1
|
Vita SM, Cruise SC, Gilpin NW, Molina PE. HISTOLOGICAL COMPARISON OF REPEATED MILD WEIGHT DROP AND LATERAL FLUID PERCUSSION INJURY MODELS OF TRAUMATIC BRAIN INJURY IN FEMALE AND MALE RATS. Shock 2024; 62:398-409. [PMID: 38813916 DOI: 10.1097/shk.0000000000002395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
ABSTRACT In preclinical traumatic brain injury (TBI) research, the animal model should be selected based on the research question and outcome measures of interest. Direct side-by-side comparisons of different injury models are essential for informing such decisions. Here, we used immunohistochemistry to compare the outcomes from two common models of TBI, lateral fluid percussion (LFP) and repeated mild weight drop (rmWD) in adult female and male Wistar rats. Specifically, we measured the effects of LFP and rmWD on markers of cerebrovascular and tight junction disruption, neuroinflammation, mature neurons, and perineuronal nets in the cortical site of injury, cortex adjacent to injury, dentate gyrus, and the CA 2/3 area of the hippocampus. Animals were randomized into the LFP or rmWD group. On day 1, the LFP group received a craniotomy, and on day 4, injury (or sham procedure; randomly assigned). The rmWD animals underwent either injury or isoflurane only (randomly assigned) on each of those 4 days. Seven days after injury, brains were harvested for analysis. Overall, our observations revealed that the most significant disruptions were evident in response to LFP, followed by craniotomy only, whereas rmWD animals showed the least residual changes compared with isoflurane-only controls, supporting consideration of rmWD as a mild injury. LFP led to longer-lasting disruptions, perhaps more representative of moderate TBI. We also report that craniotomy and LFP produced greater disruptions in females relative to males. These findings will assist the field in the selection of animal models based on target severity of postinjury outcomes and support the inclusion of both sexes and appropriate control groups.
Collapse
Affiliation(s)
| | - Shealan C Cruise
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | | | | |
Collapse
|
2
|
Vita SM, Cruise SC, Gilpin NW, Molina PE. Histological comparison of repeated mild weight drop and lateral fluid percussion injury models of traumatic brain injury (TBI) in female and male rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578177. [PMID: 38352449 PMCID: PMC10862833 DOI: 10.1101/2024.01.31.578177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Traumatic brain injury (TBI) heterogeneity has led to the development of several preclinical models, each modeling a distinct subset of outcomes. Selection of an injury model should be guided by the research question and the specific outcome measures of interest. Consequently, there is a need for conducting direct comparisons of different TBI models. Here, we used immunohistochemistry to directly compare the outcomes from two common models, lateral fluid percussion (LFP) and repeat mild weight drop (rmWD), on neuropathology in adult female and male Wistar rats. Specifically, we used immunohistochemistry to measure the effects of LFP and rmWD on cerebrovascular and tight junction disruption, inflammatory markers, mature neurons and perineuronal nets in the cortical site of injury, cortex adjacent to injury, dentate gyrus, and the CA2/3 area of the hippocampus. Animals were randomized into either LFP or rmWD groups. The LFP group received a craniotomy prior to LFP (or corresponding sham procedure) three days later, while rmWD animals underwent either weight drop or sham (isoflurane only) on each of those four days. After a recovery period of 7 days, animals were euthanized, and brains were harvested for analysis of RECA-1, claudin-5, GFAP, Iba-1, CD-68, NeuN, and wisteria floribunda lectin. Overall, our observations revealed that the most significant disruptions were evident in response to LFP, followed by craniotomy-only, while rmWD animals showed the least residual changes compared to isoflurane-only controls. These findings support consideration of rmWD as a mild, transient injury. LFP leads to longer-lasting disruptions that are more closely associated with a moderate TBI. We further show that both craniotomy and LFP produced greater disruptions in females relative to males at 7 days post-injury. These findings support the inclusion of a time-matched experimentally-naïve or anesthesia-only control group in preclinical TBI research to enhance the validity of data interpretation and conclusions.
Collapse
|
3
|
Chao CM, Hsu CC, Huang CC, Wang CH, Lin MT, Chang CP, Lin HJ, Chio CC. Selective brain cooling achieves peripheral organs protection in hemorrhagic shock resuscitation via preserving the integrity of the brain-gut axis. Int J Med Sci 2021; 18:2920-2929. [PMID: 34220319 PMCID: PMC8241763 DOI: 10.7150/ijms.61191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/23/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Although whole-body cooling has been reported to improve the ischemic/reperfusion injury in hemorrhagic shock (HS) resuscitation, it is limited by its adverse reactions following therapeutic hypothermia. HS affects the experimental and clinical bowel disorders via activation of the brain-gut axis. It is unknown whether selective brain cooling achieves beneficial effects in HS resuscitation via preserving the integrity of the brain-gut axis. Methods: Male Sprague-Dawley rats were bled to hypovolemic HS and resuscitated with blood transfusion followed by retrograde jugular vein flush (RJVF) with 4 °C or 36 °C normal saline. The mean arterial blood pressure, cerebral blood flow, and brain and core temperature were measured. The integrity of intestinal tight junction proteins and permeability, blood pro-inflammatory cytokines, and multiple organs damage score were determined. Results: Following blood transfusion resuscitation, HS rats displayed gut barrier disruption, increased blood levels of pro-inflammatory cytokines, and peripheral vital organ injuries. Intrajugular-based infusion cooled the brain robustly with a minimal effect on body temperature. This brain cooling significantly reduced the HS resuscitation-induced gut disruption, systemic inflammation, and peripheral vital organ injuries in rats. Conclusion: Resuscitation with selective brain cooling achieves peripheral vital organs protection in hemorrhagic shock resuscitation via preserving the integrity of the brain-gut axis.
