1
|
Wang X, Zang J, Yang Y, Li K, Ye D, Wang Z, Wang Q, Wu Y, Luan Z. Human neural stem cells transplanted during the sequelae phase alleviate motor deficits in a rat model of cerebral palsy. Cytotherapy 2024:S1465-3249(24)00804-1. [PMID: 39186025 DOI: 10.1016/j.jcyt.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
AIMS Cerebral palsy (CP) is the most common physical disability in children, yet lacks an ideal animal model or effective treatment. This study aimed to develop a reliable CP model in neonatal rats and explore the effectiveness and underlying mechanisms of human neural stem cells (hNSCs) transplantation during the sequelae phase of CP. METHODS Vasoconstrictor endothelin-1 (ET-1) was administered intracranially to the motor cortex and striatum of rats on postnatal day 5 to establish a CP model. hNSCs (5 × 105/5 μL) pretreated with hypoxia (5% O2 for 24 h) were transplanted near the infarct 3 weeks after ET-1 injury (the sequelae phase). The distribution and differentiation of hNSCs were observed after transplantation. Changes in neurotrophic factors, neurogenesis, angiogenesis, axonal plasticity, and motor function were analyzed. RESULTS Neurobehavioral tests showed poor muscle strength and postural control in young ET-1 rats. Motor deficits of the left forelimb and gait abnormalities persisted into adulthood. Histopathological findings and MRI indicated the atrophy of the cortex, striatum, and adjacent corpus callosum in ET-1 rats. At 56 days after transplantation, hNSCs were widely distributed in the ipsilateral hemisphere, and differentiated into neurons, oligodendrocytes and astrocytes. Transplantation of hNSCs increased BDNF and VEGF expression, EdU+ cell number in the SVZ area, RECA-1+ vessel density and GAP-43 intensity around the lesion in ET-1 rats. The cylinder test revealed a significant increase in the left forelimb motor function from 28 days after transplantation, and the staircase and CatWalk tests showed improvements in fine motor function and gait parameters. CONCLUSIONS Intracerebral injection of ET-1 modelled key functional and histopathological features of CP. hNSCs transplanted during the sequelae phase of CP resulted in long-term improvement in motor performance, possibly attributed to its capacity to stimulate neurotrophic factors, facilitate neurogenesis, angiogenesis, and promote axonal plasticity.
Collapse
Affiliation(s)
- Xiaohua Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048; Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 226001
| | - Jing Zang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Yinxiang Yang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Ke Li
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Dou Ye
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Zhaoyan Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Qian Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Youjia Wu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 226001.
| | - Zuo Luan
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048.
| |
Collapse
|
2
|
Ourednik J, Ourednik V, Ghosh N, Snyder EY. Protocol to optimize the Rice-Vannucci rat pup model of perinatal asphyxia to ensure predictable hypoxic-ischemic cerebral lesions. STAR Protoc 2024; 5:103025. [PMID: 38852156 PMCID: PMC11217776 DOI: 10.1016/j.xpro.2024.103025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/01/2024] [Accepted: 04/04/2024] [Indexed: 06/11/2024] Open
Abstract
The Rice-Vannucci model in rodent pups is subject to substantial loss of animals, result inconsistency, and high lab-to-lab variability in extent and composition of induced injury. This protocol allows for highly predictable and reproducible hypoxic-ischemic cerebral injury lesions in post-natal day 10 Wistar rat pups with no mortality. We describe steps for common carotid artery ligation, brief post-operative normothermia, exposure to hypoxia, and post-hypoxic normothermia. Precise timing and temperature control in each step are crucial for a successful procedure. For complete details on the use and execution of this protocol, please refer to Hartman et al.1.
Collapse
Affiliation(s)
- Jitka Ourednik
- Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.
| | - Václav Ourednik
- Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Nirmalya Ghosh
- Department of Electrical Engineering, Indian Institute of Technology (IIT) Kharagpur, Kharagpur, West Bengal 721302, India
| | - Evan Y Snyder
- Center for Stem Cells & Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Biomedical Sciences Graduate Program, University of California San Diego, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
3
|
Drobyshevsky A, Synowiec S, Goussakov I, Fabres R, Lu J, Caplan M. Intestinal microbiota modulates neuroinflammatory response and brain injury after neonatal hypoxia-ischemia. Gut Microbes 2024; 16:2333808. [PMID: 38533575 PMCID: PMC10978030 DOI: 10.1080/19490976.2024.2333808] [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: 08/10/2023] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
Premature infants lack a normal intestinal microbial community and also at risk of perinatal hypoxic-ischemic (HI) brain injury, which is considered to be one of the major factors for motor, sensory, and cognitive deficits. We hypothesized that neonatal gut microbiota composition modulated the immune reaction and severity of neonatal H-I brain injury. Neonatal C57BL/6J mouse pups were exposed to H-I protocol consisting of permanent left carotid artery ligation, followed by 8% hypoxia for 60 min. Microbial manipulation groups included 1) antibiotic treatment, E18 (maternal) to P5; 2) antibiotic treatment E18 to P5 + E. coli gavage; 3) antibiotic treatment E18 to P5 + B. infantis gavage; and 4) saline to pups with dams getting fresh water. The extent of brain injury and recovery was measured on MRI. Edematous injury volume was significantly higher in E. coli group than that in B. infantis group and in fresh water group. Gene expression in brains of pro-inflammatory cytokines (IL1β, IL6, IL2, TNF-α and toll-like receptors 2-6) were elevated to a greater extent in the E. coli group at P10, no injury, and at P13, 72 hours after H-I relative to sham control and B. infantis groups. Significant effects of microbiome and brain injury and interaction of these factors were found in abundance of major phyla. The neuroinflammatory response and brain injury after neonatal hypoxia-ischemia are affected by intestinal microbiota, providing opportunities for therapeutic intervention through targeting the early colonization and development of the gut microbiota.
Collapse
Affiliation(s)
| | - Sylvia Synowiec
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Ivan Goussakov
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Rafael Fabres
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Jing Lu
- Department of Pediatrics, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Michael Caplan
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| |
Collapse
|
4
|
Ueda K, Sato Y, Shimizu S, Suzuki T, Onoda A, Miura R, Go S, Mimatsu H, Kitase Y, Yamashita Y, Irie K, Tsuji M, Mishima K, Mizuno M, Takahashi Y, Dezawa M, Hayakawa M. Systemic administration of clinical-grade multilineage-differentiating stress-enduring cells ameliorates hypoxic-ischemic brain injury in neonatal rats. Sci Rep 2023; 13:14958. [PMID: 37696826 PMCID: PMC10495445 DOI: 10.1038/s41598-023-41026-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are endogenous reparative pluripotent stem cells present in the bone marrow, peripheral blood, and organ connective tissues. We assessed the homing and therapeutic effects of systemically administered nafimestrocel, a clinical-grade human Muse cell-based product, without immunosuppressants in a neonatal hypoxic-ischemic (HI) rat model. HI injury was induced on postnatal day 7 (P7) and was confirmed by T2-weighted magnetic resonance imaging on P10. HI rats received a single dose nafimestrocel (1 × 106 cells/body) or Hank's balanced salt solution (vehicle group) intravenously at either three days (on P10; M3 group) or seven days (on P14; M7 group) after HI insult. Radioisotope experiment demonstrated the homing of chromium-51-labeled nafimestrocel to the both cerebral hemispheres. The cylinder test (M3 and M7 groups) and open-field test (M7 group) showed significant amelioration of paralysis and hyperactivity at five weeks of age compared with those in the vehicle group. Nafimestrocel did not cause adverse events such as death or pathological changes in the lung at ten weeks in the both groups. Nafimestrocel attenuated the production of tumor necrosis factor-α and inducible nitric oxide synthase from activated cultured microglia in vitro. These results demonstrate the potential therapeutic benefits and safety of nafimestrocel.
Collapse
Affiliation(s)
- Kazuto Ueda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan.
| | - Shinobu Shimizu
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Toshihiko Suzuki
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Atsuto Onoda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
- Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Ryosuke Miura
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Shoji Go
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Haruka Mimatsu
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Yuma Kitase
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| | - Yuta Yamashita
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Keiichi Irie
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Masahiro Tsuji
- Department of Food and Nutrition, Faculty of Home Economics, Kyoto Women's University, Kyoto, Japan
| | - Kenichi Mishima
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Masaaki Mizuno
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, 65 Tsurumai-cho Showa-ku, Nagoya, 466-8560, Japan
| |
Collapse
|
5
|
Dumont U, Sanchez S, Repond C, Beauvieux MC, Chateil JF, Pellerin L, Bouzier-Sore AK, Roumes H. Neuroprotective Effect of Maternal Resveratrol Supplementation in a Rat Model of Neonatal Hypoxia-Ischemia. Front Neurosci 2021; 14:616824. [PMID: 33519368 PMCID: PMC7844160 DOI: 10.3389/fnins.2020.616824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/23/2020] [Indexed: 02/04/2023] Open
Abstract
Neonatal hypoxia-ischemia (nHI) is a major cause of death or subsequent disabilities in infants. Hypoxia-ischemia causes brain lesions, which are induced by a strong reduction in oxygen and nutrient supply. Hypothermia is the only validated beneficial intervention, but not all newborns respond to it and today no pharmacological treatment exists. Among possible therapeutic agents to test, trans-resveratrol is an interesting candidate as it has been reported to exhibit neuroprotective effects in some neurodegenerative diseases. This experimental study aimed to investigate a possible neuroprotection by resveratrol in rat nHI, when administered to the pregnant rat female, at a nutritional dose. Several groups of pregnant female rats were studied in which resveratrol was added to drinking water either during the last week of pregnancy, the first week of lactation, or both. Then, 7-day old pups underwent a hypoxic-ischemic event. Pups were followed longitudinally, using both MRI and behavioral testing. Finally, a last group was studied in which breastfeeding females were supplemented 1 week with resveratrol just after the hypoxic-ischemic event of the pups (to test the curative rather than the preventive effect). To decipher the molecular mechanisms of this neuroprotection, RT-qPCR and Western blots were also performed on pup brain samples. Data clearly indicated that when pregnant and/or breastfeeding females were supplemented with resveratrol, hypoxic-ischemic offspring brain lesions were significantly reduced. Moreover, maternal resveratrol supplementation allowed to reverse sensorimotor and cognitive deficits caused by the insult. The best recoveries were observed when resveratrol was administered during both gestation and lactation (2 weeks before the hypoxic-ischemic event in pups). Furthermore, neuroprotection was also observed in the curative group, but only at the latest stages examined. Our hypothesis is that resveratrol, in addition to the well-known neuroprotective benefits via the sirtuin’s pathway (antioxidant properties, inhibition of apoptosis), has an impact on brain metabolism, and more specifically on the astrocyte-neuron lactate shuttle (ANLS) as suggested by RT-qPCR and Western blot data, that contributes to the neuroprotective effects.
