1
|
Gao H, Nepovimova E, Adam V, Heger Z, Valko M, Wu Q, Kuca K. Age-associated changes in innate and adaptive immunity: role of the gut microbiota. Front Immunol 2024; 15:1421062. [PMID: 39351234 PMCID: PMC11439693 DOI: 10.3389/fimmu.2024.1421062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Aging is generally regarded as an irreversible process, and its intricate relationship with the immune system has garnered significant attention due to its profound implications for the health and well-being of the aging population. As people age, a multitude of alterations occur within the immune system, affecting both innate and adaptive immunity. In the realm of innate immunity, aging brings about changes in the number and function of various immune cells, including neutrophils, monocytes, and macrophages. Additionally, certain immune pathways, like the cGAS-STING, become activated. These alterations can potentially result in telomere damage, the disruption of cytokine signaling, and impaired recognition of pathogens. The adaptive immune system, too, undergoes a myriad of changes as age advances. These include shifts in the number, frequency, subtype, and function of T cells and B cells. Furthermore, the human gut microbiota undergoes dynamic changes as a part of the aging process. Notably, the interplay between immune changes and gut microbiota highlights the gut's role in modulating immune responses and maintaining immune homeostasis. The gut microbiota of centenarians exhibits characteristics akin to those found in young individuals, setting it apart from the microbiota observed in typical elderly individuals. This review delves into the current understanding of how aging impacts the immune system and suggests potential strategies for reversing aging through interventions in immune factors.
Collapse
Affiliation(s)
- Haoyu Gao
- College of Life Science, Yangtze University, Jingzhou, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, Slovakia
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| |
Collapse
|
2
|
Morais A, Imai T, Jin X, Locascio JJ, Boisserand L, Herman AL, Chauhan A, Lamb J, Nagarkatti K, Diniz MA, Kumskova M, Dhanesha N, Kamat PK, Khan MB, Dhandapani KM, Patel RB, Sutariya B, Shi Y, van Leyen K, Kimberly WT, Hess DC, Aronowski J, Leira EC, Koehler RC, Chauhan AK, Sansing LH, Lyden PD, Ayata C. Biological and Procedural Predictors of Outcome in the Stroke Preclinical Assessment Network (SPAN) Trial. Circ Res 2024; 135:575-592. [PMID: 39034919 PMCID: PMC11428171 DOI: 10.1161/circresaha.123.324139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND The SPAN trial (Stroke Preclinical Assessment Network) is the largest preclinical study testing acute stroke interventions in experimental focal cerebral ischemia using endovascular filament middle cerebral artery occlusion (MCAo). Besides testing interventions against controls, the prospective design captured numerous biological and procedural variables, highlighting the enormous heterogeneity introduced by the multicenter structure that might influence stroke outcomes. Here, we leveraged the unprecedented sample size achieved by the SPAN trial and the prospective design to identify the biological and procedural variables that affect experimental stroke outcomes in transient endovascular filament MCAo. METHODS The study cohort included all mice enrolled and randomized in the SPAN trial (N=1789). Mice were subjected to 60-minute MCAo and followed for a month. Thirteen biological and procedural independent variables and 4 functional (weight loss and 4-point neuroscore on days 1 and 2, corner test on days 7 and 28, and mortality) and 3 tissue (day 2, magnetic resonance imaging infarct volumes and swelling; day 30, magnetic resonance imaging tissue loss) outcome variables were prospectively captured. Multivariable regression with stepwise elimination was used to identify the predictors and their effect sizes. RESULTS Older age, active circadian stage at MCAo, and thinner and longer filament silicone tips predicted higher mortality. Older age, larger body weight, longer anesthesia duration, and longer filament tips predicted worse neuroscores, while high-fat diet and blood flow monitoring predicted milder neuroscores. Older age and a high-fat diet predicted worse corner test performance. While shorter filament tips predicted more ipsiversive turning, longer filament tips appeared to predict contraversive turning. Age, sex, and weight interacted when predicting the infarct volume. Older age was associated with smaller infarcts on day 2 magnetic resonance imaging, especially in animals with larger body weights; this association was most conspicuous in females. High-fat diet also predicted smaller infarcts. In contrast, the use of cerebral blood flow monitoring and more severe cerebral blood flow drop during MCAo, longer anesthesia, and longer filament tips all predicted larger infarcts. Bivariate analyses among the dependent variables highlighted a disconnect between tissue and functional outcomes. CONCLUSIONS Our analyses identified variables affecting endovascular filament MCAo outcome, an experimental stroke model used worldwide. Multiple regression refuted some commonly reported predictors and revealed previously unrecognized associations. Given the multicenter prospective design that represents a sampling of real-world conditions, the degree of heterogeneity mimicking clinical trials, the large number of predictors adjusted for in the multivariable model, and the large sample size, we think this is the most definitive analysis of the predictors of preclinical stroke outcome to date. Future multicenter experimental stroke trials should standardize or at least ensure a balanced representation of the biological and procedural variables identified herein as potential confounders.
Collapse
Affiliation(s)
- Andreia Morais
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Takahiko Imai
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xuyan Jin
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Joseph J Locascio
- Harvard Catalyst Biostatistical Consulting Unit, Department of Biostatistics, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Harvard Medical School, Boston MA, USA
| | - Ligia Boisserand
- Department of Neurology, Yale University School of Medicine, New Haven, CT USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT USA
| | - Alison L. Herman
- Department of Neurology, Yale University School of Medicine, New Haven, CT USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT USA
| | - Anjali Chauhan
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX, USA
| | - Jessica Lamb
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Los Angeles, CA USA
- Department of Neurology, Keck School of Medicine at USC, Los Angeles, CA, USA
| | - Karisma Nagarkatti
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Los Angeles, CA USA
- Department of Neurology, Keck School of Medicine at USC, Los Angeles, CA, USA
| | - Marcio A. Diniz
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mariia Kumskova
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nirav Dhanesha
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Pradip K. Kamat
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA
| | | | | | - Rakesh B. Patel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Brijesh Sutariya
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yanrong Shi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD
| | - Klaus van Leyen
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - W. Taylor Kimberly
- Department of Neurology, Massachusetts General Hospital, Boston, Harvard Medical School, Boston MA, USA
| | - David C. Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX, USA
| | - Enrique C. Leira
- Departments of Neurology, Neurosurgery, Carver College of Medicine, and Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Raymond C. Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD
| | - Anil K. Chauhan
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT USA
| | - Patrick D. Lyden
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Los Angeles, CA USA
- Department of Neurology, Keck School of Medicine at USC, Los Angeles, CA, USA
| | - Cenk Ayata
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Harvard Medical School, Boston MA, USA
| |
Collapse
|
3
|
Nolta NF, Christensen MB, Tresco PA. Advanced age is not a barrier to chronic intracortical single-unit recording in rat cortex. Front Neurosci 2024; 18:1389556. [PMID: 38817909 PMCID: PMC11138162 DOI: 10.3389/fnins.2024.1389556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/02/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction Available evidence suggests that as we age, our brain and immune system undergo changes that increase our susceptibility to injury, inflammation, and neurodegeneration. Since a significant portion of the potential patients treated with a microelectrode-based implant may be older, it is important to understand the recording performance of such devices in an aged population. Methods We studied the chronic recording performance and the foreign body response (FBR) to a clinically used microelectrode array implanted in the cortex of 18-month-old Sprague Dawley rats. Results and discussion To the best of our knowledge, this is the first preclinical study of its type in the older mammalian brain. Here, we show that single-unit recording performance was initially robust then gradually declined over a 12-week period, similar to what has been previously reported using younger adult rats and in clinical trials. In addition, we show that FBR biomarker distribution was similar to what has been previously described for younger adult rats implanted with multi-shank recording arrays in the motor cortex. Using a quantitative immunohistochemcal approach, we observed that the extent of astrogliosis and tissue loss near the recording zone was inversely related to recording performance. A comparison of recording performance with a younger cohort supports the notion that aging, in and of itself, is not a limiting factor for the clinical use of penetrating microelectrode recording arrays for the treatment of certain CNS disorders.
Collapse
Affiliation(s)
- Nicholas F. Nolta
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Michael B. Christensen
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
- Department of Otolaryngology – Head & Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Patrick A. Tresco
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
4
|
Bahire KL, Maļuhins R, Bello F, Upīte J, Makarovs A, Jansone B. Long-Term Region-Specific Mitochondrial Functionality Changes in Both Cerebral Hemispheres after fMCAo Model of Ischemic Stroke. Antioxidants (Basel) 2024; 13:416. [PMID: 38671864 PMCID: PMC11047464 DOI: 10.3390/antiox13040416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Cerebral ischemia/reperfusion (I/R) refers to a secondary brain injury that results in mitochondrial dysfunction of variable extent, leading to neuronal cell damage. The impact of this process has mainly been studied in the short term, from the early hours up to one week after blood flow reperfusion, and in the ischemic hemisphere only. The focus of this study was to assess the long-term impacts of I/R on mitochondrial functionality using high-resolution fluorespirometry to evaluate state-dependent activities in both ischemic (ipsilateral) and non-ischemic (contralateral) hemispheres of male mice 60, 90, 120, and 180 days after I/R caused by 60-min-long filament-induced middle cerebral artery occlusion (fMCAo). Our results indicate that in cortical tissues, succinate-supported oxygen flux (Complex I&II OXPHOS state) and H2O2 production (Complex II LEAK state) were significantly decreased in the fMCAo (stroke) group ipsilateral hemisphere compared to measurements in the contralateral hemisphere 60 and 90 days after stroke. In hippocampal tissues, during the Complex I&II ET state, mitochondrial respiration was generally lower in the ipsilateral compared to the contralateral hemisphere 90 days following stroke. An aging-dependent impact on mitochondria oxygen consumption following I/R injury was observed 180 days after surgery, wherein Complex I&II activities were lowest in both hemispheres. The obtained results highlight the importance of long-term studies in the field of ischemic stroke, particularly when evaluating mitochondrial bioenergetics in specific brain regions within and between separately affected cerebral hemispheres.
Collapse
Affiliation(s)
- Ksenija Lūcija Bahire
- Department of Pharmacology, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia; (R.M.); (F.B.); (J.U.); (A.M.)
| | | | | | | | | | - Baiba Jansone
- Department of Pharmacology, Faculty of Medicine, University of Latvia, LV-1586 Riga, Latvia; (R.M.); (F.B.); (J.U.); (A.M.)
| |
Collapse
|
5
|
Laser Speckle Flowmetry for the Prognostic Estimation Study of Permanent Focal Ischemia in Mice. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1729255. [PMID: 36177058 PMCID: PMC9514945 DOI: 10.1155/2022/1729255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/21/2022] [Accepted: 09/07/2022] [Indexed: 11/22/2022]
Abstract
The distal middle cerebral artery occlusion (dMCAO) model that mainly targets the cortex and causes low mortality is developed for the study of permanent focal ischemia, and it is highly appropriate for the study in the aged population. The two most common methods used to establish dMCAO models are dMCAO alone and dMCAO plus ipsilateral common carotid artery occlusion (CCAO). Up to now, studies on the prognosis of the two types of dMCAO models and the accuracy of cerebral blood flow (CBF) in predicting prognosis have not yet been reported. In the present study, we established permanent focal ischemia models in two groups of aged mice by dMCAO alone or by dMCAO plus ipsilateral common carotid artery occlusion (CCAO). CBF was evaluated by laser speckle flowmetry (LSF) before and after surgery. Cerebral infarction was assessed by TTC staining at day 2 after surgery and MAP2 staining at day 21 after surgery. In addition, behavioral outcomes were evaluated using the modified Garcia scoring system, adhesive removal test, and foot-fault test. Our results showed that compared with those in the dMCAO alone group, the mice in the dMCAO plus CCAO group had a larger cerebral infarct size and more severe neurological deficits. According to the results of the correlation analysis, the area of the ischemic core region on CBF imaging in the dMCAO group was helpful in predicting the infarct volume. In addition, the total CBF of the ischemic area in the dMCAO plus CCAO group showed a significant correlation with Garcia scores 3 days after surgery, but there was no significant correlation of CBF imaging with the foot-fault test 7 days after surgery. These results suggest that the total CBF of the ischemic area might be helpful to predict the severity of neurological damage at the acute stage.
