Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Signal ; 8(388): ra77, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26243191

ABSTRACT

Most patients with pancreatic ductal adenocarcinoma (PDA) present with metastatic disease at the time of diagnosis or will recur with metastases after surgical treatment. Semaphorin-plexin signaling mediates the migration of neuronal axons during development and of blood vessels during angiogenesis. The expression of the gene encoding semaphorin 3D (Sema3D) is increased in PDA tumors, and the presence of antibodies against the pleiotropic protein annexin A2 (AnxA2) in the sera of some patients after surgical resection of PDA is associated with longer recurrence-free survival. By knocking out AnxA2 in a transgenic mouse model of PDA (KPC) that recapitulates the progression of human PDA from premalignancy to metastatic disease, we found that AnxA2 promoted metastases in vivo. The expression of AnxA2 promoted the secretion of Sema3D from PDA cells, which coimmunoprecipitated with the co-receptor plexin D1 (PlxnD1) on PDA cells. Mouse PDA cells in which SEMA3D was knocked down or ANXA2-null PDA cells exhibited decreased invasive and metastatic potential in culture and in mice. However, restoring Sema3D in AnxA2-null cells did not entirely rescue metastatic behavior in culture and in vivo, suggesting that AnxA2 mediates additional prometastatic mechanisms. Patients with primary PDA tumors that have abundant Sema3D have widely metastatic disease and decreased survival compared to patients with tumors that have relatively low Sema3D abundance. Thus, AnxA2 and Sema3D may be new therapeutic targets and prognostic markers of metastatic PDA.


Subject(s)
Annexin A2/genetics , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Semaphorins/genetics , Signal Transduction/genetics , Animals , Annexin A2/metabolism , Autocrine Communication/genetics , Blotting, Western , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence/classification , Neoplasm Metastasis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Semaphorins/metabolism , Survival Analysis , Tumor Cells, Cultured , Pancreatic Neoplasms
2.
Am J Hum Genet ; 96(4): 581-96, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25839327

ABSTRACT

Innervation of the gut is segmentally lost in Hirschsprung disease (HSCR), a consequence of cell-autonomous and non-autonomous defects in enteric neuronal cell differentiation, proliferation, migration, or survival. Rare, high-penetrance coding variants and common, low-penetrance non-coding variants in 13 genes are known to underlie HSCR risk, with the most frequent variants in the ret proto-oncogene (RET). We used a genome-wide association (220 trios) and replication (429 trios) study to reveal a second non-coding variant distal to RET and a non-coding allele on chromosome 7 within the class 3 Semaphorin gene cluster. Analysis in Ret wild-type and Ret-null mice demonstrates specific expression of Sema3a, Sema3c, and Sema3d in the enteric nervous system (ENS). In zebrafish embryos, sema3 knockdowns show reduction of migratory ENS precursors with complete ablation under conjoint ret loss of function. Seven candidate receptors of Sema3 proteins are also expressed within the mouse ENS and their expression is also lost in the ENS of Ret-null embryos. Sequencing of SEMA3A, SEMA3C, and SEMA3D in 254 HSCR-affected subjects followed by in silico protein structure modeling and functional analyses identified five disease-associated alleles with loss-of-function defects in semaphorin dimerization and binding to their cognate neuropilin and plexin receptors. Thus, semaphorin 3C/3D signaling is an evolutionarily conserved regulator of ENS development whose dys-regulation is a cause of enteric aganglionosis.


Subject(s)
Epistasis, Genetic/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation , Hirschsprung Disease/genetics , Proto-Oncogene Proteins c-ret/genetics , Semaphorins/genetics , Animals , Base Sequence , Genome-Wide Association Study , Mice , Molecular Sequence Data , Semaphorins/deficiency , Semaphorins/metabolism , Sequence Analysis, DNA
3.
Neurobiol Aging ; 33(1): 197.e21-32, 2012 Jan.
Article in English | MEDLINE | ID: mdl-20696495

