Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Kidney Int ; 105(2): 293-311, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37995909

ABSTRACT

The kidney medulla is a specialized region with important homeostatic functions. It has been implicated in genetic and developmental disorders along with ischemic and drug-induced injuries. Despite its role in kidney function and disease, the medulla's baseline gene expression and epigenomic signatures have not been well described in the adult human kidney. Here we generated and analyzed gene expression (RNA-seq), chromatin accessibility (ATAC-seq), chromatin conformation (Hi-C) and spatial transcriptomic data from the adult human kidney cortex and medulla. Tissue samples were obtained from macroscopically dissected cortex and medulla of tumor-adjacent normal material in nephrectomy specimens from five male patients. We used these carefully annotated specimens to reassign incorrectly labeled samples in the larger public Genotype-Tissue Expression (GTEx) Project, and to extract meaningful medullary gene expression signatures. Using integrated analysis of gene expression, chromatin accessibility and conformation profiles, we found insights into medulla development and function and then validated this by spatial transcriptomics and immunohistochemistry. Thus, our datasets provide a valuable resource for functional annotation of variants from genome-wide association studies and are freely accessible through an epigenome browser portal.


Subject(s)
Genome-Wide Association Study , Multiomics , Adult , Humans , Male , Chromatin , Kidney , Transcriptome
2.
Nephrol Dial Transplant ; 37(5): 847-859, 2022 04 25.
Article in English | MEDLINE | ID: mdl-34865099

ABSTRACT

BACKGROUND: The sodium-glucose cotransporter-2 (SGLT2) inhibitor empagliflozin lowers blood glucose via reduced tubular reabsorption of filtered glucose and is an important new therapy for diabetic nephropathy (DN). This study tested whether treatment with empagliflozin would ameliorate proteinuria and the pathologic alterations of DN including podocyte number and integrity in the leptin-deficient BTBR ob/ob mouse model of DN. METHODS: Study cohorts included wild-type (WT) BTBR mice, untreated diabetic BTBR ob/ob mice and mice treated with empagliflozin for 6 weeks after development of established DN at 18 weeks of age. RESULTS: Hyperglycemia, proteinuria, serum creatinine, accumulation of mesangial matrix and the extent of mesangiolysis were reversed with empagliflozin treatment. Treatment with empagliflozin resulted in an increased podocyte number and podocyte density, improvement in the degree of podocyte foot process effacement and parietal epithelial cell activation. SGLT2 inhibition reduced renal oxidative stress, measured by urinary excretion of markers of RNA/DNA damage and in situ demonstration of decreased carbonyl oxidation. There was no discernable difference in accumulations of advanced glycation end-products by immunohistochemistry. CONCLUSION: The structural improvements seen in BTBR ob/ob mice treated with empagliflozin provide insights into potential long-term benefits for humans with DN, for whom there is no comparable biopsy information to identify structural changes effected by SGLT2 inhibition. The findings suggest SGLT2 inhibition may ameliorate DN through glucose lowering-dependent and -independent mechanisms that lead to podocyte restoration and delay or reversal of disease progress.


Subject(s)
Benzhydryl Compounds , Diabetes Mellitus , Diabetic Nephropathies , Glucosides , Sodium-Glucose Transporter 2 Inhibitors , Animals , Benzhydryl Compounds/therapeutic use , Blood Glucose , Diabetes Mellitus/drug therapy , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Glucosides/therapeutic use , Mice , Mice, Inbred Strains , Proteinuria , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
3.
Front Neurol ; 12: 624696, 2021.
Article in English | MEDLINE | ID: mdl-33796061

