Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Rep ; 8(1): 16165, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385782

ABSTRACT

Accumulation of alpha-synuclein (ASYN) in neurons and other CNS cell types may contribute to the underlying pathology of synucleinopathies including Parkinson's disease (PD), dementia with Lewy bodies (DLB) and Multiple Systems Atrophy (MSA). In support of this hypothesis for PD, ASYN immunopositive aggregates are a prominent pathological feature of PD, and mutations and gene multiplications of human wild type (WT) ASYN cause rare familial autosomal-dominant forms of PD. Targeted therapeutics that reduce the accumulation of ASYN could prevent or slow the neurodegenerative processes in PD and other synucleinopathies. NPT200-11 is a novel small molecule inhibitor of ASYN misfolding and aggregation. The effects of NPT200-11 on ASYN neuropathology were evaluated in animal models over expressing human alpha synuclein. Longitudinal studies using retinal imaging in mice expressing a hASYN::GFP fusion protein revealed that 2 months of once daily administration of NPT200-11 (5 mg/kg IP) resulted in a time-dependent and progressive reduction in retinal ASYN pathology. The effects of NPT200-11 on ASYN pathology in cerebral cortex and on other disease-relevant endpoints was evaluated in the Line 61 transgenic mouse model overexpressing human wild type ASYN. Results from these studies demonstrated that NPT200-11 reduced alpha-synuclein pathology in cortex, reduced associated neuroinflammation (astrogliosis), normalized striatal levels of the dopamine transporter (DAT) and improved motor function. To gain insight into the relationship between dose, exposure, and therapeutic benefit pharmacokinetic studies were also conducted in mice. These studies demonstrated that NPT200-11 is orally bioavailable and brain penetrating and established target plasma and brain exposures for future studies of potential therapeutic benefit.


Subject(s)
Inflammation/drug therapy , Parkinson Disease/drug therapy , Piperidines/pharmacology , Protein Aggregation, Pathological/drug therapy , Pyrazines/pharmacology , Pyrimidines/pharmacology , alpha-Synuclein/genetics , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Lewy Body Disease/drug therapy , Lewy Body Disease/genetics , Lewy Body Disease/pathology , Mice , Mice, Transgenic , Multiple System Atrophy/drug therapy , Multiple System Atrophy/genetics , Multiple System Atrophy/pathology , Neurons/drug effects , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Piperidines/therapeutic use , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Folding/drug effects , Pyrazines/therapeutic use , Pyrimidines/therapeutic use , Retina/diagnostic imaging , Retina/drug effects , Retina/pathology , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/chemistry
2.
FASEB J ; 29(6): 2315-26, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25681462

ABSTRACT

Conventional T (Tcon) cells and Foxp3(+) T-regulatory (Treg) cells are thought to have differing metabolic requirements, but little is known of mitochondrial functions within these cell populations in vivo. In murine studies, we found that activation of both Tcon and Treg cells led to myocyte enhancer factor 2 (Mef2)-induced expression of genes important to oxidative phosphorylation (OXPHOS). Inhibition of OXPHOS impaired both Tcon and Treg cell function compared to wild-type cells but disproportionally affected Treg cells. Deletion of Pgc1α or Sirt3, which are key regulators of OXPHOS, abrogated Treg-dependent suppressive function and impaired allograft survival. Mef2 is inhibited by histone/protein deacetylase-9 (Hdac9), and Hdac9 deletion increased Treg suppressive function. Hdac9(-/-) Treg showed increased expression of Pgc1α and Sirt3, and improved mitochondrial respiration, compared to wild-type Treg cells. Our data show that key OXPHOS regulators are required for optimal Treg function and Treg-dependent allograft acceptance. These findings provide a novel approach to increase Treg function and give insights into the fundamental mechanisms by which mitochondrial energy metabolism regulates immune cell functions in vivo.


