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










Publication year range
1.
Front Immunol ; 14: 1130735, 2023.
Article in English | MEDLINE | ID: mdl-37033925

ABSTRACT

Diabetic retinopathy, a microvascular disease characterized by irreparable vascular damage, neurodegeneration and neuroinflammation, is a leading complication of diabetes mellitus. There is no cure for DR, and medical interventions marginally slow the progression of disease. Microglia-mediated inflammation in the diabetic retina is regulated via CX3CR1-FKN signaling, where FKN serves as a calming signal for microglial activation in several neuroinflammatory models. Polymorphic variants of CX3CR1, hCX3CR1I249/M280 , found in 25% of the human population, result in a receptor with lower binding affinity for FKN. Furthermore, disrupted CX3CR1-FKN signaling in CX3CR1-KO and FKN-KO mice leads to exacerbated microglial activation, robust neuronal cell loss and substantial vascular damage in the diabetic retina. Thus, studies to characterize the effects of hCX3CR1I249/M280 -expression in microglia-mediated inflammation in the diseased retina are relevant to identify mechanisms by which microglia contribute to disease progression. Our results show that hCX3CR1I249/M280 mice are significantly more susceptible to microgliosis and production of Cxcl10 and TNFα under acute inflammatory conditions. Inflammation is exacerbated under diabetic conditions and coincides with robust neuronal loss in comparison to CX3CR1-WT mice. Therefore, to further investigate the role of hCX3CR1I249/M280 -expression in microglial responses, we pharmacologically depleted microglia using PLX-5622, a CSF-1R antagonist. PLX-5622 treatment led to a robust (~70%) reduction in Iba1+ microglia in all non-diabetic and diabetic mice. CSF-1R antagonism in diabetic CX3CR1-WT prevented TUJ1+ axonal loss, angiogenesis and fibrinogen deposition. In contrast, PLX-5622 microglia depletion in CX3CR1-KO and hCX3CR1I249/M280 mice did not alleviate TUJ1+ axonal loss or angiogenesis. Interestingly, PLX-5622 treatment reduced fibrinogen deposition in CX3CR1-KO mice but not in hCX3CR1I249/M280 mice, suggesting that hCX3CR1I249/M280 expressing microglia influences vascular pathology differently compared to CX3CR1-KO microglia. Currently CX3CR1-KO mice are the most commonly used strain to investigate CX3CR1-FKN signaling effects on microglia-mediated inflammation and the results in this study indicate that hCX3CR1I249/M280 receptor variants may serve as a complementary model to study dysregulated CX3CR1-FKN signaling. In summary, the protective effects of microglia depletion is CX3CR1-dependent as microglia depletion in CX3CR1-KO and hCX3CR1I249/M280 mice did not alleviate retinal degeneration nor microglial morphological activation as observed in CX3CR1-WT mice.


Subject(s)
Diabetes Mellitus, Experimental , Microglia , Humans , Mice , Animals , Microglia/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Diabetes Mellitus, Experimental/pathology , Inflammation/metabolism , Retina/pathology , Carrier Proteins/metabolism , Fibrinogen/metabolism
2.
Cell Rep ; 36(12): 109727, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34551293

ABSTRACT

In traumatic brain injury (TBI), a diversity of brain resident and peripherally derived myeloid cells have the potential to worsen damage and/or to assist in healing. We define the heterogeneity of microglia and macrophage phenotypes during TBI in wild-type (WT) mice and Ccr2-/- mice, which lack macrophage influx following TBI and are resistant to brain damage. We use unbiased single-cell RNA sequencing methods to uncover 25 microglia, monocyte/macrophage, and dendritic cell subsets in acute TBI and normal brains. We find alterations in transcriptional profiles of microglia subsets in Ccr2-/- TBI mice compared to WT TBI mice indicating that infiltrating monocytes/macrophages influence microglia activation to promote a type I IFN response. Preclinical pharmacological blockade of hCCR2 after injury reduces expression of IFN-responsive gene, Irf7, and improves outcomes. These data extend our understanding of myeloid cell diversity and crosstalk in brain trauma and identify therapeutic targets in myeloid subsets.


