Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Inflamm (Lond) ; 13(1): 29, 2016.
Article in English | MEDLINE | ID: mdl-27563282

ABSTRACT

BACKGROUND: Interleukin (IL)-1ß is involved in the pathology of intervertebral disc degeneration. Under normal conditions, IL-1ß is present in cells in an inactive form (pro-IL-1ß). However, under pathological conditions, pro-IL-1ß is turned into its active form (IL-1ß) by the inflammasome, a multi-protein complex of the innate immune response that activates caspase-1. Under conditions of degeneration, the disc experiences an environment of increased acidification. However, the implications of acidification on the innate immune response remain poorly explored. METHODS: Here we have studied how pH changes in human nucleus pulposus cells affect inflammasome activation by immunoblot analysis of protein lysates obtained from nucleus pulposus cells that were exposed to different pH levels in culture. RESULTS: In this study, we have found that in nucleus pulposus cells, with increased acidification, there was a decrease in inflammasome activation consistent with lower levels of active IL-1ß. However, this effect at a pH of 6.5, the lowest pH level tested, was abrogated when cells were treated with IL-1ß. CONCLUSIONS: Taken together, these findings suggest that the inflammatory response through IL-1ß experienced by the human disc is not initiated in nucleus pulposus cells when the stimulus is acidification.

2.
J Neurochem ; 136(3): 492-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26490364

ABSTRACT

Periodic treatments with estrogen receptor subtype-ß (ER-ß) agonist reduce post-ischemic hippocampal injury in ovariectomized rats. However, the underlying mechanism of how ER-ß agonists protect the brain remains unknown. Global cerebral ischemia activates the innate immune response, and a key component of the innate immune response is the inflammasome. This study tests the hypothesis that ER-ß regulates inflammasome activation in the hippocampus, thus reducing ischemic hippocampal damage in reproductively senescent female rats that received periodic ER-ß agonist treatments. First, we determined the effect of hippocampal ER-ß silencing on the expression of the inflammasome proteins caspase 1, apoptosis-associated speck-like protein containing a CARD (ASC), and interleukin (IL)-1ß. Silencing of ER-ß attenuated 17ß-estradiol mediated decrease in caspase 1, ASC, and IL-1ß. Next, we tested the hypothesis that periodic ER-ß agonist treatment reduces inflammasome activation and ischemic damage in reproductively senescent female rats. Periodic ER-ß agonist treatments significantly decreased inflammasome activation and increased post-ischemic live neuronal counts by 32% (p < 0.05) as compared to the vehicle-treated, reproductively senescent rats. Current findings demonstrated that ER-ß activation regulates inflammasome activation and protects the brain from global ischemic damage in reproductively senescent female rats. Further investigation on the role of a periodic ER-ß agonist regimen to reduce the innate immune response in the brain could help reduce the incidence and the impact of global cerebral ischemia in post-menopausal women. We propose that estrogen receptor subtype-ß (ER-ß) activation regulates inflammasome activation and protects the brain from global ischemic damage in reproductively senescent female rats.


Subject(s)
Aging , Brain Ischemia/complications , Estrogen Receptor beta/metabolism , Hippocampus/metabolism , Inflammasomes/metabolism , Signal Transduction/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Hippocampus/drug effects , Hippocampus/pathology , Immunity, Innate/drug effects , NAD/pharmacology , Neurons/drug effects , Neurons/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Ovariectomy , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
3.
J Inflamm (Lond) ; 12: 52, 2015.
Article in English | MEDLINE | ID: mdl-26379474

