflammation in Sprague dawley rats. Exp Eye Res 2020; 194:107996. [PMID: 32156652 DOI: 10.1016/j.exer.2020.107996] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/25/2020] [Accepted: 03/06/2020] [Indexed: 12/22/2022]
Abstract
Endothelin-1 (ET-1), a potent vasoconstrictor, plays a significant role in the pathophysiology of ocular conditions like glaucoma. Glaucoma is characterized by apoptotic loss of retinal ganglion cells (RGCs) and loss of visual fields and is a leading cause of irreversible blindness. In glaucomatous eyes, retinal ischemia causes release of pro-inflammatory mediators such as interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α and promotes activation of transcription factors such as nuclear factor kappa B (NFKB) and c-Jun. Magnesium acetyltaurate (MgAT) has previously been shown to protect against ET-1 induced retinal and optic nerve damage. Current study investigated the mechanisms underlying these effects of MgAT, which so far remain unknown. Sprague dawley rats were intravitreally injected with ET-1 with or without pretreatment with MgAT. Seven days post-injection, retinal expression of IL-1β, IL-6, TNF-α, NFKB and c-Jun protein and genes was determined using multiplex assay, Western blot and PCR. Animals were subjected to retrograde labeling of RGCs to determine the extent of RGC survival. RGC survival was also examined using Brn3A staining. Furthermore, visual functions of rats were determined using Morris water maze. It was observed that pre-treatment with MgAT protects against ET-1 induced increase in the retinal expression of IL-1β, IL-6 and TNF-α proteins and genes. It also protected against ET-1 induced activation of NFKB and c-Jun. These effects of MgAT were associated with greater RGC survival and preservation of visual functions in rats. In conclusion, MgAT prevents ET-1 induced RGC loss and loss of visual functions by suppressing neuroinflammatory reaction in rat retinas.
Collapse
Affiliation(s)
- Natasha Najwa Nor Arfuzir
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia.
| | - Igor Iezhitsa
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia; Volgograd State Medical University, Research Centre for Innovative Medicines, Volgograd, Russian Federation; Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Puneet Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Nafeeza Mohd Ismail
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Singh D, Reeta K, Sharma U, Jagannathan N, Dinda A, Gupta Y. Neuro-protective effect of monomethyl fumarate on ischemia reperfusion injury in rats: Role of Nrf2/HO1 pathway in peri-infarct region. Neurochem Int 2019; 126:96-108. [DOI: 10.1016/j.neuint.2019.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 02/28/2019] [Accepted: 03/11/2019] [Indexed: 12/30/2022]
|
14
|
Peng T, Jiang Y, Farhan M, Lazarovici P, Chen L, Zheng W. Anti-inflammatory Effects of Traditional Chinese Medicines on Preclinical in vivo Models of Brain Ischemia-Reperfusion-Injury: Prospects for Neuroprotective Drug Discovery and Therapy. Front Pharmacol 2019; 10:204. [PMID: 30930774 PMCID: PMC6423897 DOI: 10.3389/fphar.2019.00204] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/18/2019] [Indexed: 12/28/2022] Open
Abstract
Acquired brain ischemia-and reperfusion-injury (IRI), including both Ischemic stroke (IS) and Traumatic Brain injury (TBI), is one of the most common causes of disability and death in adults and represents a major burden in both western and developing countries worldwide. China’s clinical neurological therapeutic experience in the use of traditional Chinese medicines (TCMs), including TCM-derived active compounds, Chinese herbs, TCM formulations and decoction, in brain IRI diseases indicated a trend of significant improvement in patients’ neurological deficits, calling for blind, placebo-controlled and randomized clinical trials with careful meta-analysis evaluation. There are many TCMs in use for brain IRI therapy in China with significant therapeutic effects in preclinical studies using different brain IRI-animal. The basic hypothesis in this field claims that in order to avoid the toxicity and side effects of the complex TCM formulas, individual isolated and identified compounds that exhibited neuroprotective properties could be used as lead compounds for the development of novel drugs. China’s efforts in promoting TCMs have contributed to an explosive growth of the preclinical research dedicated to the isolation and identification of TCM-derived neuroprotective lead compounds. Tanshinone, is a typical example of TCM-derived lead compounds conferring neuroprotection toward IRI in animals with brain middle cerebral artery occlusion (MCAO) or TBI models. Recent reports show the significance of the inflammatory response accompanying brain IRI. This response appears to contribute to both primary and secondary ischemic pathology, and therefore anti-inflammatory strategies have become popular by targeting pro-inflammatory and anti-inflammatory cytokines, other inflammatory mediators, reactive oxygen species, nitric oxide, and several transcriptional factors. Here, we review recent selected studies and discuss further considerations for critical reevaluation of the neuroprotection hypothesis of TCMs in IRI therapy. Moreover, we will emphasize several TCM’s mechanisms of action and attempt to address the most promising compounds and the obstacles to be overcome before they will enter the clinic for IRI therapy. We hope that this review will further help in investigations of neuroprotective effects of novel molecular entities isolated from Chinese herbal medicines and will stimulate performance of clinical trials of Chinese herbal medicine-derived drugs in IRI patients.
Collapse
Affiliation(s)
- Tangming Peng
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China.,Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
| | - Yizhou Jiang
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Mohd Farhan
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Philip Lazarovici
- Faculty of Medicine, School of Pharmacy, The Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ligang Chen
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China.,Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
| | - Wenhua Zheng
- Center of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
15
|
Hao Y, Qi Z, Ding Y, Yu X, Pang L, Zhao T. Effect of Interventional Therapy on IL-1β, IL-6, and Neutrophil-Lymphocyte Ratio (NLR) Levels and Outcomes in Patients with Ischemic Cerebrovascular Disease. Med Sci Monit 2019; 25:610-617. [PMID: 30664615 PMCID: PMC6350451 DOI: 10.12659/msm.912064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background This study investigated the clinical effect of interventional therapy in ischemic cerebrovascular disease (ICD). Material/Methods A retrospective analysis was performed on 260 ICD patients who were divided into a control group (122 patients, conventional drug treatment) and an observation group (138 patients, interventional therapy plus conventional drug treatment). Enzyme-linked immunosorbent assay was used to examine the expression of IL-1β, IL-6, and NLR. Furthermore, neurological deficit scores and Barthel index scores as well as the correlation of IL-1β, IL-6 and NLR were examined in these 2 groups. Results The expression of IL-1β, IL-6, and NLR significantly decreased in both groups after 1 week or 4 weeks of treatment compared with before treatment (P<0.05). Significant differences in neurological impairment scores were detected between these 2 groups after 4 weeks of treatment (P<0.05), and the control group showed higher neurological deficit scores than did the observation group (P<0.05). Barthel index scores were significantly higher after treatment than before treatment in the control and observation group (P<0.05), and the control group had lower Barthel index scores than did the observation group (P<0.05). Pearson correlation analysis showed that IL-1β, IL-6, and NLR expression were positively correlated in ICD patients (P<0.05). Conclusions Interventional surgery combined with conventional drug therapy can reduce serum IL-1β and IL-6 levels, decrease neurological impairment, and improve the quality of life of patients. The combined treatment group showed better outcomes than did the group that received the drug alone; therefore, combined therapy is suitable for promoting better clinical outcomes.
