Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Blood Purif ; 53(4): 306-315, 2024.
Article in English | MEDLINE | ID: mdl-38228100

ABSTRACT

INTRODUCTION: Accumulation of ß2-microglobulin (B2M) in dialysis patients contributes to several comorbidities of end-stage kidney disease (ESKD). The LIXELLE® device adsorbs B2M from blood using sorbent bead technology. Studies in Japan showed that LIXELLE treatment during hemodialysis (HD) at blood flow rates up to 250 mL/min removes B2M above HD alone and is well tolerated. We investigated tolerance for LIXELLE treatment during HD at higher HD blood flow rates standard in the USA. METHODS: A prospective, open-label, non-randomized, single-arm, early-feasibility study (EFS) assessed tolerance and safety of LIXELLE treatment during HD at blood flow rates up to 450 mL/min. ESKD patients (40-75 years old) on thrice weekly outpatient HD were eligible. After a 1-week HD run-in, patients received LIXELLE plus HD at a blood flow rate of 250 mL/min (1 week), followed by LIXELLE plus HD at a blood flow rate up to 450 mL/min (1 week). These blood flow rates were tested with three LIXELLE column sizes in sequence (treatment = 6 weeks). B2M removal was assessed for each combination. RESULTS: Ten patients with a historic intradialytic hypotension (IDH) rate of 0.42 events/HD session/patient were enrolled. Nine patients completed all combinations without IDH events (treatment IDH rate: 0.56 events/HD session/patient). No treatment-emergent serious adverse events or significant changes in red blood cell, platelet, or complement indices except haptoglobin were reported. B2M reduction ratios and removal of select proteins (<40 kDa) increased with escalating column size and blood flow rate. CONCLUSION: LIXELLE plus HD across all column sizes was safe and well tolerated at blood flow rates up to 450 mL/min. Extent of B2M removal corresponded to column size-blood flow rate combinations. This EFS provides a risk profile to guide further studies of LIXELLE in ESKD patients at US-standard blood flow rates.


Subject(s)
Kidney Failure, Chronic , Renal Dialysis , Humans , Adult , Middle Aged , Aged , Renal Dialysis/adverse effects , Outpatients , Prospective Studies , Adsorption , beta 2-Microglobulin , Kidney Failure, Chronic/therapy , Kidney Failure, Chronic/etiology
2.
Sci Adv ; 9(46): eadf8764, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37976357

ABSTRACT

Leukotrienes, a class of inflammatory bioactive lipids, are well studied in the periphery, but less is known of their importance in the brain. We identified that the enzyme leukotriene A4 hydrolase (LTA4H) is expressed in healthy mouse neurons, and inhibition of LTA4H in aged mice improves hippocampal dependent memory. Single-cell nuclear RNA sequencing of hippocampal neurons after inhibition reveals major changes to genes important for synaptic organization, structure, and activity. We propose that LTA4H inhibition may act to improve cognition by directly inhibiting the enzymatic activity in neurons, leading to improved synaptic function. In addition, LTA4H plasma levels are increased in both aging and Alzheimer's disease and correlated with cognitive impairment. These results identify a role for LTA4H in the brain, and we propose that LTA4H inhibition may be a promising therapeutic strategy to treat cognitive decline in aging related diseases.


Subject(s)
Cognitive Dysfunction , Epoxide Hydrolases , Mice , Animals , Epoxide Hydrolases/chemistry , Cognitive Dysfunction/drug therapy
3.
eNeuro ; 10(6)2023 Jun.
Article in English | MEDLINE | ID: mdl-37321845

ABSTRACT

Peripheral administration of tissue inhibitor of metalloproteinases 2 (TIMP2), a protein inhibitor of matrix metalloproteinases (MMPs), has previously been shown to have beneficial effects on cognition and neurons in aged mice. Here, to better understand the potential of recombinant TIMP2 proteins, an IgG4Fc fusion protein (TIMP2-hIgG4) was developed to extend the plasma half-life of TIMP2. Following one month of administration of TIMP2 or TIMP2-hIgG4 via intraperitoneal injections, 23-month-old male C57BL/6J mice showed improved hippocampal-dependent memory in a Y-maze, increased hippocampal cfos gene expression, and increased excitatory synapse density in the CA1 and dentate gyrus (DG) of the hippocampus. Thus, fusion to hIgG4 extended the half-life of TIMP2 while retaining the beneficial cognitive and neuronal effects. Moreover, it retained its ability to cross the blood-brain barrier. To deepen the mechanistic understanding of the beneficial function of TIMP2 on neuronal activity and cognition, a TIMP2 construct lacking MMP inhibitory activity, Ala-TIMP2, was generated, which provides steric hindrance that prevents inhibition of MMPs by the TIMP2 protein while still allowing MMP binding. A comprehensive assessment of the MMP inhibitory and binding capacity of these engineered proteins is outlined. Surprisingly, MMP inhibition by TIMP2 was not essential for its beneficial effects on cognition and neuronal function. These findings both confirm previously published research, expand on the potential mechanism for the beneficial effects of TIMP2, and provide important details for a therapeutic path forward for TIMP2 recombinant proteins in aging-related cognitive decline.