ABSTRACT

The late neuropathological effects of traumatic brain injury have yet to be fully elucidated, particularly with respect to community-based cohorts. To contribute to this critical gap in knowledge, we designed a multimodal neuropathological study, integrating traditional and quantitative approaches to detect pathologic changes in 532 consecutive brain autopsies from participants in the Adult Changes in Thought (ACT) study. Diagnostic evaluation including assessment for chronic traumatic encephalopathy (CTE) and quantitative immunoassay-based methods were deployed to examine levels of pathological (hyperphosphorylated) tau (pTau) and amyloid (A) ß in brains from ACT participants with (n = 107) and without (n = 425) history of remote TBI with loss of consciousness (w/LOC). Further neuropathological assessments included immunohistochemistry for α-synuclein and phospho-TDP-43 pathology and astro- (GFAP) and micro- (Iba1) gliosis, mass spectrometry analysis of free radical injury, and gene expression evaluation (RNA sequencing) in a smaller sub-cohort of matched samples (49 cases with TBI and 49 non-exposed matched controls). Out of 532 cases, only 3 (0.6%-none with TBI w/LOC history) showed evidence of the neuropathologic signature of chronic traumatic encephalopathy (CTE). Across the entire cohort, the levels of pTau and Aß showed expected differences for brain region (higher levels in temporal cortex), neuropathological diagnosis (higher in participants with Alzheimer's disease), and APOE genotype (higher in participants with one or more APOE ε4 allele). However, no differences in PHF-tau or Aß1-42 were identified by Histelide with respect to the history of TBI w/LOC. In a subset of TBI cases with more carefully matched control samples and more extensive analysis, those with TBI w/LOC history had higher levels of hippocampal pTau but no significant differences in Aß, α-synuclein, pTDP-43, GFAP, Iba1, or free radical injury. RNA-sequencing also did not reveal significant gene expression associated with any measure of TBI exposure. Combined, these findings suggest long term neuropathological changes associated with TBI w/LOC may be subtle, involve non-traditional pathways of neurotoxicity and neurodegeneration, and/or differ from those in autopsy cohorts specifically selected for neurotrauma exposure.

4.
Elife ; 62017 11 09.
Article in English | MEDLINE | ID: mdl-29120328

ABSTRACT

As more people live longer, age-related neurodegenerative diseases are an increasingly important societal health issue. Treatments targeting specific pathologies such as amyloid beta in Alzheimer's disease (AD) have not led to effective treatments, and there is increasing evidence of a disconnect between traditional pathology and cognitive abilities with advancing age, indicative of individual variation in resilience to pathology. Here, we generated a comprehensive neuropathological, molecular, and transcriptomic characterization of hippocampus and two regions cortex in 107 aged donors (median = 90) from the Adult Changes in Thought (ACT) study as a freely-available resource (http://aging.brain-map.org/). We confirm established associations between AD pathology and dementia, albeit with increased, presumably aging-related variability, and identify sets of co-expressed genes correlated with pathological tau and inflammation markers. Finally, we demonstrate a relationship between dementia and RNA quality, and find common gene signatures, highlighting the importance of properly controlling for RNA quality when studying dementia.


Subject(s)
Aging/pathology , Cerebral Cortex/pathology , Gene Expression Profiling , Hippocampus/pathology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Dementia/pathology , Female , Humans , Male
5.
FASEB J ; 29(5): 1754-62, 2015 May.
Article in English | MEDLINE | ID: mdl-25593125

ABSTRACT

Several lines of evidence support immune response in brain as a mechanism of injury in Alzheimer disease (AD). Moreover, immune activation is heightened in apolipoprotein E (APOE) ε4 carriers; inhibitors of prostaglandin (PG) synthesis show a partially protective effect on AD risk from APOE ε4; and genetic variants in triggering receptor expressed on myeloid cells 2 (TREM2) are a rare but potent risk for AD. We tested the hypothesis that APOE ε4 inheritance modulates both the PGE2 pathway and TREM2 expression using primary murine microglia from targeted replacement (TR) APOE3/3 and APOE4/4 mice. Microglial cyclooxygenase-2, microsomal PGE synthase, and PGE2 expression were increased 2- to 25-fold in both genotypes by TLR activators; however, this induction was significantly (P < 0.01) greater in TR APOE4/4 microglia with TLR3 and TLR4 activators. Microglial TREM2 expression was reduced approximately 85% by all TLR activators; this reduction was approximately one-third greater in microglia from TR APOE4/4 mice. Importantly, both receptor-associated protein and a nuclear factor κ-light-chain-enhancer inhibitor blocked TR APOE4/4-dependent effects on the PGE2 pathway but not on TREM2 expression. These data demonstrate complementary, but mechanistically distinct, regulation of pro- and anti-inflammatory mediators in TR APOE4/4 murine microglia that yields a more proinflammatory state than with TR APOE3/3.


Subject(s)
Apolipoprotein E3/physiology , Apolipoprotein E4/physiology , Apolipoproteins E/metabolism , Dinoprostone/metabolism , Membrane Glycoproteins/metabolism , Microglia/metabolism , Myeloid Cells/metabolism , Receptors, Immunologic/metabolism , Animals , Blotting, Western , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/immunology , Myeloid Cells/cytology , Prostaglandin-E Synthases , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
6.
Am J Pathol ; 185(1): 230-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25452117

