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










Publication year range
1.
JTO Clin Res Rep ; 4(7): 100527, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37521368

ABSTRACT

Introduction: Relapse is common after resection of lung adenocarcinoma (LUAD). Features of the tumor microenvironment (TME) which influence postsurgical survival outcomes are poorly characterized. Here, we analyzed the TME of more than 1500 LUAD specimens to identify the relationship between B-cell infiltration and prognosis. Methods: Whole exome sequencing and bulk RNA sequencing were performed on LUADs and adjacent normal lung tissue. Relapse-free survival and overall survival (OS) were retrospectively correlated with characteristics of the tumor and TME in three data sets. Results: High B-cell content (defined as >10% B cells) was associated with improved OS in both a The Cancer Genome Atlas-resected LUAD data set (p = 0.01) and a separate institutional stage II LUAD data set (p = 0.04, median not reached versus 89.5 mo). A validation cohort consisting of pooled microarray data representing more than 1400 resected stage I to III LUADs confirmed the association between greater B-cell abundance, specifically higher B-cell expression, and longer postsurgical survival (median OS 90 versus 71 mo, p < 0.01). Relapse-free survival was longer for patients with adenocarcinomas with high B-cell content across data sets, but it did not reach statistical significance. Subcategorization of B-cell subsets indicated that high naive B-cell content was most predictive of survival. There was no correlation between programmed death-ligand 1 expression, lymphoid aggregates, or overall immune infiltrate density and survival outcomes across the cohorts. Conclusions: The growing adjuvant immunotherapy repertoire has increased the urgency for identifying prognostic and predictive biomarkers. Comprehensive profiling of more than 1500 LUADs suggests that high tumor-infiltrating B-cell content is a favorable prognostic marker.

2.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37364933

ABSTRACT

BACKGROUND: PD-1 checkpoint blockade therapy (CBT) has greatly benefited patients with select solid tumors and lymphomas but has limited efficacy against diffuse large B-cell lymphoma (DLBCL). Because numerous inhibitory checkpoint receptors have been implicated in driving tumor-specific T cell dysfunction, we hypothesized that combinatorial CBT would enhance the activity of anti-PD-1-based therapy in DLBCL. T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) is a coinhibitory receptor expressed on dysfunctional tumor-infiltrating T cells, and TIGIT blockade has demonstrated encouraging activity in combination with PD-1 blockade in murine tumor models and in clinical studies. However, the degree to which TIGIT mediates T cell dysfunction in DLBCL has not been fully explored. RESULTS: Here, we demonstrate that TIGIT is broadly expressed on lymphoma-infiltrating T cells (LITs) across a variety of human lymphomas and is frequently coexpressed with PD-1. TIGIT expression is particularly common on LITs in DLBCL, where TIGIT+ LITs often form distinct cellular communities and exhibit significant contact with malignant B cells. TIGIT+/PD-1+ LITs from human DLBCL and murine lymphomas exhibit hypofunctional cytokine production on ex vivo restimulation. In mice with established, syngeneic A20 B-cell lymphomas, TIGIT or PD-1 mono-blockade leads to modest delays in tumor outgrowth, whereas PD-1 and TIGIT co-blockade results in complete rejection of A20 lymphomas in most mice and significantly prolongs survival compared with mice treated with monoblockade therapy. CONCLUSIONS: These results provide rationale for clinical investigation of TIGIT and PD-1 blockade in lymphomas, including DLBCL.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Programmed Cell Death 1 Receptor , Humans , Animals , Mice , Receptors, Immunologic/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology
3.
J Biomark ; 2016: 1274603, 2016.
Article in English | MEDLINE | ID: mdl-27247826

ABSTRACT

Objective. Use of tyramide signal amplification (TSA) to detect autophagy biomarkers in formalin fixed and paraffin embedded (FFPE) xenograft tissue. Materials and Methods. Autophagy marker regulation was studied in xenograft tissues using Amp HQ IHC and standard IHC methods. Results. The data demonstrate the feasibility of using high sensitivity TSA IHC assays to measure low abundant autophagy markers in FFPE xenograft tissue.

