Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Curr Alzheimer Res ; 20(7): 515-522, 2023.
Article in English | MEDLINE | ID: mdl-37702232

ABSTRACT

INTRODUCTION: The present study has examined microglial and astrocyte activation in association with neuronal degeneration in an animal model using an injection of amyloid-beta peptide Aß1-42 (Aß42) plus fibrinogen into rat hippocampus. METHODS: The combination of stimuli is suggested as a novel and potent perturbation to induce gliosis and the production of glial-derived neurotoxic factors in an animal model exhibiting a leaky BBB (blood-brain barrier). Specifically, Aß42 + fibrinogen stimulation elevated levels of COX-2 (cyclooxygenase-2) and iNOS (inducible nitric oxide synthase) with a considerable extent of neuronal loss associated with microglia and astrocyte activation. RESULTS: Treatment of injected rats with the broad spectrum anti-inflammatory agent, minocycline or the iNOS inhibitor, 1400 W inhibited gliosis, reduced levels of COX-2 and iNOS, and demonstrated efficacy for neuroprotection. CONCLUSION: The findings suggest the utility of combining amyloid beta peptide plus fibrinogen as a potent and understudied neuroinflammatory stimulus for the induction of glial-derived neurotoxic factors in BBB-compromised AD brain.


Subject(s)
Amyloid beta-Peptides , Gliosis , Rats , Animals , Amyloid beta-Peptides/metabolism , Cyclooxygenase 2/metabolism , Gliosis/drug therapy , Neuroinflammatory Diseases , Microglia/metabolism , Nitric Oxide Synthase Type II/metabolism , Fibrinogen , Hippocampus/metabolism , Peptide Fragments/metabolism
2.
Curr Alzheimer Res ; 18(12): 925-938, 2021.
Article in English | MEDLINE | ID: mdl-34789126

ABSTRACT

Astrocytes contribute to brain development and homeostasis and support diverse functions of neurons. These cells also respond to the pathological processes in Alzheimer's disease (AD). There is still considerable debate concerning the overall contribution of astrocytes to AD pathogenesis since both the protective and harmful effects of these cells on neuronal survival have been documented. This review focuses exclusively on the neurotoxic potential of astrocytes while acknowledging that these cells can contribute to neurodegeneration through other mechanisms, for example, by lowered neurotrophic support. We identify reactive oxygen and nitrogen species, tumor necrosis factor α (TNF-α), glutamate, and matrix metalloproteinase (MMP)-9 as molecules that can be directly toxic to neurons and are released by reactive astrocytes. There is also considerable evidence suggesting their involvement in AD pathogenesis. We further discuss the signaling molecules that trigger the neurotoxic response of astrocytes with a focus on human cells. We also highlight microglia, the immune cells of the brain, as critical regulators of astrocyte neurotoxicity. Nuclear imaging and magnetic resonance spectroscopy (MRS) could be used to confirm the contribution of astrocyte neurotoxicity to AD progression. The molecular mechanisms discussed in this review could be targeted in the development of novel therapies for AD.


Subject(s)
Alzheimer Disease , Astrocytes , Alzheimer Disease/pathology , Humans , Microglia/pathology , Neurons , Tumor Necrosis Factor-alpha
3.
Antioxidants (Basel) ; 11(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35052606

ABSTRACT

The intactness of blood-brain barrier (BBB) is compromised in Alzheimer's disease (AD). Importantly, evidence suggests that the perturbation and abnormalities appearing in BBB can manifest early in the progression of the disease. The disruption of BBB allows extravasation of the plasma protein, fibrinogen, to enter brain parenchyma, eliciting immune reactivity and response. The presence of amyloid-ß (Aß) peptide leads to the formation of abnormal aggregates of fibrin resistant to degradation. Furthermore, Aß deposits act on the contact system of blood coagulation, altering levels of thrombin, fibrin clots and neuroinflammation. The neurovascular unit (NVU) comprises an ensemble of brain cells which interact with infiltrating fibrinogen. In particular, interaction of resident immune cell microglia with fibrinogen, fibrin and Aß results in the production of reactive oxygen species (ROS), a neurotoxic effector in AD brain. Overall, fibrinogen infiltration through a leaky BBB in AD animal models and in human AD tissue is associated with manifold abnormalities including persistent fibrin aggregation and clots, microglial-mediated production of ROS and diminished viability of neurons and synaptic connectivity. An objective of this review is to better understand how processes associated with BBB leakiness to fibrinogen link vascular pathology with neuronal and synaptic damage in AD.

