Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Country/Region as subject
Publication year range
1.
FASEB J ; 31(1): 29-34, 2017 01.
Article in English | MEDLINE | ID: mdl-27682203

ABSTRACT

In June 2015, the National Institutes of Health (NIH) released a Guide notice (NOT-OD-15-102) that highlighted the expectation of the NIH that the possible role of sex as a biologic variable be factored into research design, analyses, and reporting of vertebrate animal and human studies. Anticipating these guidelines, the NIH Office of Research on Women's Health, in October 2014, convened key stakeholders to discuss methods and techniques for integrating sex as a biologic variable in preclinical research. The workshop focused on practical methods, experimental design, and approaches to statistical analyses in the use of both male and female animals, cells, and tissues in preclinical research. Workshop participants also considered gender as a modifier of biology. This article builds on the workshop and is meant as a guide to preclinical investigators as they consider methods and techniques for inclusion of both sexes in preclinical research and is not intended to prescribe exhaustive/specific approaches for compliance with the new NIH policy.-Miller, L. R., Marks, C., Becker, J. B., Hurn, P. D., Chen, W.-J., Woodruff, T., McCarthy, M. M., Sohrabji, F., Schiebinger, L., Wetherington, C. L., Makris, S., Arnold, A. P., Einstein, G., Miller, V. M., Sandberg, K., Maier, S., Cornelison, T. L., Clayton, J. A. Considering sex as a biological variable in preclinical research.


Subject(s)
Biomedical Research/standards , Drug Evaluation, Preclinical , National Institutes of Health (U.S.)/standards , Female , Humans , Male , Sex Factors , United States
6.
J Neurosci ; 31(23): 8556-63, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21653859

ABSTRACT

Evaluation of infarct volumes and infiltrating immune cell populations in mice after middle cerebral artery occlusion (MCAO) strongly implicates a mixture of both pathogenic and regulatory immune cell subsets in stroke pathogenesis and recovery. Our goal was to evaluate the contribution of B cells to the development of MCAO by comparing infarct volumes and functional outcomes in wild-type (WT) versus B-cell-deficient µMT(-/-) mice. The results clearly demonstrate larger infarct volumes, higher mortality, more severe functional deficits, and increased numbers of activated T cells, macrophages, microglial cells, and neutrophils in the affected brain hemisphere of MCAO-treated µMT(-/-) versus WT mice. These MCAO-induced changes were completely prevented in B-cell-restored µMT(-/-) mice after transfer of highly purified WT GFP(+) B cells that were detected in the periphery, but not the CNS. In contrast, transfer of B cells from IL-10(-/-) mice had no effect on infarct volume when transferred into µMT(-/-) mice. These findings strongly support a previously unrecognized activity of IL-10-secreting WT B cells to limit infarct volume, mortality rate, recruitment of inflammatory cells, and functional neurological deficits 48 h after MCAO. Our novel observations are the first to implicate IL-10-secreting B cells as a major regulatory cell type in stroke and suggest that enhancement of regulatory B cells might have application as a novel therapy for this devastating neurologic condition.


Subject(s)
B-Lymphocytes/immunology , Brain/immunology , Encephalitis/immunology , Infarction, Middle Cerebral Artery/immunology , Adoptive Transfer , Animals , B-Lymphocytes/pathology , Brain/pathology , Brain/physiopathology , Encephalitis/pathology , Encephalitis/physiopathology , Encephalitis/prevention & control , Flow Cytometry , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Interleukin-10/immunology , Mice , Mice, Knockout , Statistics, Nonparametric
7.
J Immunol ; 184(8): 4087-94, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20304826