Collapse
|
6
|
Decoding the Transcriptional Response to Ischemic Stroke in Young and Aged Mouse Brain. Cell Rep 2021; 31:107777. [PMID: 32553170 DOI: 10.1016/j.celrep.2020.107777] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/25/2020] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
Ischemic stroke is a well-recognized disease of aging, yet it is unclear how the age-dependent vulnerability occurs and what are the underlying mechanisms. To address these issues, we perform a comprehensive RNA-seq analysis of aging, ischemic stroke, and their interaction in 3- and 18-month-old mice. We assess differential gene expression across injury status and age, estimate cell type proportion changes, assay the results against a range of transcriptional signatures from the literature, and perform unsupervised co-expression analysis, identifying modules of genes with varying response to injury. We uncover downregulation of axonal and synaptic maintenance genetic program, and increased activation of type I interferon (IFN-I) signaling following stroke in aged mice. Together, these results paint a picture of ischemic stroke as a complex age-related disease and provide insights into interaction of aging and stroke on cellular and molecular level.
Collapse
|
7
|
Ageing as a risk factor for cerebral ischemia: Underlying mechanisms and therapy in animal models and in the clinic. Mech Ageing Dev 2020; 190:111312. [PMID: 32663480 DOI: 10.1016/j.mad.2020.111312] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/24/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022]
Abstract
Age is the only one non-modifiable risk of cerebral ischemia. Advances in stroke medicine and behavioral adaptation to stroke risk factors and comorbidities was successful in decreasing stroke incidence and increasing the number of stroke survivors in western societies. Comorbidities aggravates the outcome after cerebral ischemia. However, due to the increased in number of elderly, the incidence of stroke has increased again paralleled by an increase in the number of stroke survivors, many with severe disabilities, that has led to an increased economic and social burden in society. Animal models of stroke often ignore age and comorbidities frequently associated with senescence. This might explain why drugs working nicely in animal models fail to show efficacy in stroke survivors. Since stroke afflicts mostly the elderly comorbid patients, it is highly desirable to test the efficacy of stroke therapies in an appropriate animal stroke model. Therefore, in this review, we make parallels between animal models of stroke und clinical data and summarize the impact of ageing and age-related comorbidities on stroke outcome.
Collapse
|
8
|
Sun M, McDonald SJ, Brady RD, Collins-Praino L, Yamakawa GR, Monif M, O'Brien TJ, Cloud GC, Sobey CG, Mychasiuk R, Loane DJ, Shultz SR. The need to incorporate aged animals into the preclinical modeling of neurological conditions. Neurosci Biobehav Rev 2019; 109:114-128. [PMID: 31877345 DOI: 10.1016/j.neubiorev.2019.12.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/04/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022]
Abstract
Neurological conditions such as traumatic brain injury, stroke, Parkinson's disease, epilepsy, multiple sclerosis, and Alzheimer's disease are serious clinical problems that affect millions of people worldwide. The majority of clinical trials for these common conditions have failed, and there is a critical need to understand why treatments in preclinical animal models do not translate to patients. Many patients with these conditions are middle-aged or older, however, the majority of preclinical studies have used only young-adult animals. Considering that aging involves biological changes that are relevant to the pathobiology of neurological diseases, the lack of aged subjects in preclinical research could contribute to translational failures. This paper details how aging affects biological processes involved in neurological conditions, and reviews aging research in the context of traumatic brain injury, stroke, Parkinson's disease, epilepsy, multiple sclerosis, and Alzheimer's disease. We conclude that aging is an important, but often overlooked, factor that influences biology and outcomes in neurological conditions, and provide suggestions to improve our understanding and treatment of these diseases in aged patients.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lyndsey Collins-Praino
- Department of Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Mastura Monif
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Geoffrey C Cloud
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Stroke Services, Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia.
| |
Collapse
|
9
|
Samandari H, Nabavizadeh F, Ashabi G. Age-related difference in protective effect of early post-conditioning on ischemic brain injury: possible involvement of MAP-2/Synaptophysin role. Metab Brain Dis 2019; 34:1771-1780. [PMID: 31471737 DOI: 10.1007/s11011-019-00484-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 08/15/2019] [Indexed: 12/16/2022]
Abstract
Brain Ischemia/Reperfusion (I/R) injury leads to the failure of the microtubules function and neuronal death. Ischemic post-conditioning is defined as a series of rapid alternating interruptions of blood flow in the first seconds of reperfusion. In the present study, the caspase-3, Microtubule-Associated Protein-2 (MAP-2), Protein Kinase C α (PKCα), c-fos, and synaptophysin were evaluated in the hippocampus of focal I/R post-conditioning model in a time -dependent study in aged and young rats. Adult and aged rats were subjected to right MCAO for 30 min and post-conditioned (10 s) for 3 cycles. Sensory-motor tests were performed, and locomotion and anxiety-like behavior were evaluated. Molecular tests were done by detection kit, RT-PCR, and Western blotting techniques. Ninety-six hours after I/R post-conditioning, neurological signs, locomotion, anxiety-like behavior, and ischemic area were improved in young rats compared to 6 h after I/R post-conditioning (P < 0.001). Caspase-3 activity declined in the hippocampus and cortex of I/R post-conditioned young rats in 96 h after I/R post-conditioning compared with 6 h after I/R post-conditioning (P < 0.001). Also, MAP-2 mRNA, MAP-2 protein level, PKCα, c-fos and synaptophysin protein levels were enhanced during post-conditioning in young rats in 96 h after I/R post-conditioning compared with 6 h after induction of I/R post-conditioning. The results of the present study suggested that, early post-conditioning might be considered as a candidate for therapeutic methods against I/R in the adult animals not aged rats. Moreover, inhibition of cell death in post-conditioned ischemic rats was found to be regulated by some neuroprotective molecules as well as MAP-2 and c-fos in young rats. Graphical abstract Graphical abstract representing the post-conditioning (PC) treatment timeline in adult and old rats.
Collapse
Affiliation(s)
- Hedayat Samandari
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabavizadeh
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Cohan CH, Youbi M, Saul I, Ruiz AA, Furones CC, Patel P, Perez E, Raval AP, Dave KR, Zhao W, Dong C, Rundek T, Koch S, Sacco RL, Perez-Pinzon MA. Sex-Dependent Differences in Physical Exercise-Mediated Cognitive Recovery Following Middle Cerebral Artery Occlusion in Aged Rats. Front Aging Neurosci 2019; 11:261. [PMID: 31619985 PMCID: PMC6759590 DOI: 10.3389/fnagi.2019.00261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/04/2019] [Indexed: 01/14/2023] Open
Abstract
Stroke remains a leading cause of death and disability in the United States. No current treatments exist to promote cognitive recovery in survivors of stroke. A previous study from our laboratory determined that an acute bout of forced treadmill exercise was able to promote cognitive recovery in 3 month old male rats after middle cerebral artery occlusion (MCAo). In this study, we tested the hypothesis that 6 days of intense acute bout of forced treadmill exercise (physical exercise – PE) promotes cognitive recovery in 11–14 month old male rats. We determined that PE was able to ameliorate cognitive deficits as determined by contextual fear conditioning. Additionally, we also tested the hypothesis that PE promotes cognitive recovery in 11–13 month old reproductive senescent female rats. In contrast to males, the same intensity of exercise that decrease cognitive deficits in males was not able to promote cognitive recovery in female rats. Additionally, we determined that exercise did not lessen infarct volume in both male and female rats. There are many factors that contribute to higher stroke mortality and morbidities in women and thus, future studies will investigate the effects of PE in aged female rats to identify sex differences.
Collapse
Affiliation(s)
- Charles H Cohan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mehdi Youbi
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Isabel Saul
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alex A Ruiz
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Concepcion C Furones
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Pujan Patel
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Edwin Perez
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kunjan R Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Weizhao Zhao
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Chuanhui Dong
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Tatjana Rundek
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sebastian Koch
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ralph L Sacco
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
11
|
Kim T, Chelluboina B, Chokkalla AK, Vemuganti R. Age and sex differences in the pathophysiology of acute CNS injury. Neurochem Int 2019; 127:22-28. [PMID: 30654116 PMCID: PMC6579702 DOI: 10.1016/j.neuint.2019.01.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/11/2019] [Indexed: 12/21/2022]
Abstract
Despite the immeasurable burden on patients and families, no effective therapies to protect the CNS after an acute injury are available yet. Furthermore, the underlying mechanisms that promote neuronal death and functional deficits after injury remain to be poorly understood. The prevalence, age of onset, pathophysiology, and symptomatology of many CNS insults differ significantly between males and females. In the case of stroke, younger males tend to show a higher risk than younger females, while this trend reverses with age. Accumulating evidence from preclinical studies have shown that sex hormones play a crucial role in providing neuroprotection following ischemic stroke and other acute CNS injuries. Estrogen, in particular, exerts a neuroprotective effect by modulating the immune responses after injury. In addition, there exists a sexual dimorphism in cell death pathways between males and females that are independent of hormones. Meanwhile, recent studies suggest that microRNAs are critically involved in the sex-specific mechanisms of cell death. This review discusses the current knowledge on the contribution of sex and age to outcome after stroke. Implication of the interplay between these two factors on other CNS injuries (spinal cord injury and traumatic brain injury) from the experimental evidence were also discussed.
Collapse
Affiliation(s)
- TaeHee Kim
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Bharath Chelluboina
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA; William S. Middleton Veterans Administration Hospital, Madison, WI, USA.
| |
Collapse
|
12
|
Dief AE, Hassan PS, Hartmut O, Jirikowski GF. Neuronal and glial regeneration after focal cerebral ischemia in rat, an immunohistochemical and electron microscopical study. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2018.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Abeer E. Dief
- Department of Physiology, University of Alexandria, Egypt
| | | | | | | |
Collapse
|
13
|
Zeng J, Wang Y, Luo Z, Chang LC, Yoo JS, Yan H, Choi Y, Xie X, Deverman BE, Gradinaru V, Gupton SL, Zlokovic BV, Zhao Z, Jung JU. TRIM9-Mediated Resolution of Neuroinflammation Confers Neuroprotection upon Ischemic Stroke in Mice. Cell Rep 2019; 27:549-560.e6. [PMID: 30970257 PMCID: PMC6485958 DOI: 10.1016/j.celrep.2018.12.055] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 08/26/2018] [Accepted: 12/12/2018] [Indexed: 12/31/2022] Open
Abstract
Excessive and unresolved neuroinflammation is a key component of the pathological cascade in brain injuries such as ischemic stroke. Here, we report that TRIM9, a brain-specific tripartite motif (TRIM) protein, was highly expressed in the peri-infarct areas shortly after ischemic insults in mice, but expression was decreased in aged mice, which are known to have increased neuroinflammation after stroke. Mechanistically, TRIM9 sequestered β-transducin repeat-containing protein (β-TrCP) from the Skp-Cullin-F-box ubiquitin ligase complex, blocking IκBα degradation and thereby dampening nuclear factor κB (NF-κB)-dependent proinflammatory mediator production and immune cell infiltration to limit neuroinflammation. Consequently, Trim9-deficient mice were highly vulnerable to ischemia, manifesting uncontrolled neuroinflammation and exacerbated neuropathological outcomes. Systemic administration of a recombinant TRIM9 adeno-associated virus that drove brain-wide TRIM9 expression effectively resolved neuroinflammation and alleviated neuronal death, especially in aged mice. These findings reveal that TRIM9 is essential for resolving NF-κB-dependent neuroinflammation to promote recovery and repair after brain injury and may represent an attractive therapeutic target.
