Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Metab Brain Dis ; 34(2): 631-640, 2019 04.
Article in English | MEDLINE | ID: mdl-30612292

ABSTRACT

The aim of this study was to determine whether leukemia inhibitory factor (LIF) exerts its neuroprotective effects through signal transduction of the transcription factor myeloid zinc finger-1 (MZF-1). According to the hypothesis of this study, MZF-1 mediates LIF-induced neuroprotective signaling during ELVO through increased expression and transcriptional activity. To determine the in vivo role of MZF-1 in LIF-induced neuroprotection, we used Genomatix software was used to MZF-1 sites in the promoter region of the rat superoxide dismutase 3 (SOD3) gene. Stroke was induced via middle cerebral artery occlusion, and animals were administered PBS or 125 µg/kg LIF at 6, 24, and 48 h after the injury. MZF-1 binding activity was measured using electrophoretic mobility shift assay (EMSA) and its expression/localization were determined using western blot and immunohistochemical analysis. To determine whether MZF-1 relays LIF-induced neuroprotection in vitro, primary cultured neurons were subjected to oxygen-glucose deprivation (OGD) after treatment with PBS or LIF. MZF-1 expression was measured in vitro using real time PCR and immunohistochemical staining. Transfection with siRNA was used to determine whether LIF protected cultured neurons against OGD after silencing MZF-1 expression. Four MZF-1 binding sites were identified by Genomatix, and EMSA confirmed in vivo binding activity in brain after MCAO. LIF significantly increased MZF-1 protein levels compared to PBS treatment at 72 h post-MCAO. In vivo nuclear localization of MZF-1 as well as co-localization of SOD3 and MZF-1 was observed in the cortical neurons of LIF-treated rats. Primary cultured neurons treated with LIF had significantly higher levels of MZF-1 mRNA and protein after LIF treatment compared to neurons treated with PBS. Finally, knockdown MZF-1 using siRNA counteracted the neuroprotective effects of LIF in vitro. These data demonstrate that LIF-mediated neuroprotection is dependent upon MZF-1 activity. Furthermore, these findings identify a novel neuroprotective pathway that employs MZF-1, a transcription factor associated with hematopoietic gene expression.


Subject(s)
Leukemia Inhibitory Factor/metabolism , Neuroprotective Agents/pharmacology , Stroke/drug therapy , Zinc Fingers/physiology , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Neuroprotection/physiology , Rats, Sprague-Dawley , Stroke/metabolism
2.
J Neuroinflammation ; 15(1): 288, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30322390

ABSTRACT

BACKGROUND: The migration of peripheral immune cells and splenocytes to the ischemic brain is one of the major causes of delayed neuroinflammation after permanent large vessel stroke. Other groups have demonstrated that leukemia inhibitory factor (LIF), a cytokine that promotes neural cell survival through upregulation of antioxidant enzymes, promotes an anti-inflammatory phenotype in several types of immune cells. The goal of this study was to determine whether LIF treatment modulates the peripheral immune response after stroke. METHODS: Young male (3 month) Sprague-Dawley rats underwent sham surgery or permanent middle cerebral artery occlusion (MCAO). Animals were administered LIF (125 µg/kg) or PBS at 6, 24, and 48 h prior to euthanization at 72 h. Bone marrow-derived macrophages were treated with LIF (20 ng/ml) or PBS after stimulation with interferon gamma + LPS. Western blot was used to measure protein levels of CD11b, IL-12, interferon inducible protein-10, CD3, and the LIF receptor in spleen and brain tissue. ELISA was used to measure IL-10, IL-12, and interferon gamma. Isolectin was used to label activated immune cells in brain tissue sections. Statistical analysis was performed using one-way ANOVA and Student's t test. A Kruskal-Wallis test followed by Bonferroni-corrected Mann-Whitney tests was performed if data did not pass the D'Agostino-Pearson normality test. RESULTS: LIF-treated rats showed significantly lower levels of the LIF receptor and interferon gamma in the spleen and CD11b levels in the brain compared to their PBS-treated counterparts. Fluorescence from isolectin-binding immune cells was more prominent in the ipsilateral cortex and striatum after PBS treatment compared to LIF treatment. MCAO + LIF significantly decreased splenic levels of CD11b and CD3 compared to sham surgery. MCAO + PBS treatment significantly elevated splenic levels of interferon inducible protein-10 at 72 h after MCAO, while LIF treatment after MCAO returned interferon inducible protein 10 to sham levels. LIF administration with interferon gamma + LPS significantly reduced the IL-12/IL-10 production ratio compared to macrophages treated with interferon gamma + LPS alone. CONCLUSIONS: These data demonstrate that LIF promotes anti-inflammatory signaling through alterations of the IL-12/interferon gamma/interferon inducible protein 10 pathway.


