Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Nat Med ; 23(4): 450-460, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28288111

ABSTRACT

Although blood-brain barrier (BBB) compromise is central to the etiology of diverse central nervous system (CNS) disorders, endothelial receptor proteins that control BBB function are poorly defined. The endothelial G-protein-coupled receptor (GPCR) Gpr124 has been reported to be required for normal forebrain angiogenesis and BBB function in mouse embryos, but the role of this receptor in adult animals is unknown. Here Gpr124 conditional knockout (CKO) in the endothelia of adult mice did not affect homeostatic BBB integrity, but resulted in BBB disruption and microvascular hemorrhage in mouse models of both ischemic stroke and glioblastoma, accompanied by reduced cerebrovascular canonical Wnt-ß-catenin signaling. Constitutive activation of Wnt-ß-catenin signaling fully corrected the BBB disruption and hemorrhage defects of Gpr124-CKO mice, with rescue of the endothelial gene tight junction, pericyte coverage and extracellular-matrix deficits. We thus identify Gpr124 as an endothelial GPCR specifically required for endothelial Wnt signaling and BBB integrity under pathological conditions in adult mice. This finding implicates Gpr124 as a potential therapeutic target for human CNS disorders characterized by BBB disruption.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Glioblastoma/genetics , Infarction, Middle Cerebral Artery/genetics , Intracranial Hemorrhages/genetics , Receptors, G-Protein-Coupled/genetics , Tight Junctions/metabolism , Animals , Blood-Brain Barrier/ultrastructure , Disease Models, Animal , Endothelial Cells/ultrastructure , Extracellular Matrix/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Glioblastoma/metabolism , Infarction, Middle Cerebral Artery/metabolism , Intracranial Hemorrhages/metabolism , Mice , Mice, Knockout , Microscopy, Electron , Microvessels , Pericytes/ultrastructure , Real-Time Polymerase Chain Reaction , Tight Junctions/ultrastructure , Wnt Signaling Pathway
3.
Acta Neurochir (Wien) ; 157(2): 155-64; discussion 164, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25526721

ABSTRACT

BACKGROUND: Maximum safe resection is the "gold standard" in surgical treatment of grade 2 gliomas (G2Gs), aiming to achieve maximal survival benefit with minimal risk of functional deficit. OBJECTIVE: To investigate the attitude of patients and experts towards more extensive surgery with a trade-off between neurological function and survival time. METHODS: Eight patients and seven experts participated in semi-structured focus group interviews. RESULTS: Both patients and experts accepted the premise of balancing neurological function versus longevity. Some patients would accept an increased risk of permanent neurological deficits in order to obtain a chance of increased survival. There was a significant variance in what constituted "quality of life" both between patients and for the individual patient over time. CONCLUSIONS: In important life-changing decisions there is no "one size fits all". We find that it is ethically acceptable to offer more extensive surgery than is possible within the concept of maximal safe surgery as a treatment option, when balancing the principles of beneficence, non-maleficience, autonomy and justice supports the decision. At the same time it must be remembered that even when the patients have made a well-informed decision, some will regret it. In that situation it will be our job as healthcare professionals to support them and help carry some of this burden.


Subject(s)
Ethics, Medical , Glioma/surgery , Neurosurgical Procedures/ethics , Postoperative Complications , Quality of Life/psychology , Survival Rate , Adult , Female , Humans , Male , Middle Aged , Neoplasm Grading
4.
Ugeskr Laeger ; 176(48)2014 Nov 24.
Article in Danish | MEDLINE | ID: mdl-25430570

ABSTRACT

International studies have shown that a significant number of children and adolescents are exposed to potentially traumatic events. Many of these children and adolescents, some of whom will experience post-traumatic stress disorder (PTSD), are submitted to health-care departments shortly after exposure. In this article the concept of trauma-informed health care is introduced in a Danish context. The most recent empirical literature on early preventive interventions is reviewed, and principles for best practice and recommendations for future policy and research are presented.