Collapse
Affiliation(s)
- Ursule Dumont
- CRMSB, UMR 5536, CNRS/University of Bordeaux, Bordeaux, France.,Département de Physiologie, University of Lausanne, Lausanne, Switzerland
| | | | - Cendrine Repond
- Département de Physiologie, University of Lausanne, Lausanne, Switzerland
| | - Marie-Christine Beauvieux
- CRMSB, UMR 5536, CNRS/University of Bordeaux, Bordeaux, France.,CHU de Bordeaux, Place Amélie Raba Léon, Bordeaux, France
| | - Jean-François Chateil
- CRMSB, UMR 5536, CNRS/University of Bordeaux, Bordeaux, France.,CHU de Bordeaux, Place Amélie Raba Léon, Bordeaux, France
| | - Luc Pellerin
- Département de Physiologie, University of Lausanne, Lausanne, Switzerland.,IRTOMIT, Inserm U1082, University of Poitiers, Poitiers, France
| | | | - Hélène Roumes
- CRMSB, UMR 5536, CNRS/University of Bordeaux, Bordeaux, France
| |
Collapse
|
6
|
Jiang Y, Bai X, Li TT, Al-Hawwas M, Jin Y, Zou Y, Hu Y, Liu LY, Zhang Y, Liu Q, Yang H, Ma J, Wang TH, Liu J, Xiong LL. COX5A over-expression protects cortical neurons from hypoxic ischemic injury in neonatal rats associated with TPI up-regulation. BMC Neurosci 2020; 21:18. [PMID: 32349668 PMCID: PMC7191708 DOI: 10.1186/s12868-020-00565-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 04/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Neonatal hypoxic-ischemic encephalopathy (HIE) represents as a major cause of neonatal morbidity and mortality. However, the underlying molecular mechanisms in brain damage are still not fully elucidated. This study was conducted to determine the specific potential molecular mechanism in the hypoxic-ischemic induced cerebral injury. METHODS Here, hypoxic-ischemic (HI) animal models were established and primary cortical neurons were subjected to oxygen-glucose deprivation (OGD) to mimic HIE model in vivo and in vitro. The HI-induced neurological injury was evaluated by Zea-longa scores, Triphenyte-trazoliumchloride (TTC) staining the Terminal Deoxynucleotidyl Transferased Utp Nick End Labeling (TUNEL) and immunofluorescent staining. Then the expression of Cytochrome c oxidase subunit 5a (COX5A) was determined by immunohistochemistry, western blotting (WB) and quantitative real time Polymerase Chain Reaction (qRT-PCR) techniques. Moreover, HSV-mediated COX5A over-expression virus was transducted into OGD neurons to explore the role of COX5A in vitro, and the underlying mechanism was predicted by GeneMANIA, then verified by WB and qRT-PCR. RESULTS HI induced a severe neurological dysfunction, brain infarction, and cell apoptosis as well as obvious neuron loss in neonatal rats, in corresponding to the decrease on the expression of COX5A in both sides of the brain. What's more, COX5A over-expression significantly promoted the neuronal survival, reduced the apoptosis rate, and markedly increased the neurites length after OGD. Moreover, Triosephosephate isomerase (TPI) was predicted as physical interactions with COX5A, and COX5A over-expression largely increased the expressional level of TPI. CONCLUSIONS The present findings suggest that COX5A plays an important role in promoting neurological recovery after HI, and this process is related to TPI up-regulation.
Collapse
Affiliation(s)
- Ya Jiang
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Xue Bai
- National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research Center, Department of Cardiac and Cerebral Diseases, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ting-Ting Li
- National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research Center, Department of Cardiac and Cerebral Diseases, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Mohammed Al-Hawwas
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, 5000, South Australia
| | - Yuan Jin
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Yu Zou
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Yue Hu
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Lin-Yi Liu
- Department of Neurosurgery, The First People's Hospital of Zhaotong, Zhaotong, 657000, China
| | - Ying Zhang
- National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research Center, Department of Cardiac and Cerebral Diseases, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Qing Liu
- National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research Center, Department of Cardiac and Cerebral Diseases, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Hao Yang
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Jun Ma
- Department of Neurosurgery, The First People's Hospital of Zhaotong, Zhaotong, 657000, China
| | - Ting-Hua Wang
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China.
| | - Jia Liu
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China.
| | - Liu-Lin Xiong
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China.
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, 5000, South Australia.
- National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research Center, Department of Cardiac and Cerebral Diseases, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
7
|
Dumont U, Sanchez S, Olivier B, Chateil JF, Deffieux D, Quideau S, Pellerin L, Beauvieux MC, Bouzier-Sore AK, Roumes H. Maternal alcoholism and neonatal hypoxia-ischemia: Neuroprotection by stilbenoid polyphenols. Brain Res 2020; 1738:146798. [PMID: 32229200 DOI: 10.1016/j.brainres.2020.146798] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/21/2020] [Accepted: 03/14/2020] [Indexed: 01/16/2023]
Abstract
The impact of maternal nutrition on neurodevelopment and neonatal neuroprotection is a research topic with increasing interest. Maternal diet can also have deleterious effects on fetal brain development. Fetal exposure to alcohol is responsible for poor neonatal global development, and may increase brain vulnerability to hypoxic-ischemic encephalopathy, one of the major causes of acute mortality and chronic neurological disability in newborns. Despite frequent prevention campaigns, about 10% of women in the general population drinks alcohol during pregnancy and breastfeeding. This study was inspired by this alarming fact. Its aim was to evaluate the beneficial effects of maternal supplementation with two polyphenols during pregnancy and breastfeeding, on hypoxic-ischemic neonate rat brain damages, sensorimotor and cognitive impairments, in a context of moderate maternal alcoholism. Both stilbenoid polyphenols, trans-resveratrol (RSV - 0.15 mg/kg/day), and its hydroxylated analog, trans-piceatannol (PIC - 0.15 mg/kg/day), were administered in the drinking water, containing or not alcohol (0.5 g/kg/day). In a 7-day post-natal rat model of hypoxia-ischemia (HI), our data showed that moderate maternal alcoholism does not increase brain lesion volumes measured by MRI but leads to higher motor impairments. RSV supplementation could not reverse the deleterious effects of HI coupled with maternal alcoholism. However, PIC supplementation led to a recovery of all sensorimotor and cognitive functions. This neuroprotection was obtained with a dose of PIC corresponding to the consumption of a single passion fruit per day for a pregnant woman.
Collapse
Affiliation(s)
- Ursule Dumont
- CRMSB, UMR 5536, CNRS/University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France.
| | - Stéphane Sanchez
- CRMSB, UMR 5536, CNRS/University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France.
| | - Benjamin Olivier
- CRMSB, UMR 5536, CNRS/University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France.
| | - Jean-François Chateil
- CRMSB, UMR 5536, CNRS/University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France.
| | | | | | - Luc Pellerin
- CRMSB, UMR 5536, CNRS/University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France; Department of Physiology, 7 Rue du Bugnon, CH1005 Lausanne, Switzerland.
| | | | - Anne-Karine Bouzier-Sore
- CRMSB, UMR 5536, CNRS/University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France.
| | - Hélène Roumes
- CRMSB, UMR 5536, CNRS/University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France.
| |
Collapse
|
8
|
Franco PN, Durrant LM, Carreon D, Haddad E, Vergara A, Cascavita C, Obenaus A, Pearce WJ. Prenatal metyrapone treatment modulates neonatal cerebrovascular structure, function, and vulnerability to mild hypoxic-ischemic injury. Am J Physiol Regul Integr Comp Physiol 2019; 318:R1-R16. [PMID: 31577477 DOI: 10.1152/ajpregu.00145.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study explored the hypothesis that late gestational reduction of corticosteroids transforms the cerebrovasculature and modulates postnatal vulnerability to mild hypoxic-ischemic (HI) injury. Four groups of Sprague-Dawley neonates were studied: 1) Sham-Control, 2) Sham-MET, 3) HI-Control, and 4) HI-MET. Metyrapone (MET), a corticosteroid synthesis inhibitor, was administered via drinking water from gestational day 11 to term. In Shams, MET administration 1) decreased reactivity of the hypothalamic-pituitary-adrenal (HPA) axis to surgical trauma in postnatal day 9 (P9) pups by 37%, 2) promoted cerebrovascular contractile differentiation in middle cerebral arteries (MCAs), 3) decreased compliance ≤46% and increased depolarization-induced calcium mobilization in MCAs by 28%, 4) mildly increased hemispheric cerebral edema by 5%, decreased neuronal degeneration by 66%, and increased astroglial and microglial activation by 10- and 4-fold, respectively, and 5) increased righting reflex times by 29%. Regarding HI, metyrapone-induced fetal transformation 1) diminished reactivity of the HPA axis to HI-induced stress in P9/P10 pups, 2) enhanced HI-induced contractile dedifferentiation in MCAs, 3) lessened the effects of HI on MCA compliance and calcium mobilization, 4) decreased HI-induced neuronal injury but unmasked regional HI-induced depression of microglial activation, and 5) attenuated the negative effects of HI on open-field exploration but enhanced the detrimental effects of HI on negative geotaxis responses by 79%. Overall, corticosteroids during gestation appear essential for normal cerebrovascular development and glial quiescence but induce persistent changes that in neonates manifest beneficially as preservation of postischemic contractile differentiation but detrimentally as worsened ischemic cerebrovascular compliance, increased ischemic neuronal injury, and compromised neurobehavior.
Collapse
Affiliation(s)
- P Naomi Franco
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Lara M Durrant
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Desirelys Carreon
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Elizabeth Haddad
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, California
| | - Adam Vergara
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Catherine Cascavita
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, California
| | - William J Pearce
- Center for Perinatal Biology, Divisions of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
9
|
Neurite orientation dispersion and density imaging for evaluating the severity of neonatal hypoxic-ischemic encephalopathy in rats. Magn Reson Imaging 2019; 62:214-219. [PMID: 31325487 DOI: 10.1016/j.mri.2019.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/27/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE To evaluate the utility of neurite orientation dispersion and density imaging (NODDI) for longitudinally assessing neonatal hypoxic-ischemic (HI) encephalopathy severity with 7.0 T magnetic resonance imaging. METHODS Thirteen 8-day-old Wistar rats underwent unilateral ligation of the left common carotid artery followed by mild (1 h; n = 6) or severe (2 h; n = 7) hypoxic exposure (8% O2, 34 °C). Diffusion-weighted, T2-weighted (T2W), and flow-sensitive alternating inversion recovery images were obtained with a horizontal 7.0 T scanner at 1, 24, 72, and 168 h after HI insult. The fractional anisotropy (FA), apparent diffusion coefficient (ADC), intracellular volume fraction (ICVF), isotropic volume fraction (ISO), orientation dispersion index (ODI), and cerebral blood flow (CBF) values were calculated for each group (mild and severe) at each time point (1, 24, 72, and 168 h). ICVF, ISO, and ODI were the NODDI parameters. RESULTS Left hemisphere brain damage was identified as slight hyperintensity on T2W images after 1 h in both groups. In the severe group only, the signal hyperintensity increased time-dependently over 168 h. The ADC and CBF were not significantly different between the groups within any region. The ICVF and ODI were significantly higher in the severe vs. mild group at various points between 1 and 168 h (cortex, striatum, or white matter), whereas the FA was significantly higher in the mild vs. severe group at 168 h (cortex and white matter). The ISO was higher in the severe vs. mild group at 72 h (striatum) and 168 h (all regions), while the ISO was significantly higher in the mild vs. severe group at 24 h (all regions). CONCLUSION Here, ODI, a NODDI metric, identified early differences between mild and severe HI injuries. Our findings support the potential utility of NODDI for determining neonatal HI encephalopathy severity in rats.
