1
|
Meng F, Zhou Y, Wagner A, Bülow JM, Köhler K, Neunaber C, Bundkirchen K, Relja B. Impact of age on liver damage, inflammation, and molecular signaling pathways in response to femoral fracture and hemorrhage. Front Immunol 2023; 14:1239145. [PMID: 37691959 PMCID: PMC10484338 DOI: 10.3389/fimmu.2023.1239145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Background Trauma causes disability and mortality globally, leading to fractures and hemorrhagic shock. This can trigger an irregular inflammatory response that damages remote organs, including liver. Aging increases the susceptibility to dysregulated immune responses following trauma, raising the risk of organ damage, infections, and higher morbidity and mortality in elderly patients. This study investigates how aging affects liver inflammation and damage post-trauma. Methods 24 male C57BL/6J mice were randomly divided into four groups. Twelve young (17-26 weeks) and 12 aged (64-72 weeks) mice were included. Mice further underwent either hemorrhagic shock (trauma/hemorrhage, TH), and femoral fracture (osteotomy) with external fixation (Fx) (THFx, n=6) or sham procedures (n=6). After 24 hours, mice were sacrificed. Liver injury and apoptosis were evaluated using hematoxylin-eosin staining and activated caspase-3 immunostaining. CXCL1 and infiltrating polymorphonuclear leukocytes (PMNL) in the liver were assessed by immunostaining, and concentrations of CXCL1, TNF, IL-1β, and IL-10 in the liver tissue were determined by ELISA. Gene expression of Tnf, Cxcl1, Il-1β, and Cxcl2 in the liver tissue was determined by qRT-PCR. Finally, western blot was used to determine protein expression levels of IκBα, Akt, and their phosphorylated forms. Results THFx caused liver damage and increased presence of active caspase-3-positive cells compared to the corresponding sham group. THFx aged group had more severe liver injury than the young group. CXCL1 and PMNL levels were significantly higher in both aged groups, and THFx caused a greater increase in CXCL and PMNL levels in aged compared to the young group. Pro-inflammatory TNF and IL-1β levels were elevated in aged groups, further intensified by THFx. Anti-inflammatory IL-10 levels were lower in aged groups. Tnf and Cxcl1 gene expression was enhanced in the aged sham group. Phosphorylation ratio of IκBα was significantly increased in the aged sham group versus young sham group. THFx-induced IκBα phosphorylation in the young group was significantly reduced in the aged THFx group. Akt phosphorylation was significantly reduced in the THFx aged group compared to the THFx young group. Conclusion The findings indicate that aging may lead to increased vulnerability to liver injury and inflammation following trauma due to dysregulated immune responses.
Collapse
Affiliation(s)
- Fanshuai Meng
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
- Department of Trauma and Reconstructive Surgery, Uniklinik RWTH Aachen, Aachen, Germany
| | - Yuzhuo Zhou
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Alessa Wagner
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
| | - Jasmin Maria Bülow
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Claudia Neunaber
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Katrin Bundkirchen
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Borna Relja
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, Translational and Experimental Trauma Research, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
2
|
Shimamura M, Yang Chang C, Hayashi H, Baba S, Yoshida S, Nakagami H. Impact of insufficient recovery of cerebral blood flow due to partial reperfusion on the pathophysiology of ischemic stroke in mice. Neurosci Lett 2023; 810:137370. [PMID: 37399972 DOI: 10.1016/j.neulet.2023.137370] [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: 05/05/2023] [Revised: 05/18/2023] [Accepted: 06/30/2023] [Indexed: 07/05/2023]
Abstract
Recent advances in endovascular treatment (EVT) dramatically changed the outcome of ischemic stroke, but partial reperfusion does not improve the outcome as well as no reperfusion. Although partial reperfusion is estimated to be more potential for therapeutic intervention than permanent occlusion due to some blood supply, their pathophysiological difference is still unknown. To answer the question, we analyzed the difference in mice, which were exposed to distal middle cerebral artery occlusion with 14-min common carotid artery (CCA) occlusion (partial reperfusion) or permanent CCA occlusion (no reperfusion). Although the final infarct volume was same between permanent and partial reperfusion, Fluoro-jade C staining showed that neurodegeneration was inhibited both in the severe and moderate ischemic region 3 h after partial reperfusion. Also, partial reperfusion increased the number of TUNEL-positive cells only in the severe ischemic region. IgG extravasation was suppressed at 24 h only in the moderate ischemic region in partial reperfusion. Injected FITC-dextran was observed in the brain parenchyma with BBB leakage at 24 h in partial reperfusion, but not in permanent occlusion. The expression of il1β and il6 mRNA was inhibited in the severe ischemic region. Thus, partial reperfusion showed region-dependent favorable pathophysiology, such as delayed neurodegeneration, suppressed BBB destruction and inflammation, and potential for drug delivery, when compared to permanent occlusion. Further studies on the molecular differences and effectiveness of drugs will shed light on the development of novel treatments for partial reperfusion in ischemic stroke.
Collapse
Affiliation(s)
- Munehisa Shimamura
- Department of Gene & Stem Cell Regenerative Therapy, Osaka University Graduate School of Medicine, 565-0871, Japan; Department of Neurology, Osaka University Graduate School of Medicine, Japan.
| | - Chin Yang Chang
- Department of Gene & Stem Cell Regenerative Therapy, Osaka University Graduate School of Medicine, 565-0871, Japan
| | - Hiroki Hayashi
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, 565-0871, Japan
| | - Satoshi Baba
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, 565-0871, Japan; Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Japan
| | - Shota Yoshida
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, 565-0871, Japan; Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, 565-0871, Japan
| |
Collapse
|
3
|
Bui TA, Jickling GC, Winship IR. Neutrophil dynamics and inflammaging in acute ischemic stroke: A transcriptomic review. Front Aging Neurosci 2022; 14:1041333. [PMID: 36620775 PMCID: PMC9813499 DOI: 10.3389/fnagi.2022.1041333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Stroke is among the leading causes of death and disability worldwide. Restoring blood flow through recanalization is currently the only acute treatment for cerebral ischemia. Unfortunately, many patients that achieve a complete recanalization fail to regain functional independence. Recent studies indicate that activation of peripheral immune cells, particularly neutrophils, may contribute to microcirculatory failure and futile recanalization. Stroke primarily affects the elderly population, and mortality after endovascular therapies is associated with advanced age. Previous analyses of differential gene expression across injury status and age identify ischemic stroke as a complex age-related disease. It also suggests robust interactions between stroke injury, aging, and inflammation on a cellular and molecular level. Understanding such interactions is crucial in developing effective protective treatments. The global stroke burden will continue to increase with a rapidly aging human population. Unfortunately, the mechanisms of age-dependent vulnerability are poorly defined. In this review, we will discuss how neutrophil-specific gene expression patterns may contribute to poor treatment responses in stroke patients. We will also discuss age-related transcriptional changes that may contribute to poor clinical outcomes and greater susceptibility to cerebrovascular diseases.
Collapse
Affiliation(s)
- Truong An Bui
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C. Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ian R. Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
4
|
Yang LX, Chen FY, Yu HL, Liu PY, Bao XY, Xia SN, Gu Y, Xu Y, Cao X. Poncirin suppresses lipopolysaccharide (LPS)-induced microglial inflammation and ameliorates brain ischemic injury in experimental stroke in mice. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1344. [PMID: 33313089 PMCID: PMC7723616 DOI: 10.21037/atm-20-3470] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Based on accumulating evidence, excessive activation of microglia-mediated inflammatory responses plays an essential role in ischemic stroke. Poncirin (Pon) exerts anti-hyperalgesic, anti-osteoporotic and anti-tumor effects on various diseases. However, the roles of Pon in microglial activation and the underlying mechanism have not been elucidated. This study aimed to explore whether Pon inhibits lipopolysaccharide (LPS)-induced microglial neuroinflammation and protects against brain ischemic injury in experimental stroke in mice. Methods Primary microglia cells were prepared from the cerebral cortices of 1- to 2-day-old C57BL/6J mice. Murine BV2 cells and primary microglia were stimulated with LPS and the effects of a non-cytotoxic concentration of Pon on LPS-stimulated pro-inflammatory factors were measured using real-time PCR and enzyme-linked immunosorbent assays (ELISAs). Western blot analyses were used for mechanistic studies. In an in vivo study, 8-week-old male C57BL/6J mice were subjected to focal cerebral ischemia through middle cerebral artery occlusion (MCAO). Pon (30 mg/kg, i.p.) or the same volume of saline was administered after the MCAO model was established, and the infarct volume was evaluated using 2,3,5-triphenyltetrazolium chloride (TTC) staining. We also evaluated animal behaviours, the expression of pro-inflammatory cytokines and microglial activation in the ischemic hemisphere. Results Pon prevented the release of nitric oxide (NO), prostaglandin E2 (PGE2), interleukin (IL)-1β, IL-6 and tumor necrosis factor-alpha (TNF-α) in both BV2 cells and primary microglia stimulated with LPS. The inhibitory effects of Pon were associated with the regulation of the ERK1/2, JNK and nuclear factor kappa B (NF-κB) signaling pathways. In mice that underwent MCAO, Pon administration decreased the lesion size and improved neurological deficits. Furthermore, Pon attenuated the production of inflammatory cytokines mainly by restraining microglial activation after ischemic stroke. Conclusions Based on the findings from the present study, Pon provides neuroprotection through its anti-inflammatory effects on microglia and it may be a useful treatment for ischemic stroke.