Subject(s)
Brain Injuries, Traumatic/pathology , Microglia/metabolism , Receptors, CCR2/genetics , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Disease Models, Animal , Down-Regulation , Humans , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferon Type I/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Monocytes/cytology , Monocytes/metabolism , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/deficiency , Receptors, CCR2/metabolism
3.
J Proteome Res ; 20(7): 3689-3700, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34085531

ABSTRACT

Novel therapies and biomarkers are needed for the treatment of acute ischemic stroke (AIS). This study aimed to provide comprehensive insights into the dynamic proteome changes and underlying molecular mechanisms post-ischemic stroke. TMT-coupled proteomic analysis was conducted on mouse brain cortex tissue from five time points up to 4 weeks poststroke in the distal hypoxic-middle cerebral artery occlusion (DH-MCAO) model. We found that nearly half of the detected proteome was altered following stroke, but only ∼8.6% of the changes were at relatively large scales. Clustering on the changed proteome defined four distinct expression patterns characterized by temporal and quantitative changes in innate and adaptive immune response pathways and cytoskeletal and neuronal remodeling. Further analysis on a subset of 309 "top hits", which temporally responded to stroke with relatively large and sustained changes, revealed that they were mostly secreted proteins, highly correlated to different cortical cytokines, and thereby potential pharmacodynamic biomarker candidates for inflammation-targeting therapies. Closer examination of the top enriched neurophysiologic pathways identified 57 proteins potentially associated with poststroke recovery. Altogether, our study generated a rich dataset with candidate proteins worthy of further validation as biomarkers and/or therapeutic targets for stroke. The proteomics data are available in the PRIDE Archive with identifier PXD025077.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Animals , Mice , Proteome/genetics , Proteomics
4.
Bioinformatics ; 37(20): 3670-3672, 2021 Oct 25.
Article in English | MEDLINE | ID: mdl-33901288

ABSTRACT

SUMMARY: We developed Quickomics, a feature-rich R Shiny-powered tool to enable biologists to fully explore complex omics statistical analysis results and perform advanced analysis in an easy-to-use interactive interface. It covers a broad range of secondary and tertiary analytical tasks after primary analysis of omics data is completed. Each functional module is equipped with customizable options and generates both interactive and publication-ready plots to uncover biological insights from data. The modular design makes the tool extensible with ease. AVAILABILITY AND IMPLEMENTATION: Researchers can experience the functionalities with their own data or demo RNA-Seq and proteomics datasets by using the app hosted at http://quickomics.bxgenomics.com and following the tutorial, https://bit.ly/3rXIyhL. The source code under GPLv3 license is provided at https://github.com/interactivereport/Quickomics for local installation. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

5.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33526652

ABSTRACT

Identifying molecular mediators of neural circuit development and/or function that contribute to circuit dysfunction when aberrantly reengaged in neurological disorders is of high importance. The role of the TWEAK/Fn14 pathway, which was recently reported to be a microglial/neuronal axis mediating synaptic refinement in experience-dependent visual development, has not been explored in synaptic function within the mature central nervous system. By combining electrophysiological and phosphoproteomic approaches, we show that TWEAK acutely dampens basal synaptic transmission and plasticity through neuronal Fn14 and impacts the phosphorylation state of pre- and postsynaptic proteins in adult mouse hippocampal slices. Importantly, this is relevant in two models featuring synaptic deficits. Blocking TWEAK/Fn14 signaling augments synaptic function in hippocampal slices from amyloid-beta-overexpressing mice. After stroke, genetic or pharmacological inhibition of TWEAK/Fn14 signaling augments basal synaptic transmission and normalizes plasticity. Our data support a glial/neuronal axis that critically modifies synaptic physiology and pathophysiology in different contexts in the mature brain and may be a therapeutic target for improving neurophysiological outcomes.