Collapse
|
10
|
Barks AK, Beeson MM, Matveeva T, Gale JJ, Rao R, Tran PV. Perinatal Ischemia Alters Global Expression of Synaptosomal Proteins Critical for Neural Plasticity in the Developing Mouse Brain. Dev Neurosci 2019; 40:1-13. [PMID: 31207599 DOI: 10.1159/000499126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/25/2019] [Indexed: 11/19/2022] Open
Abstract
Ischemic perinatal stroke (IPS) affects 1 in 2,300-5,000 live births. Despite a survival rate >95%, approximately 60% of IPS infants develop motor and cognitive impairments. Given the importance of axonal growth and synaptic plasticity in neurocognitive development, our objective was to identify the molecular pathways underlying IPS-associated synaptic dysfunction using a mouse model. IPS was induced by unilateral ligation of the common carotid artery of postnatal day 10 (P10) mice. Five days after ischemia, sensorimotor and motor functions were assessed by vibrissae-evoked forepaw placement and the tail suspension test respectively, showing evidence of greater impairments in male pups than in female pups. Twenty-four hours after ischemia, both hemispheres were collected and synaptosomal proteins then prepared for quantification, using isobaric tags for relative and absolute quantitation. Seventy-two of 1,498 qualified proteins were altered in the ischemic hemisphere. Ingenuity Pathway Analysis was used to map these proteins onto molecular networks indicative of reduced neuronal proliferation, survival, and synaptic plasticity, accompanied by reduced PKCα signaling in male, but not female, pups. These effects also occurred in the non-ischemic hemisphere when compared with sham controls. The altered signaling effects may contribute to the sex-specific neurodevelopmental dysfunction following IPS, highlighting potential pathways for targeting during treatment.
Collapse
Affiliation(s)
- Amanda K Barks
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Montana M Beeson
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Tatyana Matveeva
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jonathan J Gale
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Raghavendra Rao
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Phu V Tran
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA,
| |
Collapse
|
11
|
Rodent Models of Developmental Ischemic Stroke for Translational Research: Strengths and Weaknesses. Neural Plast 2019; 2019:5089321. [PMID: 31093271 PMCID: PMC6476045 DOI: 10.1155/2019/5089321] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 12/19/2018] [Accepted: 02/06/2019] [Indexed: 12/25/2022] Open
Abstract
Cerebral ischemia can occur at any stage in life, but clinical consequences greatly differ depending on the developmental stage of the affected brain structures. Timing of the lesion occurrence seems to be critical, as it strongly interferes with neuronal circuit development and determines the way spontaneous plasticity takes place. Translational stroke research requires the use of animal models as they represent a reliable tool to understand the pathogenic mechanisms underlying the generation, progression, and pathological consequences of a stroke. Moreover, in vivo experiments are instrumental to investigate new therapeutic strategies and the best temporal window of intervention. Differently from adults, very few models of the human developmental stroke have been characterized, and most of them have been established in rodents. The models currently used provide a better understanding of the molecular factors involved in the effects of ischemia; however, they still hold many limitations due to matching developmental stages across different species and the complexity of the human disorder that hardly can be described by segregated variables. In this review, we summarize the key factors contributing to neonatal brain vulnerability to ischemic strokes and we provide an overview of the advantages and limitations of the currently available models to recapitulate different aspects of the human developmental stroke.
Collapse
|
12
|
Zhang ZB, Tan YX, Zhao Q, Xiong LL, Liu J, Xu FF, Xu Y, Bobrovskaya L, Zhou XF, Wang TH. miRNA-7a-2-3p Inhibits Neuronal Apoptosis in Oxygen-Glucose Deprivation (OGD) Model. Front Neurosci 2019; 13:16. [PMID: 30728764 PMCID: PMC6351497 DOI: 10.3389/fnins.2019.00016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 01/08/2019] [Indexed: 02/05/2023] Open
Abstract
Neuronal apoptosis is a major pathological hallmark of the neonatal hypoxic-ischemic brain damage (HIBD); however, the role of miR-7a-2-3p in the regulation of HIBD remains unknown. The purpose of this study was to explore the possible roles of miR-7a-2-3p in brain injury using a hypoxia-ischemia model in rats and oxygen-glucose deprivation (OGD) model in vitro. Firstly, we established the hypoxia-ischemia (HI) model and verified the model using Zea Longa scores and MRI in rats. Next, the changes of miR-7a-2-3p were screened in the ischemic cortex of neonatal rats by qRT-PCR at 12, 48, and 96 h after HIBD. We have found that the expression of miR-7a-2-3p in the HI rats decreased significantly, compared with the sham group (P < 0.01). Then, we established the OGD model in PC12 cells, SH-SY5Y cells and primary cortical neurons in vitro and qRT-PCR was used to confirm the changes of miR-7a-2-3p in these cells after the OGD. In order to determine the function of miR-7a-2-3p, PC12 cells, SH-SY5Y cells and rat primary cortical neurons were randomly divided into normal, OGD, mimic negative control (mimic-NC) and miR-7a-2-3p groups. Then, Tuj1+ (neuronal marker) staining, TUNEL assay (to detect apoptotic cells) and MTT assay (to investigate cell viability) were performed. We have found that the number of PC12 cells, SH-SY5Y cells and cortical neurons in the miR-7a-2-3p groups increased significantly (P < 0.01) in comparison to the OGD groups. The survival of cortical neurons in the miR-7a-2-3p group was improved markedly (P < 0.01), while the apoptosis of neurons in the miR-7a-2-3p group was significantly decreased (P < 0.01), compared with the normal group. Lastly, we investigated the target genes of miR-7a-2-3p by using the prediction databases (miRDB, TargetScan, miRWalk, and miRmap) and verified the target genes with qRT-PCR in the HI rats. Bioinformatics prediction showed that Vimentin (VIM), pleiomorphic adenoma gene 1(PLAG1), dual specificity phosphatase 10 (DUSP10), NAD(P)H dehydrogenase, quinone 1 (NQO1) and tumor necrosis factor receptor superfamily member 1B (TNFRSF1B) might be the targets of miR-7a-2-3p and the qRT-PCR confirmed that VIM increased in the HI rats (P < 0.01). In conclusion, miR-7a-2-3p plays a crucial role in the hypoxic-ischemic injury, and is associated with regulation of VIM.
Collapse
Affiliation(s)
- Zi-Bin Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, China.,Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ya-Xin Tan
- Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Qiong Zhao
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liu-Lin Xiong
- School of Pharmacy and Medical Sciences, Faculty of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jia Liu
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Fei-Fei Xu
- Institute of Neurological Diseases, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Xu
- Institute of Neurological Diseases, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, Faculty of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Faculty of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Ting-Hua Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, China
| |
Collapse
|
13
|
Faustino-Mendes T, Machado-Pereira M, Castelo-Branco M, Ferreira R. The Ischemic Immature Brain: Views on Current Experimental Models. Front Cell Neurosci 2018; 12:277. [PMID: 30210301 PMCID: PMC6123378 DOI: 10.3389/fncel.2018.00277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 08/08/2018] [Indexed: 01/13/2023] Open
Affiliation(s)
| | - Marta Machado-Pereira
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - Miguel Castelo-Branco
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal.,Hospital Center of Cova da Beira, Covilhã, Portugal
| | - Raquel Ferreira
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
14
|
Charriaut-Marlangue C, Baud O. A Model of Perinatal Ischemic Stroke in the Rat: 20 Years Already and What Lessons? Front Neurol 2018; 9:650. [PMID: 30131764 PMCID: PMC6090994 DOI: 10.3389/fneur.2018.00650] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/19/2018] [Indexed: 12/18/2022] Open
Abstract
Neonatal hypoxia-ischemia (HI) and ischemia are a common cause of neonatal brain injury resulting in cerebral palsy with subsequent learning disabilities and epilepsy. Recent data suggest a higher incidence of focal ischemia-reperfusion located in the middle cerebral artery (MCA) territory in near-term and newborn babies. Pre-clinical studies in the field of cerebral palsy research used, and still today, the classical HI model in the P7 rat originally described by Rice et al. (1). At the end of the 90s, we designed a new model of focal ischemia in the P7 rat to explore the short and long-term pathophysiology of neonatal arterial ischemic stroke, particularly the phenomenon of reperfusion injury and its sequelae (reported in 1998). Cerebral blood-flow and cell death/damage correlates have been fully characterized. Pharmacologic manipulations have been applied to the model to test therapeutic targets. The model has proven useful for the study of seizure occurrence, a clinical hallmark for neonatal ischemia in babies. Main pre-clinical findings obtained within these 20 last years are discussed associated to clinical pattern of neonatal brain damage.
Collapse
Affiliation(s)
| | - Olivier Baud
- INSERM U1141 PROTECT, Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France.,Division of Neonatology and Pediatric Intensive Care, Children's Hospital, Geneva University Hospitals (HUG), University of Geneva, Geneva, Switzerland
| |
Collapse
|
15
|
Pushie MJ, Crawford AM, Sylvain NJ, Hou H, Hackett MJ, George GN, Kelly ME. Revealing the Penumbra through Imaging Elemental Markers of Cellular Metabolism in an Ischemic Stroke Model. ACS Chem Neurosci 2018; 9:886-893. [PMID: 29370523 DOI: 10.1021/acschemneuro.7b00382] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Stroke exacts a heavy financial and economic burden, is a leading cause of death, and is the leading cause of long-term disability in those who survive. The penumbra surrounds the ischemic core of the stroke lesion and is composed of cells that are stressed and vulnerable to death, which is due to an altered metabolic, oxidative, and ionic environment within the penumbra. Without therapeutic intervention, many cells within the penumbra will die and become part of the growing infarct, however, there is hope that appropriate therapies may allow potential recovery of cells within this tissue region, or at least slow the rate of cell death, therefore, slowing the spread of the ischemic infarct and minimizing the extent of tissue damage. As such, preserving the penumbra to promote functional brain recovery is a central goal in stroke research. While identification of the ischemic infarct, and the infarct/penumbra boundary is relatively trivial using classical histology and microscopy techniques, accurately assessing the penetration of the penumbra zone into undamaged brain tissue, and evaluating the magnitude of chemical alterations in the penumbra, has long been a major challenge to the stroke research field. In this study, we have used synchrotron-based X-ray fluorescence imaging to visualize the elemental changes in undamaged, penumbra, and infarct brain tissue, following ischemic stroke. We have employed a Gaussian mixture model to cluster tissue areas based on their elemental characteristics. The method separates the core of the infarct from healthy tissue, and also demarcates discrete regions encircling the infarct. These regions of interest can be combined with elemental and metabolic data, as well as with conventional histology. The cell populations defined by clustering provide a reproducible means of visualizing the size and extent of the penumbra at the level of the single cell and provide a critically needed tool to track changes in elemental status and penumbra size.