Collapse
Affiliation(s)
- Yongnan Hao
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Ziyou Qi
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Ying Ding
- Department of Radiology, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Xiangli Yu
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China (mainland)
| | - Li Pang
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Teng Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
16
|
Jank L, Pinto-Espinoza C, Duan Y, Koch-Nolte F, Magnus T, Rissiek B. Current Approaches and Future Perspectives for Nanobodies in Stroke Diagnostic and Therapy. Antibodies (Basel) 2019; 8:antib8010005. [PMID: 31544811 PMCID: PMC6640704 DOI: 10.3390/antib8010005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/20/2018] [Accepted: 12/27/2018] [Indexed: 12/15/2022] Open
Abstract
Antibody-based biologics are the corner stone of modern immunomodulatory therapy. Though highly effective in dampening systemic inflammatory processes, their large size and Fc-fragment mediated effects hamper crossing of the blood brain barrier (BBB). Nanobodies (Nbs) are single domain antibodies derived from llama or shark heavy-chain antibodies and represent a new generation of biologics. Due to their small size, they display excellent tissue penetration capacities and can be easily modified to adjust their vivo half-life for short-term diagnostic or long-term therapeutic purposes or to facilitate crossing of the BBB. Furthermore, owing to their characteristic binding mode, they are capable of antagonizing receptors involved in immune signaling and of neutralizing proinflammatory mediators, such as cytokines. These qualities combined make Nbs well-suited for down-modulating neuroinflammatory processes that occur in the context of brain ischemia. In this review, we summarize recent findings on Nbs in preclinical stroke models and how they can be used as diagnostic and therapeutic reagents. We further provide a perspective on the design of innovative Nb-based treatment protocols to complement and improve stroke therapy.
Collapse
Affiliation(s)
- Larissa Jank
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Carolina Pinto-Espinoza
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Yinghui Duan
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
17
|
Saeedi Saravi SS, Saeedi Saravi SS, Arefidoust A, Dehpour AR. The beneficial effects of HMG-CoA reductase inhibitors in the processes of neurodegeneration. Metab Brain Dis 2017; 32:949-965. [PMID: 28578514 DOI: 10.1007/s11011-017-0021-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 04/28/2017] [Indexed: 12/13/2022]
Abstract
Statins, cholesterol lowering drugs, have been demonstrated to exert beneficial effects in other conditions such as primary and progressing neurodegenerative diseases beyond their original role. Observation that statins ameliorate the neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS) and cerebral ischemic stroke, the neuroprotective effects of these drugs are thought to be linked to their anti-inflammatory, anti-oxidative, and anti-excitotoxic properties. Despite the voluminous literature on the clinical advantages of 3-hydroxy-3-methylglutaryl Co-enzyme A reductase (HMGCR) inhibitors (statins) in cardiovascular system, the neuroprotective effects and the underlying mechanisms are little understood. Hence, the present review tries to provide a critical overview on the statin-induced neuroprotection, which are presumed to be associated with the ability to reduce cholesterol, Amyloid-β and apolipoprotein E (ApoE) levels, decrease reactive oxygen and nitrogen species (ROS and RNS) formation, inhibit excitotoxicity, modulate matrix metalloproteinases (MMPs), stimulate endothelial nitric oxide synthase (eNOS), and increase cerebral blood perfusion. This review is also aimed to illustrate that statins protect neurons against the neuro-inflammatory processes through balancing pro-inflammatory/anti-inflammatory cytokines. Ultimately, the beneficial role of statins in ameliorating the development of PD, AD, MS and cerebral ischemic stroke has been separately reviewed.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Sobhan Saeedi Saravi
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Arefidoust
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Hurst J, Kuehn S, Jashari A, Tsai T, Bartz-Schmidt KU, Schnichels S, Joachim SC. A novel porcine ex vivo retina culture model for oxidative stress induced by H₂O₂. Altern Lab Anim 2017; 45:11-25. [PMID: 28409994 DOI: 10.1177/026119291704500105] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Oxidative stress is a key player in many ophthalmic diseases. However, the role of oxidative stress in most degenerative processes is not yet known. Therefore, accurate and practical models are required to efficiently screen for therapeutics. Porcine eyes are closely related to the human eye, and can be obtained from the abattoir as a by-product of the food industry. Therefore, they offer excellent opportunities for the development of culture models with which to pre-screen potential therapies, while reducing the use of laboratory animals. To induce oxidative stress, organotypic cultures of porcine retina were treated with different doses of hydrogen peroxide (H₂O₂; 100, 300 and 500μM) for three hours. On days 3 and 8, the retinas were conserved for histological and Western blotting analyses and for evaluation of gene expression, which determined the number of retinal ganglion cells (RGCs), the activation state of glial cells, and the expression levels of several oxidative stress markers. H₂O₂ treatment led to a reduction in the number of RGCs and to an increase in apoptotic RGCs. In addition, a dose-dependent increase of microglia and an elevation of CD11b expression was observed. On day 3, a reduction of IL-1β, and an increase of iNOS, as well as of HSP70 mRNA were found. On day 8, an increase in TNF-α and IL-1β mRNA expression was detected. In conclusion, this ex vivo model offers an opportunity to study the molecular mechanisms underlying certain eye disorders and to test new therapeutic approaches to diminish the effects of oxidative stress.