4.
J Huntingtons Dis ; 2(1): 101-5, 2013.
Article in English | MEDLINE | ID: mdl-25063433

ABSTRACT

The coactivator PGC1α plays a role in the transcriptional regulation of energy metabolism and its deficiency has been implicated in abnormalities of myelination and oligodendrocyte differentiation in Huntington's disease (HD). In an effort to activate PGC1α in oligodendrocytes, we found that overexpression of SIRT1 or treatment with resveratrol (RSV) and SRT1720 upregulated PGC1α expression and increased oligodendrocyte differentiation. Interestingly, in oligodendrocytes expressing mutant huntingtin, SRT1720 but not RSV was able to restore PGC1α expression and the differentiation. These results suggest that pharmacological activation of PGC1α in oligodendrocytes by SRT1720 may provide a therapeutic opportunity for correction of deficient myelination in HD.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Huntington Disease/metabolism , Oligodendroglia/metabolism , Transcription Factors/biosynthesis , Animals , Cell Differentiation/drug effects , Cells, Cultured , Mice , Mutation , Oligodendroglia/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Resveratrol , Serotonin Plasma Membrane Transport Proteins/genetics , Sirtuin 1/metabolism , Stilbenes/pharmacology , Up-Regulation
5.
J Neurosci ; 31(26): 9544-53, 2011 Jun 29.
Article in English | MEDLINE | ID: mdl-21715619

ABSTRACT

The peroxisome-proliferator-activated receptor gamma coactivator 1 α (PGC1α) has been implicated in the pathogenesis of several neurodegenerative disorders, including Huntington's disease (HD). Recent data demonstrating white matter abnormalities in PGC1α knock-out (KO) mice prompted us to examine the role of PGC1α in CNS myelination and its relevance to HD pathogenesis. We found deficient postnatal myelination in the striatum of PGC1α KO mice, accompanied by a decrease in myelin basic protein (MBP). In addition, brain cholesterol, its precursors, and the rate-limiting enzymes for cholesterol synthesis, HMG CoA synthase (HMGCS1) and HMG CoA reductase (HMGCR), were also reduced in PGC1α KO mice. Moreover, knockdown of PGC1α in oligodendrocytes by lentiviral shRNA led to a decrease in MBP, HMGCS1, and Hmgcr mRNAs. Chromatin immunoprecipitations revealed the recruitment of PGC1α to MBP promoter in mouse brain, and PGC1α over-expression increased MBP and SREBP-2 promoter activity, suggesting that PGC1α regulates MBP and cholesterol synthesis at the transcriptional level. Importantly, expression of mutant huntingtin (Htt) in primary oligodendrocytes resulted in decreased expression of PGC1α and its targets HmgcS1, Hmgcr, and MBP. Decreased expression of MBP and deficient myelination were found postnatally and in adult R6/2 mouse model of HD. Diffusion tensor imaging detected white matter abnormalities in the corpus callosum of R6/2 mice, and electron microscopy revealed thinner myelin sheaths and increased myelin periodicity in BACHD [bacterial artificial chromosome (BAC)-mediated transgenic model for Huntington's disease] mice expressing full-length mutant Htt. Together, these data suggest that PGC1α plays a role in postnatal myelination and that deficient PGC1α activity in oligodendrocytes may contribute to abnormal myelination in HD.


Subject(s)
Demyelinating Diseases/metabolism , Huntington Disease/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Transcription Factors/metabolism , Analysis of Variance , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Chromatin Immunoprecipitation , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Diffusion Tensor Imaging , Disease Models, Animal , Huntington Disease/genetics , Huntington Disease/pathology , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Synthase/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/pathology , Oligodendroglia/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
6.
ACS Chem Biol ; 6(6): 540-6, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21370928

ABSTRACT

Sirtuin 2 (SIRT2) deacetylase-dependent inhibition mediates neuroprotective reduction of cholesterol biosynthesis in an in vitro Huntington's disease model. This study sought to identify the first brain-permeable SIRT2 inhibitor and to characterize its cholesterol-reducing properties in neuronal models. Using biochemical sirtuin deacetylation assays, we screened a brain-permeable in silico compound library, yielding 3-(1-azepanylsulfonyl)-N-(3-bromphenyl)benzamide as the most potent and selective SIRT2 inhibitor. Pharmacokinetic studies demonstrated brain-permeability but limited metabolic stability of the selected candidate. In accordance with previous observations, this SIRT2 inhibitor stimulated cytoplasmic retention of sterol regulatory element binding protein-2 and subsequent transcriptional downregulation of cholesterol biosynthesis genes, resulting in reduced total cholesterol in primary striatal neurons. Furthermore, the identified inhibitor reduced cholesterol in cultured naïve neuronal cells and brain slices from wild-type mice. The outcome of this study provides a clear opportunity for lead optimization and drug development, targeting metabolic dysfunctions in CNS disorders where abnormal cholesterol homeostasis is implicated.