Collapse
Affiliation(s)
- M. Jake Pushie
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Andrew M. Crawford
- Geological Sciences, College of Arts & Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Nicole J. Sylvain
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Huishu Hou
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Mark J. Hackett
- Curtin Institute for Functional Molecules and Interfaces, Department of Chemistry, Faculty of Science & Engineering, Curtin University, Kent Street, Bentley, Perth, Western Australia 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia 6102, Australia
| | - Graham N. George
- Geological Sciences, College of Arts & Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Michael E. Kelly
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| |
Collapse
|
16
|
Amtul Z, Yang J, Nikolova S, Lee TY, Bartha R, Cechetto DF. The Dynamics of Impaired Blood-Brain Barrier Restoration in a Rat Model of Co-morbid Injury. Mol Neurobiol 2018; 55:8071-8083. [PMID: 29508280 DOI: 10.1007/s12035-018-0904-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/10/2018] [Indexed: 12/31/2022]
Abstract
Defect in brain microperfusion is increasingly recognized as an antecedent event to Alzheimer's disease (AD) and ischemia. Nevertheless, studies on the role of impaired microperfusion as a pathological trigger to neuroinflammation, Aβ deposition as well as blood-brain barrier (BBB) disruption, and the etiological link between AD and ischemia are lacking. In this study, we employ in vivo sequential magnetic resonance imaging (MRI) and computed tomography (CT) imaging in a co-morbid rat model of β-amyloid toxicity (Aβ) and ischemia (ET1) with subsequent histopathology of striatal lesion core and penumbra at 1, 7, and 28 days post injury. Within 24 h, cerebral injury resulted in increased BBB permeability due to the dissolution of β-dystroglycan (β-DG) and basement membrane laminin by active matrix metalloproteinase9 (MMP9). As a result, net flow of circulating IgG down a hydrostatic gradient into the parenchyma led to vasogenic edema and impaired perfusion, thus increasing the apparent hyperintensity in true fast imaging with steady-state free precession (true FISP) imaging and acute hypoperfusion in CT. This was followed by a slow recruitment of reactive astroglia to the affected brain and depolarization of aquaporin4 (AQP4) expression resulting in cytotoxic edema-in an attempt to resolve vasogenic edema. On d28, functional BBB was restored in ET1 rats as observed by astrocytic MMP9 release, β-DG stabilization, and new vessel formation. This was confirmed by reduced hyperintensity on true FISP imaging and normalized cerebral blood flow in CT. While, Aβ toxicity alone was not detrimental enough, Aβ+ET1 rats showed delayed differential expression of MMP9, late recruitment of astroglial cells, protracted loss of AQP4 depolarization, and thus delayed BBB restoration and cerebral perfusion.
Collapse
Affiliation(s)
- Zareen Amtul
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 5C1, Canada.
| | - Jun Yang
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K7, Canada
| | - Simona Nikolova
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K7, Canada
| | - Ting-Yim Lee
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K7, Canada
| | - Robert Bartha
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K7, Canada.,Department of Medical Biophysics, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - David F Cechetto
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, N6A 5C1, Canada
| |
Collapse
|
17
|
Modification to the Rice-Vannucci perinatal hypoxic-ischaemic encephalopathy model in the P7 rat improves the reliability of cerebral infarct development after 48hours. J Neurosci Methods 2017. [PMID: 28648719 DOI: 10.1016/j.jneumeth.2017.06.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The Rice-Vannucci model of hypoxic-ischaemic encephalopathy (HIE) has been associated with a high degree of variability with respect to the development of cerebral infarction and infarct lesion volume. For this reason, we examined the occurrence of communicational blood flow within the common carotid (CCA), internal (ICA), and external (ECA) carotid arteries following CCA occlusion as a source of variability in the model. NEW METHOD We propose a novel modification to the Rice-Vannucci model, whereby both the CCA and ECA are permanently ligated; mitigating communicational blood flow. RESULTS Using magnetic resonance angiography, phase-contrast velocity encoding, and pulsed arterial spin labelling, the modified Rice-Vannucci model (CCA/ECA occlusion) was demonstrated to mitigate communicational blood flow, whilst significantly reducing ipsilateral hemispherical cerebral blood flow (CBF). Comparatively, the original Rice-Vannucci model (CCA occlusion) demonstrated anterograde and retrograde blood flow within the ICA and CCA, respectively, with a non-significant reduction in ipsilateral CBF. Furthermore, CCA/ECA occlusion plus hypoxia (8% O2/92% N2; 2.5h) resulted in 100% of animals presenting with an infarct (vs 87%), significantly larger infarct volume at 48h (18.5% versus 10.0%; p<0.01), reduced standard deviation (±10% versus ±15%), and significantly worsened functional outcomes when compared to CCA occlusion plus hypoxia. COMPARISON WITH EXISTING METHOD We compared a modified Rice-Vannucci model (CCA/ECA occlusion±hypoxia) to the commonly used Rice-Vannucci model (CCA occlusion±hypoxia). CONCLUSION This study demonstrates that CCA/ECA occlusion in the Rice-Vannucci model of HIE reduces infarct volume variability by limiting communicational blood flow.
Collapse
|
18
|
Hirt L, Fukuda AM, Ambadipudi K, Rashid F, Binder D, Verkman A, Ashwal S, Obenaus A, Badaut J. Improved long-term outcome after transient cerebral ischemia in aquaporin-4 knockout mice. J Cereb Blood Flow Metab 2017; 37:277-290. [PMID: 26767580 PMCID: PMC5363745 DOI: 10.1177/0271678x15623290] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/07/2023]
Abstract
A hallmark of stroke is water accumulation (edema) resulting from dysregulation of osmotic homeostasis. Brain edema contributes to tissue demise and may lead to increased intracranial pressure and lethal herniation. Currently, there are only limited treatments to prevent edema formation following stroke. Aquaporin 4 (AQP4), a brain water channel, has become a focus of interest for therapeutic approaches targeting edema. At present, there are no pharmacological tools to block AQP4. The role of AQP4 in edema after brain injury remains unclear with conflicting results from studies using AQP4-/- mice and of AQP4 expression following stroke. Here, we studied AQP4 and its role in edema formation by testing AQP4-/- mice in a model of middle cerebral artery occlusion using novel quantitative MRI water content measurements, histology and behavioral changes as outcome measures. Absence of AQP4 was associated with decreased mortality and increased motor recovery 3 to 14 days after stroke. Behavioral improvement was associated with decreased lesion volume, neuronal cell death and neuroinflammation in AQP4-/- compared to wild type mice. Our data suggest that the lack of AQP4 confers an overall beneficial role at long term with improved neuronal survival and reduced neuroinflammation, but without a direct effect on edema formation.
Collapse
Affiliation(s)
- Lorenz Hirt
- Department of Clinical Neurosciences, Neurology Service, Centre Hospitalier Universitaire Vaudois and Lausanne University, Switzerland
| | - Andrew M Fukuda
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Kamalakar Ambadipudi
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Faisil Rashid
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Devin Binder
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, University of California, Riverside, Riverside, CA, USA
| | - Alan Verkman
- Medicine and Physiology, Cardiovascular Research Institute, University of California San Francisco, CA, USA
| | - Stephen Ashwal
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Andre Obenaus
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, University of California, Riverside, Riverside, CA, USA
| | - Jerome Badaut
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA, USA .,Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA.,CNRS UMR5287, University of Bordeaux, Bordeaux, France
| |
Collapse
|
19
|
Imaging microstructural damage and plasticity in the hippocampus during epileptogenesis. Neuroscience 2015; 309:162-72. [DOI: 10.1016/j.neuroscience.2015.04.054] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/28/2015] [Accepted: 04/21/2015] [Indexed: 12/19/2022]
|
20
|
Dixon BJ, Reis C, Ho WM, Tang J, Zhang JH. Neuroprotective Strategies after Neonatal Hypoxic Ischemic Encephalopathy. Int J Mol Sci 2015; 16:22368-401. [PMID: 26389893 PMCID: PMC4613313 DOI: 10.3390/ijms160922368] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/31/2015] [Accepted: 09/06/2015] [Indexed: 12/21/2022] Open
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) is a devastating disease that primarily causes neuronal and white matter injury and is among the leading cause of death among infants. Currently there are no well-established treatments; thus, it is important to understand the pathophysiology of the disease and elucidate complications that are creating a gap between basic science and clinical translation. In the development of neuroprotective strategies and translation of experimental results in HIE, there are many limitations and challenges to master based on an appropriate study design, drug delivery properties, dosage, and use in neonates. We will identify understudied targets after HIE, as well as neuroprotective molecules that bring hope to future treatments such as melatonin, topiramate, xenon, interferon-beta, stem cell transplantation. This review will also discuss some of the most recent trials being conducted in the clinical setting and evaluate what directions are needed in the future.
Collapse
Affiliation(s)
- Brandon J Dixon
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - Cesar Reis
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Wing Mann Ho
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Medical University Innsbruck, Tyrol 6020, Austria.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|
21
|
Park WS, Sung SI, Ahn SY, Yoo HS, Sung DK, Im GH, Choi SJ, Chang YS. Hypothermia augments neuroprotective activity of mesenchymal stem cells for neonatal hypoxic-ischemic encephalopathy. PLoS One 2015; 10:e0120893. [PMID: 25816095 PMCID: PMC4376738 DOI: 10.1371/journal.pone.0120893] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 01/27/2015] [Indexed: 01/21/2023] Open
Abstract
Though hypothermia is the only clinically available treatment for neonatal hypoxic-ischemic encephalopathy (HIE), it is not completely effective in severe cases. We hypothesized that combined treatment with hypothermia and transplantation of human umbilical cord blood (UCB)-derived mesenchymal stem cells (MSCs) would synergistically attenuate severe HIE compared to stand-alone therapy. To induce hypoxia-ischemia (HI), male Sprague-Dawley rats were subjected to 8% oxygen for 120 min after unilateral carotid artery ligation on postnatal day (P) 7. After confirmation of severe HIE involving >50% of the ipsilateral hemisphere volume as determined by diffusion-weighted brain magnetic resonance imaging (MRI) within 2 h after HI, intraventricular MSC transplantation (1 × 105 cells) and/or hypothermia with target temperature at 32°C for 24 h were administered 6 h after induction of HI. Follow-up brain MRI at P12 and P42, sensorimotor function tests at P40–42, evaluation of cytokines in the cerebrospinal fluid (CSF) at P42, and histologic analysis of peri-infarct tissues at P42 were performed. Severe HI resulted in progressively increased brain infarction over time as assessed by serial MRI, increased number of cells positive for terminal deoxynucleotidyl transferase nick-end labeling, microgliosis and astrocytosis, increased CSF cytokine levels, and impaired function in behavioral tests such as rotarod and cylinder tests. All of the abnormalities observed in severe HIE showed greater improvement after combined treatment with hypothermia and MSC transplantation than with either therapy alone. Overall, these findings suggest that combined treatment with hypothermia and human UCB-derived MSC transplantation might be a novel therapeutic modality to improve the prognosis of severe HIE, an intractable disease that currently has no effective treatment.