ABSTRACT

Recent studies underline the potential relevance of microglial innate immune activation in Alzheimer disease. Primary mouse microglia that lack prostaglandin E2 receptor subtype 2 (EP2) show decreased innate immune-mediated neurotoxicity and increased amyloid ß (Aß) peptide phagocytosis, features that were replicated in vivo. Here, we tested the hypothesis that scavenger receptor CD36 is an effector of EP2-regulated Aß phagocytosis. CD36 expression was 143-fold greater in mouse primary microglia than in primary astrocytes. Three different means of suppressing EP2 signaling increased and an agonist of EP2 decreased CD36 expression in primary wild-type microglia. Activation of Toll-like receptor (TLR) 3, TLR4, and TLR7, but not TLR2 or TLR9, reduced primary microglial CD36 transcription and cell surface CD36 protein and reduced Aß42 phagocytosis as well. At each step, the effects of innate immune activation on CD36 were reversed by at least 50% by an EP2 antagonist, and this partial rescue of microglia Aß42 phagocytosis was largely mediated by CD36 activity. Finally, we showed in hippocampus of wild-type mice that innate immune activation suppressed CD36 expression by an EP2-dependent mechanism. Taken together with results of others that found brain clearance of Aß peptides and behavioral improvements mediated by CD36 in mice, regulation of CD36-mediated Aß phagocytosis by suppression of EP2 signaling may provide a new approach to suppressing some aspects of Alzheimer disease pathogenesis.


Subject(s)
Amyloid beta-Peptides/metabolism , CD36 Antigens/metabolism , Microglia/metabolism , Peptide Fragments/metabolism , Phagocytosis , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Alzheimer Disease/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain/embryology , Brain/metabolism , CHO Cells , Cricetulus , Disease Models, Animal , Hippocampus/metabolism , Immunity, Innate , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Neurotoxins/chemistry , Toll-Like Receptors/metabolism
7.
ACS Appl Mater Interfaces ; 6(23): 20597-602, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25375633

ABSTRACT

Although numerous kinds of waterborne, nacre-mimetic films with excellent properties have been fabricated via different assembly methods, it remains difficult to put those kinds of lightweight materials into practical applications because they are sensitive to water in the environment. Herein, a simple superhydrophobic modification method was used to enhance the repellency of film to water and/or corrosive liquids in the environment. Furthermore, it lowered the gas transmission rate of the films dramatically and improved the heat and flame shield capabilities. This approach could also be applied to other kinds of nacre-mimetic films, proving to be a versatile, low-cost, fast, and facile method to produce large-area and thick, waterborne, multifunctional films with excellent repellency to water and some corrosive liquids in the environment, which will pave the road for the practical applications of nacre-mimetic films.

8.
Arterioscler Thromb Vasc Biol ; 33(11): 2625-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23968976

ABSTRACT

OBJECTIVE: Elevated serum phosphate has emerged as a major risk factor for vascular calcification. The sodium-dependent phosphate cotransporter, PiT-1, was previously shown to be required for phosphate-induced osteogenic differentiation and calcification of cultured human vascular smooth muscle cells (VSMCs), but its importance in vascular calcification in vivo and the potential role of its homologue, PiT-2, have not been determined. We investigated the in vivo requirement for PiT-1 in vascular calcification using a mouse model of chronic kidney disease and the potential compensatory role of PiT-2 using in vitro knockdown and overexpression strategies. APPROACH AND RESULTS: Mice with targeted deletion of PiT-1 in VSMCs were generated (PiT-1(Δsm)). PiT-1 mRNA levels were undetectable, whereas PiT-2 mRNA levels were increased 2-fold in the vascular aortic media of PiT-1(Δsm) compared with PiT-1(flox/flox) control. When arterial medial calcification was induced in PiT-1(Δsm) and PiT-1(flox/flox) by chronic kidney disease followed by dietary phosphate loading, the degree of aortic calcification was not different between genotypes, suggesting compensation by PiT-2. Consistent with this possibility, VSMCs isolated from PiT-1(Δsm) mice had no PiT-1 mRNA expression, increased PiT-2 mRNA levels, and no difference in sodium-dependent phosphate uptake or phosphate-induced matrix calcification compared with PiT-1(flox/flox) VSMCs. Knockdown of PiT-2 decreased phosphate uptake and phosphate-induced calcification of PiT-1(Δsm) VSMCs. Furthermore, overexpression of PiT-2 restored these parameters in human PiT-1-deficient VSMCs. CONCLUSIONS: PiT-2 can mediate phosphate uptake and calcification of VSMCs in the absence of PiT-1. Mechanistically, PiT-1 and PiT-2 seem to serve redundant roles in phosphate-induced calcification of VSMCs.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Renal Insufficiency, Chronic/physiopathology , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Vascular Calcification/physiopathology , Animals , Aorta/cytology , Aorta/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Phosphates/metabolism , RNA, Messenger/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Uremia/genetics , Uremia/metabolism , Uremia/physiopathology , Vascular Calcification/genetics , Vascular Calcification/metabolism
9.
Am J Pathol ; 183(3): 905-17, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23831297