Subject(s)
Brain/metabolism , Cholesterol/biosynthesis , Enzyme Inhibitors/pharmacology , Neurons/drug effects , Neurons/metabolism , Sirtuin 2/antagonists & inhibitors , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Mice , Models, Neurological , Molecular Structure , Neurons/enzymology , Permeability , Sirtuin 2/metabolism , Small Molecule Libraries , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Proc Natl Acad Sci U S A ; 107(17): 7927-32, 2010 Apr 27.
Article in English | MEDLINE | ID: mdl-20378838

ABSTRACT

Huntington's disease (HD), an incurable neurodegenerative disorder, has a complex pathogenesis including protein aggregation and the dysregulation of neuronal transcription and metabolism. Here, we demonstrate that inhibition of sirtuin 2 (SIRT2) achieves neuroprotection in cellular and invertebrate models of HD. Genetic or pharmacologic inhibition of SIRT2 in a striatal neuron model of HD resulted in gene expression changes including significant down-regulation of RNAs responsible for sterol biosynthesis. Whereas mutant huntingtin fragments increased sterols in neuronal cells, SIRT2 inhibition reduced sterol levels via decreased nuclear trafficking of SREBP-2. Importantly, manipulation of sterol biosynthesis at the transcriptional level mimicked SIRT2 inhibition, demonstrating that the metabolic effects of SIRT2 inhibition are sufficient to diminish mutant huntingtin toxicity. These data identify SIRT2 inhibition as a promising avenue for HD therapy and elucidate a unique mechanism of SIRT2-inhibitor-mediated neuroprotection. Furthermore, the ascertainment of SIRT2's role in regulating cellular metabolism demonstrates a central function shared with other sirtuin proteins.


Subject(s)
Brain/metabolism , Gene Expression Regulation/drug effects , Huntington Disease/prevention & control , Neuroprotective Agents/pharmacology , Sirtuin 2/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 2/metabolism , Sterols/biosynthesis , Analysis of Variance , Animals , Blotting, Western , Caenorhabditis elegans , Drosophila , Gene Expression Profiling , Immunohistochemistry , Mice , Microscopy, Confocal
8.
Exp Neurol ; 215(1): 41-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18929563

ABSTRACT

Statins (inhibitors of HMG-CoA reductase) have shown promise in treating multiple sclerosis (MS). However, their effect on oligodendrocyte remyelination of demyelinated axons has not been clarified. Since developmental myelination shares many features with the remyelination process, we investigated the effect of lipophilic simvastatin on developmental myelination in organotypic cerebellar slice cultures (CSC). In this study, we first characterized developmental myelination in CSC from postnatal day (P)5 and P10 mice that express enhanced green fluorescence protein (eGFP) in oligodendrocyte-lineage cells. We then examined the effect of simvastatin on three developmental myelination stages: early myelination (P5 CSC, 2DIV), late myelination (P10 CSC, 2DIV) and full myelination (P10 CSC, 10DIV). We found that treatment with simvastatin (0.1 microM) for 6 days decreased the survival of Purkinje cells and oligodendrocytes drastically during the early myelination stage, while moderately during the late and full myelination stages. Oligodendrocytes are more resistant than Purkinje cells. The toxic effect of simvastatin could be rescued by the product of HMG-CoA reductase mevalonate but not low-density lipoprotein (LDL). Additionally, this toxic effect is independent of isoprenylation since farnesyl pyrophosphate (Fpp) but not geranylgeranyl pyrophosphate (GGpp) provided partial rescue. Our findings therefore suggest that inhibition of cholesterol synthesis is detrimental to neuronal tissue.


Subject(s)
Cerebellum/cytology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myelin Sheath/metabolism , Neurons/drug effects , Simvastatin/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Cell Death/drug effects , Dose-Response Relationship, Drug , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/metabolism , Mevalonic Acid/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Myelin Basic Protein/metabolism , Oligodendroglia/drug effects , Tissue Culture Techniques
9.
Curr Neurovasc Res ; 4(4): 274-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18045153