Subject(s)
Cytokines/metabolism , Infarction, Middle Cerebral Artery , Leukemia Inhibitory Factor/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Cell Culture Techniques , Disease Models, Animal , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Interferon-gamma/therapeutic use , Lectins/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Rats , Rats, Sprague-Dawley , Spleen/drug effects , Spleen/pathology , Statistics, Nonparametric , Time Factors
3.
J Stroke Cerebrovasc Dis ; 27(10): 2746-2754, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30068479

ABSTRACT

BACKGROUND AND PURPOSE: Acid/base and electrolytes could provide clinically valuable information about cerebral infarct core and penumbra. We evaluated associations between acid/base and electrolyte changes and outcomes in 2 rat models of stroke, permanent, and transient middle cerebral artery occlusion. METHODS: Three-month old Sprague-Dawley rats underwent permanent or transient middle cerebral artery occlusion. Pre- and post-middle cerebral artery occlusion venous samples for permanent and transient models provided pH, carbon dioxide, oxygen, glucose, and electrolyte values of ionized calcium, potassium, and sodium. Multiple regression determined predictors of infarct volume from these values, and Kaplan-Meier curve analyzed morality between permanent and transient middle cerebral artery occlusion models. RESULTS: Analysis indicated significant differences in the blood gas and electrolytes between pre- to post-middle cerebral artery occlusion. A decrease in pH and sodium with increases in carbon dioxide, potassium, ionized calcium, and glucose changes were found in both middle cerebral artery occlusion models; while hematocrit and hemoglobin were significant in the transient model. pH and ionized calcium were predictors of infarct volume in the permanent model, as changes in pH and ionized calcium decreased, infarct volume increased. CONCLUSIONS: There are acute changes in acid/base balance and electrolytes during stroke in transient and permanent rodent models. Additionally, we found pH and ionized calcium changes predicted stroke volume in the permanent middle cerebral artery occlusion model. These preliminary findings are novel, and warrant further exploration in human conditions.


Subject(s)
Acid-Base Equilibrium , Infarction, Middle Cerebral Artery/physiopathology , Water-Electrolyte Balance , Animals , Biomarkers/blood , Blood Glucose/metabolism , Calcium/blood , Carbon Dioxide/blood , Disease Models, Animal , Hemoglobins/metabolism , Hydrogen-Ion Concentration , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/pathology , Oxygen/blood , Potassium/blood , Rats, Sprague-Dawley , Sodium/blood , Time Factors
4.
Mol Neurobiol ; 54(1): 608-622, 2017 01.
Article in English | MEDLINE | ID: mdl-26746670

ABSTRACT

Leukemia inhibitory factor (LIF) has been shown to protect oligodendrocytes from ischemia by upregulating endogenous antioxidants. The goal of this study was to determine whether LIF protects neurons during stroke by upregulating superoxide dismutase 3 (SOD3). Animals were administered phosphate-buffered saline (PBS) or 125 µg/kg LIF at 6, 24, and 48 h after middle cerebral artery occlusion or sham surgery. Neurons were isolated from rat pups on embryonic day 18 and used between 7 and 15 days in culture. Cells were treated with LIF and/or 10 µM Akt inhibitor IV with PBS and 0.1 % DMSO acting as vehicle controls. Neurons transfected with scrambled or SOD3 small interfering RNA (siRNA) were subjected to 24-h ischemia after PBS or LIF treatment. LIF significantly increased superoxide dismutase activity and SOD3 expression in ipsilateral brain tissue compared to PBS. Following 24-h ischemia, LIF reduced cell death and increased SOD3 messenger RNA (mRNA) in vitro compared to PBS. Adding Akt inhibitor IV with LIF counteracted the decrease in cell death. Partially silencing the expression of SOD3 using siRNA prior to LIF treatment counteracted the protective effect of LIF-alone PBS treatment. These results indicate that LIF protects neurons in vivo and in vitro via upregulation of SOD3.


Subject(s)
Cerebral Cortex/enzymology , Leukemia Inhibitory Factor/pharmacology , Neurons/enzymology , Neuroprotective Agents/pharmacology , Superoxide Dismutase/biosynthesis , Up-Regulation/physiology , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Dose-Response Relationship, Drug , Male , Neurons/drug effects , Neurons/pathology , Rats , Up-Regulation/drug effects
5.
Am J Physiol Heart Circ Physiol ; 308(6): H583-91, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25576625