Collapse
Affiliation(s)
- Jianxiong Zeng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yaoming Wang
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Zhifei Luo
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Lin-Chun Chang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ji Seung Yoo
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Huan Yan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Younho Choi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Xiaochun Xie
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Benjamin E Deverman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Stephanie L Gupton
- Neuroscience Center and Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Zhen Zhao
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Jae U Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
14
|
Schosserer M, Banks G, Dogan S, Dungel P, Fernandes A, Marolt Presen D, Matheu A, Osuchowski M, Potter P, Sanfeliu C, Tuna BG, Varela-Nieto I, Bellantuono I. Modelling physical resilience in ageing mice. Mech Ageing Dev 2018; 177:91-102. [PMID: 30290161 PMCID: PMC6445352 DOI: 10.1016/j.mad.2018.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 09/12/2018] [Accepted: 10/01/2018] [Indexed: 02/06/2023]
Abstract
Geroprotectors, a class of drugs targeting multiple deficits occurring with age, necessitate the development of new animal models to test their efficacy. The COST Action MouseAGE is a European network whose aim is to reach consensus on the translational path required for geroprotectors, interventions targeting the biology of ageing. In our previous work we identified frailty and loss of resilience as a potential target for geroprotectors. Frailty is the result of an accumulation of deficits, which occurs with age and reduces the ability to respond to adverse events (physical resilience). Modelling frailty and physical resilience in mice is challenging for many reasons. There is no consensus on the precise definition of frailty and resilience in patients or on how best to measure it. This makes it difficult to evaluate available mouse models. In addition, the characterization of those models is poor. Here we review potential models of physical resilience, focusing on those where there is some evidence that the administration of acute stressors requires integrative responses involving multiple tissues and where aged mice showed a delayed recovery or a worse outcome then young mice in response to the stressor. These models include sepsis, trauma, drug- and radiation exposure, kidney and brain ischemia, exposure to noise, heat and cold shock.
Collapse
Affiliation(s)
- Markus Schosserer
- University of Natural Resources and Life Sciences, Vienna, Department of Biotechnology, Vienna, Austria
| | - Gareth Banks
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, United Kingdom
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Peter Dungel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria
| | - Adelaide Fernandes
- Neuron-Glia Biology in Health and Disease, iMed.ULisboa, Research Institute for Medicines, Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria
| | - Ander Matheu
- Oncology Department, Biodonostia Research Institute, San Sebastián, Spain
| | - Marcin Osuchowski
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria
| | - Paul Potter
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, United Kingdom
| | - Coral Sanfeliu
- Institute of Biomedical Research of Barcelona (IIBB) CSIC, IDIBAPS, CIBERESP, Barcelona, Spain
| | - Bilge Guvenc Tuna
- Department of Medical Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | | | - Ilaria Bellantuono
- MRC/Arthritis Research-UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Oncology and Metabolism, The Medical School, Beech Hill Road, Sheffield, S10 2RX, United Kingdom.
| |
Collapse
|
15
|
Sun Y, Sun X, Qu H, Zhao S, Xiao T, Zhao C. Neuroplasticity and behavioral effects of fluoxetine after experimental stroke. Restor Neurol Neurosci 2018; 35:457-468. [PMID: 28854520 DOI: 10.3233/rnn-170725] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The brain can undergo self-repair and has the ability to compensate for functions lost after a stroke. The plasticity of the ischemic brain is influenced by several factors including aging and pharmacotherapy. Fluoxetine is an antidepressant which enhances serotonergic neurotransmission through selective inhibition of neuronal reuptake of serotonin. In clinical practice, fluoxetine alleviates the symptoms of post-stroke depression (PSD), helps motor recovery in stroke patients. In animal experiments, chronic administration of fluoxetine induces increased excitability of mature granule cells (GCs), enhancing axonal and dendritic reorganization, as well as promoting neurogenesis or angiogenesis in the dentate gurus (DG), but the effect of fluoxetine in the subventricular zone (SVZ) remains controversial. Meanwhile, chronic treatment with fluoxetine did not reverse age-dependent suppression of proliferation cells in the DG. Interestingly, although fluoxetine has been found to enhance neurogenesis in the DG in stroke rats, this property is not consistent with the behavioral recovery. More studies into this issue will be required to reveal how to translate enhanced neuronal plasticity into behavioral benefits. This review provides an update of the current knowledge about the neurogenesis and the fate of the newly generated cells after the use of fluoxetine, as well as its ability to promote a behavioral recovery after stroke in clinical and experimental results and attempts to define the therapeutic properties of fluoxetine in regenerative neuroscience.
Collapse
Affiliation(s)
- Yefei Sun
- Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Xiaoyu Sun
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Huiling Qu
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Shanshan Zhao
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Ting Xiao
- Dermatology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Health, Ministry of Education, Shenyang, China
| | - Chuansheng Zhao
- Neurology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Kelly KM, Jukkola PI, Yin G, Miller ER, Kharlamov EA, Shiau DS, Strong R, Aronowski J. Poststroke epilepsy following transient unilateral middle cerebral and common carotid artery occlusion in young adult and aged F344 rats. Epilepsy Res 2018; 141:38-47. [PMID: 29455049 PMCID: PMC5879023 DOI: 10.1016/j.eplepsyres.2018.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/29/2017] [Accepted: 02/07/2018] [Indexed: 01/14/2023]
Abstract
The mechanisms of injured brain that establish poststroke seizures and epilepsy are not well understood, largely because animal modeling has had limited development. The main objective of this study was to determine whether an arterial occlusion model of cortical stroke in young adult and aged rats was capable of generating either focal or generalized epileptic seizures within 2 months of lesioning. Four- and 20-month-old male Fischer 344 (F344) sham-operated controls and those lesioned by transient (3 h) unilateral middle cerebral artery (MCA) and common carotid artery (CCA) occlusion (MCA/CCAo) were studied by video-EEG recordings up to 2 months post-procedure. The main findings were: 1) seizures (grade 3 and above) were recorded within 2 months in both young (4-month; 0.23/h) and aged (20-month; 1.93/h) MCA/CCAo rat groups; both MCA/CCAo rat groups had more seizures recorded than the respective control groups, i.e., no seizures in young controls and 0.52/h in old controls; 2) both age and infarction independently had effects on seizure frequency; however, there was no demonstrated interaction between the two factors; and 3) there was no difference in infarct volumes comparing 4- to 20-month-old MCA/CCAo animals. In addition, all lesioned and sham-operated animals demonstrated intermittent solitary myoclonic convulsions arising out of sleep. Morbidity and mortality of animals limited the extent to which the animals could be evaluated, especially 20-month-old animals. These results suggest that transient unilateral MCA/CCAo can result in poststroke epileptic seizures in both young adult and aged F344 rats within a relatively brief period of time following lesioning.
Collapse
Affiliation(s)
- Kevin M Kelly
- Department of Neurology and Allegheny Health Network Research Institute, Allegheny General Hospital, Pittsburgh, PA, USA; Departments of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA; Departments of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Peter I Jukkola
- Department of Neurology and Allegheny Health Network Research Institute, Allegheny General Hospital, Pittsburgh, PA, USA
| | - Guo Yin
- Department of Neurology and Allegheny Health Network Research Institute, Allegheny General Hospital, Pittsburgh, PA, USA
| | - Eric R Miller
- Department of Neurology and Allegheny Health Network Research Institute, Allegheny General Hospital, Pittsburgh, PA, USA
| | - Elena A Kharlamov
- Department of Neurology and Allegheny Health Network Research Institute, Allegheny General Hospital, Pittsburgh, PA, USA; Departments of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | - Roger Strong
- Stroke Program, Department of Neurology, University of Texas at Houston McGovern Medical School, Houston, TX, USA
| | - Jaroslaw Aronowski
- Stroke Program, Department of Neurology, University of Texas at Houston McGovern Medical School, Houston, TX, USA
| |
Collapse
|
17
|
Zhou HJ, Li H, Shi MQ, Mao XN, Liu DL, Chang YR, Gan YM, Kuang X, Du JR. Protective Effect of Klotho against Ischemic Brain Injury Is Associated with Inhibition of RIG-I/NF-κB Signaling. Front Pharmacol 2018; 8:950. [PMID: 29403373 PMCID: PMC5778393 DOI: 10.3389/fphar.2017.00950] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/14/2017] [Indexed: 01/19/2023] Open
Abstract
Aging is the greatest independent risk factor for the occurrence of stroke and poor outcomes, at least partially through progressive increases in oxidative stress and inflammation with advanced age. Klotho is an antiaging gene, the expression of which declines with age. Klotho may protect against neuronal oxidative damage that is induced by glutamate. The present study investigated the effects of Klotho overexpression and knockdown by an intracerebroventricular injection of a lentiviral vector that encoded murine Klotho (LV-KL) or rat Klotho short-hairpin RNA (LV-KL shRNA) on cerebral ischemia injury and the underlying anti-neuroinflammatory mechanism. The overexpression of Klotho induced by LV-KL significantly improved neurobehavioral deficits and increased the number of live neurons in the hippocampal CA1 and caudate putamen subregions 72 h after cerebral hypoperfusion that was induced by transient bilateral common carotid artery occlusion (2VO) in mice. The overexpression of Klotho significantly decreased the immunoreactivity of glial fibrillary acidic protein and ionized calcium binding adaptor molecule-1, the expression of retinoic-acid-inducible gene-I, the nuclear translocation of nuclear factor-κB, and the production of proinflammatory cytokines (tumor necrosis factor α and interleukin-6) in 2VO mice. The knockdown of Klotho mediated by LV-KL shRNA in the brain exacerbated neurological dysfunction and cerebral infarct after 22 h of reperfusion following 2 h middle cerebral artery occlusion in rats. These findings suggest that Klotho itself or enhancers of Klotho may compensate for its aging-related decline, thus providing a promising therapeutic approach for acute ischemic stroke during advanced age.
Collapse
Affiliation(s)
- Hong-Jing Zhou
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hui Li
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Meng-Qi Shi
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xiao-Na Mao
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Dong-Ling Liu
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yi-Ran Chang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yu-Miao Gan
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xi Kuang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery System, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Kawai T, Okochi Y, Ozaki T, Imura Y, Koizumi S, Yamazaki M, Abe M, Sakimura K, Yamashita T, Okamura Y. Unconventional role of voltage‐gated proton channels (
VSOP
/Hv1) in regulation of microglial
ROS
production. J Neurochem 2017. [DOI: 10.1111/jnc.14106] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Takafumi Kawai
- Integrative Physiology Department of Physiology Graduate School of Medicine & Frontier Biosciences Osaka University Suita Osaka Japan
| | - Yoshifumi Okochi
- Integrative Physiology Department of Physiology Graduate School of Medicine & Frontier Biosciences Osaka University Suita Osaka Japan
| | - Tomohiko Ozaki
- Department of Molecular Neuroscience Graduate School of Medicine Osaka University Suita Osaka Japan
| | - Yoshio Imura
- Department of Neuropharmacology Interdisciplinary Graduate School of Medicine University of Yamanashi Chuo Yamanashi Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology Interdisciplinary Graduate School of Medicine University of Yamanashi Chuo Yamanashi Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology Brain Research Institute Niigata University Niigata Japan
| | - Manabu Abe
- Department of Cellular Neurobiology Brain Research Institute Niigata University Niigata Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology Brain Research Institute Niigata University Niigata Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience Graduate School of Medicine Osaka University Suita Osaka Japan
| | - Yasushi Okamura
- Integrative Physiology Department of Physiology Graduate School of Medicine & Frontier Biosciences Osaka University Suita Osaka Japan
| |
Collapse
|
19
|
Sandu RE, Balseanu AT, Bogdan C, Slevin M, Petcu E, Popa-Wagner A. Stem cell therapies in preclinical models of stroke. Is the aged brain microenvironment refractory to cell therapy? Exp Gerontol 2017; 94:73-77. [PMID: 28093317 DOI: 10.1016/j.exger.2017.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 01/01/2023]
Abstract
Stroke is a devastating disease demanding vigorous search for new therapies. Initial enthusiasm to stimulate restorative processes in the ischemic brain by means of cell-based therapies has meanwhile converted into a more balanced view recognizing impediments that may be related to unfavorable age-associated environments. Recent results using a variety of drug, cell therapy or combination thereof suggest that, (i) treatment with Granulocyte-Colony Stimulating Factor (G-CSF) in aged rats has primarily a beneficial effect on functional outcome most likely via supportive cellular processes such as neurogenesis; (ii) the combination therapy, G-CSF with mesenchymal cells (G-CSF+BM-MSC or G-CSF+BM-MNC) did not further improve behavioral indices, neurogenesis or infarct volume as compared to G-CSF alone in aged animals; (iii) better results with regard to integration of transplanted cells in the aged rat environment have been obtained using iPS of human origin; (iv) mesenchymal cells may be used as drug carriers for the aged post-stroke brains. CONCLUSION While the middle aged brain does not seem to impair drug and cell therapies, in a real clinical practice involving older post-stroke patients, successful regenerative therapies would have to be carried out for a much longer time.