ABSTRACT

Cyclooxygenase-2 (COX-2) is up-regulated during ischemia. However, the role of COX-2 in neuronal injury is still unclear. In this study we tested whether neuronal overexpression of human COX-2 in a transgenic mouse model potentiates neuronal injury after global ischemic insult. Further, we tested whether the neuronal injury could be ameliorated by intra-ischemic mild hypothermia (33-34 degrees C) alone or in combination with diet treatment of rofecoxib, a COX-2 specific inhibitor. Global ischemia with intra-ischemic normothermia (36-37 degrees C) resulted in significantly higher neuronal damage in the CA1 region of hippocampus of transgenic mice than in wild type controls, confirming a deleterious role of COX-2 in ischemic neuronal damage. Hypothermia significantly reduced neuronal damage in both transgenic mice and wild type controls to the same extent, suggesting that the aggravating effect of COX-2 could be largely eliminated by hypothermia. When hypothermia was combined with rofecoxib treatment, neuronal damage was further reduced in response to global ischemia. The results suggest that COX-2 inhibition by prophylactic treatment with rofecoxib coupled with hypothermia at the time of acute stroke insult could be an effective therapeutic approach in early stages of stroke treatment in high risk patients.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/biosynthesis , Hypothermia/enzymology , Lactones/pharmacology , Neurons/physiology , Sulfones/pharmacology , Animals , Apoptosis/drug effects , Brain Ischemia/physiopathology , Carotid Artery, Common/physiology , Cerebrovascular Circulation/drug effects , Dinoprostone/metabolism , Humans , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism
10.
J Neuroinflammation ; 4: 18, 2007 Jul 16.
Article in English | MEDLINE | ID: mdl-17634127

ABSTRACT

BACKGROUND: Prostaglandin (PG) production is associated with inflammation, a major feature in multiple sclerosis (MS) that is characterized by the loss of myelinating oligodendrocytes in the CNS. While PGs have been shown to have relevance in MS, it has not been determined whether PGs have a direct effect on cells within the oligodendrocyte lineage. METHODS: Undifferentiated or differentiated mouse oligodendrocyte precursor (mOP) cells were treated with PGE2, PGF2alpha, PGD2 or 15-deoxy-Delta12,14-PGJ2 (15d-PGJ2). Cell growth and survival following treatment were examined using cytotoxicity assays and apoptosis criteria. The membrane receptors for PGD2 and the nuclear receptor peroxisome proliferator-activated receptor (PPAR)gamma, as well as reactive oxygen species (ROS) in the death mechanism were examined. RESULTS: PGE2 and PGF2alpha had minimal effects on the growth and survival of mOP cells. In contrast, PGD2 and 15d-PGJ2 induced apoptosis of undifferentiated mOP cells at relatively low micromolar concentrations. 15d-PGJ2 was less toxic to differentiated mOP cells. Apoptosis was independent of membrane receptors for PGD2 and the nuclear receptor PPARgamma. The cytotoxicity of 15d-PGJ2 was associated with the production of ROS and was inversely related to intracellular glutathione (GSH) levels. However, the cytotoxicity of 15d-PGJ2 was not decreased by the free radical scavengers ascorbic acid or alpha-tocopherol. CONCLUSION: Taken together, these results demonstrated that 15d-PGJ2 is toxic to early stage OP cells, suggesting that 15d-PGJ2 may represent a deleterious factor in the natural remyelination process in MS.


Subject(s)
Apoptosis/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Prostaglandin D2/analogs & derivatives , Stem Cells/cytology , Stem Cells/physiology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Mice , Oligodendroglia/drug effects , Prostaglandin D2/physiology , Prostaglandin D2/toxicity , Rats , Stem Cells/drug effects
11.
Neurobiol Aging ; 28(6): 824-30, 2007 Jun.
Article in English | MEDLINE | ID: mdl-16769157

ABSTRACT

In this study we report that the membrane-bound, but not cytosolic insulin degrading enzyme (IDE) protein concentration and IDE activity are significantly decreased in the hippocampal formation of cases affected by mild cognitive impairment (MCI) which are at high risk to develop Alzheimer's disease (AD), relative to normal neurological controls. Membrane-bound IDE protein concentrations and activity in the hippocampal formation continued to decrease during the conversion from MCI to mild-severe AD. This selective decrease in hippocampal membrane-bound, but not cytosolic, IDE concentration and activity was tissue specific since no changes in either membrane-bound or cytosolic IDE were found in the occipital cortex of the same cases examined. Most interestingly, the decreased hippocampal membrane-bound IDE protein activity negatively correlated with brain beta-amyloid (Abeta)X-42 content in MCI and in AD brain. The study tentatively suggests that interventions aimed at promoting membrane-bound IDE activities in the brain of MCI cases may help to prevent the onset and possibly the progression into AD through mechanisms involving the clearance of monomeric Abeta from the brain.