Subject(s)
Nerve Degeneration/metabolism , Signal Transduction , Stroke/metabolism , Synapses/metabolism , TWEAK Receptor/metabolism , Animals , Cytokine TWEAK/metabolism , Disease Models, Animal , Female , Hippocampus/physiopathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/physiopathology , Neuronal Plasticity/physiology , Presynaptic Terminals/metabolism , Stroke/physiopathology , Synaptic Transmission/physiology
6.
Life Sci Alliance ; 2(6)2019 12.
Article in English | MEDLINE | ID: mdl-31792059

ABSTRACT

CX3CR1, one of the highest expressed genes in microglia in mice and humans, is implicated in numerous microglial functions. However, the molecular mechanisms underlying Cx3cr1 signaling are not well understood. Here, we analyzed transcriptomes of Cx3cr1-deficient microglia under varying conditions by RNA-sequencing (RNA-seq). In 2-mo-old mice, Cx3cr1 deletion resulted in the down-regulation of a subset of immune-related genes, without substantial epigenetic changes in markers of active chromatin. Surprisingly, Cx3cr1-deficient microglia from young mice exhibited a transcriptome consistent with that of aged Cx3cr1-sufficient animals, suggesting a premature aging transcriptomic signature. Immunohistochemical analysis of microglia in young and aged mice revealed that loss of Cx3cr1 modulates microglial morphology in a comparable fashion. Our results suggest that CX3CR1 may regulate microglial function in part by modulating the expression levels of a subset of inflammatory genes during chronological aging, making Cx3cr1-deficient mice useful for studying aged microglia.


Subject(s)
Aging, Premature/genetics , CX3C Chemokine Receptor 1/deficiency , Microglia/metabolism , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Female , Gene Deletion , Genetic Profile , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/metabolism , Receptors, Chemokine/deficiency , Signal Transduction , Transcriptome
7.
Methods Mol Biol ; 1936: 23-36, 2019.
Article in English | MEDLINE | ID: mdl-30820891

ABSTRACT

Remyelination is the regenerative process whereby myelin sheaths are restored around axons following nervous system injury, allowing reinstatement of electrical impulse conduction, trophic/metabolic support, and axon health. Failure of remyelination in progressive multiple sclerosis is considered to contribute to axon loss, a correlate of clinical decline. Lack of approved pro-regenerative therapies for MS highlights the need to understand the cellular and molecular mechanisms underpinning successful remyelination. One approach is to conduct nonbiased gene expression analyses of cell types which regulate remyelination, such as microglia and monocyte-derived macrophages. Recent technological advances address the challenges of RNA sequencing of small tissue samples, thus allowing relatively small numbers of cells to be isolated from discrete lesions for analysis. Here, we present methods for FACS-based isolation of cells from focal remyelinating lesions of the adult mouse brain and subsequent RNA extraction for sequencing, using isolation of microglia/macrophages as an example.


Subject(s)
Brain/cytology , Remyelination , Sequence Analysis, RNA/methods , Animals , Cell Separation , Central Nervous System/chemistry , Flow Cytometry , Gene Expression Regulation , Macrophages/chemistry , Macrophages/cytology , Mice , Microglia/chemistry , Microglia/cytology
8.
Neuroscience ; 330: 410-20, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27288150

ABSTRACT

Alzheimer's disease (AD), the most common form of dementia in the elderly, is characterized by the presence of extracellular plaques composed of amyloid ß (Aß) peptides and intracellular tau aggregates. The plaques are surrounded by microglia, the brain's resident immune cells, which likely participate in the clearance of Aß by phagocytosis. The microglia that are associated with plaques display an abnormal ameboid morphology and do not respond to tissue damage, in contrast to microglia in healthy brains. Here, we used time lapse confocal microscopy to perform a detailed real-time examination of microglial motility in acute hippocampal brain slices from the 5xFAD mouse model of AD, which was crossed to Cx3cr1(GFP/GFP) mice to achieve microglia-specific GFP expression for visualization. During baseline conditions, microglia around plaques appeared hypermotile, moving the processes that were pointing away from plaques at higher speed than microglia not associated with plaques. Yet, neither plaque-associated, nor plaque-free microglia were able to extend processes toward sites of modest mechanical damage. Application of the selective adenosine A2A receptor antagonist preladenant, which restores microglial response to cellular damage in a mouse model of Parkinson's disease, reduced the hypermotility of plaque-associated microglia, but did not restore motility toward damaged cells in slices from 5xFAD mice. Our results suggest that process hypermotility and resistance to A2A antagonism during response to tissue damage may represent unique functional phenotypes of plaque-associated microglia that impair their ability to function properly in the AD brain.