ABSTRACT

Considerable evidence points to important roles for inflammation in Alzheimer's disease (AD) pathophysiology. Epidemiological studies have suggested that long-term nonsteroidal anti-inflammatory drug (NSAID) therapy reduces the risk for Alzheimer's disease; however, the mechanism remains unknown. We report that a 9-month treatment of aged R1.40 mice resulted in 90% decrease in plaque burden and a similar reduction in microglial activation. Ibuprofen treatment reduced levels of lipid peroxidation, tyrosine nitration, and protein oxidation, demonstrating a dramatic effect on oxidative damage in vivo. Fibrillar ß-amyloid (Aß) stimulation has previously been demonstrated to induce the assembly and activation of the microglial nicotinamide adenine dinucleotide phosphate (NADPH) oxidase leading to superoxide production through a tyrosine kinase-based signaling cascade. Ibuprofen treatment of microglia or monocytes with racemic or S-ibuprofen inhibited Aß-stimulated Vav tyrosine phosphorylation, NADPH oxidase assembly, and superoxide production. Interestingly, Aß-stimulated Vav phosphorylation was not inhibited by COX inhibitors. These findings suggest that ibuprofen acts independently of cyclooxygenase COX inhibition to disrupt signaling cascades leading to microglial NADPH oxidase (NOX2) activation, preventing oxidative damage and enhancing plaque clearance in the brain.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Ibuprofen/pharmacology , NADPH Oxidases/antagonists & inhibitors , Amyloid beta-Peptides , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cells, Cultured , Enzyme Activation/drug effects , Ibuprofen/therapeutic use , Male , Mice , Mice, Transgenic , Microglia/enzymology , Microglia/metabolism , Microglia/pathology , Monocytes/metabolism , NADPH Oxidases/physiology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Plaque, Amyloid , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-vav , Signal Transduction/physiology
4.
J Neurosci ; 29(13): 4252-62, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19339619

ABSTRACT

Alzheimer's disease is characterized by the progressive deposition of beta-amyloid (Abeta) within the brain parenchyma and its subsequent accumulation into senile plaques. Pathogenesis of the disease is associated with perturbations in Abeta homeostasis and the inefficient clearance of these soluble and insoluble peptides from the brain. Microglia have been reported to mediate the clearance of fibrillar Abeta (fAbeta) through receptor-mediated phagocytosis; however, their participation in clearance of soluble Abeta peptides (sAbeta) is largely unknown. We report that microglia internalize sAbeta from the extracellular milieu through a nonsaturable, fluid phase macropinocytic mechanism that is distinct from phagocytosis and receptor-mediated endocytosis both in vitro and in vivo. The uptake of sAbeta is dependent on both actin and tubulin dynamics and does not involve clathrin assembly, coated vesicles or membrane cholesterol. Upon internalization, fluorescently labeled sAbeta colocalizes to pinocytic vesicles. Microglia rapidly traffic these soluble peptides into late endolysosomal compartments where they are subject to degradation. Additionally, we demonstrate that the uptake of sAbeta and fAbeta occurs largely through distinct mechanisms and upon internalization are segregated into separate subcellular vesicular compartments. Significantly, we found that upon proteolytic degradation of fluorescently labeled sAbeta, the fluorescent chromophore is retained by the microglial cell. These studies identify an important mechanism through which microglial cells participate in the maintenance of Abeta homeostasis, through their capacity to constitutively clear sAbeta peptides from the brain.


Subject(s)
Amyloid beta-Peptides/metabolism , Microglia/physiology , Peptide Fragments/metabolism , Pinocytosis/physiology , Amyloid beta-Peptides/administration & dosage , Animals , Animals, Newborn , Brain/cytology , Cells, Cultured , Coculture Techniques , Cytochalasin D/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Flow Cytometry/methods , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Neurons/drug effects , Neurons/physiology , Nocodazole/pharmacology , Peptide Fragments/administration & dosage , Phagocytosis/drug effects , Phagocytosis/physiology , Pinocytosis/genetics , Protein Transport/drug effects , Spectrum Analysis , Tubulin Modulators/pharmacology
5.
Neurotherapeutics ; 5(3): 481-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18625459

ABSTRACT

Alzheimer's disease (AD) is characterized by the deposition of beta-amyloid within the brain parenchyma and is accompanied by the impairment of neuronal metabolism and function, leading to extensive neuronal loss. The disease involves the perturbation of synaptic function, energy, and lipid metabolism. The development of amyloid plaques results in the induction of a microglial-mediated inflammatory response. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) is a ligand-activated transcription factor whose biological actions are to regulate glucose and lipid metabolism and suppress inflammatory gene expression. Thus, agonists of this receptor represent an attractive therapeutic target for AD. There is now an extensive body of evidence that has demonstrated the efficacy of PPARgamma agonists in ameliorating disease-related pathology and improved learning and memory in animal models of AD. Recent clinical trials of the PPARgamma agonist rosiglitazone have shown significant improvement in memory and cognition in AD patients. Thus, PPARgamma represents an important new therapeutic target in treating AD.