Collapse
Affiliation(s)
- Chien-Ming Chao
- Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan.,Department of Nursing, Min-Hwei College of Health Care Management, Tainan, Taiwan
| | - Chien-Chin Hsu
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chien-Cheng Huang
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan.,Department of Senior Services, Southern Taiwan University of Science and Technology, Tainan, Taiwan.,Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chung-Han Wang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Hung-Jung Lin
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan.,Department of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chung-Ching Chio
- Division of Neurosurgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
4
|
Kalisvaart ACJ, Prokop BJ, Colbourne F. Hypothermia: Impact on plasticity following brain injury. Brain Circ 2019; 5:169-178. [PMID: 31950092 PMCID: PMC6950515 DOI: 10.4103/bc.bc_21_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
Therapeutic hypothermia (TH) is a potent neuroprotectant against multiple forms of brain injury, but in some cases, prolonged cooling is needed. Such cooling protocols raise the risk that TH will directly or indirectly impact neuroplasticity, such as after global and focal cerebral ischemia or traumatic brain injury. TH, depending on the depth and duration, has the potential to broadly affect brain plasticity, especially given the spatial, temporal, and mechanistic overlap with the injury processes that cooling is used to treat. Here, we review the current experimental and clinical evidence to evaluate whether application of TH has any adverse or positive effects on postinjury plasticity. The limited available data suggest that mild TH does not appear to have any deleterious effect on neuroplasticity; however, we emphasize the need for additional high-quality preclinical and clinical work in this area.
Collapse
|
5
|
Kurisu K, Kim JY, You J, Yenari MA. Therapeutic Hypothermia and Neuroprotection in Acute Neurological Disease. Curr Med Chem 2019; 26:5430-5455. [PMID: 31057103 PMCID: PMC6913523 DOI: 10.2174/0929867326666190506124836] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/24/2018] [Accepted: 04/11/2019] [Indexed: 01/07/2023]
Abstract
Therapeutic hypothermia has consistently been shown to be a robust neuroprotectant in many labs studying different models of neurological disease. Although this therapy has shown great promise, there are still challenges at the clinical level that limit the ability to apply this routinely to each pathological condition. In order to overcome issues involved in hypothermia therapy, understanding of this attractive therapy is needed. We review methodological concerns surrounding therapeutic hypothermia, introduce the current status of therapeutic cooling in various acute brain insults, and review the literature surrounding the many underlying molecular mechanisms of hypothermic neuroprotection. Because recent work has shown that body temperature can be safely lowered using pharmacological approaches, this method may be an especially attractive option for many clinical applications. Since hypothermia can affect multiple aspects of brain pathophysiology, therapeutic hypothermia could also be considered a neuroprotection model in basic research, which would be used to identify potential therapeutic targets. We discuss how research in this area carries the potential to improve outcome from various acute neurological disorders.
Collapse
Affiliation(s)
- Kota Kurisu
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, California 94121, USA
| | - Jong Youl Kim
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, California 94121, USA
- Departments of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Jesung You
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, California 94121, USA
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Midori A. Yenari
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, California 94121, USA
| |
Collapse
|
6
|
Liska MG, Crowley MG, Tuazon JP, Borlongan CV. Neuroprotective and neuroregenerative potential of pharmacologically-induced hypothermia with D-alanine D-leucine enkephalin in brain injury. Neural Regen Res 2018; 13:2029-2037. [PMID: 30323116 PMCID: PMC6199924 DOI: 10.4103/1673-5374.241427] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 10/27/2017] [Indexed: 12/21/2022] Open
Abstract
Neurovascular disorders, such as traumatic brain injury and stroke, persist as leading causes of death and disability - thus, the search for novel therapeutic approaches for these disorders continues. Many hurdles have hindered the translation of effective therapies for traumatic brain injury and stroke primarily because of the inherent complexity of neuropathologies and an inability of current treatment approaches to adapt to the unique cell death pathways that accompany the disorder symptoms. Indeed, developing potent treatments for brain injury that incorporate dynamic and multiple disorder-engaging therapeutic targets are likely to produce more effective outcomes than traditional drugs. The therapeutic use of hypothermia presents a promising option which may fit these criteria. While regulated temperature reduction has displayed great promise in preclinical studies of brain injury, clinical trials have been far less consistent and associated with adverse effects, especially when hypothermia is pursued via systemic cooling. Accordingly, devising better methods of inducing hypothermia may facilitate the entry of this treatment modality into the clinic. The use of the delta opioid peptide D-alanine D-leucine enkephalin (DADLE) to pharmacologically induce temperature reduction may offer a potent alternative, as DADLE displays both the ability to cause temperature reduction and to confer a broad profile of other neuroprotective and neuroregenerative processes. This review explores the prospect of DADLE-mediated hypothermia to treat neurovascular brain injuries, emphasizing the translational steps necessary for its clinical translation.
Collapse
Affiliation(s)
- M. Grant Liska
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Marci G. Crowley
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Julian P. Tuazon
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| |
Collapse
|
7
|
Fan YY, Nan F, Guo BL, Liao Y, Zhang MS, Guo J, Niu BL, Liang YQ, Yang CH, Zhang Y, Zhang XP, Pang XF. Effects of long-term rapamycin treatment on glial scar formation after cryogenic traumatic brain injury in mice. Neurosci Lett 2018; 678:68-75. [DOI: 10.1016/j.neulet.2018.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 04/17/2018] [Accepted: 05/01/2018] [Indexed: 01/11/2023]
|
8
|
Xiong Y, Mahmood A, Chopp M. Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin J Traumatol 2018; 21:137-151. [PMID: 29764704 PMCID: PMC6034172 DOI: 10.1016/j.cjtee.2018.02.003] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) remains a major cause of death and disability worldwide. Increasing evidence indicates that TBI is an important risk factor for neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and chronic traumatic encephalopathy. Despite improved supportive and rehabilitative care of TBI patients, unfortunately, all late phase clinical trials in TBI have yet to yield a safe and effective neuroprotective treatment. The disappointing clinical trials may be attributed to variability in treatment approaches and heterogeneity of the population of TBI patients as well as a race against time to prevent or reduce inexorable cell death. TBI is not just an acute event but a chronic disease. Among many mechanisms involved in secondary injury after TBI, emerging preclinical studies indicate that posttraumatic prolonged and progressive neuroinflammation is associated with neurodegeneration which may be treatable long after the initiating brain injury. This review provides an overview of recent understanding of neuroinflammation in TBI and preclinical cell-based therapies that target neuroinflammation and promote functional recovery after TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA.