Subject(s)
Alzheimer Disease/pathology , Hippocampus/enzymology , Insulysin/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Analysis of Variance , Cell Membrane/enzymology , Cognition Disorders/complications , Cognition Disorders/pathology , Cytosol/enzymology , Entorhinal Cortex/metabolism , Female , Humans , Insulin/metabolism , Iodine Isotopes/metabolism , Male , Postmortem Changes
12.
J Neurosci Methods ; 157(2): 187-94, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-16720048

ABSTRACT

Oligodendrocyte precursor (OP) cells give rise to mature oligodendrocytes (OL), which are necessary for myelination of axons during CNS development and following damage to the myelin sheath that occurs in demyelinating diseases. To facilitate studies designed to understand OP maturation and OL function, we have developed OP cells that can be grown continuously, expanded, and differentiated into mature OLs. Cultures of late passage mOP cells grown in proliferation medium are highly pure early stage oligodendrocyte precursors where > 90% assume a characteristic bipolar morphology. Immunocytochemical analysis using antibodies that recognize progressive stages of OP maturation (A2B5, NG2, GD3 and O4) confirmed that mOP cells have a stable early stage OP cell phenotype. In addition, mOP cells can be induced to differentiate into mature forms of oligodendrocytes in vitro and in vivo, as characterized morphologically by the presence of multiple processes with secondary and tertiary branches, and by immunostaining and quantitative real-time PCR for the mature oligodendrocyte markers MBP, MAG, PLP, and MOBP. Finally, differentiation of mOP cells was accompanied by up-regulation of mRNA encoding Olig2 but not Olig1, which is consistent with previous findings showing that Olig2 is necessary for specification of oligodendrocytes. These new mOP cells should significantly benefit in vitro and in vivo studies on OP maturation and function.


Subject(s)
Cells, Cultured , Oligodendroglia/cytology , Stem Cells/cytology , Animals , Apoptosis , Brain/cytology , Cell Differentiation , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction
13.
Dis Markers ; 22(1-2): 95-102, 2006.
Article in English | MEDLINE | ID: mdl-16410654

ABSTRACT

The role of microglia-mediated inflammation in the progression of Alzheimer's disease (AD) neuropathology remains unclear. In this study, postmortem brain sections from AD and control cases were subjected to Human Leukocyte Antigen (HLA)-DR immunohistochemistry to examine microglia activation in the progression of AD assessed by pre-mortem clinical dementia rating (CDR) and postmortem pathological manifestations of neuritic plaque (NP) and neurofibrillary tangle (NT) according to the Consortium to Establish a Registry for Alzheimer's Disease (CERAD). In both gray and white matter of the entorhinal cortex (EC) and HLA-DR immunostaining increased with the progression of CDR or CERAD NP, and to a lesser degree with CERAD NT. Between CDR stages HLA-DR significance was found in moderate (CDR 2) to severe dementia (CDR 5) where as between CERAD NP stages staining increased significantly from NP 0 (no plaque) to NP 1 (sparse plaques), suggesting increased microglia activation begins with amyloid NP deposition. In the hippocampus, a significant increase in microglia immunostaining was found in the pyramidal cell layer of CA1 as early as CDR 1, and in the upper molecular layer of the dentate gyrus in CDR 0.5. This increase continues with the progression of CDR and reaches maximum in CDR 5. When assessed by CERAD NP stages however, a significant increase in microglia immunostaining was found only in mid-to-late stages (NP 3) and reduced staining was seen in NP 5. These results suggest that microglia activation increases with the progression of AD, with the increase varying depending on the involved brain region.


Subject(s)
Alzheimer Disease/diagnosis , Encephalitis/diagnosis , HLA-DR Antigens/analysis , Microglia/immunology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Biomarkers/analysis , Brain/pathology , Dementia/diagnosis , Disease Progression , Female , Humans , Male , Microglia/pathology
14.
J Histochem Cytochem ; 53(12): 1511-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16046669

ABSTRACT

Current methods for myelin staining in tissue sections include both histological and immunohistochemical techniques. Fluorescence immunohistochemistry, which uses antibodies against myelin components such as myelin basic protein, is often used because of the convenience for multiple labeling. To facilitate studies on myelin, this paper describes a quick and easy method for direct myelin staining in rodent and human tissues using novel near-infrared myelin (NIM) dyes that are comparable to other well-characterized histochemical reagents. The near-infrared fluorescence spectra of these probes allow fluorescent staining of tissue sections in multiple channels using visible light fluorophores commonly used in immunocytochemistry. These dyes have been used successfully to detect normal myelin structure and myelin loss in a mouse model of demyelination disease.