ABSTRACT

The consumption of flavan-3-ol-containing foods, including (-)-epicatechin (EC), has been linked to lower incidence of cardiovascular disease and stroke. We previously demonstrated nuclear transcription factor erythroid 2p45-related factor-2 (Nrf2) -dependent EC efficacy in reducing stroke-induced deficits in 2-mo-old mice; yet stroke is primarily a disease of the elderly. Because neuroinflammation, oxidative stress, and vascular dysfunction are hallmarks of aging, we tested whether Nrf2 mediates EC efficacy in aging mice through modulation of glial responses and blood brain barrier permeability. First, we compared anastomosis in naïve wild-type and C57BL/6 Nrf2(-/-) mice to identify potential differences in cerebrovascular architecture. Data showed no significant differences in the number of anastomoses or mean intersection points, indicating similar gross vascular physiology. To assess efficacy and mechanisms of protection, wild-type or Nrf2(-/-) mice were administered the minimum effective EC dose established in our previous studies before the permanent distal middle cerebral artery occlusion. Similar to previous results with young mice, 12-mo-old wild types also showed significant reductions in infarct volume (41.01 ± 29.57%) and improved performance in removing adhesive tape relative to vehicle-treated controls, whereas a trend toward protection was observed in Nrf2(-/-). However, EC did not reduce immunoreactivity for the microglia/macrophage marker anti-ionized calcium-binding adapter molecule 1, suggesting that dampened activation/recruitment did not account for EC protection. Furthermore, there were no differences in mouse IgG extravasation or spontaneous hemorrhage between EC-treated groups. These data demonstrate that EC protection occurs independent of microglia/macrophage modulation or blood brain barrier preservation, suggesting that the glial cell responses in young mice are compensatory to another, and potentially novel, protective mechanism.


Subject(s)
Brain/blood supply , Brain/drug effects , Flavonoids/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Age Factors , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Brain/physiopathology , Capillary Permeability/drug effects , Disease Models, Animal , Gait/drug effects , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Neuroglia/metabolism , Neuroglia/pathology , Time Factors
6.
Eur J Neurosci ; 40(7): 3111-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25041106

ABSTRACT

Human umbilical cord blood (HUCB) cells have shown efficacy in rodent models of focal ischemia and in vitro systems that recapitulate stroke conditions. One potential mechanism of protection is through secretion of soluble factors that protect neurons and oligodendrocytes (OLs) from oxidative stress. To overcome practical issues with cellular therapies, identification of soluble factors released by HUCB and other stem cells may pave the way for treatment modalities that are safer for a larger percentage of stroke patients. Among these soluble factors is leukemia inhibitory factor (LIF), a cytokine that exerts pleiotropic effects on cell survival. Here, data show that LIF effectively reduced infarct volume, reduced white matter injury and improved functional outcomes when administered to rats following permanent middle cerebral artery occlusion. To further explore downstream signaling, primary oligodendrocyte cultures were exposed to oxygen-glucose deprivation to mimic stroke conditions. LIF significantly reduced lactate dehydrogenase release from OLs, reduced superoxide dismutase activity and induced peroxiredoxin 4 (Prdx4) transcript. Additionally, the protective and antioxidant capacity of LIF was negated by both Akt inhibition and co-incubation with Prdx4-neutralising antibodies, establishing a role for the Akt signaling pathway and Prdx4-mediated antioxidation in LIF protection.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Leukemia Inhibitory Factor/therapeutic use , Neuroprotective Agents/therapeutic use , Oligodendroglia/drug effects , Recovery of Function/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Leukemia Inhibitory Factor/pharmacology , Neuroprotective Agents/pharmacology , Oncogene Protein v-akt/metabolism , Peroxiredoxins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stroke/drug therapy , White Matter/drug effects
7.
Eur J Neurosci ; 38(11): 3659-68, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24112193

ABSTRACT

Consumption of flavan-3-ols, notably (-)-epicatechin (EC), has been highly recommended in complementary and alternative medicine (CAM) due to reports that flavan-3-ols boost antioxidant activity, support vascular function, and prevent cardiovascular disease. To date, in vivo efficacy and mechanisms of action for many CAM therapies, including EC, remain elusive in brain ischemia. In contrast to its purported direct antioxidant role, we hypothesized protection through activation of the endogenous transcriptional factor Nrf2. To screen cellular protection and investigate Nrf2 activation, we adopted a pretreatment paradigm using enriched primary neuronal cultures from mice and washed out EC prior to oxygen glucose deprivation to attenuate direct antioxidant effects. EC protected primary neurons from oxygen glucose deprivation by increasing neuronal viability (40.2 ± 14.1%) and reducing protein oxidation, effects that occurred concomitantly with increased Nrf2-responsive antioxidant protein expression. We also utilized wildtype and Nrf2 C57BL/6 knockout mice in a permanent model of focal brain ischemia to evaluate glial cell regulation and complex sensorimotor functioning. EC-treated wildtype mice displayed a reduction or absence of forelimb motor coordination impairments that were evident in vehicle-treated mice. This protection was associated with reduced anatomical injury (54.5 ± 8.3%) and microglia/macrophage activation/recruitment (56.4 ± 13.0%). The protective effects elicited by EC in both model systems were abolished in tissues and neuronal cultures from Nrf2 knockout mice. Together, these data demonstrate EC protection through Nrf2 and extend the benefits to improved performance on a complex sensorimotor task, highlighting the potential of flavan-3-ols in CAM approaches in minimizing subsequent stroke injury.