Subject(s)
Alzheimer Disease/drug therapy , Antipsychotic Agents/therapeutic use , PPAR gamma/agonists , PPAR gamma/therapeutic use , Alzheimer Disease/metabolism , Animals , Humans
6.
Neuron ; 58(5): 681-93, 2008 Jun 12.
Article in English | MEDLINE | ID: mdl-18549781

ABSTRACT

Apolipoprotein E is associated with age-related risk for Alzheimer's disease and plays critical roles in Abeta homeostasis. We report that ApoE plays a role in facilitating the proteolytic clearance of soluble Abeta from the brain. The endolytic degradation of Abeta peptides within microglia by neprilysin and related enzymes is dramatically enhanced by ApoE. Similarly, Abeta degradation extracellularly by insulin-degrading enzyme is facilitated by ApoE. The capacity of ApoE to promote Abeta degradation is dependent upon the ApoE isoform and its lipidation status. The enhanced expression of lipidated ApoE, through the activation of liver X receptors, stimulates Abeta degradation. Indeed, aged Tg2576 mice treated with the LXR agonist GW3965 exhibited a dramatic reduction in brain Abeta load. GW3965 treatment also reversed contextual memory deficits. These data demonstrate a mechanism through which ApoE facilitates the clearance of Abeta from the brain and suggest that LXR agonists may represent a novel therapy for AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoproteins E/pharmacology , Microglia/drug effects , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Aging , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Animals, Newborn , Behavior, Animal/drug effects , Benzoates/pharmacology , Benzylamines/pharmacology , Brain/pathology , Cells, Cultured , DNA-Binding Proteins/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Liver X Receptors , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Orphan Nuclear Receptors , Peptide Fragments/metabolism , Plaque, Amyloid/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Time Factors
7.
J Neuroimmune Pharmacol ; 3(2): 130-40, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18247125

ABSTRACT

Removal of apoptotic cells is an essential process for normal development and tissue maintenance. Importantly, apoptotic cells stimulate their phagocytosis by macrophages while actively suppressing inflammatory responses. Growth arrest specific gene 6 (Gas6) is involved in this process, bridging phosphatidylserine residues on the surface of apoptotic cells to the Axl/Mer family of tyrosine kinases which stimulate phagocytosis. Animals with mutations or loss of these receptors exhibit phenotypes reflective of impaired phagocytosis and a hyperactive immune response. We report that Gas6 induces phagocytosis in microglia through a novel non-classical phagocytic mechanism. Gas6 stimulates a type-II-related phagocytic response, but requires Vav phosphorylation and Rac activation, distinguishing it from the classical type II mechanism. Importantly, Gas6 suppressed lipopolysaccharide-induced expression of the inflammatory molecules IL-1beta and iNOS. Gas6 inhibited iNOS expression through suppression of promoter activity. The present data provide direct evidence for the role of Gas6 receptors in mediating an anti-inflammatory response to ligands found on apoptotic cells with the simultaneous stimulation of phagocytosis. These data provide a mechanistic explanation for the phenotype observed in animals lacking Axl/Mer receptors.


Subject(s)
Inflammation/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Microglia/drug effects , Oncogene Proteins/physiology , Phagocytosis/drug effects , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain/metabolism , Cell Line/drug effects , Cell Line/enzymology , Cell Line/physiology , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Gene Expression Regulation/drug effects , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Lipopolysaccharides/toxicity , Mice , Microglia/enzymology , Microglia/physiology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Phagocytosis/physiology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-vav/metabolism , Recombinant Fusion Proteins/biosynthesis , Transcription, Genetic/drug effects , c-Mer Tyrosine Kinase , rac GTP-Binding Proteins/metabolism , Axl Receptor Tyrosine Kinase
8.
CNS Drugs ; 22(1): 1-14, 2008.
Article in English | MEDLINE | ID: mdl-18072811