Subject(s)
Cognition , Matrix Metalloproteinases , Animals , Male , Mice , Aging , Matrix Metalloproteinases/metabolism , Mice, Inbred C57BL
4.
Brain Behav ; 12(9): e2736, 2022 09.
Article in English | MEDLINE | ID: mdl-35971662

ABSTRACT

INTRODUCTION: Increasing age is the number one risk factor for developing cognitive decline and neurodegenerative disease. Aged humans and mice exhibit numerous molecular changes that contribute to a decline in cognitive function and increased risk of developing age-associated diseases. Here, we characterize multiple age-associated changes in male C57BL/6J mice to understand the translational utility of mouse aging. METHODS: Male C57BL/6J mice from various ages between 2 and 24 months of age were used to assess behavioral, as well as, histological and molecular changes across three modalities: neuronal, microgliosis/neuroinflammation, and the neurovascular unit (NVU). Additionally, a cohort of 4- and 22-month-old mice was used to assess blood-brain barrier (BBB) breakdown. Mice in this cohort were treated with a high, acute dose of lipopolysaccharide (LPS, 10 mg/kg) or saline control 6 h prior to sacrifice followed by tail vein injection of 0.4 kDa sodium fluorescein (100 mg/kg) 2 h later. RESULTS: Aged mice showed a decline in cognitive and motor abilities alongside decreased neurogenesis, proliferation, and synapse density. Further, neuroinflammation and circulating proinflammatory cytokines were increased in aged mice. Additionally, we found changes at the BBB, including increased T cell infiltration in multiple brain regions and an exacerbation in BBB leakiness following chemical insult with age. There were also a number of readouts that were unchanged with age and have limited utility as markers of aging in male C57BL/6J mice. CONCLUSIONS: Here we propose that these changes may be used as molecular and histological readouts that correspond to aging-related behavioral decline. These comprehensive findings, in the context of the published literature, are an important resource toward deepening our understanding of normal aging and provide an important tool for studying aging in mice.


Subject(s)
Cognitive Dysfunction , Neurodegenerative Diseases , Aging/physiology , Animals , Cognitive Dysfunction/pathology , Cytokines/metabolism , Fluorescein/metabolism , Hippocampus/metabolism , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL
5.
J Exp Med ; 214(4): 1081-1092, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28298456

ABSTRACT

Recent genetic evidence supports a link between microglia and the complement system in Alzheimer's disease (AD). In this study, we uncovered a novel role for the microglial complement receptor 3 (CR3) in the regulation of soluble ß-amyloid (Aß) clearance independent of phagocytosis. Unexpectedly, ablation of CR3 in human amyloid precursor protein-transgenic mice results in decreased, rather than increased, Aß accumulation. In line with these findings, cultured microglia lacking CR3 are more efficient than wild-type cells at degrading extracellular Aß by secreting enzymatic factors, including tissue plasminogen activator. Furthermore, a small molecule modulator of CR3 reduces soluble Aß levels and Aß half-life in brain interstitial fluid (ISF), as measured by in vivo microdialysis. These results suggest that CR3 limits Aß clearance from the ISF, illustrating a novel role for CR3 and microglia in brain Aß metabolism and defining a potential new therapeutic target in AD.