Subject(s)
Catechin/therapeutic use , Infarction, Middle Cerebral Artery/prevention & control , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Administration, Oral , Animals , Catechin/administration & dosage , Catechin/pharmacology , Cell Survival , Cells, Cultured , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Inbred C57BL , Movement , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/physiology , Neurons/metabolism , Neurons/physiology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Oxidative Stress
8.
Front Neurol ; 3: 85, 2012.
Article in English | MEDLINE | ID: mdl-22661966

ABSTRACT

Preclinical stroke models provide insights into mechanisms of cellular injury and potential therapeutic targets. Renewed efforts to standardize preclinical practices and adopt more rigorous approaches reflect the assumption that a better class of compounds will translate into clinical efficacy. While the need for novel therapeutics is clear, it is also critical that diagnostics be improved to allow for more rapid treatment upon hospital admission. Advances in imaging techniques have aided in the diagnosis of stroke, yet current limitations and expenses demonstrate the need for new and complementary approaches. Intracerebral hemorrhage (ICH) exhibits the highest mortality rate, displays unique pathology and requires specialized treatment strategies relative to other forms of stroke. The aggressive nature and severe consequences of ICH underscore the need for novel therapeutic approaches as well as accurate and expeditious diagnostic tools. The use of experimental models will continue to aid in addressing these important issues as the field attempts to translate basic science findings into the clinical setting. Several preclinical models of ICH have been developed and are widely used to recapitulate human pathology. Because each model has limitations, the burden lies with the investigator to clearly define the question being asked and select the model system that is most relevant to that question. It may also be necessary to optimize and refine pre-existing paradigms, or generate new paradigms, as the future success of translational research is dependent upon the ability to mimic human sequelae and assess clinically relevant outcome measures as means to evaluate therapeutic efficacy.

9.
Metab Brain Dis ; 27(2): 131-41, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22354752

ABSTRACT

Delayed neuronal death associated with stroke has been increasingly linked to the immune response to the injury. Splenectomy prior to middle cerebral artery occlusion (MCAO) is neuroprotective and significantly reduces neuroinflammation. The present study investigated whether splenic signaling occurs through interferon gamma (IFNγ). IFNγ was elevated early in spleens but later in the brains of rats following MCAO. Splenectomy decreased the amount of IFNγ in the infarct post-MCAO. Systemic administration of recombinant IFNγ abolished the protective effects of splenectomy with a concurrent increase in INFγ expression in the brain. These results suggest a role for spleen-derived IFNγ in stroke pathology.


Subject(s)
Interferon-gamma/physiology , Nerve Degeneration/physiopathology , Spleen/physiopathology , Stroke/physiopathology , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Hypoxia , Cells, Cultured , Female , Fluoresceins , Fluorescent Dyes , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Interferon-gamma/pharmacology , Laser-Doppler Flowmetry , Ligation , Male , Middle Cerebral Artery/physiology , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/metabolism , Organic Chemicals , Pregnancy , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Signal Transduction/physiology , Spleen/metabolism , Splenectomy
10.
Transl Stroke Res ; 3(3): 357-63, 2012 Sep.
Article in English | MEDLINE | ID: mdl-24323811

ABSTRACT

Traumatic brain injury (TBI) is complex and involves multiple processes that contribute to functional decline. Progressive neuropathies result from delayed cellular death following the initial impact. Although the precise mechanisms responsible for delayed injury are unknown, numerous data implicate a role for the peripheral immune system in perpetuating neuroinflammation after TBI. A previous report demonstrated that splenic CCL20 chemokine expression was upregulated 24 h after lateral fluid percussive impact (LFPI), prior to neuronal expression but consistent with neurodegeneration. Here, we expand on those data to report increased CCL20 protein expression in white matter 48 h after LFPI and demonstrate that CCL20 is directly toxic to primary neurons and oligodendrocytes subjected to oxygen glucose deprivation. The temporal expression profile of CCL20, coupled with in vitro toxicity to primary cells, suggests that this chemokine exerts deleterious effects on cell viability following TBI. These findings warrant further investigations into the use of CCL20 as a potential biomarker and/or therapeutic target.