ABSTRACT

Reduced risk and severity of stroke in adult females is thought to depend on normal endogenous levels of estrogen, a well-known neuroprotectant and immunomodulator. In male mice, experimental stroke induces immunosuppression of the peripheral immune system, characterized by a reduction in spleen size and cell numbers and decreased cytokine and chemokine expression. However, stroke-induced immunosuppression has not been evaluated in female mice. To test the hypothesis that estradiol (E2) deficiency exacerbates immunosuppression after focal stroke in females, we evaluated the effect of middle cerebral artery occlusion on infarct size and peripheral and CNS immune responses in ovariectomized mice with or without sustained, controlled levels of 17-beta-E2 administered by s.c. implant or the putative membrane estrogen receptor agonist, G1. Both E2- and G1-replacement decreased infarct volume and partially restored splenocyte numbers. Moreover, E2-replacement increased splenocyte proliferation in response to stimulation with anti-CD3/CD28 Abs and normalized aberrant mRNA expression for cytokines, chemokines, and chemokine receptors and percentage of CD4(+)CD25(+)FoxP3(+) T regulatory cells observed in E2-deficient animals. These beneficial changes in peripheral immunity after E2 replacement were accompanied by a profound reduction in expression of the chemokine, MIP-2, and a 40-fold increased expression of CCR7 in the lesioned brain hemisphere. These results demonstrate for the first time that E2 replacement in ovariectomized female mice improves stroke-induced peripheral immunosuppression.


Subject(s)
Benzodioxoles/administration & dosage , Estradiol/administration & dosage , Immunosuppressive Agents/administration & dosage , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Quinolines/administration & dosage , Receptors, G-Protein-Coupled/administration & dosage , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/deficiency , Animals , Benzodioxoles/metabolism , Cell Proliferation , Estradiol/deficiency , Female , Growth Inhibitors/administration & dosage , Growth Inhibitors/deficiency , Immunosuppressive Agents/metabolism , Infarction, Middle Cerebral Artery/prevention & control , Lymphocyte Count , Mice , Mice, Inbred C57BL , Ovariectomy , Quinolines/metabolism , Receptors, Estrogen , Receptors, G-Protein-Coupled/agonists , Severity of Illness Index , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/immunology
8.
Metab Brain Dis ; 27(1): 7-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21989743

ABSTRACT

Although inflammatory responses increase stroke severity, the role of immune cells specific for central nervous system (CNS) antigens remains controversial. Disruption of the blood-brain barrier (BBB) during stroke allows CNS antigens to leak into the peripheral circulation and enhances access of circulating leukocytes to the brain, including those specific for CNS antigens such as myelin oligodendrocyte glycoprotein (MOG) that can induce experimental autoimmune encephalomyelitis (EAE). We here demonstrate for the first time that myelin reactive splenocytes specific for MOG transferred into severe combined immunodeficient (SCID) mice can migrate into the infarct hemisphere of recipients subjected to 60 min middle cerebral artery occlusion (MCAO) and 96 h reperfusion; moreover these cells exacerbate infarct volume and worsen neurological deficits compared to animals transferred with naïve splenocytes. These findings indicate that autoimmunity in the CNS can exert detrimental injury on brain cells and worsen the damage from ischemic stroke.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Infarction, Middle Cerebral Artery/immunology , Myelin Proteins/immunology , Spleen/transplantation , Stroke/immunology , Adoptive Transfer , Animals , Autoimmunity/immunology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Brain/immunology , Brain/metabolism , Cell Culture Techniques , Encephalomyelitis, Autoimmune, Experimental/pathology , Infarction, Middle Cerebral Artery/pathology , Inflammation/immunology , Inflammation/pathology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Myelin Sheath/immunology , Myelin Sheath/metabolism , Myelin-Oligodendrocyte Glycoprotein , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Stroke/pathology , Transplantation, Homologous
9.
Stroke ; 42(9): 2578-83, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21737801

ABSTRACT

BACKGROUND AND PURPOSE: Evaluation of infarct volumes and infiltrating immune cell populations in mice after middle cerebral artery occlusion strongly implicates a mixture of both pathogenic and regulatory immune cell subsets that affect stroke outcome. Our goal was to evaluate the contribution of the well-described coinhibitory pathway, programmed death (PD)-1, to the development of middle cerebral artery occlusion. METHODS: Infarct volumes, functional outcomes, and effects on infiltrating immune cell populations were compared in wild-type C57BL/6 versus PD-1-deficient mice after 60 minutes middle cerebral artery occlusion and 96 hours reperfusion. RESULTS: The results clearly demonstrate a previously unrecognized activity of the PD-1 pathway to limit infarct volume, recruitment of inflammatory cells from the periphery, activation of macrophages and central nervous system microglia, and functional neurological deficits. These regulatory functions were associated with increased percentages of circulating PD-ligand-1 and PD-ligand-2 expressing CD19(+) B-cells in blood, the spleen, and central nervous system with the capacity to inhibit activation of inflammatory T-cells and central nervous system macrophages and microglial cells through upregulated PD-1. CONCLUSIONS: Our novel observations are the first to implicate PD-1 signaling as a major protective pathway for limiting central nervous system inflammation in middle cerebral artery occlusion. This inhibitory circuit would likely be pivotal in reducing stroke-associated Toll-like receptor-2- and Toll like receptor-4-mediated release of neurotoxic factors by activated central nervous system microglia.