Collapse
Affiliation(s)
- José Hurst
- University Eye Hospital Tübingen, Centre for Ophthalmology Tübingen, Tübingen, Germany
| | - Sandra Kuehn
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Adelina Jashari
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Teresa Tsai
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | | | - Sven Schnichels
- University Eye Hospital Tübingen, Centre for Ophthalmology Tübingen, Tübingen, Germany
| | - Stephanie C Joachim
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
19
|
Abstract
The cytokine interleukin-1 (IL-1) has been implicated in many forms of neurodegeneration. Expression of IL-1 is increased in the brain (mainly by microglia) of animals and humans in response to acute insults (e.g., stroke and brain injury) and in chronic neurodegenerative conditions. Although IL-1 does not kill otherwise healthy neurons, small quantities of the cytokine dramatically enhance ischemic, traumatic, or excitotoxic damage in animals. Inhibition of the synthesis, release, or action of IL-1 (e.g., by administration of IL-1 receptor antagonist) markedly reduces all of these forms of experimental neurodegeneration, indicating that approaches to block or inhibit IL-1 activity may be of benefit in clinical neurodegenerative disease. NEURO SCIENTIST 4:195-201, 1998
Collapse
Affiliation(s)
- Nancy J. Rothwell
- School of Biological Sciences University of Manchester
Manchester, United Kmgdom
| |
Collapse
|
20
|
Abstract
Global and focal ischemias induce a variety of gene families, including immediate early genes, cytokines, neurotransmitter receptors, and heat-shock proteins. The Janus-like effects of several of these gene prod ucts promote neuronal survival and degeneration. Therefore, determining the molecular pathways respon sible for the differential regulation of these genes is of paramount importance. The discovery of apoptosis as a mediator of delayed neuronal death has led to the identification of a number of other genes involved in postischemic brain damage. Future neuroprotective therapies for cerebral ischemia may be directed at preventing alterations in gene expression. NEUROSCIENTIST 5:238-253, 1999
Collapse
Affiliation(s)
- Sean I. Savitz
- Department of Neurology, Neuroscience, Albert Einstein
College of Medicine Bronx, New York
| | - Daniel M. Rosenbaum
- Department of Neurology, Neuroscience and Ophthalmology
Albert Einstein College of Medicine Bronx, New York
| |
Collapse
|
21
|
Effects of Shaoyao-Gancao Decoction on Infarcted Cerebral Cortical Neurons: Suppression of the Inflammatory Response following Cerebral Ischemia-Reperfusion in a Rat Model. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1859254. [PMID: 27413737 PMCID: PMC4931082 DOI: 10.1155/2016/1859254] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/26/2016] [Accepted: 05/26/2016] [Indexed: 11/21/2022]
Abstract
The mechanisms by which Shaoyao-Gancao decoction (SGD) inhibits the production of inflammatory cytokines in serum and brain tissue after cerebral ischemia-reperfusion (CI-RP) in rats were investigated. A right middle cerebral artery occlusion was used to induce CI-RP after which the rats were divided into model (n = 39), SGD (n = 28), clopidogrel (n = 25) and sham operated (n = 34) groups. The Bederson scale was used to evaluate changes in behavioral indices. The levels of IL-1β, TNF-α, MCP-1, IL-10, RANTES, VEGF, and TGF-β1 in the serum and infarcted brain tissues were measured. Nissl body and immunohistochemical staining methods were used to detect biochemical changes in neurons, microglial cells, and astrocytes. Serum levels of VEGF, TNF-α, MCP-1, IL-1β, and IL-10 increased significantly 24 h after CI-RP. In brain tissue, levels of TNF-α and IL-1β significantly increased 24 h after CI-RP, whereas levels of TGF-β1 and MCP-1 were significantly higher 96 h after CI-RP (P < 0.05). SGD or clopidogrel after CI-RP reduced TNF-α and IL-1β levels in brain tissue and serum levels of MCP-1, IL-1β, and IL-10. SGD increased the number of NeuN-positive cells in infarcted brain tissue and reduced the number of IBA1-positive and GFAP-positive cells. The efficacy of SGD was significantly higher than that of clopidogrel.
Collapse
|
22
|
del Zoppo GJ, Moskowitz M, Nedergaard M. The Neurovascular Unit and Responses to Ischemia. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
23
|
Anrather J, Iadecola C, Hallenbeck J. Inflammation and Immune Response. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
24
|
Pielecka-Fortuna J, Kalogeraki E, Greifzu F, Löwel S. A Small Motor Cortex Lesion Abolished Ocular Dominance Plasticity in the Adult Mouse Primary Visual Cortex and Impaired Experience-Dependent Visual Improvements. PLoS One 2015; 10:e0137961. [PMID: 26368569 PMCID: PMC4569386 DOI: 10.1371/journal.pone.0137961] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/24/2015] [Indexed: 12/01/2022] Open
Abstract
It was previously shown that a small lesion in the primary somatosensory cortex (S1) prevented both cortical plasticity and sensory learning in the adult mouse visual system: While 3-month-old control mice continued to show ocular dominance (OD) plasticity in their primary visual cortex (V1) after monocular deprivation (MD), age-matched mice with a small photothrombotically induced (PT) stroke lesion in S1, positioned at least 1 mm anterior to the anterior border of V1, no longer expressed OD-plasticity. In addition, in the S1-lesioned mice, neither the experience-dependent increase of the spatial frequency threshold (“visual acuity”) nor of the contrast threshold (“contrast sensitivity”) of the optomotor reflex through the open eye was present. To assess whether these plasticity impairments can also occur if a lesion is placed more distant from V1, we tested the effect of a PT-lesion in the secondary motor cortex (M2). We observed that mice with a small M2-lesion restricted to the superficial cortical layers no longer expressed an OD-shift towards the open eye after 7 days of MD in V1 of the lesioned hemisphere. Consistent with previous findings about the consequences of an S1-lesion, OD-plasticity in V1 of the nonlesioned hemisphere of the M2-lesioned mice was still present. In addition, the experience-dependent improvements of both visual acuity and contrast sensitivity of the open eye were severely reduced. In contrast, sham-lesioned mice displayed both an OD-shift and improvements of visual capabilities of their open eye. To summarize, our data indicate that even a very small lesion restricted to the superficial cortical layers and more than 3mm anterior to the anterior border of V1 compromised V1-plasticity and impaired learning-induced visual improvements in adult mice. Thus both plasticity phenomena cannot only depend on modality-specific and local nerve cell networks but are clearly influenced by long-range interactions even from distant brain regions.
Collapse
Affiliation(s)
- Justyna Pielecka-Fortuna
- Department of Systems Neuroscience, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie and Bernstein Fokus Neurotechnologie, Georg-August-Universität Göttingen, Göttingen, Germany
- * E-mail:
| | - Evgenia Kalogeraki
- Department of Systems Neuroscience, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie and Bernstein Fokus Neurotechnologie, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Franziska Greifzu
- Department of Systems Neuroscience, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie and Bernstein Fokus Neurotechnologie, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Siegrid Löwel
- Department of Systems Neuroscience, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie and Bernstein Fokus Neurotechnologie, Georg-August-Universität Göttingen, Göttingen, Germany
| |
Collapse
|
25
|
Walberer M, Rueger MA. The macrosphere model-an embolic stroke model for studying the pathophysiology of focal cerebral ischemia in a translational approach. ANNALS OF TRANSLATIONAL MEDICINE 2015. [PMID: 26207251 DOI: 10.3978/j.issn.2305-5839.2015.04.02] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
The main challenge of stroke research is to translate promising experimental findings from the bench to the bedside. Many suggestions have been made how to achieve this goal, identifying the need for appropriate experimental animal models as one key issue. We here discuss the macrosphere model of focal cerebral ischemia in the rat, which closely resembles the pathophysiology of human stroke both in its acute and chronic phase. Key pathophysiological processes such as brain edema, cortical spreading depolarizations (CSD), neuroinflammation, and stem cell-mediated regeneration are observed in this stroke model, following characteristic temporo-spatial patterns. Non-invasive in vivo imaging allows studying the macrosphere model from the very onset of ischemia up to late remodeling processes in an intraindividual and longitudinal fashion. Such a design of pre-clinical stroke studies provides the basis for a successful translation into the clinic.