ABSTRACT

Apolipoprotein E4 (APOE4) genotype is the strongest genetic risk factor for late-onset Alzheimer disease and confers a proinflammatory, neurotoxic phenotype to microglia. Here, we tested the hypothesis that bone marrow cell APOE genotype modulates pathological progression in experimental Alzheimer disease. We performed bone marrow transplants (BMT) from green fluorescent protein-expressing human APOE3/3 or APOE4/4 donor mice into lethally irradiated 5-month-old APPswe/PS1ΔE9 mice. Eight months later, APOE4/4 BMT-recipient APPswe/PS1ΔE9 mice had significantly impaired spatial working memory and increased detergent-soluble and plaque Aß compared with APOE3/3 BMT-recipient APPswe/PS1ΔE9 mice. BMT-derived microglia engraftment was significantly reduced in APOE4/4 recipients, who also had correspondingly less cerebral apoE. Gene expression analysis in cerebral cortex of APOE3/3 BMT recipients showed reduced expression of tumor necrosis factor-α and macrophage migration inhibitory factor (both neurotoxic cytokines) and elevated immunomodulatory IL-10 expression in APOE3/3 recipients compared with those that received APOE4/4 bone marrow. This was not due to detectable APOE-specific differences in expression of microglial major histocompatibility complex class II, C-C chemokine receptor (CCR) type 1, CCR2, CX3C chemokine receptor 1 (CX3CR1), or C5a anaphylatoxin chemotactic receptor (C5aR). Together, these findings suggest that BMT-derived APOE3-expressing cells are superior to those that express APOE4 in their ability to mitigate the behavioral and neuropathological changes in experimental Alzheimer disease.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Apolipoprotein E3/metabolism , Apolipoprotein E4/metabolism , Behavior, Animal , Bone Marrow Transplantation , Alzheimer Disease/immunology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Animals, Newborn , Cells, Cultured , Chimera/metabolism , Disease Models, Animal , Green Fluorescent Proteins/metabolism , Habituation, Psychophysiologic , Hematopoiesis , Hippocampus/pathology , Humans , Immunity, Innate , Immunomodulation/immunology , Memory, Short-Term , Mice , Mice, Inbred C57BL , Microglia/pathology , Monocytes/pathology , Phenotype , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
10.
Exp Mol Pathol ; 94(2): 366-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23318649

ABSTRACT

Alzheimer's disease (AD) neuropathology is characterized by innate immune activation primarily through prostaglandin E2 (PGE2) signaling. Dedicator of cytokinesis 2 (DOCK2) is a guanyl nucleotide exchange factor expressed exclusively in microglia in the brain and is regulated by PGE2 receptor EP2. DOCK2 modulates microglia cytokine secretion, phagocytosis, and paracrine neurotoxicity. EP2 ablation in experimental AD results in reduced oxidative damage and amyloid beta (Aß) burden. This discovery led us to hypothesize that genetic ablation of DOCK2 would replicate the anti-Aß effects of loss of EP2 in experimental AD. To test this hypothesis, we crossed mice that lacked DOCK2 (DOCK2-/-), were hemizygous for DOCK2 (DOCK2+/-), or that expressed two DOCK2 genes (DOCK2+/+) with APPswe-PS1Δe9 mice (a model of AD). While we found no DOCK2-dependent differences in cortex or in hippocampal microglia density or morphology in APPswe-PS1Δe9 mice, cerebral cortical and hippocampal Aß plaque area and size were significantly reduced in 10-month-old APPswe-PS1Δe9/DOCK2-/- mice compared with APPswe-PS1Δe9/DOCK2+/+ controls. DOCK2 hemizygous APPswe-PS1Δe9 mice had intermediate Aß plaque levels. Interestingly, soluble Aß42 was not significantly different among the three genotypes, suggesting the effects were mediated specifically in fibrillar Aß. In combination with earlier cell culture results, our in vivo results presented here suggest DOCK2 contributes to Aß plaque burden via regulation of microglial innate immune function and may represent a novel therapeutic target for AD.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , GTPase-Activating Proteins/metabolism , Plaque, Amyloid/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Brain/pathology , Dinoprostone/metabolism , Disease Models, Animal , GTPase-Activating Proteins/genetics , Genotype , Guanine Nucleotide Exchange Factors , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism
11.
J Neuroimmune Pharmacol ; 8(1): 87-93, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22718277