Collapse
Affiliation(s)
- Li-Xuan Yang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Fang-Yu Chen
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Hai-Long Yu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Pin-Yi Liu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Xin-Yu Bao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Sheng-Nan Xia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yue Gu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| |
Collapse
|
5
|
Vezzani B, Carinci M, Patergnani S, Pasquin MP, Guarino A, Aziz N, Pinton P, Simonato M, Giorgi C. The Dichotomous Role of Inflammation in the CNS: A Mitochondrial Point of View. Biomolecules 2020; 10:E1437. [PMID: 33066071 PMCID: PMC7600410 DOI: 10.3390/biom10101437] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/07/2020] [Accepted: 10/10/2020] [Indexed: 12/14/2022] Open
Abstract
Innate immune response is one of our primary defenses against pathogens infection, although, if dysregulated, it represents the leading cause of chronic tissue inflammation. This dualism is even more present in the central nervous system, where neuroinflammation is both important for the activation of reparatory mechanisms and, at the same time, leads to the release of detrimental factors that induce neurons loss. Key players in modulating the neuroinflammatory response are mitochondria. Indeed, they are responsible for a variety of cell mechanisms that control tissue homeostasis, such as autophagy, apoptosis, energy production, and also inflammation. Accordingly, it is widely recognized that mitochondria exert a pivotal role in the development of neurodegenerative diseases, such as multiple sclerosis, Parkinson's and Alzheimer's diseases, as well as in acute brain damage, such in ischemic stroke and epileptic seizures. In this review, we will describe the role of mitochondria molecular signaling in regulating neuroinflammation in central nervous system (CNS) diseases, by focusing on pattern recognition receptors (PRRs) signaling, reactive oxygen species (ROS) production, and mitophagy, giving a hint on the possible therapeutic approaches targeting mitochondrial pathways involved in inflammation.
Collapse
Affiliation(s)
- Bianca Vezzani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Marianna Carinci
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Simone Patergnani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Matteo P. Pasquin
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Annunziata Guarino
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Department of BioMedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Nimra Aziz
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Department of BioMedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy
| | - Michele Simonato
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Department of BioMedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy
- School of Medicine, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| |
Collapse
|
6
|
The Effects of Bee Venom Pharmacopuncture on Middle Cerebral Artery Occlusion Ischemic Cerebral Damage in Mice. JOURNAL OF ACUPUNCTURE RESEARCH 2019. [DOI: 10.13045/jar.2019.00080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
7
|
Bieber M, Gronewold J, Scharf AC, Schuhmann MK, Langhauser F, Hopp S, Mencl S, Geuss E, Leinweber J, Guthmann J, Doeppner TR, Kleinschnitz C, Stoll G, Kraft P, Hermann DM. Validity and Reliability of Neurological Scores in Mice Exposed to Middle Cerebral Artery Occlusion. Stroke 2019; 50:2875-2882. [PMID: 31412755 DOI: 10.1161/strokeaha.119.026652] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background and Purpose- The selection of appropriate neurological scores and tests is crucial for the evaluation of stroke consequences. The validity and reliability of neurological deficit scores and tests has repeatedly been questioned in ischemic stroke models in the past. Methods- In 198 male mice exposed to transient intraluminal middle cerebral artery occlusion, we examined the validity and reliability of 11 neurological scores (Bederson score 0-3, Bederson score 0-4, Bederson score 0-5, modified neurological severity [0-14], subjective overall impression [0-10], or simple neurological tests: grip test, latency to move body length test, pole test, wire hanging test, negative geotaxis test, and elevated body swing test) in the acute stroke phase, that is, after 24 hours. Combinations of neurological scores or tests for predicting infarct volume were statistically analyzed. Results- Infarct volume was left skewed (median [Q1-Q3], 78.4 [54.8-101.3] mm3). Among all tests, the Bederson (0-5; r=0.63, P<0.001), modified neurological severity (r=0.80, P<0.001), and subjective overall impression (r=-0.63, P<0.001) scores had the highest test validities, using infarct volume as external reference. Subjective overall impression had the best agreement between 5 raters (Kendall W=0.11, P<0.001). The Bederson (0-5) score discriminated infarct volume in mice with small (≤50 mm3; r=0.33, P=0.027) and large (>50 mm3; r=0.48, P<0.001) brain infarcts, all other tests only in mice with large infarcts. Combining subjective overall impression with Bederson (0-5) score explained 47.6% of the variance of infarct volume. Conclusions- Despite their simplicity, the Bederson (0-5) score, modified neurological severity score, and subjective overall impression have reasonable validity and reliability in the acute stroke phase. The Bederson (0-5) score equally distinguishes infarct volume in small and large infarcts. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Michael Bieber
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany
| | - Janine Gronewold
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Germany (J. Gronewold, A.-C.S., F.L., S.M., C.K., D.M.H.)
| | - Anne-Carina Scharf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Germany (J. Gronewold, A.-C.S., F.L., S.M., C.K., D.M.H.)
| | - Michael K Schuhmann
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany
| | - Friederike Langhauser
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany.,Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Germany (J. Gronewold, A.-C.S., F.L., S.M., C.K., D.M.H.)
| | - Sarah Hopp
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany.,Neurosurgery (S.H.), University Hospital Würzburg, Germany
| | - Stine Mencl
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany.,Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Germany (J. Gronewold, A.-C.S., F.L., S.M., C.K., D.M.H.)
| | - Eva Geuss
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany
| | - Jonas Leinweber
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany
| | - Josua Guthmann
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University of Göttingen Medical School, Germany (T.R.D.)
| | - Christoph Kleinschnitz
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany.,Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Germany (J. Gronewold, A.-C.S., F.L., S.M., C.K., D.M.H.)
| | - Guido Stoll
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany
| | - Peter Kraft
- From the Departments of Neurology (M.B., M.K.S., F.L., S.M., E.G., J.L., J. Guthmann, C.K., G.S., P.K.), University Hospital Würzburg, Germany.,Department of Neurology, Klinikum Main-Spessart, Lohr, Germany (P.K.)
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Germany (J. Gronewold, A.-C.S., F.L., S.M., C.K., D.M.H.)
| |
Collapse
|
8
|
Enhancing Base Excision Repair of Mitochondrial DNA to Reduce Ischemic Injury Following Reperfusion. Transl Stroke Res 2018; 10:664-671. [PMID: 30535792 PMCID: PMC6842339 DOI: 10.1007/s12975-018-0680-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/19/2018] [Accepted: 11/19/2018] [Indexed: 01/19/2023]
Abstract
We hypothesize that enhancing mitochondrial base excision repair (BER) capability in brain will reduce reperfusion-associated ischemic brain injury. Post-stroke reperfusion was modeled in mice via transient filament occlusion of the middle cerebral artery (60 min) (transient MCAO). Administration of a TAT-modified form of a DNA glycosylase (EndoIII) following reperfusion of the brain reduced resultant brain infarct volume. Protection was dose-dependent, BER enzyme specific, and regionally specific (more effective via the jugular vein). EndoIII is compatible with tissue plasminogen activator (tPA). The time window of a single dose of EndoIII effect is 3 h following reperfusion onset. These data suggest a novel approach to enhance protection of reperfused brain in the setting of revascularization procedures (thrombectomy or thrombolytic therapy) following stroke.
Collapse
|
9
|
Behrouzifar S, Vakili A, Barati M. The Effects of Mouse Recombinant Resistin on mRNA Expression of Proinflammatory and Anti-Inflammatory Cytokines and Heat Shock Protein-70 in Experimental Stroke Model. J Stroke Cerebrovasc Dis 2018; 27:3272-3279. [PMID: 30120034 DOI: 10.1016/j.jstrokecerebrovasdis.2018.07.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/04/2018] [Accepted: 07/16/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Our recent research showed that resistin has a neuroprotective effect against stroke-induced injury through suppressing apoptosis and oxidative stress. However, the molecular mechanism of neuroprotection of resistin is unclear. This work was designed to examine the effect of mouse recombinant resistin on mRNA expression of Tumor necrosis factor-α (TNF-α), Interleukin-1β (IL-1β), Interleukin-10 (IL-10), Transforming growth factor-β1 (TGF- β1), and Heat shock protein-70 (HSP-70) in mouse model of stroke. MATERIALS AND METHODS Transient focal cerebral ischemia was induced by the middle cerebral artery occlusion (MCAO) in mice. TNF-α, IL-1β, IL-10, TGF-β1, and HSP-70 mRNA were detected at sham (0 hour), 3 hours, 6 hours, 12 hours, and 24 hours after MCAO using real-time QRT-PCR method. Moreover, animals were treated with resistin at the dose of 400ng/mouse at the commencement of MCAO, and mRNA expression of the cytokines and HSP-70 was measured 24 hours after MCAO. RESULTS Tumor necrosis factor-α and IL-1β mRNA expression markedly increased at 12-hour time point and then returned to the basal level at 24 hours after MCAO; but HSP-70 mRNA expression increased at 24-hour time point. Furthermore, resistin (400 ng/mouse) significantly increased TGF-β1 and IL-10 and decreased HSP-70 gene expression at 24 hours after MCAO. CONCLUSIONS Our findings revealed that a molecular mechanism of attenuating ischemic damage by resistin administration probably is increased mRNA expression of anti-inflammatory cytokines. However, applying resistin in the clinical settings for the treatment of stroke deserves further researches in the future.
Collapse
Affiliation(s)
- Sedigheh Behrouzifar
- Research Center and Department of Physiology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Abedin Vakili
- Research Center and Department of Physiology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Mehdi Barati
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
10
|
Ischemic Brain Injury Leads to Brain Edema via Hyperthermia-Induced TRPV4 Activation. J Neurosci 2018; 38:5700-5709. [PMID: 29793978 DOI: 10.1523/jneurosci.2888-17.2018] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 01/27/2023] Open
Abstract
Brain edema is characterized by an increase in net brain water content, which results in an increase in brain volume. Although brain edema is associated with a high fatality rate, the cellular and molecular processes of edema remain largely unclear. Here, we developed an in vitro model of ischemic stroke-induced edema in which male mouse brain slices were treated with oxygen-glucose deprivation (OGD) to mimic ischemia. We continuously measured the cross-sectional area of the brain slice for 150 min under macroscopic microscopy, finding that OGD induces swelling of brain slices. OGD-induced swelling was prevented by pharmacologically blocking or genetically knocking out the transient receptor potential vanilloid 4 (TRPV4), a member of the thermosensitive TRP channel family. Because TRPV4 is activated at around body temperature and its activation is enhanced by heating, we next elevated the temperature of the perfusate in the recording chamber, finding that hyperthermia induces swelling via TRPV4 activation. Furthermore, using the temperature-dependent fluorescence lifetime of a fluorescent-thermosensitive probe, we confirmed that OGD treatment increases the temperature of brain slices through the activation of glutamate receptors. Finally, we found that brain edema following traumatic brain injury was suppressed in TRPV4-deficient male mice in vivo Thus, our study proposes a novel mechanism: hyperthermia activates TRPV4 and induces brain edema after ischemia.SIGNIFICANCE STATEMENT Brain edema is characterized by an increase in net brain water content, which results in an increase in brain volume. Although brain edema is associated with a high fatality rate, the cellular and molecular processes of edema remain unclear. Here, we developed an in vitro model of ischemic stroke-induced edema in which mouse brain slices were treated with oxygen-glucose deprivation. Using this system, we showed that the increase in brain temperature and the following activation of the thermosensitive cation channel TRPV4 (transient receptor potential vanilloid 4) are involved in the pathology of edema. Finally, we confirmed that TRPV4 is involved in brain edema in vivo using TRPV4-deficient mice, concluding that hyperthermia activates TRPV4 and induces brain edema after ischemia.