Subject(s)
Amyloid beta-Peptides/analysis , Brain/metabolism , Macrophage-1 Antigen/physiology , Microglia/physiology , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , Benzoates/pharmacology , Mice , Mice, Inbred C57BL , Proteolysis , Thiohydantoins/pharmacology
6.
JAMA Neurol ; 73(11): 1325-1333, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27598869

ABSTRACT

IMPORTANCE: Alzheimer disease (AD) pathology starts long before clinical symptoms manifest, and there is no therapy to treat, delay, or prevent the disease. A shared blood circulation between 2 mice (aka parabiosis) or repeated injections of young blood plasma (plasma from 2- to 3-month-old mice) into old mice has revealed benefits of young plasma on synaptic function and behavior. However, to our knowledge, the potential benefit of young blood has not been tested in preclinical models of neurodegeneration or AD. OBJECTIVES: To determine whether young blood plasma ameliorates pathology and cognition in a mouse model for AD and could be a possible future treatment for the disease. DESIGN, SETTING, AND PARTICIPANTS: In this preclinical study, mice that harbor a human mutant APP gene, which causes familial AD, were aged to develop AD-like disease including accumulation of amyloid plaques, loss of synaptic and neuronal proteins, and behavioral deficits. The initial parabiosis studies were done in 2010, and the final studies were conducted in 2014. Alzheimer disease model mice were then treated either by surgically connecting them with a young healthy mouse, thus providing a shared blood circulation through parabiosis, or through repeated injections of plasma from young mice. MAIN OUTCOMES AND MEASURES: Neuropathological parameters and changes in hippocampal gene expression in response to the treatment were assessed. In addition, cognition was tested in AD model mice intravenously injected with young blood plasma. RESULTS: Aged mutant amyloid precursor protein mice with established disease showed a near complete restoration in levels of synaptic and neuronal proteins after exposure to young blood in parabiosis (synaptophysin P = .02; calbindin P = .02) or following intravenous plasma administration (synaptophysin P < .001; calbindin P = .14). Amyloid plaques were not affected, but the beneficial effects in neurons in the hippocampus were accompanied by a reversal of abnormal extracellular receptor kinase signaling (P = .05), a kinase implicated in AD. Moreover, young plasma administration was associated with improved working memory (P = .01) and associative memory (P = .02) in amyloid precursor protein mice. CONCLUSIONS AND RELEVANCE: Factors in young blood have the potential to ameliorate disease in a model of AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Blood Component Transfusion/methods , Cross Circulation/methods , Hippocampus/metabolism , Age Factors , Amyloid beta-Protein Precursor , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
Pain ; 155(11): 2377-89, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25218828

ABSTRACT

Complex regional pain syndrome (CRPS) is a painful, disabling, chronic condition whose etiology remains poorly understood. The recent suggestion that immunological mechanisms may underlie CRPS provides an entirely novel framework in which to study the condition and consider new approaches to treatment. Using a murine fracture/cast model of CRPS, we studied the effects of B-cell depletion using anti-CD20 antibodies or by performing experiments in genetically B-cell-deficient (µMT) mice. We observed that mice treated with anti-CD20 developed attenuated vascular and nociceptive CRPS-like changes after tibial fracture and 3 weeks of cast immobilization. In mice with established CRPS-like changes, the depletion of CD-20+ cells slowly reversed nociceptive sensitization. Correspondingly, µMT mice, deficient in producing immunoglobulin M (IgM), failed to fully develop CRPS-like changes after fracture and casting. Depletion of CD20+ cells had no detectable effects on nociceptive sensitization in a model of postoperative incisional pain, however. Immunohistochemical experiments showed that CD20+ cells accumulate near the healing fracture but few such cells collect in skin or sciatic nerves. On the other hand, IgM-containing immune complexes were deposited in skin and sciatic nerve after fracture in wild-type, but not in µMT fracture/cast, mice. Additional experiments demonstrated that complement system activation and deposition of membrane attack complexes were partially blocked by anti-CD20+ treatment. Collectively, our results suggest that CD20-positive B cells produce antibodies that ultimately support the CRPS-like changes in the murine fracture/cast model. Therapies directed at reducing B-cell activity may be of use in treating patients with CRPS.


Subject(s)
Autoimmunity/physiology , B-Lymphocyte Subsets/pathology , Complex Regional Pain Syndromes/complications , Complex Regional Pain Syndromes/immunology , Hyperalgesia/physiopathology , Nociception/physiology , Animals , Antigens, CD20/metabolism , Complex Regional Pain Syndromes/etiology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fractures, Bone , Hindlimb/physiopathology , Immunoglobulin M/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pain Measurement , Pain Threshold , Time Factors
8.
Sci Transl Med ; 6(243): 243ra86, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-24990881