Collapse
Affiliation(s)
- Maureen Walberer
- 1 Department of Neurology, University Hospital of Cologne, Cologne, Germany ; 2 Max-Planck-Institute for Metabolism Research, Cologne, Germany ; 3 Animal Welfare Office, University of Cologne, Germany
| | - Maria Adele Rueger
- 1 Department of Neurology, University Hospital of Cologne, Cologne, Germany ; 2 Max-Planck-Institute for Metabolism Research, Cologne, Germany ; 3 Animal Welfare Office, University of Cologne, Germany
| |
Collapse
|
26
|
Murray KN, Parry-Jones AR, Allan SM. Interleukin-1 and acute brain injury. Front Cell Neurosci 2015; 9:18. [PMID: 25705177 PMCID: PMC4319479 DOI: 10.3389/fncel.2015.00018] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/12/2015] [Indexed: 01/05/2023] Open
Abstract
Inflammation is the key host-defense response to infection and injury, yet also a major contributor to a diverse range of diseases, both peripheral and central in origin. Brain injury as a result of stroke or trauma is a leading cause of death and disability worldwide, yet there are no effective treatments, resulting in enormous social and economic costs. Increasing evidence, both preclinical and clinical, highlights inflammation as an important factor in stroke, both in determining outcome and as a contributor to risk. A number of inflammatory mediators have been proposed as key targets for intervention to reduce the burden of stroke, several reaching clinical trial, but as yet yielding no success. Many factors could explain these failures, including the lack of robust preclinical evidence and poorly designed clinical trials, in addition to the complex nature of the clinical condition. Lack of consideration in preclinical studies of associated co-morbidities prevalent in the clinical stroke population is now seen as an important omission in previous work. These co-morbidities (atherosclerosis, hypertension, diabetes, infection) have a strong inflammatory component, supporting the need for greater understanding of how inflammation contributes to acute brain injury. Interleukin (IL)-1 is the prototypical pro-inflammatory cytokine, first identified many years ago as the endogenous pyrogen. Research over the last 20 years or so reveals that IL-1 is an important mediator of neuronal injury and blocking the actions of IL-1 is beneficial in a number of experimental models of brain damage. Mechanisms underlying the actions of IL-1 in brain injury remain unclear, though increasing evidence indicates the cerebrovasculature as a key target. Recent literature supporting this and other aspects of how IL-1 and systemic inflammation in general contribute to acute brain injury are discussed in this review.
Collapse
Affiliation(s)
- Katie N Murray
- Faculty of Life Sciences, University of Manchester Manchester, UK
| | | | - Stuart M Allan
- Faculty of Life Sciences, University of Manchester Manchester, UK
| |
Collapse
|
27
|
Doll D, Barr TL, Simpkins JW. Cytokines: their role in stroke and potential use as biomarkers and therapeutic targets. Aging Dis 2014; 5:294-306. [PMID: 25276489 DOI: 10.14336/ad.2014.0500294] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 05/16/2014] [Accepted: 05/27/2014] [Indexed: 12/30/2022] Open
Abstract
Inflammatory mechanisms both in the periphery and in the CNS are important in the pathophysiologic processes occurring after the onset of ischemic stroke (IS). Cytokines are key players in the inflammatory mechanism and contribute to the progression of ischemic damage. This literature review focuses on the effects of inflammation on ischemic stroke, and the role pro-inflammatory and anti-inflammatory cytokines play on deleterious or beneficial stroke outcome. The discovery of biomarkers and novel therapeutics for stroke has been the focus of extensive research recently; thus, understanding the roles of pro-inflammatory and anti-inflammatory cytokines that are up-regulated during stroke will help us further understand how inflammation contributes to the progression of ischemic damage and provide potential targets for novel therapeutics and biomarkers for diagnosis and prognosis of stroke.
Collapse
Affiliation(s)
| | - Taura L Barr
- School of Nursing, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA ; Center for Basic and Translational Stroke Research, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - James W Simpkins
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA ; Center for Basic and Translational Stroke Research, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| |
Collapse
|
28
|
Famakin BM. The Immune Response to Acute Focal Cerebral Ischemia and Associated Post-stroke Immunodepression: A Focused Review. Aging Dis 2014; 5:307-26. [PMID: 25276490 DOI: 10.14336/ad.2014.0500307] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022] Open
Abstract
It is currently well established that the immune system is activated in response to transient or focal cerebral ischemia. This acute immune activation occurs in response to damage, and injury, to components of the neurovascular unit and is mediated by the innate and adaptive arms of the immune response. The initial immune activation is rapid, occurs via the innate immune response and leads to inflammation. The inflammatory mediators produced during the innate immune response in turn lead to recruitment of inflammatory cells and the production of more inflammatory mediators that result in activation of the adaptive immune response. Under ideal conditions, this inflammation gives way to tissue repair and attempts at regeneration. However, for reasons that are just being understood, immunosuppression occurs following acute stroke leading to post-stroke immunodepression. This review focuses on the current state of knowledge regarding innate and adaptive immune activation in response to focal cerebral ischemia as well as the immunodepression that can occur following stroke. A better understanding of the intricate and complex events that take place following immune response activation, to acute cerebral ischemia, is imperative for the development of effective novel immunomodulatory therapies for the treatment of acute stroke.
Collapse
Affiliation(s)
- Bolanle M Famakin
- National Institutes of Health, National Institute of Neurological Diseases and Stroke, Stroke Branch, Branch, Bethesda, MD, 20892, USA
| |
Collapse
|
29
|
Xie L, Sun F, Wang J, Mao X, Xie L, Yang SH, Su DM, Simpkins JW, Greenberg DA, Jin K. mTOR signaling inhibition modulates macrophage/microglia-mediated neuroinflammation and secondary injury via regulatory T cells after focal ischemia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:6009-19. [PMID: 24829408 PMCID: PMC4128178 DOI: 10.4049/jimmunol.1303492] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Signaling by the mammalian target of rapamycin (mTOR) plays an important role in the modulation of both innate and adaptive immune responses. However, the role and underlying mechanism of mTOR signaling in poststroke neuroinflammation are largely unexplored. In this study, we injected rapamycin, a mTOR inhibitor, by the intracerebroventricular route 6 h after focal ischemic stroke in rats. We found that rapamycin significantly reduced lesion volume and improved behavioral deficits. Notably, infiltration of γδ T cells and granulocytes, which are detrimental to the ischemic brain, was profoundly reduced after rapamycin treatment, as was the production of proinflammatory cytokines and chemokines by macrophages and microglia. Rapamycin treatment prevented brain macrophage polarization toward the M1 type. In addition, we also found that rapamycin significantly enhanced anti-inflammation activity of regulatory T cells (Tregs), which decreased production of proinflammatory cytokines and chemokines by macrophages and microglia. Depletion of Tregs partially elevated macrophage/microglia-induced neuroinflammation after stroke. Our data suggest that rapamycin can attenuate secondary injury and motor deficits after focal ischemia by enhancing the anti-inflammation activity of Tregs to restrain poststroke neuroinflammation.