Subject(s)
Energy Metabolism/immunology , Forkhead Transcription Factors/immunology , Graft Survival/immunology , Mitochondria/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Blotting, Western , Energy Metabolism/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Graft Survival/genetics , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Histone Deacetylases/metabolism , MEF2 Transcription Factors/immunology , MEF2 Transcription Factors/metabolism , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Repressor Proteins/immunology , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 3/genetics , Sirtuin 3/immunology , Sirtuin 3/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
3.
Neuron ; 82(2): 380-97, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24742461

ABSTRACT

The colony-stimulating factor 1 receptor (CSF1R) is a key regulator of myeloid lineage cells. Genetic loss of the CSF1R blocks the normal population of resident microglia in the brain that originates from the yolk sac during early development. However, the role of CSF1R signaling in microglial homeostasis in the adult brain is largely unknown. To this end, we tested the effects of selective CSF1R inhibitors on microglia in adult mice. Surprisingly, extensive treatment results in elimination of ∼99% of all microglia brain-wide, showing that microglia in the adult brain are physiologically dependent upon CSF1R signaling. Mice depleted of microglia show no behavioral or cognitive abnormalities, revealing that microglia are not necessary for these tasks. Finally, we discovered that the microglia-depleted brain completely repopulates with new microglia within 1 week of inhibitor cessation. Microglial repopulation throughout the CNS occurs through proliferation of nestin-positive cells that then differentiate into microglia.


Subject(s)
Adult Stem Cells/physiology , Brain/metabolism , Microglia/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/physiology , Adult Stem Cells/drug effects , Animals , Animals, Newborn , Brain/cytology , Brain/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Signal Transduction/drug effects , Time Factors
4.
Ther Hypothermia Temp Manag ; 3(2): 74-83, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23781399

ABSTRACT

Therapeutic hypothermia has shown neuroprotective promise, but whether it can be used to improve outcome in stroke has yet to be determined in patients. Recombinant tissue plasminogen activator (rt-PA) is only given to a minority of patients with acute ischemic stroke, and is not without risk, namely significant brain hemorrhage.We explored whether mild hypothermia, in combination with rt-PA, influences the safety of rt-PA. Mice were subjected to middle cerebral artery occlusion (MCAO) using a filament model, followed by 24 hours reperfusion.Two paradigms were studied. In the first paradigm, cooling and rt-PA treatment began at the same time upon reperfusion, whereas in the second paradigm, cooling began soon after ischemia onset, and rt-PA began after rewarming and upon reperfusion. Experimental groups included: tPA treatment at normothermia (37°C), rt-PA treatment at hypothermia (33°C), no rt-PA at normothermia, and no rt-PA treatment at hypothermia. Infarct size, neurological deficit scores, blood brain barrier (BBB) permeability, brain hemorrhage, and expression of endogenous tissue plasminogen activator (tPA) and its inhibitor, plasminogen activator inhibitor (PAI-1) were assessed. For both paradigms, hypothermia reduced infarct size and neurological deficits compared to normothermia, regardless of whether rt-PA was given. rt-PA treatment increased brain hemorrhage and BBB disruption compared to normothermia, and this was prevented by cooling. However, mortality was higher when rt-PA and cooling were administered at the same time, beginning 1­2 hours post MCAO. Endogenous tPA expression was reduced in hypothermic mice, whereas PAI-1 levels were unchanged by cooling. In the setting of rt-PA treatment, hypothermia reduces brain hemorrhage, and BBB disruption, suggesting that combination therapy with mild hypothermia and rt-PA appears safe.

5.
Cell ; 151(2): 333-343, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063123

ABSTRACT

Maternal inheritance of mtDNA is the rule in most animals, but the reasons for this pattern remain unclear. To investigate the consequence of overriding uniparental inheritance, we generated mice containing an admixture (heteroplasmy) of NZB and 129S6 mtDNAs in the presence of a congenic C57BL/6J nuclear background. Analysis of the segregation of the two mtDNAs across subsequent maternal generations revealed that proportion of NZB mtDNA was preferentially reduced. Ultimately, this segregation process produced NZB-129 heteroplasmic mice and their NZB or 129 mtDNA homoplasmic counterparts. Phenotypic comparison of these three mtDNA lines demonstrated that the NZB-129 heteroplasmic mice, but neither homoplasmic counterpart, had reduced activity, food intake, respiratory exchange ratio; accentuated stress response; and cognitive impairment. Therefore, admixture of two normal but different mouse mtDNAs can be genetically unstable and can produce adverse physiological effects, factors that may explain the advantage of uniparental inheritance of mtDNA.


