151
|
What initiates the formation of senile plaques? The origin of Alzheimer-like dementias in capillary haemorrhages. Med Hypotheses 2008; 71:347-59. [DOI: 10.1016/j.mehy.2008.04.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Revised: 03/24/2008] [Accepted: 04/03/2008] [Indexed: 01/18/2023]
|
152
|
Yao SY, Soutto M, Sriram S. Bacterial cell wall products increases stabilization of HIF-1 alpha in an oligodendrocyte cell line preconditioned by cobalt chloride or desferrioxamine. J Neuroimmunol 2008; 200:17-26. [PMID: 18715655 DOI: 10.1016/j.jneuroim.2008.05.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Revised: 05/27/2008] [Accepted: 05/28/2008] [Indexed: 01/15/2023]
Abstract
We examined the effect of lipopolysaccharide (LPS) or lipotechoic acid (LTA) on the regulation of hypoxia inducible factor (HIF-1) alpha on the MO3.13 cells, a human oligodendroglial cell line. Our study shows that MO3.13 cells express the toll like receptors (TLR's) but do not increase cellular levels of HIF-1 alpha following exposure to bacterial cell wall products. When MO3.13 cells were preconditioned by desferrioxamine (DFO) or cobalt chloride (CoCl(2)) and then treated with either LPS or LTA, HIF-1 alpha levels were higher than that induced by DFO or CoCl(2) alone. The increase in HIF-1 alpha was due to increased protein stability that was mediated by activation of the ERK-MAP kinase pathway.
Collapse
Affiliation(s)
- Song-yi Yao
- Department of Neurology, Multiple Sclerosis Research Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, USA
| | | | | |
Collapse
|
153
|
Yenari M, Kitagawa K, Lyden P, Perez-Pinzon M. Metabolic downregulation: a key to successful neuroprotection? Stroke 2008; 39:2910-7. [PMID: 18658035 DOI: 10.1161/strokeaha.108.514471] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND PURPOSE The search for effective neuroprotectants remains frustrating, particularly with regard to specific pharmaceuticals. However, laboratory studies have consistently shown remarkable neuroprotection with 2 nonpharmacological strategies-therapeutic hypothermia and ischemic preconditioning. Recent studies have shown that the mechanism of protection underlying both of these treatments is correlated to downregulation of cellular and tissue metabolism. Thus, understanding the mechanisms underlying such robust protective effects could lead to appropriate translation at the clinical level. In fact, hypothermia is already being used at many centers to improve neurological outcome from cardiac arrest. METHODS A systematic review of both topics is presented in terms of underlying pathophysiological mechanisms and application at the clinical level. RESULTS Although the mechanisms of protection for both therapeutic strategies are multifold, both share features of downregulating metabolism. Both therapeutic strategies are robust neuroprotectants, but translating them to the clinical arena is challenging, though not impossible, and clinical studies have shown or suggest benefits of both treatments. CONCLUSIONS The strategy of metabolic downregulation should be further explored to identify effective neuroprotectants that can be easily applied clinically.
Collapse
Affiliation(s)
- Midori Yenari
- Department of Neurology, University of California, San Francisco, San Francisco Veterans Affairs Medical Center, San , San Francisco, CA 94121, USA.
| | | | | | | |
Collapse
|
154
|
HIF-1alpha inhibition ameliorates neonatal brain injury in a rat pup hypoxic-ischemic model. Neurobiol Dis 2008; 31:433-41. [PMID: 18602008 DOI: 10.1016/j.nbd.2008.05.020] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 05/17/2008] [Accepted: 05/28/2008] [Indexed: 01/03/2023] Open
Abstract
Hypoxia-inducible factor-1alpha (HIF-1alpha) has been considered as a regulator of both prosurvival and prodeath pathways in the nervous system. The present study was designed to elucidate the role of HIF-1alpha in neonatal hypoxic-ischemic (HI) brain injury. Rice-Vannucci model of neonatal hypoxic-ischemic brain injury was used in seven-day-old rats, by subjecting unilateral carotid artery ligation followed by 2 h of hypoxia (8% O2 at 37 degrees C). HIF-1alpha activity was inhibited by 2-methoxyestradiol (2ME2) and enhanced by dimethyloxalylglycine (DMOG). Results showed that 2ME2 exhibited dose-dependent neuroprotection by decreasing infarct volume and reducing brain edema at 48 h post HI. The neuroprotection was lost when 2ME2 was administered 3 h post HI. HIF-1alpha upregulation by DMOG increased the permeability of the BBB and brain edema compared with HI group. 2ME2 attenuated the increase in HIF-1alpha and VEGF 24 h after HI. 2ME2 also had a long-term effect of protecting against the loss of brain tissue. The study showed that the early inhibition of HIF-1alpha acutely after injury provided neuroprotection after neonatal hypoxia-ischemia which was associated with preservation of BBB integrity, attenuation of brain edema, and neuronal death.
Collapse
|
155
|
Beck H, Semisch M, Culmsee C, Plesnila N, Hatzopoulos AK. Egr-1 regulates expression of the glial scar component phosphacan in astrocytes after experimental stroke. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:77-92. [PMID: 18556777 DOI: 10.2353/ajpath.2008.070648] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Ischemic brain injury causes tissue damage and neuronal death. The deficits can often be permanent because adult neurons fail to regenerate. One barrier to neuronal regeneration is the formation of the glial scar, a repair mechanism that is otherwise necessary to seal off necrotic areas. The process of gliosis has been well described, but the mechanisms regulating the robust production of scar components after injury remain poorly understood. Here we show that the early growth response 1 transcriptional factor (Egr-1, also called Krox24, Zif268, and NGFI-A) is expressed in astrocytes in the ventricular wall, corpus callosum, and striatum of normal mouse brain. After experimental stroke caused by permanent occlusion of the middle cerebral artery, Egr-1 was expressed long term in reactive astrocytes that accumulate around the injury site. Gain- and loss-of-function studies in primary astrocytes indicated that Egr-1 regulates the transcription of chondroitin sulfate proteoglycans genes, the main extracellular matrix proteins of the glial scar. Egr-1 bound to a site within the phosphacan promoter and transactivated its expression. Egr-1-deficient mice accumulated lower levels of phosphacan RNA and protein than wild-type mice after stroke, but there were no measurable differences in neurite outgrowth toward the infarct area between the two groups. Our findings suggest that Egr-1 is an important component of the transcriptional network regulating genes involved in gliosis after ischemic injury.