Collapse
|
11
|
Shang JL, Cheng Q, Duan SJ, Li L, Jia LY. Cognitive improvement following ischemia/reperfusion injury induced by voluntary running‑wheel exercise is associated with LncMALAT1‑mediated apoptosis inhibition. Int J Mol Med 2018; 41:2715-2723. [PMID: 29436629 PMCID: PMC5846661 DOI: 10.3892/ijmm.2018.3484] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 01/19/2018] [Indexed: 01/16/2023] Open
Abstract
Previous human and animal studies demonstrated that voluntary exercise may improve cognitive function and facilitate neuronal plasticity in ischemia/reperfusion (I/R) models. However, the possible underlying mechanisms remain to be elucidated. Metastasis‑associated lung adenocarcinoma transcript 1 (MALAT1), a long noncoding RNA (lncRNA), may be associated with the functions and dysfunctions of endothelial cells. The present study investigated whether spontaneous running‑wheel (RW) exercise‑induced MALAT1 expression changes may be associated with the cognitive improvement of mice following I/R injury. The expression of MALAT1 was evaluated using reverse transcription‑quantitative polymerase chain reaction. Artificial MALAT1 and MALAT1 lentiviral mall interfering (siRNA) were used to alter MALAT1 expression levels in vivo. The Morris Water Maze test was performed to evaluate spatial learning and memory retention in the mice. Changes in the apoptotic rates of hippocampal neurons and levels of apoptosis‑associated proteins were also detected. The data revealed that MALAT1 increased in the hippocampus of mice in the RW‑treated I/R group and that this was associated with neurological, learning and memory improvement, reduced infarction volumes, decreased apoptosis and alterations to expression levels of apoptosis‑associated proteins. Following RW training in I/R‑injured mice, lentiviral MALAT1 siRNA conduction partially attenuated the protections induced by voluntary RW. However, exogenous MALAT1 treatment increased the protection. The current findings suggested that voluntary RW protected hippocampal neurons from I/R injury and promoted cognitive restoration, which was associated with lncRNA MALAT1‑mediated apoptosis inhibition.
Collapse
Affiliation(s)
- Jin-Lin Shang
- Neurology Department, Heji Hospital Affiliated to Changzhi Medical College, Changzhi, Shaanxi 046011, P.R. China
| | - Qing Cheng
- Geriatrics Department, Changzhi People Hospital, Changzhi, Shaanxi 046011, P.R. China
| | - Sheng-Jie Duan
- Neurology Department, Heji Hospital Affiliated to Changzhi Medical College, Changzhi, Shaanxi 046011, P.R. China
| | - Lu Li
- Neurology Department, Heji Hospital Affiliated to Changzhi Medical College, Changzhi, Shaanxi 046011, P.R. China
| | - Li-Ya Jia
- Neurology Department, Heji Hospital Affiliated to Changzhi Medical College, Changzhi, Shaanxi 046011, P.R. China
| |
Collapse
|
12
|
Wang Q, Zhu Z, Liu Y, Tu X, He J. Relationship between serum vitamin D levels and inflammatory markers in acute stroke patients. Brain Behav 2018; 8:e00885. [PMID: 29484258 PMCID: PMC5822590 DOI: 10.1002/brb3.885] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/13/2017] [Accepted: 11/01/2017] [Indexed: 02/06/2023] Open
Abstract
Introduction Low serum vitamin D levels are associated with the development of poststroke depression (PSD). Inflammatory markers play an important role in pathophysiology of PSD. The relationship between vitamin D levels and inflammatory markers has been discussed in nonstroke individuals. The purposes of this study were to explore the relationship between vitamin D levels and inflammatory markers in acute stroke patients and examine the effect of vitamin D and inflammatory markers on PSD. Methods A total of 152 acute stroke patients were recruited. Serum levels of 25-hydroxyvitamin D and inflammatory markers were measured by standardized laboratory methods. Depression symptoms were assessed with the 17-item Hamilton Depression Scale (HAMD-17). Patients with the HAMD-17 scores ≥7 were identified to have depression symptoms. Results Serum vitamin D levels were negatively correlated with serum levels of interleukin-6 and high-sensitivity C-reactive protein (hsCRP) (r = -.244, p = .002; r = -.231, p = .004). Multiple regression analysis showed that interleukin-6 and hsCRP levels were associated with vitamin D levels (B = -0.355, p = .003; B = -2.085, p = .006), whereas age, height, weight, leukocyte count, neutrophil ratio, and lymphocyte rate could be omitted without changing the results. In multivariate analyses, the serum levels of vitamin D and interleukin-6 were associated with the development of PSD after adjusted possible variables (OR = 0.976, 95% CI: 0.958-0.994, p = .009; OR = 1.029, 95% CI: 1.003-1.055, p = .027). Conclusions Serum vitamin D levels are inversely associated with the levels of interleukin-6 and hsCRP, suggesting a potential anti-inflammatory role for vitamin D in stroke individuals.
Collapse
Affiliation(s)
- Qiongzhang Wang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Zhuoying Zhu
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Yuntao Liu
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Xinjie Tu
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| | - Jincai He
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang ProvinceChina
| |
Collapse
|
13
|
Boddaert J, Bielen K, ’s Jongers B, Manocha E, Yperzeele L, Cras P, Pirici D, Kumar-Singh S. CD8 signaling in microglia/macrophage M1 polarization in a rat model of cerebral ischemia. PLoS One 2018; 13:e0186937. [PMID: 29342151 PMCID: PMC5771556 DOI: 10.1371/journal.pone.0186937] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 10/10/2017] [Indexed: 12/16/2022] Open
Abstract
Classical or M1 activity of microglia/macrophages has been described in several neurodegenerative and brain inflammatory conditions and has also been linked to expansion of ischemic injury in post-stroke brain. While different pathways of M1 polarization have been suggested to occur in the post-stroke brain, the precise underlying mechanisms remain undefined. Using a transient middle cerebral artery occlusion (MCAO) rat model, we showed a progressive M2 to M1 polarization in the perilesional brain region with M1 cells becoming one of the dominant subsets by day 4 post-stroke. Comparing key receptors involved in M1 polarization (CD8, IFNγR, Clec4, FcγR, TLR3 and TLR4) and their signal transducers (Syk, Stat1, Irf3, and Traf6) at the day 4 time point, we showed a strong upregulation of CD8 along with SYK transducer in dissected perilesional brain tissue. We further showed that CD8 expression in the post-stroke brain was associated with activated (CD68+) macrophages and that progressive accumulation of CD8+CD68+ cells in the post-stroke brain coincided with increased iNOS (M1 marker) and reduced Arg1 (M2 marker) expression on these cells. In vitro ligand-based stimulation of the CD8 receptor caused increased iNOS expression and an enhanced capacity to phagocytose E. coli particles; and interestingly, CD8 stimulation was also able to repolarize IL4-treated M2 cells to an M1 phenotype. Our data suggest that increased CD8 signaling in the post-stroke brain is primarily associated with microglia/macrophages and can independently drive M1 polarization, and that modulation of CD8 signaling could be a potential target to limit secondary post-stroke brain damage.
Collapse
Affiliation(s)
- Jan Boddaert
- Molecular Pathology Group, Cell Biology and Histology, Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Kenny Bielen
- Molecular Pathology Group, Cell Biology and Histology, Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Bart ’s Jongers
- Molecular Pathology Group, Cell Biology and Histology, Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Ekta Manocha
- Molecular Pathology Group, Cell Biology and Histology, Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Laetitia Yperzeele
- Department of Neurology, Universitair Ziekenhuis Antwerpen, Edegem, Belgium
| | - Patrick Cras
- Department of Neurology, Universitair Ziekenhuis Antwerpen, Edegem, Belgium
- Translational Neuroscience – Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Daniel Pirici
- Department of Research Methodology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Samir Kumar-Singh
- Molecular Pathology Group, Cell Biology and Histology, Faculty of Medicine and Health Sciences, Wilrijk, Belgium
- Translational Neuroscience – Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| |
Collapse
|
14
|
Allen RS, Sayeed I, Oumarbaeva Y, Morrison KC, Choi PH, Pardue MT, Stein DG. Progesterone treatment shows greater protection in brain vs. retina in a rat model of middle cerebral artery occlusion: Progesterone receptor levels may play an important role. Restor Neurol Neurosci 2018; 34:947-963. [PMID: 27802245 DOI: 10.3233/rnn-160672] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND/OBJECTIVE To determine whether inflammation increases in retina as it does in brain following middle cerebral artery occlusion (MCAO), and whether the neurosteroid progesterone, shown to have protective effects in both retina and brain after MCAO, reduces inflammation in retina as well as brain. METHODS MCAO rats treated systemically with progesterone or vehicle were compared with shams. Protein levels of cytosolic NF-κB, nuclear NF-κB, phosphorylated NF-κB, IL-6, TNF-α, CD11b, progesterone receptor A and B, and pregnane X receptor were assessed in retinas and brains at 24 and 48 h using western blots. RESULTS Following MCAO, significant increases were observed in the following inflammatory markers: pNF-κB and CD11b at 24 h in both brain and retina, nuclear NF-κB at 24 h in brain and 48 h in retina, and TNF-α at 24 h in brain.Progesterone treatment in MCAO animals significantly attenuated levels of the following markers in brain: pNF-κB, nuclear NF-κB, IL-6, TNF-α, and CD11b, with significantly increased levels of cytosolic NF-κB. Retinas from progesterone-treated animals showed significantly reduced levels of nuclear NF-κB and IL-6 and increased levels of cytosolic NF-κB, with a trend for reduction in other markers. Post-MCAO, progesterone receptors A and B were upregulated in brain and downregulated in retina. CONCLUSION Inflammatory markers increased in both brain and retina after MCAO, with greater increases observed in brain. Progesterone treatment reduced inflammation, with more dramatic reductions observed in brain than retina. This differential effect may be due to differences in the response of progesterone receptors in brain and retina after injury.
