101
|
Gravante G, Palmieri MB, Esposito G, Delogu D, Santeusanio G, Filingeri V, Montone A. Apoptotic Death in Deep Partial Thickness Burns vs. Normal Skin of Burned Patients. J Surg Res 2007; 141:141-5. [PMID: 17559878 DOI: 10.1016/j.jss.2006.07.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Revised: 07/14/2006] [Accepted: 07/20/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND Deep partial thickness burns have an ambiguous behavior evolving either into spontaneous healing or full thickness burns. The aim of this study was to investigate these lesions for the presence of apoptosis thereby giving a possible cellular explanation to their peculiar clinical progression. METHODS We used colocalization of DNA fragments (terminal deoxynucleotidyl transferase mediated dUTP nick end labeling) and Fas ligand CD95 antibodies to calculate the apoptotic rate of deep partial thickness burns and normal skin in 21 patients after acute thermal injuries (significant difference considered: P < 0.05). RESULTS Deep partial thickness burns were associated with a higher apoptotic rate than normal skin (48.15% +/- 17.22% versus 18.6% +/- 7.2%; P = 0.0002). There were no apparent significant correlations of apoptotic rate with age, days from injury, total burn surface area or deep burn area except for a slight correlation with sex (r = 0.484; P = 0.02). CONCLUSIONS A higher apoptotic rate was present in dermal cells of deep partial thickness burns if compared to that of the unburned skin. These data would suggest that deep partial thickness progression derive from apoptosis. Specific studies are required to confirm this hypothesis and to investigate its clinical and therapeutic significance.
Collapse
Affiliation(s)
- Gianpiero Gravante
- Department of General Surgery, University of Rome Tor Vergata, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
102
|
Childs EW, Tharakan B, Hunter FA, Tinsley JH, Cao X. Apoptotic signaling induces hyperpermeability following hemorrhagic shock. Am J Physiol Heart Circ Physiol 2007; 292:H3179-89. [PMID: 17307990 DOI: 10.1152/ajpheart.01337.2006] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hemorrhagic shock (HS) disrupts the endothelial cell barrier, resulting in microvascular hyperpermeability. Recent studies have also demonstrated that activation of the apoptotic signaling cascade is involved in endothelial dysfunction, which may result in hyperpermeability. Here we report involvement of the mitochondrial "intrinsic" pathway in microvascular hyperpermeability following HS in rats. HS resulted in the activation of the mitochondrial intrinsic pathway, as is evident from an increase in the proapoptotic Bcl-2 family member BAK, release of mitochondrial cytochrome c into the cytoplasm, and activation of caspase-3. This, along with the in vivo transfection of the proapoptotic peptide BAK (BH3), resulted in hyperpermeability (as visualized by intravital microscopy), release of mitochondrial cytochrome c into the cytoplasm, and activation of caspase-3. Conversely, transfection of the BAK (BH3) mutant had no effect on hyperpermeability. Together, these results demonstrate involvement of the mitochondrial intrinsic apoptotic pathway in HS-induced hyperpermeability and that the attenuation of this pathway may provide an alternative strategy in preserving vascular barrier integrity.
Collapse
MESH Headings
- Animals
- Apoptosis
- Capillary Permeability
- Caspase 3/metabolism
- Caspase Inhibitors
- Cysteine Proteinase Inhibitors/pharmacology
- Cytochromes c/metabolism
- Disease Models, Animal
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Enzyme Activation
- Male
- Membrane Potential, Mitochondrial
- Mesentery/blood supply
- Microcirculation/metabolism
- Microcirculation/pathology
- Microcirculation/physiopathology
- Microscopy, Video
- Mitochondria/metabolism
- Mitochondria/pathology
- Oligopeptides/pharmacology
- Peptide Fragments/genetics
- Peptide Fragments/metabolism
- Rats
- Rats, Sprague-Dawley
- Shock, Hemorrhagic/metabolism
- Shock, Hemorrhagic/pathology
- Shock, Hemorrhagic/physiopathology
- Signal Transduction
- Transfection
- bcl-2 Homologous Antagonist-Killer Protein/genetics
- bcl-2 Homologous Antagonist-Killer Protein/metabolism
- von Willebrand Factor/metabolism
Collapse
Affiliation(s)
- Ed W Childs
- Department of Surgery, The Texas A & M University, HSC College of Medicine, Scott & White Memorial Hospital, 2401 South 31st St., Temple, TX 76508, USA.
| | | | | | | | | |
Collapse
|
103
|
Denker SP, Ji S, Dingman A, Lee SY, Derugin N, Wendland MF, Vexler ZS. Macrophages are comprised of resident brain microglia not infiltrating peripheral monocytes acutely after neonatal stroke. J Neurochem 2007; 100:893-904. [PMID: 17212701 PMCID: PMC2262099 DOI: 10.1111/j.1471-4159.2006.04162.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Macrophages can be both beneficial and detrimental after CNS injury. We previously showed rapid accumulation of macrophages in injured immature brain acutely after ischemia-reperfusion. To determine whether these macrophages are microglia or invading monocytes, we subjected post-natal day 7 (P7) rats to transient 3 h middle cerebral artery (MCA) occlusion and used flow cytometry at 24 and 48 h post-reperfusion to distinguish invading monocytes (CD45high/CD11b+) from microglia (CD45low/medium/CD11b+). Inflammatory cytokines and chemokines were determined in plasma, injured and contralateral tissue 1-24 h post-reperfusion using ELISA-based cytokine multiplex assays. At 24 h, the number of CD45+/CD11b+ cells increased 3-fold in injured compared to uninjured brain tissue and CD45 expression shifted from low to medium with less than 10% of the population expressing CD45high. MCA occlusion induced rapid and transient asynchronous increases in the pro-inflammatory cytokine IL-beta and chemokines cytokine-induced neutrophil chemoattractant protein 1 (CINC-1) and monocyte-chemoattractant protein 1 (MCP-1), first in systemic circulation and then in injured brain. Double immunofluorescence with cell-type specific markers showed that multiple cell types in the injured brain produce MCP-1. Our findings show that despite profound increases in MCP-1 in injured regions, monocyte infiltration is low and the majority of macrophages in acutely injured regions are microglia.