ABSTRACT

Genetic variants in the triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to Nasu-Hakola disease, Alzheimer's disease (AD), Parkinson's disease, amyotrophic lateral sclerosis, frontotemporal dementia (FTD), and FTD-like syndrome without bone involvement. TREM2 is an innate immune receptor preferentially expressed by microglia and is involved in inflammation and phagocytosis. Whether and how TREM2 missense mutations affect TREM2 function is unclear. We report that missense mutations associated with FTD and FTD-like syndrome reduce TREM2 maturation, abolish shedding by ADAM proteases, and impair the phagocytic activity of TREM2-expressing cells. As a consequence of reduced shedding, TREM2 is virtually absent in the cerebrospinal fluid (CSF) and plasma of a patient with FTD-like syndrome. A decrease in soluble TREM2 was also observed in the CSF of patients with AD and FTD, further suggesting that reduced TREM2 function may contribute to increased risk for two neurodegenerative disorders.


Subject(s)
Membrane Glycoproteins/genetics , Neurodegenerative Diseases/genetics , Phagocytosis/physiology , Receptors, Immunologic/genetics , Alzheimer Disease/genetics , Biological Transport/genetics , Biological Transport/physiology , Cell Line , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Frontotemporal Dementia/genetics , Humans , Mutation , Phagocytosis/genetics
9.
Neuron ; 79(5): 873-86, 2013 Sep 04.
Article in English | MEDLINE | ID: mdl-24012002

ABSTRACT

Phagocytosis controls CNS homeostasis by facilitating the removal of unwanted cellular debris. Accordingly, impairments in different receptors or proteins involved in phagocytosis result in enhanced inflammation and neurodegeneration. While various studies have identified extrinsic factors that modulate phagocytosis in health and disease, key intracellular regulators are less understood. Here we show that the autophagy protein beclin 1 is required for efficient phagocytosis in vitro and in mouse brains. Furthermore, we show that beclin 1-mediated impairments in phagocytosis are associated with dysfunctional recruitment of retromer to phagosomal membranes, reduced retromer levels, and impaired recycling of phagocytic receptors CD36 and Trem2. Interestingly, microglia isolated from human Alzheimer's disease (AD) brains show significantly reduced beclin 1 and retromer protein levels. These findings position beclin 1 as a link between autophagy, retromer trafficking, and receptor-mediated phagocytosis and provide insight into mechanisms by which phagocytosis is regulated and how it may become impaired in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apoptosis Regulatory Proteins/metabolism , Brain/metabolism , Membrane Proteins/metabolism , Microglia/metabolism , Phagocytosis/physiology , Vesicular Transport Proteins/metabolism , Alzheimer Disease/physiopathology , Animals , Apoptosis Regulatory Proteins/physiology , Autophagy/physiology , Beclin-1 , CD36 Antigens/metabolism , Cell Line , Humans , Membrane Glycoproteins/metabolism , Membrane Proteins/physiology , Mice , Microglia/physiology , Receptors, Immunologic/metabolism , Vesicular Transport Proteins/physiology
10.
J Clin Invest ; 122(4): 1156-63, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22466657

ABSTRACT

While immune responses in neurodegeneration were regarded as little more than a curiosity a decade ago, they are now increasingly moving toward center stage. Factors driving this movement include the recognition that most of the relevant immune molecules are produced within the brain, that microglia are proficient immune cells shaping neuronal circuitry and fate, and that systemic immune responses affect brain function. We will review this complex field from the perspective of neurons, extra-neuronal brain cells, and the systemic environment and highlight the possibility that cell intrinsic innate immune molecules in neurons may function in neurodegenerative processes.


Subject(s)
Neurodegenerative Diseases/immunology , Neuroimmunomodulation/physiology , Animals , Brain/immunology , Brain/pathology , Cell Communication , Cytokines/physiology , Humans , Inflammation/immunology , Inflammation/physiopathology , Mice , Mice, Neurologic Mutants , Microglia/immunology , Models, Immunological , Models, Neurological , Neurodegenerative Diseases/pathology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Neurotransmitter Agents/physiology , Phagocytosis , Receptors, Immunologic/physiology , Toll-Like Receptors/physiology
11.
Nature ; 477(7362): 90-4, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21886162