Collapse
Affiliation(s)
- Heike Beck
- Institute for Clinical Molecular Biology and Tumor Genetics, German Research Center for Environmental Health, Helmholtz Center Munich, Munich, Germany
| | | | | | | | | |
Collapse
|
156
|
Jung ME, Simpkins JW, Wilson AM, Downey HF, Mallet RT. Intermittent hypoxia conditioning prevents behavioral deficit and brain oxidative stress in ethanol-withdrawn rats. J Appl Physiol (1985) 2008; 105:510-7. [PMID: 18499779 DOI: 10.1152/japplphysiol.90317.2008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intermittent hypoxia (IH) has been found to protect brain from ischemic injury. We investigated whether IH mitigates brain oxidative stress and behavioral deficits in rats subjected to ethanol intoxication and abrupt ethanol withdrawal (EW). The effects of IH on overt EW behavioral signs, superoxide generation, protein oxidation, and mitochondrial permeability transition pore (PTP) opening were examined. Male rats consumed dextrin or 6.5% (wt/vol) ethanol for 35 days. During the last 20 days, rats were treated with repetitive (5-8 per day), brief (5-10 min) cycles of hypoxia (9.5-10% inspired O2) separated by 4-min normoxia exposures. Cerebellum, cortex, and hippocampus were biopsied on day 35 of the diet or at 24 h of EW. Superoxide and protein carbonyl contents in tissue homogenates and absorbance decline at 540 nm in mitochondrial suspensions served as indicators of oxidative stress, protein oxidation, and PTP opening, respectively. Although IH altered neither ethanol consumption nor blood ethanol concentration, it sharply lowered the severity of EW signs including tremor, tail rigidity, and startle response. Compared with dextrin and ethanol per se, in the three brain regions, EW increased superoxide and protein carbonyl contents and accelerated PTP opening in a manner ameliorated by IH. Administration of antioxidant N-acetylcysteine throughout the IH program abrogated the reductions in EW signs and superoxide content, implicating IH-induced ROS as mediators of the salutary adaptations. We conclude that IH conditioning during chronic ethanol consumption attenuates oxidative damage to the brain and mitigates behavioral abnormalities during subsequent EW. IH-induced ROS may evoke this powerful protection.
Collapse
Affiliation(s)
- Marianna E Jung
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107-2699, USA
| | | | | | | | | |
Collapse
|
157
|
Carp JS, Tennissen AM, Mongeluzi DL, Dudek CJ, Chen XY, Wolpaw JR. An in vitro protocol for recording from spinal motoneurons of adult rats. J Neurophysiol 2008; 100:474-81. [PMID: 18463177 DOI: 10.1152/jn.90422.2008] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
In vitro slice preparations of CNS tissue are invaluable for studying neuronal function. However, up to now, slice protocols for adult mammal spinal motoneurons--the final common pathway for motor behaviors--have been available for only limited portions of the spinal cord. In most cases, these preparations have not been productive due to the poor viability of motoneurons in vitro. This report describes and validates a new slice protocol that for the first time provides reliable intracellular recordings from lumbar motoneurons of adult rats. The key features of this protocol are: preexposure to 100% oxygen; laminectomy prior to perfusion; anesthesia with ketamine/xylazine; embedding the spinal cord in agar prior to slicing; and, most important, brief incubation of spinal cord slices in a 30% solution of polyethylene glycol to promote resealing of the many motoneuron dendrites cut during sectioning. Together, these new features produce successful recordings in 76% of the experiments and an average action potential amplitude of 76 mV. Motoneuron properties measured in this new slice preparation (i.e., voltage and current thresholds for action potential initiation, input resistance, afterhyperpolarization size and duration, and onset and offset firing rates during current ramps) are comparable to those recorded in vivo. Given the mechanical stability and precise control over the extracellular environment afforded by an in vitro preparation, this new protocol can greatly facilitate electrophysiological and pharmacological study of these uniquely important neurons and other delicate neuronal populations in adult mammals.
Collapse
Affiliation(s)
- Jonathan S Carp
- Wadsworth Center, New York State Department of Health, Albany, New York 12201-0509, USA.
| | | | | | | | | | | |
Collapse
|
158
|
Neuroprotective properties and mechanisms of erythropoietin in in vitro and in vivo experimental models for hypoxia/ischemia. ACTA ACUST UNITED AC 2008; 59:22-33. [PMID: 18514916 DOI: 10.1016/j.brainresrev.2008.04.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Revised: 04/23/2008] [Accepted: 04/24/2008] [Indexed: 11/22/2022]
Abstract
Besides its established function in erythropoiesis, erythropoietin (EPO) is currently also appreciated for its neuroprotective effects. The detrimental sequelae of prolonged cerebral hypoxia and ischemia have been shown to attenuate by EPO treatment. After binding to the EPO receptor, EPO is capable of initiating a cascade of events which--via different pathways--may lead to neuroprotection. The circumstances that determine which specific signalling route(s) are activated by EPO are largely unknown. We aim to provide the reader with a timely overview on the use of EPO in models of stroke and hypoxia-ischemia and to discuss the molecular events that underlie its neuroprotection.
Collapse
|
159
|
Kobayashi MS, Asai S, Ishikawa K, Nishida Y, Nagata T, Takahashi Y. Global profiling of influence of intra-ischemic brain temperature on gene expression in rat brain. ACTA ACUST UNITED AC 2008; 58:171-91. [PMID: 18440647 DOI: 10.1016/j.brainresrev.2008.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2007] [Revised: 02/08/2008] [Accepted: 03/08/2008] [Indexed: 12/20/2022]
Abstract
Mild to moderate differences in brain temperature are known to greatly affect the outcome of cerebral ischemia. The impact of brain temperature on ischemic disorders has been mainly evaluated through pathological analysis. However, no comprehensive analyses have been conducted at the gene expression level. Using a high-density oligonucleotide microarray, we screened 24000 genes in the hippocampus under hypothermic (32 degrees C), normothermic (37 degrees C), and hyperthermic (39 degrees C) conditions in a rat ischemia-reperfusion model. When the ischemic group at each intra-ischemic brain temperature was compared to a sham-operated control group, genes whose expression levels changed more than three-fold with statistical significance could be detected. In our screening condition, thirty-three genes (some of them novel) were obtained after screening, and extensive functional surveys and literature reviews were subsequently performed. In the hypothermic condition, many neuroprotective factor genes were obtained, whereas cell death- and cell damage-associated genes were detected as the brain temperature increased. At all intra-ischemic brain temperatures, multiple molecular chaperone genes were obtained. The finding that intra-ischemic brain temperature affects the expression level of many genes related to neuroprotection or neurotoxicity coincides with the different pathological outcomes at different brain temperatures, demonstrating the utility of the genetic approach.