Collapse
Affiliation(s)
- Sheryl P. Denker
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Shaoquan Ji
- Linco Research, Inc., St Charles, Missouri, USA
| | - Andra Dingman
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Sarah Y. Lee
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Nikita Derugin
- Department of Neurosurgery, University of California, San Francisco, San Francisco, California, USA
| | - Michael F. Wendland
- Department of Radiology, University of California, San Francisco, San Francisco, California, USA
| | - Zinaida S. Vexler
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
104
|
Acarin L, Villapol S, Faiz M, Rohn TT, Castellano B, González B. Caspase-3 activation in astrocytes following postnatal excitotoxic damage correlates with cytoskeletal remodeling but not with cell death or proliferation. Glia 2007; 55:954-65. [PMID: 17487878 DOI: 10.1002/glia.20518] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Caspase-3 has classically been defined as the main executioner of programmed cell death. However, recent data supports the participation of this protease in non-apoptotic cellular events including cell proliferation, cell cycle regulation, and cellular differentiation. In this study, astroglial cleavage of caspase-3 was analyzed following excitotoxic damage in postnatal rats to determine if its presence is associated with apoptotic cell death, cell proliferation, or cytoskeletal remodeling. A well-characterized in vivo model of excitotoxicity was studied, where damage was induced by intracortical injection of N-methyl-D-asparate (NMDA) in postnatal day 9 rats. Our results demonstrate that cleaved caspase-3 was mainly observed in the nucleus of activated astrocytes in the lesioned hemisphere as early as 4 h postlesion and persisted until the glial scar was formed at 7-14 days, and it was not associated with TUNEL labeling. Caspase-3 enzymatic activity was detected at 10 h and 1 day postlesion in astrocytes, and co-localized with caspase-cleaved fragments of glial fibrillary acidic protein (CCP-GFAP). However, at longer survival times, when astroglial hypertrophy was observed, astroglial caspase-3 did not generally correlate with GFAP cleavage, but instead was associated with de novo expression of vimentin. Moreover, astroglial caspase-3 cleavage was not associated with BrdU incorporation. These results provide further evidence for a nontraditional role of caspases in cellular function that is independent of cell death and suggest that caspase activation is important for astroglial cytoskeleton remodeling following cellular injury.
Collapse
Affiliation(s)
- Laia Acarin
- Medical Histology, Department of Cell Biology, Physiology and Immunology, Faculty of Medicine and Institute of Neurosciences, Autonomous University of Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
105
|
Ashwal S, Tone B, Tian HR, Chong S, Obenaus A. Comparison of two neonatal ischemic injury models using magnetic resonance imaging. Pediatr Res 2007; 61:9-14. [PMID: 17211133 DOI: 10.1203/01.pdr.0000251612.16069.4b] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Using an 11.7-Tesla magnetic resonance imaging (MRI) scanner in 10-d-old rat pups we report on the evolution of injury over 28 d in a model of neonatal stroke (transient filament middle cerebral artery occlusion, tfMCAO) and a model of hypoxic-ischemic injury (Rice-Vannucci model, RVM). In both models, diffusion-weighted imaging (DWI) was more sensitive in the early detection of ischemia than T2-weighted imaging (T2WI). Injury volumes in both models were greater on d 1 for DWI and d 3 for T2WI, decreased over time and by d 28 T2WI injury volumes (tfMCAO 10.3% of ipsilateral hemisphere; RVM 23.9%) were definable. The distribution of injury with tfMCAO was confined to the vascular territory of the middle cerebral artery and a definable core and penumbra evolved over time. Ischemic injury in the RVM was more generalized and greater in cortical regions. Contralateral hemispheric involvement was only observed in the RVM. Our findings demonstrate that high-field MRI over extended periods of time is possible in a small animal model of neonatal brain injury and that the tfMCAO model should be used for studies of neonatal stroke and that the RVM does not reflect the vascular distribution of injury seen with focal ischemia.
Collapse
Affiliation(s)
- Stephen Ashwal
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92320, USA.
| | | | | | | | | |
Collapse
|
106
|
Askalan R, Deveber G, Ho M, Ma J, Hawkins C. Astrocytic-inducible nitric oxide synthase in the ischemic developing human brain. Pediatr Res 2006; 60:687-92. [PMID: 17065568 DOI: 10.1203/01.pdr.0000246226.89215.a6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Variability in the expression of apoptotic and inflammatory mediators with time after an ischemic insult and their role in the expansion of cerebral infarcts are still controversial. This study examines DNA degradation and the expression of activated caspase-3 and iNOS, inducible nitric oxide (iNOS) in the human developing brain. Autopsy specimens from children with a neuropathologic diagnosis of focal ischemic infarct were included in the study. The specimens were classified based on the clinical history as acute (< 24 h, n = 5), subacute (24-72 h, n = 8), or old (> 72 h, n = 6) infarcts. Immunohistochemical staining for caspase-3, iNOS and TUNEL were then performed on all infarcts alongside age-matched controls. TUNEL staining was detected in regions of all infarcts. Expression of iNOS was significantly higher than that of caspase-3 in the penumbra of subacute infarcts (p = 0.02). Glial fibrillary acidic protein and iNOS staining co-localized in the penumbra of acute and subacute infarcts. These results suggest that cell death continues to occur for more than 3 d post ischemic insult. Cell death in the penumbra of subacute infarcts is partially caspase-3 independent and may be attributed to nitric oxide. Astrocytes are a source of iNOS and may play a role in the evolution of pediatric brain injury days after the initial insult.