Subject(s)
Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/metabolism , Brain Infarction/metabolism , Microglia/metabolism , Signal Transduction , Stroke/metabolism , Animals , Antigens, Surface/genetics , Apoptosis Regulatory Proteins/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , B7-H1 Antigen , Brain Infarction/genetics , Brain Infarction/pathology , Disease Models, Animal , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Microglia/pathology , Peptides/genetics , Peptides/metabolism , Programmed Cell Death 1 Ligand 2 Protein , Programmed Cell Death 1 Receptor , Stroke/genetics , Stroke/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
10.
Stroke ; 42(6): 1736-41, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21512172

ABSTRACT

BACKGROUND AND PURPOSE: Treatment of ischemic stroke by activation of endogenous plasminogen using tissue plasminogen activator is limited by bleeding side effects. In mice, treatment of experimental ischemic stroke with activated protein C improves outcomes; however, activated protein C also has bleeding side effects. In contrast, activation of endogenous protein C using thrombin mutant W215A/E217A (WE) is antithrombotic without hemostasis impairment in primates. Therefore, we investigated the outcome of WE-treated experimental ischemic stroke in mice. METHODS: The middle cerebral artery was occluded with a filament for 60 minutes to induce ischemic stroke. Vehicle, recombinant WE, or tissue plasminogen activator was administered during middle cerebral artery occlusion or 2 hours after middle cerebral artery occlusion. Neurological performance was scored daily. Intracranial bleeding and cerebral infarct size, defined by 2,3,5-triphenyltetrazolium chloride exclusion, were determined on autopsy. Hemostasis was evaluated using tail bleeding tests. RESULTS: WE improved neurological performance scores, increased laser Doppler flowmetry-monitored post-middle cerebral artery occlusion reperfusion of the parietal cortex, and reduced 2,3,5-triphenyltetrazolium chloride-defined cerebral infarct size versus vehicle controls. However, unlike tissue plasminogen activator, WE did not increase tail bleeding or intracranial hemorrhage. CONCLUSIONS: WE treatment is neuroprotective without hemostasis impairment in experimental acute ischemic stroke in mice and thus may provide an alternative to tissue plasminogen activator for stroke treatment.


Subject(s)
Anticoagulants/therapeutic use , Brain Ischemia , Cerebral Infarction/drug therapy , Cerebral Infarction/pathology , Stroke , Thrombin/genetics , Thrombin/therapeutic use , Animals , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Cerebral Infarction/physiopathology , Disease Models, Animal , Hemostasis , Humans , Infarction, Middle Cerebral Artery/drug therapy , Laser-Doppler Flowmetry , Male , Mice , Mice, Inbred C57BL , Mutation , Stroke/drug therapy , Stroke/pathology , Tissue Plasminogen Activator/therapeutic use , Treatment Outcome
11.
Brain Behav Immun ; 25(4): 685-95, 2011 May.
Article in English | MEDLINE | ID: mdl-21262338

ABSTRACT

Stroke is a sexually dimorphic disease with male gender considered a disadvantage in terms of risk and disease outcome. In intact males, stroke induces peripheral immunosuppression, characterized by decreased splenocyte numbers and proliferation and altered percentages of viable T, B, and CD11b+ cells. To investigate whether the potent androgen and known immunomodulator, dihydrotestosterone (DHT), exacerbates post-stroke immunosuppression in castrated male mice after focal stroke, we evaluated the effect of middle cerebral artery occlusion (MCAO) on peripheral and central nervous system (CNS) immune responses in castrated mice with or without controlled levels of DHT. MCAO reduced spleen cell numbers in both groups, but altered T cell and B cell percentages in remaining splenocytes and concomitantly increased the percentage of CD11b+ blood cells solely in DHT-replaced animals at 24 h. Furthermore, DHT-replacement reduced splenocyte proliferation which was accompanied by an increased percentage of immunosuppressive regulatory T cells relative to castrates 96 h post-MCAO. In brain, the percentages of immune cell populations in the ischemic hemisphere relative to the non-ischemic hemisphere were similar between castrated and DHT-replaced mice after MCAO. These data suggest DHT modulates peripheral immunosuppression after MCAO but with relatively little effect on early immune response of the recovering CNS.