ABSTRACT

BACKGROUND: Focal cerebral ischemia induces an inflammatory response that when exacerbated contributes to deleterious outcomes. The molecular basis regarding the regulation of the innate immune response after focal cerebral ischemia remains poorly understood. METHODS: In this study we examined the expression of retinoic acid-inducible gene (RIG)-like receptor-I (RIG-I) and its involvement in regulating inflammation after ischemia in the brain of rats subjected to middle cerebral artery occlusion (MCAO). In addition, we studied the regulation of RIG-I after oxygen glucose deprivation (OGD) in astrocytes in culture. RESULTS: In this study we show that in the hippocampus of rats, RIG-I and IFN-α are elevated after MCAO. Consistent with these results was an increased in RIG-I and IFN-α after OGD in astrocytes in culture. These data are consistent with immunohistochemical analysis of hippocampal sections, indicating that in GFAP-positive cells there was an increase in RIG-I after MCAO. In addition, in this study we have identified n-propyl gallate as an inhibitor of IFN-α signaling in astrocytes. CONCLUSION: Our findings suggest a role for RIG-I in contributing to the innate immune response after focal cerebral ischemia.

4.
J Neurotrauma ; 32(4): 228-36, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25111533

ABSTRACT

The innate immune response contributes to the inflammatory activity after traumatic brain injury (TBI). In the present study we identify macrophage-inducible C-type lectin (mincle) as a pattern recognition receptor that contributes to innate immunity in neurons after TBI. Here we report that mincle is activated by SAP130 in cortical neurons in culture, resulting in production of the inflammatory cytokine TNF. In addition, mincle and SAP130 are elevated in the brain and cerebrospinal fluid of humans after TBI and the brain of rodents after fluid percussion brain injury. Thus, these findings suggest the involvement of mincle to the pathology of TBI. Importantly, blocking mincle with a neutralizing antibody against mincle in cortical neurons in culture treated with SAP130 resulted in inhibition of mincle signaling and decreased TNF production. Therefore, our findings identify mincle as a contributor to the inflammatory response after TBI.


Subject(s)
Brain Injuries/immunology , Immunity, Innate/immunology , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , Signal Transduction/immunology , Adolescent , Adult , Animals , Female , Humans , Immunoblotting , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged , Rats , Rats, Sprague-Dawley , Young Adult
5.
J Neuroinflammation ; 11: 67, 2014 Apr 02.
Article in English | MEDLINE | ID: mdl-24694234

ABSTRACT

BACKGROUND: Neuroinflammation plays a critical role in the pathogenesis of Alzheimer's disease (AD) and involves activation of the innate immune response via recognition of diverse stimuli by pattern recognition receptors (PRRs). The inflammatory inducers and precise innate signaling pathway contributing to AD pathology remain largely undefined. RESULTS: In the present study we analyzed expression levels of innate immune proteins in temporal and occipital cortices from preclinical (no cognitive impairment, NCI, N = 22) to mild cognitive impairment (MCI, N = 20) associated with AD pathology (N = 20) and AD patients (N = 23). We found that retinoic acid-inducible gene-I (RIG-1) is significantly elevated in the temporal cortex and plasma in patients with MCI. In addition, primary human astrocytes stimulated with the RIG-1 ligand 5'ppp RNA showed increased expression of amyloid precursor protein (APP) and amyloid-ß (Aß), supporting the idea that RIG-1 is involved in the pathology of MCI associated with early progression to AD. CONCLUSION: These findings suggest that RIG-1 may play a critical role in incipient AD.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/pathology , DEAD-box RNA Helicases/metabolism , Occipital Lobe/metabolism , Temporal Lobe/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Cognitive Dysfunction/blood , Cognitive Dysfunction/pathology , DEAD Box Protein 58 , Dose-Response Relationship, Drug , Female , Gene Expression/drug effects , Humans , Interferon Regulatory Factor-3/metabolism , Male , Middle Aged , Occipital Lobe/cytology , Peptide Fragments/pharmacology , RNA, Viral/pharmacology , Receptors, Immunologic , Temporal Lobe/cytology
6.
J Cereb Blood Flow Metab ; 34(4): 621-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24398937