ABSTRACT

Multiple lines of evidence indicate that regional brain eicosanoid signaling is important in initiation and progression of neurodegenerative conditions that have neuroinflammatory pathologic component, such as AD. We hypothesized that PGE(2) receptor subtype 1 (EP1) signaling (linked to intracellular Ca(2+) release) regulates Aß peptide neurotoxicity and tested this in two complementary in vitro models: a human neuroblastoma cell line (MC65) producing Aß(1-40) through conditional expression of the APP C-terminal portion, and murine primary cortical neuron cultures exposed to Aß(1-42). In MC65 cells, EP1 receptor antagonist SC-51089 reduced Aß neurotoxicity ~50 % without altering high molecular weight Aß immunoreactive species formation. Inositol-3-phosphate receptor antagonist 2-aminoethoxy-diphenyl borate offered similar protection. SC-51089 largely protected the neuron cultures from synthetic Aß(1-42) neurotoxicity. Nimodipine, a Ca(2+) channel blocker, was completely neuroprotective in both models. Based on these data, we conclude that suppressing neuronal EP1 signaling may represent a promising therapeutic approach to ameliorate Aß peptide neurotoxicity.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Dinoprostone/metabolism , Neurotoxicity Syndromes/prevention & control , Prostaglandin Antagonists/pharmacology , Receptors, Prostaglandin E, EP1 Subtype/drug effects , Animals , Blotting, Western , Calcium Channel Blockers/pharmacology , Cell Line, Tumor , Cells, Cultured , Coloring Agents , Humans , Hydrazines/pharmacology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurotoxicity Syndromes/pathology , Nimodipine/pharmacology , Oxazepines/pharmacology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/toxicity , Tetrazolium Salts , Thiazoles
12.
J Neuroinflammation ; 9: 192, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22883744

ABSTRACT

BACKGROUND: Inheritance of the human ε4 allele of the apolipoprotein (apo) E gene (APOE) significantly increases the risk of developing Alzheimer's disease (AD), in addition to adversely influencing clinical outcomes of other neurologic diseases. While apoE isoforms differentially interact with amyloid ß (Aß), a pleiotropic neurotoxin key to AD etiology, more recent work has focused on immune regulation in AD pathogenesis and on the mechanisms of innate immunomodulatory effects associated with inheritance of different APOE alleles. APOE genotype modulates expression of proximal genes including APOC1, which encodes a small apolipoprotein that is associated with Aß plaques. Here we tested the hypothesis that APOE-genotype dependent innate immunomodulation may be mediated in part by apoC-I. METHODS: ApoC-I concentration in cerebrospinal fluid from control subjects of differing APOE genotypes was quantified by ELISA. Real-time PCR and ELISA were used to analyze apoC-I mRNA and protein expression, respectively, in liver, serum, cerebral cortex, and cultured primary astrocytes derived from mice with targeted replacement of murine APOE for human APOE ε3 or ε4. ApoC-I direct modulation of innate immune activity was investigated in cultured murine primary microglia and astrocytes, as well as human differentiated macrophages, using specific toll-like receptor agonists LPS and PIC as well as Aß. RESULTS: ApoC-I levels varied with APOE genotype in humans and in APOE targeted replacement mice, with ε4 carriers showing significantly less apoC-I in both species. ApoC-I potently reduced pro-inflammatory cytokine secretion from primary murine microglia and astrocytes, and human macrophages, stimulated with LPS, PIC, or Aß. CONCLUSIONS: ApoC-I is immunosuppressive. Our results illuminate a novel potential mechanism for APOE genotype risk for AD; one in which patients with an ε4 allele have decreased expression of apoC-I resulting in increased innate immune activity.


Subject(s)
Apolipoprotein C-I/metabolism , Gene Expression Regulation/genetics , Neuroglia/metabolism , Aged , Amyloid beta-Peptides/pharmacology , Animals , Animals, Newborn , Apolipoprotein C-I/cerebrospinal fluid , Apolipoprotein C-I/genetics , Apolipoprotein C-I/pharmacology , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/drug effects , Genotype , Glial Fibrillary Acidic Protein/metabolism , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/drug effects , Poly I-C/pharmacology
13.
FASEB J ; 26(7): 3075-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22499581