ABSTRACT

Alzheimer's disease is a complex neurodegenerative disorder, with aging, genetic and environmental factors contributing to its development and progression. The complexity of Alzheimer's disease presents substantial challenges for the development of new therapeutic agents. Alzheimer's disease is typified by pathological depositions of beta-amyloid peptides and neurofibrillary tangles within the diseased brain. It has also been demonstrated to be associated with a significant microglia-mediated inflammatory component, dysregulated lipid homeostasis and regional deficits in glucose metabolism within the brain. The peroxisome proliferator-activated receptor-gamma (PPARgamma) is a prototypical ligand-activated nuclear receptor that coordinates lipid, glucose and energy metabolism, and is found in elevated levels in the brains of individuals with Alzheimer's disease. A recently appreciated physiological function of this type of receptor is its ability to modulate inflammatory responses. In animal models of Alzheimer's disease, PPARgamma agonist treatment results in the reduction of amyloid plaque burden, reduced inflammation and reversal of disease-related behavioural impairment. In a recent phase II clinical trial, the use of the PPARgamma agonist rosiglitazone was associated with improved cognition and memory in patients with mild to moderate Alzheimer's disease. Thus, PPARgamma may act to modulate multiple pathophysiological mechanisms that contribute to Alzheimer's disease, and represents an attractive therapeutic target for the treatment of the disease.


Subject(s)
Alzheimer Disease/physiopathology , PPAR gamma/physiology , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Animals , Gene Expression/drug effects , Gene Expression/physiology , Humans , Inflammation/pathology , Insulin Resistance/physiology , PPAR gamma/agonists , PPAR gamma/genetics
9.
Proc Natl Acad Sci U S A ; 104(25): 10601-6, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17563384

ABSTRACT

Alzheimer's disease (AD) is an age-dependent neurodegenerative disease that causes progressive cognitive impairment. The initiation and progression of AD has been linked to cholesterol metabolism and inflammation, processes that can be modulated by liver x receptors (LXRs). We show here that endogenous LXR signaling impacts the development of AD-related pathology. Genetic loss of either Lxralpha or Lxrbeta in APP/PS1 transgenic mice results in increased amyloid plaque load. LXRs regulate basal and inducible expression of key cholesterol homeostatic genes in the brain and act as potent inhibitors of inflammatory gene expression. Ligand activation of LXRs attenuates the inflammatory response of primary mixed glial cultures to fibrillar amyloid beta peptide (fAbeta) in a receptor-dependent manner. Furthermore, LXRs promote the capacity of microglia to maintain fAbeta-stimulated phagocytosis in the setting of inflammation. These results identify endogenous LXR signaling as an important determinant of AD pathogenesis in mice. We propose that LXRs may be tractable targets for the treatment of AD due to their ability to modulate both lipid metabolic and inflammatory gene expression in the brain.


Subject(s)
Alzheimer Disease/pathology , DNA-Binding Proteins/physiology , Inflammation/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Amyloid beta-Peptides/pharmacology , Animals , Brain/metabolism , Brain/pathology , Cells, Cultured , Cholesterol/genetics , Cholesterol/metabolism , DNA-Binding Proteins/genetics , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation/physiology , Immunohistochemistry , Inflammation/pathology , Lipid Metabolism , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/cytology , Microglia/drug effects , Microglia/physiology , Orphan Nuclear Receptors , Phagocytosis/drug effects , Phagocytosis/physiology , Plaque, Amyloid/pathology , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction
10.
Neurochem Int ; 49(2): 136-44, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16766086

ABSTRACT

Diseases of the central nervous system present a challenge for the development of new therapeutic agents. Nuclear receptors are ligand-activated transcription factors that have proven to be valuable targets for development of new drugs owing to their ability to directly regulate gene expression. The nuclear receptor, peroxisome proliferator-activated receptor gamma (PPARgamma), has been investigated for its action in ameliorating the development and progression of a number of CNS diseases. PPARgamma agonists exhibit potent anti-inflammatory effects and appear to have direct neuroprotective actions. PPARgamma agonists have been shown to be efficacious in animal models of Alzheimer's disease, stroke, multiple sclerosis, Parkinson's disease and amyotrophic lateral sclerosis. The availability of FDA-approved agonists of this receptor will facilitate the rapid translation of these findings into clinical trials for a number of CNS diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain Diseases/drug therapy , Brain/drug effects , Encephalitis/drug therapy , Neuroprotective Agents/pharmacology , PPAR gamma/agonists , Animals , Anti-Inflammatory Agents/therapeutic use , Brain/immunology , Brain/physiopathology , Brain Diseases/immunology , Brain Diseases/physiopathology , Disease Models, Animal , Encephalitis/genetics , Encephalitis/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Neuroprotective Agents/therapeutic use , PPAR gamma/immunology , PPAR gamma/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Transcriptional Activation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...