Collapse
Affiliation(s)
- Raluca Elena Sandu
- University of Medicine and Pharmacy of Craiova, Chair of Biochemistry, Neurobiology of Aging Group, Romania
| | - Adrian Tudor Balseanu
- University of Medicine and Pharmacy of Craiova, Chair of Biochemistry, Neurobiology of Aging Group, Romania
| | - Catalin Bogdan
- University of Medicine and Pharmacy of Craiova, Chair of Biochemistry, Neurobiology of Aging Group, Romania
| | - Mark Slevin
- Department of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Eugen Petcu
- Griffith University School of Medicine, Gold Coast Campus, QLD 4222, Australia
| | - Aurel Popa-Wagner
- Department of Psychiatry, University Hospital Rostock, Germany; University of Medicine and Pharmacy of Craiova, Chair of Biochemistry, Neurobiology of Aging Group, Romania.
| |
Collapse
|
20
|
Turner RC, DiPasquale K, Logsdon AF, Tan Z, Naser ZJ, Huber JD, Rosen CL, Lucke-Wold BP. The role for infarct volume as a surrogate measure of functional outcome following ischemic stroke. JOURNAL OF SYSTEMS AND INTEGRATIVE NEUROSCIENCE 2016; 2. [PMID: 28299202 PMCID: PMC5347398 DOI: 10.15761/jsin.1000136] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The failed translation of proposed therapeutic agents for ischemic stroke from preclinical to clinical studies has led to increased scrutiny of preclinical studies, namely the model and outcome measures utilized. Preclinical studies routinely use infarct volume as an experimental endpoint or measure in studies employing young-adult, healthy male animals despite the fact that clinically, ischemic stroke is a disease of the elderly and improvements in functional outcome from pre- to post-intervention remains the most widely utilized assessment. The validity of infarct volume as a surrogate measure for functional outcome remains unclear in clinical studies as well as preclinical studies, particularly those utilizing a more clinically relevant aged thromboembolic model. In this work, we will address the relationship between acute and chronic functional outcome and infarct volume using a variety of functional assessments ranging from more simplistic, subjective measurements such as the modified Neurologic Severity Score (mNSS), to more complex, objective measurements such as grip strength and inclined plane.
Collapse
Affiliation(s)
- Ryan C Turner
- Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA; Center for Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA
| | - Kenneth DiPasquale
- Center for Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA; Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia 26506, USA
| | - Aric F Logsdon
- Center for Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA; Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia 26506, USA
| | - Zhenjun Tan
- Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA; Center for Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA
| | - Zachary J Naser
- Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA; Center for Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA
| | - Jason D Huber
- Center for Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA; Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia 26506, USA
| | - Charles L Rosen
- Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA; Center for Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA
| | - Brandon P Lucke-Wold
- Department of Neurosurgery, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA; Center for Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia 26506, USA
| |
Collapse
|
21
|
Kent TA, Mandava P. Embracing Biological and Methodological Variance in a New Approach to Pre-Clinical Stroke Testing. Transl Stroke Res 2016; 7:274-83. [PMID: 27018014 PMCID: PMC5425098 DOI: 10.1007/s12975-016-0463-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/08/2016] [Accepted: 03/15/2016] [Indexed: 12/12/2022]
Abstract
High-profile failures in stroke clinical trials have discouraged clinical translation of neuroprotectants. While there are several plausible explanations for these failures, we believe that the fundamental problem is the way clinical and pre-clinical studies are designed and analyzed for heterogeneous disorders such as stroke due to innate biological and methodological variability that current methods cannot capture. Recent efforts to address pre-clinical rigor and design, while important, are unable to account for variability present even in genetically homogenous rodents. Indeed, efforts to minimize variability may lessen the clinical relevance of pre-clinical models. We propose a new approach that recognizes the important role of baseline stroke severity and other factors in influencing outcome. Analogous to clinical trials, we propose reporting baseline factors that influence outcome and then adapting for the pre-clinical setting a method developed for clinical trial analysis where the influence of baseline factors is mathematically modeled and the variance quantified. A new therapy's effectiveness is then evaluated relative to the pooled outcome variance at its own baseline conditions. In this way, an objective threshold for robustness can be established that must be overcome to suggest its effectiveness when expanded to broader populations outside of the controlled environment of the PI's laboratory. The method is model neutral and subsumes sources of variance as reflected in baseline factors such as initial stroke severity. We propose that this new approach deserves consideration for providing an objective method to select agents worthy of the commitment of time and resources in translation to clinical trials.
Collapse
Affiliation(s)
- Thomas A Kent
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, McNair Campus, 7200 Cambridge St. 9th Floor, MS: BCM609, Houston, TX, 77030, USA.
- Michael E. DeBakey VA Medical Center Stroke Program and Center for Translational Research on Inflammatory Diseases, Houston, TX, USA.
| | - Pitchaiah Mandava
- Stroke Outcomes Laboratory, Department of Neurology, Baylor College of Medicine, McNair Campus, 7200 Cambridge St. 9th Floor, MS: BCM609, Houston, TX, 77030, USA
- Michael E. DeBakey VA Medical Center Stroke Program and Center for Translational Research on Inflammatory Diseases, Houston, TX, USA
| |
Collapse
|
22
|
Livingston-Thomas J, Nelson P, Karthikeyan S, Antonescu S, Jeffers MS, Marzolini S, Corbett D. Exercise and Environmental Enrichment as Enablers of Task-Specific Neuroplasticity and Stroke Recovery. Neurotherapeutics 2016; 13:395-402. [PMID: 26868018 PMCID: PMC4824016 DOI: 10.1007/s13311-016-0423-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Improved stroke care has resulted in greater survival, but >50% of patients have chronic disabilities and 33% are institutionalized. While stroke rehabilitation is helpful, recovery is limited and the most significant gains occur in the first 2-3 months. Stroke triggers an early wave of gene and protein changes, many of which are potentially beneficial for recovery. It is likely that these molecular changes are what subserve spontaneous recovery. Two interventions, aerobic exercise and environmental enrichment, have pleiotropic actions that influence many of the same molecular changes associated with stroke injury and subsequent spontaneous recovery. Enrichment paradigms have been used for decades in adult and neonatal animal models of brain injury and are now being adapted for use in the clinic. Aerobic exercise enhances motor recovery and helps reduce depression after stroke. While exercise attenuates many of the signs associated with normal aging (e.g., hippocampal atrophy), its ability to reverse cognitive impairments subsequent to stroke is less evident. It may be that stroke, like other diseases such as cancer, needs to use multimodal treatments that augment complimentary neurorestorative processes.
Collapse
Affiliation(s)
- Jessica Livingston-Thomas
- Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Paul Nelson
- Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Sudhir Karthikeyan
- Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Sabina Antonescu
- Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Matthew Strider Jeffers
- Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Susan Marzolini
- Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, ON, Canada
- Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Dale Corbett
- Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada.
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
23
|
Chisholm NC, Sohrabji F. Astrocytic response to cerebral ischemia is influenced by sex differences and impaired by aging. Neurobiol Dis 2016; 85:245-253. [PMID: 25843666 PMCID: PMC5636213 DOI: 10.1016/j.nbd.2015.03.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/16/2015] [Accepted: 03/26/2015] [Indexed: 12/21/2022] Open
Abstract
Ischemic stroke occurs more often among the elderly, and within this demographic, women are at an increased risk for stroke and have poorer functional recovery than men. This is also well replicated in animal studies where aging females are shown to have more extensive brain tissue loss as compared to adult females. Astrocytes provide nutrients for neurons, regulate glutamate levels, and release neurotrophins and thus play a key role in the events that occur following ischemia. In addition, astrocytes express receptors for gonadal hormones and synthesize several neurosteroids suggesting that the sex differences in stroke outcome may be mediated through astrocytes. This review discusses key astrocytic responses to ischemia including, reactive gliosis, excitotoxicity, and neuroinflammation. In light of the age and sex differences in stroke outcomes, this review highlights how aging and gonadal hormones influence these responses. Lastly, astrocyte specific changes in gene expression and epigenetic modifications during aging and following ischemia are discussed as possible molecular mechanisms for impaired astrocytic functioning.
Collapse
Affiliation(s)
- Nioka C Chisholm
- Department of Neuroscience and Experimental Therapeutics, Texas A & M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Texas A & M Health Science Center, College of Medicine, Bryan, TX 77807, USA.