Subject(s)
DNA, Mitochondrial/genetics , Mice/genetics , Animals , Behavior, Animal , Cognition , Female , Inheritance Patterns , Male , Mice/physiology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NZB , Species Specificity
6.
PLoS One ; 7(8): e42665, 2012.
Article in English | MEDLINE | ID: mdl-22912719

ABSTRACT

The incidence of Alzheimer's disease increases in people who have had an ischemic episode. Furthermore, APP expression is increased following ischemic or hypoxic conditions, as is the production of the Aß peptide. To address the question of why APP and Aß are increased in hypoxic and ischemic conditions we induced an ischemic episode in APP knockout mice (APP-/-) and BACE1 knockout mice (BACE-/-). We find that both APP-/- and BACE-/- mice have a dramatically increased risk of mortality as a result of cerebral ischemia. Furthermore, APP knockout mice have reduced cerebral blood flow in response to hypoxia, while wild-type mice maintain or increase cerebral blood flow to the same conditions. The transcription factor, serum response factor (SRF), and calcium-binding molecule, calsequestrin, both involved in vascular regulation, are significantly altered in the brains of APP-/- mice compared to wild type controls. These results show that APP regulates cerebral blood flow in response to hypoxia, and that it, and its cleavage fragments, are crucial for rapid adaptation to ischemic conditions.


Subject(s)
Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Brain Ischemia/genetics , Brain Ischemia/mortality , Gene Knockout Techniques , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/deficiency , Aspartic Acid Endopeptidases/genetics , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Calsequestrin/metabolism , Cell Hypoxia , Cerebrovascular Circulation/genetics , Hypercapnia/physiopathology , Mice , Oxidative Stress/genetics , Peptide Fragments/metabolism , Proteolysis , Serum Response Factor/metabolism
7.
J Immunol ; 187(12): 6539-49, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22095718

ABSTRACT

Inflammation is a key pathological hallmark of Alzheimer's disease (AD), although its impact on disease progression and neurodegeneration remains an area of active investigation. Among numerous inflammatory cytokines associated with AD, IL-1ß in particular has been implicated in playing a pathogenic role. In this study, we sought to investigate whether inhibition of IL-1ß signaling provides disease-modifying benefits in an AD mouse model and, if so, by what molecular mechanisms. We report that chronic dosing of 3xTg-AD mice with an IL-1R blocking Ab significantly alters brain inflammatory responses, alleviates cognitive deficits, markedly attenuates tau pathology, and partly reduces certain fibrillar and oligomeric forms of amyloid-ß. Alterations in inflammatory responses correspond to reduced NF-κB activity. Furthermore, inhibition of IL-1 signaling reduces the activity of several tau kinases in the brain, including cdk5/p25, GSK-3ß, and p38-MAPK, and also reduces phosphorylated tau levels. We also detected a reduction in the astrocyte-derived cytokine, S100B, and in the extent of neuronal Wnt/ß-catenin signaling in 3xTg-AD brains, and provided in vitro evidence that these changes may, in part, provide a mechanistic link between IL-1 signaling and GSK-3ß activation. Taken together, our results suggest that the IL-1 signaling cascade may be involved in one of the key disease mechanisms for AD.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , Cognition Disorders/immunology , Interleukin-1beta/antagonists & inhibitors , Neurons/immunology , Signal Transduction/immunology , beta Catenin/physiology , tau Proteins/antagonists & inhibitors , Alzheimer Disease/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Cognition Disorders/genetics , Cognition Disorders/pathology , Disease Models, Animal , Female , Humans , Interleukin-1beta/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/physiology , Neurons/metabolism , Neurons/pathology , Receptors, Interleukin-1/antagonists & inhibitors , S100 Calcium Binding Protein beta Subunit , S100 Proteins/antagonists & inhibitors , S100 Proteins/physiology , Signal Transduction/genetics , beta Catenin/antagonists & inhibitors , tau Proteins/physiology
8.
Ann Biomed Eng ; 39(4): 1349-57, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21331663