Subject(s)
Stress Disorders, Post-Traumatic/prevention & control , Adolescent , Child , Early Medical Intervention , Evidence-Based Medicine , Health Personnel/education , Hospitals , Humans , Social Support
5.
J Cereb Blood Flow Metab ; 33(1): 106-14, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23032483

ABSTRACT

Rosiglitazone, a synthetic peroxisome proliferator-activated receptor-γ (PPARγ) agonist, prevents cell death after cerebral ischemia in animal models, but the underlying mechanism has not been clarified. In this study, we examined how rosiglitazone protects neurons against ischemia. Mice treated with rosiglitazone were subjected to 60 minutes of focal ischemia followed by reperfusion. Rosiglitazone reduced infarct volume after ischemia and reperfusion. We show that this neuroprotective effect was reversed with a PPARγ antagonist. Western blot analysis showed a significant increase in expression of phosphorylated stress-activated protein kinases (c-Jun N-terminal kinase (JNK) and p38) in ischemic brain tissue. Rosiglitazone blocked this increase. Furthermore, we observed that rosiglitazone increased expression of the dual-specificity phosphatase 8 (DUSP8) protein and messenger RNA in ischemic brain tissue. Dual-specificity phosphatase 8 is a mitogen-activated protein kinase phosphatase that can dephosphorylate JNK and p38. Another key finding of the present study was that knockdown of DUSP8 in primary cultured cortical neurons that were subjected to oxygen-glucose deprivation diminished rosiglitazone's effect on downregulation of JNK phosphorylation. Thus, rosiglitazone's neuroprotective effect after ischemia is mediated by blocking JNK phosphorylation induced by ischemia via DUSP8 upregulation.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Ischemic Attack, Transient/drug therapy , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Thiazolidinediones/therapeutic use , Animals , Blotting, Western , Cell Death , Disease Models, Animal , Enzyme Activation , Glucose/metabolism , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/pathology , Male , Mice , Mice, Inbred C57BL , Neurons/enzymology , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Oxygen/metabolism , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Phosphorylation , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology
6.
Brain ; 135(Pt 11): 3298-310, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23169920

ABSTRACT

Transplantation of neural stem cells provides a promising therapy for stroke. Its efficacy, however, might be limited because of massive grafted-cell death after transplantation, and its insufficient capability for tissue repair. Interleukin 6 is a pro-inflammatory cytokine involved in the pathogenesis of various neurological disorders. Paradoxically, interleukin 6 promotes a pro-survival signalling pathway through activation of signal transducer and activator of transcription 3. In this study, we investigated whether cellular reprogramming of neural stem cells with interleukin 6 facilitates the effectiveness of cell transplantation therapy in ischaemic stroke. Neural stem cells harvested from the subventricular zone of foetal mice were preconditioned with interleukin 6 in vitro and transplanted into mouse brains 6 h or 7 days after transient middle cerebral artery occlusion. Interleukin 6 preconditioning protected the grafted neural stem cells from ischaemic reperfusion injury through signal transducer and activator of transcription 3-mediated upregulation of manganese superoxide dismutase, a primary mitochondrial antioxidant enzyme. In addition, interleukin 6 preconditioning induced secretion of vascular endothelial growth factor from the neural stem cells through activation of signal transducer and activator of transcription 3, resulting in promotion of angiogenesis in the ischaemic brain. Furthermore, transplantation of interleukin 6-preconditioned neural stem cells significantly attenuated infarct size and improved neurological performance compared with non-preconditioned neural stem cells. This interleukin 6-induced amelioration of ischaemic insults was abolished by transfecting the neural stem cells with signal transducer and activator of transcription 3 small interfering RNA before transplantation. These results indicate that preconditioning with interleukin 6, which reprograms neural stem cells to tolerate oxidative stress after ischaemic reperfusion injury and to induce angiogenesis through activation of signal transducer and activator of transcription 3, is a simple and beneficial approach for enhancing the effectiveness of cell transplantation therapy in ischaemic stroke.


Subject(s)
Interleukin-6/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/transplantation , Recovery of Function/drug effects , Stroke/therapy , Angiogenesis Inducing Agents/pharmacology , Animals , Brain/metabolism , Brain/physiopathology , Brain/surgery , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Interleukin-6/antagonists & inhibitors , Interleukin-6/therapeutic use , Male , Mice , Mice, Transgenic , Neurologic Examination/methods , Neurologic Examination/statistics & numerical data , RNA, Small Interfering/pharmacology , Reperfusion Injury/drug therapy , STAT3 Transcription Factor/physiology , Stroke/physiopathology , Superoxide Dismutase/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
7.
Stroke ; 43(9): 2423-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22713489