Subject(s)
Fluorescent Dyes , Myelin Sheath/metabolism , Animals , Brain/metabolism , Demyelinating Diseases/metabolism , Disease Models, Animal , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Spectrometry, Fluorescence , Spectroscopy, Near-Infrared
15.
FASEB J ; 18(7): 902-4, 2004 May.
Article in English | MEDLINE | ID: mdl-15033922

ABSTRACT

Recent epidemiological evidence indicates that insulin resistance, a proximal cause of Type II diabetes [a non-insulin dependent form of diabetes mellitus (NIDDM)], is associated with an increased relative risk for Alzheimer's disease (AD). In this study we examined the role of dietary conditions leading to NIDDM-like insulin resistance on amyloidosis in Tg2576 mice, which model AD-like neuropathology. We found that diet-induced insulin resistance promoted amyloidogenic beta-amyloid (Abeta) Abeta1-40 and Abeta1-42 peptide generation in the brain that corresponded with increased gamma-secretase activities and decreased insulin degrading enzyme (IDE) activities. Moreover, increased Abeta production also coincided with increased AD-type amyloid plaque burden in the brain and impaired performance in a spatial water maze task. Further exploration of the apparent interrelationship of insulin resistance to brain amyloidosis revealed a functional decrease in insulin receptor (IR)-mediated signal transduction in the brain, as suggested by decreased IR beta-subunit (IRbeta) Y1162/1163 autophosphorylation and reduced phosphatidylinositol 3 (PI3)-kinase/pS473-AKT/Protein kinase (PK)-B in these same brain regions. This latter finding is of particular interest given the known inhibitory role of AKT/PKB on glycogen synthase kinase (GSK)-3alpha activity, which has previously been shown to promote Abeta peptide generation. Most interestingly, we found that decreased pS21-GSK-3alpha and pS9-GSK-3beta phosphorylation, which is an index of GSK activation, positively correlated with the generation of brain C-terminal fragment (CTF)-gamma cleavage product of amyloid precursor protein, an index of gamma-secretase activity, in the brain of insulin-resistant relative to normoglycemic Tg2576 mice. Our study is consistent with the hypothesis that insulin resistance may be an underlying mechanism responsible for the observed increased relative risk for AD neuropathology, and presents the first evidence to suggest that IR signaling can influence Abeta production in the brain.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/biosynthesis , Amyloidosis/etiology , Brain/metabolism , Dietary Fats/toxicity , Insulin Resistance , Peptide Fragments/biosynthesis , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/genetics , Amyloidosis/genetics , Animals , Aspartic Acid Endopeptidases , Brain/pathology , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Endopeptidases/metabolism , Female , Glycogen Synthase Kinase 3/metabolism , Humans , Insulysin/deficiency , Insulysin/metabolism , Maze Learning , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Plaque, Amyloid , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Insulin/physiology , Signal Transduction , Spatial Behavior
16.
Exp Neurol ; 186(1): 70-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14980811

ABSTRACT

During the initial minutes of cerebral ischemia, lactic acid accumulates and acidifies brain pH to 6.0-6.7. Glutamate is also released during ischemia that activates glutamate receptors and induces excitotoxicity. While glutamate excitotoxicity is well established to induce ischemic injury, a role of lactic acidosis in ischemic brain damage is poorly understood. This study analyzes acidosis neurotoxicity in hippocampal slice cultures in the presence or absence of lactate. At pH 6.7, neuronal loss was similar whether or not lactate was present. At pH 6.4, neuronal loss was significantly greater in the presence of lactate suggesting that lactate potentiates the acidosis toxicity. At pH 6.4 in the presence of lactate, NMDA or non-NMDA receptor antagonists reduced neuronal loss, while in the absence of lactate, NMDA or non-NMDA receptor antagonists had little effect. [3H]-Glutamate uptake was inhibited by acidic pH, and the amount of inhibition was significantly greater in the presence of lactate. These findings suggest that lactate plays a role in acidosis neurotoxicity by inducing excitotoxicity. Lactic acidosis and excitotoxicity have been previously thought to be independent events during ischemia. This study suggests that during ischemia, lactic acidosis contributes to excitotoxic neuronal loss.