4.
Curr Alzheimer Res ; 17(12): 1057-1064, 2020.
Article in English | MEDLINE | ID: mdl-33463465

ABSTRACT

The dysregulation of calcium signaling mechanisms in neurons has been considered a contributing factor to the pathogenesis evident in early-onset Alzheimer's Disease (AD). However, considerably less is known concerning the possible impairment of Ca2+ mobilization in resident immune cell microglia. This review considers findings which suggest that a prominent pathway for non-excitable microglial cells, store-operated calcium entry (SOCE), is altered in the sporadic form of AD. The patterns of Ca2+ mobilization are first discussed with platelet-activating factor (PAF) stimulation of SOCE in adult, fetal and immortalized cell-line, human microglia in the healthy brain. In all cases, PAF was found to induce a rapid transient depletion of Ca2+ from endoplasmic reticulum (ER) stores, followed by a sustained entry of Ca2+ (SOCE). A considerably attenuated duration of SOCE is observed with ATP stimulation of human microglia, suggested as due to agonist actions on differential subtype purinergic receptors. Microglia obtained from AD brain tissue, or microglia treated with full-length amyloid-ß peptide (Aß42), show significant reductions in the amplitude of SOCE relative to controls. In addition, AD brain and Aß42-treated microglia exhibit decreased levels of Ca2+ release from ER stores compared to controls. Changes in properties of SOCE in microglia could lead to altered immune cell response and neurovascular unit dysfunction in the inflamed AD brain.


Subject(s)
Alzheimer Disease/physiopathology , Calcium Signaling/physiology , Calcium/metabolism , Microglia/metabolism , Amyloid beta-Peptides/metabolism , Endoplasmic Reticulum/metabolism , Humans , Neurons/pathology
5.
Curr Alzheimer Res ; 16(11): 1007-1017, 2019.
Article in English | MEDLINE | ID: mdl-31692444

ABSTRACT

A combinatorial cocktail approach is suggested as a rationale intervention to attenuate chronic inflammation and confer neuroprotection in Alzheimer's disease (AD). The requirement for an assemblage of pharmacological compounds follows from the host of pro-inflammatory pathways and mechanisms present in activated microglia in the disease process. This article suggests a starting point using four compounds which present some differential in anti-inflammatory targets and actions but a commonality in showing a finite permeability through Blood-brain Barrier (BBB). A basis for firstchoice compounds demonstrated neuroprotection in animal models (thalidomide and minocycline), clinical trial data showing some slowing in the progression of pathology in AD brain (ibuprofen) and indirect evidence for putative efficacy in blocking oxidative damage and chemotactic response mediated by activated microglia (dapsone). It is emphasized that a number of candidate compounds, other than ones suggested here, could be considered as components of the cocktail approach and would be expected to be examined in subsequent work. In this case, systematic testing in AD animal models is required to rigorously examine the efficacy of first-choice compounds and replace ones showing weaker effects. This protocol represents a practical approach to optimize the reduction of microglial-mediated chronic inflammation in AD pathology. Subsequent work would incorporate the anti-inflammatory cocktail delivery as an adjunctive treatment with ones independent of inflammation as an overall preventive strategy to slow the progression of AD.


Subject(s)
Alzheimer Disease , Drug Therapy, Combination/methods , Inflammation , Neuroprotective Agents/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Animals , Dapsone/pharmacology , Humans , Ibuprofen/pharmacology , Inflammation/complications , Inflammation/drug therapy , Minocycline/pharmacology , Thalidomide/pharmacology
6.
Cancer Lett ; 402: 93-99, 2017 08 28.
Article in English | MEDLINE | ID: mdl-28536012