Collapse
Affiliation(s)
- Megumi Sugahara Kobayashi
- Division of Genomic Epidemiology and Clinical Trials, Advanced Medical Research Center, Nihon University School of Medicine, Oyaguchi-Kami Machi, Itabashi-ku, Tokyo 173-8610, Japan
| | | | | | | | | | | |
Collapse
|
160
|
Dirnagl U, Meisel A. Endogenous neuroprotection: mitochondria as gateways to cerebral preconditioning? Neuropharmacology 2008; 55:334-44. [PMID: 18402985 DOI: 10.1016/j.neuropharm.2008.02.017] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2007] [Revised: 02/24/2008] [Accepted: 02/26/2008] [Indexed: 01/06/2023]
Abstract
From single to multicellular organisms, protective mechanisms have evolved against endogenous and exogenous noxious stimuli. Preconditioning paradigms, in which stimulation below the threshold of injury results in subsequent protection of the brain, have played an important role in elucidating such endogenous protective mechanisms. Consequently, over the past decades numerous signaling pathways have been discovered by which the brain senses and reacts to such insults as neurotoxins, substrate deprivation, or inflammation. Research on preconditioning is aimed at understanding endogenous neuroprotection to boost it, or to supplement its effectors therapeutically once damage to the brain has occurred, such as after stroke or brain trauma. Another goal of establishing preconditioning protocols is to induce endogenous neuroprotection in anticipation of incipient brain damage. Currently several endogenous neuroprotectants are being investigated in controlled clinical trials. In the present review we will give a short overview on the signals, sensors, transducers, and effectors of endogenous neuroprotection. We will first focus on common mechanisms, on which pathways of endogenous neuroprotection converge, and in particular on mitochondria, which may be considered master integrators of endogenous neuroprotection. We will then discuss various applications of preconditioning, including pharmacological and anesthetic preconditioning, as well as postconditioning, and explore the prospects of endogenous neuroprotective therapeutic approaches.
Collapse
Affiliation(s)
- Ulrich Dirnagl
- Department of Experimental Neurology, Center for Stroke Research Berlin, Berlin, Germany.
| | | |
Collapse
|
161
|
Gutsaeva DR, Carraway MS, Suliman HB, Demchenko IT, Shitara H, Yonekawa H, Piantadosi CA. Transient hypoxia stimulates mitochondrial biogenesis in brain subcortex by a neuronal nitric oxide synthase-dependent mechanism. J Neurosci 2008; 28:2015-24. [PMID: 18305236 PMCID: PMC6671843 DOI: 10.1523/jneurosci.5654-07.2008] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Accepted: 12/27/2007] [Indexed: 02/06/2023] Open
Abstract
The adaptive mechanisms that protect brain metabolism during and after hypoxia, for instance, during hypoxic preconditioning, are coordinated in part by nitric oxide (NO). We tested the hypothesis that acute transient hypoxia stimulates NO synthase (NOS)-activated mechanisms of mitochondrial biogenesis in the hypoxia-sensitive subcortex of wild-type (Wt) and neuronal NOS (nNOS) and endothelial NOS (eNOS)-deficient mice. Mice were exposed to hypobaric hypoxia for 6 h, and changes in immediate hypoxic transcriptional regulation of mitochondrial biogenesis was assessed in relation to mitochondrial DNA (mtDNA) content and mitochondrial density. There were no differences in cerebral blood flow or hippocampal PO2 responses to acute hypoxia among these strains of mice. In Wt mice, hypoxia increased mRNA levels for peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1 alpha), nuclear respiratory factor-1, and mitochondrial transcription factor A. After 24 h, new mitochondria, localized in reporter mice expressing mitochondrial green fluorescence protein, were seen primarily in hippocampal neurons. eNOS-/- mice displayed lower basal levels but maintained hypoxic induction of these transcripts. In contrast, nuclear transcriptional regulation of mitochondrial biogenesis in nNOS-/- mice was normal at baseline but did not respond to hypoxia. After hypoxia, subcortical mtDNA content increased in Wt and eNOS-/- mice but not in nNOS-/- mice. Hypoxia stimulated PGC-1alpha protein expression and phosphorylation of protein kinase A and cAMP response element binding (CREB) protein in Wt mice, but CREB only was activated in eNOS-/- mice and not in nNOS-/- mice. These findings demonstrate that hypoxic preconditioning elicits subcortical mitochondrial biogenesis by a novel mechanism that requires nNOS regulation of PGC-1alpha and CREB.
Collapse
Affiliation(s)
- Diana R. Gutsaeva
- Departments of Medicine, Anesthesiology, and Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina 27710
- Institute of Evolutionary Physiology and Biochemistry RAS, St. Petersburg 197376, Russia, and
| | - Martha Sue Carraway
- Departments of Medicine, Anesthesiology, and Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Hagir B. Suliman
- Departments of Medicine, Anesthesiology, and Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Ivan T. Demchenko
- Departments of Medicine, Anesthesiology, and Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina 27710
- Institute of Evolutionary Physiology and Biochemistry RAS, St. Petersburg 197376, Russia, and
| | - Hiroshi Shitara
- Department of Laboratory Animal Science, Tokyo Metropolitan Institute of Medical Science, Tokyo 113 8613, Japan
| | - Hiromichi Yonekawa
- Department of Laboratory Animal Science, Tokyo Metropolitan Institute of Medical Science, Tokyo 113 8613, Japan
| | - Claude A. Piantadosi
- Departments of Medicine, Anesthesiology, and Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
162
|
Goñi-Allo B, Puerta E, Ramos M, Lasheras B, Jordán J, Aguirre N. Minoxidil prevents 3,4-methylenedioxymethamphetamine-induced serotonin depletions: role of mitochondrial ATP-sensitive potassium channels, Akt and ERK. J Neurochem 2008; 104:914-25. [DOI: 10.1111/j.1471-4159.2007.05042.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
163
|
In vitro hypoxic preconditioning of embryonic stem cells as a strategy of promoting cell survival and functional benefits after transplantation into the ischemic rat brain. Exp Neurol 2008; 210:656-70. [PMID: 18279854 DOI: 10.1016/j.expneurol.2007.12.020] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 11/24/2007] [Accepted: 12/16/2007] [Indexed: 01/11/2023]
Abstract
Hypoxic preconditioning (HP) and stem cell transplantation have been extensively studied as individual therapies for ischemic stroke. The present investigation is an initial effort to combine these methods to achieve increased therapeutic effects after brain ischemia. Sublethal in vitro hypoxia pretreatment significantly enhanced the tolerance of neurally-differentiating embryonic stem (ES) cells and primary bone marrow mesenchymal stem cells (BMSC) to apoptotic cell death (40-50% reduction in cell death and caspase-3 activation). The HP protective effects on cultured cells lasted for at least 6 days. HP increased secretion of erythropoietin (EPO) and upregulated expression of bcl-2, hypoxia-inducible factor (HIF-1alpha), erythropoietin receptor (EPOR), neurofilament (NF), and synaptophysin in ES cell-derived neural progenitor cells (ES-NPCs). The HP cytoprotective effect was diminished by blocking EPOR, while pretreatment of ES-NPCs with recombinant human EPO mimicked the HP effect. HP-primed ES-NPCs survived better 3 days after transplantation into the ischemic brain (30-40% reduction in cell death and caspase-3 activation). Finally, transplanted HP-primed ES-NPCs exhibited extensive neuronal differentiation in the ischemic brain, accelerated and enhanced recovery of sensorimotor function when compared to transplantation of non-HP-treated ES-NPCs. The cell-priming strategy aimed to promote transplanted cell survival and their tissue repair capability provides a simple yet effective way of optimizing cell transplantation therapy.