Subject(s)
Acidosis/complications , Hippocampus/drug effects , Lactic Acid/toxicity , Neurotoxicity Syndromes/etiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/therapeutic use , Acidosis/chemically induced , Acidosis/drug therapy , Analysis of Variance , Animals , Animals, Newborn , Cell Death/drug effects , Dizocilpine Maleate/therapeutic use , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/therapeutic use , Glucose/toxicity , Glutamic Acid/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Hydrogen-Ion Concentration , In Vitro Techniques , Intracellular Space/metabolism , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/pathology , Propidium , Rats , Rats, Sprague-Dawley , Time Factors , Tritium/metabolism
17.
J Biol Chem ; 278(51): 50970-7, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14507922

ABSTRACT

In previous studies we found that overexpression of the inducible form of cyclooxygenase, COX-2, in the brain exacerbated beta-amyloid (A beta) neuropathology in a transgenic mouse model of Alzheimer's disease. To explore the mechanism through which COX may influence A beta amyloidosis, we used an adenoviral gene transfer system to study the effects of human (h)COX-1 and hCOX-2 isoform expression on A beta peptide generation. We found that expression of hCOXs in human amyloid precursor protein (APP)-overexpressing (Chinese hamster ovary (CHO)-APPswe) cells or human neuroglioma (H4-APP751) cells resulting in 10-25 nM prostaglandin (PG)-E2 concentration in the conditioned medium coincided with an approximately 1.8-fold elevation of A beta-(1-40) and A beta-(1-42) peptide generation and an approximately 1.8-fold induction of the C-terminal fragment (CTF)-gamma cleavage product of the APP, an index of gamma-secretase activity. Treatment of APP-overexpressing cells with the non-selective COX inhibitor ibuprofen (1 microM, 48 h) or with the specific gamma-secretase inhibitor L-685,458 significantly attenuated hCOX-1- and hCOX-2-mediated induction of A beta peptide generation and CTF-gamma cleavage product formation. Based on this evidence, we next tested the hypothesis that COX expression might promote A beta peptide generation via a PG-E2-mediated mechanism. We found that exposure of CHO-APPswe or human embryonic kidney (HEK-APPswe) cells to PG-E2 (11-deoxy-PG-E2) at a concentration (10 nM) within the range of PG-E2 found in hCOX-expressing cells similarly promoted (approximately 1.8-fold) the generation of the CTF-gamma cleavage product of APP and commensurate A beta-(1-40) and A beta-(1-42) peptide elevation. The study suggests that expression of COXs may influence A beta peptide generation through mechanisms that involve PG-E2-mediated potentiation of gamma-secretase activity, further supporting a role for COX-2 and COX-1 in Alzheimer's disease neuropathology.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Endopeptidases/metabolism , Isoenzymes/physiology , Peptide Fragments/biosynthesis , Prostaglandin-Endoperoxide Synthases/physiology , Alzheimer Disease/etiology , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Cell Line , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Endopeptidases/drug effects , Humans , Ibuprofen/pharmacology , Membrane Proteins , Transfection
18.
Arch Neurol ; 60(3): 369-76, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12633148

ABSTRACT

BACKGROUND: Caspase gene expression has previously been reported in terminal Alzheimer disease (AD) brain, but, currently, little is known about the temporal pattern of caspase gene expression relative to the onset and clinical progression of AD. OBJECTIVE: To derive a profile of caspase gene expression and proapoptotic indexes as a function of the clinical and neuropathologic progression of AD dementia. SETTING AND PATIENTS: Postmortem survey of nursing home patients characterized clinically by Clinical Dementia Rating (CDR) and neuropathologically by Consortium to Establish a Registry for Alzheimer's Disease criteria. DESIGN AND OUTCOME MEASURES: To assess messenger RNA expression of caspase-1, -2L, -2S, -3, -5, -6, -7, -8, and -9; apoptotic cell death by TUNEL assay; and poly (ADP-ribose) polymerase cleavage in postmortem brain tissue samples from cognitively normal (CDR 0), high risk of developing AD dementia (CDR 0.5), and severe dementia (CDR 5) cases. RESULTS: Compared with CDR 0 cases, elevated messenger RNA expression of caspase-1 and caspase-7 in the entorhinal cortex of CDR 0.5 cases coincided with increased poly (ADP-ribose) polymerase cleavage but not apoptotic cell injury. In the entorhinal cortex of CDR 5 cases, we found elevation of caspase-1, -2L, -3, -5, -6, -7, -8, and -9 and a greater than 4-fold increase in TUNEL-positive cells. Caspase messenger RNA expression was closely associated with neurofibrillary tangle and, to a lesser extent, neuritic plaque density. CONCLUSIONS: Proapoptotic mechanisms may be at play early in the onset of AD (before overt signs of apoptosis) and may be a conditional factor for later apoptotic cell injury or death. These data have relevance to potential therapeutic interventions for AD using selective caspase inhibitors.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Brain/enzymology , Brain/pathology , Caspases/genetics , Aged , Aged, 80 and over , Apoptosis , Biomarkers , Cognition , Disease Progression , Female , Gene Expression Regulation, Enzymologic , Humans , In Situ Nick-End Labeling , Male , Poly(ADP-ribose) Polymerases/metabolism , RNA, Messenger/analysis
19.
Gene Expr ; 10(5-6): 271-8, 2002.
Article in English | MEDLINE | ID: mdl-12450219