ABSTRACT

Extensive changes in neural tissue structure and function accompanying Alzheimer's disease (AD) suggest that intrinsic signal optical imaging can provide new contrast mechanisms and insight for assessing AD appearance and progression. In this work, we report the development of a wide-field spatial frequency domain imaging (SFDI) method for non-contact, quantitative in vivo optical imaging of brain tissue composition and function in a triple transgenic mouse AD model (3xTg). SFDI was used to generate optical absorption and scattering maps at up to 17 wavelengths from 650 to 970 nm in 20-month-old 3xTg mice (n = 4) and age-matched controls (n = 6). Wavelength-dependent optical properties were used to form images of tissue hemoglobin (oxy-, deoxy-, and total), oxygen saturation, and water. Significant baseline contrast was observed with 13-26% higher average scattering values and elevated water content (52 ± 2% vs. 31 ± 1%); reduced total tissue hemoglobin content (127 ± 9 µM vs. 174 ± 6 µM); and lower tissue oxygen saturation (57 ± 2% vs. 69 ± 3%) in AD vs. control mice. Oxygen inhalation challenges (100% oxygen) resulted in increased levels of tissue oxy-hemoglobin (ctO(2)Hb) and commensurate reductions in deoxy-hemoglobin (ctHHb), with ~60-70% slower response times and ~7 µM vs. ~14 µM overall changes for 3xTg vs. controls, respectively. Our results show that SFDI is capable of revealing quantitative functional contrast in an AD model and may be a useful method for studying dynamic alterations in AD neural tissue composition and physiology.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Diagnostic Imaging/methods , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Biomedical Engineering , Brain/metabolism , Brain/pathology , Diagnostic Imaging/instrumentation , Disease Models, Animal , Hemoglobins/metabolism , Humans , Hyperoxia/metabolism , Hyperoxia/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Optical Phenomena , Oxygen/metabolism , Recombinant Proteins/genetics , Scattering, Radiation , tau Proteins/genetics
9.
Am J Pathol ; 177(1): 300-10, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20472896

ABSTRACT

Decreased blood flow to the brain in humans is associated with altered Alzheimer's disease (AD)-related pathology, although the underlying mechanisms by which hypoperfusion influences AD neuropathology remains unknown. To try to address this question, we developed an oligemic model of cerebral hypoperfusion in the 3xTg-AD mouse model of AD. We bilaterally and transiently occluded the common carotid artery and then examined the molecular and cellular pathways by which hypoperfusion influenced tau and amyloid-beta proteins. We report the novel finding that a single, mild, transient hypoperfusion insult acutely increases Abeta levels by enhancing beta-secretase protein expression. In contrast, transient hypoperfusion markedly decreases total tau levels, coincident with activation of macroautophagy and ubiquitin-proteosome pathways. Furthermore, we find that oligemia results in a significant increase specifically in tau phosphorylated at serine(212) and threonine(214), a tau epitope associated with paired helical filaments in AD patients. Despite the mild and transient nature of this hypoperfusion injury, the pattern of decreased total tau, altered phosphorylated tau, and increased amyloid-beta persisted for several weeks postoligemia. Our study indicates that a single, mild, cerebral hypoperfusion event produces profound and long lasting effects on both tau and amyloid-beta. This finding may have implications for the pathogenesis of AD, as it indicates for the first time that total tau and amyloid-beta are differentially impacted by mild hypoperfusion.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain Ischemia , Brain/metabolism , Brain/pathology , Cerebrovascular Circulation/physiology , Regional Blood Flow/physiology , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Biomarkers/metabolism , Brain/anatomy & histology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Humans , Lysosomes/metabolism , Mice , Mice, Transgenic
10.
J Neurochem ; 109(4): 1144-56, 2009 May.
Article in English | MEDLINE | ID: mdl-19302484