Collapse
Affiliation(s)
- Rachael S Allen
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA.,Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| | - Yuliya Oumarbaeva
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| | | | - Paul H Choi
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| | - Machelle T Pardue
- Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Donald G Stein
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
15
|
Moghaddasi M, Taati M, Asadian P, Khalatbary AR, Asaei R, Pajouhi N. The effects of two-stage carotid occlusion on spatial memory and pro-inflammatory markers in the hippocampus of rats. J Physiol Sci 2017; 67:415-423. [PMID: 27470129 PMCID: PMC10717598 DOI: 10.1007/s12576-016-0474-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 07/13/2016] [Indexed: 02/07/2023]
Abstract
The purpose of this study was to evaluate the effects of cerebral hypoperfusion on cognitive ability, TNFα, IL1β and PGE2 levels in both hippocampi in a modified two-vessel occlusion model. Both common carotid arteries of adult male Wistar rats were permanently occluded with an interval of 1 week between occlusions. Learning and memory were significantly decreased after 1 month. This reduction was not significant after 2 months, which may be attributed to blood flow compensation. The TNFα level was significantly increased after 3 h and 1 day. IL1β was significantly increased after 1 day. After a week there was no significant difference in pro-inflammatory levels. Furthermore, there was no difference between right and left hippocampi. It is possible that TNFα and IL1β elevation initiates pathologic processes that contribute to memory impairment.
Collapse
Affiliation(s)
- Mehrnoush Moghaddasi
- Department of Physiology, Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Majid Taati
- Department of Pathobiology, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Payman Asadian
- Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - Ali Reza Khalatbary
- Department of Anatomy, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Raheleh Asaei
- Department of Physiology, Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Naser Pajouhi
- Department of Physiology, Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
16
|
Hsuan YCY, Lin CH, Chang CP, Lin MT. Mesenchymal stem cell-based treatments for stroke, neural trauma, and heat stroke. Brain Behav 2016; 6:e00526. [PMID: 27781140 PMCID: PMC5064338 DOI: 10.1002/brb3.526] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/05/2016] [Accepted: 06/08/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) transplantation has been reported to improve neurological function following neural injury. Many physiological and molecular mechanisms involving MSC therapy-related neuroprotection have been identified. METHODS A review is presented of articles that pertain to MSC therapy and diverse brain injuries including stroke, neural trauma, and heat stroke, which were identified using an electronic search (e.g., PubMed), emphasize mechanisms of MSC therapy-related neuroprotection. We aim to discuss neuroprotective mechanisms that underlie the beneficial effects of MSCs in treating stroke, neural trauma, and heatstroke. RESULTS MSC therapy is promising as a means of augmenting brain repair. Cell incorporation into the injured tissue is not a prerequisite for the beneficial effects exerted by MSCs. Paracrine signaling is believed to be the most important mediator of MSC therapy in brain injury. The multiple mechanisms of action of MSCs include enhanced angiogenesis and neurogenesis, immunomodulation, and anti-inflammatory effects. Microglia are the first source of the inflammatory cascade during brain injury. Cytokines, including tumor necrosis factor-α, interleukin-1β, and interleukin-6, are significantly produced by microglia in the brain after experimental brain injury. The proinflammatory M1 phenotype of microglia is associated with tissue destruction, whereas the anti-inflammatory M2 phenotype of microglia facilitates repair and regeneration. MSC therapy may improve outcomes of ischemic stroke, neural trauma, and heatstroke by inhibiting the activity of M1 phenotype of microglia but augmenting the activity of M2 phenotype of microglia. CONCLUSION This review offers a testable platform for targeting microglial-mediated cytokines in clinical trials based upon the rational design of MSC therapy in the future. MSCs that are derived from the placenta provide a great choice for stem cell therapy. Although targeting the microglial activation is an important approach to reduce the burden of the injury, it is not the only one. This review focuses on this specific aspect.
Collapse
Affiliation(s)
| | | | - Ching-Ping Chang
- Department of Medical Research Chi Mei Medical Center Tainan Taiwan
| | - Mao-Tsun Lin
- Department of Medical Research Chi Mei Medical Center Tainan Taiwan
| |
Collapse
|
17
|
Xie W, Fang L, Gan S, Xuan H. Interleukin-19 alleviates brain injury by anti-inflammatory effects in a mice model of focal cerebral ischemia. Brain Res 2016; 1650:172-177. [PMID: 27608956 DOI: 10.1016/j.brainres.2016.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 09/01/2016] [Accepted: 09/04/2016] [Indexed: 11/17/2022]
Abstract
Stroke causes brain injury with neuroinflammation which exacerbates the neuronal damage. Recent studies show that anti-inflammatory cytokine interleukin-19 (IL-19) plays a critical part in the inflammatory and ischemic vascular diseases, yet its potential role in ischemic stroke is unknown. Here, we tested the hypothesis that IL-19 exerts protective effects against brain ischemia by modulating inflammation after stroke. Mice were injected intraperitoneally with 10ng/g per day recombinant mouse IL-19 starting pre-stroke, and were subjected to transient middle cerebral artery occlusion. Infarct volume was assessed by triphenyltetrazolium chloride and neurobehavioral outcome by neurological scores. Inflammation was measured using real-time quantitative PCR, immunochemistry, and fluorescence-activated cell sorting. Infarct volume at 72h after stroke was significantly smaller in IL-19 treated group and focal neurological score was significantly better. IL-19 treatment markedly attenuated elevation of the expression of TNF-α and IL-6 mRNA, suppressed increases in the number of microglia, macrophages, CD4+ T cells, CD8+ T cells as well as B cells, and blocked activation of macrophages and neutrophils in the ischemic brain. In peripheral blood, IL-19 injection helped to robustly preserve the reduced immune cells, including macrophages, CD4+ T cells, CD8+ T cells and B cells, compared to control group. IL-19 reduced brain infarction and attenuated neurological deficits following stroke in mice, probably by inhibiting infiltration and activation of immune cells, and by suppressing increases in gene expression of proinflammatory cytokines. This may identify IL-19 as a new therapeutic to limit neuroinflammation after stroke.
Collapse
Affiliation(s)
- Weiying Xie
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Fang
- Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shuyuan Gan
- Department of Anesthesiology, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Haojun Xuan
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, China.
| |
Collapse
|
18
|
Madsen PM, Clausen BH, Degn M, Thyssen S, Kristensen LK, Svensson M, Ditzel N, Finsen B, Deierborg T, Brambilla R, Lambertsen KL. Genetic ablation of soluble tumor necrosis factor with preservation of membrane tumor necrosis factor is associated with neuroprotection after focal cerebral ischemia. J Cereb Blood Flow Metab 2016; 36:1553-69. [PMID: 26661199 PMCID: PMC5012516 DOI: 10.1177/0271678x15610339] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/07/2015] [Indexed: 11/16/2022]
Abstract
Microglia respond to focal cerebral ischemia by increasing their production of the neuromodulatory cytokine tumor necrosis factor, which exists both as membrane-anchored tumor necrosis factor and as cleaved soluble tumor necrosis factor forms. We previously demonstrated that tumor necrosis factor knockout mice display increased lesion volume after focal cerebral ischemia, suggesting that tumor necrosis factor is neuroprotective in experimental stroke. Here, we extend our studies to show that mice with intact membrane-anchored tumor necrosis factor, but no soluble tumor necrosis factor, display reduced infarct volumes at one and five days after stroke. This was associated with improved functional outcome after experimental stroke. No changes were found in the mRNA levels of tumor necrosis factor and tumor necrosis factor-related genes (TNFR1, TNFR2, TACE), pro-inflammatory cytokines (IL-1β, IL-6) or chemokines (CXCL1, CXCL10, CCL2); however, protein expression of TNF, IL-1β, IL-6 and CXCL1 was reduced in membrane-anchored tumor necrosis factor(Δ/Δ) compared to membrane-anchored tumor necrosis factor(wt/wt) mice one day after experimental stroke. This was paralleled by reduced MHCII expression and a reduction in macrophage infiltration in the ipsilateral cortex of membrane-anchored tumor necrosis factor(Δ/Δ) mice. Collectively, these findings indicate that membrane-anchored tumor necrosis factor mediates the protective effects of tumor necrosis factor signaling in experimental stroke, and therapeutic strategies specifically targeting soluble tumor necrosis factor could be beneficial in clinical stroke therapy.
Collapse
Affiliation(s)
- Pernille M Madsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Bettina H Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Matilda Degn
- Molecular Sleep Lab, Department of Diagnostics, Glostrup Hospital, Glostrup, Denmark
| | - Stine Thyssen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lotte K Kristensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Martina Svensson
- Department of Experimental Medical Sciences, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Nicholas Ditzel
- KMEB, Molecular Endocrinology, Odense University Hospital, Odense, Denmark
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Tomas Deierborg
- Department of Experimental Medical Sciences, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Kate L Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
19
|
Miao W, Bao TH, Han JH, Yin M, Zhang J, Yan Y, Zhu YH. Neuroprotection induced by post-conditioning following ischemia/reperfusion in mice is associated with altered microRNA expression. Mol Med Rep 2016; 14:2582-8. [PMID: 27485299 PMCID: PMC4991681 DOI: 10.3892/mmr.2016.5576] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 06/24/2016] [Indexed: 12/05/2022] Open
Abstract
Ischemic preconditioning and ischemic postconditioning (IPostC) represent promising strategies to reduce ischemia-reperfusion (I/R) injury and attenuate the lethal ischemic damage following stroke. However, the mechanism underlying this attenuation remains to be elucidated. It was hypothesized that alterations in microRNA (miRNA) expression in the cerebral cortex and hippocampus of mice following I/R is associated with the functional improvement induced by IPostC. Behavioral changes were assessed in a mouse model of I/R in the absence or presence of IPostC, followed by microarray analyses to investigate the expressional alterations of miRNAs in the cerebral cortex and hippocampus of mice. The results of the present study revealed that IPostC abrogated the neurological impairment and hippocampus-associated cognitive deficits induced by I/R, and upregulated or downregulated the expression levels of numerous miRNAs. Furthermore, the upregulation of miR-19a, and the downregulation of miR-1, let-7f and miR-124 expression levels following IPostC was confirmed utilizing reverse transcription-quantitative polymerase chain reaction. The results of the present study demonstrated that alterations in miRNA expression in the cerebral cortex and hippocampus of mice following I/R was associated with the neuroprotection induced by IPostC.