Subject(s)
Alzheimer Disease/physiopathology , Cell Movement/physiology , Hippocampus/physiopathology , Microglia/physiology , Plaque, Amyloid/physiopathology , Adenosine A2 Receptor Antagonists/pharmacology , Alzheimer Disease/pathology , Animals , Cell Movement/drug effects , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/pathology , Humans , Immunohistochemistry , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Microscopy, Confocal , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Plaque, Amyloid/pathology , Pyrimidines/pharmacology , Receptor, Adenosine A2A/metabolism , Tissue Culture Techniques , Triazoles/pharmacology
9.
J Biol Chem ; 291(3): 1277-88, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26565023

ABSTRACT

Amyloid propagation requires high levels of sequence specificity so that only molecules with very high sequence identity can form cross-ß-sheet structures of sufficient stringency for incorporation into the amyloid fibril. This sequence specificity presents a barrier to the transmission of prions between two species with divergent sequences, termed a species barrier. Here we study the relative effects of protein sequence, seed conformation, and environment on the species barrier strength and specificity for the yeast prion protein Sup35p from three closely related species of the Saccharomyces sensu stricto group; namely, Saccharomyces cerevisiae, Saccharomyces bayanus, and Saccharomyces paradoxus. Through in vivo plasmid shuffle experiments, we show that the major characteristics of the transmission barrier and conformational fidelity are determined by the protein sequence rather than by the cellular environment. In vitro data confirm that the kinetics and structural preferences of aggregation of the S. paradoxus and S. bayanus proteins are influenced by anions in accordance with their positions in the Hofmeister series, as observed previously for S. cerevisiae. However, the specificity of the species barrier is primarily affected by the sequence and the type of anion present during the formation of the initial seed, whereas anions present during the seeded aggregation process typically influence kinetics rather than the specificity of prion conversion. Therefore, our work shows that the protein sequence and the conformation variant (strain) of the prion seed are the primary determinants of cross-species prion specificity both in vivo and in vitro.


Subject(s)
Fungal Proteins/metabolism , Host Specificity , Prions/chemistry , Saccharomyces/metabolism , Biomarkers/metabolism , Chlorides/chemistry , Fungal Proteins/chemistry , Fungal Proteins/genetics , Gene Deletion , Kinetics , Mutation , Peptide Termination Factors/metabolism , Perchlorates/chemistry , Prions/genetics , Prions/metabolism , Prions/pathogenicity , Protein Aggregates , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces/classification , Saccharomyces/growth & development , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sequence Analysis, Protein , Species Specificity , Sulfates/chemistry
10.
J Neuroinflammation ; 12: 228, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26634348

ABSTRACT

BACKGROUND: Millions of people experience traumatic brain injury (TBI) as a result of falls, car accidents, sports injury, and blast. TBI has been associated with the development of neurodegenerative conditions such as Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). In the initial hours and days, the pathology of TBI comprises neuronal injury, breakdown of the blood-brain barrier, and inflammation. At the cellular level, the inflammatory reaction consists of responses by brain-resident microglia, astrocytes, and vascular elements as well as infiltration of peripheral cells. After TBI, signaling by chemokine (C-C motif) ligand 2 (CCL2) to the chemokine (C-C motif) receptor 2 (CCR2) is a key regulator of brain infiltration by monocytes. METHODS: We utilized mice with one or both copies of Ccr2 disrupted by red fluorescent protein (RFP, Ccr2 (RFP/+) and Ccr2 (RFP/RFP) ). We subjected these mice to the mild lateral fluid percussion model of TBI and examined several pathological outcomes 3 days later in order to determine the effects of altered monocyte entry into the brain. RESULTS: Ccr2 deletion reduced monocyte infiltration, diminished lesion cavity volume, and lessened axonal damage after mild TBI, but the microglial reaction to the lesion was not affected. We further examined phosphorylation of the microtubule-associated protein tau, which aggregates in brains of people with TBI, AD, and CTE. Surprisingly, Ccr2 deletion was associated with increased tau mislocalization to the cell body in the cortex and hippocampus by tissue staining and increased levels of phosphorylated tau in the hippocampus by Western blot. CONCLUSIONS: Disruption of CCR2 enhanced tau pathology and reduced cavity volume in the context of TBI. The data reveal a complex role for CCR2(+) monocytes in TBI, as monitored by cavity volume, axonal damage, and tau phosphorylation.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/pathology , Gene Deletion , Receptors, CCR2/deficiency , tau Proteins/metabolism , Animals , Brain Injuries/genetics , Female , Male , Mice , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Receptors, CCR2/genetics , tau Proteins/genetics
11.
Trends Pharmacol Sci ; 36(7): 471-80, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25979813