Collapse
Affiliation(s)
- Rand Askalan
- Division of Neurology, Hospital for Sick Children, M5G 1X8 Toronto, Ontario, Canada.
| | | | | | | | | |
Collapse
|
107
|
Zhang ZJ, Li P, Wang Z, Li PT, Zhang WS, Sun ZH, Zhang XJ, Wang YY. A comparative study on the individual and combined effects of baicalin and jasminoidin on focal cerebral ischemia-reperfusion injury. Brain Res 2006; 1123:188-95. [PMID: 17069775 DOI: 10.1016/j.brainres.2006.09.063] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 09/07/2006] [Accepted: 09/08/2006] [Indexed: 11/18/2022]
Abstract
To compare the individual effects of baicalin and jasminoidin with the combined effect of them on cerebral ischemia-reperfusion injury, and test whether the combined administration of baicalin and jasminoidin can improve the therapeutic effect. Male Sprague-Dawley rats underwent focal cerebral ischemia for 1.5 h and reperfusion for 24 h. Just before reperfusion, tested drugs (baicalin, jasminoidin, a drug combination consisting of baicalin and jasminoidin, or nimodipine) were intravenously treated. Diffusion weighted imaging (DWI) of magnetic resonance imaging (MRI), behavior examination, 2,3,5-triphenyltetrazolium chloride (TTC) staining, histological examination, and real-time PCR for BDNF and caspase-3 were performed. All of the drug treatments could significantly ameliorate the results of TTC and histological examination, and the baicalin/jasminoidin combination did so most prominently. This combination could also significantly ameliorate DWI of MRI and behavior examination results, and promote the expression of BDNF and inhibit the expression of caspase-3. On the whole, both baicalin and jasminoidin have a preventive effect against ischemic stroke, although their effects are not very strong. However, the combination of baicalin and jasminoidin can significantly improve their effectiveness. This may be related to its better regulation on the BDNF and caspase-3.
Collapse
Affiliation(s)
- Zhan-Jun Zhang
- Beijing Normal University, The Key Laboratory of Traditional Chinese Medicine Protection and Utilization, 19 XinJieKouWai Street, HaiDian District, Beijing, PA 100875, China.
| | | | | | | | | | | | | | | |
Collapse
|
108
|
Northington FJ. Brief update on animal models of hypoxic-ischemic encephalopathy and neonatal stroke. ILAR J 2006; 47:32-8. [PMID: 16391429 DOI: 10.1093/ilar.47.1.32] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The discovery of safe and effective therapies for perinatal hypoxia ischemia (HI) and stroke remains an unmet goal of neonatal-perinatal medicine. Because of the many developmental and functional differences between the neonatal brain and the adult brain, the ability to extrapolate adult data to the neonatal condition is very limited. For this reason, it is incumbent on scientists in the field of neonatal brain injury to address the questions of therapeutic efficacy of an array of potential therapies in a developmentally appropriate model. Toward that end, a number of new models of neonatal HI and stroke have been introduced recently. Additionally, some of the established models have been adapted to different species and different ages, giving scientists a greater choice of models for the study of neonatal HI and stroke. Many of these models are now also being used for functional and behavioral testing, an absolute necessity for preclinical therapeutic trials. This review focuses primarily on the newly developed models, recent adaptations to established models, and the studies of functional outcome that have been published since 2000.
Collapse
Affiliation(s)
- Frances J Northington
- Department of Pediatrics, Eudowood Neonatal Pulmonary Division, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
109
|
Gravante G, Palmieri MB, Esposito G, Delogu D, Santeusanio G, Filingeri V, Montone A. Apoptotic Cells Are Present in Ischemic Zones of Deep Partial-Thickness Burns. J Burn Care Res 2006; 27:688-93. [PMID: 16998402 DOI: 10.1097/01.bcr.0000238101.94950.ec] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Deep partial-thickness burns exhibit ambiguous behavior, either spontaneously healing or evolving into full-thickness burns. The aim of this study was to investigate these lesions for the presence of apoptotic cells and to compare their rate with that of superficial and full-thickness burns. We used colocalization of DNA fragments (ie, terminal deoxynucleotidyl transferase Biotin-dUTP nick end labeling) and Fas ligand CD95 antibodies to calculate the apoptotic rate of superficial, deep partial-thickness and full-thickness burns in 45 patients after the thermal injury. Biopsies were collected mainly during the acute postburn phase (first week of hospitalization). Deep partial-thickness burns presented apoptotic cells, both in the dermis and in cutaneous adnexa, and showed a higher apoptotic rate than superficial and full-thickness burns (44.5% in deep partial thickness, interquartile range 6.3-90.5%; 5.6% in superficial partial thickness, interquartile range: 0-13%; 0% for full-thickness burn; P = .000243). A significant greater apoptotic rate was present in cells of deep partial-thickness burns when compared with superficial and full thickness. These data would suggest that deep burns sustain an ischemic damage that forces cells to undergo apoptosis and could represent the biologic basis for their clinical evolution into full-thickness burns. Further correlation studies are now required to confirm this hypothesis.
Collapse
|
110
|
Vexler ZS, Sharp FR, Feuerstein GZ, Ashwal S, Thoresen M, Yager JY, Ferriero DM. Translational stroke research in the developing brain. Pediatr Neurol 2006; 34:459-63. [PMID: 16765824 DOI: 10.1016/j.pediatrneurol.2005.10.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Revised: 08/15/2005] [Accepted: 10/06/2005] [Indexed: 11/26/2022]
Abstract
Preclinical animal models can help guide the development of clinical pediatric and newborn stroke trials. Data obtained using currently available models of hypoxia-ischemia and focal stroke have demonstrated the need for age-appropriate models. There are age-related differences in susceptibility of the immature brain to oxidative stress and inflammation, as well as in the rate and degree of apoptotic neuronal death. These issues need to be carefully addressed in designing future clinical trials.