Subject(s)
Brain/immunology , Dihydrotestosterone/pharmacology , Immune Tolerance/immunology , Immunologic Factors/pharmacology , Lymphocyte Subsets/cytology , Stroke/immunology , Analysis of Variance , Androgens/pharmacology , Animals , Brain/pathology , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Male , Matched-Pair Analysis , Mice , Mice, Inbred C57BL , Spleen/cytology , Spleen/immunology , Stroke/etiology , Stroke/pathology
12.
Metab Brain Dis ; 26(1): 87-90, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21082336

ABSTRACT

Experimental cerebral ischemic stroke is exacerbated by inflammatory T-cells and is accompanied by systemic increases in CD4+CD25+Foxp3+ regulatory T-cells (Treg). To determine their effect on ischemic brain injury, Treg were depleted in Foxp3(DTR) mice prior to stroke induction. In contrast to a recent Nature Medicine report, our results demonstrate unequivocally that Treg depletion did not affect stroke infarct volume, thus failing to implicate this regulatory pathway in limiting stroke damage.


Subject(s)
Inflammation/immunology , Stroke/immunology , T-Lymphocytes, Regulatory , Animals , Animals, Genetically Modified , CD4 Antigens/immunology , Disease Models, Animal , Female , Forkhead Transcription Factors , Inflammation/metabolism , Interleukin-2 Receptor alpha Subunit , Male , Mice , Mice, Inbred C57BL/genetics , Stroke/physiopathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
13.
Metab Brain Dis ; 26(2): 123-33, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21472429

ABSTRACT

Stroke induces a biphasic effect on the peripheral immune response that involves early activation of peripheral leukocytes followed by severe immunosuppression and atrophy of the spleen. Peripheral immune cells, including T lymphocytes, migrate to the brain and exacerbate the developing infarct. Recombinant T-cell receptor (TCR) Ligand (RTL)551 is designed as a partial TCR agonist for myelin oligodendrocyte glycoprotein (MOG)-reactive T cells and has demonstrated the capacity to limit infarct volume and inflammation in brain when administered to mice undergoing middle cerebral artery occlusion (MCAO). The goal of this study was to determine if RTL551 could retain protection when given within the therapeutically relevant 4 h time window currently in clinical practice for stroke patients. RTL551 was administered subcutaneously 4 h after MCAO, with repeated doses every 24 h until the time of euthanasia. Cell numbers were assessed in the brain, blood, spleen and lymph nodes and infarct size was measured after 24 and 96 h reperfusion. RTL551 reduced infarct size in both cortex and striatum at 24 h and in cortex at 96 h after MCAO and inhibited the accumulation of inflammatory cells in brain at both time points. At 24 h post-MCAO, RTL551 reduced the frequency of the activation marker, CD44, on T-cells in blood and in the ischemic hemisphere. Moreover, RTL551 reduced expression of the chemokine receptors, CCR5 in lymph nodes and spleen, and CCR7 in the blood and lymph nodes. These data demonstrate effective treatment of experimental stroke with RTL551 within a therapeutically relevant 4 h time window through immune regulation of myelin-reactive inflammatory T-cells.


Subject(s)
Brain , Infarction, Middle Cerebral Artery , Myelin Proteins , Receptors, Antigen, T-Cell/agonists , Recombinant Fusion Proteins/therapeutic use , Animals , Blood/immunology , Blood/metabolism , Brain/immunology , Brain/metabolism , Disease Models, Animal , Humans , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/therapy , Lymph Nodes/immunology , Lymph Nodes/metabolism , Male , Mice , Mice, Inbred C57BL , Myelin Proteins/agonists , Myelin Proteins/immunology , Myelin-Oligodendrocyte Glycoprotein , Receptors, Antigen, T-Cell/immunology , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Receptors, CCR7/immunology , Receptors, CCR7/metabolism , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , Time Factors , Treatment Outcome
14.
Anesthesiology ; 112(2): 395-405, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20068453