Collapse
|
164
|
Pateliya BB, Singh N, Jaggi AS. Possible Role of Opioids and KATP Channels in Neuroprotective Effect of Postconditioning in Mice. Biol Pharm Bull 2008; 31:1755-60. [DOI: 10.1248/bpb.31.1755] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University
| | | |
Collapse
|
165
|
Ran R, Pan R, Lu A, Xu H, Davis RR, Sharp FR. A novel 165-kDa Golgin protein induced by brain ischemia and phosphorylated by Akt protects against apoptosis. Mol Cell Neurosci 2007; 36:392-407. [PMID: 17888676 DOI: 10.1016/j.mcn.2007.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 07/18/2007] [Accepted: 07/30/2007] [Indexed: 01/02/2023] Open
Abstract
A cDNA encoding a novel protein was cloned from ischemic rat brain and found to be homologous to testis Mea-2 Golgi-associated protein (Golga3). The sequence predicted a 165-kDa protein, and in vitro translated protein exhibited a molecular mass of 165-170 kDa. Because brain ischemia induced the mRNA, and the protein localized to the Golgi apparatus, this protein was designated Ischemia-Inducible Golgin Protein 165 (IIGP165). In HeLa cells, serum and glucose deprivation-induced caspase-dependent cleavage of the IIGP165 protein, after which the IIGP165 fragments translocated to the nucleus. The C-terminus of IIGP165, which contains a LXXLL motif, appears to function as a transcriptional co-regulator. Akt co-localizes with IIGP165 protein in the Golgi in vivo, and phosphorylates IIGP165 on serine residues 345 and 134. Though transfection of IIGP165 cDNA alone does not protect HeLa cells from serum deprivation or Brefeldin-A-triggered cell death, co-transfection of both Akt and IIGP165 cDNA or combined IIGP165-transfection with PDGF treatment significantly protects HeLa cells better than either treatment alone. These data show that Akt phosphorylation of IIGP165 protects against apoptotic cell death, and add to evidence that the Golgi apparatus also plays a role in regulating apoptosis.
Collapse
Affiliation(s)
- Ruiqiong Ran
- M.I.N.D. Institute and Department of Neurology, University of California at Davis Medical Center, University of California at Davis, Sacramento, CA 95817, USA.
| | | | | | | | | | | |
Collapse
|
166
|
Qutub AA, Popel AS. Three autocrine feedback loops determine HIF1 alpha expression in chronic hypoxia. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1511-25. [PMID: 17720260 PMCID: PMC2094118 DOI: 10.1016/j.bbamcr.2007.07.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 06/13/2007] [Accepted: 07/12/2007] [Indexed: 11/30/2022]
Abstract
Hypoxia occurs in cancer, prolonged exercise, and long-term ischemia with durations of several hours or more, and the hypoxia-inducible factor 1 (HIF1) pathway response to these conditions differs from responses to transient hypoxia. We used computational modeling, validated by experiments, to gain a quantitative, temporal understanding of the mechanisms driving HIF1 response. To test the hypothesis that HIF1 alpha protein levels during chronic hypoxia are tightly regulated by a series of molecular feedbacks, we took into account protein synthesis and product inhibition, and analyzed HIF1 system changes in response to hypoxic exposures beyond 3 to 4 h. We show how three autocrine feedback loops together regulate HIF 1 alpha hydroxylation in different microenvironments. Results demonstrate that prolyl hydroxylase, succinate and HIF1 alpha feedback determine intracellular HIF1 alpha levels over the course of hours to days. The model provides quantitative insight critical for characterizing molecular mechanisms underlying a cell's response to long-term hypoxia.
Collapse
Affiliation(s)
- Amina A Qutub
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, 613 Traylor Building, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | | |
Collapse
|
167
|
Baranova O, Miranda LF, Pichiule P, Dragatsis I, Johnson RS, Chavez JC. Neuron-specific inactivation of the hypoxia inducible factor 1 alpha increases brain injury in a mouse model of transient focal cerebral ischemia. J Neurosci 2007; 27:6320-32. [PMID: 17554006 PMCID: PMC6672155 DOI: 10.1523/jneurosci.0449-07.2007] [Citation(s) in RCA: 297] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In the present study, we show a biphasic activation of hypoxia inducible factor 1alpha (HIF-1) after stroke that lasts for up to 10 d, suggesting the involvement of the HIF pathway in several aspects of the pathophysiology of cerebral ischemia. We provide evidence that HIF-1-mediated responses have an overall beneficial role in the ischemic brain as indicated by increased tissue damage and reduced survival rate of mice with neuron-specific knockdown of HIF-1alpha that were subjected to transient focal cerebral ischemia. In addition, we demonstrated that drugs known to activate HIF-1 in cultured cells as well as in vivo had neuroprotective properties in this model of cerebral ischemia. This protective effect was significantly attenuated but not completely abolished in neuron-specific HIF-1alpha-deficient mice, suggesting that alternative mechanisms of neuroprotection are also implicated. Last, our study showed that hypoxia-induced tolerance to ischemia was preserved in neuron-specific HIF-1alpha-deficient mice, indicating that the neuroprotective effects of hypoxic preconditioning do not depend on neuronal HIF-1 activation.