Collapse
Affiliation(s)
- Zinaida S Vexler
- Department of Neurology, University of California San Francisco, San Francisco, California 94143-0663, USA.
| | | | | | | | | | | | | |
Collapse
|
111
|
Grant PE, Yu D. Acute Injury to the Immature Brain with Hypoxia with or Without Hypoperfusion. Magn Reson Imaging Clin N Am 2006; 14:271-85. [PMID: 16873014 DOI: 10.1016/j.mric.2006.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hypoxia with or without hypoperfusion is a common mechanism of injury in the immature brain and can result in normal or minimally abnormal imaging studies in the first 12 hours. Often, injury progresses over time suggesting a central role for delayed cell death pathways. An awareness of the patterns of injury associated with different mechanisms of injury and the central role of delayed cell death pathways in injury evolution is important for the radiologist to understand the significance and potential outcome of these injuries.
Collapse
Affiliation(s)
- P Ellen Grant
- Division of Pediatric Radiology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | |
Collapse
|
112
|
Wei L, Han BH, Li Y, Keogh CL, Holtzman DM, Yu SP. Cell death mechanism and protective effect of erythropoietin after focal ischemia in the whisker-barrel cortex of neonatal rats. J Pharmacol Exp Ther 2006; 317:109-16. [PMID: 16357210 DOI: 10.1124/jpet.105.094391] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cell death induced by the combined insult of hypoxia-ischemia in neonatal rodents has been extensively investigated. Ischemia-only-induced cell death, however, has been much less characterized. Based on the notion that 1) ischemic stroke is a relatively common disorder in human neonates, and 2) developing cells are more susceptible to apoptosis, the present study examined whether typical apoptosis was induced by cerebral ischemia in a new neonatal rat model. Erythropoietin (EPO; Epoetin) was tested for its protective effect against ischemia-induced cell death. Postnatal day 7 rats were subjected to permanent occlusion of the middle cerebral artery branch supplying the right whisker-barrel cortex. Terminal deoxynucleotidyl transferase biotin-dUTP nick end-labeled-positive cells in the ischemic region were detectable 4 h after ischemia and reached a peak level 16 h later. The cell death was preceded by caspase activation and cytochrome c release. Cell body shrinkage was evident among damaged cells. Agarose gel electrophoresis showed DNA damage with a smear pattern as well as DNA laddering. Electron microscopy demonstrated apoptotic features such as cell shrinkage, chromatin condensation, and fragmentation; meanwhile, necrotic alterations coexisted in the cytoplasm. EPO treatment increased signal transducers and activators of transcription-5 and Bcl-2 levels, markedly attenuated apoptotic cell death, and reduced ischemic infarct in the cortex. It is suggested that focal ischemia in the developing brain causes cell death with prominent apoptotic features coexisting with some characteristics of necrosis. This is consistent with the concept of hybrid death described previously in cultures and adult or developing brain. EPO may be explored as a potential therapy for neonatal ischemic stroke.
Collapse
Affiliation(s)
- Ling Wei
- Departments of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, USA
| | | | | | | | | | | |
Collapse
|
113
|
West T, Atzeva M, Holtzman DM. Caspase-3 deficiency during development increases vulnerability to hypoxic-ischemic injury through caspase-3-independent pathways. Neurobiol Dis 2006; 22:523-37. [PMID: 16480886 DOI: 10.1016/j.nbd.2005.12.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2005] [Revised: 12/17/2005] [Accepted: 12/22/2005] [Indexed: 02/01/2023] Open
Abstract
Neonatal hypoxia-ischemia (H-I) is a common cause of perinatal morbidity and mortality leading to prominent activation of caspase-3 in the brain. Previous studies have shown that acute inhibition of caspase-3 can protect against neonatal H-I in rats. In this study, we investigated brain injury following neonatal H-I in mice deficient in caspase-3. Wild-type, caspase-3+/- and caspase-3-/- mice underwent unilateral carotid ligation at postnatal day (P) 7, followed by 45 min of exposure to 8% oxygen. Surprisingly, tissue loss at P14 was significantly higher in caspase-3-/- mice when compared to wild-type littermates. As in rats, we found that acute inhibition of caspase-3 in mice leads to decrease in tissue loss at P14. There was no difference in nuclear morphology, DNA laddering or calpain activation between caspase-3-/-caspase-3+/- and wild-type mice subjected to H-I, and there was no evidence for compensatory activation of other caspases in caspase-3-/- mice. Also, all genotypes showed evidence of mitochondrial dysfunction after H-I, suggesting that this is a critical point in regulation of neuronal cell death following neonatal H-I. Our results suggest that long-term inhibition of caspase-3 during development, unlike acute inhibition, leads to upregulation of caspase-3-independent cell death pathways and increases the vulnerability of the developing brain to neonatal H-I injury.
Collapse
Affiliation(s)
- Tim West
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
114
|
Dingman A, Lee SY, Derugin N, Wendland MF, Vexler ZS. Aminoguanidine inhibits caspase-3 and calpain activation without affecting microglial activation following neonatal transient cerebral ischemia. J Neurochem 2006; 96:1467-79. [PMID: 16464234 DOI: 10.1111/j.1471-4159.2006.03672.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Microglial cells, the resident macrophages of the CNS, can be both beneficial and detrimental to the brain. These cells play a central role as mediators of neuroinflammation associated with many neurodegenerative states, including cerebral ischemia. Because microglial cells are both a major source of inducible nitric oxide synthase (iNOS)/nitric oxide (NO) production locally in the injured brain and are activated by NO-mediated injury, we tested whether iNOS inhibition reduces microglial activation and ischemic injury in a neonatal focal ischemia-reperfusion model. Post-natal day 7 rats were subjected to a 2 h transient middle cerebral artery (MCA) occlusion. Pups with confirmed injury on diffusion-weighted magnetic resonance imaging (MRI) during occlusion were administered 300 mg/kg/dose aminoguanidine (AG) or vehicle at 0, 4 and 18 h after reperfusion, and animals were killed at 24 or 72 h post-reperfusion. The effect of AG on microglial activation as judged by the acquisition of ED1 immunoreactivity and proliferation of ED1-positive cells, on activation of cell death pathways and on injury volume, was determined. The study shows that while AG attenuates caspase 3 and calpain activation in the injured tissue, treatment does not affect the rapidly occurring activation and proliferation of microglia following transient MCA occlusion in the immature rat, or reduce injury size.
