Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Neurol Res ; 31(9): 947-58, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19203442

ABSTRACT

UNLABELLED: Insulin is neuroprotective following transient global brain ischemia; however, the mechanisms by which insulin exerts its salutary effects remain unclear. OBJECTIVE: We assessed insulin's effect on the PI3K-Akt survival system and consequent modulation of the pro-apoptotic proteins Bim, Bad and FoxO3a. METHODS: We utilized rats subjected to 10 minutes of global brain ischemia, with or without insulin administered at the onset of reperfusion. RESULTS: In sham-operated animals, minimal pAkt immunofluorescence was detected in the CA1. Moreover, at 30 minute reperfusion, there was no change in pAkt in CA1 neurons. Single bolus high-dose insulin treatment resulted in an early increase in pAkt after 30 minutes, preservation of CA1 neurons to 14 days of reperfusion and preservation of spatial learning ability. Insulin treatment increased cytoplasmic and nuclear staining for pAkt in both CA1 and cortex. Insulin-induced Akt phosphorylation was suppressed by the PI3K inhibitor wortmannin. Neither reperfusion nor insulin induced any change in the phosphorylation or subcellular localization of FoxO3a, Bim or Bad. A single bolus of high-dose insulin reduced CA1 neuronal cell death and thus represents a potential therapeutic intervention for global brain ischemia. DISCUSSION: These results reveal that proximal elements of a known cell-survival pathway are triggered by high-dose insulin during early reperfusion. Insulin induces robust PI3K-dependent phosphorylation of Akt by 30 minute reperfusion and results in improvement of hippocampal structure and function. However, the Akt substrates FoxO3a, Bim and Bad do not undergo corresponding changes in phosphorylation or subcellular localization in this model of global brain ischemia. The downstream components of insulin-induced Akt survival signaling after transient global brain ischemia remain to be identified.


Subject(s)
Brain Ischemia/drug therapy , Insulin/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Animals , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Brain Ischemia/enzymology , Brain Ischemia/physiopathology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/physiopathology , Cell Survival/drug effects , Cell Survival/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Forkhead Box Protein O3 , Forkhead Transcription Factors/drug effects , Forkhead Transcription Factors/metabolism , Insulin/metabolism , Insulin/therapeutic use , Male , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Memory Disorders/drug therapy , Memory Disorders/enzymology , Memory Disorders/physiopathology , Neurons/enzymology , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Long-Evans , Reperfusion Injury/drug therapy , Reperfusion Injury/enzymology , Reperfusion Injury/physiopathology , Signal Transduction/drug effects , Signal Transduction/physiology , bcl-Associated Death Protein/drug effects , bcl-Associated Death Protein/metabolism
2.
J Neurochem ; 106(3): 1248-58, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18518905

ABSTRACT

The critical event of the intrinsic pathway of apoptosis following transient global brain ischemia is the release of cytochrome c from the mitochondria. In vitro studies have shown that insulin can signal specifically via phosphatidylinositol-3-OH-kinase (PI3-K) and Akt to prevent cytochrome c release. Therefore, insulin may exert its neuroprotective effects during brain reperfusion by blocking cytochrome c release. We hypothesized that insulin acts through PI3-K, Akt, and Bcl-2 family proteins to inhibit cytochrome c release following transient global brain ischemia. We found that a single bolus of insulin given immediately upon reperfusion inhibited cytochrome c release for at least 24 h, and produced a fivefold improvement in neuronal survival at 14 days. Moreover, insulin's ability to inhibit cytochrome c release was completely dependent on PI3-K signaling and insulin induces phosphorylation of Akt through PI3-K. In untreated animals, there was an increase in mitochondrial Bax at 6 h of reperfusion, and Bax binding to Bcl-X(L) was disrupted at the mitochondria. Insulin prevented both these events in a PI3-K-dependent manner. In summary, insulin regulates cytochrome c release through PI3-K likely by activating Akt, promoting the binding between Bax and Bcl-X(L), and by preventing Bax translocation to the mitochondria.


Subject(s)
Brain Ischemia/metabolism , Cytochromes c/antagonists & inhibitors , Insulin/physiology , Phosphatidylinositol 3-Kinases/physiology , Reperfusion Injury/metabolism , Signal Transduction/physiology , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Animals , Brain Ischemia/enzymology , Cytochromes c/metabolism , Insulin/therapeutic use , Male , Protein Binding/physiology , Rats , Rats, Long-Evans , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control
3.
Brain Res ; 1075(1): 36-47, 2006 Feb 23.
Article in English | MEDLINE | ID: mdl-16469303