Collapse
Affiliation(s)
- Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Soo Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong Kyung Sung
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Geun Ho Im
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul, Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
22
|
Clowry GJ, Basuodan R, Chan F. What are the Best Animal Models for Testing Early Intervention in Cerebral Palsy? Front Neurol 2014; 5:258. [PMID: 25538677 PMCID: PMC4255621 DOI: 10.3389/fneur.2014.00258] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/21/2014] [Indexed: 11/13/2022] Open
Abstract
Interventions to treat cerebral palsy should be initiated as soon as possible in order to restore the nervous system to the correct developmental trajectory. One drawback to this approach is that interventions have to undergo exceptionally rigorous assessment for both safety and efficacy prior to use in infants. Part of this process should involve research using animals but how good are our animal models? Part of the problem is that cerebral palsy is an umbrella term that covers a number of conditions. There are also many causal pathways to cerebral palsy, such as periventricular white matter injury in premature babies, perinatal infarcts of the middle cerebral artery, or generalized anoxia at the time of birth, indeed multiple causes, including intra-uterine infection or a genetic predisposition to infarction, may need to interact to produce a clinically significant injury. In this review, we consider which animal models best reproduce certain aspects of the condition, and the extent to which the multifactorial nature of cerebral palsy has been modeled. The degree to which the corticospinal system of various animal models human corticospinal system function and development is also explored. Where attempts have already been made to test early intervention in animal models, the outcomes are evaluated in light of the suitability of the model.
Collapse
Affiliation(s)
- Gavin John Clowry
- Institute of Neuroscience, Newcastle University , Newcastle upon Tyne , UK
| | - Reem Basuodan
- Institute of Neuroscience, Newcastle University , Newcastle upon Tyne , UK
| | - Felix Chan
- Institute of Neuroscience, Newcastle University , Newcastle upon Tyne , UK
| |
Collapse
|
23
|
Lin EP, Miles L, Hughes EA, McCann JC, Vorhees CV, McAuliffe JJ, Loepke AW. A Combination of Mild Hypothermia and Sevoflurane Affords Long-Term Protection in a Modified Neonatal Mouse Model of Cerebral Hypoxia-Ischemia. Anesth Analg 2014; 119:1158-73. [DOI: 10.1213/ane.0000000000000262] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
24
|
Bellot B, Peyronnet-Roux J, Gire C, Simeoni U, Vinay L, Viemari JC. Deficits of brainstem and spinal cord functions after neonatal hypoxia-ischemia in mice. Pediatr Res 2014; 75:723-30. [PMID: 24618565 DOI: 10.1038/pr.2014.42] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 12/30/2013] [Indexed: 01/18/2023]
Abstract
BACKGROUND Perinatal cerebral hypoxia-ischemia (HI) can lead to severe neurodevelopmental disorders. Studies in humans and animal models mainly focused on cerebral outcomes, and little is known about the mechanisms that may affect the brainstem and the spinal cord. Dysfunctions of neuromodulatory systems, such as the serotonergic (5-HT) projections, critical for the development of neural networks, have been postulated to underlie behavioral and motor deficits, as well as metabolic changes. METHODS The aim of this study was to investigate brainstem and spinal cord functions by means of plethysmography and sensorimotor tests in a neonatal Rice-Vanucci model of HI in mice. We also evaluated bioaminergic contents in central regions dedicated to the motor control of autonomic functions. RESULTS Mice with cerebral infarct expressed motor disturbances and had a lower body weight and a decreased respiratory frequency than SHAM, suggesting defects of brainstem neural network involved in the motor control of feeding, suckling, swallowing, and respiration. Moreover, our study revealed changes of monoamine and amino acid contents in the brainstem and the spinal cord of HI mice. CONCLUSION Our results suggest that monoaminergic neuromodulation plays an important role in the physiopathology of HI brain injury that may represent a good therapeutic target.
Collapse
Affiliation(s)
- Blandine Bellot
- 1] Institut de Neurosciences de la Timone (P3M Team), UMR 7289, CNRS, Aix Marseille Université, Marseille, France [2] Pôle de Médecine et Réanimation Néonatales, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Julie Peyronnet-Roux
- Institut de Neurosciences de la Timone (P3M Team), UMR 7289, CNRS, Aix Marseille Université, Marseille, France
| | - Catherine Gire
- Pôle de Médecine et Réanimation Néonatales, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Umberto Simeoni
- 1] Pôle de Médecine et Réanimation Néonatales, Assistance Publique Hôpitaux de Marseille, Marseille, France [2] Aix Marseille Université, Marseille, France
| | - Laurent Vinay
- Institut de Neurosciences de la Timone (P3M Team), UMR 7289, CNRS, Aix Marseille Université, Marseille, France
| | - Jean-Charles Viemari
- Institut de Neurosciences de la Timone (P3M Team), UMR 7289, CNRS, Aix Marseille Université, Marseille, France
| |
Collapse
|
25
|
Mechanisms of perinatal arterial ischemic stroke. J Cereb Blood Flow Metab 2014; 34:921-32. [PMID: 24667913 PMCID: PMC4050239 DOI: 10.1038/jcbfm.2014.41] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 12/30/2013] [Accepted: 01/02/2014] [Indexed: 01/21/2023]
Abstract
The incidence of perinatal stroke is high, similar to that in the elderly, and produces a significant morbidity and severe long-term neurologic and cognitive deficits, including cerebral palsy, epilepsy, neuropsychological impairments, and behavioral disorders. Emerging clinical data and data from experimental models of cerebral ischemia in neonatal rodents have shown that the pathophysiology of perinatal brain damage is multifactorial. These studies have revealed that, far from just being a smaller version of the adult brain, the neonatal brain is unique with a very particular and age-dependent responsiveness to hypoxia-ischemia and focal arterial stroke. In this review, we discuss fundamental clinical aspects of perinatal stroke as well as some of the most recent and relevant findings regarding the susceptibility of specific brain cell populations to injury, the dynamics and the mechanisms of neuronal cell death in injured neonates, the responses of neonatal blood-brain barrier to stroke in relation to systemic and local inflammation, and the long-term effects of stroke on angiogenesis and neurogenesis. Finally, we address translational strategies currently being considered for neonatal stroke as well as treatments that might effectively enhance repair later after injury.
Collapse
|
26
|
Ghosh N, Sun Y, Bhanu B, Ashwal S, Obenaus A. Automated detection of brain abnormalities in neonatal hypoxia ischemic injury from MR images. Med Image Anal 2014; 18:1059-69. [PMID: 25000294 DOI: 10.1016/j.media.2014.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 04/23/2014] [Accepted: 05/10/2014] [Indexed: 11/30/2022]
Abstract
We compared the efficacy of three automated brain injury detection methods, namely symmetry-integrated region growing (SIRG), hierarchical region splitting (HRS) and modified watershed segmentation (MWS) in human and animal magnetic resonance imaging (MRI) datasets for the detection of hypoxic ischemic injuries (HIIs). Diffusion weighted imaging (DWI, 1.5T) data from neonatal arterial ischemic stroke (AIS) patients, as well as T2-weighted imaging (T2WI, 11.7T, 4.7T) at seven different time-points (1, 4, 7, 10, 17, 24 and 31 days post HII) in rat-pup model of hypoxic ischemic injury were used to assess the temporal efficacy of our computational approaches. Sensitivity, specificity, and similarity were used as performance metrics based on manual ('gold standard') injury detection to quantify comparisons. When compared to the manual gold standard, automated injury location results from SIRG performed the best in 62% of the data, while 29% for HRS and 9% for MWS. Injury severity detection revealed that SIRG performed the best in 67% cases while 33% for HRS. Prior information is required by HRS and MWS, but not by SIRG. However, SIRG is sensitive to parameter-tuning, while HRS and MWS are not. Among these methods, SIRG performs the best in detecting lesion volumes; HRS is the most robust, while MWS lags behind in both respects.
Collapse
Affiliation(s)
- Nirmalya Ghosh
- Department of Pediatrics, Loma Linda University, School of Medicine, Loma Linda, CA 92354, USA
| | - Yu Sun
- Center for Research in Intelligent Systems (CRIS), University of California, Riverside, CA 92521, USA
| | - Bir Bhanu
- Center for Research in Intelligent Systems (CRIS), University of California, Riverside, CA 92521, USA
| | - Stephen Ashwal
- Department of Pediatrics, Loma Linda University, School of Medicine, Loma Linda, CA 92354, USA
| | - Andre Obenaus
- Department of Pediatrics, Loma Linda University, School of Medicine, Loma Linda, CA 92354, USA; Cell, Molecular and Developmental Biology Program and Department of Neuroscience, University of California, 1140 Bachelor Hall, Riverside, CA 92521, USA.
| |
Collapse
|
27
|
Tsuji M, Ohshima M, Taguchi A, Kasahara Y, Ikeda T, Matsuyama T. A novel reproducible model of neonatal stroke in mice: Comparison with a hypoxia–ischemia model. Exp Neurol 2013; 247:218-25. [DOI: 10.1016/j.expneurol.2013.04.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 04/01/2013] [Accepted: 04/18/2013] [Indexed: 11/16/2022]
|
28
|
Abstract
Neonatal hypoxic-ischemic brain injury (HII) and arterial ischemic stroke (AIS) result in irreversibly injured (core) and salvageable (penumbral) tissue regions. Identification and reliable quantification of salvageable tissue is pivotal to any effective and safe intervention. Magnetic resonance imaging (MRI) is the current standard to distinguish core from penumbra using diffusion-perfusion mismatch (DPM). However, subtle MR signal variations between core-penumbral regions make their visual delineation difficult. We hypothesized that computational analysis of MRI data provides a more accurate assessment of core and penumbral tissue evolution in HII/AIS. We used two neonatal rat-pup models of HII/AIS (unilateral and global hypoxic-ischemia) and clinical data sets from neonates with AIS to test our noninvasive, automated computational approach, Hierarchical Region Splitting (HRS), to detect and quantify ischemic core-penumbra using only a single MRI modality (T2- or diffusion-weighted imaging, T2WI/DWI). We also validated our approach by comparing core-penumbral images (from HRS) to DPM with immunohistochemical validation of HII tissues. Our translational and clinical data results showed that HRS could accurately and reliably distinguish the ischemic core from penumbra and their spatiotemporal evolution, which may aid in the vetting and execution of effective therapeutic interventions as well as patient selection.