ABSTRACT

This review considers evidence suggesting that activation of the ionotropic purinergic receptor P2X7 (P2X7R) is a contributing factor in the growth of brain tumors. Importantly, expression of P2X7R may be upregulated in both glioma cells and in immune responding microglial cells with possible differential effects on tumor progression. The recruitment of immune cells into tumor regions may not only be involved in supporting an immunosuppressive environment aiding tumor growth but activated microglia could secrete inflammatory factors promoting neoangiogenesis in expanding tumors. The subtype P2X7R exhibits a number of unique properties including activation of the receptor in pathological conditions associated with developing brain tumors. In particular, the tumor microenvironment includes elevated levels of ATP required for activation of P2X7R and the sustained tumor and immune cell P2X7R-mediated responses which in total contribute to overall tumor growth and viability. Studies on cultured rat and human glioma show marked increases in expression of P2X7R and enhanced cell mobility relative to control. Glioma cell animal models demonstrate enhanced expression of P2X7R in both glioma and microglia with antagonism of receptor showing differential effects on tumor growth. Overall, P2X7R activation is associated with a complexity of modulatory actions on tumor growth in part due to ubiquitous expression of the receptor in glioma and immune responsive cells.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Microglia/metabolism , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Proliferation , Glioma/drug therapy , Glioma/pathology , Humans , Microglia/drug effects , Microglia/pathology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/drug effects , Signal Transduction , Tumor Burden , Tumor Microenvironment
7.
Am J Neurodegener Dis ; 5(1): 69-73, 2016.
Article in English | MEDLINE | ID: mdl-27073744

ABSTRACT

The effects of pyruvate, the end metabolite of glycolysis, on blood-brain barrier (BBB) impairment and immune reactivity were examined in the quinolinic acid (QA)-injected rat striatum. Extensive disruption of BBB was observed at 7 d post QA-injection as demonstrated by increased immunohistochemical staining using antibody against immunoglobulin G (IgG). Animals receiving pyruvate administration (500 mg/kg) with QA-injection exhibited reduced lgG immunoreactivity (by 45%) relative to QA alone. QA intrastriatal injection also resulted in marked increases in the number of infiltrating T-lymphocytes (by 70-fold) and expression of major histocompatibility complex (MHC-class II) (by 45-fold) relative to unlesioned control. Treatment with pyruvate significantly reduced infiltration of T-cells (by 68%) and MHC class II expression (by 48%) induced by QA. These results indicate that QA injection into rat striatum leads to impairment in BBB function with pyruvate administration reducing immune response and BBB leakiness in excitotoxic injury.

8.
J Neuroinflammation ; 12: 144, 2015 Aug 09.
Article in English | MEDLINE | ID: mdl-26255110

ABSTRACT

BACKGROUND: The chemokine interleukin-8 (IL-8) and its receptor CXCR2 contribute to chemotactic responses in Alzheimer's disease (AD); however, properties of the ligand and receptor have not been characterized in animal models of disease. The primary aim of our study was to examine effects of pharmacological antagonism of CXCR2 as a strategy to inhibit receptor-mediated inflammatory reactivity and enhance neuronal viability in animals receiving intrahippocampal injection of amyloid-beta (Aß1-42). METHODS: In vivo studies used an animal model of Alzheimer's disease incorporating injection of full-length Aß1-42 into rat hippocampus. Immunohistochemical staining of rat brain was used to measure microgliosis, astrogliosis, neuronal viability, and oxidative stress. Western blot and Reverse Transcription PCR (RT-PCR) were used to determine levels of CXCR2 in animal tissue with the latter also used to determine expression of pro-inflammatory mediators. Immunostaining of human AD and non-demented (ND) tissue was also undertaken. RESULTS: We initially determined that in the human brain, AD relative to ND tissue exhibited marked increases in expression of CXCR2 with cell-specific receptor expression prominent in microglia. In Aß1-42-injected rat brain, CXCR2 and IL-8 showed time-dependent increases in expression, concomitant with enhanced gliosis, relative to controls phosphate-buffered saline (PBS) or reverse peptide Aß42-1 injection. Administration of the competitive CXCR2 antagonist SB332235 to peptide-injected rats significantly reduced expression of CXCR2 and microgliosis, with astrogliosis unchanged. Double staining studies demonstrated localization of CXCR2 and microglial immunoreactivity nearby deposits of Aß1-42 with SB332235 effective in inhibiting receptor expression and microgliosis. The numbers of neurons in granule cell layer (GCL) were reduced in rats receiving Aß1-42, compared with PBS, with administration of SB332235 to peptide-injected animals conferring neuroprotection. Oxidative stress was indicated in the animal model since both 4-hydroxynonenal (4-HNE) and hydroethidine (HEt) were markedly elevated in Aß1-42 vs. PBS-injected rat brain and diminished with SB332235 treatment. CONCLUSION: Overall, the findings suggest critical roles for CXCR2-dependent inflammatory responses in an AD animal model with pharmacological modulation of the receptor effective in inhibiting inflammatory reactivity and conferring neuroprotection against oxidative damage.