ABSTRACT

To discover candidate genes in the pathogenesis of congenital hydrocephalus, gene arrays were utilized to analyze transcripts from the midbrain region of 5-day-old H-Tx rats; these animals develop hydrocephalus due to closure of their cerebral aqueduct between embryonic day 18 and post-natal day 5. Of the 15,924 transcripts assayed, we detected 47 differentially expressed transcripts representing 23 genes and 24 expressed sequence tags (ESTs); 17 transcripts (7 genes and 10 ESTs) were upregulated and 30 (16 genes and 14 ESTs) were downregulated in the hydrocephalic animals relative to control non-hydrocephalic animals. Seven of these genes, Cck, Nfix, Lgals3, Gsta1, Xdh, Tnf, and Tfpi-2, can be linked to hydrocephalus. In addition, 17 genes that displayed altered expression in our study are not currently known to be associated with the presence or development of hydrocephalus. These results indicate that a relatively few number of transcripts were found to be altered in the development of hydrocephalus in this model. This is the first experiment of its kind to identify changes in gene expression in a congenital model of rodent hydrocephalus that are occurring locally in the area surrounding the cerebral aqueduct. Studies are now needed to examine these candidate genes and their cognate proteins to delineate their role in hydrocephalus.


Subject(s)
Gene Expression Profiling , Hydrocephalus/genetics , Animals , Animals, Newborn , Chromosome Mapping , DNA, Complementary/genetics , Expressed Sequence Tags , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , RNA/genetics , Rats
4.
J Neurochem ; 94(5): 1235-42, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16000157

ABSTRACT

Reperfusion after global brain ischemia results initially in a widespread suppression of protein synthesis in neurons that is due to inhibition of translation initiation as a result of the phosphorylation of the alpha-subunit of eukaryotic initiation factor 2 (eIF2). To address the role of the eIF2alpha kinase RNA-dependent protein kinase-like endoplasmic reticulum kinase (PERK) in the reperfused brain, transgenic mice with a targeted disruption of the Perk gene were subjected to 20 min of forebrain ischemia followed by 10 min of reperfusion. In wild-type mice, phosphorylated eIF2alpha was detected in the non-ischemic brain and its levels were elevated threefold after 10 min of reperfusion. Conversely, there was no phosphorylated eIF2alpha detected in the non-ischemic transgenic mice and there was no sizeable rise in phosphorylated eIF2alpha levels in the forebrain after ischemia and reperfusion. Moreover, there was a substantial rescue of protein translation in the reperfused transgenic mice. Neither group showed any change in total eIF2alpha, phosphorylated eukaryotic elongation factor 2 or total eukaryotic elongation factor 2 levels. These data demonstrate that PERK is responsible for the large increase in phosphorylated eIF2alpha and the suppression of translation early in reperfusion after transient global brain ischemia.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Ischemic Attack, Transient/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , eIF-2 Kinase/metabolism , Animals , Mice , Mice, Knockout , Phosphorylation , Reperfusion Injury/metabolism
5.
Ann Emerg Med ; 42(1): 66-80, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12827125

ABSTRACT

In Williamsburg, VA, April 17 to 20, 1994, the Josiah Macy, Jr. Foundation sponsored a conference entitled "The Role of Emergency Medicine in the Future of American Medical Care," a report on which was published in Annals in 1995. This report promulgated recommendations for the development and enhancement of academic departments of emergency medicine and a conference to develop an agenda for research in emergency medicine. The American College of Emergency Physicians' Research Committee, along with several ad hoc members, presents updates in several of the areas addressed by the Macy Report and subsequent conferences, as a status report for the development of emergency medicine research as a whole, as of late 2002.


Subject(s)
Emergency Medicine , Research , Congresses as Topic , Emergency Medicine/economics , Emergency Medicine/education , Emergency Medicine/trends , Humans , Research/economics , Research/statistics & numerical data , Research/trends
6.
J Cereb Blood Flow Metab ; 23(4): 462-71, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12679723

ABSTRACT

A variety of endoplasmic reticulum (ER) stresses trigger the unfolded protein response (UPR), a compensatory response whose most proximal sensors are the ER membrane-bound proteins ATF6, IRE1alpha, and PERK. The authors simultaneously examined the activation of ATF6, IRE1alpha, and PERK, as well as components of downstream UPR pathways, in the rat brain after reperfusion after a 10-minute cardiac arrest. Although ATF6 was not activated, PERK was maximally activated at 10-minute reperfusion, which correlated with maximal eIF2alpha phosphorylation and protein synthesis inhibition. By 4-h reperfusion, there was 80% loss of PERK immunostaining in cortex and 50% loss in brain stem and hippocampus. PERK was degraded in vitro by mu-calpain. Although inactive IRE1alpha was maximally decreased by 90-minute reperfusion, there was no evidence that its substrate xbp-1 messenger RNA had been processed by removal of a 26-nt sequence. Similarly, there was no expression of the UPR effector proteins 55-kd XBP-1, CHOP, or ATF4. These data indicate that there is dysfunction in several key components of the UPR that abrogate the effects of ER stress. In other systems, failure to mount the UPR results in increased cell death. As other studies have shown evidence for ER stress after brain ischemia and reperfusion, the failure of the UPR may play a significant role in reperfusion neuronal death.