ABSTRACT

A major therapeutic target for Parkinson's disease (PD) is providing increased glial-derived neurotrophic factor (GDNF) to dopaminergic neurons. We tested the hypothesis that innate immune activation increases astrocyte GDNF production and that this is regulated by specific eicosanoid receptors. Innate immune-activated primary murine astrocytes were assayed for GDNF expression and secretion. Controls were agent vehicle exposure and wild-type mice. Rank order for up to 10-fold selectively increased GDNF expression was activators of TLR3 > TLR2 or TLR4 > TLR9. TLR3 activator-stimulated GDNF expression was selectively JNK-dependent, followed cyclooxygenase (COX)-2, was coincident with membranous PGE(2) synthase, and was not significantly altered by a nonspecific COX- or a COX-2-selective inhibitor. Specific eicosanoid receptors had opposing effects on TLR3 activator-induced GDNF expression: ∼60% enhancement by blocking or ablating of PGE(2) receptor subtype 1 (EP1), ∼30% enhancement by activating PGF(2α) receptor or thromboxane receptor, or ∼15% enhancement by activating EP4. These results demonstrate functionally antagonistic eicosanoid receptor subtype regulation of innate immunity-induced astrocyte GDNF expression and suggest that selective inhibition of EP1 signaling might be a means to augment astrocyte GDNF secretion in the context of innate immune activation in diseased regions of brain in PD.


Subject(s)
Astrocytes/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Receptors, Eicosanoid/metabolism , Toll-Like Receptors/metabolism , Animals , Astrocytes/immunology , Base Sequence , Cells, Cultured , DNA Primers/genetics , Glial Cell Line-Derived Neurotrophic Factor/deficiency , Glial Cell Line-Derived Neurotrophic Factor/genetics , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Receptors, Eicosanoid/classification , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism
14.
Curr Opin Neurobiol ; 21(6): 920-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21907569

ABSTRACT

The largest genetic risk for late-onset Alzheimer's disease (AD) resides at the apolipoprotein E gene (APOE) locus, which has three common alleles (ɛ2, ɛ3, ɛ4) that encode three isoforms (apoE2, apoE3, apoE4). The very strong association of the APOE ɛ4 allele with AD risk and its role in the accumulation of amyloid ß in brains of people and animal models solidify the biological relevance of apoE isoforms but do not provide mechanistic insight. The innate immune response is consistently observed in AD and is a likely contributor to neuronal injury and response to injury. Here we review emerging data showing that apoE isoform regulation of multiple facets of the innate immune response in the brain may alter AD not only through amyloid ß-dependent mechanisms, but also through other, amyloid ß-independent mechanisms.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/immunology , Apolipoproteins E/genetics , Apolipoproteins E/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Alzheimer Disease/metabolism , Animals , Genetic Predisposition to Disease , Humans , Protein Isoforms/genetics , Protein Isoforms/immunology
15.
FASEB J ; 25(6): 2082-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21385991

ABSTRACT

Complement component C5a and ATP are potent effectors of microglial movement and are increased in diverse neurodegenerative diseases and at sites of injury. Apolipoprotein E (apoE) influences microglial function, and different human apoE isoforms confer variable risk for development of neurodegenerative disorders, especially Alzheimer's disease. The purpose of this investigation was to test the hypothesis that mouse apoE and human apoE isoforms influence microglial migration. Using primary wild-type and apoE-deficient microglia, we show that C5a- and ATP-stimulated chemotaxis are largely apoE-dependent processes with different molecular bases. Although the C5a-dependent chemotaxis of wild-type microglia was completely blocked by receptor-associated protein (RAP), suggesting apoE receptor involvement, ATP-stimulated migration was unaffected by RAP but was associated with differential ERK phosphorylation. Studies using primary microglia derived from targeted replacement mice "humanized" for the coding exons (protein isoform) of human ε2 (apoE2), ε3 (apoE3), or ε4 (apoE4) allele of APOE revealed that primary mouse microglia expressing apoE4 or apoE2 exhibited significantly reduced C5a- and ATP-stimulated migration compared with microglia expressing human apoE3. This study, for the first time, demonstrates apoE dependence and apoE isoform-specific modulation of microglial migration in response to distinct chemotactic stimuli commonly associated with neurodegenerative disease.


Subject(s)
Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cell Movement/physiology , Microglia/cytology , Microglia/physiology , Animals , Cells, Cultured , Chemotaxis , Gene Expression Regulation/physiology , Genotype , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Protein Isoforms
16.
Glia ; 59(4): 569-76, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21319223