| | - Asim Mahmood
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA; Department of Physics, Oakland University, Rochester, MI, 48309, USA
| |
Collapse
|
9
|
Stewart EM, Wu Z, Huang XF, Kapsa RMI, Wallace GG. Use of conducting polymers to facilitate neurite branching in schizophrenia-related neuronal development. Biomater Sci 2018; 4:1244-51. [PMID: 27376413 DOI: 10.1039/c6bm00212a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Schizophrenia (SCZ) is a debilitating mental disorder which results in high healthcare and loss of productivity costs to society. This disease remains poorly understood, however there is increasing evidence suggesting a role for oxidative damage in the disease etiology. We aimed to examine the effect of the conducting polymer polypyrrole on the growth and morphology of both wildtype and neuregulin-1 knock out (NRG-1 +/-) explant cells. Polypyrrole is an organic conducting polymer known to be cytocompatible and capable of acting as a platform for effective stimulation of neurons. Here we demonstrate for the first time the ability of this material to mediate processes occurring in disease affected neurons: schizophrenic model cortical neurons. Prefrontal cortical cells were grown on conducting polymer scaffolds of specific composition and showed significantly increased neurite branching and outgrowth length on the polymers compared to controls. Concurrently, a more significant enhancement was seen in both parameters in the NRG-1 +/- model cells. This finding implies that conducting polymers such as polypyrrole may be utilised to overcome neuro-functional deficits associated with neurological disease in humans.
Collapse
Affiliation(s)
- Elise M Stewart
- ARC Centre of Excellence for Electromaterials Science, University of Wollongong, NSW, Australia.
| | - Zhixiang Wu
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia
| | - Xu Feng Huang
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia
| | - Robert M I Kapsa
- ARC Centre of Excellence for Electromaterials Science, University of Wollongong, NSW, Australia.
| | - Gordon G Wallace
- ARC Centre of Excellence for Electromaterials Science, University of Wollongong, NSW, Australia.
| |
Collapse
|
10
|
Klahr AC, Fagan K, Aziz JR, John R, Colbourne F. Mild Contralesional Hypothermia Reduces Use of the Unimpaired Forelimb in a Skilled Reaching Task After Motor Cortex Injury in Rats. Ther Hypothermia Temp Manag 2018; 8:90-98. [PMID: 29298129 DOI: 10.1089/ther.2017.0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Therapeutic hypothermia (TH) mitigates neuronal injury in models of ischemic stroke. Although this therapy is meant for injured tissue, most protocols cool the whole body, including the contralesional hemisphere. Neuroplasticity responses within this hemisphere can affect functional outcome. Thus, cooling the contralesional hemisphere serves no clear neuroprotective function and may instead be detrimental. In this study, we cooled the contralesional hemisphere to determine whether this harms behavioral recovery after cortical injury in rats. All rats were trained on skilled reaching and walking tasks. Rats then received a motor cortex insult contralateral to their dominant paw after which they were randomly assigned to focal contralesional TH (∼33°C) for 1-48, 1-97, or 48-96 hours postinjury, or to a normothermic control group. Contralesional cooling did not impact lesion volume (p = 0.371) and had minimal impact on neurological outcome of the impaired limb. However, rats cooled early were significantly less likely to shift paw preference to the unimpaired paw (p ≤ 0.043), suggesting that cooling reduced learned nonuse. In a second experiment, we tested whether cooling impaired learning of the skilled reaching task in naive rats. Localized TH applied to the hemisphere contralateral or ipsilateral to the preferred paw did not impair learning (p ≥ 0.677) or dendritic branching/length in the motor cortex (p ≥ 0.105). In conclusion, localized TH did not impair learning or plasticity in the absence of neural injury, but contralesional TH may reduce unwanted shifts in limb preference after stroke.
Collapse
Affiliation(s)
- Ana C Klahr
- 1 Neuroscience and Mental Health Institute, University of Alberta, Edmonton , Canada
| | - Kelly Fagan
- 2 Department of Psychology, MacEwan University , Edmonton, Canada
| | - Jasmine R Aziz
- 3 Department of Psychology, University of Alberta, Edmonton, Canada
| | - Roseleen John
- 1 Neuroscience and Mental Health Institute, University of Alberta, Edmonton , Canada
| | - Frederick Colbourne
- 1 Neuroscience and Mental Health Institute, University of Alberta, Edmonton , Canada .,3 Department of Psychology, University of Alberta, Edmonton, Canada
| |
Collapse
|
11
|
Shido O, Matsuzaki K, Katakura M. Neurogenesis in the thermoregulatory system. HANDBOOK OF CLINICAL NEUROLOGY 2018; 156:457-463. [PMID: 30454607 DOI: 10.1016/b978-0-444-63912-7.00028-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In response to various internal and external stimuli, neuronal progenitor cells in the hypothalamic area proliferate and differentiate to functionally working neurons even in adult animals. This is the case in the thermoregulatory system, especially in the process of heat acclimation. The heat acclimation process presents two different patterns, namely short-term and long-term heat acclimation. In rats, long-term heat acclimation is attained by exposing subjects to constant heat for more than 4 weeks, while short-term heat acclimation is established within several days of heat exposure. Heat exposure for more than 6 days facilitates cell proliferation in the ependymal layer of the third ventricle. The newborn cells then migrate into the hypothalamic parenchyma. After 33 days of heat exposure, the newborn cells abruptly differentiate to mature neurons. A part of the newborn cells are incorporated in a neuronal circuit in the hypothalamus. However, only 6 days of heat exposure hardly promote neuronal differentiation. An administration of mitosis inhibitor interferes with cell proliferation in the hypothalamic area and attenuates heat acclimation-induced improvement of heat tolerance. Long-term, but not short-term, heat acclimation may be established by generating new functional neurons in the hypothalamic area, which is where an important part of the thermoregulatory circuitry exists in rats.