ABSTRACT

In the central nervous system, ageing results in a precipitous decline in adult neural stem/progenitor cells and neurogenesis, with concomitant impairments in cognitive functions. Interestingly, such impairments can be ameliorated through systemic perturbations such as exercise. Here, using heterochronic parabiosis we show that blood-borne factors present in the systemic milieu can inhibit or promote adult neurogenesis in an age-dependent fashion in mice. Accordingly, exposing a young mouse to an old systemic environment or to plasma from old mice decreased synaptic plasticity, and impaired contextual fear conditioning and spatial learning and memory. We identify chemokines--including CCL11 (also known as eotaxin)--the plasma levels of which correlate with reduced neurogenesis in heterochronic parabionts and aged mice, and the levels of which are increased in the plasma and cerebrospinal fluid of healthy ageing humans. Lastly, increasing peripheral CCL11 chemokine levels in vivo in young mice decreased adult neurogenesis and impaired learning and memory. Together our data indicate that the decline in neurogenesis and cognitive impairments observed during ageing can be in part attributed to changes in blood-borne factors.


Subject(s)
Chemokines/blood , Chemokines/metabolism , Learning/physiology , Neurogenesis/physiology , Aging , Animals , Chemokine CCL11/blood , Chemokine CCL11/cerebrospinal fluid , Chemokine CCL11/metabolism , Chemokine CCL11/pharmacology , Chemokines/cerebrospinal fluid , Female , Learning/drug effects , Learning Disabilities/blood , Learning Disabilities/cerebrospinal fluid , Learning Disabilities/physiopathology , Male , Memory Disorders/blood , Memory Disorders/cerebrospinal fluid , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Parabiosis , Plasma/chemistry , Time Factors
12.
J Neurochem ; 116(3): 385-95, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21091478

ABSTRACT

γ-Secretase modulators (GSMs) inhibit the generation of amyloidogenic Aß42 peptides and are promising agents for treatment or prevention of Alzheimer's disease (AD). Recently, a second generation of GSMs with favorable pharmacological properties has emerged, but preclinical studies to assess their efficacy in vivo are lacking. Such studies rely on transgenic mouse models that express amyloid precursor protein (APP) and presenilin (PSEN) mutations associated with early-onset familial AD. Previously, we have shown that certain PSEN1 mutations attenuated the response of cultured cells to GSMs and potentially confound in vivo studies in AD mouse models. However, different combinations of familial AD mutations might have synergistic or opposing effects, and we have now systematically determined the response of APP and PSEN1 mutations present in current AD models. Using a potent acidic GSM, we found that APP mutations, either single mutations or in combination, did not affect the potency of GSMs. In contrast, all PSEN1 mutations that have been used to accelerate pathological changes in AD models strongly attenuated the Aß42-lowering activity of GSMs with two exceptions (M146L, A246E). Similar results were obtained with potent non-acidic GSMs indicating that the attenuating effect of PSEN1 mutations cannot simply be overcome by increased potency or structural changes. Notably, two non-acidic compounds fully compensated the attenuating effect of the PSEN1-G384A mutation. Taken together, our findings indicate that most AD models with rapid pathology and advanced phenotypes are unsuitable for preclinical GSM studies. However, we also provide evidence that additional compound screens could discover GSMs that are able to break the attenuating effects of PSEN mutations.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Mutation/genetics , Presenilin-1/genetics , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Presenilin-1/physiology , Structure-Activity Relationship
13.
J Neurochem ; 111(6): 1369-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19804379

ABSTRACT

The Swedish mutation within the amyloid precursor protein (APP) causes early-onset Alzheimer's disease due to increased cleavage of APP by BACE1. While beta-secretase shedding of Swedish APP (APPswe) largely results from an activity localized in the late secretory pathway, cleavage of wild-type APP occurs mainly in endocytic compartments. However, we show that liberation of Abeta from APPswe is still dependent on functional internalization from the cell surface. Inspite the unchanged overall beta-secretase cleaved soluble APP released from APP(swe) secretion, mutations of the APPswe internalization motif strongly reduced C99 levels and substantially decreased Abeta secretion. We point out that alpha-secretase activity-mediated conversion of C99 to C83 is the main cause of this Abeta reduction. Furthermore, we demonstrate that alpha-secretase cleavage of C99 even contributes to the reduction of Abeta secretion of internalization deficient wild-type APP. Therefore, inhibition of alpha-secretase cleavage increased Abeta secretion through diminished conversion of C99 to C83 in APP695, APP695swe or C99 expressing cells.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Peptide Fragments/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , Biotinylation/methods , CHO Cells , Cricetinae , Cricetulus , Gene Expression Regulation/genetics , Humans , Mutation/genetics , Peptide Fragments/genetics , Protein Interaction Domains and Motifs/physiology , Transfection
14.
Neurodegener Dis ; 6(1-2): 1-8, 2009.
Article in English | MEDLINE | ID: mdl-18349521