ABSTRACT

Activation of innate immunity via toll-like receptors (TLRs) is associated with neurodegenerative diseases, and some effectors, like tumor necrosis factor alpha (TNFα) and interleukin 6 (IL-6), directly contribute to neurodegeneration. We tested the hypothesis that prostaglandin (PG) E(2) receptor subtype 1 (EP1) was necessary for the induction of microglial cytokines following the activation of innate immunity. Primary murine microglia had cytokine secretion by activators of TLR3 > TLR9 > TLR4 > TLR2. TLR3 activation induced early expression of cyclooxygenase 2 (COX2) and delayed expression of membranous PGE synthase and secretion of PGE(2) . Nonselective and COX2-selective inhibitors blocked TLR3 induction of TNFα and IL-6. Moreover, of the nine of twenty cytokines and chemokines induced by TLR3 activation, only TNFα and IL-6 were significantly dependent on EP1 signaling as determined using microglia from mice homozygous deficient for EP1 gene or wild-type microglia coincubated with an EP1 antagonist. These results were confirmed by blocking intracellular Ca(2+) release with 2-aminoethoxy-diphenyl borate or Xestospongin C, inhibitors of IP3 receptors. Our results show that suppression of microglial EP1 signaling achieves much of the desired effect of COX inhibitors by selectively blocking TLR3-induced microglial secretion of two major effectors of paracrine neuron damage. In combination with the ability of EP1 suppression to ameliorate excitotoxicity, these data point to blockade of EP1 as an attractive candidate therapeutic for neurodegenerative diseases.


Subject(s)
Interleukin-6/metabolism , Microglia/metabolism , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Toll-Like Receptor 3/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Immunity, Innate/physiology , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
17.
Am J Pathol ; 177(1): 346-54, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20522650

ABSTRACT

A complex therapeutic challenge for Alzheimer's disease (AD) is minimizing deleterious aspects of microglial activation while maximizing beneficial actions, including phagocytosis/clearance of amyloid beta (Abeta) peptides. One potential target is selective suppression of microglial prostaglandin E(2) receptor subtype 2 (EP2) function, which influences microglial phagocytosis and elaboration of neurotoxic cytokines. To test this hypothesis, we transplanted bone marrow cells derived from wild-type mice or mice homozygous deficient for EP2 (EP2(-/-)) into lethally irradiated 5-month-old wild-type or APPswe-PS1DeltaE9 double transgenic AD mouse model recipients. We found that cerebral engraftment by bone marrow transplant (BMT)-derived wild-type or EP2(-/-) microglia was more efficient in APPswe-PS1DeltaE9 than in wild-type mice, and APPswe-PS1DeltaE9 mice that received EP2(-/-) BMT had increased cortical microglia compared with APPswe-PS1DeltaE9 mice that received wild-type BMT. We found that myeloablative irradiation followed by bone marrow transplant-derived microglia engraftment, rather than cranial irradiation or BMT alone, was responsible for the approximate one-third reduction in both Abeta plaques and potentially more neurotoxic soluble Abeta species. An additional 25% reduction in cerebral cortical Abeta burden was achieved in mice that received EP2(-/-) BMT compared with mice that received wild-type BMT. Our results provide a foundation for an adult stem cell-based therapy to suppress soluble Abeta peptide and plaque accumulation in the cerebrum of patients with AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cerebral Cortex/pathology , Mice, Transgenic , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Animals , Bone Marrow Transplantation/methods , Cerebral Cortex/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Receptors, Prostaglandin E, EP2 Subtype/genetics
18.
J Cell Biochem ; 110(4): 935-47, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20564193

ABSTRACT

Vascular calcification is a major risk factor for cardiovascular morbidity and mortality. Smooth muscle cells (SMCs) may play an important role in vascular cartilaginous metaplasia and calcification via reprogramming to the osteochondrogenic state. To study whether SM lineage reprogramming and thus matrix calcification is reversible and what the necessary regulatory factors are to reverse this process, we used cells isolated from calcifying arterial medias of 4-week-old matrix Gla protein knockout mice (MGP-/-SMCs). We found that vascular cells with an osteochondrogenic phenotype regained SMC properties (positive for SM22alpha and SM alpha-actin) and down-regulated osteochondrogenic gene expression (Runx2/Cbfa1 and osteopontin) upon culture in medium that favors SMC differentiation. Over time, the MGP-/- SMCs no longer expressed osteochondrogenic proteins and became indistinguishable from wild-type SMCs. Moreover, phenotypic switch of the restored SMCs to the osteochondrogenic state was re-induced by the pro-calcific factor, inorganic phosphate. Finally, loss- and gain-of-function studies of myocardin, a SM-specific transcription co-activator, and Runx2/Cbfa1, an osteochondrogenic transcription factor, revealed that upregulation of Runx2/Cbfa1, but not loss of myocardin, played a critical role in phosphate-induced SMC lineage reprogramming and calcification. These results are the first to demonstrate reversibility of vascular SMCs in the osteochondrogenic state in response to local environmental cues, and that myocardin-enforced SMC lineage allocation was not sufficient to block vascular calcification. On the other hand, Runx2/Cbfa1 was found to be a decisive factor identified in the process.