Collapse
Affiliation(s)
- Andra Dingman
- Department of Neurology, University of California San Francisco, California 94143-0663, USA
| | | | | | | | | |
Collapse
|
115
|
Gravante G, Esposito G, Filingeri V, Delogu D, Castrì F, Montone A. Apoptotic cell death in toxic epidermal necrolysis: the Fas ligand assumption. Arch Dermatol Res 2006; 297:430-1; author reply 432. [PMID: 16429298 DOI: 10.1007/s00403-006-0639-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2005] [Accepted: 12/27/2005] [Indexed: 10/25/2022]
|
116
|
Grant PE, Yu D. Acute injury to the immature brain with hypoxia with or without hypoperfusion. Radiol Clin North Am 2006; 44:63-77, viii. [PMID: 16297682 DOI: 10.1016/j.rcl.2005.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
This article reviews the imaging features and evolution of immature brain injury caused by hypoxia with or without hypoperfusion in the neonate and young child. Clinical presentations and available literature on mechanisms and clinical outcomes are discussed. In many of these cases, diffusion-weighted imaging does not show the full extent of the injury but detects a pattern of injury that is important in guiding clinical care. Awareness of the delayed cell death mechanisms is essential to understand diffusion-weighted imaging sensitivity and evolution and to provide the most accurate clinical interpretation, especially in cases of hypoxia with or without hypoperfusion.
Collapse
Affiliation(s)
- P Ellen Grant
- Division of Pediatric Radiology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | |
Collapse
|
117
|
Derugin N, Dingman A, Wendland MF, Fox C, Bollen A, Vexler ZS. Magnetic resonance imaging as a surrogate measure for histological sub-chronic endpoint in a neonatal rat stroke model. Brain Res 2005; 1066:49-56. [PMID: 16336947 DOI: 10.1016/j.brainres.2005.10.043] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Revised: 10/07/2005] [Accepted: 10/11/2005] [Indexed: 11/22/2022]
Abstract
INTRODUCTION It is becoming increasingly recognized that CNS immaturity at birth affects ischemic injury and recovery, and that the consequences of neonatal stroke need to be studied using age-appropriate focal stroke models. The inclusion of magnetic resonance imaging (MRI) as a surrogate measure of stroke progression has provided useful information in adult models, but the benefit for neonatal stroke studies is yet to be established. METHODS Postnatal 7-day (P7) rats were subjected to a 3-h transient occlusion of the middle cerebral artery (MCA) which was produced either by inserting a filament via the external carotid artery or via the internal carotid artery. MRI was used to delineate the size and pattern of injury acutely, during MCA occlusion, and 7 days following reperfusion. RESULTS The size of the diffusion-weighted (DW) MRI-detectable injury during MCA occlusion was similar following both surgical procedures and resulted in histological lesions 7 days later in all animals. The extent of spontaneous recovery in individual animals varied substantially 7 days later within each group, as was depicted by a combination of DW- and T2W-MRI and confirmed by the corresponding histology. CONCLUSIONS The ability of MRI to provide accurate information on the size of histological outcome at 7 days after neonatal focal transient ischemia suggests that MRI is useful as an intermediate surrogate measure of injury progression in long-term neonatal stroke studies.
Collapse
Affiliation(s)
- Nikita Derugin
- Department of Neurosurgery, University of California, San Francisco, CA 94143, USA
| | | | | | | | | | | |
Collapse
|
118
|
Affiliation(s)
- Michael V Johnston
- Department of Neurology, Kennedy Krieger Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|
119
|
Northington FJ, Graham EM, Martin LJ. Apoptosis in perinatal hypoxic-ischemic brain injury: how important is it and should it be inhibited? ACTA ACUST UNITED AC 2005; 50:244-57. [PMID: 16216332 DOI: 10.1016/j.brainresrev.2005.07.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Revised: 07/12/2005] [Accepted: 07/14/2005] [Indexed: 01/21/2023]
Abstract
The discovery of safe and effective therapies for perinatal hypoxia-ischemia (HI) and stroke remains an unmet goal of perinatal medicine. Hypothermia and antioxidants such as allopurinol are currently under investigation as treatments for neonatal HI. Drugs targeting apoptotic mechanisms are currently being studied in adult diseases such as cancer, stroke, and trauma and have been proposed as potential therapies for perinatal HI and stroke. Before developing antiapoptosis therapies for perinatal brain injury, we must determine whether this form of cell death plays an important role in these injuries and if the inhibition of these pathways promotes more benefit than harm. This review summarizes current evidence for apoptotic mechanisms in perinatal brain injury and addresses issues pertinent to the development of antiapoptosis therapies for perinatal HI and stroke.
Collapse
Affiliation(s)
- Frances J Northington
- Department of Pediatrics, Eudowood Neonatal Pulmonary Division, Dept. of Pediatrics, CMSC 6-104, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA.