Collapse
Affiliation(s)
- Wei Miao
- Department of Neurology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Tian-Hao Bao
- Department of Geratology, Psychiatric Hospital of Yunnan Kunming, Yunnan 650224, P.R. China
| | - Jian-Hong Han
- Department of Neurology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Mei Yin
- Department of Neurology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Jie Zhang
- Department of Neurology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Yong Yan
- Department of Neurology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Yu-Hong Zhu
- Department of Neurology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| |
Collapse
|
20
|
Yu JH, Seo JH, Lee JY, Lee MY, Cho SR. Induction of Neurorestoration From Endogenous Stem Cells. Cell Transplant 2016; 25:863-82. [PMID: 26787093 DOI: 10.3727/096368916x690511] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Neural stem cells (NSCs) persist in the subventricular zone lining the ventricles of the adult brain. The resident stem/progenitor cells can be stimulated in vivo by neurotrophic factors, hematopoietic growth factors, magnetic stimulation, and/or physical exercise. In both animals and humans, the differentiation and survival of neurons arising from the subventricular zone may also be regulated by the trophic factors. Since stem/progenitor cells present in the adult brain and the production of new neurons occurs at specific sites, there is a possibility for the treatment of incurable neurological diseases. It might be feasible to induce neurogenesis, which would be particularly efficacious in the treatment of striatal neurodegenerative conditions such as Huntington's disease, as well as cerebrovascular diseases such as ischemic stroke and cerebral palsy, conditions that are widely seen in the clinics. Understanding of the molecular control of endogenous NSC activation and progenitor cell mobilization will likely provide many new opportunities as therapeutic strategies. In this review, we focus on endogenous stem/progenitor cell activation that occurs in response to exogenous factors including neurotrophic factors, hematopoietic growth factors, magnetic stimulation, and an enriched environment. Taken together, these findings suggest the possibility that functional brain repair through induced neurorestoration from endogenous stem cells may soon be a clinical reality.
Collapse
Affiliation(s)
- Ji Hea Yu
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
21
|
Korhonen P, Kanninen KM, Lehtonen Š, Lemarchant S, Puttonen KA, Oksanen M, Dhungana H, Loppi S, Pollari E, Wojciechowski S, Kidin I, García-Berrocoso T, Giralt D, Montaner J, Koistinaho J, Malm T. Immunomodulation by interleukin-33 is protective in stroke through modulation of inflammation. Brain Behav Immun 2015; 49:322-36. [PMID: 26111431 DOI: 10.1016/j.bbi.2015.06.013] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/05/2015] [Accepted: 06/18/2015] [Indexed: 01/19/2023] Open
Abstract
Cerebral stroke induces massive Th1-shifted inflammation both in the brain and the periphery, contributing to the outcome of stroke. A Th1-type response is neurotoxic whereas a Th2-type response is accompanied by secretion of anti-inflammatory cytokines, such as interleukin-4 (IL-4). Interleukin-33 (IL-33) is a cytokine known to induce a shift towards the Th2-type immune response, polarize macrophages/microglia towards the M2-type, and induce production of anti-inflammatory cytokines. We found that the plasma levels of the inhibitory IL-33 receptor, sST2, are increased in human stroke and correlate with a worsened stroke outcome, suggesting an insufficient IL-33-driven Th2-type response. In mouse, peripheral administration of IL-33 reduced stroke-induced cell death and improved the sensitivity of the contralateral front paw at 5days post injury. The IL-33-treated mice had increased levels of IL-4 in the spleen and in the peri-ischemic area of the cortex. Neutralization of IL-4 by administration of an IL-4 antibody partially prevented the IL-33-mediated protection. IL-33 treatment also reduced astrocytic activation in the peri-ischemic area and increased the number of Arginase-1 immunopositive microglia/macrophages at the lesion site. In human T-cells, IL-33 treatment induced IL-4 secretion, and the conditioned media from IL-33-exposed T-cells reduced astrocytic activation. This study demonstrates that IL-33 is protective against ischemic insult by induction of IL-4 secretion and may represent a novel therapeutic approach for the treatment of stroke.
Collapse
Affiliation(s)
- Paula Korhonen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Katja M Kanninen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Šárka Lehtonen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sighild Lemarchant
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Katja A Puttonen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Minna Oksanen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Hiramani Dhungana
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sanna Loppi
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Eveliina Pollari
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sara Wojciechowski
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Iurii Kidin
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Teresa García-Berrocoso
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Dolors Giralt
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Joan Montaner
- Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain
| | - Jari Koistinaho
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland.
| | - Tarja Malm
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| |
Collapse
|
22
|
|
23
|
Allen RS, Olsen TW, Sayeed I, Cale HA, Morrison KC, Oumarbaeva Y, Lucaciu I, Boatright JH, Pardue MT, Stein DG. Progesterone treatment in two rat models of ocular ischemia. Invest Ophthalmol Vis Sci 2015; 56:2880-91. [PMID: 26024074 DOI: 10.1167/iovs.14-16070] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE To determine whether the neurosteroid progesterone, shown to have protective effects in animal models of traumatic brain injury, stroke, and spinal cord injury, is also protective in ocular ischemia animal models. METHODS Progesterone treatment was tested in two ocular ischemia models in rats: a rodent anterior ischemic optic neuropathy (rAION) model, which induces permanent monocular optic nerve stroke, and the middle cerebral artery occlusion (MCAO) model, which causes transient ischemia in both the retina and brain due to an intraluminal filament that blocks the ophthalmic and middle cerebral arteries. Visual function and retinal histology were assessed to determine whether progesterone attenuated retinal injury in these models. Additionally, behavioral testing and 2% 2,3,5-triphenyltetrazolium chloride (TTC) staining in brains were used to compare progesterone's neuroprotective effects in both retina and brain using the MCAO model. RESULTS Progesterone treatment showed no effect on visual evoked potential (VEP) reduction and retinal ganglion cell loss in the permanent rAION model. In the transient MCAO model, progesterone treatment reduced (1) electroretinogram (ERG) deficits, (2) MCAO-induced upregulation of glutamine synthetase (GS) and glial fibrillary acidic protein (GFAP), and (3) retinal ganglion cell loss. As expected, progesterone treatment also had significant protective effects in behavioral tests and a reduction in infarct size in the brain. CONCLUSIONS Progesterone treatment showed protective effects in the retina following MCAO but not rAION injury, which may result from mechanistic differences with injury type and the therapeutic action of progesterone.
Collapse
Affiliation(s)
- Rachael S Allen
- Emergency Medicine Emory University, Atlanta, Georgia, United States 2Department of Ophthalmology, Emory University, Atlanta, Georgia, United States 3Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - Timothy W Olsen
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Iqbal Sayeed
- Emergency Medicine Emory University, Atlanta, Georgia, United States
| | - Heather A Cale
- Emergency Medicine Emory University, Atlanta, Georgia, United States
| | | | - Yuliya Oumarbaeva
- Emergency Medicine Emory University, Atlanta, Georgia, United States
| | - Irina Lucaciu
- Emergency Medicine Emory University, Atlanta, Georgia, United States
| | - Jeffrey H Boatright
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States 3Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - Machelle T Pardue
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States 3Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - Donald G Stein
- Emergency Medicine Emory University, Atlanta, Georgia, United States
| |
Collapse
|
24
|
Li P, Su L, Li X, Di W, Zhang X, Zhang C, He T, Zhu X, Zhang Y, Li Y. Remote limb ischemic postconditioning protects mouse brain against cerebral ischemia/reperfusion injury via upregulating expression of Nrf2, HO-1 and NQO-1 in mice. Int J Neurosci 2015; 126:552-559. [DOI: 10.3109/00207454.2015.1042973] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
25
|
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: 67] [Impact Index Per Article: 6.7] [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
|
26
|
Jeon JH, Jung HW, Jang HM, Moon JH, Park KT, Lee HC, Lim HY, Sur JH, Kang BT, Ha J, Jung DI. Canine model of ischemic stroke with permanent middle cerebral artery occlusion: clinical features, magnetic resonance imaging, histopathology, and immunohistochemistry. J Vet Sci 2014; 16:75-85. [PMID: 25269716 PMCID: PMC4367152 DOI: 10.4142/jvs.2015.16.1.75] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 09/26/2014] [Indexed: 01/17/2023] Open
Abstract
The purpose of this study was to identify time-related changes in clinical, MRI, histopathologic, and immunohistochemical findings associated with ischemic stroke in dogs. Additionally, the association of cerebrospinal fluid (CSF) and tissue levels of interleukin (IL)-6 with clinical prognosis was assessed. Ischemic stroke was induced by permanent middle cerebral artery occlusion (MCAO) in nine healthy experimental dogs. The dogs were divided into three groups according to survival time and duration of the experimental period: group A (survived only 1 day), group B (1-week experimental period), and group C (2-week experimental period). Neurologic status was evaluated daily. Magnetic resonance imaging (MRI) was performed according to a predetermined schedule. Concentration of IL-6 in CSF was measured serially after ischemic stroke. Postmortem examination was performed for all experimental dogs. During histopathological examination, variable degrees of cavitation and necrosis due to neuronal cytopathic effects, such as pyknotic nuclei and cytoplasmic shrinkage, were observed on the affected side of the cerebral cortex in all dogs. Immunohistochemistry specific for IL-6 showed increased expression in the ischemic lesions. CSF IL-6 concentrations and ischemic lesion volumes 1 day after ischemic stroke were significantly higher in group A compared to groups B and C.