Subject(s)
Alzheimer Disease/drug therapy , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Neuroprotective Agents/therapeutic use , Receptors, Interleukin-8B/antagonists & inhibitors , Aged , Aged, 80 and over , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amyloid beta-Peptides , Animals , Brain/pathology , Female , Gliosis/pathology , Hippocampus , Humans , Inflammation Mediators , Male , Microinjections , Middle Aged , Oxidative Stress/drug effects , Peptide Fragments , Rats
9.
Biomed Res Int ; 2014: 923670, 2014.
Article in English | MEDLINE | ID: mdl-24822221

ABSTRACT

Animal models of Alzheimer's disease (AD) which emphasize activation of microglia may have particular utility in correlating proinflammatory activity with neurodegeneration. This paper reviews injection of amyloid- ß (A ß ) into rat brain as an alternative AD animal model to the use of transgenic animals. In particular, intrahippocampal injection of Aß 1-42 peptide demonstrates prominent microglial mobilization and activation accompanied by a significant loss of granule cell neurons. Furthermore, pharmacological inhibition of inflammatory reactivity is demonstrated by a broad spectrum of drugs with a common endpoint in conferring neuroprotection in peptide-injected animals. Peptide-injection models provide a focus on glial cell responses to direct peptide injection in rat brain and offer advantages in the study of the mechanisms underlying neuroinflammation in AD brain.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/administration & dosage , Disease Models, Animal , Alzheimer Disease/chemically induced , Alzheimer Disease/immunology , Alzheimer Disease/physiopathology , Animals , Brain/immunology , Brain/physiopathology , Brain Chemistry , Rats
10.
BMC Neurosci ; 15: 18, 2014 Jan 22.
Article in English | MEDLINE | ID: mdl-24447580

ABSTRACT

BACKGROUND: The properties of Ca2+ signaling mediated by purinergic receptors are intrinsically linked with functional activity of astrocytes. At present little is known concerning Ca2+-dependent purinergic responses in adult human astrocytes. This work has examined effects of purinergic stimulation to alter levels of intracellular Ca2+ in adult human astrocytes. Ca2+-sensitive spectrofluorometry was carried out to determine mobilization of intracellular Ca2+ following adenosine triphosphate (ATP) or 3'-O-(4-benzoyl)benzoyl-ATP (Bz-ATP) stimulation of adult human astrocytes. In some experiments pharmacological modulation of Ca2+ pathways was applied to help elucidate mechanisms of Ca2+ signaling. RT-PCR was also performed to confirm human astrocyte expression of specific purinoceptors which were indicated from imaging studies. RESULTS: The endogenous P2 receptor agonist ATP (at 100 µM or 1 mM) applied in physiological saline solution (PSS) evoked a rapid increase of [Ca2+]i to a peak amplitude with the decay phase of response exhibiting two components. The two phases of decay consisted of an initial rapid component which was followed by a secondary slower component. In the presence of Ca2+-free solution, the secondary phase of decay was absent indicating this prolonged component was due to influx of Ca2+. This prolonged phase of decay was also attenuated with the store-operated channel (SOC) inhibitor gadolinium (at 2 µM) added to standard PSS, suggesting this component was mediated by SOC activation. These results are consistent with ATP activation of P2Y receptor (P2YR) in adult human astrocytes leading to respective rapid [Ca2+]i mobilization from intracellular stores followed by Ca2+ entry through SOC. An agonist for P2X7 receptor (P2X7R), BzATP induced a very different response compared with ATP whereby BzATP (at 300 µM) elicited a slowly rising increase in [Ca2+]i to a plateau level which was sustained in duration. The BzATP-induced increase in [Ca2+]i was not enhanced with lipopolysaccharide pre-treatment of cells as previously found for P2X7R mediated response in human microglia. RT-PCR analysis showed that adult human astrocytes in vitro constitutively express mRNA for P2Y1R, P2Y2R and P2X7R. CONCLUSION: These results suggest that activation of metabotropic P2YR (P2Y1R and/or P2Y2R) and ionotropic P2X7R could mediate purinergic responses in adult human astrocytes.