ABSTRACT

The central nervous system (CNS) is an active participant in the innate immune response to infection and injury. In these studies, we show embryonic cortical neurons express a functional, deoxyribonucleic acid (DNA)-responsive, absent in melanoma 2 (AIM2) inflammasome that activates caspase-1. Neurons undergo pyroptosis, a proinflammatory cell death mechanism characterized by the following: (a) oligomerization of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC); (b) caspase-1 dependency; (c) formation of discrete pores in the plasma membrane; and (d) release of the inflammatory cytokine interleukin-1ß (IL-1ß). Probenecid and Brilliant Blue FCF, inhibitors of the pannexin1 channel, prevent AIM2 inflammasome-mediated cell death, identifying pannexin1 as a cell death effector during pyroptosis and probenecid as a novel pyroptosis inhibitor. Furthermore, we show activation of the AIM2 inflammasome in neurons by cerebrospinal fluid (CSF) from traumatic brain injury (TBI) patients and oligomerization of ASC. These findings suggest neuronal pyroptosis is an important cell death mechanism during CNS infection and injury that may be attenuated by probenecid.


Subject(s)
Apoptosis , Inflammasomes/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Adolescent , Adult , Aged , Animals , Apoptosis/drug effects , Apoptosis/immunology , Apoptosis Regulatory Proteins/metabolism , Brain Injuries/cerebrospinal fluid , Brain Injuries/immunology , Brain Injuries/metabolism , Brain Injuries/pathology , Caspase 1/metabolism , Cell Culture Techniques , Cell Death , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , DNA-Binding Proteins , Female , Humans , Immunity, Innate/drug effects , Inflammasomes/immunology , Male , Middle Aged , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Poly dA-dT/pharmacology , Probenecid/pharmacology , Rats , Rats, Sprague-Dawley , Young Adult
7.
Stroke ; 42(5): 1404-11, 2011 May.
Article in English | MEDLINE | ID: mdl-21454816

ABSTRACT

BACKGROUND AND PURPOSE: Stroke and heart disease are the most serious complications of diabetes accounting for >65% of mortality among diabetics. Although intensive insulin therapy has significantly improved the prognosis of diabetes and its complications, it is associated with an elevated risk of recurrent hypoglycemia (RH). We tested the hypothesis that RH exacerbates cerebral ischemic damage in a rodent model of diabetes. METHOD: We determined the extent of neuronal death in CA1 hippocampus after global cerebral ischemia in control and streptozotocin-induced diabetic rats. Diabetic animals included an insulin-treated streptozotocin-diabetic (ITD) group and a group of ITD rats exposed also to 10 episodes of hypoglycemia (ITD+recurrent hypoglycemia: RH). Hypoglycemia (55 to 65 mg/dL blood glucose) was induced twice daily for 5 consecutive days. RESULTS: As expected, uncontrolled diabetes (streptozotocin-diabetes, untreated animals) resulted in a 70% increase in ischemic damage as compared with the control group. Insulin treatment was able to lower ischemic damage by 64% as compared with the diabetic group. However, ITD+RH rats had 44% more damage when compared with the ITD group. We also observed that free radical release from mitochondria is increased in ITD+RH rats. CONCLUSIONS: This is the first report on the impact of RH in exacerbating cerebral ischemic damage in diabetic animals. Our results suggest that increased free radical release from mitochondria may be responsible for observed increased ischemic damage in ITD+RH rats. RH thus may be an unexplored but important factor responsible for increased ischemic damage in diabetes.


Subject(s)
Brain Ischemia/pathology , Brain Ischemia/physiopathology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , Hypoglycemia/complications , Hypoglycemia/physiopathology , Animals , Brain Ischemia/etiology , CA1 Region, Hippocampal/pathology , Cell Death , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Free Radicals/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Male , Mitochondria/metabolism , Neurons/pathology , Rats , Rats, Wistar , Recurrence , Risk Factors , Streptozocin
SELECTION OF CITATIONS
SEARCH DETAIL
...