ABSTRACT

Traumatic brain injury (TBI) affects millions of people worldwide every year. The primary impact initiates the secretion of pro- and anti-inflammatory factors, subsequent recruitment of peripheral immune cells, and activation of brain-resident microglia and astrocytes. Chemokines are major mediators of peripheral blood cell recruitment to damaged tissue, including the TBI brain. Here we review the involvement of specific chemokine pathways in TBI pathology and attempts to modulate these pathways for therapeutic purposes. We focus on chemokine (C-C motif) ligand 2/chemokine (C-C motif) receptor 2 (CCL2/CCR2) and chemokine (C-X-C motif) ligand 12/chemokine (C-X-C motif) receptor 4 (CXCL12/CXCR4). Recent microarray and multiplex expression profiling have also implicated CXCL10 and CCL5 in TBI pathology. Chemokine (C-X3-C motif) ligand 1/chemokine (C-X3-C motif) receptor 1 (CX3CL1/CX3CR1) signaling in the context of TBI is also discussed. Current literature suggests that modulating chemokine signaling, especially CCL2/CCR2, may be beneficial in TBI treatment.


Subject(s)
Brain Injuries/immunology , Brain Injuries/therapy , Cell Communication/immunology , Chemokines/immunology , Animals , Astrocytes/immunology , Brain Injuries/metabolism , Brain Injuries/pathology , Chemokines/metabolism , Disease Models, Animal , Humans , Microglia/immunology , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Signal Transduction
13.
Glia ; 62(8): 1345-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24807189

ABSTRACT

Microglia, the resident immune cells of the central nervous system, exist in either a "resting" state associated with physiological tissue surveillance or an "activated" state in neuroinflammation. We recently showed that ATP is the primary chemoattractor to tissue damage in vivo and elicits opposite effects on the motility of activated microglia in vitro through activation of adenosine A2A receptors. However, whether systemic inflammation affects microglial responses to tissue damage in vivo remains largely unknown. Using in vivo two-photon imaging of mice, we show that injection of lipopolysaccharide (LPS) at levels that can produce both clear neuroinflammation and some features of sepsis significantly reduced the rate of microglial response to laser-induced ablation injury in vivo. Under proinflammatory conditions, microglial processes initially retracted from the ablation site, but subsequently moved toward and engulfed the damaged area. Analyzing the process dynamics in 3D cultures of primary microglia indicated that only A2A , but not A1 or A3 receptors, mediate process retraction in LPS-activated microglia. The A2A receptor antagonists caffeine and preladenant reduced adenosine-mediated process retraction in activated microglia in vitro. Finally, administration of preladenant before induction of laser ablation in vivo accelerated the microglial response to injury following systemic inflammation. The regulation of rapid microglial responses to sites of injury by A2A receptors could have implications for their ability to respond to the neuronal death occurring under conditions of neuroinflammation in neurodegenerative disorders.