ABSTRACT

BACKGROUND: Severe ischemia induces renal injury less frequently in women than men. In this study, cardiac arrest and cardiopulmonary resuscitation were used to assess whether estradiol is renoprotective via an estrogen receptor (ER)-dependent mechanism. MATERIALS AND METHODS: Male and female C57BL/6 and ER gene-deleted mice underwent 10 min of cardiac arrest followed by cardiopulmonary resuscitation. Serum chemistries and renal stereology were measured 24 h after arrest. RESULTS: Estrogen did not affect mean arterial pressure, regional renal cortical blood flow, and arterial blood gases. Hence, female kidneys were protected (mean +/- SEM: blood urea nitrogen, 65+/- 21 vs.149+/- 27 mg/dl, P = 0.04; creatinine, 0.14 +/- 0.05 vs. 0.73 +/- 0.16 mg/dl, P = 0.01; volume of necrotic tubules, 7 +/- 1% vs. 10 +/- 0%, P = 0.04). Estrogen also reduced renal injury. In intact females (n = 5), ovariectomized/vehicle-treated (n = 8), and ovariectomized/estrogen-treated (n = 8) animals, blood urea nitrogen was 65 +/- 21, 166 +/- 28, and 50 +/- 14 mg/dl (P = 0.002); creatinine was 0.14 +/- 0.05, 0.74 +/- 0.26, and 0.23 +/- 0.27 mg/dl (P = 0.014); necrotic tubules were 2.5 +/- 0.25%, 12.0 +/- 1.9%, and 5.0 +/- 1.6% (P = 0.004), respectively. In ER-[alpha] and ER-[beta] gene-deleted mice and controls estradiol-reduced functional injury (blood urea nitrogen: estradiol 117 +/- 71, vehicle 167 +/- 56, P = 0.007; creatinine: estradiol 0.5 +/- 0.5, vehicle 1.0 +/- 0.4, P = 0.013), but the effect of estradiol was not different between ER-[alpha] or ER-[beta] gene-deleted mice. Adding ICI 182,780 to estradiol did not alter injury. CONCLUSIONS: In women, kidneys were protected from cardiac arrest through estrogen. Estradiol-mediated renoprotection was not affected by ER deletion or blockade. Estradiol is renoprotective after cardiac arrest. The results indicate that estradiol renoprotection is ER-[alpha] and ER-[beta] independent.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Estrogens/physiology , Heart Arrest/complications , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Protective Agents , Acute-Phase Proteins/metabolism , Acute-Phase Proteins/urine , Animals , Blood Chemical Analysis , Blood Pressure/drug effects , Blood Urea Nitrogen , Creatinine/blood , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/genetics , Estrogen Receptor beta/physiology , Female , Kidney/pathology , Kidney Cortex/blood supply , Kidney Diseases/pathology , Lipocalin-2 , Lipocalins/metabolism , Lipocalins/urine , Male , Mice , Mice, Inbred C57BL , Oncogene Proteins/metabolism , Oncogene Proteins/urine , Ovariectomy , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Reperfusion Injury/prevention & control , Sex Characteristics
16.
Stroke ; 40(7): 2539-45, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19443805

ABSTRACT

BACKGROUND AND PURPOSE: Experimental stroke induces a biphasic effect on the immune response that involves early activation of peripheral leukocytes followed by severe immunodepression and atrophy of the spleen and thymus. In tandem, the developing infarct is exacerbated by influx of numerous inflammatory cell types, including T and B lymphocytes. These features of stroke prompted our use of recombinant T cell receptor ligands (RTL), partial major histocompatibility complex Class II molecules covalently bound to myelin peptides. We tested the hypothesis that RTL would improve ischemic outcome in the brain without exacerbating defects in the peripheral immune system function. METHODS: Four daily doses of RTL were administered subcutaneously to C57BL/6 mice after middle cerebral artery occlusion, and lesion size and cellular composition were assessed in the brain and cell numbers were assessed in the spleen and thymus. RESULTS: Treatment with RTL551 (I-A(b) molecule linked to MOG-35-55 peptide) reduced cortical and total stroke lesion size by approximately 50%, inhibited the accumulation of inflammatory cells, particularly macrophages/activated microglial cells and dendritic cells, and mitigated splenic atrophy. Treatment with RTL1000 (HLA-DR2 moiety linked to human MOG-35-55 peptide) similarly reduced the stroke lesion size in HLA-DR2 transgenic mice. In contrast, control RTL with a nonneuroantigen peptide or a mismatched major histocompatibility complex Class II moiety had no effect on stroke lesion size. CONCLUSIONS: These data are the first to demonstrate successful treatment of experimental stroke using a neuroantigen-specific immunomodulatory agent administered after ischemia, suggesting therapeutic potential in human stroke.