Collapse
Affiliation(s)
- Osamu Shido
- Department of Environmental Physiology, School of Medicine, Shimane University, Izumo, Japan.
| | - Kentaro Matsuzaki
- Department of Environmental Physiology, School of Medicine, Shimane University, Izumo, Japan
| | - Masanori Katakura
- Department of Nutritional Physiology, Faculty of Pharmaceutical Sciences, Josai University, Saitama, Japan
| |
Collapse
|
12
|
Kurisu K, Yenari MA. Therapeutic hypothermia for ischemic stroke; pathophysiology and future promise. Neuropharmacology 2017; 134:302-309. [PMID: 28830757 DOI: 10.1016/j.neuropharm.2017.08.025] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/12/2017] [Accepted: 08/17/2017] [Indexed: 01/08/2023]
Abstract
Therapeutic hypothermia, or cooling of the body or brain for the purposes of preserving organ viability, is one of the most robust neuroprotectants at both the preclinical and clinical levels. Although therapeutic hypothermia has been shown to improve outcome from related clinical conditions, the significance in ischemic stroke is still under investigation. Numerous pre-clinical studies of therapeutic hypothermia has suggested optimal cooling conditions, such as depth, duration, and temporal therapeutic window for effective neuroprotection. Several studies have also explored mechanisms underlying the mechanisms of neuroprotection by therapeutic hypothermia. As such, it appears that cooling affects multiple aspects of brain pathophysiology, and regulates almost every pathway involved in the evolution of ischemic stroke. This multifaceted mechanism is thought to contribute to its strong neuroprotective effect. In order to carry out this therapy in optimal clinical settings, methodological and pathophysiological understanding is crucial. However, more investigation is still needed to better understand the underlying mechanisms of this intervention, and to overcome clinical barriers which seem to preclude the routine use therapeutic hypothermia in stroke. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Kota Kurisu
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Midori A Yenari
- Department of Neurology, University of California, San Francisco and Veterans Affairs Medical Center, San Francisco, CA 94121, USA.
| |
Collapse
|
13
|
Singh DP, Barani Lonbani Z, Woodruff MA, Parker TJ, Steck R, Peake JM. Effects of Topical Icing on Inflammation, Angiogenesis, Revascularization, and Myofiber Regeneration in Skeletal Muscle Following Contusion Injury. Front Physiol 2017; 8:93. [PMID: 28326040 PMCID: PMC5339266 DOI: 10.3389/fphys.2017.00093] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/06/2017] [Indexed: 01/01/2023] Open
Abstract
Contusion injuries in skeletal muscle commonly occur in contact sport and vehicular and industrial workplace accidents. Icing has traditionally been used to treat such injuries under the premise that it alleviates pain, reduces tissue metabolism, and modifies vascular responses to decrease swelling. Previous research has examined the effects of icing on inflammation and microcirculatory dynamics following muscle injury. However, whether icing influences angiogenesis, collateral vessel growth, or myofiber regeneration remains unknown. We compared the effects of icing vs. a sham treatment on the presence of neutrophils and macrophages; expression of CD34, von Willebrands factor (vWF), vascular endothelial growth factor (VEGF), and nestin; vessel volume; capillary density; and myofiber regeneration in skeletal after muscle contusion injury in rats. Muscle tissue was collected 1, 3, 7, and 28 d after injury. Compared with uninjured rats, muscles in rats that sustained the contusion injury exhibited major necrosis, inflammation, and increased expression of CD34, vWF, VEGF, and nestin. Compared with the sham treatment, icing attenuated and/or delayed neutrophil and macrophage infiltration; the expression of vWF, VEGF, and nestin; and the change in vessel volume within muscle in the first 7 d after injury (P < 0.05). By contrast, icing did not influence capillary density in muscle 28 d after injury (P = 0.59). The percentage of immature myofibers relative to the total number of fibers was greater in the icing group than in the sham group 28 d after injury (P = 0.026), but myofiber cross-sectional area did not differ between groups after 7 d (P = 0.35) and 28 d (P = 0.30). In conclusion, although icing disrupted inflammation and some aspects of angiogenesis/revascularization, these effects did not result in substantial differences in capillary density or muscle growth.
Collapse
Affiliation(s)
- Daniel P Singh
- Tissue Repair and Regeneration Group, Institute of Health and Biomedical Innovation, Queensland University of Technology Brisbane, QLD, Australia
| | - Zohreh Barani Lonbani
- Tissue Repair and Regeneration Group, Institute of Health and Biomedical Innovation, Queensland University of Technology Brisbane, QLD, Australia
| | - Maria A Woodruff
- Biofabrication and Tissue Morphology Group, Institute of Health and Biomedical Innovation, Queensland University of Technology Brisbane, QLD, Australia
| | - Tony J Parker
- Tissue Repair and Regeneration Group, Institute of Health and Biomedical Innovation, Queensland University of TechnologyBrisbane, QLD, Australia; School of Biomedical Sciences, Queensland University of TechnologyBrisbane, QLD, Australia
| | - Roland Steck
- Medical Engineering Research Facility, Queensland University of Technology Brisbane, QLD, Australia
| | - Jonathan M Peake
- Tissue Repair and Regeneration Group, Institute of Health and Biomedical Innovation, Queensland University of TechnologyBrisbane, QLD, Australia; School of Biomedical Sciences, Queensland University of TechnologyBrisbane, QLD, Australia
| |
Collapse
|
14
|
Szczygielski J, Müller A, Mautes AE, Sippl C, Glameanu C, Schwerdtfeger K, Steudel WI, Oertel J. Selective Brain Hypothermia Mitigates Brain Damage and Improves Neurological Outcome after Post-Traumatic Decompressive Craniectomy in Mice. J Neurotrauma 2017; 34:1623-1635. [PMID: 27799012 DOI: 10.1089/neu.2016.4615] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hypothermia and decompressive craniectomy (DC) have been considered as treatment for traumatic brain injury. The present study investigates whether selective brain hypothermia added to craniectomy could improve neurological outcome after brain trauma. Male CD-1 mice were assigned into the following groups: sham; DC; closed head injury (CHI); CHI followed by craniectomy (CHI+DC); and CHI+DC followed by focal hypothermia (CHI+DC+H). At 24 h post-trauma, animals were subjected to Neurological Severity Score (NSS) test and Beam Balance Score test. At the same time point, magnetic resonance imaging using a 9.4 Tesla scanner and subsequent volumetric evaluation of edema and contusion were performed. Thereafter, the animals were sacrificed and subjected to histopathological analysis. According to NSS, there was a significant impairment among all the groups subjected to trauma. Animals with both trauma and craniectomy performed significantly worse than animals with craniectomy alone. This deleterious effect disappeared when additional hypothermia was applied. BBS was significantly worse in the CHI and CHI+DC groups, but not in the CHI+DC+H group, compared to the sham animals. Edema and contusion volumes were significantly increased in CHI+DC animals, but not in the CHI+DC+H group, compared to the DC group. Histopathological analysis showed that neuronal loss and contusional blossoming could be attenuated by application of selective brain hypothermia. Selective brain cooling applied post-trauma and craniectomy improved neurological function and reduced structural damage and may be therefore an alternative to complication-burdened systemic hypothermia. Clinical studies are recommended in order to explore the potential of this treatment.