Collapse
Affiliation(s)
- Oxana Baranova
- Burke Medical Research Institute, White Plains, New York 10605
| | - Luis F. Miranda
- Burke Medical Research Institute, White Plains, New York 10605
| | - Paola Pichiule
- Department of Pediatrics, Morgan Stanley Children's Hospital, Columbia University, New York, New York 10032
| | - Ioannis Dragatsis
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, Tennessee 38163, and
| | - Randall S. Johnson
- Molecular Biology Section, Division of Biology, University of California, San Diego, La Jolla, California 92093
| | - Juan C. Chavez
- Burke Medical Research Institute, White Plains, New York 10605
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, New York 10021
| |
Collapse
|
168
|
Mukerji SS, Katsman EA, Wilber C, Haner NA, Selman WR, Hall AK. Activin is a neuronal survival factor that is rapidly increased after transient cerebral ischemia and hypoxia in mice. J Cereb Blood Flow Metab 2007; 27:1161-72. [PMID: 17133227 DOI: 10.1038/sj.jcbfm.9600423] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
One approach for developing targeted stroke therapies is to identify the neuronal protective and destructive signaling pathways and gene expression that follow ischemic insult. In some neural injury models, the transforming growth factor-beta family member activin can provide neuroprotective effects in vivo and promote neuronal survival. This study tests if activin supports cortical neurons after ischemic challenge in vitro and if signals after cerebral ischemia involve activin in vivo. In a defined cell culture model that uses hydrogen peroxide (H(2)O(2))-free radical stress, activin addition maintained neuronal survival. H(2)O(2) treatment increased activin mRNA twofold in surviving cortical neurons, and inhibition of activin with neutralizing antibodies caused neuronal death. These data identify activin gene changes as a rapid response to oxidative stress, and indicate that endogenous activin acts as a protective factor for cortical neurons in vitro. Similarly, after transient focal cerebral ischemia in adult mice, activin mRNA increased at 1 and 4 h ipsilateral to the infarct but returned to control values at 24 h after reperfusion. Intracellular activated smad signals were detected in neurons adjacent to the infarct. Activin was also increased after 2 h of 11% hypoxia. Activin mRNA increased at 1 h but not 4 or 24 h after hypoxia, similar to the time course of erythropoietin and vascular endothelial growth factor induction. These findings identify activin as an early-regulated gene response to transient ischemia and hypoxia, and its function in cortical neuron survival during oxidative challenge provides a basis to test activin as a potential therapeutic in stroke injury.
Collapse
Affiliation(s)
- Shibani S Mukerji
- Department of Neuroscience, Case Western Reserve University School of Medicine and University Hospitals of Cleveland, Cleveland, Ohio 44106, USA
| | | | | | | | | | | |
Collapse
|
169
|
Shi LB, Huang JH, Han BS. Hypoxia inducible factor-1α mediates protective effects of ischemic preconditioning on ECV-304 endothelial cells. World J Gastroenterol 2007; 13:2369-73. [PMID: 17511040 PMCID: PMC4147150 DOI: 10.3748/wjg.v13.i16.2369] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether hypoxia inducible factor-1α (HIF-1α) is linked to the protective effects of ischemic preconditioning (IP) on sinusoidal endothelial cells against ischemia/reperfusion injury.
METHODS: Sinusoidal endothelial cell lines ECV-304 were cultured and divided into four groups: control group, cells were cultured in complete DMEM medium; cold anoxia/warm reoxygenation (A/R) group, cells were preserved in a 4°C UW solution in a mixture of 95% N2 and 5% CO2 for 24 h; anoxia-preconditioning (APC) group, cells were treated with 4 cycles of short anoxia and reoxygenation before prolonged anoxia-preconditioning treatment; and anoxia-preconditioning and hypoxia inducible factor-1α (HIF-1α) inhibitor (I-HIF-1) group, cells were pretreated with 5 μm of HIF-1α inhibitor NS398 in DMEM medium before subjected to the same treatment as group APC. After the anoxia treatment, each group was reoxygenated in a mixture of 95% air and 5% CO2 incubator for 6 h. Cytoprotections were evaluated by cell viabilities from Trypan blue, lactate dehydrogenase (LDH) release rates, and intracellular cell adhesion molecule-1 (ICAM-1) expressions. Expressions of HIF-1α mRNA and HIF-1α protein from each group were determined by the RT-PCR method and Western blotting, respectively.
RESULTS: Ischemia preconditioning increased cell viability, and reduced LDH release and ICAM-1 expressions. Ischemia preconditioning also upregulated the HIF-1α mRNA level and HIF-1α protein expression. However, all of these changes were reversed by HIF-1α inhibitor NS398.
CONCLUSION: Ischemia preconditioning effectively inhibited cold hypoxia/warm reoxygenation injury to endothelial cells, and the authors showed for the first time HIF-1α is causally linked to the protective effects of ischemic preconditioning on endothelial cells.
Collapse
Affiliation(s)
- Liu-Bin Shi
- Department of Organ Transplantation, Huashan Hospital, Fudan University, No.12, Wulumuqi Road, Shanghai 200040, China.
| | | | | |
Collapse
|
170
|
Liu XS, Zhang ZG, Zhang RL, Gregg SR, Meng H, Chopp M. Comparison of in vivo and in vitro gene expression profiles in subventricular zone neural progenitor cells from the adult mouse after middle cerebral artery occlusion. Neuroscience 2007; 146:1053-61. [PMID: 17428613 PMCID: PMC1942046 DOI: 10.1016/j.neuroscience.2007.02.056] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2006] [Revised: 01/25/2007] [Accepted: 02/22/2007] [Indexed: 11/28/2022]
Abstract
Stroke stimulates neurogenesis in the adult rodent brain. The molecules that mediate stroke-induced neurogenesis are not definitely known. Using microarrays containing approximately 400 known genes associated with stem cell and angiogenesis, we compared transcriptional profiles of subventricular zone (SVZ) tissue with cultured neural progenitor cells isolated from the SVZ 7 days after ischemic stroke in the adult mouse. In SVZ tissue, we found that stroke upregulated 58 genes which are involved in multiple signaling pathways during embryonic development, suggesting that stroke recaptures embryonic molecular signals. In neural progenitor cells cultured in growth medium, 23 gene expressions were increased after stroke and 8 of 23 genes overlapped with upregulated genes in stroke SVZ tissue. Expression alterations of selected genes were confirmed by real-time RT-PCR and immunohistochemistry. These in vivo and in vitro data provide new insight into the genetic program of adult SVZ neural progenitor cells after stroke and demonstrate gene expression differences between SVZ tissue and cultured SVZ cells.
Collapse
Affiliation(s)
- Xian Shuang Liu
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, MI 48202
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, MI 48202
| | - Rui Lan Zhang
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, MI 48202
| | - Sara R. Gregg
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, MI 48202
| | - He Meng
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, MI 48202
| | - Michael Chopp
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, MI 48202
- Department of Physics, Oakland University, Rochester, MI 48309
| |
Collapse
|
171
|
Lee HJ, Kim KS, Park IH, Kim SU. Human neural stem cells over-expressing VEGF provide neuroprotection, angiogenesis and functional recovery in mouse stroke model. PLoS One 2007; 2:e156. [PMID: 17225860 PMCID: PMC1764718 DOI: 10.1371/journal.pone.0000156] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Accepted: 11/27/2006] [Indexed: 12/27/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) is a lethal stroke type. As mortality approaches 50%, and current medical therapy against ICH shows only limited effectiveness, an alternative approach is required, such as stem cell-based cell therapy. Previously we have shown that intravenously transplanted human neural stem cells (NSCs) selectively migrate to the brain and induce behavioral recovery in rat ICH model, and that combined administration of NSCs and vascular endothelial growth factor (VEGF) results in improved structural and functional outcome from cerebral ischemia. Methods and Findings We postulated that human NSCs overexpressing VEGF transplanted into cerebral cortex overlying ICH lesion could provide improved survival of grafted NSCs, increased angiogenesis and behavioral recovery in mouse ICH model. ICH was induced in adult mice by unilateral injection of bacterial collagenase into striatum. HB1.F3.VEGF human NSC line produced an amount of VEGF four times higher than parental F3 cell line in vitro, and induced behavioral improvement and 2–3 fold increase in cell survival at two weeks and eight weeks post-transplantation. Conclusions Brain transplantation of F3 human NSCs over-expressing VEGF near ICH lesion sites provided differentiation and survival of grafted human NSCs and renewed angiogenesis of host brain and functional recovery of ICH animals. These results suggest a possible application of the human neural stem cell line, which is genetically modified to over-express VEGF, as a therapeutic agent for ICH-stroke.