11.
J Biol Chem ; 287(6): 4177-87, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22158864

ABSTRACT

Human umbilical cord blood (HUCB) cells protect the brain against ischemic injury, yet the mechanism of protection remains unclear. Using both in vitro and in vivo paradigms, this study examined the role of Akt signaling and peroxiredoxin 4 expression in human umbilical cord blood cell-mediated protection of oligodendrocytes from ischemic conditions. As previously reported, the addition of HUCB cells to oligodendrocyte cultures prior to oxygen glucose deprivation significantly enhanced oligodendrocyte survival. The presence of human umbilical cord blood cells also increased Akt phosphorylation and elevated peroxiredoxin 4 expression in oligodendrocytes. Blocking either Akt or peroxiredoxin 4 activity with Akt Inhibitor IV or a peroxiredoxin 4-neutralizing antibody, respectively, negated the protective effects of human umbilical cord blood cells. In vivo, systemic administration of human umbilical cord blood cells 48 h after middle cerebral artery occlusion increased Akt phosphorylation and peroxiredoxin 4 protein expression while reducing proteolytic cleavage of caspase 3 in oligodendrocytes residing in the ipsilateral external capsule. Moreover, human umbilical cord blood cells protected striatal white matter bundles from degeneration following middle cerebral artery occlusion. These results suggest that the soluble factors released from human umbilical cord blood cells converge on Akt to elevate peroxiredoxin 4 levels, and these effects contribute to oligodendrocyte survival.


Subject(s)
Brain Ischemia/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Umbilical Cord/cytology , Animals , Brain Ischemia/pathology , Brain Ischemia/therapy , Caspase 3/biosynthesis , Cell Survival , Humans , Oligodendroglia/pathology , Peroxiredoxins/biosynthesis , Phosphorylation , Rats , Rats, Sprague-Dawley
12.
J Neuroinflammation ; 8: 148, 2011 Oct 31.
Article in English | MEDLINE | ID: mdl-22040257

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) evokes a systemic immune response including leukocyte migration into the brain and release of pro-inflammatory cytokines; however, the mechanisms underlying TBI pathogenesis and protection are poorly understood. Due to the high incidence of head trauma in the sports field, battlefield and automobile accidents identification of the molecular signals involved in TBI progression is critical for the development of novel therapeutics. METHODS: In this report, we used a rat lateral fluid percussion impact (LFPI) model of TBI to characterize neurodegeneration, apoptosis and alterations in pro-inflammatory mediators at two time points within the secondary injury phase. Brain histopathology was evaluated by fluoro-jade (FJ) staining and terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay, polymerase chain reaction (qRT PCR), enzyme linked immunosorbent assay (ELISA) and immunohistochemistry were employed to evaluate the CCL20 gene expression in different tissues. RESULTS: Histological analysis of neurodegeneration by FJ staining showed mild injury in the cerebral cortex, hippocampus and thalamus. TUNEL staining confirmed the presence of apoptotic cells and CD11b+ microglia indicated initiation of an inflammatory reaction leading to secondary damage in these areas. Analysis of spleen mRNA by PCR microarray of an inflammation panel led to the identification of CCL20 as an important pro-inflammatory signal upregulated 24 h after TBI. Although, CCL20 expression was observed in spleen and thymus after 24 h of TBI, it was not expressed in degenerating cortex or hippocampal neurons until 48 h after insult. Splenectomy partially but significantly decreased the CCL20 expression in brain tissues. CONCLUSION: These results demonstrate that the systemic inflammatory reaction to TBI starts earlier than the local brain response and suggest that spleen- and/ or thymus-derived CCL20 might play a role in promoting neuronal injury and central nervous system inflammation in response to mild TBI.


Subject(s)
Brain Injuries/immunology , Brain Injuries/pathology , Brain/immunology , Brain/pathology , Chemokine CCL20/immunology , Inflammation/pathology , Nerve Degeneration/pathology , Animals , Apoptosis/physiology , Brain/anatomy & histology , Chemokine CCL20/genetics , DNA Fragmentation , Humans , In Situ Nick-End Labeling , Inflammation/immunology , Male , Nerve Degeneration/immunology , Percussion/adverse effects , Rats , Rats, Sprague-Dawley , Spleen/immunology , Splenectomy , Thymus Gland/immunology
13.
Nutr Neurosci ; 14(5): 226-36, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22005287

ABSTRACT

Epidemiological studies have demonstrated that the consumption of fruits and vegetables is associated with reduced risk for cardiovascular disease and stroke. Detailed investigations into the specific dietary components of these foods have revealed that many polyphenolic constituents exert anti-oxidant effects on key substrates involved in the pathogenesis and progression of ischemic injury. These data have perpetuated the belief that the protective effects of flavonoids result from direct anti-oxidant actions at the levels of the cerebral vasculature and brain parenchyma. While many in vitro studies using purified extracts support this contention, first-pass metabolism alters the bioavailability of flavonoids such that the achievable concentrations after oral consumption are not consistent with this mechanism. Importantly, oral consumption of flavonoids may promote neural protection by facilitating the expression of gene products responsible for detoxifying the ischemic microenvironment through both anti-oxidative and anti-inflammatory actions. In particular, the transcriptional factor nuclear factor erythroid 2-related factor 2 has emerged as a critical regulator of flavonoid-mediated protection through the induction of various cytoprotective genes. The pleiotropic effects associated with potent transcriptional regulation likely represent the primary mechanisms of neural protection, as the flavonoid concentrations reaching ischemic tissues in vivo are sufficient to alter intracellular signal transduction but likely preclude the one-to-one stoichiometry necessary to confer protection by direct anti-oxidation. These data reflect an exciting new direction in the study of complementary and alternative medicine that may lead to the development of novel therapies for ischemic/hemorrhagic stroke, traumatic brain injury, and other neurological disorders.