Collapse
|
29
|
Yan J, Khatibi NH, Han H, Hu Q, Chen C, Li L, Yang X, Zhou C. p53-induced uncoupling expression of aquaporin-4 and inwardly rectifying K+ 4.1 channels in cytotoxic edema after subarachnoid hemorrhage. CNS Neurosci Ther 2012; 18:334-42. [PMID: 22420318 PMCID: PMC6493666 DOI: 10.1111/j.1755-5949.2012.00299.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Revised: 11/28/2011] [Accepted: 12/16/2011] [Indexed: 11/30/2022] Open
Abstract
AIMS To investigate the mechanism behind cytotoxic edema formation following subarachnoid hemorrhage (SAH). METHODS We explored the role of aquaporin-4 (AQP4), inwardly rectifying K(+) 4.1 (Kir4.1) channels and their upstream orchestrators p53 and p38MAPK in this process. A p53 inhibitor, pifithrin-α (PFT-α) was administered intraperitoneally to rats undergoing SAH by endovascular perforation. Totally, 98 male SD rats were categorized into sham, SAH, SAH+ dimethyl sulfoxide (DMSO), SAH+ 0.2 or 2.0 mg/kg PFT-α groups. At 24 h after SAH, MRI (diffusion-weighted imaging [DWI]), immunohistochemistry, and Western blot were used. RESULTS MRI (DWI) showed a significant cytotoxic edema in the brain following SAH with PFT-α therapy reducing it. Immunohistochemistry and Western blot showed an increased level of p53, phosphorylated-p38MAPK and AQP4 and a reduced level of Kir4.1; all of which could be reversed following PFT-α treatment. Treble labeling staining revealed colocalization of p53 with phosphorylated-p38MAPK and unmatched expression of AQP4 and Kir4.1 within astrocyte cells. CONCLUSION These results indicated p53 mediates the formation of cytotoxic edema in the brain following SAH; an uncoupling expression of AQP4 and Kir4.1 on astrocytic end feet orchestrated by p38MAPK was partly responsible.
Collapse
Affiliation(s)
- Jun‐hao Yan
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Physiology and Pharmacology, Loma Linda University, Medical Center, Loma Linda, California
| | - Nikan H. Khatibi
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, California
| | - Hong‐bin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Qin Hu
- Department of Physiology and Pharmacology, Loma Linda University, Medical Center, Loma Linda, California
| | - Chun‐hua Chen
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Physiology and Pharmacology, Loma Linda University, Medical Center, Loma Linda, California
| | - Li Li
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiao‐mei Yang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chang‐man Zhou
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
30
|
Superior Neuroprotective Efficacy of LAU-0901, a Novel Platelet-Activating Factor Antagonist, in Experimental Stroke. Transl Stroke Res 2011; 3:154-63. [PMID: 22408693 PMCID: PMC3284672 DOI: 10.1007/s12975-011-0116-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 10/10/2011] [Accepted: 10/12/2011] [Indexed: 01/05/2023]
Abstract
Platelet-activating factor (PAF) accumulates during cerebral ischemia, and inhibition of this process plays a critical role in neuronal survival. Recently, we demonstrated that LAU-0901, a novel PAF receptor antagonist, is neuroprotective in experimental stroke. We used magnetic resonance imaging in conjunction with behavior and immunohistopathology to expand our understanding of this novel therapeutic approach. Sprague–Dawley rats received 2 h middle cerebral artery occlusion (MCAo) and were treated with LAU-0901 (60 mg/kg) or vehicle 2 h from MCAo onset. Behavioral function, T2-weighted imaging (T2WI), and apparent diffusion coefficients were performed on days 1, 3, and 7 after MCAo. Infarct volume and number of GFAP, ED-1, and NeuN-positive cells were conducted on day 7. Behavioral deficit was significantly improved by LAU-0901 treatment compared to vehicle on days 1, 3, and 7. Total lesion volumes computed from T2WI were significantly reduced by LAU-0901 on days 1, 3, and 7 (by 83%, 90%, and 96%, respectively), which was consistent with decreased edema formation. Histopathology revealed that LAU-0901 treatment resulted in significant reduction of cortical and subcortical infarct volumes, attenuated microglial infiltration, and promoted astrocytic and neuronal survival. These findings suggest LAU-0901 is a promising neuroprotectant and provide the basis for future therapeutics in patients suffering ischemic stroke.
Collapse
|
31
|
Abstract
We investigated whether morphine plays a neuroprotective role in a neonatal rat pup model of bilateral carotid artery occlusion with hypoxia. At postnatal day 10, rats received either morphine (n = 7), naloxone (n = 7), or saline placebo (n = 15) after hypoxic-ischemic injury. Survival (days), weight gain and animal testing (negative geotaxis, surface righting, and rotarod) were compared between treatment groups. Lesion volume was delineated with magnetic resonance imaging at days 7 and 28-57 after injury. Survival in rats treated with morphine, naloxone, or saline was, respectively, 14, 29, and 73%. Median number of days of survival after bilateral carotid artery occlusion with hypoxia treated with morphine was 4 (95% confidence interval 4 to 22), with naloxone was 3 (95% confidence interval -1.4 to 21), and with placebo was 28 (95% confidence interval 18 to 28). There were no statistically significant differences in magnetic resonance imaging-derived ischemic lesion volumes, weight gain, or behavioral testing measures between the groups. Morphine was ineffective as a neuroprotectant in rat pups with severe hypoxic-ischemic injury and may have contributed to their decreased survival.
Collapse
|
32
|
Brain irradiation improves focal cerebral ischemia recovery in aged rats. J Neurol Sci 2011; 306:143-53. [DOI: 10.1016/j.jns.2011.02.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 01/21/2011] [Accepted: 02/28/2011] [Indexed: 12/14/2022]
|
33
|
Yan J, Li L, Khatibi NH, Yang L, Wang K, Zhang W, Martin RD, Han J, Zhang J, Zhou C. Blood-brain barrier disruption following subarchnoid hemorrhage may be faciliated through PUMA induction of endothelial cell apoptosis from the endoplasmic reticulum. Exp Neurol 2011; 230:240-7. [PMID: 21586287 DOI: 10.1016/j.expneurol.2011.04.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 04/17/2011] [Accepted: 04/28/2011] [Indexed: 12/26/2022]
Abstract
The blood-brain barrier (BBB) plays a vital role as both a physiologic and physical barrier in regulating the movement of water from the vasculature to the brain. During a subarachnoid hemorrhage (SAH), the BBB is disrupted by a variety of mediators, one of which can result in endothelial cell death. As a result, in the present study, we investigated the role of PUMA (p53 upregulated modulator of apoptosis) following SAH injury in rats. Specifically evaluating whether through the endoplasmic reticulum (ER), PUMA could orchestrate the induction of endothelial cell apoptosis and cause a disruption in the blood-brain barrier integrity. One hundred twelve male Sprague-Dawley rats were randomly divided into 4 groups: sham, SAH, SAH+control siRNA, SAH+PUMA siRNA. Outcomes measured include mortality rate, brain edema, BBB disruption, and neurobehavioral testing. We also used Western blotting techniques to measure the expression of key pro-apoptotic proteins such as BAX, BAK, and DRP1. PUMA siRNA treatment significantly reduced the mortality rate, cerebral edema, neurobehavioral deficits, and BBB disruption as measured by Evans blue assay following SAH injury. The T2WI images showed there was an increase in vasogenic edema in the brain following SAH, which could be alleviated by PUMA siRNA. Immunohistochemical staining and Western blot analysis demonstrated an increased expression of PUMA, BAX, BAK, GRP78 and DRP1 in the microvascular endothelial cells of the hippocampus, which was accompanied with endothelium apoptosis. This study showed that PUMA induced endothelial cell apoptosis may in fact play a significant role in BBB disruption following SAH and its mediation may be through the endoplasmic reticulum. By blocking the activity of PUMA using siRNA, we were able to prevent the accumulation of cerebral edema that occurs following BBB disruption. This translated into a preservation of functional integrity and an improvement in mortality.
Collapse
Affiliation(s)
- Junhao Yan
- Department of Anatomy and Histology, Peking University Health Science Center, Beijing 100191, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ghosh N, Recker R, Shah A, Bhanu B, Ashwal S, Obenaus A. Automated ischemic lesion detection in a neonatal model of hypoxic ischemic injury. J Magn Reson Imaging 2011; 33:772-81. [DOI: 10.1002/jmri.22488] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
35
|
Ferrari DC, Nesic O, Perez-Polo JR. Perspectives on neonatal hypoxia/ischemia-induced edema formation. Neurochem Res 2010; 35:1957-65. [PMID: 21136160 DOI: 10.1007/s11064-010-0308-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2010] [Indexed: 11/30/2022]
Abstract
Neonatal hypoxia/ischemia (HI) is the most common cause of developmental neurological, cognitive and behavioral deficits in children, with hyperoxia (HHI) treatment being a clinical therapy for newborn resuscitation. Although cerebral edema is a common outcome after HI, the mechanisms leading to excessive fluid accumulation in the brain are poorly understood. Given the rigid nature of the bone-encased brain matter, knowledge of edema formation in the brain as a consequence of any injury, as well as the importance of water clearance mechanisms and water and ion homeostasis is important to our understanding of its detrimental effects. Knowledge of the pathological process underlying the appearance of dysfunctional outcomes after development of cerebral edema after neonatal HI in the developing brain and the molecular events triggered will allow a rational assessment of HHI therapy for neonatal HI and determine whether this treatment is beneficial or harmful to the developing infant.