ABSTRACT

Following neuronal injury, microglia initiate repair by phagocytosing dead neurons without eliciting inflammation. Prior evidence indicates triggering receptor expressed by myeloid cells-2 (TREM2) promotes phagocytosis and retards inflammation. However, evidence that microglia and neurons directly interact through TREM2 to orchestrate microglial function is lacking. We here demonstrate that TREM2 interacts with endogenous ligands on neurons. Staining with TREM2-Fc identified TREM2 ligands (TREM2-L) on Neuro2A cells and on cultured cortical and dopamine neurons. Apoptosis greatly increased the expression of TREM2-L. Furthermore, apoptotic neurons stimulated TREM2 signaling, and an anti-TREM2 mAb blocked stimulation. To examine the interaction between TREM2 and TREM2-L in phagocytosis, we studied BV2 microglial cells and their engulfment of apoptotic Neuro2A. One of our anti-TREM2 mAb, but not others, reduced engulfment, suggesting the presence of a functional site on TREM2 interacting with neurons. Further, Chinese hamster ovary cells transfected with TREM2 conferred phagocytic activity of neuronal cells demonstrating that TREM2 is both required and sufficient for competent uptake of apoptotic neuronal cells. Finally, while TREM2-L are expressed on neurons, TREM2 is not; in the brain, it is found on microglia. TREM2 and TREM2-L form a receptor-ligand pair connecting microglia with apoptotic neurons, directing removal of damaged cells to allow repair.


Subject(s)
Apoptosis/physiology , Membrane Glycoproteins/physiology , Microglia/physiology , Neurons/physiology , Phagocytosis/physiology , Receptors, Immunologic/physiology , Animals , Antibodies/chemistry , CHO Cells , Cell Communication , Cell Separation , Cricetinae , Cricetulus , Lentivirus/genetics , Ligands , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/physiology , RNA, Messenger/genetics , Receptors, Immunologic/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transfection
11.
Neurobiol Dis ; 33(2): 301-12, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19063968

ABSTRACT

We previously showed that hypothermia attenuates inflammation in focal cerebral ischemia (FCI) by suppressing activating kinases of nuclear factor-kappa B (NFkappaB). Here we characterize the inflammatory response in global cerebral ischemia (GCI), and the influence of mild hypothermia. Rodents were subjected to GCI by bilateral carotid artery occlusion. The inflammatory response was accompanied by microglial activation, but not neutrophil infiltration, or blood brain barrier disruption. Mild hypothermia reduced CA1 damage, decreased microglial activation and decreased nuclear NFkappaB translocation and activation. Similar anti-inflammatory effects of hypothermia were observed in a model of pure brain inflammation that does not cause brain cell death. Primary microglial cultures subjected to oxygen glucose deprivation (OGD) or stimulated with LPS under hypothermic conditions also experienced less activation and less NFkappaB translocation. However, NFkappaB regulatory proteins were not affected by hypothermia. The inflammatory response following GCI and hypothermia's anti-inflammatory mechanism is different from that observed in FCI.


Subject(s)
Brain Ischemia/immunology , Brain Ischemia/therapy , Hypothermia, Induced , NF-kappa B/metabolism , Animals , Blood-Brain Barrier/physiology , Carotid Arteries , Cell Nucleus/metabolism , Cells, Cultured , Glucose/deficiency , Hippocampus/physiopathology , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/physiology , Neutrophils/physiology , Rats , Rats, Sprague-Dawley
12.
J Neurochem ; 106(2): 541-50, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18410517