Collapse
Affiliation(s)
- Luokun Xie
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Fen Sun
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Jixian Wang
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107; Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - XiaoOu Mao
- Buck Institute for Research on Aging, Novato, CA 94945; and
| | - Lin Xie
- Buck Institute for Research on Aging, Novato, CA 94945; and
| | - Shao-Hua Yang
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Dong-Ming Su
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - James W Simpkins
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107; Department of Physiology and Pharmacology, Center for Neuroscience, Health Science Center, West Virginia University, Morgantown, WV 26506
| | | | - Kunlin Jin
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX 76107;
| |
Collapse
|
30
|
Dacho AK, Dietz A, Mueller K. Histological effect on the adipocutaneous flap in rats after preconditioning with 2-chloro-N(6) -cyclopentyladenosine. Head Neck 2013; 36:1189-99. [PMID: 23893540 DOI: 10.1002/hed.23433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 03/03/2013] [Accepted: 07/04/2013] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND 2-chloro-N(6) -cyclopentyladenosine (CCPA) was proven to be a protective factor in ischemic reperfusion injury. The purpose of this study was to determine how CCPA would affect the single tissue layers of the adipocutaneous flap. METHODS Seventy male Wistar rats were divided into 5 experimental groups. Samples were taken of the area of flap necrosis and the wound margin after classical or pharmacological preconditioning on the fifth postoperative day. All samples were fixed in formaldehyde, embedded in paraplast, and analyzed in 3- to 4-μm sections (hemalaun-eosin stain and light microscopy). RESULTS In general, wound healing was alike and remained unaffected by the experimental design. The most sensitive part of the flap during preconditioning is the subcutis. The number of neutrophils and of plasma cells is reduced significantly (p < .05). CONCLUSION CCPA has an effect on each tissue layer of the flap. Subcutis became apparent as the most sensitive layer. CCPA influences complement pathway and neutrophils directly and indirectly.
Collapse
Affiliation(s)
- Andreas K Dacho
- Department of Plastic Surgery, St. Josef Medical Center, Essen, Germany
| | | | | |
Collapse
|
31
|
Bayat M, Azami Tameh A, Hossein Ghahremani M, Akbari M, Mehr SE, Khanavi M, Hassanzadeh G. Neuroprotective properties of Melissa officinalis after hypoxic-ischemic injury both in vitro and in vivo. ACTA ACUST UNITED AC 2012; 20:42. [PMID: 23351182 PMCID: PMC3555743 DOI: 10.1186/2008-2231-20-42] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 09/26/2012] [Indexed: 01/13/2023]
Abstract
Background Brain ischemia initiates several metabolic events leading to neuronal death. These events mediate large amount of damage that arises after some neurodegenerative disorders as well as transient brain ischemia. Melissa officinalis is considered as a helpful herbal plant in the prevention of various neurological diseases like Alzheimer that is related with oxidative stress. Methods We examined the effect of Melissa officinalis on hypoxia induced neuronal death in a cortical neuronal culture system as in vitro model and transient hippocampal ischemia as in vivo model. Transient hippocampal ischemia was induced in male rats by tow vessel-occlusion for 20 min. After reperfusion, the histopathological changes and the levels inflammation, oxidative stress status, and caspase-3 activity in hippocampus were measured. Results Cytotoxicity assays showed a significant protection of a 10 μg/ml dose of Melissa against hypoxia in cultured neurons which was confirmed by a conventional staining (P<0.05). Melissa treatment decrease caspase3 activity (P<0.05) and TUNEL-positive cells significantly (P<0.01). Melissa oil has also inhibited malon dialdehyde level and attenuated decrease of Antioxidant Capacity in the hippocampus. Pro-inflammatory cytokines TNF-α, IL-1β and HIF-1α mRNA levels were highly increased after ischemia and treatment with Melissa significantly suppressed HIF-1α gene expression (P<0.05). Discussion Results showed that Melissa officinalis could be considered as a protective agent in various neurological diseases associated with ischemic brain injury.
Collapse
Affiliation(s)
- Mohammad Bayat
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Inflammation is a hallmark of stroke pathology. The cytokines, tumor necrosis factor (TNF), interleukin (IL)-1, and IL-6, modulate tissue injury in experimental stroke and are therefore potential targets in future stroke therapy. The effect of these cytokines on infarct evolution depends on their availability in the ischemic penumbra in the early phase after stroke onset, corresponding to the therapeutic window (<4.5 hours), which is similar in human and experimental stroke. This review summarizes a large body of literature on the spatiotemporal and cellular production of TNF, IL-1, and IL-6, focusing on the early phase in experimental and human stroke. We also review studies of cytokines in blood and cerebrospinal fluid in stroke. Tumor necrosis factor and IL-1 are upregulated early in peri-infarct microglia. Newer literature suggests that IL-6 is produced by microglia, in addition to neurons. Tumor necrosis factor- and IL-1-producing macrophages infiltrate the infarct and peri-infarct with a delay. This information is discussed in the context of suggestions that neuronal sensitivity to ischemia may be modulated by cytokines. The fact that TNF and IL-1, and suppossedly also IL-6, are produced by microglia within the therapeutic window place these cells centrally in potential future stroke therapy.
Collapse
Affiliation(s)
- Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | | |
Collapse
|
33
|
Oliveira-Filho J, Dias JDS, Jesus PA, Neto NJ, Aras R, Reis FJ, Furie KL. Clinical assessment, neuroimaging and immunomarkers in Chagas disease study (CLINICS): Rationale, study design and preliminary findings. Dement Neuropsychol 2012; 6:180-187. [PMID: 29213794 PMCID: PMC5618967 DOI: 10.1590/s1980-57642012dn06030012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 05/20/2012] [Indexed: 11/21/2022] Open
Abstract
Chagas disease (CD) is an important cause of cardiomyopathy and stroke in Brazil. Brain infarcts and atrophy seem to occur independently of cardiomyopathy severity and cognitive impairment is understudied. OBJECTIVE Compare the prevalence of brain magnetic resonance imaging abnormalities between patients with or without CD; determine if inflammatory biomarkers are increased in CD; and determine the efficacy of aspirin in reducing the rate of microembolization in these patients. METHODS 500 consecutive patients with heart failure will undergo a structured cognitive evaluation, biomarker collection and search for microembolic signals on transcranial Doppler. The first 90 patients are described, evaluated with cognitive tests and brain magnetic resonance imaging to measure N-acetyl aspartate (NAA), choline (Cho), myo-inositol (MI) and creatine (Cr). RESULTS Mean age was 55±11 years, 51% female, 38 (42%) with CD. Mean NAA/Cr ratio was lower in patients with CD as compared to other cardiomyopathies. Long-term memory and clock-drawing test were also significantly worse in CD patients. In the multivariable analysis correcting for ejection fraction, age, sex and educational level, reduced NAA/Cr (p=0.006) and cognitive dysfunction (long-term memory, p=0.023; clock-drawing test, p=0.015) remained associated with CD. CONCLUSION In this preliminary sample, CD was associated with cognitive impairment and decreased NAA/Cr independently of cardiac function or educational level.