Subject(s)
Astrocytes/physiology , Calcium Channels/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Ion Channel Gating/physiology , Receptors, Purinergic/metabolism , Adult , Cells, Cultured , Humans
11.
PLoS One ; 8(10): e76860, 2013.
Article in English | MEDLINE | ID: mdl-24146937

ABSTRACT

Neural stem cell (NSC) replacement therapy is considered a promising cell replacement therapy for various neurodegenerative diseases. However, the low rate of NSC survival and neurogenesis currently limits its clinical potential. Here, we examined if hippocampal long-term potentiation (LTP), one of the most well characterized forms of synaptic plasticity, promotes neurogenesis by facilitating proliferation/survival and neuronal differentiation of NSCs. We found that the induction of hippocampal LTP significantly facilitates proliferation/survival and neuronal differentiation of both endogenous neural progenitor cells (NPCs) and exogenously transplanted NSCs in the hippocampus in rats. These effects were eliminated by preventing LTP induction by pharmacological blockade of the N-methyl-D-aspartate glutamate receptor (NMDAR) via systemic application of the receptor antagonist, 3-[(R)-2-carboxypiperazin-4-yl]-propyl-1-phosphonic acid (CPP). Moreover, using a NPC-neuron co-culture system, we were able to demonstrate that the LTP-promoted NPC neurogenesis is at least in part mediated by a LTP-increased neuronal release of brain-derived neurotrophic factor (BDNF) and its consequent activation of tropomysosin receptor kinase B (TrkB) receptors on NSCs. Our results indicate that LTP promotes the neurogenesis of both endogenous and exogenously transplanted NSCs in the brain. The study suggests that pre-conditioning of the host brain receiving area with a LTP-inducing deep brain stimulation protocol prior to NSC transplantation may increase the likelihood of success of using NSC transplantation as an effective cell therapy for various neurodegenerative diseases.


Subject(s)
Cell Differentiation , Long-Term Potentiation/physiology , Neural Stem Cells/cytology , Neurons/cytology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Separation , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Dentate Gyrus/cytology , HEK293 Cells , Humans , Long-Term Potentiation/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptor, trkB/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Stem Cell Transplantation
12.
J Alzheimers Dis ; 37(2): 367-77, 2013.
Article in English | MEDLINE | ID: mdl-23948889

ABSTRACT

Immunohistochemical staining has been used to determine expression patterns of the angiogenic transcription factor, Ets-1, in the brains of Alzheimer's disease (AD) individuals. Brain tissue from non-demented controls showed little expression of Ets-1 whereas in AD brain tissue, Ets-1 was ubiquitously expressed in cortex and hippocampus. Double immunostaining with von Willerbrand factor demonstrated prominent Ets-1 intravascular immunoreactivity (ir) in AD cortical microvessels. In addition, Ets-1 also exhibited extravascular expression characterized by a diffuse pattern of Ets-1 ir in AD brain. Double staining also showed Ets-1 colocalization in microvasculature with the potent angiogenic agent, vascular endothelial growth factor (VEGF). Cell-associated tumor necrosis factor-α (TNF-α), a pro-inflammatory cytokine with pro-angiogenic activity, was primarily associated with diffuse extravascular Ets-1 ir. Clusters of HLA-DR positive microglia, resident immune cells of brain which release TNF-α, were also localized with diffuse Ets-1. Intravascular Ets-1 ir was maximally co-localized with soluble amyloid-ß peptide (Aß), Aß1-40, in microvasculature whereas diffuse extravascular Ets-1 ir appeared in proximity to Aß plaques in brain parenchyma. Similar overall results were obtained for patterns of Ets-1 staining in AD hippocampal tissue. This work provides novel findings on expression of the angiogenic transcription factor, Ets-1, in vascular remodeling and its association with pro-angiogenic factors, reactive microglia, and Aß deposition in AD brain.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Proto-Oncogene Protein c-ets-1/cerebrospinal fluid , Up-Regulation/physiology , Age Factors , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Analysis of Variance , Creutzfeldt-Jakob Syndrome/cerebrospinal fluid , Depression/cerebrospinal fluid , Enzyme-Linked Immunosorbent Assay , Female , Fluorometry , Frontotemporal Dementia/cerebrospinal fluid , Humans , Linear Models , Male , Middle Aged , Neprilysin/blood , Neprilysin/metabolism , tau Proteins/cerebrospinal fluid
13.
Curr Alzheimer Res ; 10(3): 252-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23534534