| |
Collapse
|
24
|
Sun X, Zhou Z, Liu T, Zhao M, Zhao S, Xiao T, Jolkkonen J, Zhao C. Fluoxetine Enhances Neurogenesis in Aged Rats with Cortical Infarcts, but This is not Reflected in a Behavioral Recovery. J Mol Neurosci 2015; 58:233-42. [PMID: 26474565 DOI: 10.1007/s12031-015-0662-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/05/2015] [Indexed: 01/28/2023]
Abstract
Age is associated with poor outcome and impaired functional recovery after stroke. Fluoxetine, which is widely used in clinical practice, can regulate hippocampal neurogenesis in young rodents. As the rate of neurogenesis is dramatically reduced during aging, we studied the effect of post-stroke fluoxetine treatment on neurogenesis in the subventricular zone (SVZ) and subgranular zone (SGZ) of dentate gyrus (DG) and whether this would be associated with any behavioral recovery after the cortical infarct in aged rats. Aged rats were randomly assigned to four groups: sham-operated rats, sham-operated rats treated with fluoxetine, rats subjected to cerebral ischemia, and rats with ischemia treated with fluoxetine. Focal cortical ischemia was induced by intracranial injection of vasoconstrictive peptide, endothelin-1 (ET-1). Fluoxetine was administered in the drinking water for 3 weeks starting 1 week after ischemia at a dose of 18 mg/kg/day. Behavioral recovery was evaluated on post-stroke days 29 to 31 after which the survival rate and fate of proliferating cells in the SVZ and DG were assessed by immunohistochemistry. Apoptosis was measured with the TUNEL assay. The results indicated that chronic fluoxetine treatment after stroke enhanced the proliferation of newborn neurons in the SVZ, but not in SGZ, and it suppressed perilesional apoptosis. Fluoxetine treatment did not affect the survival or differentiation of newly generated cells in the SVZ i.e., the enhanced neurogenesis was not translated into a behavioral outcome.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China
| | - Zhike Zhou
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China
| | - Tingting Liu
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China
| | - Mei Zhao
- Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shanshan Zhao
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China
| | - Ting Xiao
- Dermatology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Immunodermatology, Ministry of Health, Ministry of Education, Shenyang, China
| | - Jukka Jolkkonen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, P. O. Box 1627, 70211, Kuopio, Finland
| | - Chuansheng Zhao
- Neurology, The First Hospital of China Medical University, No.155, North Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
25
|
White matter injury and microglia/macrophage polarization are strongly linked with age-related long-term deficits in neurological function after stroke. Exp Neurol 2015; 272:109-19. [PMID: 25836044 DOI: 10.1016/j.expneurol.2015.03.021] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 03/19/2015] [Accepted: 03/21/2015] [Indexed: 01/04/2023]
Abstract
Most of the successes in experimental models of stroke have not translated well to the clinic. One potential reason for this failure is that stroke mainly afflicts the elderly and the majority of experimental stroke studies rely on data gathered from young adult animals. Therefore, in the present study we established a reliable, reproducible model of stroke with low mortality in aged (18month) male mice and contrasted their pathophysiological changes with those in young (2month) animals. To this end, mice were subjected to permanent tandem occlusion of the left distal middle cerebral artery (dMCAO) with ipsilateral common carotid artery occlusion (CCAO). Cerebral blood flow (CBF) was evaluated repeatedly during and after stroke. Reduction of CBF was more dramatic and sustained in aged mice. Aged mice exhibited more severe long-term sensorimotor deficits, as manifested by deterioration of performance in the Rotarod and hanging wire tests up to 35d after stroke. Aged mice also exhibited significantly worse long-term cognitive deficits after stroke, as measured by the Morris water maze test. Consistent with these behavioral observations, brain infarct size and neuronal tissue loss after dMCAO were significantly larger in aged mice at 2d and 14d, respectively. The young versus aged difference in neuronal tissue loss, however, did not persist until 35d after dMCAO. In contrast to the transient difference in neuronal tissue loss, we found significant and long lasting deterioration of white matter in aged animals, as revealed by the loss of myelin basic protein (MBP) staining in the striatum at 35d after dMCAO. We further examined the expression of M1 (CD16/CD32) and M2 (CD206) markers in Iba-1(+) microglia by double immunofluorescent staining. In both young and aged mice, the expression of M2 markers peaked around 7d after stroke whereas the expression of M1 markers peaked around 14d after stroke, suggesting a progressive M2-to-M1 phenotype shift in both groups. However, aged mice exhibited significantly reduced M2 polarization compared to young adults. Remarkably, we discovered a strong positive correlation between favorable neurological outcomes after dMCAO and MBP levels or the number of M2 microglia/macrophages. In conclusion, our studies suggest that the distal MCAO stroke model consistently results in ischemic brain injury with long-term behavioral deficits, and is therefore suitable for the evaluation of long-term stroke outcomes. Furthermore, aged mice exhibit deterioration of functional outcomes after stroke and this deterioration is linked to white matter damage and reductions in M2 microglia/macrophage polarization.
Collapse
|
26
|
Qu H, Zhao M, Zhao S, Xiao T, Song C, Cao Y, Jolkkonen J, Zhao C. Forced limb-use enhanced neurogenesis and behavioral recovery after stroke in the aged rats. Neuroscience 2015; 286:316-24. [DOI: 10.1016/j.neuroscience.2014.11.040] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/21/2014] [Accepted: 11/10/2014] [Indexed: 11/25/2022]
|
27
|
Neuroprotection for ischaemic stroke: Current status and challenges. Pharmacol Ther 2015; 146:23-34. [DOI: 10.1016/j.pharmthera.2014.09.003] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 12/31/2022]
|
28
|
Lucke-Wold BP, Logsdon AF, Turner RC, Rosen CL, Huber JD. Aging, the metabolic syndrome, and ischemic stroke: redefining the approach for studying the blood-brain barrier in a complex neurological disease. ADVANCES IN PHARMACOLOGY 2014; 71:411-49. [PMID: 25307225 DOI: 10.1016/bs.apha.2014.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier (BBB) has many important functions in maintaining the brain's immune-privileged status. Endothelial cells, astrocytes, and pericytes have important roles in preserving vasculature integrity. As we age, cell senescence can contribute to BBB compromise. The compromised BBB allows an influx of inflammatory cytokines to enter the brain. These cytokines lead to neuronal and glial damage. Ultimately, the functional changes within the brain can cause age-related disease. One of the most prominent age-related diseases is ischemic stroke. Stroke is the largest cause of disability and is third largest cause of mortality in the United States. The biggest risk factors for stroke, besides age, are results of the metabolic syndrome. The metabolic syndrome, if unchecked, quickly advances to outcomes that include diabetes, hypertension, cardiovascular disease, and obesity. The contribution from these comorbidities to BBB compromise is great. Some of the common molecular pathways activated include: endoplasmic reticulum stress, reactive oxygen species formation, and glutamate excitotoxicity. In this chapter, we examine how age-related changes to cells within the central nervous system interact with comorbidities. We then look at how comorbidities lead to increased risk for stroke through BBB disruption. Finally, we discuss key molecular pathways of interest with a focus on therapeutic targets that warrant further investigation.
Collapse
Affiliation(s)
- Brandon P Lucke-Wold
- Department of Neurosurgery, West Virginia University, School of Medicine, Morgantown, West Virginia, USA; The Center for Neuroscience, West Virginia University, School of Medicine, Morgantown, West Virginia, USA
| | - Aric F Logsdon
- The Center for Neuroscience, West Virginia University, School of Medicine, Morgantown, West Virginia, USA; Department of Basic Pharmaceutical Sciences, West Virginia University, School of Pharmacy, Morgantown, West Virginia, USA
| | - Ryan C Turner
- Department of Neurosurgery, West Virginia University, School of Medicine, Morgantown, West Virginia, USA; The Center for Neuroscience, West Virginia University, School of Medicine, Morgantown, West Virginia, USA
| | - Charles L Rosen
- Department of Neurosurgery, West Virginia University, School of Medicine, Morgantown, West Virginia, USA; The Center for Neuroscience, West Virginia University, School of Medicine, Morgantown, West Virginia, USA
| | - Jason D Huber
- The Center for Neuroscience, West Virginia University, School of Medicine, Morgantown, West Virginia, USA; Department of Basic Pharmaceutical Sciences, West Virginia University, School of Pharmacy, Morgantown, West Virginia, USA.
| |
Collapse
|
29
|
The influence of aging on poststroke depression using a rat model via middle cerebral artery occlusion. COGNITIVE AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2014; 13:847-59. [PMID: 23761136 DOI: 10.3758/s13415-013-0177-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Poststroke depression (PSD) is the most frequent psychological sequela following stroke. While previous studies describe the impact of age on brain infarct volume, brain edema, and blood-brain barrier (BBB) breakdown following ischemia, the role of age on PSD has yet to be described. Here, we examine the influence of age on PSD progression in a rat model of PSD by middle cerebral artery occlusion (MCAO). One hundred forty-three rats were divided into three groups. 48 rats 20 weeks of age underwent a sham procedure, 51 rats 20 weeks of age had MCAO, and 44 rats 22-26 months of age had MCAO. Groups were further divided into two subgroups. The first subgroup was used to measure infarct lesion volume, brain edema, and BBB breakdown at 24 h. In the second subgroup at 3 weeks after MCAO, rats were subjected to a sucrose preference test, two-way shuttle avoidance task, forced swimming test, and a brain-derived neurotrophic factor (BDNF) protein level measurement. Total and striatal infarct volume, brain edema, and BBB breakdown in the striatum were increased in older rats, as compared with younger rats. While both old and young rats exhibited depressive-like behaviors on each of the behavioral tests and lower BDNF levels post-MCAO, as compared with control rats, there were no differences between old and young rats. Although older rats suffered from larger infarct volumes, increased brain edema and more BBB disruption following MCAO, the lack of behavioral differences between young and old rats suggests that there was no effect of rat age on the incidence of PSD.
Collapse
|
30
|
Dong P, Zhao J, Zhang Y, Dong J, Zhang L, Li D, Li L, Zhang X, Yang B, Lei W. Aging causes exacerbated ischemic brain injury and failure of sevoflurane post-conditioning: role of B-cell lymphoma-2. Neuroscience 2014; 275:2-11. [PMID: 24929064 DOI: 10.1016/j.neuroscience.2014.05.064] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/15/2014] [Accepted: 05/29/2014] [Indexed: 11/26/2022]
Abstract
Aging is associated with exacerbated brain injury after ischemic stroke. Herein, we explored the possible mechanisms underlying the age-associated exacerbated brain injury after ischemic stroke and determined whether therapeutic intervention with anesthetic post-conditioning would provide neuroprotection in aged rats. Male Fisher 344 rats (young, 4 months; aged, 24 months) underwent 2h of middle cerebral artery occlusion (MCAO) followed by 24-h reperfusion, with or without sevoflurane post-conditioning for 15 min immediately at the onset of reperfusion. Compared with young rats, aged rats showed larger infarct size, worse neurological scores and more TUNEL-positive cells in the penumbral cerebral cortex at 24h after MCAO. However, edema formation and motor coordination were similar in both groups. Sevoflurane reduced the infarct size, edema formation, and TUNEL-positive cells, and improved the neurological outcome in young rats but not in aged rats. Molecular studies revealed that basal expression of the anti-apoptotic molecule B-cell lymphoma-2 (Bcl-2) in the brain was lower in aged rats compared with young rats before MCAO, while basal expression of the pro-apoptotic molecule Bcl-2-associated X protein (Bax) showed similar levels in both groups. MCAO reduced Bcl-2 expression and increased Bax expression in both groups; however, Bax increase was more pronounced in aged rats. In young rats, sevoflurane reversed the above MCAO-induced changes. In contrast, sevoflurane failed to enhance Bcl-2 expression but decreased Bax expression in aged rats. These findings suggest that aging-associated reduction in basal Bcl-2 expression in the brain contributes to increased neuronal injury by enhancing cell apoptosis after ischemic stroke. Sevoflurane post-conditioning failed to provide neuroprotection in aged rats, probably due to its inability to increase Bcl-2 levels and prevent apoptosis in the brain.
Collapse
Affiliation(s)
- P Dong
- Department of Anesthesiology, Qilu Hospital, Shandong University, No. 44 Wenhua Xi Road, Jinan City, Shandong Province 250012, China
| | - J Zhao
- Department of Anesthesiology, The People's Hospital of Chiping, No. 136 Wenhua Road, Chiping City, Shandong Province 252100, China
| | - Y Zhang
- Department of Anesthesiology, The First People's Hospital of Jining, No. 6 Jiankang Road, Jining City, Shandong Province 272011, China
| | - J Dong
- Department of Gynaecology and Obstetrics, Qilu Hospital, Shandong University, No. 44 Wenhua Xi Road, Jinan City, Shandong Province 250012, China
| | - L Zhang
- Department of Anesthesiology, Qilu Hospital, Shandong University, No. 44 Wenhua Xi Road, Jinan City, Shandong Province 250012, China
| | - D Li
- Department of Anesthesiology, Qilu Hospital, Shandong University, No. 44 Wenhua Xi Road, Jinan City, Shandong Province 250012, China
| | - L Li
- Department of Anesthesiology, Qilu Hospital, Shandong University, No. 44 Wenhua Xi Road, Jinan City, Shandong Province 250012, China
| | - X Zhang
- Department of Anesthesiology, Qilu Hospital, Shandong University, No. 44 Wenhua Xi Road, Jinan City, Shandong Province 250012, China
| | - B Yang
- Department of Anesthesiology, Qilu Hospital, Shandong University, No. 44 Wenhua Xi Road, Jinan City, Shandong Province 250012, China
| | - W Lei
- Department of Anesthesiology, Qilu Hospital, Shandong University, No. 44 Wenhua Xi Road, Jinan City, Shandong Province 250012, China.
| |
Collapse
|
31
|
Piccin D, Tufford A, Morshead CM. Neural stem and progenitor cells in the aged subependyma are activated by the young niche. Neurobiol Aging 2014; 35:1669-79. [PMID: 24559648 DOI: 10.1016/j.neurobiolaging.2014.01.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 01/09/2014] [Accepted: 01/23/2014] [Indexed: 11/25/2022]
Abstract
Previous studies have demonstrated an age related decline in the size of the neural stem cell (NSC) pool and a decrease in neural progenitor cell proliferation, however, the mechanisms underlying these changes are unclear. In contrast to previous reports, we report that the numbers of NSCs is unchanged in the old age subependyma and the apparent loss is because of reduced proliferative potential in the aged stem cell niche. Transplantation studies reveal that the proliferation kinetics and migratory behavior of neural precursor cells are dependent on the age of the host animal and independent of the age of the donor cells suggesting that young and old age neural precursors are not intrinsically different. Factors from the young stem cell niche rescue the numbers of NSC colonies derived from old age subependyma and enhance progenitor cell proliferation in vivo in old age mice. Finally, we report a loss of Wnt signaling in the old age stem cell niche that underlies the lack of expansion of the NSC pool after stroke.