Collapse
Affiliation(s)
- Jamary Oliveira-Filho
- From the Stroke and Cardiomyopathy Clinics of the Hospital Universitario
Professor Edgard Santos, Federal University of Bahia, BA, Brazil; and the Stroke
Service of Massachusetts General Hospital, Harvard University, USA
| | - Jesângeli de S. Dias
- From the Stroke and Cardiomyopathy Clinics of the Hospital Universitario
Professor Edgard Santos, Federal University of Bahia, BA, Brazil; and the Stroke
Service of Massachusetts General Hospital, Harvard University, USA
| | - Pedro A.P. Jesus
- From the Stroke and Cardiomyopathy Clinics of the Hospital Universitario
Professor Edgard Santos, Federal University of Bahia, BA, Brazil; and the Stroke
Service of Massachusetts General Hospital, Harvard University, USA
| | - Nestor J.S.B. Neto
- From the Stroke and Cardiomyopathy Clinics of the Hospital Universitario
Professor Edgard Santos, Federal University of Bahia, BA, Brazil; and the Stroke
Service of Massachusetts General Hospital, Harvard University, USA
| | - Roque Aras
- From the Stroke and Cardiomyopathy Clinics of the Hospital Universitario
Professor Edgard Santos, Federal University of Bahia, BA, Brazil; and the Stroke
Service of Massachusetts General Hospital, Harvard University, USA
| | - Francisco J.F.B. Reis
- From the Stroke and Cardiomyopathy Clinics of the Hospital Universitario
Professor Edgard Santos, Federal University of Bahia, BA, Brazil; and the Stroke
Service of Massachusetts General Hospital, Harvard University, USA
| | - Karen L. Furie
- From the Stroke and Cardiomyopathy Clinics of the Hospital Universitario
Professor Edgard Santos, Federal University of Bahia, BA, Brazil; and the Stroke
Service of Massachusetts General Hospital, Harvard University, USA
| |
Collapse
|
34
|
Stress and social isolation increase vulnerability to stroke. Exp Neurol 2012; 233:33-9. [DOI: 10.1016/j.expneurol.2011.01.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Accepted: 01/24/2011] [Indexed: 01/18/2023]
|
35
|
Greifzu F, Schmidt S, Schmidt KF, Kreikemeier K, Witte OW, Löwel S. Global impairment and therapeutic restoration of visual plasticity mechanisms after a localized cortical stroke. Proc Natl Acad Sci U S A 2011; 108:15450-5. [PMID: 21873250 PMCID: PMC3174619 DOI: 10.1073/pnas.1016458108] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We tested the influence of a photothrombotic lesion in somatosensory cortex on plasticity in the mouse visual system and the efficacy of anti-inflammatory treatment to rescue compromised learning. To challenge plasticity mechanisms, we induced monocular deprivation (MD) in 3-mo-old mice. In control animals, MD induced an increase of visual acuity of the open eye and an ocular dominance (OD) shift towards this eye. In contrast, after photothrombosis, there was neither an enhancement of visual acuity nor an OD-shift. However, OD-plasticity was present in the hemisphere contralateral to the lesion. Anti-inflammatory treatment restored sensory learning but not OD-plasticity, as did a 2-wk delay between photothrombosis and MD. We conclude that (i) both sensory learning and cortical plasticity are compromised in the surround of a cortical lesion; (ii) transient inflammation is responsible for impaired sensory learning, suggesting anti-inflammatory treatment as a useful adjuvant therapy to support rehabilitation following stroke; and (iii) OD-plasticity cannot be conceptualized solely as a local process because nonlocal influences are more important than previously assumed.
Collapse
Affiliation(s)
- Franziska Greifzu
- Institut für Allgemeine Zoologie und Tierphysiologie, Friedrich-Schiller-Universität Jena, D-07743 Jena, Germany; and
| | - Silvio Schmidt
- Hans-Berger Klinik für Neurologie, Universitätsklinikum Jena, D-07747 Jena, Germany
| | - Karl-Friedrich Schmidt
- Institut für Allgemeine Zoologie und Tierphysiologie, Friedrich-Schiller-Universität Jena, D-07743 Jena, Germany; and
| | - Klaus Kreikemeier
- Institut für Allgemeine Zoologie und Tierphysiologie, Friedrich-Schiller-Universität Jena, D-07743 Jena, Germany; and
| | - Otto W. Witte
- Hans-Berger Klinik für Neurologie, Universitätsklinikum Jena, D-07747 Jena, Germany
| | - Siegrid Löwel
- Institut für Allgemeine Zoologie und Tierphysiologie, Friedrich-Schiller-Universität Jena, D-07743 Jena, Germany; and
| |
Collapse
|
36
|
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
|
37
|
Inácio AR, Bucala R, Deierborg T. Lack of macrophage migration inhibitory factor in mice does not affect hallmarks of the inflammatory/immune response during the first week after stroke. J Neuroinflammation 2011; 8:75. [PMID: 21714902 PMCID: PMC3152909 DOI: 10.1186/1742-2094-8-75] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Accepted: 06/29/2011] [Indexed: 11/10/2022] Open
Abstract
Background Macrophage migration inhibitory factor (MIF) has been proposed to play a detrimental role in stroke. We recently showed that MIF promotes neuronal death and aggravates neurological deficits during the first week after experimental stroke, in mice. Since MIF regulates tissue inflammation, we studied the putative role of MIF in post-stroke inflammation. Methods We subjected C57BL/6 mice, Mif-/- (MIF-KO) or Mif+/+ (WT), to a transient occlusion of the right middle cerebral artery (tMCAo) or sham-surgery. We studied MIF expression, GFAP expression and the number of CD74-positive cells in the ischemic brain hemisphere 7 days after tMCAo using primarily immunohistochemistry. We determined IFN-γ, IL-2, IL-4, IL-5, IL-10, IL-12, KC/CXCL-1 and TNF-α protein levels in the brain (48 h after surgery) and serum (48 h and 7 days after surgery) by a multiplex immunoassay. Results We observed that MIF accumulates in neurons and astrocytes of the peri-infarct region, as well as in microglia/macrophages of the infarct core up to 7 days after stroke. Among the inflammatory mediators analyzed, we found a significant increase in cerebral IL-12 and KC levels after tMCAo, in comparison to sham-surgery. Importantly, the deletion of Mif did not significantly affect the levels of the cytokines evaluated, in the brain or serum. Moreover, the spleen weight 48 h and 7 days subsequent to tMCAo was similar in WT and MIF-KO mice. Finally, the extent of GFAP immunoreactivity and the number of MIF receptor (CD74)-positive cells within the ischemic brain hemisphere did not differ significantly between WT and MIF-KO mice subjected to tMCAo. Conclusions We conclude that MIF does not affect major components of the inflammatory/immune response during the first week after experimental stroke. Based on present and previous evidence, we propose that the deleterious MIF-mediated effects in stroke depend primarily on an intraneuronal and/or interneuronal action.