| | | | | |
Collapse
|
120
|
Pabello NG, Tracy SJ, Snyder-Keller A, Keller RW. Regional expression of constitutive and inducible transcription factors following transient focal ischemia in the neonatal rat: influence of hypothermia. Brain Res 2005; 1038:11-21. [PMID: 15748868 DOI: 10.1016/j.brainres.2004.12.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2004] [Revised: 12/13/2004] [Accepted: 12/17/2004] [Indexed: 10/25/2022]
Abstract
Ischemia is a potent modulator of gene expression. Differential expression of transcription factors after focal ischemia may reflect the potential for neuronal recovery in peri-ischemic regions. Previously, we demonstrated that hypothermia reduces the volume of damage in a model of neonatal focal ischemia. In the present study, immunocytochemistry was used to assess the temporal and spatial profiles of the transcription factors Fos and pCREB under normal and hypothermic conditions in this neonatal model of focal ischemia. At 7 days of age, rat pups underwent a permanent middle cerebral artery occlusion (MCAo) coupled with a temporary 1-h occlusion of the common carotid artery (CCAo). They were maintained at 37 degrees C throughout ischemia and reperfusion (Normothermic), or given 1 h of hypothermic conditions (28 degrees C) either during the occlusion (Intraischemic Hypothermia) or during the second hour of reperfusion (postischemic hypothermia). In normothermic pups, Fos immunoreactivity peaked at early time points (4-8 h post-ischemia) in a narrow band in peri-ischemic regions. By later stages of reperfusion (12-24 h), there was a more widespread induction in peri-ischemic regions including the ipsilateral cortex. In contrast with Fos, the constitutive transcription factor pCREB was reduced in core regions at all time points examined. Both the c-fos induction in peri-ischemic regions and the reduction of pCREB in the core were attenuated by intraischemic hypothermia. Postischemic hypothermia altered the distribution of Fos immunoreactivity without significantly changing the number of Fos- and pCREB-immunoreactive cells compared to normothermic rats. Both intra- and postischemic hypothermia reduced the number of caspase-immunoreactive cells. Thus, focal ischemia in the P7 rat produces different distributions of Fos and pCREB than what has been observed in adult rats subjected to focal ischemia, and expression of these transcription factors can be altered by hypothermia.
Collapse
Affiliation(s)
- Nina G Pabello
- Center for Neuropharmacology and Neuroscience, MC-136, Albany Medical College, Albany, NY 12208, USA
| | | | | | | |
Collapse
|
121
|
Fox C, Dingman A, Derugin N, Wendland MF, Manabat C, Ji S, Ferriero DM, Vexler ZS. Minocycline confers early but transient protection in the immature brain following focal cerebral ischemia-reperfusion. J Cereb Blood Flow Metab 2005; 25:1138-49. [PMID: 15874975 PMCID: PMC2262097 DOI: 10.1038/sj.jcbfm.9600121] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The incidence of neonatal stroke is high and currently there are no strategies to protect the neonatal brain from stroke or reduce the sequelae. Agents capable of modifying inflammatory processes hold promise. We set out to determine whether delayed administration of one such agent, minocycline, protects the immature brain in a model of transient middle cerebral artery (MCA) occlusion in 7-day-old rat pups. Injury volume in minocycline (45 mg/kg/dose, beginning at 2 h after MCA occlusion) and vehicle-treated pups was determined 24 h and 7 days after onset of reperfusion. Accumulation of activated microglia/macrophages, phosphorylation of mitogen-activated protein kinase (MAPK) p38 in the brain, and concentrations of inflammatory mediators in plasma and brain were determined at 24 h. Minocycline significantly reduced the volume of injury at 24 h but not 7 days after transient MCA occlusion. The beneficial effect of minocycline acutely after reperfusion was not associated with changed ED1 phenotype, nor was the pattern of MAPK p38 phosphorylation altered. Minocycline reduced accumulation of IL-1beta and CINC-1 in the systemic circulation but failed to affect the increased levels of IL-1beta, IL-18, MCP-1 or CINC-1 in the injured brain tissue. Therefore, minocycline provides early but transient protection, which is largely independent of microglial activation or activation of the MAPK p38 pathway.
Collapse
Affiliation(s)
- Christine Fox
- Department of Neurology, University of California, San Francisco, California, USA
| | - Andra Dingman
- Department of Neurology, University of California, San Francisco, California, USA
| | - Nikita Derugin
- Department of Neurosurgery, University of California, San Francisco, California, USA
| | - Michael F Wendland
- Department of Radiology, University of California, San Francisco, California, USA
| | - Catherine Manabat
- Department of Neurology, University of California, San Francisco, California, USA
| | - Shaoquan Ji
- Linco Research, Inc., St. Charles, Missouri, USA
| | - Donna M Ferriero
- Department of Neurology, University of California, San Francisco, California, USA
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - Zinaida S Vexler
- Department of Neurology, University of California, San Francisco, California, USA
| |
Collapse
|
122
|
Chang YS, Mu D, Wendland M, Sheldon RA, Vexler ZS, McQuillen PS, Ferriero DM. Erythropoietin improves functional and histological outcome in neonatal stroke. Pediatr Res 2005; 58:106-11. [PMID: 15879287 DOI: 10.1203/01.pdr.0000163616.89767.69] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neonatal stroke is a condition that leads to disability in later life, and as yet there is no effective treatment. Recently, erythropoietin (EPO) has been shown to be cytoprotective following brain injury and may promote neurogenesis. However, the effect of EPO on functional outcome and on morphologic changes in neonatal subventricular zone (SVZ) following experimental neonatal stroke has not been described. We used a transient focal model of neonatal stroke in P10 rat. Injury was documented by diffusion weighted MRI during occlusion. Immediately upon reperfusion, either EPO (5U/gm) or vehicle was administered intraperitoneally and animals were allowed to grow for 2 wk. Sensorimotor function was assessed using the cylinder rearing test and then brains were processed for volumetric analysis of the SVZ. Stroke induced SVZ expansion proportional to hemispheric volume loss. EPO treatment markedly preserved hemispheric volume and decreased the expansion of SVZ unilaterally. Furthermore, EPO treatment significantly improved the asymmetry of forelimb use following neonatal stroke. This functional improvement directly correlated with the amount of preserved hemispheric volume. These results suggest EPO may be a candidate in the treatment of neonatal stroke.