ABSTRACT

BACKGROUND: Epidemiological studies have suggested that long-term use of nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with a reduced incidence of Alzheimer's disease (AD). Several mechanisms have been proposed to explain these findings including increased shedding of the soluble ectodomain of the amyloid precursor protein (sAPP), which functions as a neurotrophic and neuroprotective factor in vitroand in vivo. OBJECTIVE: To clarify whether NSAIDs consistently stimulate sAPP secretion. METHODS: 293-EBNA cells with stable overexpression of an APP-alkaline phosphatase fusion protein (APP-AP), SH-SY5Y and PC12 cells or primary telencephalic chicken neurons were treated with ibuprofen or indomethacin. APP shedding was then determined by measuring AP activity in conditioned media, Western blot analysis with antibodies against total sAPP or specific for sAPP-alpha, or in a pulse-chase paradigm. RESULTS: AP activity in conditioned media was not increased after NSAID treatment of 293-EBNA cells whereas it was elevated by phorbol ester. Surprisingly, ibuprofen or indomethacin treatment of SH-SY5Y and PC12 cells expressing endogenous APP did not cause changes in sAPP or sAPP-alpha secretion or downregulation of cellular APP. These findings were further corroborated in primary chicken neuronal cultures. CONCLUSIONS: Using various experimental settings, we were unable to confirm sAPP or sAPP-alpha stimulation with the NSAIDs ibuprofen and indomethacin in transfected and nontransfected cells of neuronal and nonneuronal origin. Importantly, these findings seem to rule out chronic sAPP stimulation as an alternative mechanism of NSAID action in AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Alkaline Phosphatase/adverse effects , Amyloid beta-Protein Precursor/drug effects , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Ibuprofen/pharmacology , Indomethacin/pharmacology , Kinetics , Neuroblastoma , PC12 Cells/drug effects , Rats , Tetradecanoylphorbol Acetate/pharmacology
15.
J Biol Chem ; 283(25): 17049-54, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18426795

ABSTRACT

Proteolytic processing of the amyloid precursor protein by beta- and gamma-secretase generates the amyloid-beta (Abeta) peptides, which are principal drug targets in Alzheimer disease therapeutics. gamma-Secretase has imprecise cleavage specificity and generates the most abundant Abeta40 and Abeta42 species together with longer and shorter peptides such as Abeta38. Several mechanisms could explain the production of multiple Abeta peptides by gamma-secretase, including sequential processing of longer into shorter Abeta peptides. A novel class of gamma-secretase modulators (GSMs) that includes some non-steroidal anti-inflammatory drugs has been shown to selectively lower Abeta42 levels without a change in Abeta40 levels. A signature of GSMs is the concomitant increase in shorter Abeta peptides, such as Abeta38, leading to the suggestion that generation of Abeta42 and Abeta38 peptide species by gamma-secretase is coordinately regulated. However, no evidence for or against such a precursor-product relationship has been provided. We have previously shown that stable overexpression of aggressive presenilin-1 (PS1) mutations associated with early-onset familial Alzheimer disease attenuated the cellular response to GSMs, resulting in greatly diminished Abeta42 reductions as compared with wild type PS1. We have now used this model system to investigate whether Abeta38 production would be similarly affected indicating coupled generation of Abeta42 and Abeta38 peptides. Surprisingly, treatment with the GSM sulindac sulfide increased Abeta38 production to similar levels in four different PS1 mutant cell lines as compared with wild type PS1 cells. This was confirmed with the structurally divergent GSMs ibuprofen and indomethacin. Mass spectrometry analysis and high resolution urea gel electrophoresis further demonstrated that sulindac sulfide did not induce detectable compensatory changes in levels of other Abeta peptide species. These data provide evidence that Abeta42 and Abeta38 species can be independently generated by gamma-secretase and argue against a precursor-product relationship between these peptides.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Ibuprofen/pharmacology , Molecular Sequence Data , Mutation , Presenilin-1/genetics , Sulindac/analogs & derivatives , Sulindac/pharmacology
16.
J Biol Chem ; 282(34): 24504-13, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17573346