Collapse
|
36
|
Obenaus A, Dilmac N, Tone B, Tian HR, Hartman R, Digicaylioglu M, Snyder EY, Ashwal S. Long-term magnetic resonance imaging of stem cells in neonatal ischemic injury. Ann Neurol 2010; 69:282-91. [PMID: 21387373 DOI: 10.1002/ana.22168] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 07/03/2010] [Accepted: 07/16/2010] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Quantitative magnetic resonance imaging (MRI) can serially and noninvasively assess the degree of injury in rat pup models of hypoxic ischemic injury (HII). It can also noninvasively monitor stem cell migration following iron oxide prelabeling. Reports have shown that neural stem cells (NSCs) may help mediate neuroprotection or stimulate neuroreparative responses in adult and neonatal models of ischemic injury. We investigated the ability of high-field MRI to monitor and noninvasively quantify the migration, proliferation, and location of iron oxide-labeled NSCs over very long time periods (58 weeks) in real time while contemporaneously correlating this activity with the evolving severity and extent of neural damage. METHODS Labeled clonal murine NSCs (mNSCs) were implanted 3 days after unilateral HII in 10-day-old rat pups into the contralateral striatum or ventricle. We developed methods for objectively quantifying key aspects of dynamic NSC behavior (eg, viability; extent, and speed of migration; degree of proliferation; extent of integration into host parenchyma). MRI images were validated with histological and immunohistochemical assessments. RESULTS mNSCs rapidly migrated (100 μm/day) to the lesion site. Chains of migrating NSCs were observed in the corpus callosum. In pups subjected to HII, though not in intact control animals, we observed a 273% increase in the MR-derived volume of mNSCs 4 weeks after implantation (correlating with the known proliferative behavior of endogenous and exogenous NSCs) that slowly declined over the 58-week time course, with no adverse consequences. Large numbers of now quiescent mNSCs remained at the site of injury, many retaining their iron oxide label. INTERPRETATION Our studies demonstrate that MRI can simultaneously monitor evolving neonatal cerebral injury as well as NSC migration and location. Most importantly, it can noninvasively monitor proliferation dynamically for prolonged time periods. To be able to pursue clinical trials in newborns using stem cell therapies it is axiomatic that safety be insured through the long-term real time monitoring of cell fate and activity, particularly with regard to observing unanticipated risks to the developing brain. This study supports the feasibility of reliably using MRI for this purpose.
Collapse
Affiliation(s)
- Andre Obenaus
- Department Radiation Medicine, Loma Linda University, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Ferrari DC, Nesic OB, Perez-Polo JR. Oxygen resuscitation does not ameliorate neonatal hypoxia/ischemia-induced cerebral edema. J Neurosci Res 2010; 88:2056-65. [PMID: 20143414 DOI: 10.1002/jnr.22358] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neonatal hypoxia/ischemia (HI) is a common cause of cognitive and behavioral deficits in children with hyperoxia treatment (HHI) being the current therapy for newborn resuscitation. HI induces cerebral edema that is associated with poor neurological outcomes. Our objective was to characterize cerebral edema after HI and determine the consequences of HHI (40% or 100% O(2)). Dry weight analyses showed cerebral edema 1 to 21 days after HI in the ipsilateral cortex; and 3 to 21 days after HI in the contralateral cortex. Furthermore, HI increased blood-brain barrier (BBB) permeability 1 to 7 days after HI, leading to bilateral cortical vasogenic edema. HHI failed to prevent HI-induced increase in BBB permeability and edema development. At the molecular level, HI increased ipsilateral, but not contralateral, AQP4 cortical levels at 3 and up to 21 days after HI. HHI treatment did not further affect HI-induced changes in AQP4. In addition, we observed developmental increases of AQP4 accompanied by significant reduction in water content and increase permeability of the BBB. Our results suggest that the ipsilateral HI-induced increase in AQP4 may be beneficial and that its absence in the contralateral cortex may account for edema formation after HI. Finally, we showed that HI induced impaired motor coordination 21 days after the insult and HHI did not ameliorate this behavioral outcome. We conclude that HHI treatment is effective as a resuscitating therapy, but does not ameliorate HI-induced cerebral edema and impaired motor coordination.
Collapse
Affiliation(s)
- Diana Carolina Ferrari
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, 301 University Boulevard, Galveston, TX 77555-1072, USA
| | | | | |
Collapse
|
38
|
Bouet V, Freret T, Ankri S, Bezault M, Renolleau S, Boulouard M, Jacotot E, Chauvier D, Schumann-Bard P. Predicting sensorimotor and memory deficits after neonatal ischemic stroke with reperfusion in the rat. Behav Brain Res 2010; 212:56-63. [DOI: 10.1016/j.bbr.2010.03.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 03/19/2010] [Indexed: 11/17/2022]
|
39
|
Brissaud O, Villega F, Pieter Konsman J, Sanchez S, Raffard G, Franconi JM, Chateil JF, Bouzier-Sore AK. Short-term effect of erythropoietin on brain lesions and aquaporin-4 expression in a hypoxic-ischemic neonatal rat model assessed by magnetic resonance diffusion weighted imaging and immunohistochemistry. Pediatr Res 2010; 68:123-7. [PMID: 20461024 DOI: 10.1203/pdr.0b013e3181e67d02] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Erythropoietin (Epo) is an endogenous cytokine that regulates hematopoiesis and is widely used to treat anemia. In addition, it has recently increased interest in the neurosciences since the new concept of Epo as a neuroprotective agent has emerged. The potential protective effect of human recombinant Epo (r-hu-Epo) on a hypoxic-ischemic (HI) pup rat model was studied. Cerebral HI was obtained by permanent left carotid artery ligature of pups followed by a 2-h hypoxia. Three hours after carotid occlusion, brain lesions were assessed by magnetic resonance diffusion weighted imaging. Intraperitoneal administration of r-hu-Epo (30,000 U/kg dose) limited both the HI-induced brain lesion area and the decrease in apparent diffusion coefficient (ADC) in the lesion. To identify potential mechanisms underlying the effects of Epo, immunohistochemical detection of caspase-3 and water channel protein aquaporin-4 (AQP4) were performed. No early apoptosis was detected, but up-regulation of AQP4 expression was observed in HI pups that received r-hu-Epo compared with HI animals without treatment. This study demonstrates an early neuroprotective effect of Epo with regard to brain lesion area and ADC values. One possible mechanism of Epo for decreasing brain edema and cellular swelling could be a better clearance of water excess in brain tissue, a process possibly mediated by AQP4.
Collapse
Affiliation(s)
- Olivier Brissaud
- Neonatal Intensive Care Unit, University Children's Hospital, Bordeaux 33076, France.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Pendharkar AV, Chua JY, Andres RH, Wang N, Gaeta X, Wang H, De A, Choi R, Chen S, Rutt BK, Gambhir SS, Guzman R. Biodistribution of neural stem cells after intravascular therapy for hypoxic-ischemia. Stroke 2010; 41:2064-70. [PMID: 20616329 DOI: 10.1161/strokeaha.109.575993] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE Intravascular transplantation of neural stem cells represents a minimally invasive therapeutic approach for the treatment of central nervous system diseases. The cellular biodistribution after intravascular injection needs to be analyzed to determine the ideal delivery modality. We studied the biodistribution and efficiency of targeted central nervous system delivery comparing intravenous and intra-arterial (IA) administration of neural stem cells after brain ischemia. METHODS Mouse neural stem cells were transduced with a firefly luciferase reporter gene for bioluminescence imaging (BLI). Hypoxic-ischemia was induced in adult mice and reporter neural stem cells were transplanted IA or intravenous at 24 hours after brain ischemia. In vivo BLI was used to track transplanted cells up to 2 weeks after transplantation and ex vivo BLI was used to determine single organ biodistribution. RESULTS Immediately after transplantation, BLI signal from the brain was 12 times higher in IA versus intravenous injected animals (P<0.0001). After IA injection, 69% of the total luciferase activity arose from the brain early after transplantation and 93% at 1 week. After intravenous injection, 94% of the BLI signal was detected in the lungs (P=0.004) followed by an overall 94% signal loss at 1 week, indicating lack of cell survival outside the brain. Ex vivo single organ analysis showed a significantly higher BLI signal in the brain than in the lungs, liver, and kidneys at 1 week (P<0.0001) and 2 weeks in IA (P=0.007). CONCLUSIONS IA transplantation results in superior delivery and sustained presence of neural stem cells in the ischemic brain in comparison to intravenous infusion.
Collapse
Affiliation(s)
- Arjun V Pendharkar
- Department of Neurosurgery, Stanford University, School of Medicine, Stanford, CA 94305-5327, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Huang L, Smith A, Badaut J, Obenaus A. Dynamic Characteristics of56Fe-Particle Radiation-Induced Alterations in the Rat Brain: Magnetic Resonance Imaging and Histological Assessments. Radiat Res 2010; 173:729-37. [DOI: 10.1667/rr1832.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
42
|
Planas AM. Noninvasive Brain Imaging in Small Animal Stroke Models: MRI and PET. NEUROMETHODS 2010. [DOI: 10.1007/978-1-60761-750-1_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
43
|
Lequin MH, Dudink J, Tong KA, Obenaus A. Magnetic resonance imaging in neonatal stroke. Semin Fetal Neonatal Med 2009; 14:299-310. [PMID: 19632909 DOI: 10.1016/j.siny.2009.07.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neonatal stroke occurs in 1 in 2300-5000 live births, the incidence of which is lower than that in adults, but still higher than that in childhood. The higher incidence of perinatal stroke in preterm and term infants compared to stroke in childhood may be partly explained by higher detection rates using routine fetal ultrasound and postnatal cranial sonography. In addition, there is greater availability of magnetic resonance imaging (MRI) for neuroimaging in preterm and full-term infants, which is due in part to the availability of MR-compatible incubators and MR systems at or near the neonatal intensive care unit. In addition, the wide range of MR techniques, such as T2-, diffusion- and susceptibility-weighted imaging allows improved visualization and quantification of neonatal stroke or hypoxic-ischemic injury. This chapter reviews the MR neuroimaging modalities that actually assist the clinician in the detection of neonatal stroke.
Collapse
Affiliation(s)
- M H Lequin
- Department of Radiology, Erasmus MC - Sophia Children's Hospital, Erasmus University Medical Center, Dr Molewaterplein 60, 3015 GJ Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
44
|
Rodent neonatal bilateral carotid artery occlusion with hypoxia mimics human hypoxic-ischemic injury. J Cereb Blood Flow Metab 2009; 29:1305-16. [PMID: 19436315 DOI: 10.1038/jcbfm.2009.56] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We report a new clinically relevant model of neonatal hypoxic-ischemic injury in a 10-day-old rat pup. Bilateral carotid artery occlusion and 8% hypoxia (1 to 15 mins, BCAO-H) was induced with varying degrees of injury (mild, moderate, severe), which was quantified using magnetic resonance imaging including diffusion-weighted and T2-weighted imaging at 24 h and 21/28 days. We developed a magnetic resonance imaging-based rat pup severity score and compared 3D ischemic injury volumes/rat pup severity score with histology and behavioral testing. At 24 h, hypoxic-ischemic injury was observed in 17/27 animals; long-term survival was 81%. Magnetic resonance imaging lesion volumes did not correlate with hypoxia duration but correlated with rat pup severity score, which was used to classify animals into mild (n=21), moderate (n=6), and severe (n=10) groups with average brain lesion volumes of 0.9%, 33.2%, and 56.3%, respectively. Histology confirmed lesion location and histologic scoring correlated with the rat pup severity score. We also found excellent correlation between injury severity and multiple behavioral tasks. Bilateral carotid artery occlusion and hypoxia in the P10 rat pup is an excellent model of neonatal hypoxic-ischemic injury because it induces diffuse global injury similar to the term infant. This model can produce graded injury severity, similar to that seen in human neonates, but manipulation with hypoxia duration is unpredictable.