Collapse
Affiliation(s)
- Yun Sil Chang
- Departments of Neurology and Pediatrics, University of California-San Francisco, 521 Parnassus Avenue, San Francisco, CA 94143-0663, USA
| | | | | | | | | | | | | |
Collapse
|
123
|
Meng S, Qiao M, Foniok T, Tuor UI. White matter damage precedes that in gray matter despite similar magnetic resonance imaging changes following cerebral hypoxia-ischemia in neonatal rats. Exp Brain Res 2005; 166:56-60. [PMID: 15968456 DOI: 10.1007/s00221-005-2340-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2004] [Accepted: 08/13/2004] [Indexed: 10/25/2022]
Abstract
We hypothesized that the cerebral injury produced by hypoxia-ischemia (HI) in neonatal rats would differ in white compared with gray matter as detected histologically or with magnetic resonance (MR) imaging methods. Maps of T2 and the apparent diffusion coefficient (ADC) of water were acquired in 1-week-old rats at times prior to cerebral HI (right carotid artery occlusion plus 1.5 h of hypoxia), within the last 5-10 min of HI, and 1 h or 24 h after HI. Near the end of HI, ADC decreased and T2 increased in both cortical gray and subcortical white matter within the cingulum of the HI hemisphere. One hour after HI, ADC partially recovered, but T2 remained increased and then increased further by 24 h post-HI. In contrast to the similar MR responses in white and gray matter, histological evidence for irreversible cell damage occurred in white matter earlier than in gray matter within the HI hemisphere. At 1 h post-HI, rarefied or disrupted nerve fibers and an increase in TUNEL-positive cells were observed within white matter in the cingulum, whereas neurons within the cortical gray matter appeared normal. By 24 h post-HI, damage was apparent in both white and gray matter. Thus, MR imaging detected acute tissue edema following cerebral HI in both gray and white matter but did not distinguish between the early irreversible tissue injury detected histologically in white but not gray matter in this rather severe model of neonatal encephalopathy.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Newborn
- Brain Edema/etiology
- Brain Edema/pathology
- Brain Edema/physiopathology
- Cell Count
- Cell Death/physiology
- Cerebral Cortex/pathology
- Cerebral Cortex/physiopathology
- Cerebral Infarction/pathology
- Cerebral Infarction/physiopathology
- Diffusion
- Disease Models, Animal
- Humans
- Hypoxia-Ischemia, Brain/pathology
- Hypoxia-Ischemia, Brain/physiopathology
- In Situ Nick-End Labeling
- Infant, Newborn
- Leukomalacia, Periventricular/pathology
- Leukomalacia, Periventricular/physiopathology
- Nerve Degeneration/etiology
- Nerve Degeneration/pathology
- Nerve Degeneration/physiopathology
- Nerve Fibers, Myelinated/metabolism
- Nerve Fibers, Myelinated/pathology
- Rats
- Rats, Wistar
Collapse
Affiliation(s)
- Shuzhen Meng
- Institute for Biodiagnostics (West), National Research Council of Canada, 3330 Hospital Drive, NW Calgary, AB, T2N 4N1, Canada
| | | | | | | |
Collapse
|
124
|
Karamlou T, Hickey E, Silliman CC, Shen I, Ungerleider RM. Reducing risk in infant cardiopulmonary bypass: the use of a miniaturized circuit and a crystalloid prime improves cardiopulmonary function and increases cerebral blood flow. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu 2005:3-11. [PMID: 15818352 DOI: 10.1053/j.pcsu.2005.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Advances in perfusion strategies have played an important role in improving outcomes following repair of complex congenital heart defects. The influence of cooling strategy, temperature, duration of circulatory arrest, and specific method of cerebral perfusion on neurologic morbidity have been extensively characterized. Similarly, the ability of pharmacologic agents to modulate the post-cardiopulmonary bypass (CPB) inflammatory response has been previously elucidated in both the laboratory and clinical arena. However, modification of the circuit and priming components have received comparably less attention. We recently showed that employment of a miniaturized circuit and a bloodless prime reduce inflammation and have salutary effects on cardiopulmonary function following hypothermic low-flow perfusion (HLF), and that this circuit may also improve cerebral protection following both deep hypothermic circulatory arrest and HLF. The current report, therefore, reviews current strategies utilized to minimize post-CPB inflammation and highlights the empirical evidence from our laboratory demonstrating the beneficial role of a miniaturized extracorporeal circuit in this context.
Collapse
Affiliation(s)
- Tara Karamlou
- Division of Cardiovascular Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
125
|
Kuo YM, Duncan JL, Westaway SK, Yang H, Nune G, Xu EY, Hayflick SJ, Gitschier J. Deficiency of pantothenate kinase 2 (Pank2) in mice leads to retinal degeneration and azoospermia. Hum Mol Genet 2004; 14:49-57. [PMID: 15525657 PMCID: PMC2117329 DOI: 10.1093/hmg/ddi005] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pantothenate kinase-associated neurodegeneration (PKAN, formerly known as Hallervorden-Spatz syndrome) is a rare but devastating neurodegenerative disorder, resulting from an inherited defect in coenzyme A biosynthesis. As pathology in the human condition is limited to the central nervous system, specifically the retina and globus pallidus, we have generated a mouse knock-out of the orthologous murine gene (Pank2) to enhance our understanding of the mechanisms of disease and to serve as a testing ground for therapies. Over time, the homozygous null mice manifest retinal degeneration, as evidenced by electroretinography, light microscopy and pupillometry response. Specifically, Pank2 mice show progressive photoreceptor decline, with significantly lower scotopic a- and b-wave amplitudes, decreased cell number and disruption of the outer segment and reduced pupillary constriction response when compared with those of wild-type littermates. Additionally, the homozygous male mutants are infertile due to azoospermia, a condition that was not appreciated in the human. Arrest occurs in spermiogenesis, with complete absence of elongated and mature spermatids. In contrast to the human, however, no changes were observed in the basal ganglia by MRI or by histological exam, nor were there signs of dystonia, even after following the mice for one year. Pank2 mice are 20% decreased in weight when compared with their wild-type littermates; however, dysphagia was not apparent. Immunohistochemistry shows staining consistent with localization of Pank2 to the mitochondria in both the retina and the spermatozoa.