Collapse
Affiliation(s)
- David Piccin
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Adele Tufford
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Cindi M Morshead
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
32
|
Moyanova SG, Mitreva RG, Kortenska LV, Nicoletti F, Ngomba RT. Age-dependence of sensorimotor and cerebral electroencephalographic asymmetry in rats subjected to unilateral cerebrovascular stroke. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2013; 5:13. [PMID: 24245542 PMCID: PMC4176494 DOI: 10.1186/2040-7378-5-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 11/06/2013] [Indexed: 01/24/2023]
Abstract
BACKGROUND The human population mostly affected by stroke is more than 65 years old. This study was designed to meet the recommendation that models of cerebral ischemia in aged animals are more relevant to the clinical setting than young animal models. Until now the majority of the pre-clinical studies examining age effects on stroke outcomes have used rats of old age. Considering the increasing incidence of stroke among younger than old human population, new translational approaches in animal models are needed to match the rejuvenation of stroke. A better knowledge of alterations in stroke outcomes in middle-aged rats has important preventive and management implications providing clues for future investigations on effects of various neuroprotective and neurorestorative drugs against cerebrovascular accidents that may occur before late senescence. METHODS We evaluated the impact of transient focal ischemia, induced by intracerebral unilateral infusion of endothelin-1 (Et-1) near the middle cerebral artery of conscious rats, on volume of brain damage and asymmetry in behavioral and electroencephalographic (EEG) output measures in middle-aged (11-12 month-old) rats. RESULTS We did not find any age-dependent difference in the volume of ischemic brain damage three days after Et-1 infusion. However, age was an important determinant of neurological and EEG outcomes after stroke. Middle-aged ischemic rats had more impaired somatosensory functions of the contralateral part of the body than young ischemic rats and thus, had greater left-right reflex/sensorimotor asymmetry. Interhemispheric EEG asymmetry was more evident in middle-aged than in young ischemic rats, and this could tentatively explain the behavioral asymmetry. CONCLUSIONS With a multiparametric approach, we have validated the endothelin model of ischemia in middle-aged rats. The results provide clues for future studies on mechanisms underlying plasticity after brain damage and motivate investigations of novel neuroprotective strategies against cerebrovascular accidents that may occur before late senescence.
Collapse
Affiliation(s)
| | | | | | | | - Richard T Ngomba
- I,R,C,C,S,, NEUROMED, Localita Camerelle, 86077, Pozzilli, (IS), Italy.
| |
Collapse
|
33
|
Tan Z, Turner RC, Leon RL, Li X, Hongpaisan J, Zheng W, Logsdon AF, Naser ZJ, Alkon DL, Rosen CL, Huber JD. Bryostatin improves survival and reduces ischemic brain injury in aged rats after acute ischemic stroke. Stroke 2013; 44:3490-7. [PMID: 24172582 DOI: 10.1161/strokeaha.113.002411] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND PURPOSE Bryostatin, a potent protein kinase C (PKC) activator, has demonstrated therapeutic efficacy in preclinical models of associative memory, Alzheimer disease, global ischemia, and traumatic brain injury. In this study, we tested the hypothesis that administration of bryostatin provides a therapeutic benefit in reducing brain injury and improving stroke outcome using a clinically relevant model of cerebral ischemia with tissue plasminogen activator reperfusion in aged rats. METHODS Acute cerebral ischemia was produced by reversible occlusion of the right middle cerebral artery (MCAO) in 18- to 20-month-old female Sprague-Dawley rats using an autologous blood clot with tissue plasminogen activator-mediated reperfusion. Bryostatin was administered at 6 hours post-MCAO, then at 3, 6, 9, 12, 15, and 18 days after MCAO. Functional assessment was conducted at 2, 7, 14, and 21 days after MCAO. Lesion volume and hemispheric swelling/atrophy were performed at 2, 7, and 21 days post-MCAO. Histological assessment of PKC isozymes was performed at 24 hours post-MCAO. RESULTS Bryostatin-treated rats showed improved survival post-MCAO, especially during the first 4 days. Repeated administration of bryostatin post-MCAO resulted in reduced infarct volume, hemispheric swelling/atrophy, and improved neurological function at 21 days post-MCAO. Changes in αPKC expression and εPKC expression in neurons were noted in bryostatin-treated rats at 24 hours post-MCAO. CONCLUSIONS Repeated bryostatin administration post-MCAO protected the brain from severe neurological injury post-MCAO. Bryostatin treatment improved survival rate, reduced lesion volume, salvaged tissue in infarcted hemisphere by reducing necrosis and peri-infarct astrogliosis, and improved functional outcome after MCAO.
Collapse
Affiliation(s)
- Zhenjun Tan
- From the Department of Neurosurgery, School of Medicine (Z.T., R.C.T., R.L.L., X.L., Z.J.N., C.L.R.), Blanchette Rockefeller Neuroscience Institute (J.H., W.Z., D.L.A.), and Department of Basic Pharmaceutical Science, School of Pharmacy (A.F.L., J.D.H.), West Virginia University Health Sciences Center, Morgantown, WV
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Dhungana H, Malm T, Denes A, Valonen P, Wojciechowski S, Magga J, Savchenko E, Humphreys N, Grencis R, Rothwell N, Koistinaho J. Aging aggravates ischemic stroke-induced brain damage in mice with chronic peripheral infection. Aging Cell 2013; 12:842-50. [PMID: 23725345 DOI: 10.1111/acel.12106] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2013] [Indexed: 01/13/2023] Open
Abstract
Ischemic stroke is confounded by conditions such as atherosclerosis, diabetes, and infection, all of which alter peripheral inflammatory processes with concomitant impact on stroke outcome. The majority of the stroke patients are elderly, but the impact of interactions between aging and inflammation on stroke remains unknown. We thus investigated the influence of age on the outcome of stroke in animals predisposed to systemic chronic infection. Th1-polarized chronic systemic infection was induced in 18-22 month and 4-month-old C57BL/6j mice by administration of Trichuris muris (gut parasite). One month after infection, mice underwent permanent middle cerebral artery occlusion and infarct size, brain gliosis, and brain and plasma cytokine profiles were analyzed. Chronic infection increased the infarct size in aged but not in young mice at 24 h. Aged, ischemic mice showed altered plasma and brain cytokine responses, while the lesion size correlated with plasma prestroke levels of RANTES. Moreover, the old, infected mice exhibited significantly increased neutrophil recruitment and upregulation of both plasma interleukin-17α and tumor necrosis factor-α levels. Neither age nor infection status alone or in combination altered the ischemia-induced brain microgliosis. Our results show that chronic peripheral infection in aged animals renders the brain more vulnerable to ischemic insults, possibly by increasing the invasion of neutrophils and altering the inflammation status in the blood and brain. Understanding the interactions between age and infections is crucial for developing a better therapeutic regimen for ischemic stroke and when modeling it as a disease of the elderly.
Collapse
Affiliation(s)
- Hiramani Dhungana
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Tarja Malm
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | | | - Piia Valonen
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Sara Wojciechowski
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Johanna Magga
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Ekaterina Savchenko
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Neil Humphreys
- Faculty of Life Sciences; University of Manchester; Oxford Road; Manchester; M13 9PT; UK
| | - Richard Grencis
- Faculty of Life Sciences; University of Manchester; Oxford Road; Manchester; M13 9PT; UK
| | - Nancy Rothwell
- Faculty of Life Sciences; University of Manchester; Oxford Road; Manchester; M13 9PT; UK
| | | |
Collapse
|
35
|
Buga AM, Di Napoli M, Popa-Wagner A. Preclinical models of stroke in aged animals with or without comorbidities: role of neuroinflammation. Biogerontology 2013; 14:651-62. [PMID: 24057280 DOI: 10.1007/s10522-013-9465-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/10/2013] [Indexed: 12/31/2022]
Abstract
Age is the principal nonmodifiable risk factor for stroke. Over the past 10 years, suitable models for stroke in aged rats have been established. At genetic and cellular level there are significant differences in behavioral, cytological and genomics responses to injury in old animals as compared with the young ones. Behaviorally, the aged rats have the capacity to recover after cortical infarcts albeit to a lower extent than the younger counterparts. Similarly, the increased vulnerability of the aged brain to stroke, together with a decreased interhemisphere synchrony after stroke, assessed by different experimental methods (MRI, fMRI, in vivo microscopy, EEG) leads to unfavorable recovery of physical and cognitive functions in aged people and may have a prognostic value for the recovery of stroke patients. Furthermore, in elderly, comorbidities like diabetes or arterial hypertension are associated with higher risk of stroke, increased mortality and disability, and poorer functional status and quality of life. Aging brain reacts strongly to ischemia-reperfusion injury with an early inflammatory response. The process of cellular senescence can be an important additional contributor to chronic post-stroke by creating a "primed" inflammatory environment in the brain. Overall, these pro-inflammatory reactions promote early scar formation associated with tissue fibrosis and reduce functional recovery. A better understanding of molecular factors and signaling pathways underlying the contribution of comorbidities to stroke-induced pathological sequelae, may be translated into successful treatment or prevention therapies for age-associated diseases which would improve lifespan and quality of life.
Collapse
Affiliation(s)
- A-M Buga
- Department of Functional Sciences, Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy, Craiova, Craiova, Romania
| | | | | |
Collapse
|
36
|
De Geyter D, Stoop W, Sarre S, De Keyser J, Kooijman R. Neuroprotective efficacy of subcutaneous insulin-like growth factor-I administration in normotensive and hypertensive rats with an ischemic stroke. Neuroscience 2013; 250:253-62. [PMID: 23872393 DOI: 10.1016/j.neuroscience.2013.07.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 07/01/2013] [Accepted: 07/03/2013] [Indexed: 10/26/2022]
Abstract
The aim of this study was to test the insulin-like growth factor-I (IGF-I) as a neuroprotective agent in a rat model for ischemic stroke and to compare its neuroprotective effects in conscious normotensive and spontaneously hypertensive rats. The effects of subcutaneous IGF-I injection were investigated in both rat strains using the endothelin-1 rat model for ischemic stroke. Motor-sensory functions were measured using the Neurological Deficit Score. Infarct size was assessed by Cresyl Violet staining. Subcutaneous administration of IGF-I resulted in significantly reduced infarct volumes and an increase in motor-sensory functions in normotensive rats. In these rats, IGF-I did not modulate blood flow in the striatum and had no effect on the activation of astrocytes as assessed by GFAP staining. In hypertensive rats, the protective effects of IGF-I were smaller and not always significant. Furthermore, IGF-I significantly reduced microglial activation in the cortex of hypertensive rats, but not in normotensive rats. More detailed studies are required to find out whether the reduction by IGF-I of microglial activation contributes to an impairment IGF-I treatment efficacy. Indeed, we have shown before that microglia in hypertensive rats have different properties compared to those in control rats, as they exhibit a reduced responsiveness to ischemic stroke and lipopolysaccharide.