Collapse
Affiliation(s)
- Jacek Szczygielski
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Andreas Müller
- 2 Department of Radiology, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Angelika E Mautes
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Christoph Sippl
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Cosmin Glameanu
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Karsten Schwerdtfeger
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Wolf-Ingo Steudel
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Joachim Oertel
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| |
Collapse
|
15
|
Quintard H, Heurteaux C, Ichai C. Adult neurogenesis and brain remodelling after brain injury: From bench to bedside? Anaesth Crit Care Pain Med 2015; 34:239-45. [PMID: 26233283 DOI: 10.1016/j.accpm.2015.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 02/19/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Brain trauma and stroke cause important disabilities. The mechanisms involved are now well described, but all therapeutics developed thus far for neuro-protection are currently unsuccessful at improving neurologic prognosis. The recently studied neuro-restorative time following brain injury may point towards a promising therapeutic approach. The purpose of this paper is to explain the mechanisms of this revolutionary concept, give an overview of related knowledge and discuss its transfer into clinical practice. DATA SOURCES AND SYNTHESIS An overview of the neurogenesis concept using MEDLINE, EMBASE and CENTRAL databases was carried out in May 2014. The clinicaltrials.gov registry was used to search for ongoing clinical trials in this domain. CONCLUSION The concept of brain remodelling upset fundamental ideas concerning the neurologic system and opened new fields of research. Therapies currently under evaluation hold promising results and could have a real prognostic impact in future years, but the translation of these therapies from the laboratory to the clinic is still far from completion.
Collapse
Affiliation(s)
- Hervé Quintard
- Intensive Care Unit, CHU Nice, 4, rue Pierre-Dévoluy, 06000 Nice, France.
| | - Catherine Heurteaux
- Institut de Pharmacologie Moléculaire et Cellulaire (CNRS), Université de Sophia-Antipolis, 660, route des Lucioles, 06560 Valbonne, France.
| | - Carole Ichai
- Intensive Care Unit, CHU Nice, 4, rue Pierre-Dévoluy, 06000 Nice, France.
| |
Collapse
|
16
|
Girisgin AS, Kalkan E, Ergin M, Keskin F, Dundar ZD, Kebapcioglu S, Kocak S, Cander B. An experimental study: does the neuroprotective effect increase when hypothermia deepens after traumatic brain injury? IRANIAN RED CRESCENT MEDICAL JOURNAL 2015; 17:e21233. [PMID: 26023335 PMCID: PMC4443303 DOI: 10.5812/ircmj.17(4)2015.21233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/16/2014] [Accepted: 11/09/2014] [Indexed: 11/16/2022]
Abstract
BACKGROUND Experimental approaches have been promising with the use of therapeutic hypothermia after Traumatic Brain Injury (TBI) whereas clinical data have not supported its efficacy. OBJECTIVES This study aimed to investigate whether using selective deeper brain cooling correlates with a more neuroprotective effect on Intracranial Pressure (ICP) increments following TBI in rats. MATERIALS AND METHODS Adult male Sprague-Dawley rats (mean weight = 300 g; n = 25) were subjected to brain injury using a modified Marmarou method. Immediately after the onset of TBI, rats were randomized into three groups. Selective brain cooling was applied around the head using ice packages. Intracranial Temperature (ICT) and ICP were continuously measured at 0, 30, 60, 120, and 180 minutes and recorded for all groups. Group 1 (n = 5) was normothermia and was assigned as the control group. Group 2 (n = 10) received moderate hypothermia with a target ICT of between 32°C - 33°C and Group 3 (n = 10) was given a deeper hypothermia with a target ICT of below 32°C. RESULTS All subjects reached the target ICT by the 30th minute of hypothermia induction. The ICT was significantly different in Group 2 compared to Group 1 only at the 120th minute (P = 0.017), while ICP was significantly lower starting from the 30th minute (P = 0.015). The ICT was significantly lower in Group 3 compared to Groups 1 and 2 starting from the 30th minute (P = 0.001 and P = 0.003, respectively). The ICP was significantly lower in Group 3 compared to Group 1 starting from 30th minute (P = 0.001); however, a significant difference in ICP between Group 3 and Group 2 was observed only at the 180th minute (P = 0.047). CONCLUSIONS Results of this study indicate that selective brain cooling is an effective method of decreasing ICP in rats; however, the deeper hypothermia caused a greater decrease in ICP three hours after hypothermia induction.
Collapse
Affiliation(s)
- Abdullah Sadik Girisgin
- Department of Emergency Medicine, Meram Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Erdal Kalkan
- Department of Neurosurgery, Meram Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Mehmet Ergin
- Department of Emergency Medicine, Meram Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Fatih Keskin
- Department of Neurosurgery, Meram Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Zerrin Defne Dundar
- Department of Emergency Medicine, Meram Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Sedat Kebapcioglu
- Department of Emergency Medicine, Medicine Faculty, Mevlana University, Konya, Turkey
| | - Sedat Kocak
- Department of Emergency Medicine, Meram Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Basar Cander
- Department of Emergency Medicine, Meram Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
17
|
Chio CC, Chang CP, Lin MT, Su FC, Yang CZ, Tseng HY, Liu ZM, Huang HS. Involvement of TG-interacting factor in microglial activation during experimental traumatic brain injury. J Neurochem 2014; 131:816-24. [PMID: 25319900 DOI: 10.1111/jnc.12971] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 09/13/2014] [Accepted: 10/06/2014] [Indexed: 11/28/2022]
Abstract
Traumatic brain injury (TBI) is a complex injury involving several physiological alterations, potentially leading to neurological impairment. Previous mouse studies using high-density oligonucleotide array analysis have confirmed the upregulation of transforming growth-interacting factor (TGIF) mRNA in TBI. TGIF is a transcriptional corepressor of transforming growth factor beta (TGF-β) signaling which plays a protective role in TBI. However, the functional roles of TGIF in TBI are not well understood. In this study, we used confocal microscopy after immunofluorescence staining to demonstrate the increase of TGIF levels in the activated microglia of the pericontusional cortex of rats with TBI. Intracerebral knockdown of TGIF in the pericontusional cortex significantly downregulated TGIF expression, attenuated microglial activation, reduced the volume of damaged brain tissue, and facilitated recovery of limb motor function. Collectively, our results indicate that TGIF is involved in TBI-induced microglial activation, resulting in secondary brain injury and motor dysfunction. This study investigated the roles of transforming growth-interacting factor (TGIF) in a traumatic brain injury (TBI)-rat model. We demonstrated the increase of TGIF levels in the activated microglia of the pericontusional cortex of rats with TBI. Intracerebral knockdown of TGIF in the pericontusional cortex of the TBI rats significantly attenuated micoglial activation, reduced the volume of damaged brain tissue, and facilitated recovery of limb motor function. We suggest that inhibition of TGIF might provide a promising therapeutic strategy for TBI.