ABSTRACT

BACKGROUND AND PURPOSE: The harsh host brain microenvironment caused by production of reactive oxygen species after ischemic reperfusion injury offers a significant challenge to survival of transplanted neural stem cells (NSCs) after ischemic stroke. Copper/zinc-superoxide dismutase (SOD1) is a specific antioxidant enzyme that counteracts superoxide anions. We have investigated whether genetic manipulation to overexpress SOD1 enhances survival of grafted stem cells and accelerates amelioration of ischemic stroke. METHODS: NSCs genetically modified to overexpress or downexpress SOD1 were administered intracerebrally 2 days after transient middle cerebral artery occlusion. Histological and behavioral tests were examined from Days 0 to 28 after stroke. RESULTS: Overexpression of SOD1 suppressed production of superoxide anions after ischemic reperfusion injury and reduced NSC death after transplantation. In contrast, downexpression of SOD1 promoted superoxide generation and increased oxidative stress-mediated NSC death. Transplantation of SOD1-overexpressing NSCs enhanced angiogenesis in the ischemic border zone through upregulation of vascular endothelial growth factor. Moreover, grafted SOD1-overexpressing NSCs reduced infarct size and improved behavioral performance compared with NSCs that were not genetically modified. CONCLUSIONS: Our findings reveal a strong involvement of SOD1 expression in NSC survival after ischemic reperfusion injury. We propose that conferring antioxidant properties on NSCs by genetic manipulation of SOD1 is a potential approach for enhancing the effectiveness of cell transplantation therapy in ischemic stroke.


Subject(s)
Brain Ischemia/therapy , Neural Stem Cells/physiology , Stem Cell Transplantation , Stroke/therapy , Superoxide Dismutase/genetics , Animals , Brain Ischemia/pathology , Cell Separation , Cell Survival/drug effects , Cells, Cultured , Glucose/deficiency , Hypoxia, Brain/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Stroke/pathology , Superoxide Dismutase/biosynthesis , Superoxides/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
8.
J Neurosci ; 32(10): 3462-73, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22399769

ABSTRACT

Transplantation of neural stem cells (NSCs) offers a novel therapeutic strategy for stroke; however, massive grafted cell death following transplantation, possibly due to a hostile host brain environment, lessens the effectiveness of this approach. Here, we have investigated whether reprogramming NSCs with minocycline, a broadly used antibiotic also known to possess cytoprotective properties, enhances survival of grafted cells and promotes neuroprotection in ischemic stroke. NSCs harvested from the subventricular zone of fetal rats were preconditioned with minocycline in vitro and transplanted into rat brains 6 h after transient middle cerebral artery occlusion. Histological and behavioral tests were examined from days 0-28 after stroke. For in vitro experiments, NSCs were subjected to oxygen-glucose deprivation and reoxygenation. Cell viability and antioxidant gene expression were analyzed. Minocycline preconditioning protected the grafted NSCs from ischemic reperfusion injury via upregulation of Nrf2 and Nrf2-regulated antioxidant genes. Additionally, preconditioning with minocycline induced the NSCs to release paracrine factors, including brain-derived neurotrophic factor, nerve growth factor, glial cell-derived neurotrophic factor, and vascular endothelial growth factor. Moreover, transplantation of the minocycline-preconditioned NSCs significantly attenuated infarct size and improved neurological performance, compared with non-preconditioned NSCs. Minocycline-induced neuroprotection was abolished by transfecting the NSCs with Nrf2-small interfering RNA before transplantation. Thus, preconditioning with minocycline, which reprograms NSCs to tolerate oxidative stress after ischemic reperfusion injury and express higher levels of paracrine factors through Nrf2 up-regulation, is a simple and safe approach to enhance the effectiveness of transplantation therapy in ischemic stroke.


Subject(s)
Brain Ischemia/prevention & control , Ischemic Preconditioning/methods , Minocycline/pharmacology , Neural Stem Cells/transplantation , Neuroprotective Agents/pharmacology , Stem Cell Transplantation/methods , Stroke/prevention & control , Animals , Brain Ischemia/pathology , Brain Ischemia/surgery , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Male , Minocycline/therapeutic use , Neural Stem Cells/drug effects , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Stroke/pathology , Stroke/surgery
9.
Antioxid Redox Signal ; 14(8): 1505-17, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-20812869