Subject(s)
Cerebral Cortex/injuries , Inflammation/physiopathology , Microglia/physiology , Neuroimmunomodulation/physiology , Animals , Cell Culture Techniques , Cell Movement/drug effects , Cell Movement/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Female , Immunologic Factors/pharmacology , Inflammation/drug therapy , Inflammation/pathology , Lasers/adverse effects , Lipopolysaccharides , Male , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Neuroimmunomodulation/drug effects , Pyrimidines/pharmacology , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A3/metabolism , Sepsis/drug therapy , Sepsis/pathology , Sepsis/physiopathology , Triazoles/pharmacology
14.
Neurobiol Dis ; 67: 191-202, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24632419

ABSTRACT

Microglia, the immune cells of the central nervous system, constantly survey the parenchyma in the healthy brain to maintain homeostasis. When a disturbance, such as cell death, results in ATP release in vivo, microglial processes respond by utilizing P2Y12 purinergic receptors to trigger extension toward the site of damage. Processes ultimately surround the injury site, preventing the spread of harmful cellular constituents and assisting with tissue repair. In contrast to the healthy brain, many neurodegenerative diseases, including Parkinson's disease, are characterized by the presence of neuroinflammation. Yet, the ability of microglia to respond to tissue damage under pro-inflammatory conditions has not been well studied. To assess the ability of microglia to respond to tissue injury and localized cell death in the context of Parkinson's disease, we performed confocal imaging of acute brain slices from mice with microglia-specific green fluorescent protein expression. Microglia in coronal slices containing the substantia nigra extend processes toward a mechanical injury in a P2Y12 receptor-dependent manner. However, microglia in mice treated for 5days with 20mg/kg/day 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) show significantly reduced process displacement toward the injury compared to microglia in control animals. Pre-treatment of slices from MPTP-injected mice with the A2A receptor-selective antagonist preladenant restores the ability of activated microglia to respond to tissue damage. These data support the hypothesis that chronic inflammation impedes microglial motility in response to further injury, such as cell death, and suggest that some aspects of the neuroprotection observed with adenosine A2A receptor antagonists may involve direct or indirect actions at microglia.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , MPTP Poisoning/immunology , MPTP Poisoning/metabolism , Microglia/drug effects , Microglia/metabolism , Substantia Nigra/metabolism , Animals , Disease Models, Animal , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Substantia Nigra/drug effects
15.
J Med Chem ; 57(6): 2334-56, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24512267

ABSTRACT

NMDA receptors are tetrameric complexes composed of GluN1 and GluN2A-D subunits that mediate a slow Ca(2+)-permeable component of excitatory synaptic transmission. NMDA receptors have been implicated in a wide range of neurological diseases and thus represent an important therapeutic target. We herein describe a novel series of pyrrolidinones that selectively potentiate only NMDA receptors that contain the GluN2C subunit. The most active analogues tested were over 100-fold selective for recombinant GluN2C-containing receptors over GluN2A/B/D-containing NMDA receptors as well as AMPA and kainate receptors. This series represents the first class of allosteric potentiators that are selective for diheteromeric GluN2C-containing NMDA receptors.


Subject(s)
Excitatory Amino Acid Agonists/chemical synthesis , Excitatory Amino Acid Agonists/pharmacology , Receptors, N-Methyl-D-Aspartate/agonists , Animals , Chromatography, High Pressure Liquid , Computational Biology , Drug Design , High-Throughput Screening Assays , Humans , Indicators and Reagents , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Oocytes/drug effects , Patch-Clamp Techniques , Pyrrolidinones/chemical synthesis , Pyrrolidinones/pharmacology , Pyruvates/chemical synthesis , Pyruvates/pharmacology , Stereoisomerism , Structure-Activity Relationship , Xenopus laevis
16.
J Biol Chem ; 288(21): 15291-302, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23548902