Collapse
Affiliation(s)
- Joon-Hyeok Jeon
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 660-701, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Reduced inflammatory phenotype in microglia derived from neonatal rat spinal cord versus brain. PLoS One 2014; 9:e99443. [PMID: 24914808 PMCID: PMC4051776 DOI: 10.1371/journal.pone.0099443] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 05/15/2014] [Indexed: 12/22/2022] Open
Abstract
Microglia are the primary immune cells of the central nervous system (CNS). Membrane bound sensors on their processes monitor the extracellular environment and respond to perturbations of the CNS such as injury or infection. Once activated, microglia play a crucial role in determining neuronal survival. Recent studies suggest that microglial functional response properties vary across different regions of the CNS. However, the activation profiles of microglia derived from the spinal cord have not been evaluated against brain microglia in vitro. Here, we studied the morphological properties and secretion of inflammatory and trophic effectors by microglia derived from the brain or spinal cord of neonatal rats under basal culture conditions and after activation with lipopolysaccharide (LPS). Our results demonstrate that spinal microglia assume a less inflammatory phenotype after LPS activation, with reduced release of the inflammatory effectors tumor necrosis factor alpha, interleukin-1 beta, and nitric oxide, a less amoeboid morphology, and reduced phagocytosis relative to brain-derived microglia. Phenotypic differences between brain and spinal microglia are an important consideration when evaluating anti-inflammatory or immunomodulatory therapies for brain versus spinal injury.
Collapse
|
28
|
Sist B, Fouad K, Winship IR. Plasticity beyond peri-infarct cortex: spinal up regulation of structural plasticity, neurotrophins, and inflammatory cytokines during recovery from cortical stroke. Exp Neurol 2013; 252:47-56. [PMID: 24291254 DOI: 10.1016/j.expneurol.2013.11.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 10/24/2013] [Accepted: 11/20/2013] [Indexed: 12/12/2022]
Abstract
Stroke induces pathophysiological and adaptive processes in regions proximal and distal to the infarct. Recent studies suggest that plasticity at the level of the spinal cord may contribute to sensorimotor recovery after cortical stroke. Here, we compare the time course of heightened structural plasticity in the spinal cord against the temporal profile of cortical plasticity and spontaneous behavioral recovery. To examine the relation between trophic and inflammatory effectors and spinal structural plasticity, spinal expression of brain derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) was measured. Growth-associated protein 43 (GAP-43), measured at 3, 7, 14, or 28 days after photothrombotic stroke of the forelimb sensorimotor cortex (FL-SMC) to provide an index of periods of heightened structural plasticity, varied as a function of lesion size and time after stroke in the cortical hemispheres and the spinal cord. Notably, GAP-43 levels in the cervical spinal cord were significantly increased after FL-SMC lesion, but the temporal window of elevated structural plasticity was more finite in spinal cord relative to ipsilesional cortical expression (returning to baseline levels by 28 post-stroke). Peak GAP-43 expression in spinal cord occurred during periods of accelerated spontaneous recovery, as measured on the Montoya Staircase reaching task, and returned to baseline as recovery plateaued. Interestingly, spinal GAP-43 levels were significantly correlated with spinal levels of the inflammatory cytokines TNF-α and IL-6 as well as the neurotrophin NT-3, while a transient increase in BDNF levels preceded elevated GAP-43 expression. These data identify a significant but time-limited window of heightened structural plasticity in the spinal cord following stroke that correlates with spontaneous recovery and the spinal expression of inflammatory cytokines and neurotrophic factors.
Collapse
Affiliation(s)
- Bernice Sist
- Centre for Neuroscience, University of Alberta, Edmonton, Alberta T6G 2R3, Canada; Neurochemical Research Unit, University of Alberta, Edmonton, Alberta T6G 2R3, Canada.
| | - Karim Fouad
- Centre for Neuroscience, University of Alberta, Edmonton, Alberta T6G 2R3, Canada; Faculty of Rehabilitative Medicine, University of Alberta, Edmonton, Alberta T6G 2R3, Canada.
| | - Ian R Winship
- Centre for Neuroscience, University of Alberta, Edmonton, Alberta T6G 2R3, Canada; Department of Psychiatry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada; Neurochemical Research Unit, University of Alberta, Edmonton, Alberta T6G 2R3, Canada.
| |
Collapse
|
29
|
Tuna M, Yılmaz DM, Erman T, Arslan A, Özgür H, Polat S. Effect of neutralization of rat interleukin 6 bioactivity on inducible nitric oxide synthase up-regulation and cerebral ischemic damage. Neurol Res 2013; 31:714-20. [DOI: 10.1179/174313209x380892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
30
|
|
31
|
Affiliation(s)
- Yi-Bing Ouyang
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA 94305-5117, USA.
| |
Collapse
|
32
|
Winters L, Winters T, Gorup D, Mitrečić D, Ćurlin M, Križ J, Gajović S. Expression analysis of genes involved in TLR2-related signaling pathway: Inflammation and apoptosis after ischemic brain injury. Neuroscience 2013; 238:87-96. [DOI: 10.1016/j.neuroscience.2013.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Revised: 01/16/2013] [Accepted: 02/02/2013] [Indexed: 11/29/2022]
|
33
|
Ischemic postconditioning protects against focal cerebral ischemia by inhibiting brain inflammation while attenuating peripheral lymphopenia in mice. Neuroscience 2013; 243:149-57. [PMID: 23590905 DOI: 10.1016/j.neuroscience.2013.03.062] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 03/26/2013] [Accepted: 03/28/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND Ischemic postconditioning (IPostC) has been shown to attenuate brain injury in rat stroke models, but a mouse model has not been reported. This study establishes an IPostC model in mice and investigates how IPostC affects infiltration of leukocytes in the ischemic brain and lymphopenia associated with stroke-induced immunodepression. MATERIAL AND METHODS A total of 125 mice were used. IPostC was performed by a repeated series of brief occlusions of the middle cerebral artery (MCA) after reperfusion, in a focal ischemia model in mice. Infarct sizes, neurological scores, inflammatory brain cells and immune cell populations in lymph nodes, spleen and bone marrow were analyzed with fluorescence-activated cell sorting (FACS). RESULTS IPostC performed immediately, 2 min and 3 h after reperfusion significantly reduced infarct sizes and attenuated neurological scores as measured up to 3 days post-stroke. In the group with strongest protection, infarct sizes were reduced from 49.6±2.8% (n=16) to 27.9±2.9% (n=10, P<.001). The spared infarct areas were seen in the ischemic penumbra or ischemic margins, i.e., the border zones between the cortical territories of the anterior cerebral artery and those of the MCA, as well as in the ventromedial and dorsolateral striata. FACS analyses showed that IPostC significantly blocked increases in the numbers of microglia (CD45intCD11b+), macrophages (CD45hiCD68+), CD4 T cells (CD45+CD4+) and CD8 T cells (CD45+CD8+) as well as B lymphocytes (CD45+CD19+) in the ischemic brain (n=5/group). Reduced-immune cell numbers in the peripheral blood and spleen were increased by IPostC while immune cell populations in the bone marrow were not altered by IPostC. CONCLUSIONS IPostC reduced brain infarction and mitigated neurological deficits in mice, likely by blocking infiltration of both innate and adaptive immune cells in the ischemic brain. In addition, IPostC robustly attenuated peripheral lymphopenia and thus improved systemic immunodepression.
Collapse
|
34
|
Sohrabji F, Selvamani A, Balden R. Revisiting the timing hypothesis: biomarkers that define the therapeutic window of estrogen for stroke. Horm Behav 2013; 63:222-30. [PMID: 22728278 PMCID: PMC3483414 DOI: 10.1016/j.yhbeh.2012.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 02/06/2023]
Abstract
Significantly extended life expectancy coupled with contemporary sedentary lifestyles and poor nutrition has created a global epidemic of cardiovascular disease and stroke. For women, this issue is complicated by the discrepant outcomes of hormone therapy (HT) for stroke incidence and severity as well as the therapeutic complications for stroke associated with advancing age. Here we propose that the impact of estrogen therapy cannot be considered in isolation, but should include age-related changes in endocrine, immune, and nucleic acid mediators that collaborate with estrogen to produce neuroprotective effects commonly seen in younger, healthier demographics. Due to their role as modulators of ischemic cell death, the post-stroke inflammatory response, and neuronal survival and regeneration, this review proposes that Insulin-like Growth Factor (IGF)-1, Vitamin D, and discrete members of the family of non-coding RNA peptides called microRNAs (miRNAs) may be crucial biochemical markers that help determine the neuroprotective "window" of HT. Specifically, IGF-1 confers neuroprotection in concert with, and independently of, estrogen and failure of the insulin/IGF-1 axis is associated with metabolic disturbances that increase the risk for stroke. Vitamin D and miRNAs regulate and complement IGF-1 mediated function and neuroprotective efficacy via modulation of IGF-1 availability and neural stem cell and immune cell proliferation, differentiation and secretions. Together, age-related decline of these factors differentially affects stroke risk, severity, and outcome, and may provide a novel therapeutic adjunct to traditional HT practices.
Collapse
Affiliation(s)
- Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Women's Health in Neuroscience Program, Texas A&M Health Science Center, College Station, TX 77843-1114, USA.
| | | | | |
Collapse
|
35
|
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
|
36
|
Toll-like receptor 2 deficiency leads to delayed exacerbation of ischemic injury. J Neuroinflammation 2012; 9:191. [PMID: 22873409 PMCID: PMC3458899 DOI: 10.1186/1742-2094-9-191] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 07/13/2012] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Using a live imaging approach, we have previously shown that microglia activation after stroke is characterized by marked and long-term induction of the Toll-like receptor (TLR) 2 biophotonic signals. However, the role of TLR2 (and potentially other TLRs) beyond the acute innate immune response and as early neuroprotection against ischemic injury is not well understood. METHODS TLR2-/- mice were subjected to transient middle cerebral artery occlusion followed by different reperfusion times. Analyses assessing microglial activation profile/innate immune response were performed using in situ hybridization, immunohistochemistry analysis, flow cytometry and inflammatory cytokine array. The effects of the TLR2 deficiency on the evolution of ischemic brain injury were analyzed using a cresyl violet staining of brain sections with appropriate lesion size estimation. RESULTS Here we report that TLR2 deficiency markedly affects post-stroke immune response resulting in delayed exacerbation of the ischemic injury. The temporal analysis of the microglia/macrophage activation profiles in TLR2-/- mice and age-matched controls revealed reduced microglia/macrophage activation after stroke, reduced capacity of resident microglia to proliferate as well as decreased levels of monocyte chemotactic protein-1 (MCP-1) and consequently lower levels of CD45(high)/CD11b(+) expressing cells as shown by flow cytometry analysis. Importantly, although acute ischemic lesions (24 to 72 h) were smaller in TLR2-/- mice, the observed alterations in innate immune response were more pronounced at later time points (at day 7) after initial stroke, which finally resulted in delayed exacerbation of ischemic lesion leading to larger chronic infarctions as compared with wild-type mice. Moreover, our results revealed that TLR2 deficiency is associated with significant decrease in the levels of neurotrophic/anti-apoptotic factor Insulin-like growth factor-1 (IGF-1), expressed by microglia in the areas both in and around ischemic lesion. CONCLUSION Our results clearly suggest that optimal and timely microglial activation/innate immune response is needed to limit neuronal damage after stroke.