Collapse
Affiliation(s)
- Ana R Inácio
- Laboratory for Experimental Brain Research, Department of Clinical Sciences, Lund University, BMC A13, 22184 Lund, Sweden.
| | | | | |
Collapse
|
38
|
Chaung WW, Wu R, Ji Y, Wang Z, Dong W, Cheyuo C, Qi L, Qiang X, Wang H, Wang P. Peripheral administration of human adrenomedullin and its binding protein attenuates stroke-induced apoptosis and brain injury in rats. Mol Med 2011; 17:1075-83. [PMID: 21695352 DOI: 10.2119/molmed.2010.00104] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 06/16/2011] [Indexed: 11/06/2022] Open
Abstract
Stroke is a leading cause of death and the primary medical cause of acquired adult disability worldwide. The progressive brain injury after acute stroke is partly mediated by ischemia-elicited inflammatory responses. The vasoactive hormone adrenomedullin (AM), upregulated under various inflammatory conditions, counterbalances inflammatory responses. However, regulation of AM activity in ischemic stroke remains largely unknown. Recent studies have demonstrated the presence of a specific AM binding protein (that is, AMBP-1) in mammalian blood. AMBP-1 potentiates AM biological activities. Using a rat model of focal cerebral ischemia induced by permanent middle cerebral artery occlusion (MCAO), we found that plasma levels of AM increased significantly, whereas plasma levels of AMBP-1 decreased significantly after stroke. When given peripherally early after MCAO, exogenous human AM in combination with human AMBP-1 reduced brain infarct volume 24 and 72 h after MCAO, an effect not observed after the treatment by human AM or human AMBP-1 alone. Furthermore, treatment of human AM/AMBP-1 reduced neuron apoptosis and morphological damage, inhibited neutrophil infiltration in the brain and decreased serum levels of S100B and lactate. Thus, human AM/AMBP-1 has the ability to reduce stroke-induced brain injury in rats. AM/AMBP-1 can be developed as a novel therapeutic agent for patients with ischemic stroke.
Collapse
Affiliation(s)
- Wayne W Chaung
- The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
The Cerebral Microvasculature and Responses to Ischemia. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
40
|
|
41
|
Saino O, Taguchi A, Nakagomi T, Nakano-Doi A, Kashiwamura SI, Doe N, Nakagomi N, Soma T, Yoshikawa H, Stern DM, Okamura H, Matsuyama T. Immunodeficiency reduces neural stem/progenitor cell apoptosis and enhances neurogenesis in the cerebral cortex after stroke. J Neurosci Res 2010; 88:2385-97. [PMID: 20623538 DOI: 10.1002/jnr.22410] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Acute inflammation in the poststroke period exacerbates neuronal damage and stimulates reparative mechanisms, including neurogenesis. However, only a small fraction of neural stem/progenitor cells survives. In this report, by using a highly reproducible model of cortical infarction in SCID mice, we examined the effects of immunodeficiency on reduction of brain injury, survival of neural stem/progenitor cells, and functional recovery. Subsequently, the contribution of T lymphocytes to neurogenesis was evaluated in mice depleted for each subset of T lymphocyte. SCID mice revealed the reduced apoptosis and enhanced proliferation of neural stem/progenitor cells induced by cerebral cortex after stroke compared with the immunocompetent wild-type mice. Removal of T lymphocytes, especially the CD4(+) T-cell population, enhanced generation of neural stem/progenitor cells, followed by accelerated functional recovery. In contrast, removal of CD25(+) T cells, a cell population including regulatory T lymphocytes, impaired functional recovery through, at least in part, suppression of neurogenesis. Our findings demonstrate a key role of T lymphocytes in regulation of poststroke neurogenesis and indicate a potential novel strategy for cell therapy in repair of the central nervous system.
Collapse
Affiliation(s)
- Orie Saino
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Chen HN, Hsieh CL. Effects of Sophora japonica flowers (Huaihua) on cerebral infarction. Chin Med 2010; 5:34. [PMID: 20875105 PMCID: PMC2954919 DOI: 10.1186/1749-8546-5-34] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 09/27/2010] [Indexed: 11/12/2022] Open
Abstract
The dried flowers and buds of Sophora japonica are used as a medicinal herb in China, Japan and Korea to treat bleeding hemorrhoids and hematemesis. This article presents an overview of the effects of Sophora japonica on cerebral infarction based on literature searched from Medline, PubMed, Cochrane Library and the China National Knowledge Infrastructure (CNKI). Sophora japonica contains both anti-hemorrhagic and anti-hemostatic substances. Sophora japonica reduces cerebral infarction partly as a result of its anti-oxidative and anti-inflammatory activities. Previous studies found that Sophora japonica reduced the size of cerebral infarction and neurological deficits and reduced microglial activation, interleukin-1β release and number of apoptotic cells in ischemia-reperfusion injured Sprague-Dawley rats. Further study is required to determine the relationship between Sophora japonica-mediated reduction in cerebral infarction size and the effects of Sophora japonica on platelet aggregation and cardiovascular function.