Subject(s)
Dietary Supplements , Flavonoids/administration & dosage , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/metabolism , Stroke/diet therapy , Complementary Therapies , Flavonoids/metabolism , Humans , Risk Factors , Stroke/epidemiology , Stroke/prevention & control
14.
Transl Stroke Res ; 2(3): 328-38, 2011 Sep.
Article in English | MEDLINE | ID: mdl-24323652

ABSTRACT

The majority of promising experimental compounds have failed in clinical trials, highlighting the need for novel approaches to treat stroke. Much research has been devoted to elucidating the signaling pathways involved in delayed neuroinflammation that can be targeted at clinically relevant time points. The field of stroke research has benefited from experiments characterizing the temporal expression profiles of candidate cytokines, chemokines, matrix metalloproteinases, and other putative pro-inflammatory molecules. Yet, these data have offered only a glimpse into the complex pathological sequelae and have not advanced the treatment of neuropathies. Upon recognition that peripheral immune cell activation is involved in penumbral expansion, the spleen has emerged as a novel target that mediates the peripheral immune response and promotes pro-inflammatory injury. Although the precise mechanisms have yet to be elucidated, accumulated evidence demonstrates that focal cerebral ischemia alters cytokine, chemokine, and immune cell profiles in the spleen. Additionally, removal of this peripheral lymphoid organ is neuroprotective, and the efficacy of several protective therapies has been linked to actions at the level of the spleen. Future experiments aimed at identifying the splenic lymphocyte populations that respond to ischemic stroke, as well as their signaling mechanisms, are critical in developing novel therapies.

15.
Transl Stroke Res ; 1(2): 135-45, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20563232

ABSTRACT

Many pharmacological treatments for stroke have afforded protection in rodent models but failed to show efficacy in clinical trials. This discrepancy may be due to the lack of long-term functional studies. Previously, delayed administration of the sigma receptor agonist 1,3-di-o-tolylguanidine (DTG) reduced infarct volume after middle cerebral artery occlusion (MCAO) in rats. The present study was conducted to determine whether the protective effects of DTG lead to improvements in behavioral functioning. Rats were subjected to MCAO and administered 7.5, 1.5, or 0.75 mg/kg DTG beginning 24 h post-surgery. Histological outcomes (96 h, 2 weeks, and 5 weeks) were compared with performance on a series of behavioral tests (2 and 4 weeks). Fluoro-Jade staining and immunohistochemistry were used to assess infarct volume and immune cell recruitment. All doses significantly reduced infarct volume and perturbation of striatal white matter tracts at 96 h. These reductions were associated with decreased numbers of CD11b-positive amoeboid microglia/macrophages. Despite short-term efficacy, DTG failed to improve behavioral outcomes or reduce infarct volumes after 96 h. While DTG may prove beneficial as a short-term therapy, these data highlight the importance of long-term functional recovery when evaluating novel therapies to treat stroke.

16.
J Neurosci Res ; 88(6): 1213-22, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19998484

ABSTRACT

Secondary neurodegeneration resulting from stroke is mediated by delayed proinflammatory signaling and immune cell activation. Although it remains unknown which cell surface markers signify a proinflammatory phenotype, increased isolectin binding occurs on CD11b-expressing immune cells within injured brain tissue. Several reports have confirmed the efficacy of human umbilical cord blood (HUCB) cell therapy in reducing ischemic injury in rat after middle cerebral artery occlusion (MCAO), and these effects were attributed in part to dampened neuroinflammation. The present study examined the time course of lectin binding to cells of microglia/macrophage lineage within 96 hr after MCAO and whether delayed HUCB cell treatment alters the migration and/or morphological characteristics of these cells throughout the period of infarct expansion. Isolectin binding was up-regulated in response to injury, was maximal at 96 hr, and colocalized with cells that expressed the putative proinflammatory markers MMP-9 and nitric oxide. Isolectin-tagged fluorescence was also significantly increased at 72 hr and localized to greater numbers of amoeboid, CD11b-expressing cells relative to 51 hr. Treatment with 1 x 10(6) HUCB cells significantly reduced total lectin binding at 72 hr, as well as the total area occupied by lectin-tagged fluorescence at both 51 and 72 hr, relative to vehicle-treated controls. This effect was accompanied by a shift in the morphology of CD11b-positive cells from amoeboid to ramified shape. These data indicate that HUCB cell therapy suppressed the recruitment of proinflammatory, isolectin-binding cells during the period of infarct expansion, thus offering a potential mechanism for the protective effects of HUCB cell therapy.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/therapy , Macrophages/physiology , Microglia/physiology , Animals , CD11b Antigen/metabolism , Cell Movement , Humans , In Vitro Techniques , Infarction, Middle Cerebral Artery/pathology , Lectins/metabolism , Macrophages/pathology , Matrix Metalloproteinase 9/metabolism , Microglia/pathology , Neuroimmunomodulation , Nitric Oxide/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Time Factors
17.
J Neuroinflammation ; 6: 13, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19368723