Subject(s)
Immunologic Factors/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Receptors, Antigen, T-Cell , Recombinant Fusion Proteins/therapeutic use , Stroke/drug therapy , Stroke/pathology , Animals , Atrophy , Disease Models, Animal , Dose-Response Relationship, Drug , Injections, Subcutaneous , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reperfusion Injury/pathology , Spleen/pathology , Thymus Gland/pathology
17.
Stroke ; 40(6): 2244-50, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19246690

ABSTRACT

The initial Stroke Therapy Academic Industry Roundtable (STAIR) recommendations published in 1999 were intended to improve the quality of preclinical studies of purported acute stroke therapies. Although recognized as reasonable, they have not been closely followed nor rigorously validated. Substantial advances have occurred regarding the appropriate quality and breadth of preclinical testing for candidate acute stroke therapies for better clinical translation. The updated STAIR preclinical recommendations reinforce the previous suggestions that reproducibly defining dose response and time windows with both histological and functional outcomes in multiple animal species with appropriate physiological monitoring is appropriate. The updated STAIR recommendations include: the fundamentals of good scientific inquiry should be followed by eliminating randomization and assessment bias, a priori defining inclusion/exclusion criteria, performing appropriate power and sample size calculations, and disclosing potential conflicts of interest. After initial evaluations in young, healthy male animals, further studies should be performed in females, aged animals, and animals with comorbid conditions such as hypertension, diabetes, and hypercholesterolemia. Another consideration is the use of clinically relevant biomarkers in animal studies. Although the recommendations cannot be validated until effective therapies based on them emerge from clinical trials, it is hoped that adherence to them might enhance the chances for success.


Subject(s)
Stroke/therapy , Animals , Brain Ischemia/complications , Disease Models, Animal , Guidelines as Topic , Health Care Sector , Humans , Research , Species Specificity , Stroke/etiology , Stroke/physiopathology , Treatment Outcome
18.
Restor Neurol Neurosci ; 27(3): 163-79, 2009.
Article in English | MEDLINE | ID: mdl-19531872

ABSTRACT

Biological sex is an important determinant of stroke risk and outcome. Women are protected from cerebrovascular disease relative to men, an observation commonly attributed to the protective effect of female sex hormones, estrogen and progesterone. However, sex differences in brain injury persist well beyond the menopause and can be found in the pediatric population, suggesting that the effects of reproductive steroids may not completely explain sexual dimorphism in stroke. We review recent advances in our understanding of sex steroids (estradiol, progesterone and testosterone) in the context of ischemic cell death and neuroprotection. Understanding the molecular and cell-based mechanisms underlying sex differences in ischemic brain injury will lead to a better understanding of basic mechanisms of brain cell death and is an important step toward designing more effective therapeutic interventions in stroke.


Subject(s)
Brain Injuries/etiology , Brain Ischemia/etiology , Brain Ischemia/pathology , Sex Characteristics , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Ischemia/metabolism , Cerebral Cortex/pathology , Gonadal Steroid Hormones/metabolism , Humans , Putamen/pathology , Risk Factors
19.
Anesth Analg ; 108(3): 964-70, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19224810