ABSTRACT

Significant amounts of oxygen free radicals (oxidants) are generated during cerebral ischemia/reperfusion, and oxidative stress plays an important role in brain damage after stroke. In addition to oxidizing macromolecules, leading to cell injury, oxidants are also involved in cell death/survival signal pathways and cause mitochondrial dysfunction. Experimental data from laboratory animals that either overexpress (transgenic) or are deficient in (knock-out) antioxidant proteins, mainly superoxide dismutase, have provided strong evidence of the role of oxidative stress in ischemic brain damage. In addition to mitochondria, recent reports demonstrate that NADPH oxidase (NOX), an important pro-oxidant enzyme, is also involved in the generation of oxidants in the brain after stroke. Inhibition of NOX is neuroprotective against cerebral ischemia. We propose that superoxide dismutase and NOX activity in the brain is a major determinant for ischemic damage/repair and that these major anti- and pro-oxidant enzymes are potential endogenous molecular targets for stroke therapy.


Subject(s)
Cytoprotection , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , NADPH Oxidases/metabolism , Oxidative Stress , Superoxide Dismutase/metabolism , Animals , Cell Death , Humans , Hypoxia-Ischemia, Brain/enzymology , NADPH Oxidases/antagonists & inhibitors , Oxidants/metabolism
10.
Mol Neurobiol ; 41(2-3): 172-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20157789

ABSTRACT

Effective stroke therapies require recanalization of occluded cerebral blood vessels. However, reperfusion can cause neurovascular injury, leading to cerebral edema, brain hemorrhage, and neuronal death by apoptosis/necrosis. These complications, which result from excess production of reactive oxygen species in mitochondria, significantly limit the benefits of stroke therapies. We have developed a focal stroke model using mice deficient in mitochondrial manganese-superoxide dismutase (SOD2-/+) to investigate neurovascular endothelial damage that occurs during reperfusion. Following focal stroke and reperfusion, SOD2-/+ mice had delayed blood-brain barrier breakdown, associated with activation of matrix metalloproteinase and high brain hemorrhage rates, whereas a decrease in apoptosis and hemorrhage was observed in SOD2 overexpressors. Thus, induction and activation of SOD2 is a novel strategy for neurovascular protection after ischemia/reperfusion. Our recent study identified the signal transducer and activator of transcription 3 (STAT3) as a transcription factor of the mouse SOD2 gene. During reperfusion, activation of STAT3 and its recruitment into the SOD2 gene were blocked, resulting in increased oxidative stress and neuronal apoptosis. In contrast, pharmacological activation of STAT3 induced SOD2 expression, which limits ischemic neuronal death. Our studies point to antioxidant-based neurovascular protective strategies as potential treatments to expand the therapeutic window of currently approved therapies.


Subject(s)
Neuroprotective Agents/therapeutic use , Reperfusion Injury/physiopathology , Reperfusion Injury/therapy , Stroke/physiopathology , Stroke/therapy , Animals , Disease Models, Animal , Humans , Mice , Mice, Knockout , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidative Stress , Reperfusion Injury/complications , Reperfusion Injury/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Stroke/complications , Stroke/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
11.
J Cereb Blood Flow Metab ; 29(9): 1589-600, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19553907

ABSTRACT

We recently showed that intraischemic moderate hypothermia (30 degrees C) reduces ischemic damage through the Akt pathway after permanent distal middle cerebral artery occlusion in rats. The only Akt pathway component preserved by hypothermia is phosphorylated phosphatase and tensin homolog deleted on chromosome 10 (p-PTEN), which suggests that p-PTEN may have a central role in neuroprotection. Reactive oxygen species (ROS) are critically involved in mediating ischemic damage after stroke by interacting with signaling molecules, including Akt, PTEN, and delta-protein kinase C (PKC). We investigated the protective mechanisms of moderate hypothermia on these signaling proteins after transient focal ischemia in rats. Early moderate hypothermia (3 h) was administered 15 mins before reperfusion, and delayed moderate hypothermia (3 h) was applied 15 mins after reperfusion. Our results indicate that early hypothermia reduced infarction, whereas delayed hypothermia did not. However, both early and delayed hypothermia maintained levels of Mn-SOD (superoxide dismutase) and phosphorylated Akt and blocked delta-PKC cleavage, suggesting that these factors may not be critical to the protection of hypothermia. Nevertheless, early hypothermia preserved p-PTEN levels after reperfusion, whereas delayed hypothermia did not. Furthermore, ROS inhibition maintained levels of p-PTEN after stroke. Together, these findings suggest that phosphorylation levels of PTEN are closely associated with the protective effect of early hypothermia against stroke.