ABSTRACT

Protection by mild hypothermia has previously been associated with better mitochondrial preservation and suppression of the intrinsic apoptotic pathway. It is also known that the brain may undergo apoptotic death via extrinsic, or receptor-mediated pathways, such as that triggered by Fas/FasL. Male Sprague-Dawley rats subjected to 2 h middle cerebral artery occlusion with 2 h intraischemic mild hypothermia (33 degrees C) were assayed for Fas, FasL and caspase-8 expression. Ischemia increased Fas, but decreased FasL by approximately 50-60% at 6 and 24 h post-insult. Mild hypothermia significantly reduced expression of Fas and processed caspase-8 both by approximately 50%, but prevented ischemia-induced FasL decreases. Fractionation revealed that soluble/shed FasL (sFasL) was decreased by hypothermia, while membrane-bound FasL (mFasL) increased. To more directly assess the significance of the Fas/FasL pathway in ischemic stroke, primary neuron cultures were exposed to oxygen glucose deprivation. Since FasL is cleaved by matrix metalloproteinases (MMPs), and mild hypothermia decreases MMP expression, treatment with a pan-MMP inhibitor also decreased sFasL. Thus, mild hypothermia is associated with reduced Fas expression and caspase-8 activation. Hypothermia prevented total FasL decreases, and most of it remained membrane-bound. These findings reveal new observations regarding the effect of mild hypothermia on the Fas/FasL and MMP systems.


Subject(s)
Fas Ligand Protein/metabolism , Gene Expression Regulation/physiology , Hypothermia, Induced/methods , Ischemia/therapy , Analysis of Variance , Animals , Caspase 8/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Embryo, Mammalian , Fas Ligand Protein/genetics , Glucose/deficiency , Hypoxia , Male , Matrix Metalloproteinases/pharmacology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , fas Receptor/metabolism
13.
J Nucl Med ; 48(11): 1822-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17942809

ABSTRACT

UNLABELLED: Minocycline is an antibiotic now recognized to have antiapoptotic and antiinflammatory properties. Because of these properties, minocycline may be of benefit in reducing neuronal apoptosis from ischemia and subsequent postischemic inflammation if administered soon after a stroke. We now explore the feasibility of using (99m)Tc-annexin V, an in vivo marker of apoptosis, with SPECT to monitor the antiapoptotic effects of minocycline therapy. METHODS: CB6/F1 adult male mice underwent unilateral distal middle cerebral artery occlusion (dMCA) occlusion and were imaged and sacrificed at 1, 3, 7, or 30 d after injury. Animals were given minocycline (or vehicle) 30 min and 12 h after dMCA occlusion and then given 22.5 mg/kg twice daily for up to 7 d. Before imaging, behavioral tests were performed to evaluate the neurologic function. After imaging, brains were collected for histology and assessed for the degree of apoptosis and microglial activation. RESULTS: (99m)Tc-Annexin V uptake in injured hemispheres was significantly decreased 2- to 3-fold by minocycline at all time points. Minocyline reduced infarct size as seen histologically and improved behavioral indices as late as 30 d. Infarct volume as seen histologically correlated with radiolabeled annexin V uptake seen by SPECT. In situ fluorescent microscopy demonstrated that annexin V bound primarily to neurons at 1 and 3 d, with a shift toward microglia by 7 and 30 d. CONCLUSION: We found that minocycline significantly reduces neuronal apoptosis and infarct size and improves neurologic outcome in mice after acute focal cortical ischemia.


Subject(s)
Annexin A5 , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Attack, Transient/prevention & control , Minocycline/therapeutic use , Neuroprotective Agents/therapeutic use , Organotechnetium Compounds , Animals , Annexin A5/chemistry , Apoptosis , Hydrazines/chemistry , Infarction, Middle Cerebral Artery/diagnostic imaging , Ischemic Attack, Transient/diagnostic imaging , Male , Mice , Microglia/diagnostic imaging , Nicotinic Acids/chemistry , Organotechnetium Compounds/chemistry , Radiopharmaceuticals , Tomography, Emission-Computed, Single-Photon
14.
Eur J Nucl Med Mol Imaging ; 33(5): 566-74, 2006 May.
Article in English | MEDLINE | ID: mdl-16477433