Subject(s)
Brain Ischemia/metabolism , Membrane Proteins , Reperfusion Injury/metabolism , Activating Transcription Factor 4 , Activating Transcription Factor 6 , Animals , Biomarkers , Brain Ischemia/pathology , CCAAT-Enhancer-Binding Proteins/genetics , Calpain/metabolism , Cell Death/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression , Male , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Folding , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Long-Evans , Regulatory Factor X Transcription Factors , Reperfusion Injury/pathology , Transcription Factor CHOP , Transcription Factors/genetics , Transcription Factors/metabolism , X-Box Binding Protein 1 , eIF-2 Kinase/metabolism
7.
Acta Neuropathol ; 106(1): 8-16, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12687390

ABSTRACT

Upon brain reperfusion following ischemia, there is widespread inhibition of neuronal protein synthesis that is due to phosphorylation of eukaryotic initiation factor 2alpha (eIF2alpha), which persists in selectively vulnerable neurons (SVNs) destined to die. Other investigators have shown that expression of mutant eIF2alpha (S51D) mimicking phosphorylated eIF2alpha induces apoptosis, and expression of non-phosphorylatable eIF2alpha (S51A) blocks induction of apoptosis. An early event in initiating apoptosis is the release of cytochrome c from mitochondria, and cytochrome c release corresponds to the selective vulnerability of hippocampal CA1 neurons in rats after transient global cerebral ischemia. At present the signaling pathways leading to this are not well defined. We hypothesized that persistent eIF2alpha(P) reflects injury mechanisms that are causally upstream of release of cytochrome c and induction of apoptosis. At 4 h of reperfusion following 10-min cardiac arrest, vulnerable neurons in the striatum, hippocampal hilus and CA1 showed colocalized intense immunostaining for both persistent eIF2alpha(P) and cytoplasmic cytochrome c, while resistant neurons in the dentate gyrus and elsewhere did not immunostain for either. A lower intensity of persistent eIF2alpha(P) immunostaining was present in cortical layer V pyramidal neurons without cytoplasmic cytochrome c, possibly reflecting the lesser vulnerability of this area to ischemia. We did not observe cytoplasmic cytochrome c in any neurons that did not also display persistent eIF2alpha(P) immunostaining. Because phosphorylation of eIF2alpha during early brain reperfusion is carried out by PERK, these findings suggest that there is prolonged activation of the unfolded protein response in the reperfused brain.


Subject(s)
Brain Ischemia/metabolism , Cytochrome c Group/metabolism , Eukaryotic Initiation Factor-2/metabolism , Hippocampus/metabolism , Neurons/metabolism , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Brain Ischemia/pathology , Cytoplasm/metabolism , Cytoplasm/pathology , Disease Models, Animal , Hippocampus/pathology , Immunohistochemistry , Male , Neurons/pathology , Rats , Rats, Long-Evans , Reperfusion/methods , Time Factors
9.
J Cereb Blood Flow Metab ; 22(2): 127-41, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11823711

ABSTRACT

Protein synthesis inhibition occurs in neurons immediately on reperfusion after ischemia and involves at least alterations in eukaryotic initiation factors 2 (eIF2) and 4 (eIF4). Phosphorylation of the alpha subunit of eIF2 [eIF2(alphaP)] by the endoplasmic reticulum transmembrane eIF2alpha kinase PERK occurs immediately on reperfusion and inhibits translation initiation. PERK activation, along with depletion of endoplasmic reticulum Ca2+ and inhibition of the endoplasmic reticulum Ca2+ -ATPase, SERCA2b, indicate that an endoplasmic reticulum unfolded protein response occurs as a consequence of brain ischemia and reperfusion. In mammals, the upstream unfolded protein response components PERK, IRE1, and ATF6 activate prosurvivial mechanisms (e.g., transcription of GRP78, PDI, SERCA2b ) and proapoptotic mechanisms (i.e., activation of Jun N-terminal kinases, caspase-12, and CHOP transcription). Sustained eIF2(alphaP) is proapoptotic by inducing the synthesis of ATF4, the CHOP transcription factor, through "bypass scanning" of 5' upstream open-reading frames in ATF4 messenger RNA; these upstream open-reading frames normally inhibit access to the ATF4 coding sequence. Brain ischemia and reperfusion also induce mu-calpain-mediated or caspase-3-mediated proteolysis of eIF4G, which shifts message selection to m 7 G-cap-independent translation initiation of messenger RNAs containing internal ribosome entry sites. This internal ribosome entry site-mediated translation initiation (i.e., for apoptosis-activating factor-1 and death-associated protein-5) can also promote apoptosis. Thus, alterations in eIF2 and eIF4 have major implications for which messenger RNAs are translated by residual protein synthesis in neurons during brain reperfusion, in turn constraining protein expression of changes in gene transcription induced by ischemia and reperfusion. Therefore, our current understanding shifts the focus from protein synthesis inhibition to the molecular pathways that underlie this inhibition, and the role that these pathways play in prosurvival and proapoptotic processes that may be differentially expressed in vulnerable and resistant regions of the reperfused brain.


Subject(s)
Brain Ischemia/physiopathology , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/physiology , Reperfusion Injury/physiopathology , Animals , Cell Survival , Nerve Tissue Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...