Collapse
|
37
|
Balden R, Selvamani A, Sohrabji F. Vitamin D deficiency exacerbates experimental stroke injury and dysregulates ischemia-induced inflammation in adult rats. Endocrinology 2012; 153:2420-35. [PMID: 22408173 PMCID: PMC3339639 DOI: 10.1210/en.2011-1783] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vitamin D deficiency (VDD) is widespread and considered a risk factor for cardiovascular disease and stroke. Low vitamin D levels are predictive for stroke and more fatal strokes in humans, whereas vitamin D supplements are associated with decreased risk of all-cause mortality. Because VDD occurs with other comorbid conditions that are also independent risk factors for stroke, this study examined the specific effect of VDD on stroke severity in rats. Adult female rats were fed control or VDD diet for 8 wk and were subject to middle cerebral artery occlusion thereafter. The VDD diet reduced circulating vitamin D levels to one fifth (22%) of that observed in rats fed control chow. Cortical and striatal infarct volumes in animals fed VDD diet were significantly larger, and sensorimotor behavioral testing indicated that VDD animals had more severe poststroke behavioral impairment than controls. VDD animals were also found to have significantly lower levels of the neuroprotective hormone IGF-I in plasma and the ischemic hemisphere. Cytokine analysis indicated that VDD significantly reduced IL-1α, IL-1β, IL-2, IL-4, IFN-γ, and IL-10 expression in ischemic brain tissue. However, ischemia-induced IL-6 up-regulation was significantly higher in VDD animals. In a separate experiment, the therapeutic potential of acute vitamin D treatments was evaluated, where animals received vitamin D injections 4 h after stroke and every 24 h thereafter. Acute vitamin D treatment did not improve infarct volume or behavioral performance. Our data indicate that VDD exacerbates stroke severity, involving both a dysregulation of the inflammatory response as well as suppression of known neuroprotectants such as IGF-I.
Collapse
Affiliation(s)
- Robyn Balden
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 228 Reynolds Medical Building, College Station, Texas 77843-1114, USA
| | | | | |
Collapse
|
38
|
Saini MG, Bix GJ. Oxygen-glucose deprivation (OGD) and interleukin-1 (IL-1) differentially modulate cathepsin B/L mediated generation of neuroprotective perlecan LG3 by neurons. Brain Res 2011; 1438:65-74. [PMID: 22244880 DOI: 10.1016/j.brainres.2011.12.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 11/24/2011] [Accepted: 12/13/2011] [Indexed: 11/15/2022]
Abstract
Brain extracellular matrix (ECM) is highly degraded after cerebral ischemia. The perlecan c-terminal fragment LG3 is generated at increased levels by proteolytic processing as long as 3 days after ischemia. It has previously been shown that oxygen-glucose deprivation (OGD), reperfusion and interleukin-1 α (IL-1α) stimulate brain cells to yield increased levels of LG3. This LG3, in turn, is neuroprotective against OGD, and may therefore represent one of the brain's defenses against ischemic injury. Here, we investigate whether, in neurons, this increased LG3 is the result of increased perlecan generation and cellular release, increased protease release (to generate LG3 from previous extracellularly deposited perlecan) or both. We found that pre-synthesized perlecan may be exocytosed by neurons during OGD and de novo synthesis of perlecan is increased during reperfusion, even 24 h after OGD. Furthermore, while cathepsin L activity was seen to be marginally important to generate LG3 during normoxic conditions, cathepsin B activity was found to be important to generate increased levels of LG3 following OGD and reperfusion. On the other hand, IL-1α treatment raised levels of cathepsin L in neuronal media, and both cathepsin L and cathepsin B were demonstrated to be important for increasing LG3 levels after IL-1α treatment.
Collapse
Affiliation(s)
- Maxim G Saini
- Department of Molecular and Cellular Medicine, Texas A&M College of Medicine, College Station, TX, USA
| | | |
Collapse
|
39
|
Saini MG, Pinteaux E, Lee B, Bix GJ. Oxygen-glucose deprivation and interleukin-1α trigger the release of perlecan LG3 by cells of neurovascular unit. J Neurochem 2011; 119:760-71. [PMID: 21919908 DOI: 10.1111/j.1471-4159.2011.07484.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Two of the main stresses faced by cells at the neurovascular unit (NVU) as an immediate result of cerebral ischemia are oxygen-glucose deprivation (OGD)/reperfusion and inflammatory stress caused by up regulation of IL-1. As a result of these stresses, perlecan, an important component of the NVU extracellular matrix, is highly proteolyzed. In this study, we describe that focal cerebral ischemia in rats results in increased generation of laminin globular domain 3 (LG3), the c-terminal bioactive fragment of perlecan. Further, in vitro study of the cells of the NVU was performed to locate the source of this increased perlecan-LG3. Neurons, astrocytes, brain endothelial cells and pericytes were exposed to OGD/reperfusion and IL-1α/β. It was observed that neurons and pericytes showed increased levels of LG3 during OGD in their culture media. During in vitro reperfusion, neurons, astrocytes and pericytes showed elevated levels of LG3, but only after exposure to brief durations of OGD. IL-1α and IL-1β treatment tended to have opposite effects on NVU cells. While IL-1α increased or had minimal to no effect on LG3 generation, high concentrations of IL-1β decreased it in most cells studied. Finally, LG3 was determined to be neuroprotective and anti-proliferative in brain endothelial cells, suggesting a possible role for the generation of LG3 in the ischemic brain.
Collapse
Affiliation(s)
- Maxim G Saini
- Department of Molecular and Cellular Medicine, Texas A&M College of Medicine, College Station, Texas 77843, USA
| | | | | | | |
Collapse
|
40
|
Cybulska-Klosowicz A, Liguz-Lecznar M, Nowicka D, Ziemka-Nalecz M, Kossut M, Skangiel-Kramska J. Matrix metalloproteinase inhibition counteracts impairment of cortical experience-dependent plasticity after photothrombotic stroke. Eur J Neurosci 2011; 33:2238-46. [PMID: 21615560 DOI: 10.1111/j.1460-9568.2011.07713.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Matrix metalloproteinases (MMPs) are fine modulators of brain plasticity and pathophysiology. The inhibition of MMPs shortly after ischaemic stroke reduces the infarct size and has beneficial effects on post-stroke behavioural recovery. Our previous studies have shown that photothrombotic cortical stroke disrupts use-dependent plasticity in the neighbouring cortex. The aim of the present study was to check whether the inhibition of MMPs after photothrombosis rescued the plastic capacity of the barrel cortex. To induce plasticity in adult mice, a unilateral deprivation of all vibrissae except row C was applied. The deprivation started immediately after stroke and lasted 7 days. This procedure, in control (non-stroke) animals, results in an enlargement of functional representation of the spared row, as shown with [(14)C]2-deoxyglucose uptake mapping. In mice with stroke induced by photothrombosis in the vicinity of the barrel cortex, vibrissae deprivation did not result in an enlargement of the cortical representation of the spared row C of vibrissae, which confirmed our previous results. However, when mice were injected with the broad-spectrum inhibitor of MMPs FN-439 (10 mg/kg, i.v.) immediately before a stroke, an enlargement of the representation of the spared row similar to the enlargement found in sham mice was observed. These results indicate the involvement of MMPs in the impairment of use-dependent plasticity in the vicinity of an ischaemic lesion.
Collapse
Affiliation(s)
- A Cybulska-Klosowicz
- Department of Molecular and Cellular Neurobiology, Nencki Institute, 3 Pasteur Street, 02-093 Warsaw, Poland
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
It has been discovered recently that toll-like receptors (TLRs) are key mediators of tissue injury in response to stroke. This revelation has identified a new target critical to understanding the underlying mechanisms of stroke injury and potential therapies. Much of the interest in TLRs centers around their ability to self regulate - a process commonly referred to as "tolerance," wherein prior exposure to low level TLR activation induces protection against a subsequent challenge that would otherwise cause damage. This endogenous process has been exploited in the setting of stroke. Recent studies show that TLR pathways can be reprogrammed via prior exposure to TLR ligands leading to decreased infarct size and improved neurological outcomes in response to ischemic injury. Efforts to understand the molecular mechanisms of TLR reprogramming have led to the identification of multiple routes of TLR regulation including inhibitors that target signaling mediators, microRNAs that suppress genes post-transcriptionally, and epigenetic changes in chromatin remodeling that affect global gene regulation. In this review, we discuss the role of TLRs in mediating injury due to stroke, evidence for TLR preconditioning-induced TLR reprogramming in response to stroke, and possible mechanisms of TLR-induced neuroprotection.
Collapse
|
42
|
Peroxisome-proliferator-activated receptors γ and peroxisome-proliferator-activated receptors β/δ and the regulation of interleukin 1 receptor antagonist expression by pioglitazone in ischaemic brain. J Hypertens 2010; 28:1488-97. [DOI: 10.1097/hjh.0b013e3283396e4e] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
Craft TKS, Devries AC. Vulnerability to stroke: implications of perinatal programming of the hypothalamic-pituitary-adrenal axis. Front Behav Neurosci 2009; 3:54. [PMID: 20057937 PMCID: PMC2802556 DOI: 10.3389/neuro.08.054.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Accepted: 11/23/2009] [Indexed: 12/11/2022] Open
Abstract
Chronic stress is capable of exacerbating each major, modifiable, endogenous risk factor for cerebrovascular and cardiovascular disease. Indeed, exposure to stress can increase both the incidence and severity of stroke, presumably through activation of the hypothalamic-pituitary-adrenal (HPA) axis. Now that characterization of the mechanisms underlying epigenetic programming of the HPA axis is well underway, there has been renewed interest in examining the role of early environment on the evolution of health conditions across the entire lifespan. Indeed, neonatal manipulations in rodents that reduce stress responsivity, and subsequent life-time exposure to glucocorticoids, are associated with a reduction in the development of neuroendocrine, neuroanatomical, and cognitive dysfunctions that typically progress with age. Although improved day to day regulation of the HPA axis also may be accompanied by a decrease in stroke risk, evidence from rodent studies suggest that an associated cost could be increased susceptibility to inflammation and neuronal death in the event that a stroke does occur and the individual is exposed to persistently elevated corticosteroids. Given its importance in regulation of health and disease states, any long-term modulation of the HPA axis is likely to be associated with both benefits and potential risks. The goals of this review article are to examine (1) the clinical and experimental data suggesting that neonatal experiences can shape HPA axis regulation, (2) the influence of stress and the HPA axis on stroke incidence and severity, and (3) the potential for neonatal programming of the HPA axis to impact adult cerebrovascular health.