Collapse
Affiliation(s)
- Hong J. Lee
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Korea
- College of Bioscience and Biotechnology, Korea University, Seoul, Korea
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, University of British Columbia, Vancouver, Canada
| | - Kwang S. Kim
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Korea
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, University of British Columbia, Vancouver, Canada
| | - In H. Park
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Seung U. Kim
- Brain Disease Research Center, Ajou University School of Medicine, Suwon, Korea
- Division of Neurology, Department of Medicine, University of British Columbia Hospital, University of British Columbia, Vancouver, Canada
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
172
|
Horowitz M. Heat acclimation and cross-tolerance against novel stressors: genomic–physiological linkage. PROGRESS IN BRAIN RESEARCH 2007; 162:373-92. [PMID: 17645928 DOI: 10.1016/s0079-6123(06)62018-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Heat acclimation (AC) is a "within lifetime" reversible phenotypic adaptation, enhancing thermotolerance and heat endurance via a transition to "efficient" cellular performance when acclimatory homeostasis is reached. An inseparable outcome of AC is the development of cross-tolerance (C-T) against novel stressors. This chapter focuses on central plasticity and the molecular-physiological linkage of acclimatory and C-T responses. A drop in temperature thresholds (T-Tsh) for activation of heat-dissipation mechanisms and an elevated T-Tsh for thermal injury development imply autonomic nervous system (ANS) and cytoprotective network involvement in these processes. During acclimation, the changes in T-Tsh for heat dissipation are biphasic. Initially T-Tsh drops, signifying the early autonomic response, and is associated with perturbed peripheral effector cellular performance. Pre-acclimation values return when acclimatory homeostasis is achieved. The changes in the ANS suggest that acclimatory plasticity involves molecular and cellular changes. These changes are manifested by the activation of central peripheral molecular networks and post-translational modifications. Sympathetic induction of elevated HSP 72 reservoirs, with faster heat shock response, is only one example of this. The global genomic response, detected using gene-chips and cluster analyses imply upregulation of genes encoding ion channels, pumps, and transporters (markers for neuronal excitability) in the hypothalamus at the onset of AC and down regulation of metabotrophic genes upon long term AC. Peripherally, the transcriptional program indicates a two-tier defense strategy. The immediate transient response is associated with the maintenance of DNA and cellular integrity. The sustained response correlates with long-lasting cytoprotective-signaling networks. C-T is recorded against cerebral hypoxia, hyperoxia, and traumatic brain injury. Using the highly developed ischemic/reperfused heart model as a baseline, it is evident that C-T stems via protective shared pathways developed with AC. These comprise constitutive elevation of HIF 1alpha and associated target pathways, HSPs, anti-apoptosis, and antioxidative pathways. Collectively the master regulators of AC and C-T are still enigmatic; however, cutting-edge investigative techniques, using a broad molecular approach, challenge current ideas, and the data accumulated will pinpoint novel pathways and provide new perspectives.
Collapse
Affiliation(s)
- Michal Horowitz
- Laboratory of Environmental Physiology, The Hebrew University, POB 12272, Jerusalem 91120, Israel.
| |
Collapse
|
173
|
Cerebral Ischemia: Molecular Mechanisms and Protective Therapies. Neurobiol Dis 2007. [DOI: 10.1016/b978-012088592-3/50028-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
174
|
Shein NA, Horowitz M, Shohami E. Heat acclimation: a unique model of physiologically mediated global preconditioning against traumatic brain injury. PROGRESS IN BRAIN RESEARCH 2007; 161:353-63. [PMID: 17618990 DOI: 10.1016/s0079-6123(06)61025-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sub-lethal exposure to practically any harmful stimulus has been shown to induce consequent protection against more severe stress. This preconditioning (PC) effect may be achieved by exposure to different stressors, indicating that the induction of tolerance involves activation of common protective pathways. Chronic exposure to moderate heat (heat acclimation, HA) is a unique PC model, since this global physiological adaptation, as opposed to discrete organ PC, has been shown to induce cross-tolerance against other stressors, including closed head injury (CHI). HA animals show accelerated functional recovery after injury which is accompanied by reduced secondary brain damage. However, the precise mechanisms underlying this phenomenon have not been thoroughly studied until recently. Here we will address the concept of PC, highlighting the unique properties of HA as a model which can be used for the study of endogenous protective pathways triggered by PC procedures. Several molecular mechanisms which are suggested to mediate HA-induced neuroprotection will also be discussed, bringing to light their potential contribution to the development of traumatic brain injury treatment strategies utilizing therapeutic augmentation of endogenous defense mechanisms.
Collapse
Affiliation(s)
- Na'ama A Shein
- Department of Pharmacology, Hebrew University, Jerusalem, Israel
| | | | | |
Collapse
|
175
|
Hu X, Rea HC, Wiktorowicz JE, Perez-Polo JR. Proteomic analysis of hypoxia/ischemia-induced alteration of cortical development and dopamine neurotransmission in neonatal rat. J Proteome Res 2006; 5:2396-404. [PMID: 16944952 PMCID: PMC3128998 DOI: 10.1021/pr060209x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Perinatal hypoxia/ischemia (HI) is a common cause of neurological deficits in children. Our goal was to elucidate the underlying mechanisms that contribute to the neurological sequelae of HI-induced brain injury. HI was induced by permanent ligation of the left carotid artery followed by 90 min of hypoxia (7.8% O2) in female P7 rats. A two-dimensional differential proteome analysis was used to assess changes in protein expression in cortex 2 h after HI. In total, 17 proteins reflecting a 2-fold or higher perturbation of expression after HI as compared to sham-treated pups were identified by mass spectrometry. Of the altered proteins, 14-3-3epsilon and TUC-2, both playing an important role in the development of the central nervous system, decrease after HI, consistent with an early disturbance of cortical development. Also affected, DARPP-32 and alpha-synuclein, two proteins important for dopamine neurotransmission, increased more than 2-fold 2 h after HI injury. The differential expression of these proteins was validated by individual Western blot assays. The expression of several metabolic enzymes and translational factors was also perturbed early after HI brain injury. These findings provide initial insights into the mechanisms underlying neurodegenerative events after HI and may allow for the rational design of therapeutic strategies that enhance neuronal adaptation and compensation after HI.