ABSTRACT

We have examined the anti-angiogenic compound, angiostatin as a modulator of inflammatory reactivity and vascular responses and for neuroprotection in an animal model of Alzheimer's disease (AD). Intra-hippocampal amyloidbeta (Aß1₋42) injection, relative to controls phosphate buffer saline (PBS) or reverse peptide Aß42₋1, increased gliosis in the molecular layer (ML) of rat hippocampus. Vascular remodeling was indicated from increased microvessel immunoreactivity (ir) in ML suggesting the possibility of an angiogenic response to peptide injection. Administration of Aß1₋42 also induced a loss of neurons in the granule cell region of hippocampus relative to controls. Treatment of peptide-injected rats with angiostatin was associated with a spectrum of modulatory effects including reduced microgliosis (by 34%), diminished microvessel ir (by 36%) and increased neuronal viability (by 31%) compared with peptide injection alone. Angiostatin treatment was ineffective in reducing astrogliosis induced by Aß1₋42 and applied alone the compound had no significant effect to alter gliosis, microvessel ir or neuronal viability compared with PBS control. In vitro, angiostatin significantly attenuated secretion of the pro-angiogenic agent, vascular endothelial growth factor (VEGF) in lipopolysaccharide (LPS)-stimulated THP-1 cells. Our findings provide novel evidence for a broad spectrum of angiostatin effects in an animal model of AD including actions to reduce inflammatory reactivity, stabilize vascular remodeling and confer neuroprotection. The overall effects of angiostatin are consistent with actions of the compound to inhibit microglial secretion of VEGF.


Subject(s)
Alzheimer Disease/pathology , Angiogenesis Inhibitors/pharmacology , Angiostatins/pharmacology , Brain/drug effects , Animals , Brain/metabolism , Brain/pathology , Cell Line , Disease Models, Animal , Humans , Immunohistochemistry , Male , Monocytes/drug effects , Monocytes/metabolism , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
14.
Neuron ; 75(6): 1094-104, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22998876

ABSTRACT

Astrocytes are proposed to participate in brain energy metabolism by supplying substrates to neurons from their glycogen stores and from glycolysis. However, the molecules involved in metabolic sensing and the molecular pathways responsible for metabolic coupling between different cell types in the brain are not fully understood. Here we show that a recently cloned bicarbonate (HCO3⁻) sensor, soluble adenylyl cyclase (sAC), is highly expressed in astrocytes and becomes activated in response to HCO3⁻ entry via the electrogenic NaHCO3 cotransporter (NBC). Activated sAC increases intracellular cAMP levels, causing glycogen breakdown, enhanced glycolysis, and the release of lactate into the extracellular space, which is subsequently taken up by neurons for use as an energy substrate. This process is recruited over a broad physiological range of [K⁺](ext) and also during aglycemic episodes, helping to maintain synaptic function. These data reveal a molecular pathway in astrocytes that is responsible for brain metabolic coupling to neurons.


Subject(s)
Adenylyl Cyclases/metabolism , Astrocytes/drug effects , Astrocytes/enzymology , Bicarbonates/pharmacology , Hippocampus/cytology , Neurons/drug effects , Neurons/enzymology , 1-Methyl-3-isobutylxanthine/pharmacology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Animals, Newborn , Astrocytes/ultrastructure , Coumaric Acids/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , Glial Fibrillary Acidic Protein/metabolism , Glucose/deficiency , Glycogen/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Lactic Acid/metabolism , Microscopy, Immunoelectron , Microtubule-Associated Proteins/metabolism , Models, Biological , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Neurons/ultrastructure , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
15.
Brain Res ; 1442: 1-8, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22297175

ABSTRACT

We have recently reported effects of Mg2+ to confer neuroprotection against toxicity of purinergic stimulated microglia and THP-1 monocytes. To examine mechanisms underlying neuroprotection, we have studied Mg2+ modulation of transient changes in intracellular Ca2+ ([Ca2+]i) in THP-1 cells induced by P2X7R agonist 2',3'-[benzoyl-4-benzoyl]-ATP (BzATP). Application of BzATP caused a rapid transient increase in [Ca2+]i followed by a prolonged component. The time course of the secondary slower phase was significantly reduced with Ca2+-free extracellular solution, with treatment of THP-1 cells by the P2X7R antagonist, oxATP or with exposure of cells to the store-operated channel (SOC) inhibitor, SKF96365. These results suggest that Ca2+ influx, mediated by both the P2X7R or by SOC, contribute to the slow component of [Ca2+]i. Treatment of THP-1 cells with 10 mMMg2+ was highly effective in reducing the time course of BzATP-induced Ca2+ decay; unlike the other modulatory protocols, Mg2+ markedly inhibited the amplitudes of slow and rapid components. In addition, acute application of Mg2+ during BzATP-induced responses elicited in the presence of either oxATP or SKF96365 to block respective P2X7R and SOC contributions, rapidly attenuated [Ca2+]i to baseline levels. Priming of cells with the inflammatory stimulus LPS/IFN-γ markedly enhanced the slower, but not rapid, phase of BzATP-induced [Ca2+]i with application of 10 mMMg2+ inhibiting both components of response. A model is proposed to account for BzATP stimulation of both ionotropic P2XR and metabotropic P2YR which provides a mechanistic basis for elevated Mg2+ anti-inflammatory and neuroprotective actions in inflamed brain.