Collapse
|
18
|
Boström M, Hellström Erkenstam N, Kaluza D, Jakobsson L, Kalm M, Blomgren K. The hippocampal neurovascular niche during normal development and after irradiation to the juvenile mouse brain. Int J Radiat Biol 2014; 90:778-89. [DOI: 10.3109/09553002.2014.931612] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
19
|
Wang CC, Chen YS, Lin BS, Chio CC, Hu CY, Kuo JR. The neuronal protective effects of local brain cooling at the craniectomy site after lateral fluid percussion injury in a rat model. J Surg Res 2013; 185:753-62. [DOI: 10.1016/j.jss.2013.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 06/08/2013] [Accepted: 07/02/2013] [Indexed: 10/26/2022]
|
20
|
Chuang CH, Hsu YC, Wang CC, Hu C, Kuo JR. Cerebral blood flow and apoptosis-associated factor with electroacupuncture in a traumatic brain injury rat model. Acupunct Med 2013; 31:395-403. [PMID: 24055977 DOI: 10.1136/acupmed-2013-010406] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Electroacupuncture (EA) has been widely used for treatment of stroke, but there is little information on the effect of EA on the neuroprotective function in traumatic brain injury (TBI). The aim of the present study was to investigate the protective effects and mechanisms of EA treatment in a TBI rat model. METHODS Male Sprague-Dawley rats were randomly divided into four groups: sham operation, TBI control, TBI+EA treated for 30 min or TBI+EA treated for 60 min. The animals were treated with EA immediately after TBI. The EA was applied at acupuncture points GV20, GV26, LI4 and KI1 with a dense-dispersed wave, frequencies of 0.2 and 1 Hz, and amplitude of 1 mA for 30 or 60 min. Regional blood flow, cell infarction volume, extent of neuronal apoptosis, expression of cell apoptosis-associated factor transforming growth-interacting factor (TGIF) were studied, and functional outcome was assessed by running speed test. All tests except regional blood flow were performed 72 h after TBI onset. RESULTS Immediately after TBI, compared with the TBI control groups, the regional blood flow was significantly increased by EA treatment for 60 min. Compared with the TBI controls 72 h after TBI, the TBI-induced run speed impairment, infarction volume, neuronal apoptosis and apoptosis-associated TGIF expression were significantly improved by EA treatment. CONCLUSIONS The treatment of TBI in the acute stage with EA for 60 min could increase the regional blood flow and attenuate the levels of TGIF in the injured cortex, might lead to a decrease in neuronal apoptosis and cell infarction volume, and might represent one mechanism by which functional recovery may occur.
Collapse
Affiliation(s)
- Chih Hsiang Chuang
- Department of Chinese Medicine, Chi-Mei Medical Center, , Tainan, Taiwan
| | | | | | | | | |
Collapse
|
21
|
Wang CC, Lin KC, Lin BS, Chio CC, Kuo JR. Resuscitation from experimental traumatic brain injury by magnolol therapy. J Surg Res 2013; 184:1045-52. [PMID: 23721932 DOI: 10.1016/j.jss.2013.04.059] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/08/2013] [Accepted: 04/24/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND The purpose of the present study was to determine whether magnolol, a free radical scavenger, mitigates the deleterious effects of traumatic brain injury (TBI). MATERIAL AND METHODS Traumatic brain injuries were induced in anesthetized male Sprague-Dawley rats using fluid percussion, and the rats were divided into groups treated with magnolol (2 mg/kg, intravenously) or vehicle. A group of rats that did not undergo TBI induction was also studied as controls. Biomarkers of TBI, including glycerol and 2,3-dihydroxybenzoic acid, were evaluated by microdialysis. Infraction volume, extent of neuronal apoptosis, and antiapoptosis factor transforming growth factor β1 (TGF-β1) were also measured. Functional outcomes were assessed by motor assays. RESULTS Compared with the rats without TBI, the animals with TBI exhibited higher hippocampal glycerol and 2,3-dihydroxybenzoic acid. Relative to the vehicle-treated group, the magnolol-treated group showed decreased hippocampal levels of glycerol and hydroxyl radical levels. The magnolol-treated rats also exhibited decreased cerebral infarction volume and neuronal apoptosis and increased antiapoptosis-associated factor TGF-β1 expression. These effects were translated into improved motor function post TBI. CONCLUSIONS Our results suggest that intravenous magnolol injection mitigates the deleterious effects of TBI in rats based on its potent free radical scavenging capability, and the mechanism of anti-neuronal apoptosis is partly due to an increase in TGF-β1 expression in the ischemic cortex.