Subject(s)
Hypothermia, Induced , PTEN Phosphohydrolase/metabolism , Reactive Oxygen Species/metabolism , Stroke/metabolism , Animals , Benzenesulfonates/metabolism , Male , Phosphorylation , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Stroke/pathology , Superoxide Dismutase/metabolism , Time Factors
12.
Breast J ; 14(5): 450-5, 2008.
Article in English | MEDLINE | ID: mdl-18657146

ABSTRACT

The mammary gland undergoes morphologic changes during the menstrual cycle. Proliferation of normal breast epithelium is most extensive during the natural luteal phase. To determine the impact of one cycle of a combined oral contraceptive (COC) on breast homeostasis, we evaluated the proliferation index (PI), determined by KI-67 expression, in normal human mammary epithelial cells and correlated it with cellular proliferation in spontaneous menstrual cycles during the same period. Normal breast tissue samples were obtained from 82 patients randomized in two groups. Forty-two women in group A received one cycle of a COC (30 mug ethinyl estradiol and 150 mug levonorgestrel) administrated daily for 21 days, beginning on the first day of the menstrual cycle. Group B patients (n = 40) experienced a natural menstrual cycle. Menstrual cycle phase characterization was based on the date of the last period and subsequent menses and on progesterone serum levels obtained at the time of biopsy. The PI (number of Ki-67-positive nuclei per 1,000 epithelial cells), was significantly larger in group A (5.47 +/- 3.87), than in group B (3.27 +/- 3.24), p < 0.01. A cyclical variation of PI was observed in COC cycles. The rise in PI in the first week of the COC cycles was significantly higher than in the natural cycle (COC = 7.02 +/- 4.94; non-COC = 1.10 +/- 0.67; p < 0.0011). There was no significant difference between the two groups during the other weeks. Additionally, there was an inverse correlation between proliferation and chronological age, irrespective of the stage of the cycle. The PI of COC (p = 0.175) and natural cycles (p = 0.466) were not statistically different in younger patients. COC users have increased proliferative activity at the beginning of the menstrual cycle. This alteration in the pattern of proliferative activity may relate to the increased risk of breast cancer that has been associated with COCs.


Subject(s)
Breast/pathology , Cell Proliferation/drug effects , Contraceptives, Oral, Combined/administration & dosage , Menstrual Cycle/drug effects , Adolescent , Adult , Age Factors , Biopsy, Needle , Breast/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Humans , Immunohistochemistry , Linear Models , Luteal Phase/drug effects , Luteal Phase/physiology , Menstrual Cycle/physiology , Probability , Progesterone/metabolism , Radioimmunoassay , Reference Values , Sensitivity and Specificity
13.
Ann Neurol ; 59(6): 929-38, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16673393

ABSTRACT

OBJECTIVE: Early reperfusion after an ischemic stroke can cause blood-brain barrier injury with subsequent cerebral edema and devastating brain hemorrhage. These complications of early reperfusion, which result from excess production of reactive oxygen species, significantly limit the benefits of stroke therapies. In this article, we use a novel animal model that facilitates identification of specific components of the reperfusion injury process, including vascular injury and secondary brain damage, and allows assessment of therapeutic interventions. METHODS: Knock-out (KO) mice containing 50% manganese-superoxide dismutase activity (SOD2-KO) and transgenic mice overexpressing SOD2 undergo transient focal ischemia and reperfusion followed by assessment of infarct, edema, hemorrhage rates, metalloproteinase activation, and microvascular injury. RESULTS: SOD2-KO mice demonstrate delayed (>24h) blood-brain barrier breakdown associated with activation of matrix metalloproteinases, inflammation, and high brain hemorrhage rates. These adverse consequences are absent in wild-type littermates and minocycline-treated SOD2-KO animals. Increased hemorrhage rates also are absent in SOD2 overexpressors, which have reduced vascular endothelial cell death. Finally, we show that the tight junction membrane protein, occludin, is an early and specific target in oxidative stress-induced microvascular injury. INTERPRETATION: This model is ideal for studying ischemia/reperfusion-induced vascular injury and secondary brain hemorrhage and offers a unique opportunity to evaluate antioxidant-based neurovascular protective strategies as potential adjunct treatments to currently approved stroke therapies such as thrombolysis and endovascular clot retrieval.