ABSTRACT

Microglia, the resident immune cells of the central nervous system (CNS), monitor the brain for disturbances of tissue homeostasis by constantly moving their fine processes. Microglia respond to tissue damage through activation of ATP/ADP receptors followed by directional process extension to the damaged area. A common feature of several neurodegenerative diseases is the loss of norepinephrine, which might contribute to the associated neuroinflammation. We carried out a high resolution analysis of the effects of norepinephrine (NE) on microglial process dynamics in acute brain slices from mice that exhibit microglia-specific enhanced green fluorescent protein expression. Bath application of NE to the slices resulted in significant process retraction in microglia. Analysis of adrenergic receptor expression with quantitative PCR indicated that resting microglia primarily express ß2 receptors but switch expression to α2A receptors under proinflammatory conditions modeled by LPS treatment. Despite the differential receptor expression, NE caused process retraction in both resting and LPS-activated microglia cultured in the gelatinous substrate Matrigel in vitro. The use of subtype-selective receptor agonists and antagonists confirmed the involvement of ß2 receptors in mediating microglial process dynamics in resting cells and α2A receptors in activated cells. Co-application of NE with ATP to resting microglia blocked the ATP-induced process extension and migration in isolated microglia, and ß2 receptor antagonists prolonged ATP effects in brain slice tissues, suggesting the presence of cross-talk between adrenergic and purinergic signaling in microglia. These data show that the neurotransmitter NE can modulate microglial motility, which could affect microglial functions in pathogenic situations of either elevated or reduced NE levels.


Subject(s)
Adenosine Triphosphate/pharmacology , Adrenergic alpha-Agonists/pharmacology , Cell Movement/drug effects , Microglia/metabolism , Norepinephrine/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic alpha-Agonists/metabolism , Animals , Cell Movement/physiology , Cells, Cultured , Mice , Mice, Transgenic , Microglia/cytology , Norepinephrine/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
17.
Cell Signal ; 24(6): 1351-60, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22306780

ABSTRACT

Protease-activated receptor 1 (PAR1) is an unusual GPCR that interacts with multiple G protein subfamilies (G(q/11), G(i/o), and G(12/13)) and their linked signaling pathways to regulate a broad range of pathophysiological processes. However, the molecular mechanisms whereby PAR1 interacts with multiple G proteins are not well understood. Whether PAR1 interacts with various G proteins at the same, different, or overlapping binding sites is not known. Here we investigated the functional and specific binding interactions between PAR1 and representative members of the G(q/11), G(i/o), and G(12/13) subfamilies. We report that G(q/11) physically and functionally interacts with specific amino acids within the second intracellular (i2) loop of PAR1. We identified five amino acids within the PAR1 i2 loop that, when mutated individually, each markedly reduced PAR1 activation of linked inositol phosphate formation in transfected COS-7 cells (functional PAR1-null cells). Among these mutations, only R205A completely abolished direct G(q/11) binding to PAR1 and also PAR1-directed inositol phosphate and calcium mobilization in COS-7 cells and PAR1-/- primary astrocytes. In stark contrast, none of the PAR1 i2 loop mutations disrupted direct PAR1 binding to either G(o) or G(12), or their functional coupling to linked pertussis toxin-sensitive ERK phosphorylation and C3 toxin-sensitive Rho activation, respectively. In astrocytes, our findings suggest that PAR1-directed calcium signaling involves a newly appreciated G(q/11)-PLCε pathway. In summary, we have identified key molecular determinants for PAR1 interactions with G(q/11), and our findings support a model where G(q/11), G(i/o) or G(12/13) each bind to distinct sites within the cytoplasmic regions of PAR1.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptor, PAR-1/metabolism , Amino Acids/chemistry , Amino Acids/genetics , Amino Acids/metabolism , Animals , Astrocytes/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , Gene Deletion , Mice , Mutation , Phosphoinositide Phospholipase C/metabolism , Receptor, PAR-1/chemistry , Receptor, PAR-1/genetics , Signal Transduction
18.
J Bone Miner Metab ; 29(5): 515-25, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21246227