Collapse
Affiliation(s)
| | | | | | - J. Regino Perez-Polo
- Corresponding Author: J. R. Perez-Polo, Ph.D., Galveston, Texas, 77555-1072 USA. Telephone: 409-772-3668. Fax: 409-772-8028.
| |
Collapse
|
176
|
Van Hoecke M, Prigent-Tessier AS, Garnier PE, Bertrand NM, Filomenko R, Bettaieb A, Marie C, Beley AG. Evidence of HIF-1 functional binding activity to caspase-3 promoter after photothrombotic cerebral ischemia. Mol Cell Neurosci 2006; 34:40-7. [PMID: 17101276 DOI: 10.1016/j.mcn.2006.09.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Revised: 09/27/2006] [Accepted: 09/29/2006] [Indexed: 01/29/2023] Open
Abstract
Hypoxia-inducible factor 1 alpha (HIF-1alpha) is a transcription factor that was suggested in vitro to promote cell death by modulation of proapoptotic genes. In this report, we tested the hypothesis of an in vivo proapoptotic role of HIF-1alpha after an ischemic insult. For this purpose, HIF-1alpha and procaspase-3 mRNA and protein expressions were examined in rat brain subjected to 12- and 24-h permanent focal ischemia and the presence of an HIF-1 binding activity to the caspase-3 gene promoter was explored. The results showed that HIF-1alpha and procaspase-3 expressions increased with a similar pattern in response to ischemia. In addition, caspase-3 activation was observed in cells that express HIF-1alpha. Moreover, electrophoretic mobility assay revealed a specific HIF-1 binding activity to the caspase-3 gene promoter. Altogether the present data provide strong arguments for a causative relationship between HIF-1alpha and caspase-3 inductions through a functional binding activity to the caspase-3 gene promoter.
Collapse
MESH Headings
- Animals
- Binding Sites/genetics
- Caspase 3/genetics
- Caspase 3/metabolism
- Disease Models, Animal
- Enzyme Activation/genetics
- Gene Expression Regulation, Enzymologic/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Ischemia, Brain/genetics
- Hypoxia-Ischemia, Brain/metabolism
- Hypoxia-Ischemia, Brain/physiopathology
- Intracranial Thrombosis/genetics
- Intracranial Thrombosis/metabolism
- Intracranial Thrombosis/physiopathology
- Male
- Oxygen/metabolism
- Promoter Regions, Genetic/genetics
- Protein Binding/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Telencephalon/metabolism
- Telencephalon/physiopathology
Collapse
Affiliation(s)
- Michaël Van Hoecke
- Laboratoire de Pharmacodynamie et Physiologie Pharmaceutique, Faculté de Pharmacie, 7 boulevard Jeanne d'Arc, BP 87900, 21079 Dijon Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
177
|
Zhao H, Sapolsky RM, Steinberg GK. Interrupting reperfusion as a stroke therapy: ischemic postconditioning reduces infarct size after focal ischemia in rats. J Cereb Blood Flow Metab 2006; 26:1114-21. [PMID: 16736038 DOI: 10.1038/sj.jcbfm.9600348] [Citation(s) in RCA: 244] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cerebral ischemic preconditioning protects against stroke, but is clinically feasible only when the occurrence of stroke is predictable. Reperfusion plays a critical role in cerebral injury after stroke; we tested the hypothesis that interrupting reperfusion lessens ischemic injury. We found for the first time that such postconditioning with a series of mechanical interruptions of reperfusion significantly reduces ischemic damage. Focal ischemia was generated by permanent distal middle cerebral artery (MCA) occlusion plus transient bilateral common carotid artery (CCA) occlusion. After 30 secs of CCA reperfusion, ischemic postconditioning was performed by occluding CCAs for 10 secs, and then allowing for another two cycles of 30 secs of reperfusion and 10 secs of CCA occlusion. Infarct size was measured 2 days later. Cerebral blood flow (CBF) was measured in animals subjected to permanent MCA occlusion plus 15 mins of bilateral CCA occlusion, which demonstrates that postconditioning disturbed the early hyperemia immediately after reperfusion. Postconditioning dose dependently reduced infarct size in animals subjected to permanent MCA occlusion combined with 15, 30, and 60 mins of bilateral CCA occlusion, by reducing infarct size approximately 80%, 51%, and 17%, respectively. In addition, postconditioning blocked terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end labeling-positive staining, a marker of apoptosis, in the penumbra 2 days after stroke. Furthermore, in situ superoxide detection using hydroethidine suggested that postconditioning attenuated superoxide products during early reperfusion after stroke. In conclusion, postconditioning reduced infarct size, most plausibly by blocking apoptosis and free radical generation. With further study it may eventually be clinically applicable for stroke treatment.
Collapse
Affiliation(s)
- Heng Zhao
- Department of Neurosurgery, Stanford University, California 94305-5327, USA.
| | | | | |
Collapse
|
178
|
Abstract
Adaptation is one of physiology's fundamental tenets, operating not only at the level of species, as Darwin proposed, but also at the level of tissues, cells, molecules and, perhaps, genes. During recent years, stroke neurobiologists have advanced a considerable body of evidence supporting the hypothesis that, with experimental coaxing, the mammalian brain can adapt to injurious insults such as cerebral ischaemia to promote cell survival in the face of subsequent injury. Establishing this protective phenotype in response to stress depends on a coordinated response at the genomic, molecular, cellular and tissue levels. Here, I summarize our current understanding of how 'preconditioning' stimuli trigger a cerebroprotective state known as cerebral 'ischaemic tolerance'.