ABSTRACT

PURPOSE: The first aim of the study was to determine whether (99m)Tc-HYNIC-annexin V, a marker of cellular stress and apoptosis, can detect ischemic injury in patients with acute stroke. Secondly, we wished to test radiolabeled annexin's ability to monitor therapy in a rodent model of focal ischemic injury. METHODS: SPECT imaging of patients was performed between 1 and 2 h after intravenous injection of 30 mCi (1,110 MBq) of tracer. Eight MFL4 (anti-FasL) antibody-treated (400 microg i.p. days 0 and 3) and 21 control adult male Sprague-Dawley rats underwent small animal SPECT imaging with 5-10 mCi (185-370 MBq) of tracer, 1 and 6 days after a 2-h intraluminal thread occlusion of the left middle cerebral artery. RESULTS: Two patients with acute stroke had regions of multifocal annexin uptake that correlated with sites of restricted diffusion on MRI. Anti-FasL antibody treatment significantly reduced annexin uptake by 92% with a 60% decrease in the number of caspase-8 staining (apoptotic) neurons on day 1. On day 6, treated animals had an 80% reduction in tracer uptake with a 75% decrease in infarct size as compared with controls. Annexin uptake in controls and treated animals (day 6) linearly correlated with infarct size (r (2)=0.603, p=0.0036) and the number of TUNEL-positive (apoptotic) nuclei (r (2)=0.728, p=0.00084). CONCLUSION: Annexin imaging shows foci of increased uptake at sites of ischemic injury in patients with acute stroke. Annexin imaging can assess the effects of therapy for ischemic cerebral injury in rats, suggesting its potential as a non-invasive indicator of drug efficacy in future clinical trials.


Subject(s)
Annexin A5 , Antibodies, Monoclonal/therapeutic use , Brain Ischemia/diagnostic imaging , Brain Ischemia/drug therapy , Organotechnetium Compounds , Stroke/diagnostic imaging , Stroke/drug therapy , Acute Disease , Animals , Antibodies, Monoclonal, Murine-Derived , Brain Ischemia/etiology , Humans , Male , Neuroprotective Agents/therapeutic use , Prognosis , Radiopharmaceuticals , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Stroke/complications , Tomography, Emission-Computed, Single-Photon/methods , Treatment Outcome
15.
Neuropathology ; 25(4): 315-25, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16382780

ABSTRACT

Progression of HIV encephalitis (HIVE) is associated with neuronal damage and loss because of infiltration of infected and/or activated macrophages into the CNS. We have previously observed increased inactivation of the retinoblastoma susceptibility gene product (pRb) by phosphorylation in neurons and glia of HIVE and the simian model of HIVE (SIVE). To determine if other pRb family members are altered in response to increased macrophage-secreted factors, we investigated expression of pRb family members p107 and p130 in SIVE. Both p130 and p107 exhibited increased staining in macrophages, but not neurons, astrocytes or T-cells in SIVE. Increased p130 and p107 immunostaining was not limited to virally infected or PCNA-expressing macrophages. Most p107-positive staining was observed in perivascular macrophages, suggesting p107 may indicate macrophages at a specific stage of differentiation soon after migration. In contrast, cytoplasmic p130 was found in the majority of macrophages present in SIVE cases and may indicate activation as it was not seen in microglia in control CNS. These findings suggest that p107 and p130 are differentially expressed in CNS macrophage populations which may have multiple derivations and/or roles in lentiviral encephalitis.


Subject(s)
Brain/metabolism , Encephalitis/physiopathology , Macrophages/virology , Retinoblastoma-Like Protein p107/biosynthesis , Retinoblastoma-Like Protein p130/biosynthesis , Simian Acquired Immunodeficiency Syndrome/physiopathology , Animals , Astrocytes/immunology , Astrocytes/metabolism , Brain/immunology , Brain/pathology , Encephalitis/etiology , Encephalitis/immunology , Fluorescent Antibody Technique , Immunohistochemistry , Macaca mulatta , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Neurons/immunology , Neurons/metabolism , Simian Acquired Immunodeficiency Syndrome/complications , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
16.
J Cell Biochem ; 96(4): 728-40, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16088944