ABSTRACT

Exposure to hypoxic-ischemic insults during the neonatal or perinatal developmental periods produces various forms of pathology. Injuries that occur in response to these events often manifest as severe cognitive and/or motor disturbances over time. Due to difficulties regarding the early diagnosis and treatment of hypoxic-ischemic injury, there is a growing need for effective therapies that can be delivered at delayed time points. Much of the research into mechanisms of neural injury has focused on molecular targets associated with excitotoxicity and free oxygen radicals. Despite repeated success in animal models, these compounds have failed to show efficacy in clinical trials. Increasing evidence indicates that hypoxic-ischemic injury in the neonate is progressive, and the resulting neuropathies are linked to the activation of neuroinflammatory processes that occur in response to the initial wave of cell death. Understanding this latter response, therefore, will be critical in the development of novel therapies to block the progression of the injury. In this review, we summarize emerging concepts from rodent models concerning the regulation of various cytokines, chemokines, and matrix metalloproteinases in response to ischemia, and the various ways in which the delayed neuroinflammatory response may contribute to the progressive nature of neonatal hypoxic-ischemic injury in rat. Finally, we discuss data that supports the potential to target these neuroinflammatory signals at clinically relevant time points.


Subject(s)
Brain/physiopathology , Hypoxia-Ischemia, Brain/physiopathology , Matrix Metalloproteinases/metabolism , Neuroimmunomodulation , Animals , Blood-Brain Barrier/physiopathology , Brain/pathology , Cytokines/metabolism , Disease Progression , Extracellular Matrix/metabolism , Free Radicals/metabolism , Humans , Hypoxia-Ischemia, Brain/drug therapy , Infant, Newborn , Matrix Metalloproteinase Inhibitors , Time Factors
18.
Exp Neurol ; 218(1): 47-55, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19371742

ABSTRACT

Recent studies have highlighted the involvement of the peripheral immune system in delayed cellular degeneration after stroke. In the permanent middle cerebral artery occlusion (MCAO) model of stroke, the spleen decreases in size. This reduction occurs through the release of splenic immune cells. Systemic treatment with human umbilical cord blood cells (HUCBC) 24 h post-stroke blocks the reduction in spleen size while significantly reducing infarct volume. Splenectomy 2 weeks prior to MCAO also reduces infarct volume, further demonstrating the detrimental role of this organ in stroke-induced neurodegeneration. Activation of the sympathetic nervous system after MCAO results in elevated catecholamine levels both at the level of the spleen, through direct splenic innervation, and throughout the systemic circulation upon release from the adrenal medulla. These catecholamines bind to splenic alpha and beta adrenoreceptors. This study examines whether catecholamines regulate the splenic response to stroke. Male Sprague-Dawley rats either underwent splenic denervation 2 weeks prior to MCAO or received injections of carvedilol, a pan adrenergic receptor blocker, prazosin, an alpha1 receptor blocker, or propranolol, a beta receptor blocker. Denervation was confirmed by reduced splenic expression of tyrosine hydroxylase. Denervation prior to MCAO did not alter infarct volume or spleen size. Propranolol treatment also had no effects on these outcomes. Treatment with either prazosin or carvedilol prevented the reduction in spleen size, yet only carvedilol significantly reduced infarct volume (p < 0.05). These results demonstrate that circulating blood borne catecholamines regulate the splenic response to stroke through the activation of both alpha and beta adrenergic receptors.


Subject(s)
Infarction, Middle Cerebral Artery/pathology , Receptors, Adrenergic/metabolism , Spleen/immunology , Spleen/pathology , Adrenergic Antagonists/pharmacology , Adrenergic alpha-Antagonists , Adrenergic beta-Antagonists/pharmacology , Animals , Carbazoles/pharmacology , Carvedilol , Cytokines/metabolism , Denervation/methods , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fetal Blood/cytology , Flow Cytometry/methods , Humans , Male , Propanolamines/pharmacology , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley , Spleen/drug effects , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
19.
J Neuroinflammation ; 5: 34, 2008 Aug 11.
Article in English | MEDLINE | ID: mdl-18694515