Collapse
Affiliation(s)
- Che-Chuan Wang
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan; Institute of Imaging and Biomedical Photonics, National Chiao-Tung University, Tainan, Taiwan
| | | | | | | | | |
Collapse
|
22
|
Etanercept attenuates traumatic brain injury in rats by reducing brain TNF- α contents and by stimulating newly formed neurogenesis. Mediators Inflamm 2013; 2013:620837. [PMID: 23710117 PMCID: PMC3654326 DOI: 10.1155/2013/620837] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 03/25/2013] [Accepted: 03/25/2013] [Indexed: 11/24/2022] Open
Abstract
It remains unclear whether etanercept penetrates directly into the contused brain and improves the outcomes of TBI by attenuating brain contents of TNF-α and/or stimulating newly formed neurogenesis. Rats that sustained TBI are immediately treated with etanercept. Acute neurological and motor injury is assessed in all rats the day prior to and 7 days after surgery. The numbers of the colocalizations of 5-bromodeoxyuridine and doublecortin specific markers in the contused brain injury that occurred during TBI were counted by immunofluorescence staining. Enzyme immunoassay for quantitative determination of TNF-α or etanercept in brain tissues is also performed. Seven days after systemic administration of etanercept, levels of etanercept can be detected in the contused brain tissues. In addition, neurological and motor deficits, cerebral contusion, and increased brain TNF-α contents caused by TBI can be attenuated by etanercept therapy. Furthermore, the increased numbers of the colocalizations of 5-bromodeoxyuridine and doublecortin specific markers in the contused brain tissues caused by TBI can be potentiated by etanercept therapy. These findings indicate that systemically administered etanercept may penetrate directly into the contused brain tissues and may improve outcomes of TBI by reducing brain contents of TNF-α and by stimulating newly formed neurogenesis.
Collapse
|
23
|
Chen SH, Wang JJ, Chen CH, Chang HK, Lin MT, Chang FM, Chio CC. Umbilical cord blood-derived CD34⁺ cells improve outcomes of traumatic brain injury in rats by stimulating angiogenesis and neurogenesis. Cell Transplant 2013; 23:959-79. [PMID: 23582375 DOI: 10.3727/096368913x667006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Human umbilical cord blood cells (HUCBCs) have been shown to be beneficial in reducing neurological deficits in rats with brain fluid percussion injury (FPI). This study aimed to assess the basic mechanisms underlying the neuroprotective effects of HUCBC-derived cluster of differentiation 34-positive (CD34⁺) cells. Rats were divided into three major groups: (i) sham-operated controls; (ii) FPI rats treated with phosphate-buffered saline (PBS); (iii) FPI rats treated with 0.2%, 50%, or 95% CD34⁺ cells (in 5 × 10⁵ cord blood lymphocytes and monocytes). Intravenous (IV) administration of 0.3 ml of PBS, 0.2% CD34⁺ cells, 50% CD34⁺ cells, or 95% CD34⁺ cells was conducted immediately after FPI. It was found that 4 days post-FPI, CD34⁺ cells could be detected in the ischemic brain tissues for 50% CD34⁺ cell- or 95% CD34⁺ cell-treated FPI rats, but not for the PBS-treated FPI rats or the 0.2% CD34⁺ cell-treated FPI rats. CD34⁺ cell (0.2%)-treated FPI rats or PBS-treated FPI rats displayed neurological and motor deficits, cerebral contusion and apoptosis [e.g., increased numbers of both TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling)-positive cells and caspase-3-positive cells], and activated inflammation (e.g., increased serum levels of tumor necrosis factor-α). FPI-induced neurological motor dysfunction, cerebral contusion and apoptosis, and activated inflammation could be attenuated by 50% CD34⁺ or 95% CD34⁺ cell therapy. In addition 50% or 95% CD34⁺ cell therapy but not PBS or 0.2% CD34⁺ cell therapy significantly promoted angiogenesis (e.g., increased numbers of both vasculoendothelial growth factor-positive cells and 5-bromodeoxyuridine (BrdU)-endothelial double-positive cells), neurogenesis (e.g., increased numbers of both glial cell line-derived neurotrophic factor-positive cells and BrdU/neuronal nuclei double-positive cells) in the ischemic brain after FPI, and migration of endothelial progenitor cells from the bone marrow. Our data suggest that IV administration of HUCBC-derived CD34⁺ cells may improve outcomes of FPI in rats by stimulating both angiogenesis and neurogenesis.
Collapse
|
24
|
ZHOU TIANEN, JIANG JUN, ZHANG MENG, FU YUE, YANG ZHENGFEI, JIANG LONGYUAN. Protective effect of mild hypothermia on oxygen-glucose deprivation injury in rat hippocampal neurons after hypoxia. Mol Med Rep 2013; 7:1859-64. [DOI: 10.3892/mmr.2013.1410] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 03/18/2013] [Indexed: 11/06/2022] Open
|
25
|
Spatial relationship between NSCs/NPCs and microvessels in rat brain along prenatal and postnatal development. Int J Dev Neurosci 2013; 31:280-5. [PMID: 23518447 DOI: 10.1016/j.ijdevneu.2013.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/25/2013] [Accepted: 03/08/2013] [Indexed: 11/22/2022] Open
Abstract
Neurogenesis and angiogenesis are two parallel processes that occur in brain development and repair, and so share some molecular signals. In order to better understand the interaction between the genesis of neural cells and vessels during brain development, the density of microvessels and the number of nestin positive neural stem/neural progenitor cells (NSCs/NPCs) around microvasculature in various brain regions was quantified. Results showed that the density of microvessels remained at a relative low level during embryonic development and dramatically increased after postnatal day 3 (P3), especially in subventricular zone. The number of nestin positive NSCs/NPCs per microvessel in neurogenic brain regions continually increased with fetal brain development and then gradually dropped down during postnatal development. The highest density of NSCs/NPCs appeared at postnatal day 1 (P1) and dramatically decreased after P3. Similar pattern was observed in striatum. In the olfactory bulb, the cerebral cortex and cerebellum, the dramatic decrease of NSCs/NPCs density appeared after P7, especially in the cerebral cortex. Our results demonstrated that anatomically, the spatial relationship between NSCs/NPCs and microvessels changed during brain development. The alteration patterns in neurogenic brain regions differed from non-neurogenic brain regions.