Subject(s)
Chondrogenesis , Core Binding Factor Alpha 1 Subunit/physiology , Muscle, Smooth/cytology , Nuclear Proteins/physiology , Osteogenesis , Trans-Activators/physiology , Animals , Base Sequence , Blotting, Western , Calcium/metabolism , Cell Lineage , Cells, Cultured , DNA Primers , Mice , Mice, Knockout , Muscle, Smooth/metabolism , RNA Interference
19.
Vascul Pharmacol ; 52(1-2): 70-6, 2010.
Article in English | MEDLINE | ID: mdl-19962453

ABSTRACT

AIMS: Internal mammary (IMA) and radial artery (RA) have different incidence of vasospasm and long-term patency rates in arterial grafting. We compared the vasoreactivity of human urotensin II (hU-II) and its receptor with mechanism investigations in IMA and RA. METHODS: IMA and RA taken from patients undergoing coronary bypass surgery were studied in organ baths. Urotensin receptor expression was determined by RT-PCR. RESULTS: hU-II contracted IMA with pD(2) of 8.57+/-0.41 and 45.4+/-9.1% E(max) of contraction to 100 mM KCl, whereas caused less contractile responses in RA (pD(2):8.30+/-0.79, E(max):20.4+/-4.8%, p<0.05). Nifedipine inhibited hU-II-contraction in IMA. In U(46619)-precontraction, hU-II elicited comparable relaxation in IMA (pD(2):8.39+/-0.43, E(max):56.1+/-4.0%) and RA (pD(2):9.03+/-0.46, E(max):65.2+/-7.1%). The relaxation was abolished by endothelium denudation and by indomethacin, oxadiazoloquinoxalinone or N(omega)-nitro-L-arginine, oxyhemoglobin, and Ca2+-activated K+ channel (K(Ca)) blockers. Urotensin receptor mRNA was detected in both arteries. CONCLUSIONS: hU-II is an important spasmogen in arterial grafts with receptors expressed in IMA and RA. hU-II elicits stronger contraction in IMA than in RA and a moderate endothelium-dependent relaxation attributable to nitric oxide, prostacyclin, and endothelium-derived hyperpolarizing factor with involvement of K(Ca) activation. The relaxant response of endothelium-intact IMA and RA to hU-II demonstrates the importance of preservation of endothelium in these grafts.


Subject(s)
Coronary Artery Bypass , Mammary Arteries/drug effects , Peptide Hormones/pharmacology , Radial Artery/drug effects , Vasodilation/drug effects , Aged , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Male , Mammary Arteries/metabolism , Middle Aged , Peptide Hormones/metabolism , Radial Artery/metabolism , Receptors, G-Protein-Coupled/metabolism , Vasodilation/physiology
20.
Arterioscler Thromb Vasc Biol ; 30(2): 321-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19948844

ABSTRACT

OBJECTIVE: Vascular calcification is highly correlated with morbidity and mortality, and it is often associated with inflammation. Vitamin D may regulate vascular calcification and has been associated with cardiovascular survival benefits. METHODS AND RESULTS: We developed a macrophage/smooth muscle cell (SMC) coculture system and examined the effects of vitamin D receptor activators (VDRA), calcitriol and paricalcitol, on SMC matrix calcification. We found that treatment of SMC alone with VDRA had little effect on phosphate-induced SMC calcification in vitro. However, coculture with macrophages promoted SMC calcification, and this was strikingly inhibited by VDRA treatment. Several VDRA-induced genes, including bone morphogenetic protein-2 (BMP2), tumor necrosis factor-alpha, and osteopontin, were identified as candidate paracrine factors for the protective effect of VDRA. Of these, osteopontin was further investigated and found to contribute significantly to the inhibitory actions of VDRA on calcification in macrophage/SMC cocultures. CONCLUSIONS: The ability of VDRA to direct a switch in the paracrine phenotype of macrophages from procalcific to anticalcific may contribute to their observed cardiovascular survival benefits.


Subject(s)
Calcinosis/prevention & control , Calcitriol/pharmacology , Ergocalciferols/pharmacology , Macrophages/drug effects , Muscle, Smooth, Vascular/drug effects , Osteopontin/metabolism , Paracrine Communication/drug effects , Receptors, Calcitriol/agonists , Animals , Bone Morphogenetic Protein 2/genetics , Calcinosis/metabolism , Calcium/metabolism , Cells, Cultured , Coculture Techniques , Gene Expression Regulation/drug effects , Humans , Macrophages/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Osteopontin/genetics , Phenotype , RNA Interference , RNA, Messenger/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Time Factors , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...