ABSTRACT

Several epidemiologic studies have reported that cyclooxygenase (COX) inhibitors prevent/delay the onset of Alzheimer's disease (AD). Recent experimental studies suggest that these compounds can also diminish amyloid-beta (Abeta) neuropathology in rodent models of AD. To explore the relationship of COX expression to Abeta neuropathology, we crossed mice expressing both mutant amyloid precursor protein [K670N/M671L (APP(swe)] and mutant PS1 (A246E) with mice expressing human COX-2 selectively in neurons. We show here that human COX-2 expression in APP(swe)/PS1/COX-2 mice induces potentiation of brain parenchymal amyloid plaque formation and a greater than twofold increase in prostaglandin E2 production, at 24 months of age. This increased amyloid plaque formation coincided with a preferential elevation of Abeta1-40 and Abeta1-42 with no change in total amyloid precursor protein (APP) expression/content in the brain. Collectively these data suggest that COX-2 influences APP processing and promotes amyloidosis in the brain.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Amyloid/metabolism , Isoenzymes/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Animals , Brain/enzymology , Brain/metabolism , Cyclooxygenase 2 , Dinoprostone/metabolism , Disease Models, Animal , Humans , Image Processing, Computer-Assisted , Inflammation , Isoenzymes/metabolism , Mass Spectrometry , Membrane Proteins , Mice , Mice, Transgenic , Mutation , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/metabolism , Time Factors
20.
Neurobiol Aging ; 23(3): 327-34, 2002.
Article in English | MEDLINE | ID: mdl-11959394

ABSTRACT

Prior studies have shown that cyclooxygenase (COX)-2, an enzyme involved in inflammatory mechanisms as well as neuronal activities, is up-regulated in the Alzheimer's disease (AD) brain and may represent a therapeutic target for anti-inflammatory treatments. We report the effect of neuronal overexpression of human (h)COX-2 in a murine model of AD neuropathology. Transgenic mice expressing both the human amyloid precursor protein mutation (APPswe) and the human presenilin (PS1-A246E) mutation, with resultant AD plaque pathology, were crossed with transgenic mice expressing human (h)COX-2 in neurons. At 12 months of age, the APPswe/PS1-A246E/hCOX-2 triple-transgenic mice showed an elevation in the number of phosphorylated retinoblastoma (pRb) tumor suppressor protein and active caspase-3 immunopositive neurons, compared to double APPswe/PS1-A246E or single hCOX-2 transgenic controls. No detectable influence of neuronal hCOX-2 on AD neuropathology was found in the brain of APPswe/PS1-A246E/hCOX-2 triple-transgenic mice, compared to double APPswe/PS1-A246E. In vitro studies revealed that hCOX-2 overexpression in primary cortico-hippocampal neurons derived from the hCOX-2 transgenics accelerates beta-amyloid (Abeta)(1-42)-mediated apoptotic damage which was prevented by the cell cycle dependent (CDK) inhibitor, flavoperidol. The data indicates that COX-2 overexpression causes alteration of neuronal cell cycle in a murine model of AD neuropathology, and provides a rational basis for targeting neuronal COX-2 in therapeutic research aimed at slowing the clinical progression of AD.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Cell Cycle/genetics , Disease Models, Animal , Isoenzymes/genetics , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Caspase 3 , Caspases/genetics , Caspases/metabolism , Cell Death/genetics , Cells, Cultured , Cyclooxygenase 2 , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes/biosynthesis , Membrane Proteins , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Neurons/enzymology , Neurons/pathology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Prostaglandin-Endoperoxide Synthases/biosynthesis , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...