ABSTRACT

The E2F1 transcription factor can initiate proliferation or apoptosis, the latter by both transcription-dependent and -independent mechanisms. Recently, an E2F1 mutant lacking the DNA binding domain, E2F1(180-437), has been implicated in degradation of MDMX and MDM2 proteins via lysosomal proteases. MDM proteins block p53 dependent apoptosis by directly inhibiting p53 stability and function. Here we demonstrate E2F1(180-437) induces death in HEK293 cells independent of E2F1 transcriptional activation and p53 stabilization. E2F1(180-437) elevates the activity of the calcium-activated protease, calpain, which is required for E2F1 induced proteolysis of MDMX and E2F1 induced cell loss. To determine if E2F1 could be activating proteolysis via calpains in neurodegeneration, we examined MDMX immunofluorescence in simian immunodeficiency virus encephalitis (SIVE). We found a reciprocal relationship between E2F1 and MDMX staining: in SIVE where E2F1 immunostaining is increased, MDMX is decreased, while in controls where E2F1 immunostaining is low, MDMX is high. Together these experiments support a new function for E2F1 in the activation of calpain proteases and suggest a role for this pathway in SIVE.


Subject(s)
Calpain/metabolism , E2F1 Transcription Factor/metabolism , Encephalitis/complications , Encephalitis/pathology , Neurons/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Simian Acquired Immunodeficiency Syndrome/complications , Animals , Cell Cycle , Cell Death , Encephalitis/genetics , Enzyme Activation , Flow Cytometry , Genes, Reporter , Macaca mulatta , Models, Biological , Mutation/genetics , Protein Processing, Post-Translational , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53
17.
Exp Neurol ; 193(2): 455-68, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15869948

ABSTRACT

The retinoblastoma susceptibility gene product (pRb) and E2F1 have been found to exhibit altered localization and increased staining in several neurodegenerative diseases. We have observed similar localization in primary murine cortical cultures treated with neurotrophic factors (NTF) or chemokines. In untreated cultures, E2F1 exhibited minimal immunostaining using the KH95 antibody, which recognizes the pRb interaction domain. In primary E16 murine cortical cultures, NTF- or chemokine-treated neurons, KH95 E2F1 staining was increased in the cytoplasm. However, an antibody recognizing the amino-terminus of E2F1 (KH20) stained the cytoplasm of both untreated and treated neurons. Taken together these results suggest that the change seen in E2F1 using the KH95 antibody is due to antigen unmasking of a carboxy-terminal epitope in response to NTF and chemokines. When we assessed staining for the hyperphosphorylated, inactive form of pRb (ppRb) in untreated cultures, ppRb was predominantly cytoplasmic. In response to NTF or chemokine treatment, staining for ppRb was observed predominantly in nuclei of neurons indicating a change in subcellular distribution. Immunoblot analysis demonstrated increased levels of ppRb in response to NTF and chemokines. Inhibitors of translation, nuclear export, and phoshpatidylinositol-3-kinase blocked NTF- and chemokine-induced nuclear ppRb localization while having no effect on E2F1 staining. Instead increased cytoplasmic KH95 E2F1 staining was dependent on cytoskeletal destabilization which did not influence ppRb localization. These findings demonstrate that alterations in ppRb distribution and E2F1 antigen availability by NTF and chemokines occur by distinct mechanisms suggesting that E2F1 function may be independent of pRb regulation in post-mitotic neurons.


Subject(s)
Cell Cycle Proteins/metabolism , Cerebral Cortex/cytology , Chemokines/pharmacology , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Nerve Growth Factors/pharmacology , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western/methods , Brain-Derived Neurotrophic Factor/pharmacology , Cell Cycle Proteins/genetics , Cells, Cultured , Chemokine CCL5/pharmacology , Cycloheximide/pharmacology , DNA/metabolism , DNA-Binding Proteins/genetics , Drug Interactions , E2F Transcription Factors , E2F1 Transcription Factor , Embryo, Mammalian , Fatty Acids, Unsaturated/pharmacology , Female , Fluorescent Antibody Technique/methods , Gene Expression Regulation/physiology , Hydrogen Peroxide/pharmacology , Indoles/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Confocal/methods , Microtubule-Associated Proteins/metabolism , Paclitaxel/pharmacology , Phosphorylation/drug effects , Pregnancy , Protein Synthesis Inhibitors/pharmacology , Retinoblastoma Protein/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...