Collapse
Affiliation(s)
- D De Geyter
- Center for Neurosciences, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussels, Belgium; Department of Pharmacology, VUB Brussel, Belgium
| | | | | | | | | |
Collapse
|
37
|
Turner RC, Dodson SC, Rosen CL, Huber JD. The science of cerebral ischemia and the quest for neuroprotection: navigating past failure to future success. J Neurosurg 2013; 118:1072-85. [PMID: 23331000 DOI: 10.3171/2012.11.jns12408] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Ischemic stroke remains a leading cause of morbidity and death for which few therapeutic options are available. The development of neuroprotective agents, a once promising field of investigation, has failed to translate from bench to bedside successfully. This work reviews the ischemic cascade, agents targeting steps within the cascade, and potential reasons for lack of translation. Additional therapeutic targets are highlighted and areas requiring further investigation are discussed. It is clear that alternative targets need to be pursued, such as the role glia play in neurological injury and recovery, particularly the interactions between neurons, astrocytes, microglia, and the vasculature. Similarly, the biphasic nature of many signaling molecules such as matrix metalloproteinases and high-mobility group box 1 protein must be further investigated to elucidate periods of detrimental versus beneficial activity.
Collapse
Affiliation(s)
- Ryan C Turner
- Department of Neurosurgery, West Virginia University, Morgantown, West Virginia 26506-9183, USA
| | | | | | | |
Collapse
|
38
|
Turner RC, Lucke-Wold B, Lucke-Wold N, Elliott AS, Logsdon AF, Rosen CL, Huber JD. Neuroprotection for ischemic stroke: moving past shortcomings and identifying promising directions. Int J Mol Sci 2013; 14:1890-1917. [PMID: 23344061 PMCID: PMC3565354 DOI: 10.3390/ijms14011890] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/04/2013] [Accepted: 01/10/2013] [Indexed: 02/05/2023] Open
Abstract
The translation of neuroprotective agents for ischemic stroke from bench-to-bedside has largely failed to produce improved treatments since the development of tissue plasminogen activator (tPA). One possible reason for lack of translation is the failure to acknowledge the greatest risk factor for stroke, age, and other common comorbidities such as hypertension, obesity, and diabetes that are associated with stroke. In this review, we highlight both mechanisms of studying these factors and results of those that have been addressed. We also discuss the potential role of other lifestyle factors associated with an increased stroke risk such as sleep fragmentation and/or deprivation. Furthermore, many proposed therapeutic agents have targeted molecular mechanisms occurring soon after the onset of ischemia despite data indicating delayed patient presentation following ischemic stroke. Modulating inflammation has been identified as a promising therapeutic avenue consistent with preliminary success of ongoing clinical trials for anti-inflammatory compounds such as minocycline. We review the role of inflammation in stroke and in particular, the role of inflammatory cell recruitment and macrophage phenotype in the inflammatory process. Emerging evidence indicates an increasing role of neuro-immune crosstalk, which has led to increased interest in identification of peripheral biomarkers indicative of neural injury. It is our hope that identification and investigation of factors influencing stroke pathophysiology may lead to improved therapeutics.
Collapse
Affiliation(s)
- Ryan C. Turner
- Department of Neurosurgery, One Medical Center Drive, West Virginia University School of Medicine, P.O. Box 9183, Morgantown, WV 26506, USA; E-Mails: (R.C.T.); (B.L.-W.); (A.S.E.)
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
| | - Brandon Lucke-Wold
- Department of Neurosurgery, One Medical Center Drive, West Virginia University School of Medicine, P.O. Box 9183, Morgantown, WV 26506, USA; E-Mails: (R.C.T.); (B.L.-W.); (A.S.E.)
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
| | - Noelle Lucke-Wold
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
- Department of Health Restoration, West Virginia University School of Nursing, Morgantown, WV 26506, USA
| | - Alisa S. Elliott
- Department of Neurosurgery, One Medical Center Drive, West Virginia University School of Medicine, P.O. Box 9183, Morgantown, WV 26506, USA; E-Mails: (R.C.T.); (B.L.-W.); (A.S.E.)
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
| | - Aric F. Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA
| | - Charles L. Rosen
- Department of Neurosurgery, One Medical Center Drive, West Virginia University School of Medicine, P.O. Box 9183, Morgantown, WV 26506, USA; E-Mails: (R.C.T.); (B.L.-W.); (A.S.E.)
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
| | - Jason D. Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA; E-Mails: (N.L.-W.); (A.F.L.); (J.D.H.)
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA
| |
Collapse
|
39
|
Liu F, McCullough LD. Interactions between age, sex, and hormones in experimental ischemic stroke. Neurochem Int 2012; 61:1255-65. [PMID: 23068990 DOI: 10.1016/j.neuint.2012.10.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 10/01/2012] [Accepted: 10/05/2012] [Indexed: 12/27/2022]
Abstract
Age, sex, and gonadal hormones have profound effects on ischemic stroke outcomes, although how these factors impact basic stroke pathophysiology remains unclear. There is a plethora of inconsistent data reported throughout the literature, primarily due to differences in the species examined, the timing and methods used to evaluate injury, the models used, and confusion regarding differences in stroke incidence as seen in clinical populations vs. effects on acute neuroprotection or neurorepair in experimental stroke models. Sex and gonadal hormone exposure have considerable independent impact on stroke outcome, but these factors also interact with each other, and the contribution of each differs throughout the lifespan. The contribution of sex and hormones to experimental stroke will be the focus of this review. Recent advances and our current understanding of age, sex, and hormone interactions in ischemic stroke with a focus on inflammation will be discussed.
Collapse
Affiliation(s)
- Fudong Liu
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | | |
Collapse
|
40
|
Leon RL, Li X, Huber JD, Rosen CL. Worsened outcome from middle cerebral artery occlusion in aged rats receiving 17β-estradiol. Endocrinology 2012; 153:3386-93. [PMID: 22581460 PMCID: PMC3380301 DOI: 10.1210/en.2011-1859] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Although estrogens are neuroprotective in young adult animal models of stroke, clinical trials demonstrate that estrogens increase the incidence and severity of stroke in aged women. We have previously shown that experimental stroke pathophysiology differs between young adult and aged rats. The aim of this study was to determine the effects of 17β-estradiol after middle cerebral artery occlusion and reperfusion in young adult and aged female rats. Focal embolic stroke was performed by middle cerebral artery occlusion with fibrin clot followed by reperfusion with i.v. human recombinant tissue plasminogen activator. Histological and functional outcomes were measured at 24 h after middle cerebral artery occlusion with fibrin clot. Aged rats treated with 17β-estradiol had significantly increased infarct volumes compared with placebo-treated aged rats. Young adult rats treated with 17β-estradiol had significantly decreased infarct volumes and improved functional outcome compared with ovariectomized young adult rats. Our results suggest that 17β-estradiol may act in an age-dependent manner in the postischemic rat brain. In young adult rats, it is neuroprotective; chronic treatment with 17β-estradiol during aging leads to worsened ischemic brain injury in aged female rats.
Collapse
Affiliation(s)
- Rachel L Leon
- West Virginia University Department of Neurosurgery, Morgantown, West Virginia 26506-9183, USA
| | | | | | | |
Collapse
|
41
|
Turner RC, Seminerio MJ, Naser ZJ, Ford JN, Martin SJ, Matsumoto RR, Rosen CL, Huber JD. Effects of aging on behavioral assessment performance: implications for clinically relevant models of neurological disease. J Neurosurg 2012; 117:629-37. [PMID: 22746378 DOI: 10.3171/2012.5.jns112224] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Despite the role of aging in development of neurological and neurodegenerative diseases, the effects of age are often disregarded in experimental design of preclinical studies. Functional assessment increases the clinical relevance of animal models of neurological disease and adds value beyond traditional histological measures. However, the relationship between age and functional impairment has not been systematically assessed through a battery of functional tests. METHODS In this study, various sensorimotor and behavioral tests were used to evaluate effects of aging on functional performance in naive animals. Sensorimotor measures included locomotor activity; Rotarod, inclined plane, and grip-strength testing; and modified Neurological Severity Score. The Morris water maze was used to examine differences in learning and memory, and the elevated plus maze and forced swim test were used to assess anxiety-like and depressive-like behaviors, respectively. RESULTS Older Sprague-Dawley rats (18-20 months) were found to perform significantly worse on the inclined plane tests, and they exhibited alterations in elevated-plus maze and forced swim test compared with young adult rats (3-4 months). Specifically, older rats exhibited reduced exploration of open arms in elevated plus maze and higher immobility time in forced swim test. Spatial acquisition and reference memory were diminished in older rats compared with those in young adult rats. CONCLUSIONS This study demonstrates clear differences between naive young adult and older animals, which may have implications in functional assessment for preclinical models of neurological disease.
Collapse
Affiliation(s)
- Ryan C Turner
- Department of Neurosurgery, West Virginia University, School of Medicine, Morgantown, West Virginia 26506-9183, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
The transcriptome of cerebral ischemia. Brain Res Bull 2012; 88:313-9. [PMID: 22381515 DOI: 10.1016/j.brainresbull.2012.02.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 07/20/2011] [Accepted: 02/13/2012] [Indexed: 01/26/2023]
Abstract
The molecular causality and response to stroke is complex. Yet, much of the literature examining the molecular response to stroke has focused on targeted pathways that have been well-characterized. Consequently, our understanding of stroke pathophysiology has made little progress by way of clinical therapeutics since tissue plasminogen activator was approved for treatment nearly a decade ago. The lack of clinical translation is in part due to neuron-focused studies, preclinical models of cerebral ischemia and the paradoxical nature of neuro-inflammation. With the evolution of the Stroke Therapy Academic Industry Roundtable criteria streamlining research efforts and broad availability of genomic technologies, the ability to decipher the molecular fingerprint of ischemic stroke is on the horizon. This review highlights preclinical microarray findings of the ischemic brain, discusses the transcriptome of cerebral preconditioning and emphasizes the importance of further characterizing the role of the neurovascular unit and peripheral white blood cells in mediating stroke damage and repair within the penumbra.
Collapse
|
43
|
Di Napoli M, Shah IM. Neuroinflammation and cerebrovascular disease in old age: a translational medicine perspective. J Aging Res 2011; 2011:857484. [PMID: 22132330 PMCID: PMC3205617 DOI: 10.4061/2011/857484] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 08/10/2011] [Indexed: 11/20/2022] Open
Abstract
The incidence of cerebrovascular disease is highest in the elderly population. However, the pathophysiological mechanisms of brain response to cerebral ischemia in old age are currently poorly understood. Ischemic changes in the commonly used young animal stroke models do not reflect the molecular changes associated with the aged brain. Neuroinflammation and oxidative stress are important pathogenic processes occurring during the acute phase of cerebral ischemia. Free radical generation is also implicated in the aging process, and the combination of these effects in elderly stroke patients could explain the higher risk of morbidity and mortality. A better understanding of stroke pathophysiology in the elderly patient would assist in the development of new therapeutic strategies for this vulnerable age group. With the increasing use of reperfusion therapies, inflammatory pathways and oxidative stress remain attractive therapeutic targets for the development of adjuvant neuroprotective agents. This paper will discuss these molecular aspects of acute stroke and senescence from a bench-to-bedside research perspective.