Subject(s)
Brain Ischemia/physiopathology , Cerebral Hemorrhage/physiopathology , Disease Models, Animal , Reperfusion Injury/physiopathology , Superoxide Dismutase/metabolism , Animals , Blood-Brain Barrier/pathology , Blotting, Western , Brain/blood supply , Brain/pathology , Brain/physiopathology , Brain Ischemia/complications , Brain Ischemia/pathology , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/pathology , Enzyme Inhibitors/pharmacology , In Situ Nick-End Labeling , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Minocycline/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Reperfusion Injury/complications , Reperfusion Injury/pathology , Superoxide Dismutase/genetics
14.
Stroke ; 37(4): 1116-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16514094

ABSTRACT

BACKGROUND AND PURPOSE: Compelling evidence supporting the role of inflammation in the development of cerebral infarction has focused attention on the potential of antiinflammatory treatment strategies for stroke. Interferon (IFN)-beta, an immunomodulatory agent approved for treatment of multiple sclerosis, is being evaluated in a phase I clinical trial in acute ischemic stroke. In the present study, we evaluated the effects of wild-type rat IFN-beta and its pegylated counterpart (PEG-IFN-beta) in a model of focal ischemia and reperfusion. METHODS: After 60 minutes of middle cerebral artery occlusion, rats (n=12/group) were treated with IV tail injections of 8 or 16 mug of IFN-beta in 300 muL of PBS once daily for 3 or 7 days or with IV or SC injections of PEG-IFN-beta for 1 day. The animals were assessed daily for weight and for neurological findings. Additional animals underwent complete hematology and chemistry profiles, as well as complete multiorgan necropsy studies. All of the brain tissue was evaluated for assessment of infarct areas, neutrophil infiltration, and presence of hemorrhagic transformations. RESULTS: IFN-beta and PEG-IFN-beta failed to protect against experimental ischemic brain injury as assessed by histopathology and neurological outcome. Furthermore, IFN-beta treatment was associated with significant weight loss and alterations in hematology and chemistry profiles. CONCLUSIONS: Our results suggest that additional preclinical studies are warranted.


Subject(s)
Immunologic Factors/pharmacology , Interferon-beta/pharmacology , Ischemic Attack, Transient/complications , Neuroprotective Agents/pharmacology , Stroke/etiology , Stroke/pathology , Animals , Brain/drug effects , Brain/pathology , Male , Polyethylene Glycols/pharmacology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Stroke/blood , Stroke/metabolism
15.
Breast Cancer Res ; 7(3): R306-13, 2005.
Article in English | MEDLINE | ID: mdl-15987425

ABSTRACT

INTRODUCTION: During the menstrual cycle, the mammary gland goes through sequential waves of proliferation and apoptosis. In mammary epithelial cells, hormonal and non-hormonal factors regulate apoptosis. To determine the cyclical effects of gonadal steroids on breast homeostasis, we evaluated the apoptotic index (AI) determined by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining in human mammary epithelial cells during the spontaneous menstrual cycle and correlated it with cellular proliferation as determined by the expression of Ki-67 during the same period. METHODS: Normal breast tissue samples were obtained from 42 randomly selected patients in the proliferative (n = 21) and luteal (n = 21) phases. Menstrual cycle phase characterization was based on the date of the last and subsequent menses, and on progesterone serum levels obtained at the time of biopsy. RESULTS: The proliferation index (PI), defined as the number of Ki-67-positive nuclei per 1,000 epithelial cells, was significantly larger in the luteal phase (30.46) than in the follicular phase (13.45; P = 0.0033). The AI was defined as the number of TUNEL-positive cells per 1,000 epithelial cells. The average AI values in both phases of the menstrual cycle were not statistically significant (P = 0.21). However, the cell renewal index (CRI = PI/AI) was significantly higher in the luteal phase (P = 0.033). A significant cyclical variation of PI, AI and CRI was observed. PI and AI peaks occurred on about the 24th day of the menstrual cycle, whereas the CRI reached higher values on the 28th day. CONCLUSIONS: We conclude that proliferative activity is dependent mainly on hormonal fluctuations, whereas apoptotic activity is probably regulated by hormonal and non-hormonal factors.