Collapse
Affiliation(s)
- Jeffrey M Gidday
- Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri 63110, USA.
| |
Collapse
|
179
|
Kaminski A, Kasch C, Zhang L, Kumar S, Sponholz C, Choi YH, Ma N, Liebold A, Ladilov Y, Steinhoff G, Stamm C. Endothelial nitric oxide synthase mediates protective effects of hypoxic preconditioning in lungs. Respir Physiol Neurobiol 2006; 155:280-5. [PMID: 16916627 DOI: 10.1016/j.resp.2006.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2005] [Revised: 06/16/2006] [Accepted: 06/19/2006] [Indexed: 02/01/2023]
Abstract
To elucidate the protective mechanism of whole-body hypoxic preconditioning (WHPC) on pulmonary ischemia-reperfusion injury focussing on nitric oxide synthases (NOS), mice were placed in a hypoxic chamber (FIO(2)=0.1) for 4h followed by 12h of normoxia. Then, pulmonary ischemia for 1h followed by 5h of reperfusion was performed by clamping the left hilum in vivo (I/R). WHPC protected WT mice from pulmonary leukocyte infiltration as assessed by myeloperoxidase (MPO) activity, associated with a mild further increase in endothelial permeability (Evans Blue extravasation). When all NOS isoforms were inhibited during WHPC by L-NAME, mortality and MPO activity after I/R markedly increased. To determine the responsible NOS isoform, quantitative RT-PCR was performed for eNOS and iNOS mRNA, showing that only eNOS was upregulated in response to WHPC. While eNOS total protein expression remained unchanged, the amount of phosphorylated eNOS also increased. The WHPC/IR experiments were then repeated with eNOS knockout mice. Here, we found that the protective effect of WHPC on pulmonary leukocyte sequestration was abrogated, and endothelial leakage was further exacerbated. We conclude that WHPC limits neutrophil sequestration via an eNOS-dependent mechanism, and that eNOS helps preserve endothelial permeability during hypoxia and I/R.
Collapse
Affiliation(s)
- A Kaminski
- Department of Cardiac Surgery, University of Rostock, Schillingallee 35, 18055 Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
180
|
Luh SP, Yang PC. Organ preconditioning: the past, current status, and related lung studies. J Zhejiang Univ Sci B 2006; 7:331-41. [PMID: 16615162 PMCID: PMC1462933 DOI: 10.1631/jzus.2006.b0331] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Accepted: 03/23/2006] [Indexed: 12/13/2022]
Abstract
Preconditioning (PC) has emerged as a powerful method for experimentally and clinically attenuating various types of organ injuries. In this paper related clinical and basic research issues on organ preconditioning issues were systemically reviewed. Since lung injuries, including ischemia-reperfusion and others, play important roles in many clinical results, including thromboembolism, trauma, thermal injury, hypovolemic and endotoxin shock, reimplantation response after organ transplantation, and many respiratory diseases in critical care. It is of interest to uncover methods, including the PCs, to protect the lung from the above injuries. However, related studies on pulmonary PC are relatively rare and still being developed, so we will review previous literature on experimental and clinical studies on pulmonary PC in the following paragraphs.
Collapse
Affiliation(s)
- Shi-ping Luh
- Department of Cardiothoracic Surgery, Taipei Tzu-Chi Medical University Hospital, Taiwan 231, China.
| | | |
Collapse
|
181
|
|
182
|
Williams JM, White CR, Chang MM, Injeti ER, Zhang L, Pearce WJ. Chronic hypoxic decreases in soluble guanylate cyclase protein and enzyme activity are age dependent in fetal and adult ovine carotid arteries. J Appl Physiol (1985) 2006; 100:1857-66. [PMID: 16469937 DOI: 10.1152/japplphysiol.00662.2005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The present study tests the hypothesis that chronic hypoxia enhances reactivity to nitric oxide (NO) through age-dependent increases in soluble guanylate cyclase (sGC) and protein kinase G (PKG) activity. In term fetal and adult ovine carotids, chronic hypoxia had no significant effect on mRNA levels for the beta1-subunit of sGC, but depressed sGC abundance by 16% in fetal and 50% in adult arteries, through possible depression of rates of mRNA translation (15% in fetal and 50% in adult) and/or increased protein turnover. Chronic hypoxia also depressed the catalytic activity of sGC, but only in fetal arteries (63%). Total sGC activity was reduced by chronic hypoxia in both fetal (69%) and adult (37%) carotid homogenates, but this effect was not observed in intact arteries when sGC activity was measured by timed accumulation of cGMP. In intact arteries treated with 300 microM 3-isobutyl-1-methylxanthine (IBMX), chronic hypoxia dramatically enhanced sGC activity in fetal (186%) but not adult (89%) arteries. This latter observation suggests that homogenization either removed an sGC activator, released an sGC inhibitor, or altered the phosphorylation state of the enzyme, resulting in reduced activity. In the absence of IBMX, chronic hypoxia had no significant effect on rates of cGMP accumulation. Chronic hypoxia also depressed the ability of the cGMP analog, 8-(p-chlorophenylthio)-cGMP, to promote vasorelaxation in both fetal (8%) and adult (12%) arteries. Together, these results emphasize the fact that intact and homogenized artery studies of sGC activity do not always yield equivalent results. The results further suggest that enhancement of reactivity to NO by chronic hypoxia must occur upstream of PKG and can only be possible if changes in cGMP occurred in functional compartments that afforded either temporal or chemical protection to the actions of phosphodiesterase. The range and age dependence of hypoxic effects observed also suggest that some responses to hypoxia must be compensatory and homeostatic, with reactivity to NO as the primary regulated variable.
Collapse
Affiliation(s)
- James M Williams
- Department of Physiology, Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | | | | | | | | | | |
Collapse
|
183
|
Ezquer ME, Valdez SR, Seltzer AM. Inflammatory responses of the substantia nigra after acute hypoxia in neonatal rats. Exp Neurol 2005; 197:391-8. [PMID: 16293246 DOI: 10.1016/j.expneurol.2005.10.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Revised: 09/29/2005] [Accepted: 10/08/2005] [Indexed: 11/29/2022]
Abstract
The neocortex and the striatum are the brain regions most known to be particularly vulnerable to acute insults like hypoxia or ischemia. In this work, we assess the possibility of cellular damage to the substantia nigra (SN) after hypoxia-reoxygenation in the new born rat. The aim of the present paper was to evaluate the expression of growth factor IGF-I, and growth factor binding proteins IGFBP-3 and IGFBP-5 genes and induction of NOS family members (nNOS, eNOS and iNOS) and TNF-alpha genes together with glia activation, in the SN at 5 and 48 h after severe hypoxia in the 7 day-old rat, a model for the term human fetus. At early time, while IGFs remain unchanged, we found a transient increase in eNOS and nNOS. Two days after the injury, nNOS expression remained high, iNOS and TNF-alpha increased and also GFAP protein expression was observed together with a profusion of reactive astrocytes distributed throughout the SN. This study on the acute effects of hypoxia on the developing brain provides additional insights into the vulnerability of the SN, a brain region involved in neurodegenerative pathologies.
Collapse
Affiliation(s)
- Marcelo E Ezquer
- IMBECU-CRICYT, Centro Regional de Investigaciones Cientificas y Tecnologicas, Mendoza 5500, Argentina
| | | | | |
Collapse
|
184
|
Affiliation(s)
- Seth Love
- Department of Neuropathology, University of Bristol Institute of Clinical Neuroscience
| |
Collapse
|