ABSTRACT

Abeta42-lowering nonsteroidal anti-inflammatory drugs (NSAIDs) constitute the founding members of a new class of gamma-secretase modulators that avoid side effects of pan-gamma-secretase inhibitors on NOTCH processing and function, holding promise as potential disease-modifying agents for Alzheimer disease (AD). These modulators are active in cell-free gamma-secretase assays indicating that they directly target the gamma-secretase complex. Additional support for this hypothesis was provided by the observation that certain mutations in presenilin-1 (PS1) associated with early-onset familial AD (FAD) change the cellular drug response to Abeta42-lowering NSAIDs. Of particular interest is the PS1-DeltaExon9 mutation, which provokes a pathogenic increase in the Abeta42/Abeta40 ratio and dramatically reduces the cellular response to the Abeta42-lowering NSAID sulindac sulfide. This FAD PS1 mutant is unusual as a splice-site mutation results in deletion of amino acids Thr(291)-Ser(319) including the endoproteolytic cleavage site of PS1, and an additional amino acid exchange (S290C) at the exon 8/10 splice junction. By genetic dissection of the PS1-DeltaExon9 mutation, we now demonstrate that a synergistic effect of the S290C mutation and the lack of endoproteolytic cleavage is sufficient to elevate the Abeta42/Abeta40 ratio and that the attenuated response to sulindac sulfide results partially from the deficiency in endoproteolysis. Importantly, a wider screen revealed that a diminished response to Abeta42-lowering NSAIDs is common among aggressive FAD PS1 mutations. Surprisingly, these mutations were also partially unresponsive to gamma-secretase inhibitors of different structural classes. This was confirmed in a mouse model with transgenic expression of the PS1-L166P mutation, in which the potent gamma-secretase inhibitor LY-411575 failed to reduce brain levels of soluble Abeta42. In summary, these findings highlight the importance of genetic background in drug discovery efforts aimed at gamma-secretase, suggesting that certain AD mouse models harboring aggressive PS mutations may not be informative in assessing in vivo effects of gamma-secretase modulators and inhibitors.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/physiology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Enzyme Inhibitors/pharmacology , Mutation , Peptide Fragments/metabolism , Peptide Fragments/physiology , Presenilin-1/genetics , Presenilin-1/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Sequence Homology, Amino Acid
17.
Neurodegener Dis ; 3(4-5): 298-304, 2006.
Article in English | MEDLINE | ID: mdl-17047371

ABSTRACT

The amyloid-beta (Abeta) peptides and specifically the highly amyloidogenic isoform Abeta42 appear to be key agents in the pathogenesis of familial and sporadic forms of Alzheimer's disease (AD). The final step in the generation of Abeta from the amyloid precursor protein is catalyzed by the multiprotein complex gamma-secretase, which constitutes a prime drug target for prevention and therapy of the disease. However, highly potent gamma-secretase inhibitors that block formation of all Abeta peptides have provoked troubling side effects in preclinical animal models of AD. This toxicity can be readily explained by the promiscuous substrate specificity of gamma-secretase and its essential role in the NOTCH signaling pathway. For that reason and because of the crucial role of Abeta42 in the pathogenesis of the disease, selective inhibition of Abeta42 production would seem to be a more promising alternative to complete inhibition of gamma-secretase activity. This theoretical concept has edged much closer to clinical reality with the surprising finding that certain nonsteroidal anti-inflammatory drugs (NSAIDs), including ibuprofen, and derived compounds display preferential Abeta42-lowering activity. In contrast to gamma-secretase inhibitors, these gamma-secretase modulators effectively suppress Abeta42 production while sparing processing of NOTCH and other gamma-secretase substrates. Although not fully resolved on the molecular level, the mechanism of action of Abeta42-lowering NSAIDs is independent of cyclooxygenase inhibition and most likely involves direct interaction with components of the gamma-secretase complex or its substrates. Current efforts to improve the pharmacological shortcomings of available gamma-secretase modulators will hopefully lead to the development of clinically useful Abeta42-lowering compounds in the near future.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/drug effects , Amyloid beta-Peptides/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Clinical Trials as Topic , Humans
18.
J Neurochem ; 96(2): 355-65, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16300630