Collapse
|
26
|
|
27
|
Kim JH, Cho YE, Seo M, Baek MC, Suk K. Glial proteome changes in response to moderate hypothermia. Proteomics 2012; 12:2571-83. [DOI: 10.1002/pmic.201200024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Jong-Heon Kim
- Department of Pharmacology; Brain Science & Engineering Institute; Kyungpook National University School of Medicine; Daegu; Republic of Korea
| | - Young-Eun Cho
- Department of Molecular Medicine; Cell & Matrix Biology Research Institute; Kyungpook National University School of Medicine; Daegu; Republic of Korea
| | - Minchul Seo
- Department of Pharmacology; Brain Science & Engineering Institute; Kyungpook National University School of Medicine; Daegu; Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine; Cell & Matrix Biology Research Institute; Kyungpook National University School of Medicine; Daegu; Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology; Brain Science & Engineering Institute; Kyungpook National University School of Medicine; Daegu; Republic of Korea
| |
Collapse
|
28
|
Prolonged gaseous hypothermia prevents the upregulation of phagocytosis-specific protein annexin 1 and causes low-amplitude EEG activity in the aged rat brain after cerebral ischemia. J Cereb Blood Flow Metab 2012; 32:1632-42. [PMID: 22617647 PMCID: PMC3421103 DOI: 10.1038/jcbfm.2012.65] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In aged humans, stroke is a major cause of disability for which no neuroprotective measures are available. In animal studies of focal ischemia, short-term hypothermia often reduces infarct size. Nevertheless, efficient neuroprotection requires long-term, regulated lowering of whole-body temperature. Previously, we reported that post-stroke exposure to hydrogen sulfide (H(2)S) effectively lowers whole-body temperature and confers neuroprotection in aged animals. In the present study using magnetic resonance imaging, electroencephalogram recording, DNA arrays, reverse transcriptase polymerase chain reaction, western blotting and immunofluorescence, we characterized the central nervous system response to H(2)S-induced hypothermia and report, for the first time, that annexin A1, a major pro-inflammatory protein that is upregulated after stroke, was consistently downregulated in polymorphonuclear cells in the peri-lesional cortex of post-ischemic, aged rat brain after 48 hours of hypothermia induced by exposure to H(2)S. Our data suggest that long-term hypothermia may be a viable clinical approach to protecting the aged brain from cerebral injury. Our findings further suggest that, in contrast to monotherapies that have thus far uniformly failed in clinical practice, hypothermia has pleiotropic effects on brain physiology that may be necessary for effective protection of the brain after stroke.
Collapse
|
29
|
Affiliation(s)
- L A Urbano
- Department of Critical Care Medicine, Lausanne University Hospital and Faculty of Biology and Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Rue du Bugnon 46, CH-1011 Lausanne, Switzerland.
| | | |
Collapse
|
30
|
Attenuating inflammation but stimulating both angiogenesis and neurogenesis using hyperbaric oxygen in rats with traumatic brain injury. J Trauma Acute Care Surg 2012; 72:650-9. [PMID: 22491549 DOI: 10.1097/ta.0b013e31823c575f] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Inflammation, angiogenesis, neurogenesis, and gliosis are involved in traumatic brain injury (TBI). Several studies provide evidence supporting the neuroprotective effect of hyperbaric oxygen (HBO2) therapy in TBI. The aim of this study was to ascertain whether inflammation, angiogenesis, neurogenesis, and gliosis during TBI are affected by HBO2 therapy. METHODS Rats were randomly divided into three groups: TBI + NBA (normobaric air: 21% O2 at 1 absolute atmospheres), TBI + HBO2, and Sham operation + NBA. TBI + HBO2 rats received 100% O2 at 2.0 absolute atmospheres for 1 hr/d for three consecutive days. Behavioral tests and biochemical and histologic evaluations were done 4 days after TBI onset. RESULTS TBI + NBA rats displayed: (1) motor and cognitive dysfunction; (2) cerebral infarction and apoptosis; (3) activated inflammation (evidenced by increased brain myeloperoxidase activity and higher serum levels of tumor necrosis factor-α); (4) neuronal loss (evidenced by fewer NeuN-positive cells); and (5) gliosis (evidenced by more glial fibrillary protein-positive cells). In TBI + HBO2 rats, HBO2 therapy significantly reduced TBI-induced motor and cognitive dysfunction, cerebral infarction and apoptosis, activated inflammation, neuronal loss, and gliosis. In addition, HBO2 therapy stimulated angiogenesis (evidenced by more bromodeoxyuridine-positive endothelial and vascular endothelial growth factor-positive cells), neurogenesis (evidenced by more bromodeoxyuridine-NeuN double-positive and glial cells-derived neurotrophic factor-positive cells), and overproduction of interleukin-10 (an anti-inflammatory cytokine). CONCLUSIONS Collectively, these results suggest that HBO2 therapy may improve outcomes of TBI in rats by inhibiting activated inflammation and gliosis while stimulating both angiogenesis and neurogenesis in the early stage.
Collapse
|
31
|
Auriat AM, Klahr AC, Silasi G, Maclellan CL, Penner M, Clark DL, Colbourne F. Prolonged hypothermia in rat: a safety study using brain-selective and systemic treatments. Ther Hypothermia Temp Manag 2012; 2:37-43. [PMID: 24717136 DOI: 10.1089/ther.2012.0005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hypothermia is an effective neuroprotectant for cardiac arrest and perinatal ischemic injury. Hypothermia also improves outcome after traumatic brain injury and stroke. Although the ideal treatment parameters (duration, delay, and depth) are not fully delineated, prolonged cooling is usually more effective than shorter periods. There is the concern that extended cooling may be hazardous to brain plasticity and cause damage. In order to evaluate this possibility, we assessed the effects of 3 days of systemic hypothermia (32°C) in rats subjected to a sham stroke surgery. There were no detrimental behavioral effects or signs of brain damage. As even longer cooling may be needed in some patients, we cooled (∼32°C) the right hemisphere of rats for 3 or 21 days. Physiological variables, functional outcome, and measures of cell injury were examined. Focal brain cooling for 21 days modestly decreased heart rate, blood pressure, and core temperature. However, focal hypothermia did not affect subsequent behavior (e.g., spontaneous limb usage), cell morphology (e.g., dendritic arborization, ultrastructure), or cause cell death. In conclusion, prolonged mild hypothermia did not harm the brain of normal animals. Further research is now needed to evaluate whether such treatments affect plasticity after brain injury.
Collapse
Affiliation(s)
- Angela M Auriat
- Department of Psychology and Center for Neuroscience, University of Alberta , Edmonton, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
32
|
Yenari MA, Han HS. Neuroprotective mechanisms of hypothermia in brain ischaemia. Nat Rev Neurosci 2012; 13:267-78. [DOI: 10.1038/nrn3174] [Citation(s) in RCA: 411] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|