Collapse
Affiliation(s)
- Mario Di Napoli
- Neurological Service, San Camillo de'Lellis General Hospital, 02100 Rieti, Italy
| | | |
Collapse
|
44
|
Sieber MW, Claus RA, Witte OW, Frahm C. Attenuated inflammatory response in aged mice brains following stroke. PLoS One 2011; 6:e26288. [PMID: 22028848 PMCID: PMC3196544 DOI: 10.1371/journal.pone.0026288] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 09/23/2011] [Indexed: 12/18/2022] Open
Abstract
Background Increased age is a major risk factor for stroke incidence, post-ischemic mortality, and severe and long-term disability. Stroke outcome is considerably influenced by post-ischemic mechanisms. We hypothesized that the inflammatory response following an ischemic injury is altered in aged organisms. Methods and Results To that end, we analyzed the expression pattern of pro-inflammatory cytokines (TNF, IL-1α, IL-1β, IL-6), anti-inflammatory cytokines (IL-10, TGFβ1), and chemokines (Mip-1α, MCP-1, RANTES) of adult (2 months) and aged (24 months) mice brains at different reperfusion times (6 h, 12 h, 24 h, 2 d, 7 d) following transient occlusion of the middle cerebral artery. The infarct size was assessed to monitor possible consequences of an altered inflammatory response in aged mice. Our data revealed an increased neuro-inflammation with age. Above all, we found profound age-related alterations in the reaction to stroke. The response of pro-inflammatory cytokines (TNF, and IL-1β) and the level of chemokines (Mip-1α, and MCP-1) were strongly diminished in the aged post-ischemic brain tissue. IL-6 showed the strongest age-dependent decrease in its post-ischemic expression profile. Anti-inflammatory cytokines (TGFβ1, and IL-10) revealed no significant age dependency after ischemia. Aged mice brains tend to develop smaller infarcts. Conclusion The attenuated inflammatory response to stroke in aged animals may contribute to their smaller infarcts. The results presented here highlight the importance of using aged animals to investigate age-associated diseases like stroke, and should be considered as a major prerequisite in the development of age-adjusted therapeutic interventions.
Collapse
Affiliation(s)
- Matthias W. Sieber
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Thuringia, Germany
- Centre for Sepsis Control and Care, Jena University Hospital, Jena, Thuringia, Germany
| | - Ralf A. Claus
- Centre for Sepsis Control and Care, Jena University Hospital, Jena, Thuringia, Germany
| | - Otto W. Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Thuringia, Germany
- * E-mail:
| | - Christiane Frahm
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Thuringia, Germany
| |
Collapse
|
45
|
Mora-Lee S, Sirerol-Piquer MS, Gutiérrez-Pérez M, López T, Casado-Nieto M, Jauquicoam C, Abizanda G, Romaguera-Ros M, Gomez-Pinedo U, Prósper F, García-Verdugo JM. Histological and ultrastructural comparison of cauterization and thrombosis stroke models in immune-deficient mice. J Inflamm (Lond) 2011; 8:28. [PMID: 22008614 PMCID: PMC3221623 DOI: 10.1186/1476-9255-8-28] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 10/18/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stroke models are essential tools in experimental stroke. Although several models of stroke have been developed in a variety of animals, with the development of transgenic mice there is the need to develop a reliable and reproducible stroke model in mice, which mimics as close as possible human stroke. METHODS BALB/Ca-RAG2-/-γc-/- mice were subjected to cauterization or thrombosis stroke model and sacrificed at different time points (48hr, 1wk, 2wk and 4wk) after stroke. Mice received BrdU to estimate activation of cell proliferation in the SVZ. Brains were processed for immunohistochemical and EM. RESULTS In both stroke models, after inflammation the same glial scar formation process and damage evolution takes place. After stroke, necrotic tissue is progressively removed, and healthy tissue is preserved from injury through the glial scar formation. Cauterization stroke model produced unspecific damage, was less efficient and the infarct was less homogeneous compared to thrombosis infarct. Finally, thrombosis stroke model produces activation of SVZ proliferation. CONCLUSIONS Our results provide an exhaustive analysis of the histopathological changes (inflammation, necrosis, tissue remodeling, scarring...) that occur after stroke in the ischemic boundary zone, which are of key importance for the final stroke outcome. This analysis would allow evaluating how different therapies would affect wound and regeneration. Moreover, this stroke model in RAG 2-/- γC -/- allows cell transplant from different species, even human, to be analyzed.
Collapse
Affiliation(s)
- Silvia Mora-Lee
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | | - María Gutiérrez-Pérez
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Tania López
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Mayte Casado-Nieto
- Department of Comparative Neurobiology. Cavanilles Institute. CIPF. CIBERNED, Valencia, Spain
| | - Carlos Jauquicoam
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Gloria Abizanda
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Miriam Romaguera-Ros
- Department of Comparative Neurobiology. Cavanilles Institute. CIPF. CIBERNED, Valencia, Spain
| | | | - Felipe Prósper
- Hematology and Cell Therapy Area, Clinica Universidad de Navarra and Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | |
Collapse
|
46
|
Quantitative MRI reveals the elderly ischemic brain is susceptible to increased early blood-brain barrier permeability following tissue plasminogen activator related to claudin 5 and occludin disassembly. J Cereb Blood Flow Metab 2011; 31:1874-85. [PMID: 21610723 PMCID: PMC3185885 DOI: 10.1038/jcbfm.2011.79] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Great uncertainty exists as to whether aging enhances the detrimental effects of tissue plasminogen activator (tPA) on vascular integrity of the ischemic brain. We hypothesized that tPA treatment would augment ischemic injury by causing increased blood-brain barrier (BBB) breakdown as determined by quantitative serial T(1) and T(2) magnetic resonance imaging (MRI), and the transfer constant for gadolinium-diethylenetriamine penta-acetic acid (Gd-DTPA) from blood to brain in aged (18 to 20 months) compared with young (3 to 4 months) Wistar rats after middle cerebral artery occlusion, mediated through the acute disassembly of claudin 5 and occludin. Increased T(2) values over the first hour of postreperfusion were independently augmented following treatment with tPA (P<0.001) and aging (P<0.01), supporting a synergistic effect of tPA on the aged ischemic brain. Blood-brain barrier permeability for Gd-DTPA (K(Gd)) was substantial following reperfusion in all animal groups and was exacerbated by tPA treatment in the elderly rat (P<0.001). The frequency of hematoma formation was proportionately increased in the elderly ischemic brain (P<0.05). Both tPA and age independently increased claudin 5 and occludin phosphorylation during ischemia. Early BBB permeability detected by quantitative MRI following ischemic stroke is enhanced by increased age and tPA and is related to claudin 5 and occludin phosphorylation.
Collapse
|
47
|
Vangilder RL, Rosen CL, Barr TL, Huber JD. Targeting the neurovascular unit for treatment of neurological disorders. Pharmacol Ther 2011; 130:239-47. [PMID: 21172386 PMCID: PMC3092634 DOI: 10.1016/j.pharmthera.2010.12.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 11/22/2010] [Indexed: 12/17/2022]
Abstract
Drug discovery for CNS disorders has been restricted by the inability for therapeutic agents to cross the blood-brain barrier (BBB). Moreover, current drugs aim to correct neuron cell signaling, thereby neglecting pathophysiological changes affecting other cell types of the neurovascular unit (NVU). Components of the NVU (pericytes, microglia, astrocytes, and neurons, and basal lamina) act as an intricate network to maintain the neuronal homeostatic microenvironment. Consequently, disruptions to this intricate cell network lead to neuron malfunction and symptoms characteristic of CNS diseases. A lack of understanding in NVU signaling cascades may explain why current treatments for CNS diseases are not curative. Current therapies treat symptoms by maintaining neuron function. Refocusing drug discovery to sustain NVU function may provide a better method of treatment by promoting neuron survival. In this review, we will examine current therapeutics for common CNS diseases, describe the importance of the NVU in cerebral homeostasis and discuss new possible drug targets and technologies that aim to improve treatment and drug delivery to the diseased brain.
Collapse
Affiliation(s)
- Reyna L Vangilder
- Department of Health Restoration, West Virginia University School of Nursing, Morgantown WV, USA
| | | | | | | |
Collapse
|
48
|
Li N, Kong X, Ye R, Yang Q, Han J, Xiong L. Age-Related Differences in Experimental Stroke: Possible Involvement of Mitochondrial Dysfunction and Oxidative Damage. Rejuvenation Res 2011; 14:261-73. [PMID: 21466386 DOI: 10.1089/rej.2010.1115] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Nanlin Li
- Department of Vascular and Endocrine Surgery, Fourth Military Medical University, Xi'an, China
| | - Xiangwei Kong
- College of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Ruidong Ye
- Department of Neurology, Fourth Military Medical University, Xi'an, China
| | - Qianzi Yang
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Junliang Han
- Department of Neurology, Fourth Military Medical University, Xi'an, China
| | - Lize Xiong
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
49
|
Leon RL, Huber JD, Rosen CL. Potential age-dependent effects of estrogen on neural injury. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2450-60. [PMID: 21641373 PMCID: PMC3124359 DOI: 10.1016/j.ajpath.2011.01.057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 12/28/2010] [Accepted: 01/07/2011] [Indexed: 12/27/2022]
Abstract
In 2000, approximately 10 million women were receiving hormone replacement therapy (HRT) for alleviation of menopausal symptoms. A number of prior animal studies suggested that HRT may be neuroprotective and cardioprotective. Then, in 2003, reports from the Women's Health Initiative (WHI) indicated that long-term estrogen/progestin supplementation led to increased incidence of stroke. A second branch of the WHI in women with prior hysterectomy found an even stronger correlation between estrogen supplementation alone and stroke incidence. Follow-up analyses of the data, as well as data from other smaller clinical trials, have also demonstrated increased stroke severity in women receiving HRT or estrogen alone. This review examines the studies indicating that estrogen is neuroprotectant in animal models and explores potential reasons why this may not be true in postmenopausal women. Specifically, age-related differences in estrogen receptors and estrogenic actions in the brain are discussed, with the conclusion that animal models of disease must closely mimic human disease to produce clinically relevant results.
Collapse
Affiliation(s)
- Rachel L. Leon
- Department of Neurosurgery, West Virginia University, Morgantown, West Virginia
| | - Jason D. Huber
- Department of Basic Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia
| | - Charles L. Rosen
- Department of Neurosurgery, West Virginia University, Morgantown, West Virginia
| |
Collapse
|
50
|
Popa-Wagner A, Buga AM, Kokaia Z. Perturbed cellular response to brain injury during aging. Ageing Res Rev 2011; 10:71-9. [PMID: 19900590 DOI: 10.1016/j.arr.2009.10.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Accepted: 10/28/2009] [Indexed: 12/22/2022]
Abstract
Old age is associated with an enhanced susceptibility to stroke and poor recovery from brain injury, but the cellular processes underlying these phenomena are only partly understood. Therefore, studying the basic mechanisms underlying structural and functional recovery after brain injury in aged subjects is of considerable clinical interest. Behavioral and cytological analyses of rodents that have undergone experimental injury show that: (a) behaviorally, aged rodents are more severely impaired by ischemia than are young animals, and older rodents also show diminished functional recovery; (b) compared to young animals, aged animals develop a larger infarct area, as well as a necrotic zone characterized by a higher rate of cellular degeneration and a larger number of apoptotic cells; (c) both astrocytes and macrophages are activated strongly and early following stroke in aged rodents; (d) in older animals, the premature, intense cytoproliferative activity following brain injury leads to the precipitous formation of growth-inhibiting scar tissue, a phenomenon amplified by the persistent expression of neurotoxic factors; (e) though the timing is altered, the regenerative capability of the brain is largely preserved in rats, at least into early old age. Whether endogenous neurogenesis contributes to spontaneous recovery after stroke has not yet been established. If neurogenesis from endogenous neuronal stem cells is to be used therapeutically, an individual approach will be required to assess the possible extent of neurogenic response as well as the possibilities to alter this response for functional improvement or prevention of further loss of brain function.
Collapse
|