ABSTRACT

BACKGROUND: Hypoxia-ischemia (H-I) can produce widespread neurodegeneration and deep cerebral white matter injury in the neonate. Resident microglia and invading leukocytes promote lesion progression by releasing reactive oxygen species, proteases and other pro-inflammatory mediators. After injury, expression of the gelatin-degrading matrix metalloproteinases (MMPs), MMP-2 and MMP-9, are thought to result in the proteolysis of extracellular matrix (ECM), activation of cytokines/chemokines, and the loss of vascular integrity. Thus, therapies targeting ECM degradation and progressive neuroinflammation may be beneficial in reducing H-I - induced neuropathy. Minocycline has MMP-inhibitory properties and is both anti-inflammatory and neuroprotective. AG3340 (prinomastat) is an MMP inhibitor with high selectivity for the gelatinases. The purpose of this study was to determine whether these compounds could limit H-I--induced injury when administered at a delayed time point. METHODS: Sprague-Dawley rats were exposed to H-I at postnatal day 7 (P7), consisting of unilateral carotid artery ligation followed by 90 min exposure to 8% O2. Minocycline, AG3340, or vehicle were administered once daily for 6 days, beginning 24 hours after insult. Animals were sacrificed at P14 for neurohistological assessments. Immunohistochemistry was performed to determine the degree of reactive astrogliosis and immune cell activation/recruitment. Neural injury was detected using the Fluoro-Jade stain, a marker that identifies degenerating cells. RESULTS: CD11b and glial fibrillary acidic protein (GFAP) immunopositive cells increased in ipsilateral cortex after treatment with vehicle alone, demonstrating microglia/macrophage recruitment and reactive astrogliosis, respectively. Fluoro-Jade staining was markedly increased throughout the fronto-parietal cortex, striatum and hippocampus. Treatment with minocycline or AG3340 inhibited microglia/macrophage recruitment, attenuated astrogliosis and reduced Fluoro-Jade staining when compared to vehicle alone. CONCLUSION: The selective gelatinase inhibitor AG3340 showed equal efficacy in reducing neural injury and dampening neuroinflammation when compared to the anti-inflammatory compound minocycline. Thus, MMP-2 and MMP-9 may be viable therapeutic targets to treat neonatal brain injury.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/prevention & control , Hypoxia-Ischemia, Brain/pathology , Matrix Metalloproteinase Inhibitors , Minocycline/therapeutic use , Organic Chemicals/therapeutic use , Animals , Animals, Newborn , Brain Injuries/etiology , Brain Injuries/pathology , Enzyme Inhibitors/therapeutic use , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/metabolism , Macrophages/cytology , Macrophages/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Microglia/cytology , Microglia/metabolism , Neuroprotective Agents/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley
20.
Exp Neurol ; 210(2): 428-40, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18178186

ABSTRACT

Aggregating proteoglycans (PG) bearing chondroitin sulfate (CS) side chains associate with hyaluronan and various secreted proteins to form a complex of extracellular matrix (ECM) that inhibits neural plasticity in the central nervous system (CNS). Chondroitinase treatment depletes PGs of their CS side chains and enhances neurite extension. Increasing evidence from in vivo models indicates that proteolytic cleavage of the PG core protein by members of the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family of glutamyl-endopeptidases also promotes neural plasticity. The purpose of this study was to determine whether proteolytic action of the ADAMTSs influences neurite outgrowth in cultured neurons. Transfection of primary rat neurons with ADAMTS4 cDNA induced longer neurites, whether the neurons were grown on a monolayer of astrocytes that secrete inhibitory PGs or on laminin/poly-L-lysine substrate alone. Similar results were found when neurons were transfected with a construct encoding a proteolytically inactive, point mutant of ADAMTS4. Addition of recombinant ADAMTS4 or ADAMTS5 protein to immature neuronal cultures also enhanced neurite extension in a dose-dependent manner, an effect demonstrated to be dependent on the activation of MAP ERK1/2 kinase. These results suggest that ADAMTS4 enhances neurite outgrowth via a mechanism that does not require proteolysis but is dependent on activation of the MAP kinase cascade. Thus a model to illustrate multimodal ADAMTS activity would entail proteolysis of CS-bearing PGs to create a loosened matrix environment more favorable for neurite outgrowth, and enhanced neurite outgrowth directly stimulated by ADAMTS signaling at the cell surface.


Subject(s)
ADAM Proteins/metabolism , Neurites/physiology , Neurons/cytology , Procollagen N-Endopeptidase/metabolism , Signal Transduction/physiology , ADAM Proteins/administration & dosage , ADAMTS4 Protein , ADAMTS5 Protein , Analysis of Variance , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/drug effects , Brain/cytology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Mutation/physiology , Neurites/drug effects , Procollagen N-Endopeptidase/administration & dosage , Rats , Rats, Sprague-Dawley , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...