Collapse
|
45
|
Huang L, Smith A, Cummings P, Kendall EJ, Obenaus A. Neuroimaging assessment of memory-related brain structures in a rat model of acute space-like radiation. J Magn Reson Imaging 2009; 29:785-92. [PMID: 19306400 DOI: 10.1002/jmri.21661] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
PURPOSE To investigate the acute effects on the central nervous system (CNS) of (56)Fe radiation, a component of high-energy charged particles (HZE) in space radiation, using quantitative magnetic resonance imaging (MRI) noninvasively. MATERIALS AND METHODS Sprague-Dawley rats were exposed to whole-brain (56)Fe (0, 1, 2, and 4 Gy). At 1 week postirradiation, MRI scans were made using T2-weighted (T2WI), diffusion-weighted (DWI), and contrast enhanced T1-(CET1) imaging. T2 relaxation time and apparent diffusion coefficient (ADC) values were obtained from memory-related brain regions of interest (ROIs). Histopathology was correlated using ex vivo tissues. RESULTS No overt abnormalities were visualized using T2WI and DWI at 1 week postradiation. CET1 values did not differ significantly between the irradiated and control animals. Compared to 0 Gy, there were significant prolongations in T2 values and reductions in ADC after irradiation. In the absence of evident neuronal pathology, immunohistochemistry revealed astrocytic activation in 4 Gy animals. CONCLUSION At 1 week after whole-brain (56)Fe exposure, T2 and ADC values can differentiate radiosensitivity in regions critical for hippocampal-related memory. MRI may provide noninvasive assessment of the initial molecular/cellular disturbances in vivo after HZE irradiation.
Collapse
Affiliation(s)
- Lei Huang
- Department of Radiation Medicine, Loma Linda University School of Medicine, Loma Linda, California, USA
| | | | | | | | | |
Collapse
|
46
|
Ashwal S, Obenaus A, Snyder EY. Neuroimaging as a basis for rational stem cell therapy. Pediatr Neurol 2009; 40:227-36. [PMID: 19218036 DOI: 10.1016/j.pediatrneurol.2008.09.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 09/04/2008] [Accepted: 09/25/2008] [Indexed: 02/02/2023]
Abstract
Neonatal global or focal hypoxic-ischemic brain injury remains a frequent and devastating condition, with serious long-term sequelae. An important issue in any neonatal clinical trial of neuroprotective agents relates to developing accurate measures of injury severity and also suitable measures of the response to treatment. Advanced magnetic resonance imaging techniques can acquire serial and noninvasive data about brain structure, metabolic activity, and the response to injury or treatment. These imaging methods need validation in appropriate animal models for translational research studies in human newborns. This review describes several approaches that use imaging as well as proton magnetic resonance spectroscopy to assess the severity of ischemic injury (e.g., for possible candidate selection) and for monitoring the progression and evolution of injury over time and as an indicator of recovery or response to treatment. Preliminary data are presented on how imaging can be used after neural stem cell implantation to characterize the migration rate, the magnitude of stem cell proliferation, and their final location. Imaging has the potential to allow monitoring of many dimensions of neuroprotective treatments and can be expected to contribute to efficacy and safety when clinical trials using neural stem cells or other neuroprotective agents become available.
Collapse
Affiliation(s)
- Stephen Ashwal
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California 92354, USA.
| | | | | |
Collapse
|
47
|
Gill MB, Bockhorst K, Narayana P, Perez-Polo JR. Bax shuttling after neonatal hypoxia-ischemia: hyperoxia effects. J Neurosci Res 2009; 86:3584-604. [PMID: 18655197 DOI: 10.1002/jnr.21795] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Perinatal hypoxia-ischemia (HI) occurs in 0.2%-0.4% of all live births, with 100% O(2) resuscitation (HHI) remaining a standard clinical treatment. HI produces a broad spectrum of neuronal death phenotypes ranging from a more noninflammatory apoptotic death to a more inflammatory necrotic cell death that may be responsible for the broad spectrum of reported dysfunctional outcomes. However, the mechanisms that would account for this phenotypic spectrum of cell death are not fully understood. Here, we provide evidence that Bcl-2-associated X protein (Bax) can shuttle to different subcellular compartments in response to HI, thus triggering the different organelle-associated cell death signaling cascades resulting in cell death phenotype diversity. There was an early increase in intranuclear and total nuclear Bax protein levels followed by a later Bax redistribution to the mitochondria and endoplasmic reticulum (ER). Associated with the organelle-specific Bax shuttling time course, there was an increase in nuclear phosphorylated p53, cytosolic cleaved caspase-3, and caspase-12. When HI-treated P7 rats were resuscitated with 100% O(2) (HHI), there were increased lesion volumes as determined by T2-weighted magnetic resonance imaging with no change in cortical apoptotic signaling compared with HI treatment alone. There was, however, increased inflammatory (cytosolic-cleaved interleukin-1beta) and necrotic (increased nuclear 55-kDa-cleaved PARP-1 [poly-ADP-ribose 1] and decreased nuclear HMGB1 [nuclear high-mobility group box 1]) after HHI. Furthermore, HHI increased ER calpain activation and ER Bax protein levels compared with HI alone. These data suggest that 100% O(2) resuscitation increases Bax-mediated activation of ER cell death signaling, inflammation, and lesion volume by increasing necrotic-like cell death. In light of these findings, the use of 100% O(2) treatment for neonatal HI should be reevaluated.
Collapse
Affiliation(s)
- Martin B Gill
- Department of Neuroscience and Cell Biology, University of Texas-Medical Branch, Galveston, TX 77555-1072, USA
| | | | | | | |
Collapse
|
48
|
Jadhav V, Sugawara T, Zhang J, Jacobson P, Obenaus A. Magnetic resonance imaging detects and predicts early brain injury after subarachnoid hemorrhage in a canine experimental model. J Neurotrauma 2008; 25:1099-106. [PMID: 18729770 DOI: 10.1089/neu.2008.0518] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The canine double hemorrhage model is an established model to study cerebral vasospasm, the late sequelae of subarachnoid hemorrhage (SAH). The present study uses magnetic resonance imaging (MRI) to examine the recently reported early brain injury after SAH. Double hemorrhage SAH modeling was obtained by injecting 0.5 mL/kg of autologous arterial blood into the cisterna magna of five adult mongrel dogs on day 0 and day 2, followed by imaging at day 2 and day 7 using a 4.7-Tesla (T) scanner. White matter (WM) showed a remarkable increase in T2 values at day 2 which resolved by day 7, whereas gray matter (GM) T2 values did not resolve. The apparent diffusion coefficient (ADC) values progressively increased in both WM and GM after SAH, suggestive of a transition from vasogenic to cytotoxic edema. Ventricular volume also increased dramatically. Prominent neuronal injury with Nissl's staining was seen in the cortical GM and in the periventricular tissue. Multimodal MRI reveals acute changes in the brain after SAH and can be used to non-invasively study early brain injury and normal pressure hydrocephalus post-SAH. MR can also predict tissue histopathology and may be useful for assessing pharmacological treatments designed to ameliorate SAH.
Collapse
Affiliation(s)
- Vikram Jadhav
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California 92354, USA
| | | | | | | | | |
Collapse
|
49
|
Chen W, Ostrowski RP, Obenaus A, Zhang JH. Prodeath or prosurvival: two facets of hypoxia inducible factor-1 in perinatal brain injury. Exp Neurol 2008; 216:7-15. [PMID: 19041643 DOI: 10.1016/j.expneurol.2008.10.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 09/12/2008] [Accepted: 10/24/2008] [Indexed: 01/13/2023]
Abstract
Hypoxia, which occurs in the brain when oxygen availability drops below the normal level, is a major cause of perinatal hypoxic-ischemic injury (HII). The transcriptional factor hypoxia inducible factor-1 (HIF-1) is a key regulator in the pathophysiological response to the stress of hypoxia. Genes regulated by HIF-1 are involved in energy metabolism, erythropoiesis, angiogenesis, vasodilatation, cell survival and apoptosis. Compared with the adult brain, the neonatal brain is different in physiological structure, function, cellular composition and signaling pathway related gene activation and response after hypoxia. The purpose of this review is to determine if developmental susceptibility of the brain after hypoxic/ischemic injury is related to HIF-1alpha, which also plays a pivotal role in the normal brain development. HIF-1alpha regulates both prosurvival and prodeath responses in the neonatal brain and various mechanisms underlie the apparent contradictory effects, including duration of ischemic injury and severity, cell-types, and/or dependent on the nature of the stimulus after HII. Studies report an excessive induction of HIF-1 in the immature brain, which suggests that a cell death promoting role of HIF may prevail. Inhibition of HIF-1alpha and targeted activation of its prosurvival genes appear as a favorable therapeutic strategy. However, a better understanding of multifaceted HIF-1 function during brain development is required to explore potential targets for further therapeutic interventions in the neonate.
Collapse
Affiliation(s)
- Wanqiu Chen
- Department of Physiology, Loma Linda University, Loma Linda, CA 92354, USA
| | | | | | | |
Collapse
|
50
|
Obenaus A, Ashwal S. Magnetic resonance imaging in cerebral ischemia: focus on neonates. Neuropharmacology 2008; 55:271-80. [PMID: 18601935 DOI: 10.1016/j.neuropharm.2008.06.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Revised: 05/30/2008] [Accepted: 06/02/2008] [Indexed: 10/21/2022]
Abstract
Magnetic resonance imaging (MRI) has dramatically changed our ability to diagnose and treat stroke as well as follow its evolution and response to treatment. Early stroke and ischemia can be visualized using diffusion-weighted imaging (DWI), which utilizes proton diffusion within tissues as a reporter for evolving neuropathology that reflects cytotoxic edema, particularly during the first several days after injury. Historically, T2-weighted imaging (T2WI) has been used for evaluation of vasogenic edema and also is a reliable indicator of injured tissue late after injury. While visual analysis of MR data can provide information about the evolution of injury, quantitative analyses allow definitive and objective evaluations of injury size and location and the effectiveness of novel therapeutic strategies. We review the clinical basis of imaging for stroke and ischemia diagnosis and the methods for post-processing of MR data that could provide novel insights into the evolution and pathophysiology of stroke in the newborn.
Collapse
Affiliation(s)
- Andre Obenaus
- Department of Radiation Medicine, Loma Linda University, Loma Linda, CA 92354, USA.
| | | |
Collapse
|