Subject(s)
Calcium/metabolism , Magnesium/pharmacology , Monocytes/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Cell Line, Tumor , Humans , Imidazoles/pharmacology , Interferon-gamma/immunology , Lipopolysaccharides/immunology , Models, Biological , Monocytes/immunology , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2Y/metabolism
16.
Neurobiol Dis ; 45(1): 438-49, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21946335

ABSTRACT

Central nervous system (CNS) inflammatory processes including microglial activation have been implicated in the pathogenesis of neurodegenerative diseases such as Huntington Disease (HD). We report age-dependent changes in striatal microglial morphology and vasculature in the YAC128 mouse model of HD. Decreases in microglial ramification along with a decrease in vessel diameter and increased vessel density and length suggest the presence of microgliosis and proangiogenic activity in YAC128 mice. Our hypothesis for this study was that the changes in microglial morphology and perturbations in vasculature may be involved in the pathogenesis of HD and that peripheral challenge with the bacterial endotoxin, lipopolysaccharide (LPS), will exacerbate these microglial and vascular changes as well as the HD phenotype in YAC128 mice at 12 months. Chronic peripheral LPS (1mg/kg) potentiated microglial activation indicated by an increase in microglial cell body size and retraction of processes. This potentiation in microglial activation with chronic peripheral LPS challenge was paralleled with vascular remodeling including dilatation, increased vessel wall thickness, increased BBB permeability and fibrinogen deposition in YAC128 striatum. Although peripheral LPS caused an increase in microglial activation and degenerative changes in cerebrovasculature, the phenotypic hallmarks of HD in YAC128 mice such as motor coordination deficits and decreased striatal volume were not exacerbated by chronic peripheral LPS exposure. This study identifies age-dependent increases in microglial activation and angiogenesis in YAC128 at 12 months. Peripheral inflammation induced by chronic LPS causes similar changes but does not influence the HD phenotype in YAC128 mice.


Subject(s)
Brain/blood supply , Brain/pathology , Huntington Disease/pathology , Microglia/pathology , Age Factors , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Cell Shape , Corpus Striatum/blood supply , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Female , Huntington Disease/genetics , Huntington Disease/metabolism , Male , Mice , Mice, Transgenic , Microglia/metabolism , Phenotype
17.
Am J Neurodegener Dis ; 1(3): 199-204, 2012.
Article in English | MEDLINE | ID: mdl-23383392

ABSTRACT

The net migration of microglia induced by deposits of amyloid beta (Aß) constitutes a chemotactic response of resident neuroimmune brain cells. This process serves to localize clusters of microglia nearby Aß deposits preparatory to cellular activation and functional responses. Microglial responses to Aß deposits localized in brain parenchyma and in blood vessels lead to acute and chronic neuroinflammation in Alzheimer's disease (AD) brain. This review summarizes studies on the prominent chemotactic factors MCP-1, MIP-1α and IL-8 and also includes recent work indicating VEGF and fractalkine as chemotactic agents. The possibility that microglial release of MCP-1 may play a role in mediating chemotactic responses of neural progenitor cells is also considered. The plethora of chemotactic factors and their cognate receptors suggests the utility in testing pharmacological modulation of chemotaxis for effects to inhibit chronic neuroinflammation and confer neuroprotection in AD animal models.

18.
Int J Alzheimers Dis ; 2011: 918280, 2011.
Article in English | MEDLINE | ID: mdl-21969915

ABSTRACT

The validity of amyloid-ß peptide (Aß(1-42)) intrahippocampal injection, as an animal model of Alzheimer's disease (AD), has previously been considered in terms of inflammatory reactivity and neuronal damage. In this work, we have extended the testing of the animal model to vasculature by comparison of selected properties of microvessels in vivo with those in human AD brain tissue. The injection of Aß(1-42), relative to control PBS (phosphate buffered saline), increased the mean number of microvessels and diminished the mean length of microvessels in the molecular layer of dentate gyrus. The animal model showed Aß(1-42), but not PBS, injection was associated with abnormalities in morphology of microvessels which were characterized as looping, fragmented, knob-like, uneven, and constricted. In particular, numbers of constricted microvessels, defined as vessels with diameters less than 3 µm, were considerably enhanced for Aß(1-42), compared to PBS, injection. In comparison, human AD brain demonstrated an elevated number of microvessels with a diminished mean length relative to nondemented (ND) brain. Additionally, microvessel perturbations in AD brain showed a similar pattern of morphological abnormalities to those observed in Aß(1-42)-injected rat hippocampus. Constricted microvessels were a prominent feature of AD brain but were rarely observed in ND tissue. These results provide the first evidence that a peptide-injection animal model exhibits a commonality in perturbations of microvessels compared with those evident in AD brain.