Collapse
Affiliation(s)
- Tara K S Craft
- Departments of Psychology, The Ohio State University Columbus, OH, USA
| | | |
Collapse
|
44
|
Cheppudira BP, Girard BM, Malley SE, Dattilio A, Schutz KC, May V, Vizzard MA. Involvement of JAK-STAT signaling/function after cyclophosphamide-induced bladder inflammation in female rats. Am J Physiol Renal Physiol 2009; 297:F1038-44. [PMID: 19625377 DOI: 10.1152/ajprenal.00110.2009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cytokines are upregulated in a variety of inflammatory conditions and cytokine/receptor interactions can activate JAK-STAT signaling. Previous studies demonstrated upregulation of numerous cytokines in the urinary bladder following cyclophosphamide (CYP)-induced cystitis. The role of JAK-STAT signaling in urinary bladder inflammation and referred somatic sensitivity has not been addressed. The contribution of JAK-STAT signaling pathways in CYP-induced bladder hyperreflexia and referred somatic hypersensitivity was determined in CYP-treated rats using a JAK2 inhibitor, AG490. Acute (4 h; 150 mg/kg ip), intermediate (48 h; 150 mg/kg ip), or chronic (75 mg/kg ip, once every 3 days for 10 days) cystitis was induced in adult, female Wistar rats with CYP treatment. Phosphorylation status of STAT-3 was increased in urinary bladder after CYP-induced cystitis (4 h, 48 h, chronic). Blockade of JAK2 with AG490 (5-15 mg/kg ip or intravesical) significantly (P < or = 0.05) reduced bladder hyperreflexia and hind paw sensitivity in CYP-treated rats. These studies demonstrate a potential role for JAK-STAT signaling pathways in bladder hyperreflexia and referred pain induced by CYP-induced bladder inflammation.
Collapse
Affiliation(s)
- Bopaiah P Cheppudira
- Dept. of Neurology, Univ. of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Kriz J, Lalancette-Hébert M. Inflammation, plasticity and real-time imaging after cerebral ischemia. Acta Neuropathol 2009; 117:497-509. [PMID: 19225790 DOI: 10.1007/s00401-009-0496-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Revised: 02/05/2009] [Accepted: 02/06/2009] [Indexed: 12/24/2022]
Abstract
With an incidence of approximately 350 in 100,000, stroke is the third leading cause of death and a major cause of disability in industrialized countries. At present, although progress has been made in understanding the molecular pathways that lead to ischemic cell death, the current clinical treatments remain poorly effective. There is mounting evidence that inflammation plays an important role in cerebral ischemia. Experimentally and clinically, brain response to ischemic injury is associated with an acute and prolonged inflammatory process characterized by the activation of resident glial cells, production of inflammatory cytokines as well as leukocyte and monocyte infiltration in the brain, events that may contribute to ischemic brain injury and affect brain recovery and plasticity. However, whether the post-ischemic inflammatory response is deleterious or beneficial to brain recovery is presently a matter of debate and controversies. Here, we summarize the current knowledge on the molecular mechanisms underlying post-ischemic neuronal plasticity and the potential role of inflammation in regenerative processes and functional recovery after stroke. Furthermore, because of the dynamic nature of the brain inflammatory response, we highlight the importance of the development of novel experimental approaches such as real-time imaging. Finally, we discuss the novel transgenic reporter mice models that have allowed us to visualize and to analyze the processes such as neuroinflammation and neuronal repair from the ischemic brains of live animals.
Collapse
Affiliation(s)
- Jasna Kriz
- Department of Anatomy and Physiology, Faculty of Medicine, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ), T3-67, Laval University, 2705 Boulevard Laurier, Quebec, QC, G1V 4G2, Canada.
| | | |
Collapse
|
46
|
Suzuki S, Tanaka K, Suzuki N. Ambivalent aspects of interleukin-6 in cerebral ischemia: inflammatory versus neurotrophic aspects. J Cereb Blood Flow Metab 2009; 29:464-79. [PMID: 19018268 DOI: 10.1038/jcbfm.2008.141] [Citation(s) in RCA: 188] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Interleukin-6 (IL-6) is pleiotropic cytokine involved in many central nervous system disorders including stroke, and elevated serum IL-6 has been found in acute stroke patients. IL-6 is implicated in the inflammation, which contributes to both injury and repair process after cerebral ischemia. However, IL-6 is one of the neurotrophic cytokines sharing a common receptor subunit, gp130, with other neurotrophic cytokines, such as leukemia inhibitory factor (LIF) and ciliary neurotrophic factor. The expression of IL-6 is most prominently identified in neurons in the peri-ischemic regions, and LIF expression shows a similar pattern. The direct injection of these cytokines into the brain after ischemia can reduce ischemic brain injury. The cytokine receptors are localized on the neuron surface, suggesting that neurons are the cytokine target. The major IL-6 downstream signaling pathway is JAK-STAT, and Stat3 activation occurs mainly in neurons during postischemic reperfusion. Further investigation is necessary to clarify the exact role of Stat3 signaling in neuroprotection. Taken together, the information suggests that IL-6 plays a double role in cerebral ischemia, as an inflammatory mediator during the acute phase and as a neurotrophic mediator between the subacute and prolonged phases.
Collapse
Affiliation(s)
- Shigeaki Suzuki
- Department of Neurology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan.
| | | | | |
Collapse
|
47
|
Fiedorowicz A, Figiel I, Zaremba M, Dzwonek K, Schliebs R, Oderfeld-Nowak B. Trimethyltin-evoked apoptosis of murine hippocampal granule neurons is accompanied by the expression of interleukin-1beta and interleukin-1 receptor antagonist in cells of ameboid phenotype, the majority of which are NG2-positive. Brain Res Bull 2008; 77:19-26. [DOI: 10.1016/j.brainresbull.2008.02.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Accepted: 02/06/2008] [Indexed: 01/02/2023]
|
48
|
Akuzawa S, Kazui T, Shi E, Yamashita K, Bashar AHM, Terada H. Interleukin-1 receptor antagonist attenuates the severity of spinal cord ischemic injury in rabbits. J Vasc Surg 2008; 48:694-700. [DOI: 10.1016/j.jvs.2008.04.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 03/23/2008] [Accepted: 04/06/2008] [Indexed: 10/21/2022]
|
49
|
Studeny S, Cheppudira BP, Meyers S, Balestreire EM, Apodaca G, Birder LA, Braas KM, Waschek JA, May V, Vizzard MA. Urinary bladder function and somatic sensitivity in vasoactive intestinal polypeptide (VIP)-/- mice. J Mol Neurosci 2008; 36:175-87. [PMID: 18561033 DOI: 10.1007/s12031-008-9100-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 05/09/2008] [Indexed: 12/18/2022]
Abstract
Vasoactive intestinal polypeptide (VIP) is an immunomodulatory neuropeptide widely distributed in neural pathways that regulate micturition. VIP is also an endogenous anti-inflammatory agent that has been suggested for the development of therapies for inflammatory disorders. In the present study, we examined urinary bladder function and hindpaw and pelvic sensitivity in VIP(-/-) and littermate wildtype (WT) controls. We demonstrated increased bladder mass and fewer but larger urine spots on filter paper in VIP(-/-) mice. Using cystometry in conscious, unrestrained mice, VIP(-/-) mice exhibited increased void volumes and shorter intercontraction intervals with continuous intravesical infusion of saline. No differences in transepithelial resistance or water permeability were demonstrated between VIP(-/-) and WT mice; however, an increase in urea permeability was demonstrated in VIP(-/-) mice. With the induction of bladder inflammation by acute administration of cyclophosphamide, an exaggerated or prolonged bladder hyperreflexia and hindpaw and pelvic sensitivity were demonstrated in VIP(-/-) mice. The changes in bladder hyperreflexia and somatic sensitivity in VIP(-/-) mice may reflect increased expression of neurotrophins and/or proinflammatory cytokines in the urinary bladder. Thus, these changes may further regulate the neural control of micturition.
Collapse
Affiliation(s)
- Simon Studeny
- Department of Neurology, University of Vermont College of Medicine, D415A Given Research Building, Burlington, VT 05405, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Doyle KP, Yang T, Lessov NS, Ciesielski TM, Stevens SL, Simon RP, King JS, Stenzel-Poore MP. Nasal administration of osteopontin peptide mimetics confers neuroprotection in stroke. J Cereb Blood Flow Metab 2008; 28:1235-48. [PMID: 18364727 PMCID: PMC6015748 DOI: 10.1038/jcbfm.2008.17] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Osteopontin (OPN), a large secreted glycoprotein with an arginine, glycine, aspartate (RGD) motif, can bind and signal through cellular integrin receptors. We have shown previously that OPN enhances neuronal survival in the setting of ischemia. Here, we sought to increase the neuroprotective potency of OPN and improve the method of delivery with the goal of identifying a treatment for stroke in humans. We show that thrombin cleavage of OPN improves its ability to ligate integrin receptors and its neuroprotective capacity in models of ischemia. Thrombin-cleaved OPN is a twofold more effective neuroprotectant than the untreated molecule. We also tested whether OPN could be administered intranasally and found that it is efficiently targeted to the brain via intranasal delivery. Furthermore, intranasal administration of thrombin-treated OPN confers protection against ischemic brain injury. Osteopontin mimetics based on the peptide sequences located either N or C terminal to the thrombin cleavage site were generated and tested in models of ischemia. Treatment with successively shorter N-terminal peptides and a phosphorylated C-terminal peptide provided significant neuroprotection against ischemic injury. These findings show that OPN mimetics offer promise for development into new drugs for the treatment of stroke.
Collapse
Affiliation(s)
- Kristian P Doyle
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | | | | | | | | | | | | | | |
Collapse
|