ABSTRACT

Certain non-steroidal anti-inflammatory drugs (NSAIDs) preferentially inhibit production of the amyloidogenic Abeta42 peptide, presumably by direct modulation of gamma-secretase activity. A recent report indicated that NSAIDs could reduce Abeta42 by inhibition of the small GTPase Rho, and a single inhibitor of Rho kinase (ROCK) mimicked the effects of Abeta42-lowering NSAIDs. To investigate whether Abeta42 reduction is a common property of ROCK inhibitors, we tested commercially available compounds in cell lines that were previously used to demonstrate the Abeta42-lowering activity of NSAIDs. Surprisingly, we found that two ROCK inhibitors reduced total Abeta secretion in a dose-dependent manner but showed no selectivity for Abeta42. In addition, ROCK inhibitors did not increase Abeta38 secretion in cell-based assays or reduce Abeta production in gamma-secretase in vitro assays, which are critical characteristics of Abeta42-lowering NSAIDs. The reduction in total Abeta levels by ROCK inhibitors was not accompanied by overall-changes in amyloid precursor protein processing. Targeting ROCK by expression of dominant-negative or constitutively active ROCK mutants failed to modulate Abeta secretion, indicating that ROCK inhibition may either be redundant or insufficient for Abeta reduction by ROCK inhibitors. Taken together, these results seem to exclude a mechanistic involvement of ROCK in the Abeta42-lowering activity of NSAIDs.


Subject(s)
Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Enzyme Inhibitors/pharmacology , Peptide Fragments/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases , CHO Cells , Cell Survival/drug effects , Cricetinae , Cricetulus , Endopeptidases/metabolism , Enzyme Inhibitors/poisoning , Intracellular Signaling Peptides and Proteins , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , rho-Associated Kinases
19.
J Cell Sci ; 117(Pt 20): 4643-51, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15340007

ABSTRACT

Outer dense fibre protein 2 (ODF2) is a major protein of sperm tail outer dense fibres which are prominent sperm tail-specific cytoskeletal structures. Moreover, ODF2 was also identified as a widespread component of the centrosomal scaffold and was found to associate preferentially with the appendages of the mother centriole [Nakagawa, Y., Yamane, Y., Okanoue, T., Tsukita, S. and Tsukita, S. (2001) Mol. Biol. Cell 12, 1687-1697]. Secondary structure predictions indicated ODF2 as an overall coiled-coil protein with a putative fibre forming capacity. To investigate its potential functions in generating the centrosomal scaffold and in microtubule nucleation we asked whether ODF2 is able to form a fibrillar structure by self-association in vivo and if it interacts with microtubules. By cytological investigation of transfected mammalian cells expressing ODF2-GFP fusion proteins and in vitro coprecipitation assays we could demonstrate that ODF2 is a self-interacting protein that forms a fibrillar structure partially linked to the microtubule network. Microtubule cosedimentation and coprecipitation assays indicated ODF2 as a microtubule-associated protein. However, we could not demonstrate a direct interaction of ODF2 with tubulin, suggesting that binding of endogenous ODF2 to the axonemal as well as to centrosomal microtubules may be mediated by, as yet, unknown proteins.


Subject(s)
Centrosome/metabolism , Heat-Shock Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Structure, Secondary , Animals , COS Cells , Chlorocebus aethiops , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Oocytes/chemistry , Protein Binding , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Xenopus laevis
20.
J Cell Sci ; 117(Pt 2): 189-98, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14676273

ABSTRACT

Histone macroH2A1.2 and the murine heterochromatin protein 1, HP1 beta, have both been implicated in meiotic sex chromosome inactivation (MSCI) and the formation of the XY-body in male meiosis. In order to get a closer insight into the function of histone macroH2A1.2 we have investigated the localisation of macroH2A1.2 in surface spread spermatocytes from normal male mice and in oocytes of XX and XYTdym1 mice. Oocytes of XYTdym1 mice have no XY-body or MSCI despite having an XY chromosome constitution, so the presence or absence of 'XY-body' proteins in association with the X and/or Y chromosome of these oocytes enables some discrimination between potential functions of XY-body located proteins. We demonstrate here that macroH2A1.2 localises to the X and Y chromatin of spermatocytes as they condense to form the XY-body but is not associated with the X and Y chromatin of XYTdym1 early pachytene oocytes. MacroH2A1.2 and HP1 beta co-localise to autosomal pericentromeric heterochromatin in spermatocytes. However, the two proteins show temporally and spatially distinct patterns of association to X and Y chromatin.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Histones/metabolism , Sex Chromatin/metabolism , Animals , Chromatin/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosome Pairing/genetics , Chromosome Pairing/physiology , Chromosomes/metabolism , Female , Histones/genetics , Male , Meiosis/physiology , Mice , Oocytes/cytology , Oocytes/metabolism , Sex Chromatin/genetics , Sex Chromosomes/genetics , Spermatocytes/cytology , Spermatocytes/metabolism , Y Chromosome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...