ABSTRACT

Voltage-dependent L-type calcium channels (VDCC) play important roles in many cellular processes. The interaction of the actin cytoskeleton with the channel in nonexcitable cells is less well understood. We performed whole-cell patch-clamp surface biotinylation and calcium imaging on different osteoblast cells to determine channel kinetics, amplitude, surface abundance, and intracellular calcium, respectively. Patch-clamp studies showed that actin polymerization by phalloidin increased the peak current density of I (Ca), whereas actin depolymerization by cytochalasin D (CD) significantly decreased the current amplitude. This result is consistent with calcium imaging, which showed that CD significantly decreased Bay K8644-induced intracellular calcium increase. Surface biotinylation studies showed that CD is not able to affect the surface expression of the pore-forming subunit α(1C). Interestingly, application of CD caused a significantly negative shift in the steady-state inactivation kinetics of I (Ca). There were decreases in the voltage at half-maximal inactivation that changed in a dose-dependent manner. CD also reduced the effect of activated vitamin D(3) (1α,25-D3) on VDCC and intracellular calcium. We conclude that in osteoblasts the actin cytoskeleton affects α(1C) by altering the channel kinetic properties, instead of changing the surface expression, and it is able to regulate 1α,25-D3 signaling through VDCC. Our study provides a new insight into calcium regulation in osteoblasts, which are essential in many physiological functions of this cell.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium Channels, L-Type/metabolism , Osteoblasts/metabolism , Actin Cytoskeleton/drug effects , Animals , Calcium/metabolism , Cell Line, Tumor , Cholecalciferol/pharmacology , Cytochalasin D/pharmacology , Mice , Osteoblasts/drug effects , Patch-Clamp Techniques , Protein Transport/drug effects , Rats
19.
Mol Microbiol ; 76(6): 1483-99, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20444092

ABSTRACT

Self-perpetuating amyloid-based protein isoforms (prions) transmit neurodegenerative diseases in mammals and phenotypic traits in yeast. Although mechanisms that control species specificity of prion transmission are poorly understood, studies of closely related orthologues of yeast prion protein Sup35 demonstrate that cross-species prion transmission is modulated by both genetic (specific sequence elements) and epigenetic (prion variants, or 'strains') factors. Depending on the prion variant, the species barrier could be controlled at the level of either heterologous co-aggregation or conversion of the aggregate-associated heterologous protein into a prion polymer. Sequence divergence influences cross-species transmission of different prion variants in opposing ways. The ability of a heterologous prion domain to either faithfully reproduce or irreversibly switch the variant-specific prion patterns depends on both sequence divergence and the prion variant. Sequence variations within different modules of prion domains contribute to transmission barriers in different cross-species combinations. Individual amino acid substitutions within short amyloidogenic stretches drastically alter patterns of cross-species prion conversion, implicating these stretches as major determinants of species specificity.


Subject(s)
Gene Expression Regulation, Fungal , Gene Transfer, Horizontal , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Polymorphism, Genetic , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Amyloid/metabolism , Molecular Sequence Data , Sequence Alignment
20.
Parkinsonism Relat Disord ; 15 Suppl 3: S195-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20082989

ABSTRACT

Microglia are motile immune-competent cells of the central nervous system. They assume a highly branched morphology and monitor the brain parenchyma under physiological conditions. In the presence of injury, microglia retract their branching processes, migrate to the site of injury, and help clear cellular debris by phagocytosis. This response appears to be mediated in part by ATP released at the site of injury. Here, we review the evidence for the involvement of ATP and the purinergic P2Y(12) receptor in microglial process extension and chemoattraction to injury. We subsequently discuss recent findings regarding a switch of this chemotactic response to ATP in activated, or proinflammatory, microglia. Specifically, in LPS-activated microglia, ATP induces process retraction and repulsive migration, effects opposite to those seen in unstimulated cells. These repulsive effects of ATP are mediated by the G(s)-coupled adenosine A(2A) receptor and depend on the breakdown of ATP to adenosine. Thus, ATP-induced repulsion by activated microglia involves upregulation of the adenosine A(2A) receptor and coincident downregulation of the P2Y(12) receptor. The roles of the A(2A) receptor in brain pathologies such as Parkinson's disease and ischemia are also examined. We propose that the effects of A(2A) receptor antagonists on brain injury may be in part due to the inactivation of A(2A) on activated microglia.


Subject(s)
Adenosine/metabolism , Adenosine/pharmacology , Cell Movement/drug effects , Microglia/drug effects , Adenosine Diphosphate , Adenosine Triphosphate , Animals , Brain Injuries/pathology , Humans , Microglia/metabolism , Receptor, Adenosine A2A/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...