Collapse
Affiliation(s)
- Yien-Ming Kuo
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Jacque L. Duncan
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - Shawn K. Westaway
- Department of Molecular and Medical Genetics, Oregon Health and Science University, OR, USA
| | - Haidong Yang
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - George Nune
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - Eugene Yujun Xu
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Susan J. Hayflick
- Department of Molecular and Medical Genetics, Oregon Health and Science University, OR, USA
| | - Jane Gitschier
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
- *To whom correspondence should be addressed at: HSE-901, PO Box 0794, University of California, San Francisco, CA 94143, USA. Tel: +1 4154768729; Fax: +1 4155020720;
| |
Collapse
|
126
|
Ren M, Leng Y, Jeong M, Leeds PR, Chuang DM. Valproic acid reduces brain damage induced by transient focal cerebral ischemia in rats: potential roles of histone deacetylase inhibition and heat shock protein induction. J Neurochem 2004; 89:1358-67. [PMID: 15189338 DOI: 10.1111/j.1471-4159.2004.02406.x] [Citation(s) in RCA: 302] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Growing evidence from in vitro studies supports that valproic acid (VPA), an anti-convulsant and mood-stabilizing drug, has neuroprotective effects. The present study investigated whether VPA reduces brain damage and improves functional outcome in a transient focal cerebral ischemia model of rats. Subcutaneous injection of VPA (300 mg/kg) immediately after ischemia followed by repeated injections every 12 h, was found to markedly decrease infarct size and reduce ischemia-induced neurological deficit scores measured at 24 and 48 h after ischemic onset. VPA treatment also suppressed ischemia-induced neuronal caspase-3 activation in the cerebral cortex. VPA treatments resulted in a time-dependent increase in acetylated histone H3 levels in the cortex and striatum of both ipsilateral and contralateral brain hemispheres of middle cerebral artery occlusion (MCAO) rats, as well as in these brain areas of normal, non-surgical rats, supporting the in vitro finding that VPA is a histone deacetylase (HDAC) inhibitor. Similarly, heat shock protein 70 (HSP70) levels were time-dependently up-regulated by VPA in the cortex and striatum of both ipsilateral and contralateral sides of MCAO rats and in these brain areas of normal rats. Altogether, our results demonstrate that VPA is neuroprotective in the cerebral ischemia model and suggest that the protection mechanisms may involve HDAC inhibition and HSP induction.
Collapse
Affiliation(s)
- Ming Ren
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, MSC 1363, Bethesda, MD 20892-1363, USA
| | | | | | | | | |
Collapse
|
127
|
Lee WT, Chang C. Magnetic resonance imaging and spectroscopy in assessing 3-nitropropionic acid-induced brain lesions: an animal model of Huntington’s disease. Prog Neurobiol 2004; 72:87-110. [PMID: 15063527 DOI: 10.1016/j.pneurobio.2004.02.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2003] [Accepted: 02/03/2004] [Indexed: 10/26/2022]
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disease, in which there is progressive motor and cognitive deterioration, and for which the pathogenesis of neuronal death remains controversial. Mitochondrial toxins like 3-nitropropionic acid (3-NP) and malonate, functioning as the inhibitors of the complex II of mitochondrial respiratory chain, have been found to effectively induce specific behavioral changes and selective striatal lesions in rats and non-human primates mimicking those in HD. Furthermore, several kinds of transgenic mouse models of HD have been recently developed, and used in the development and assessment of novel treatments for HD. In the past, most studies evaluating the animal models for HD were based on histological changes or in vitro neuronal cultures. With the emergence of advanced magnetic resonance technologies, non-invasive magnetic resonance imaging (MRI) and spectroscopy provide more detail of cerebral alterations, including the changes of cerebral structure, function and metabolites. These studies support the hypothesis that mitochondrial dysfunction with increased excitation of N-methyl-D-aspartate (NMDA) receptors can replicate the neurobehavioral changes, selective brain injury and neurochemical alterations in HD. The present review focuses on our work as well as that of others regarding 3-NP-induced neurotoxicity and other animal models of HD. Using both conventional and advanced MRI and spectroscopy, we summarize the pathogenesis and possible therapeutic strategies in chemical and transgenic models of HD. The results show magnetic resonance techniques to be powerful techniques in the evaluation of pathogenesis and therapeutic intervention for both chemical and transgenic models of HD.
Collapse
Affiliation(s)
- Wang-Tso Lee
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | | |
Collapse
|
128
|
Clinical trials for cytoprotection in stroke. Neurotherapeutics 2004. [DOI: 10.1007/bf03206567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
129
|
Abstract
To date, many cytoprotective drugs have reached the stage of pivotal phase 3 efficacy trials in acute stroke patients. (Table 1) Unfortunately, throughout the neuroprotective literature, the phrase "failure to demonstrate efficacy" prevails as a common thread among the many neutral or negative trials, despite the largely encouraging results encountered in preclinical studies. The reasons for this discrepancy are multiple, and have been discussed by Dr. Zivin in his review. Many of the recent trials have addressed deficiencies of the previous ones with more rigorous trial design, including more specific patient selection criteria (ensure homogeneity of stroke location and severity), stratified randomization algorithms (time-to-treat), narrowed therapeutic time-window and pharmacokinetic monitoring. Current trials have also incorporated biologic surrogate markers of toxicity and outcome such as drug levels and neuroimaging. Lastly, multi-modal therapies and coupled cytoprotection/reperfusion strategies are being investigated to optimize tissue salvage. This review will focus on individual therapeutic strategies and we will emphasize what we have learned from these trials both in terms of trial design and the biologic effect (or lack thereof) of these agents.
Collapse
Affiliation(s)
- Lise A Labiche
- Stroke Program, University of Texas at Houston Medical School, 6431 Fannin Street, Houston, Texas 77030, USA
| | | |
Collapse
|