Collapse
Affiliation(s)
- Hsiang-Ni Chen
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40402, Taiwan.
| | | |
Collapse
|
43
|
Denes A, Thornton P, Rothwell NJ, Allan SM. Inflammation and brain injury: acute cerebral ischaemia, peripheral and central inflammation. Brain Behav Immun 2010; 24:708-23. [PMID: 19770034 DOI: 10.1016/j.bbi.2009.09.010] [Citation(s) in RCA: 226] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 09/15/2009] [Accepted: 09/15/2009] [Indexed: 12/18/2022] Open
Abstract
Inflammation is a classical host defence response to infection and injury that has many beneficial effects. However, inappropriate (in time, place and magnitude) inflammation is increasingly implicated in diverse disease states, now including cancer, diabetes, obesity, atherosclerosis, heart disease and, most relevant here, CNS disease. A growing literature shows strong correlations between inflammatory status and the risk of cerebral ischaemia (CI, most commonly stroke), as well as with outcome from an ischaemic event. Intervention studies to demonstrate a causal link between inflammation and CI (or its consequences) are limited but are beginning to emerge, while experimental studies of CI have provided direct evidence that key inflammatory mediators (cytokines, chemokines and inflammatory cells) contribute directly to ischaemic brain injury. However, it remains to be determined what the relative importance of systemic (largely peripheral) versus CNS inflammation is in CI. Animal models in which CI is driven by a CNS intervention may not accurately reflect the clinical condition; stroke being typically induced by atherosclerosis or cardiac dysfunction, and hence current experimental paradigms may underestimate the contribution of peripheral inflammation. Experimental studies have already identified a number of potential anti-inflammatory therapeutic interventions that may limit ischaemic brain damage, some of which have been tested in early clinical trials with potentially promising results. However, a greater understanding of the contribution of inflammation to CI is still required, and this review highlights some of the key mechanism that may offer future therapeutic targets.
Collapse
Affiliation(s)
- A Denes
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
44
|
Jiang L, Womble T, Saporta S, Chen N, Sanberg CD, Sanberg PR, Willing AE. Human umbilical cord blood cells decrease microglial survival in vitro. Stem Cells Dev 2010; 19:221-8. [PMID: 19788371 DOI: 10.1089/scd.2009.0170] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
When human umbilical cord blood (HUCB) cells are systemically administered following middle cerebral artery occlusion (MCAO) in rats, they produce a reduction in infarct size resulting in recovery of motor function. Rats receiving HUCB cells have a less severe inflammatory response compared to MCAO stroke rats. The purpose of this study was to determine the interaction between HUCB cells and the main resident immune cells of the brain (microglia) under normoxic and hypoxic conditions in vitro. Primary microglial cultures were incubated for 2 h in no oxygen (95% N, 5% CO(2)) and low glucose (1%) media. Mononuclear HUCB cells were added to half the cultures at the beginning of the hypoxia conditions. Microglial viability was determined using fluorescein diacetate/propidium iodide (FDA/PI) labeling and cytokine expression using ELISA. In some studies, CD11b+ or CD19+ cells isolated from the HUCB mononuclear fraction with magnetic antibody cell sorting (MACS) were used instead of the mononuclear fraction. Co-culturing mononuclear HUCB cells with microglia decreased viability of the microglia during hypoxia. In the microglial monocultures, hypoxia significantly increased release of IL-1beta compared to normoxia, while adding HUCB cells in the hypoxia condition decreased IL-1beta concentrations to the same level as in the normoxia monocultures. Both CD11b+ and CD19+ HUCB cells decreased microglial viability during normoxia and hypoxia. Our data suggest that HUCB cells may produce a soluble factor that decreases viability of microglia.
Collapse
Affiliation(s)
- Lixian Jiang
- Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, Florida 33612, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The cellular mechanisms underlying neuronal loss and neurodegeneration have been an area of interest in the last decade. Although neurodegenerative diseases such as Alzheimer disease, Parkinson disease, and Huntington disease each have distinct clinical symptoms and pathologies, they all share common mechanisms such as protein aggregation, oxidative injury, inflammation, apoptosis, and mitochondrial injury that contribute to neuronal loss. Although cerebrovascular disease has different causes from the neurodegenerative disorders, many of the same common disease mechanisms come into play following a stroke. Novel therapies that target each of these mechanisms may be effective in decreasing the risk of disease, abating symptoms, or slowing down their progression. Although most of these therapies are experimental, and require further investigation, a few seem to offer promise.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, 63108
- Department of Neurology, Washington University School of Medicine, St Louis, MO, 63108
| | - James E. Galvin
- Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, 63108
- Department of Neurobiology, Washington University School of Medicine, St Louis, MO, 63108
| |
Collapse
|
46
|
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]
|
47
|
Laskowitz DT, Grocott H, Hsia A, Copeland KR. Serum markers of cerebral ischemia. J Stroke Cerebrovasc Dis 2009; 7:234-41. [PMID: 17895090 DOI: 10.1016/s1052-3057(98)80032-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/1997] [Accepted: 01/30/1998] [Indexed: 12/20/2022] Open
Abstract
Rapid diagnosis and management of stroke patients is becoming increasingly important with the emergence of new interventional strategies for acute cerebral ischemia. A biochemical surrogate of cerebral ischemia, rapidly detectable in the serum before radiological diagnosis, might have clinical utility in the setting of acute stroke, high-risk cardiovascular procedures, and subarachnoid hemorrhage. Such a marker might also aid in the neurological prognosis of anoxic brain injury. Several serum markers have been evaluated in acute cerebral ischemia. These include neuronal enzymes such as neuron-specific enolase; markers of glial injury and activation, such as protein S100beta; and mediators of inflammation, such as interleukin-6. The clinical and preclinical data supporting the use of these biochemical surrogates of cerebral ischemia are reviewed.
Collapse
Affiliation(s)
- D T Laskowitz
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | | | | | |
Collapse
|
48
|
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]
|
49
|
Offner H, Vandenbark AA, Hurn PD. Effect of experimental stroke on peripheral immunity: CNS ischemia induces profound immunosuppression. Neuroscience 2009; 158:1098-111. [PMID: 18597949 PMCID: PMC2666964 DOI: 10.1016/j.neuroscience.2008.05.033] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Revised: 05/12/2008] [Accepted: 05/13/2008] [Indexed: 11/27/2022]
Abstract
The profound damage to the CNS caused by ischemic lesions has been well documented. Yet, relatively little is known about the contribution to and effects on the immune system during stroke. We have focused on both early and late events in the peripheral immune system during stroke in mice and have observed an early activation of splenocytes that conceivably could result in immune-mediated damage in the developing CNS lesion, followed by global immunosuppression that affects the spleen, thymus, lymph nodes and circulation. While this second immunosuppressive phase may not directly enhance infarction size, it without doubt leads to an inability to respond to antigenic challenges, thereby enhancing the risk for crippling systemic infection and septicemia in stroke survivors. These novel findings advocate the need to develop or effectively utilize agents that can block early neural splenic activation and modulate immune cells specific for brain antigens as a means to prevent mobilization of T and B cells carrying a cytokine death warrant to the brain. Equally important for the recovering stroke patient are approaches that can derail the second phase of immune dysfunction and restore the ability to mount a defense against systemic infectious insults.
Collapse
Affiliation(s)
- H Offner
- Neuroimmunology Research R&D-31, Portland Veterans Affairs Medical Center, 3710 Southwest US Veterans Hospital Road, and Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | |
Collapse
|
50
|
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
|