19.
J Alzheimers Dis ; 27(3): 531-41, 2011.
Article in English | MEDLINE | ID: mdl-21841251

ABSTRACT

Pyrazole derivatives are well documented to possess anti-inflammatory activity but their effects on microglial activation are unknown. We determined the efficacy of the novel pyrazole compound 2-MBAPA (R/S-(±)-2-Methylbenzylamino 2-oxo-N-[4-cyano-1-phenyl-1H-pyrazol-5-yl] acetamide) on activated microglia under conditions relevant to inflammation in Alzheimer's disease (AD) brain. The compound at a non-toxic concentration inhibited secretion of tumor necrosis factor (TNF)-α by activated human microglia and attenuated toxicity of conditioned medium from activated human microglia towards human SH-SY5Y neuroblastoma cells in vitro. The 2-MBAPA neuroprotection was further demonstrated in vivo using an animal model of AD. The compound inhibited microgliosis, but not astrogliosis, in amyloid-ß peptide (Aß)(1-42)-injected rat brain. 2-MBAPA also diminished neuronal loss in the dentate gyrus caused by Aß(1-42) injection. These results indicate that this novel pyrazole compound confers neuroprotection by inhibiting microglial activation. Therefore, further studies with 2-MBAPA and novel analogues based on this lead compound are warranted in an effort to develop new pharmacological agents that may be useful for slowing down progression of AD and other neuroinflammatory disorders associated with activated microglia.


Subject(s)
Microglia/drug effects , Microglia/metabolism , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/pathology , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Humans , Male , Microglia/pathology , Neurotoxicity Syndromes/metabolism , Rats , Rats, Sprague-Dawley
20.
Neurosci Lett ; 497(2): 80-4, 2011 Jun 22.
Article in English | MEDLINE | ID: mdl-21540076

ABSTRACT

We have examined activation of purinergic P2Y1 receptor-dependent Ca²âº-signaling pathways in mediating C6 glioma cell migration. The administration of 2-methylthioadenosine 5'-diphosphate (2MeSADP), a selective agonist for P2Y1R, induced marked increases in patterns of glioma migration in both scratch wound and Boyden chamber assays. Antagonism of P2Y1R with either the broad spectrum purinergic blocker, pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate (PPADS) or the specific P2Y1R antagonist, 2'-deoxy-N6-methyladenosine-3',5'-bisphosphate (MRS2179), significantly inhibited C6 cell migration. Calcium-sensitive spectrofluorometry showed 2MeSADP stimulation of glioma cells caused a biphasic change in intracellular Ca²âº ([Ca²âº]i). The rapid transient phase was unchanged in Ca²âº-free solution reflecting a [Ca²âº]i component due to intracellular stores release subsequent to activation of a metabotropic P2Y subtype receptor. The secondary prolonged phase of [Ca²âº]i was abolished in Ca²âº-free solution or in glioma cells treated with the store-operated channel (SOC) blocker, SKF96365. Treatment of glioma with either MRS2179 or PPADS significantly attenuated both the rapid and prolonged phases of [Ca²âº]i. These results suggest critical roles for activation of P2Y1R in mediating glioma cell mobility and migration with changes in [Ca²âº]i contributing as a mechanistic link between activated receptor and functional response. Our findings suggest that pharmacological modulation of metabotropic P2Y1R-dependent signaling pathways may serve as a novel therapeutic procedure to slow glioma progression.


Subject(s)
Brain Neoplasms/metabolism , Calcium Signaling/physiology , Cell Movement/physiology , Glioma/metabolism , Receptors, Purinergic P2Y1/physiology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Brain Neoplasms/pathology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Glioma/pathology , Intracellular Fluid/metabolism , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Neoplasm Invasiveness/pathology , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Rats , Receptors, Purinergic P2Y1/drug effects , Thionucleotides/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...