Subject(s)
Apoptosis , Cell Proliferation , Follicular Phase/physiology , Luteal Phase/physiology , Mammary Glands, Human/cytology , Adolescent , Adult , Female , Homeostasis , Humans , In Situ Nick-End Labeling , Ki-67 Antigen/analysis , Kinetics , Mammary Glands, Human/physiology , Progesterone/blood , Progesterone/physiology
16.
Mol Neurobiol ; 31(1-3): 105-16, 2005.
Article in English | MEDLINE | ID: mdl-15953815

ABSTRACT

It has been demonstrated by numerous studies that apoptotic cell death pathways are implicated in ischemic cerebral injury in ischemia models in vivo. Experimental ischemia and reperfusion models, such as transient focal/global ischemia in rodents, have been thoroughly studied and the numerous reports suggest the involvement of cell survival/death signaling pathways in the pathogenesis of apoptotic cell death in ischemic lesions. In these models, reoxygenation during reperfusion provides oxygen as a substrate for numerous enzymatic oxidation reactions and for mitochondrial oxidative phosphorylation to produce adenosine triphosphate. Oxygen radicals, the products of these biochemical and physiological reactions, are known to damage cellular lipids, proteins, and nucleic acids and to initiate cell signaling pathways after cerebral ischemia. Genetic manipulation of intrinsic antioxidants and factors in the signaling pathways has provided substantial understanding of the mechanisms involved in cell death/survival signaling pathways and the role of oxygen radicals in ischemic cerebral injury. Future studies of these pathways could provide novel therapeutic strategies in clinical stroke.


Subject(s)
Brain Ischemia/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Animals , Cell Death/physiology , Cell Survival/physiology , Humans
17.
Neurobiol Dis ; 20(2): 491-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15896972

ABSTRACT

The serine-threonine kinase Akt is a cell survival signaling pathway that inactivates the proapoptotic BCL-2 family protein Bad and promotes cell survival in cerebral ischemia. Involvement of the Akt/Bad signaling pathway after spinal cord injury (SCI) is, however, uncertain. Our results showed that phospho-Akt (serine-473) and phospho-Bad (serine-136) were significantly upregulated at 1 day after SCI. In addition, phospho-Akt and phospho-Bad were colocalized in motor neurons that survived SCI and inhibition of PI3-K reduced expression of phospho-Akt and phospho-Bad. Dimerization of Bad with 14-3-3 in the cytosol was increased whereas Bad/Bcl-XL binding in the mitochondria was decreased after SCI. We further found that reduced oxidative stress by SOD1 overexpression in rats enhanced the expression of phospho-Akt, phospho-Bad, Bad/14-3-3 binding and reduced Bad/Bcl-XL binding after SCI, as compared to wild-type rats. We conclude that oxidative stress may play a role in modulating Akt/Bad signaling and subsequent motor neuron survival after SCI.


Subject(s)
Motor Neurons/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , bcl-Associated Death Protein/metabolism , 14-3-3 Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Survival/genetics , Disease Models, Animal , Female , Humans , Motor Neurons/pathology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Up-Regulation/genetics , bcl-Associated Death Protein/genetics , bcl-X Protein/metabolism
18.
J Cereb Blood Flow Metab ; 25(10): 1312-24, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15829914

ABSTRACT

Proinflammatory cytokines and chemokines are quickly upregulated in response to ischemia/reperfusion (I/R) injury; however, the relationship between I/R-induced oxidative stress and cytokine/chemokine expression has not been elucidated. We investigated the temporal profile of cytokine and chemokine gene expression in transient focal cerebral ischemia using complementary DNA array technology. Among 96 genes studied, 10, 4, 11, and 5 genes were increased at 6, 12, 24, and 72 h of reperfusion, respectively, whereas, 4, 11, 8, and 21 genes, respectively, were decreased. To clarify the relationship between chemokines and oxidative stress, we compared the gene and protein expression of monocyte chemoattractant protein 1 (MCP-1) and macrophage inflammatory protein-1 alpha (MIP-1 alpha) in wild-type (WT) mice and copper/zinc-superoxide dismutase (SOD 1) transgenic (Tg) mice. Monocyte chemoattractant protein-1 and MIP-1 alpha mRNA were significantly upregulated at 6 to 12 h of reperfusion. In the SOD 1 Tg mice, however, MCP-1 and MIP-1 alpha mRNA expression was significantly decreased 12 h postinsult. In the WT mice, MCP-1 and MIP-1 alpha protein expression peaked 24 h after onset of reperfusion determined by immunohistochemistry. In the SOD 1 Tg mice, MCP-1 and MIP-1 alpha immunopositive cells were reduced, as were concentrations of these proteins (measured by enzyme-linked immunosorbent assay) at 24 h of reperfusion. Our results suggest that MCP-1 and MIP-1 alpha expression is influenced by I/R-induced oxidative stress after transient focal stroke.