ABSTRACT

BACKGROUND: Previous studies show that the potent, prototypical sigma(1)-receptor agonist 4-phenyl-1-(4-phenylbutyl) piperidine (PPBP) prevents cell death after oxygen-glucose deprivation (OGD) in primary cortical neuronal cultures. We tested the hypothesis that PPBP protects neurons by a mechanism involving activation of the transcription factor cyclic adenosine monophosphate response element-binding protein (CREB). METHODS: Primary cultured cortical neurons were exposed to 2 h of OGD and allowed to recover for 24 h, and PPBP treatment was initiated 15 min before the insult in the presence and absence of the sigma(1)-receptor antagonist rimcazole and inhibitors against protein kinases known to activate signal transduction cascades that result in CREB phosphorylation, such as H89 (protein kinase A inhibitor), LY294002 (PI3K inhibitor), U0126 (MEK1/2 inhibitor), or KN62 calmodulin kinase II inhibitor). Neuronal cell death was assayed by lactate dehydrogenase measurement 24 h after OGD. CREB phosphorylation was measured by immunoblot analysis at 30 min, 1 h, and 3 h of reoxygenation. Blots were quantitatively analyzed using Quantity One image analysis software. RESULTS: PPBP increased CREB phosphorylation at 1 h after recovery from OGD, which was abolished by rimcazole (1.7 +/- 0.2 in PPBP and 0.8 +/- 0.1 in PPBP plus rimcazole with OGD compared with 0.9 +/- 0.1 in OGD alone, p-CREB/CREB). The PPBP-induced increase in CREB phosphorylation was blocked by H89 (0.5 +/- 0.07) but not U0126, KN62, or LY294002. PPBP treatment prevented OGD-induced cell death and pretreatment with H89 blocked this protection (0.18 +/- 0.02 in PPBP and 0.27 +/- 0.03 in PPBP plus H89 with OGD compared with 0.33 +/- 0.02 in OGD alone, lactate dehydrogenase assay). Pretreatment with LY294002, UO126, or KN62 had no effect on neuronal protection by PPBP. CONCLUSIONS: These data suggest that the mechanism of neuroprotection by PPBP may be linked to CREB phosphorylation.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Haloperidol/analogs & derivatives , Neuroprotective Agents/pharmacology , Animals , Blotting, Western , Butadienes/pharmacology , Cell Death/drug effects , Cell Hypoxia/drug effects , Cells, Cultured , Chromones/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Glucose/deficiency , Haloperidol/pharmacology , Isoquinolines/pharmacology , Morpholines/pharmacology , Neurons/drug effects , Neurons/metabolism , Nitriles/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, sigma/drug effects , Sulfonamides/pharmacology , Sigma-1 Receptor
20.
J Neurosci ; 27(27): 7268-74, 2007 Jul 04.
Article in English | MEDLINE | ID: mdl-17611279

ABSTRACT

Estradiol is protective in experimental cerebral ischemia, but the precise mechanisms remain unknown. Signal transducer and activator of transcription-3 (STAT3) is a transcription factor that is activated by estrogen, translocates to the nucleus, and induces the transcription of neuroprotective genes, such as bcl-2. We determined whether estradiol increases STAT3 activation in female rat brain after focal cerebral ischemia and whether STAT3 activation contributes to estradiol-mediated neuroprotection against ischemic brain injury. Ovariectomized (OVX) female rats with and without estradiol replacement were subjected to 2 h of middle cerebral artery occlusion (MCAO), and phosphorylated STAT3 (P-STAT3) and total STAT3 (T-STAT3) were quantified by Western blot analysis at 3 and 22 h of reperfusion. STAT3 activation was colocalized with neuronal and survival markers microtubule-associated protein 2 (MAP2) and Bcl-2 using immunohistochemistry. Infarct size was measured at 22 h after MCAO in estradiol-treated OVX animals in the presence and absence of STAT3 inhibitor cucurbitacin I (JSI-124) using 2,3,5-triphenyltetrazolium chloride staining. Estradiol increased P-STAT3 in the ischemic cortex cytosolic fraction at 3 h after MCAO without affecting T-STAT3. This was associated with increased P-STAT3 in the nuclear fraction, which remained elevated at 22 h after MCAO. The nuclear P-STAT3 colocalized with MAP2 and Bcl-2 within the peri-infarct zone. The P-STAT3 inhibitor JSI-124 abolished the protective effect of estradiol without affecting infarct size in untreated OVX rats. We conclude that estradiol increases STAT3 phosphorylation in neurons after MCAO and that STAT3 activation plays an important role in estradiol-mediated neuroprotection.


Subject(s)
Brain Ischemia/metabolism , Estradiol/therapeutic use , Neuroprotective Agents/therapeutic use , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Animals , Brain Ischemia/prevention & control , Female , Rats
SELECTION OF CITATIONS
SEARCH DETAIL