Subject(s)
Chemokine CCL2/genetics , Ischemic Attack, Transient/genetics , Macrophage Inflammatory Proteins/genetics , Superoxide Dismutase/pharmacology , Animals , Chemokine CCL4 , Chemokines/genetics , Cytokines/genetics , Gene Expression Regulation/drug effects , Kinetics , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Reperfusion Injury , Superoxide Dismutase/genetics , Superoxide Dismutase-1
19.
Stroke ; 35(5): 1169-74, 2004 May.
Article in English | MEDLINE | ID: mdl-15060315

ABSTRACT

BACKGROUND AND PURPOSE: Expression of matrix metalloproteinases (MMPs), proteolytic enzymes that degrade extracellular proteins, is altered after ischemia/reperfusion injury and may contribute to blood-brain barrier (BBB) breakdown. Neutrophils, a source of reactive oxygen species and MMP-9, infiltrate damaged tissue 6 to 24 hours after ischemia and have also been implicated in delayed secondary tissue damage. Here we examined the spatial-temporal relation between MMP-9 expression and neutrophil infiltration after stroke. METHODS: Knockout mice containing 50% manganese superoxide dismutase activity (SOD2-KOs), which are more susceptible to ischemic damage than wild-type (WT) littermates, underwent quantitative antigen (MMP-9, myeloperoxidase) immunohistochemistry (24 and 72 hours) analysis and protein expression by Western blotting (6, 12, 24, 48, and 72 hours) after transient focal cerebral ischemia. BBB breakdown was determined by Evans blue extravasation. RESULTS: There was a clear spatial relation between MMP-9 expression and Evans blue extravasation. MMP-9-positive cell and vessel counts for SOD2-KOs (72 hours) were significantly different from SOD2-KO (24 hours, P=0.004), WT (24 hours, P=0.01), and WT (72 hours, P=0.007) mice. In contrast, MMP-9-positive neutrophil counts were comparatively low and did not differ by time or animal type. MMP-9 expression was biphasic in SOD2-KOs but not in WT littermates, with a significant increase observed 6 to 12 hours after ischemic insult and again at 48 to 72 hours. SOD2-KOs showed increased MMP-9 expression compared with WT littermates at all time points studied (P< or =0.05). CONCLUSIONS: In this model, neutrophils are not the primary source of MMP-9 protein and thus are unlikely the key contributor to BBB breakdown observed in SOD2-KOs.


Subject(s)
Brain Ischemia/metabolism , Matrix Metalloproteinase 9/metabolism , Peroxidase/metabolism , Stroke/metabolism , Animals , Blotting, Western , Brain Ischemia/immunology , Brain Ischemia/pathology , Cell Movement/immunology , Evans Blue , Extravasation of Diagnostic and Therapeutic Materials , Immunohistochemistry , Mice , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/metabolism , Stroke/immunology
20.
NeuroRx ; 1(1): 17-25, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15717004

ABSTRACT

Cumulative evidence suggests that apoptosis plays a pivotal role in cell death in vitro after hypoxia. Apoptotic cell death pathways have also been implicated in ischemic cerebral injury in in vivo ischemia models. Experimental ischemia and reperfusion models, such as transient focal/global ischemia in rodents, have been thoroughly studied and the numerous reports suggest the involvement of cell survival/death signaling pathways in the pathogenesis of apoptotic cell death in ischemic lesions. In these models, reoxygenation during reperfusion provides a substrate for numerous enzymatic oxidation reactions. Oxygen radicals damage cellular lipids, proteins and nucleic acids, and initiate cell signaling pathways after cerebral ischemia. Genetic manipulation of intrinsic antioxidants and factors in the signaling pathways has provided substantial understanding of the mechanisms involved in cell death/survival signaling pathways and the role of oxygen radicals in ischemic cerebral injury. Future studies of these pathways may provide novel therapeutic strategies in clinical stroke.


Subject(s)
Brain Ischemia/pathology , Cell Death/physiology , Neurons/pathology , Signal Transduction/physiology , Animals , Brain/pathology , Brain/physiopathology , Brain Ischemia/physiopathology , Cell Survival/physiology , Humans , Oxidative Stress/physiology , Reactive Oxygen Species/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL