42 results on '"Tesseur I"'
Search Results
2. Screening and Recruitment Experience When Using Biomarker Based Population Definition in Alzheimer’s Disease Studies
- Author
-
Smekens, P., Mesens, S., Engelborghs, Sebastiaan, Börjesson-Hanson, Anne, Christoph, L., Prins, N.D., Hugon, J., Andreassen, N., cuevas Jiméne, A.I., Timmers, M., Tesseur, I., Van Nueten, L., Streffer, J., Clinical sciences, and Neurology
- Subjects
recruitment experience ,Medicine(all) ,Screening ,biomarker ,Alzheimer's disease - Published
- 2016
3. Pharmacodynamics of the Oral BACE Inhibitor JNJ-54861911 in Early Alzheimer's Disease
- Author
-
Streffer, J., Börjesson-Hanson, Anne, Van Broek, B., Smekens, P., Timmers, M., Tesseur, I., Tatikola, Kanaka, Russo, A., Sinha, Vivek, Salvadore, G., Aguilar, M., Frank, A., Matias-Guiu, Jorge, Boada, Merce, Baquero, M., Tristmans, L., Van Nueten, L., Andreassen, N., Engelborghs, Sebastiaan, Clinical sciences, and Neurology
- Subjects
Medicine(all) ,Alzheimer Disease - Published
- 2016
4. The Disintegrin/Metalloproteinase ADAM10 Is Essential for the Establishment of the Brain Cortex
- Author
-
Jorissen, E., primary, Prox, J., additional, Bernreuther, C., additional, Weber, S., additional, Schwanbeck, R., additional, Serneels, L., additional, Snellinx, A., additional, Craessaerts, K., additional, Thathiah, A., additional, Tesseur, I., additional, Bartsch, U., additional, Weskamp, G., additional, Blobel, C. P., additional, Glatzel, M., additional, De Strooper, B., additional, and Saftig, P., additional
- Published
- 2010
- Full Text
- View/download PDF
5. No evidence for cholinergic problems in apolipoprotein E knockout and apolipoprotein E4 transgenic mice
- Author
-
Bronfman, F.C, primary, Tesseur, I, additional, Hofker, M.H, additional, Havekens, L.M, additional, and Van Leuven, F, additional
- Published
- 2000
- Full Text
- View/download PDF
6. Lipoprotein receptor-related protein in brain and in cultured neurons of mice deficient in receptor-associated protein and transgenic for apolipoprotein E4 or amyloid precursor protein
- Author
-
Umans, L, primary, Serneels, L, additional, Lorent, K, additional, Dewachter, I, additional, Tesseur, I, additional, Moechars, D, additional, and Van Leuven, F, additional
- Published
- 1999
- Full Text
- View/download PDF
7. Early phenotypic changes in transgenic mice that overexpress different mutants of amyloid precursor protein in brain.
- Author
-
Moechars, D, Dewachter, I, Lorent, K, Reversé, D, Baekelandt, V, Naidu, A, Tesseur, I, Spittaels, K, Haute, C V, Checler, F, Godaux, E, Cordell, B, and Van Leuven, F
- Abstract
Transgenic mice overexpressing different forms of amyloid precursor protein (APP), i.e. wild type or clinical mutants, displayed an essentially comparable early phenotype in terms of behavior, differential glutamatergic responses, deficits in maintenance of long term potentiation, and premature death. The cognitive impairment, demonstrated in F1 hybrids of the different APP transgenic lines, was significantly different from nontransgenic littermates as early as 3 months of age. Biochemical analysis of secreted and membrane-bound APP, C-terminal "stubs," and Abeta(40) and Abeta(42) peptides in brain indicated that no single intermediate can be responsible for the complex of phenotypic dysfunctions. As expected, the Abeta(42) levels were most prominent in APP/London transgenic mice and correlated directly with the formation of amyloid plaques in older mice of this line. Plaques were associated with immunoreactivity for hyperphosphorylated tau, eventually signaling some form of tau pathology. In conclusion, the different APP transgenic mouse lines studied display cognitive deficits and phenotypic traits early in life that dissociated in time from the formation of amyloid plaques and will be good models for both early and late neuropathological and clinical aspects of Alzheimer's disease.
- Published
- 1999
8. Highly sensitive and specific bioassay for measuring bioactive TGF-β
- Author
-
Berber Elisabeth, Zou Kun, Tesseur Ina, Zhang Hui, and Wyss-Coray Tony
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Background Transforming Growth Factor-β (TGF-β) regulates key biological processes during development and in adult tissues and has been implicated in many diseases. To study the biological functions of TGF-β, sensitive, specific, and convenient bioassays are necessary. Here we describe a new cell-based bioassay that fulfills these requirements. Results Embryonic fibroblasts from Tgfb1-/- mice were stably transfected with a reporter plasmid consisting of TGF-β responsive Smad-binding elements coupled to a secreted alkaline phosphatase reporter gene (SBE-SEAP). Clone MFB-F11 showed more than 1000-fold induction after stimulation with 1 ng/ml TGF-β1, and detected as little as 1 pg/ml TGF-β1. MFB-F11 cells were highly induced by TGF-β1, TGF-β2 and TGF-β3, but did not show induction with related family members activin, nodal, BMP-2 and BMP-6 or with trophic factors bFGF and BDNF. MFB-F11 cells can detect and quantify TGF-β in biological samples without prior enrichment of TGF-βs, and can detect biologically activated TGF-β in a cell co-culture system. Conclusion MFB-F11 cells can be used to rapidly and specifically measure TGF-β with high sensitivity.
- Published
- 2006
- Full Text
- View/download PDF
9. Concordance of Alzheimer's Disease-Related Biomarkers Between Intraventricular and Lumbar Cerebrospinal Fluid in Idiopathic Normal Pressure Hydrocephalus.
- Author
-
Lukkarinen H, Vanninen A, Tesseur I, Pemberton D, Van Der Ark P, Kokkola T, Herukka SK, Rauramaa T, Hiltunen M, Blennow K, Zetterberg H, and Leinonen V
- Subjects
- Humans, tau Proteins cerebrospinal fluid, Amyloid beta-Peptides cerebrospinal fluid, Biomarkers cerebrospinal fluid, Peptide Fragments cerebrospinal fluid, Alzheimer Disease diagnosis, Alzheimer Disease cerebrospinal fluid, Hydrocephalus, Normal Pressure cerebrospinal fluid
- Abstract
Background: Alzheimer's disease cerebrospinal fluid (CSF) biomarkers amyloid-β 1-42 (Aβ42), total tau (T-tau), and phosphorylated tau 181 (P-tau181) are widely used. However, concentration gradient of these biomarkers between intraventricular (V-CSF) and lumbar CSF (L-CSF) has been demonstrated in idiopathic normal pressure hydrocephalus (iNPH), potentially affecting clinical utility., Objective: Here we aim to provide conversion factors for clinical and research use between V-CSF and L-CSF., Methods: Altogether 138 iNPH patients participated. L-CSF samples were obtained prior to shunt surgery. Intraoperative V-CSF samples were obtained from 97 patients. Post-operative follow-up L- and V-CSF (shunt reservoir) samples of 41 patients were obtained 1-73 months after surgery and then after 3, 6, and 18 months. CSF concentrations of Aβ42, T-tau, and P-tau181 were analyzed using commercial ELISA assays., Results: Preoperative L-CSF Aβ42, T-tau, and P-tau181 correlated to intraoperative V-CSF (ρ= 0.34-0.55, p < 0.001). Strong correlations were seen between postoperative L- and V-CSF for all biomarkers in every follow-up sampling point (ρs Aβ42: 0.77-0.88, T-tau: 0.91-0.94, P-tau181: 0.94-0.96, p < 0.0001). Regression equations were determined for intraoperative V- and preoperative L-CSF (Aβ42: V-CSF = 185+0.34*L-CSF, T-tau: Ln(V-CSF) = 3.11+0.49*Ln(L-CSF), P-tau181: V-CSF = 8.2+0.51*L-CSF), and for postoperative V- and L-CSF (Aβ42: V-CSF = 86.7+0.75*L-CSF, T-tau: V-CSF = 86.9+0.62*L-CSF, P-tau181: V-CSF = 2.6+0.74*L-CSF)., Conclusion: Aβ42, T-tau, and P-tau181 correlate linearly in-between V- and L-CSF, even stronger after CSF shunt surgery. Equations presented here, provide a novel tool to use V-CSF for diagnostic and prognostic entities relying on the L-CSF concentrations and can be applicable to clinical use when L-CSF samples are not available or less invasively obtained shunt reservoir samples should be interpreted.
- Published
- 2023
- Full Text
- View/download PDF
10. Time Trends of Cerebrospinal Fluid Biomarkers of Neurodegeneration in Idiopathic Normal Pressure Hydrocephalus.
- Author
-
Lukkarinen H, Tesseur I, Pemberton D, Van Der Ark P, Timmers M, Slemmon R, Janssens L, Streffer J, Van Nueten L, Bottelbergs A, Rauramaa T, Koivisto AM, Herukka SK, Korhonen VE, Junkkari A, Hiltunen M, Engelborghs S, Blennow K, Zetterberg H, Kolb HC, and Leinonen V
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease diagnosis, Biomarkers cerebrospinal fluid, Cerebrospinal Fluid Shunts, Female, Humans, Hydrocephalus, Normal Pressure pathology, Hydrocephalus, Normal Pressure surgery, Male, Middle Aged, Phosphorylation, Regression Analysis, Risk Assessment, Alzheimer Disease cerebrospinal fluid, Amyloid beta-Peptides cerebrospinal fluid, Hydrocephalus, Normal Pressure cerebrospinal fluid, Neurofilament Proteins cerebrospinal fluid, Peptide Fragments cerebrospinal fluid, tau Proteins cerebrospinal fluid
- Abstract
Background: Longitudinal changes in cerebrospinal fluid (CSF) biomarkers are seldom studied. Furthermore, data on biomarker gradient between lumbar (L-) and ventricular (V-) compartments seems to be discordant., Objective: To examine alteration of CSF biomarkers reflecting Alzheimer's disease (AD)-related amyloid-β (Aβ) aggregation, tau pathology, neurodegeneration, and early synaptic degeneration by CSF shunt surgery in idiopathic normal pressure hydrocephalus (iNPH) in relation to AD-related changes in brain biopsy. In addition, biomarker levels in L- and V-CSF were compared., Methods: L-CSF was collected prior to shunt placement and, together with V-CSF, 3-73 months after surgery. Thereafter, additional CSF sampling took place at 3, 6, and 18 months after the baseline sample from 26 iNPH patients with confirmed Aβ plaques in frontal cortical brain biopsy and 13 iNPH patients without Aβ pathology. CSF Amyloid-β42 (Aβ42), total tau (T-tau), phosphorylated tau (P-tau181), neurofilament light (NFL), and neurogranin (NRGN) were analyzed with customized ELISAs., Results: All biomarkers but Aβ42 increased notably by 140-810% in L-CSF after CSF diversion and then stabilized. Aβ42 instead showed divergent longitudinal decrease between Aβ-positive and -negative patients in L-CSF, and thereafter increase in Aβ-negative iNPH patients in both L- and V-CSF. All five biomarkers correlated highly between V-CSF and L-CSF (Aβ42 R = 0.87, T-tau R = 0.83, P-tau R = 0.92, NFL R = 0.94, NRGN R = 0.9; all p < 0.0001) but were systematically lower in V-CSF (Aβ42 14 %, T-tau 22%, P-tau 20%, NFL 32%, NRGN 19%). With APOE genotype-grouping, only Aβ42 showed higher concentration in non-carriers of allele ɛ4., Conclusion: Longitudinal follow up shows that after an initial post-surgery increase, T-tau, P-tau, and NRGN are stable in iNPH patients regardless of brain biopsy Aβ pathology, while NFL normalized toward its pre-shunt levels. Aβ42 as biomarker seems to be the least affected by the surgical procedure or shunt and may be the best predictor of AD risk in iNPH patients. All biomarker concentrations were lower in V- than L-CSF yet showing strong correlations.
- Published
- 2021
- Full Text
- View/download PDF
11. Long-term safety and tolerability of atabecestat (JNJ-54861911), an oral BACE1 inhibitor, in early Alzheimer's disease spectrum patients: a randomized, double-blind, placebo-controlled study and a two-period extension study.
- Author
-
Novak G, Streffer JR, Timmers M, Henley D, Brashear HR, Bogert J, Russu A, Janssens L, Tesseur I, Tritsmans L, Van Nueten L, and Engelborghs S
- Subjects
- Amyloid beta-Peptides, Aspartic Acid Endopeptidases, Double-Blind Method, Humans, Pyridines, Thiazines, Treatment Outcome, Alzheimer Disease drug therapy, Amyloid Precursor Protein Secretases
- Abstract
Background: Atabecestat, a potent brain-penetrable inhibitor of BACE1 activity that reduces CSF amyloid beta (Aβ), was developed for oral treatment for Alzheimer's disease (AD). The long-term safety and effect of atabecestat on cognitive performance in participants with predementia AD in two phase 2 studies were assessed., Methods: In the placebo-controlled double-blind parent ALZ2002 study, participants aged 50 to 85 years were randomized (1:1:1) to placebo or atabecestat 10 or 50 mg once daily (later reduced to 5 and 25 mg) for 6 months. Participants entered ALZ2004, a 12-month treatment extension with placebo or atabecestat 10 or 25 mg, followed by an open-label phase. Safety, changes in CSF biomarker levels, brain volume, and effects on cognitive performance were assessed., Results: Of 114 participants randomized in ALZ2002, 99 (87%) completed, 90 entered the ALZ2004 double-blind phase, and 77 progressed to the open-label phase. CSF Aβ fragments and sAPPβ were reduced dose-proportionately. Decreases in whole brain and hippocampal volumes were greater in participants with mild cognitive impairment (MCI) due to AD than in preclinical AD, but were not affected by treatment. In ALZ2004, change from baseline in RBANS trended toward worse scores for atabecestat versus placebo. Elevated liver enzyme adverse events reported in 12 participants on atabecestat resulted in dosage modification and increased frequency of safety monitoring. Treatment discontinuation normalized ALT or AST in all except one with pretreatment elevation, which remained mildly elevated. No case met ALT/AST > 3× ULN and total bilirubin > 2× ULN (Hy's law)., Conclusion: Atabecestat was associated with trend toward declines in cognition, and elevation of liver enzymes., Trial Registration: ALZ2002: ClinicalTrials.gov, NCT02260674, registered October 9, 2014; ALZ2004: ClinicalTrials.gov, NCT02406027, registered April 1, 2015.
- Published
- 2020
- Full Text
- View/download PDF
12. Relevance of the interplay between amyloid and tau for cognitive impairment in early Alzheimer's disease.
- Author
-
Timmers M, Tesseur I, Bogert J, Zetterberg H, Blennow K, Börjesson-Hanson A, Baquero M, Boada M, Randolph C, Tritsmans L, Van Nueten L, Engelborghs S, and Streffer JR
- Subjects
- Aged, Alzheimer Disease diagnosis, Biomarkers cerebrospinal fluid, Cross-Sectional Studies, Female, Humans, Male, Middle Aged, Neuropsychological Tests, Alzheimer Disease psychology, Amyloid beta-Peptides cerebrospinal fluid, Cognition, Peptide Fragments cerebrospinal fluid, tau Proteins cerebrospinal fluid
- Abstract
Amyloid β (Aβ) and tau are key hallmark features of Alzheimer's disease (AD) neuropathology. The interplay of Aβ and tau for cognitive impairment in early AD was examined with cross-sectional analysis, measured by cerebrospinal fluid biomarkers (Aβ
1-42 , total tau [t-tau], and phosphorylated tau [p-tau181P]), and on cognitive performance by the repeatable battery for assessment of neuropsychological status (RBANS). Participants (n = 246) included cognitively normal (Aβ-), mild cognitively impaired (Aβ-), preclinical AD (Aβ+), and prodromal AD (Aβ+). Overall, cognitive scores (RBANS total scale score) had a moderate negative correlation to t-tau (n = 246; r = -0.434; p < 0.001) and p-tau181P (r = -0.389; p < 0.001). When classified by Aβ status, this correlation to t-tau was applicable only in Aβ+ participants (n = 139; r = -0.451, p < 0.001) but not Aβ- participants (n = 107; r = 0.137, p = 0.16), with identical findings for p-tau. Both tau (p < 0.0001) and interaction of Aβ1-42 with tau (p = 0.006) affected RBANS, but not Aβ1-42 alone. Cognitive/memory performance correlated well with cerebrospinal fluid tau levels across early stages of AD, although the correlation is Aβ dependent., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
13. Pharmacodynamics of atabecestat (JNJ-54861911), an oral BACE1 inhibitor in patients with early Alzheimer's disease: randomized, double-blind, placebo-controlled study.
- Author
-
Timmers M, Streffer JR, Russu A, Tominaga Y, Shimizu H, Shiraishi A, Tatikola K, Smekens P, Börjesson-Hanson A, Andreasen N, Matias-Guiu J, Baquero M, Boada M, Tesseur I, Tritsmans L, Van Nueten L, and Engelborghs S
- Subjects
- Administration, Oral, Aged, Alzheimer Disease cerebrospinal fluid, Asian People, Biomarkers cerebrospinal fluid, Double-Blind Method, Female, Humans, Male, Pyridines administration & dosage, Thiazines administration & dosage, Treatment Outcome, White People, tau Proteins cerebrospinal fluid, Alzheimer Disease drug therapy, Amyloid Precursor Protein Secretases antagonists & inhibitors, Amyloid beta-Peptides cerebrospinal fluid, Aspartic Acid Endopeptidases antagonists & inhibitors, Peptide Fragments cerebrospinal fluid, Pyridines pharmacology, Thiazines pharmacology
- Abstract
Background: β-Secretase enzyme (BACE) inhibition has been proposed as a priority treatment mechanism for Alzheimer's disease (AD), but treatment initiation may need to be very early. We present proof of mechanism of atabecestat (also known as JNJ-54861911), an oral BACE inhibitor for the treatment of AD, in Caucasian and Japanese populations with early AD who do not show signs of dementia., Methods: In two similarly designed phase I studies, a sample of amyloid-positive elderly patients comprising 45 Caucasian patients with early AD diagnosed as preclinical AD (n = 15, Clinical Dementia Rating [CDR] = 0) or with mild cognitive impairment due to AD (n = 30, CDR = 0.5) and 18 Japanese patients diagnosed as preclinical AD (CDR-J = 0) were randomized 1:1:1 to atabecestat 10 or 50 mg or placebo (n = 6-8/treatment) daily for 4 weeks. Safety, pharmacokinetics (PK), and pharmacodynamics (PD) (i.e., reduction of cerebrospinal fluid [CSF] amyloid beta 1-40 [Aβ
1-40 ] levels [primary endpoint] and effect on other AD biomarkers) of atabecestat were evaluated., Results: In both populations, atabecestat was well tolerated and characterized by linear PK and high central nervous system penetrance of unbound drug. Atabecestat significantly reduced CSF Aβ1-40 levels from baseline at day 28 in both the 10-mg (67-68%) and 50-mg (87-90%) dose groups compared with placebo. For Caucasians with early AD, the least squares mean differences (95% CI) were - 69.37 (- 72.25; - 61.50) and - 92.74 (- 100.08; - 85.39), and for Japanese with preclinical AD, they were - 62.48 (- 78.32; - 46.64) and - 80.81 (- 96.13; - 65.49), respectively. PK/PD model simulations confirmed that once-daily 10 mg and 50 mg atabecestat can attain 60-70% and 90% Aβ1-40 reductions, respectively. The trend of the reduction was similar across the Aβ1-37 , Aβ1-38 , and Aβ1-42 fragments in both atabecestat dose groups, consistent with Aβ1-40 . CSF amyloid precursor protein fragment (sAPPβ) levels declined from baseline, regardless of patient population, whereas CSF sAPPα levels increased compared with placebo. There were no relevant changes in either CSF total tau or phosphorylated tau 181P over a 4-week treatment period., Conclusions: JNJ-54861911 at 10 and 50 mg daily doses after 4 weeks resulted in mean CSF Aβ1-40 reductions of 67% and up to 90% in both Caucasian and Japanese patients with early stage AD, confirming results in healthy elderly adults., Trial Registration: ALZ1005: ClinicalTrials.gov, NCT01978548. Registered on 7 November 2013. ALZ1008: ClinicalTrials.gov, NCT02360657. Registered on 10 February 2015.- Published
- 2018
- Full Text
- View/download PDF
14. S-[18F]THK-5117-PET and [11C]PIB-PET Imaging in Idiopathic Normal Pressure Hydrocephalus in Relation to Confirmed Amyloid-β Plaques and Tau in Brain Biopsies.
- Author
-
Leinonen V, Rauramaa T, Johansson J, Bottelbergs A, Tesseur I, van der Ark P, Pemberton D, Koivisto AM, Jääskeläinen JE, Hiltunen M, Herukka SK, Blennow K, Zetterberg H, Jokinen P, Rokka J, Helin S, Haaparanta-Solin M, Solin O, Okamura N, Kolb HC, and Rinne JO
- Subjects
- Aged, Aged, 80 and over, Brain Mapping, Female, Humans, Hydrocephalus, Normal Pressure cerebrospinal fluid, Magnetic Resonance Imaging, Male, Middle Aged, Positron-Emission Tomography, Aniline Compounds pharmacokinetics, Brain diagnostic imaging, Brain drug effects, Brain metabolism, Hydrocephalus, Normal Pressure diagnostic imaging, Plaque, Amyloid metabolism, Quinolines pharmacokinetics, Thiazoles pharmacokinetics, tau Proteins metabolism
- Abstract
Background: Detection of pathological tau aggregates could facilitate clinical diagnosis of Alzheimer's disease (AD) and monitor drug effects in clinical trials. S-[18F]THK-5117 could be a potential tracer to detect pathological tau deposits in brain. However, no previous study have correlated S-[18F]THK-5117 uptake in PET with brain biopsy verified tau pathology in vivo., Objective: Here we aim to evaluate the association between cerebrospinal fluid (CSF) AD biomarkers, S-[18F]THK-5117, and [11C]PIB PET against tau and amyloid lesions in brain biopsy., Methods: Fourteen patients with idiopathic normal pressure hydrocephalus (iNPH) with previous shunt surgery including right frontal cortical brain biopsy and CSF Aβ1 - 42, total tau, and P-tau181 measures, underwent brain MRI, [11C]PIB PET, and S-[18F]THK-5117 PET imaging., Results: Seven patients had amyloid-β (Aβ, 4G8) plaques, two both Aβ and phosphorylated tau (Pτ, AT8) and one only Pτ in biopsy. As expected, increased brain biopsy Aβ was well associated with higher [11C]PIB uptake in PET. However, S-[18F]THK-5117 uptake did not show any statistically significant correlation with either brain biopsy Pτ or CSF P-tau181 or total tau., Conclusions: S-[18F]THK-5117 lacked clear association with neuropathologically verified tau pathology in brain biopsy probably, at least partially, due to off-target binding. Further studies with larger samples of patients with different tau tracers are urgently needed. The detection of simultaneous Aβ and tau pathology in iNPH is important since that may indicate poorer and especially shorter response for CSF shunt surgery compared with no pathology.
- Published
- 2018
- Full Text
- View/download PDF
15. Deficiency in Neuronal TGF-β Signaling Leads to Nigrostriatal Degeneration and Activation of TGF-β Signaling Protects against MPTP Neurotoxicity in Mice.
- Author
-
Tesseur I, Nguyen A, Chang B, Li L, Woodling NS, Wyss-Coray T, and Luo J
- Subjects
- Animals, Cell Survival genetics, Gait Disorders, Neurologic chemically induced, Gait Disorders, Neurologic physiopathology, Maze Learning, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neurodegenerative Diseases chemically induced, Postural Balance, Protein Serine-Threonine Kinases genetics, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta genetics, MPTP Poisoning physiopathology, Neostriatum physiopathology, Neurodegenerative Diseases physiopathology, Signal Transduction, Substantia Nigra physiopathology, Transforming Growth Factor beta deficiency
- Abstract
Transforming growth factor-β (TGF-β) plays an important role in the development and maintenance of embryonic dopaminergic (DA) neurons in the midbrain. To study the function of TGF-β signaling in the adult nigrostriatal system, we generated transgenic mice with reduced TGF-β signaling in mature neurons. These mice display age-related motor deficits and degeneration of the nigrostriatal system. Increasing TGF-β signaling in the substantia nigra through adeno-associated virus expressing a constitutively active type I receptor significantly reduces 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration and motor deficits. These results suggest that TGF-β signaling is critical for adult DA neuron survival and that modulating this signaling pathway has therapeutic potential in Parkinson disease. SIGNIFICANCE STATEMENT We show that reducing Transforming growth factor-β (TGF-β) signaling promotes Parkinson disease-related pathologies and motor deficits, and increasing TGF-β signaling reduces neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, a parkinsonism-inducing agent. Our results provide a rationale to pursue a means of increasing TGF-β signaling as a potential therapy for Parkinson's disease., (Copyright © 2017 the authors 0270-6474/17/374584-09$15.00/0.)
- Published
- 2017
- Full Text
- View/download PDF
16. Hallmarks of Alzheimer's Disease in Stem-Cell-Derived Human Neurons Transplanted into Mouse Brain.
- Author
-
Espuny-Camacho I, Arranz AM, Fiers M, Snellinx A, Ando K, Munck S, Bonnefont J, Lambot L, Corthout N, Omodho L, Vanden Eynden E, Radaelli E, Tesseur I, Wray S, Ebneth A, Hardy J, Leroy K, Brion JP, Vanderhaeghen P, and De Strooper B
- Subjects
- Alzheimer Disease diagnosis, Animals, Cell Death physiology, Humans, Mice, Phosphorylation, Alzheimer Disease pathology, Brain metabolism, Brain pathology, Cell Differentiation physiology, Neurites metabolism, Neurons metabolism, Pluripotent Stem Cells cytology, tau Proteins metabolism
- Abstract
Human pluripotent stem cells (PSCs) provide a unique entry to study species-specific aspects of human disorders such as Alzheimer's disease (AD). However, in vitro culture of neurons deprives them of their natural environment. Here we transplanted human PSC-derived cortical neuronal precursors into the brain of a murine AD model. Human neurons differentiate and integrate into the brain, express 3R/4R Tau splice forms, show abnormal phosphorylation and conformational Tau changes, and undergo neurodegeneration. Remarkably, cell death was dissociated from tangle formation in this natural 3D model of AD. Using genome-wide expression analysis, we observed upregulation of genes involved in myelination and downregulation of genes related to memory and cognition, synaptic transmission, and neuron projection. This novel chimeric model for AD displays human-specific pathological features and allows the analysis of different genetic backgrounds and mutations during the course of the disease., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
17. BACE1 Dynamics Upon Inhibition with a BACE Inhibitor and Correlation to Downstream Alzheimer's Disease Markers in Elderly Healthy Participants.
- Author
-
Timmers M, Barão S, Van Broeck B, Tesseur I, Slemmon J, De Waepenaert K, Bogert J, Shaw LM, Engelborghs S, Moechars D, Mercken M, Van Nueten L, Tritsmans L, de Strooper B, and Streffer JR
- Subjects
- Aged, Alzheimer Disease cerebrospinal fluid, Apolipoprotein E4 genetics, Biomarkers cerebrospinal fluid, Double-Blind Method, Female, Follow-Up Studies, Humans, Male, Middle Aged, Phosphorylation drug effects, Protease Inhibitors adverse effects, Protease Inhibitors pharmacokinetics, Amyloid Precursor Protein Secretases antagonists & inhibitors, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Peptides cerebrospinal fluid, Aspartic Acid Endopeptidases antagonists & inhibitors, Aspartic Acid Endopeptidases metabolism, Protease Inhibitors therapeutic use, tau Proteins cerebrospinal fluid
- Abstract
The β-site amyloid-β protein precursor (AβPP) cleaving enzyme-1 (BACE1) is the rate limiting enzyme in the generation of amyloid-β peptide (Aβ) from AβPP, one of the major pathways in Alzheimer's disease (AD) pathology. Increased BACE1 levels and activity have been reported in the brain of patients with sporadic AD. Therefore, changes of BACE1 levels in the cerebrospinal fluid (CSF) have also been investigated as a possible biomarker of the disease. We analyzed BACE1 levels in CSF of elderly healthy participants before and after chronic treatment with a BACE inhibitor (BACEi) and evaluated the correlation between BACE1 levels and downstream AD markers. Overall, BACE1 CSF levels showed strong correlations to all downstream AD markers investigated. This is the first reported finding that shows BACE1 levels in CSF were well correlated to its end product Aβ1 - 42. As previously described, BACE1 levels were strongly correlated to total-tau and phosphorylated tau levels in CSF. Generally, chronic BACE inhibition did not influence BACE1 CSF protein levels. Follow-up studies including early-stage AD pathophysiology and prodromal AD patients will help to understand the importance of measuring BACE1 routinely in daily clinical practice and AD clinical trials.
- Published
- 2017
- Full Text
- View/download PDF
18. Regulation of amyloid precursor protein processing by serotonin signaling.
- Author
-
Pimenova AA, Thathiah A, De Strooper B, and Tesseur I
- Subjects
- Adenylyl Cyclases metabolism, Alzheimer Disease metabolism, Alzheimer Disease physiopathology, Animals, Arrestins metabolism, Casein Kinase II metabolism, Cell Line, Tumor, Cyclic AMP metabolism, GTP-Binding Proteins metabolism, Humans, Inositol Polyphosphate 5-Phosphatases, Mice, Mice, Transgenic metabolism, Mice, Transgenic physiology, Phosphoric Monoester Hydrolases metabolism, Proteolysis, Receptors, Serotonin, 5-HT4 metabolism, Type C Phospholipases metabolism, beta-Arrestins, src-Family Kinases metabolism, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Peptides metabolism, Serotonin metabolism, Signal Transduction physiology
- Abstract
Proteolytic processing of the amyloid precursor protein (APP) by the β- and γ-secretases releases the amyloid-β peptide (Aβ), which deposits in senile plaques and contributes to the etiology of Alzheimer's disease (AD). The α-secretase cleaves APP in the Aβ peptide sequence to generate soluble APPα (sAPPα). Upregulation of α-secretase activity through the 5-hydroxytryptamine 4 (5-HT4) receptor has been shown to reduce Aβ production, amyloid plaque load and to improve cognitive impairment in transgenic mouse models of AD. Consequently, activation of 5-HT4 receptors following agonist stimulation is considered to be a therapeutic strategy for AD treatment; however, the signaling cascade involved in 5-HT4 receptor-stimulated proteolysis of APP remains to be determined. Here we used chemical and siRNA inhibition to identify the proteins which mediate 5-HT4d receptor-stimulated α-secretase activity in the SH-SY5Y human neuronal cell line. We show that G protein and Src dependent activation of phospholipase C are required for α-secretase activity, while, unexpectedly, adenylyl cyclase and cAMP are not involved. Further elucidation of the signaling pathway indicates that inositol triphosphate phosphorylation and casein kinase 2 activation is also a prerequisite for α-secretase activity. Our findings provide a novel route to explore the treatment of AD through 5-HT4 receptor-induced α-secretase activation.
- Published
- 2014
- Full Text
- View/download PDF
19. Assay to probe proteolytic processing of Notch by γ-secretase.
- Author
-
Serneels L, Tesseur I, and De Strooper B
- Subjects
- Animals, Blotting, Western methods, HEK293 Cells, Humans, Mice, Receptors, Notch analysis, Receptors, Notch genetics, Transfection methods, Amyloid Precursor Protein Secretases metabolism, Proteolysis, Receptors, Notch metabolism
- Abstract
With the increasing appreciation of the role of Notch in development and disease, measuring its cleavage and signaling activity in cellular systems has become important. Here we describe a cell-based method to analyze the cleavage of Notch at the S3 site by γ-secretase. HEK cells are transfected with an N-terminal truncated and myc-labeled mNotchΔE construct which can be easily and quantitatively detected by western blotting.
- Published
- 2014
- Full Text
- View/download PDF
20. Dose-dependent improvements in learning and memory deficits in APPPS1-21 transgenic mice treated with the orally active Aβ toxicity inhibitor SEN1500.
- Author
-
Lo AC, Tesseur I, Scopes DI, Nerou E, Callaerts-Vegh Z, Vermaercke B, Treherne JM, De Strooper B, and D'Hooge R
- Subjects
- Administration, Oral, Alzheimer Disease genetics, Amyloid beta-Protein Precursor genetics, Aniline Compounds chemistry, Aniline Compounds pharmacology, Aniline Compounds therapeutic use, Animals, Avoidance Learning drug effects, Disease Models, Animal, Gene Expression Regulation drug effects, Humans, Maze Learning drug effects, Mice, Mice, Transgenic, Mutation genetics, Nitriles chemistry, Presenilin-1 genetics, Pyrimidines administration & dosage, Pyrimidines chemistry, Pyrimidines pharmacology, Pyrimidines therapeutic use, Synaptophysin metabolism, Taste drug effects, Alzheimer Disease complications, Amyloid beta-Peptides antagonists & inhibitors, Learning Disabilities drug therapy, Learning Disabilities etiology, Memory Disorders etiology, Nitriles administration & dosage, Peptide Fragments antagonists & inhibitors
- Abstract
In the Alzheimer's disease (AD) brain, accumulation of Aβ1-42 peptides is suggested to initiate a cascade of pathological events. To date, no treatments are available that can reverse or delay AD-related symptoms in patients. In the current study, we introduce a new Aβ toxicity inhibitor, SEN1500, which in addition to its block effect on Aβ1-42 toxicity in synaptophysin assays, can be administered orally and cross the blood-brain barrier without adverse effects in mice. In a different set of animals, APPPS1-21 mice were fed with three different doses of SEN1500 (1 mg/kg, 5 mg/kg and 20 mg/kg) for a period of 5 months. Cognition was assessed in a variety of behavioral tests (Morris water maze, social recognition, conditioned taste aversion and passive avoidance). Results suggest a positive effect on cognition with 20 mg/kg SEN1500 compared to control APPPS1-21 mice. However, no changes in soluble or insoluble Aβ1-40 and Aβ1-42 were detected in the brains of SEN1500-fed mice. SEN1500 also attenuated the effect of Aβ1-42 on synaptophysin levels in mouse cortical neurons, which indicated that the compound blocked the synaptic toxicity of Aβ1-42. In vitro and in vivo effects presented here suggest that SEN1500 could be an interesting AD therapeutic., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
21. When the dust settles: what did we learn from the bexarotene discussion?
- Author
-
Tesseur I and De Strooper B
- Abstract
With 27 million people affected by Alzheimer's disease (AD), any proposal of a novel avenue for drug development is hot news. When Cramer and colleagues proposed last year that they could tackle AD pathology in an AD mouse model with bexarotene, a drug already in use in the clinic for other diseases, the news was covered worldwide by the popular press. Apolipoprotein E4 is the strongest genetic risk factor for AD and bexarotene appeared to exert spectacular effects on AD pathology when tested in APP/PS1 transgenic mice. One year later the slumbering discussion on the use of bexarotene in AD exploded in a flurry of papers. Four papers question the initial optimistic claims, while two others can only partially support the original work. We summarize here the available data and try to make sense out of the controversy. The major question is what we can learn from the experiments and what these studies imply for the further development of bexarotene in the clinic.
- Published
- 2013
- Full Text
- View/download PDF
22. Chronic 5-HT4 receptor activation decreases Aβ production and deposition in hAPP/PS1 mice.
- Author
-
Tesseur I, Pimenova AA, Lo AC, Ciesielska M, Lichtenthaler SF, De Maeyer JH, Schuurkes JA, D'Hooge R, and De Strooper B
- Subjects
- Alzheimer Disease drug therapy, Alzheimer Disease enzymology, Alzheimer Disease metabolism, Amyloid beta-Protein Precursor antagonists & inhibitors, Amyloid beta-Protein Precursor biosynthesis, Amyloid beta-Protein Precursor genetics, Animals, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Amyloid beta-Peptides antagonists & inhibitors, Amyloid beta-Peptides biosynthesis, Amyloid beta-Peptides metabolism, Presenilin-1 genetics, Receptors, Serotonin, 5-HT4 metabolism, Serotonin Receptor Agonists pharmacology, Serotonin Receptor Agonists therapeutic use
- Abstract
Lowering the production and accumulation of Aβ has been explored as treatment for Alzheimer's disease (AD), because Aβ is postulated to play an important role in the pathogenesis of AD. 5-HT4 receptors are an interesting drug target in this regard, as their activation might stimulate α-secretase processing, which increases sAPPα and reduces Aβ, at least according to the central dogma in APP processing. Here we describe a novel high-affinity 5-HT4 receptor agonist SSP-002392 that, in cultured human neuroblastoma cells, potently increases the levels of cAMP and sAPPα at 100-fold lower concentrations than the effective concentrations of prucalopride, a known selective 5-HT4 receptor agonist. Chronic administration of this compound in a hAPP/PS1 mouse model of Alzheimer's disease decreased soluble and insoluble Aβ in hippocampus, but the potential mechanisms underlying these observations seem to be complex. We found no evidence for direct α-secretase stimulation in the brain in vivo, but observed decreased APP and BACE-1 expression and elevated astroglia and microglia responses. Taken together these results provide support for a potential disease-modifying aspect when stimulating central 5-HT4 receptors; however, the complexity of the phenomena warrants further research., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
23. Comment on "ApoE-directed therapeutics rapidly clear β-amyloid and reverse deficits in AD mouse models".
- Author
-
Tesseur I, Lo AC, Roberfroid A, Dietvorst S, Van Broeck B, Borgers M, Gijsen H, Moechars D, Mercken M, Kemp J, D'Hooge R, and De Strooper B
- Subjects
- Animals, Male, Alzheimer Disease drug therapy, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Apolipoproteins E metabolism, Brain metabolism, Tetrahydronaphthalenes pharmacology, Tetrahydronaphthalenes therapeutic use
- Abstract
Cramer et al. (Reports, 23 March 2012, p. 1503; published online 9 February 2012) tested bexarotene as a potential β-amyloid-lowering drug for Alzheimer's disease (AD). We were not able to reproduce the described effects in several animal models. Drug formulation appears very critical. Our data call for extreme caution when considering this compound for use in AD patients.
- Published
- 2013
- Full Text
- View/download PDF
24. Amyloid and tau neuropathology differentially affect prefrontal synaptic plasticity and cognitive performance in mouse models of Alzheimer's disease.
- Author
-
Lo AC, Iscru E, Blum D, Tesseur I, Callaerts-Vegh Z, Buée L, De Strooper B, Balschun D, and D'Hooge R
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease psychology, Amyloid beta-Protein Precursor genetics, Animals, Cognition Disorders genetics, Cognition Disorders psychology, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neuronal Plasticity physiology, Plaque, Amyloid genetics, Plaque, Amyloid pathology, Presenilin-1 genetics, Synapses genetics, Alzheimer Disease pathology, Cognition Disorders pathology, Disease Models, Animal, Prefrontal Cortex pathology, Synapses pathology, tau Proteins genetics
- Abstract
Alzheimer's disease (AD) is a consequence of degenerative brain pathology with amyloid plaque deposition and neurofibrillary tangle formation. These distinct aspects of AD neuropathology have been suggested to induce a cascade of pathological events ultimately leading to neurodegeneration as well as cognitive and behavioral decline. Amyloid and tau neuropathology is known to develop along distinct stages and affect parts of the brain differentially. In this study, we examined two mouse AD lines (AβPPPS1-21 and Tau22 mice), which mimic different partial aspects of AD pathology, at comparable stages of their pathology. Since prefrontal cortex (PFC) is one of the first regions to be affected in clinical AD, we compared long-term potentiation (LTP) of synaptic responses in medial PFC of AβPPPS1-21 and Tau22 mice. Frontal LTP was impaired in AβPPPS1-21 mice, but not in Tau22 mice. Consequently, we observed different behavioral defects between AβPPPS1-21 and Tau22 animals. Apart from spatial learning deficits, AβPPPS1-21 transgenic mice were impaired in fear learning, aversion learning, and extinction learning, whereas THY-Tau22 were impaired in appetitive responding. Discriminant function analysis identified critical behavioral variables that differentiated AβPPPS1-21 and THY-Tau22 mice from wild type littermates, and further confirmed that amyloid- versus tau-pathology differentially affects brain function.
- Published
- 2013
- Full Text
- View/download PDF
25. ADAM9 inhibition increases membrane activity of ADAM10 and controls α-secretase processing of amyloid precursor protein.
- Author
-
Moss ML, Powell G, Miller MA, Edwards L, Qi B, Sang QX, De Strooper B, Tesseur I, Lichtenthaler SF, Taverna M, Zhong JL, Dingwall C, Ferdous T, Schlomann U, Zhou P, Griffith LG, Lauffenburger DA, Petrovich R, and Bartsch JW
- Subjects
- ADAM Proteins chemistry, ADAM10 Protein, Biocatalysis drug effects, Cell Line, Tumor, Cell Membrane metabolism, Humans, Membrane Proteins chemistry, Protease Inhibitors pharmacology, Protein Array Analysis, Protein Structure, Tertiary, ADAM Proteins antagonists & inhibitors, ADAM Proteins metabolism, Amyloid Precursor Protein Secretases metabolism, Amyloid beta-Protein Precursor metabolism, Cell Membrane drug effects, Cell Membrane enzymology, Membrane Proteins antagonists & inhibitors, Membrane Proteins metabolism, Peptide Fragments pharmacology
- Abstract
Prodomains of A disintegrin and metalloproteinase (ADAM) metallopeptidases can act as highly specific intra- and intermolecular inhibitors of ADAM catalytic activity. The mouse ADAM9 prodomain (proA9; amino acids 24-204), expressed and characterized from Escherichia coli, is a competitive inhibitor of human ADAM9 catalytic/disintegrin domain with an overall inhibition constant of 280 ± 34 nM and high specificity toward ADAM9. In SY5Y neuroblastoma cells overexpressing amyloid precursor protein, proA9 treatment reduces the amount of endogenous ADAM10 enzyme in the medium while increasing membrane-bound ADAM10, as shown both by Western and activity assays with selective fluorescent peptide substrates using proteolytic activity matrix analysis. An increase in membrane-bound ADAM10 generates higher levels of soluble amyloid precursor protein α in the medium, whereas soluble amyloid precursor protein β levels are decreased, demonstrating that inhibition of ADAM9 increases α-secretase activity on the cell membrane. Quantification of physiological ADAM10 substrates by a proteomic approach revealed that substrates, such as epidermal growth factor (EGF), HER2, osteoactivin, and CD40-ligand, are increased in the medium of BT474 breast tumor cells that were incubated with proA9, demonstrating that the regulation of ADAM10 by ADAM9 applies for many ADAM10 substrates. Taken together, our results demonstrate that ADAM10 activity is regulated by inhibition of ADAM9, and this regulation may be used to control shedding of amyloid precursor protein by enhancing α-secretase activity, a key regulatory step in the etiology of Alzheimer disease.
- Published
- 2011
- Full Text
- View/download PDF
26. Bioactive TGF-beta can associate with lipoproteins and is enriched in those containing apolipoprotein E3.
- Author
-
Tesseur I, Zhang H, Brecht W, Corn J, Gong JS, Yanagisawa K, Michikawa M, Weisgraber K, Huang Y, and Wyss-Coray T
- Subjects
- Animals, Apolipoprotein E3 genetics, Apolipoprotein E4 metabolism, Cell Line, Cells, Cultured, Humans, Lipid Metabolism physiology, Mice, Mice, Knockout, Protein Isoforms metabolism, Protein Structure, Secondary physiology, Protein Structure, Tertiary physiology, Signal Transduction physiology, Transforming Growth Factor beta1 chemistry, Transforming Growth Factor beta1 genetics, Apolipoprotein E3 metabolism, Astrocytes metabolism, Hepatocytes metabolism, Lipoproteins metabolism, Transforming Growth Factor beta1 metabolism
- Abstract
Transforming growth factor-beta1 (TGF-beta1) has central functions in development, tissue maintenance, and repair and has been implicated in major diseases. We discovered that TGF-beta1 contains several amphipathic helices and hydrophobic domains similar to apolipoprotein E (apoE), a protein involved in lipoprotein metabolism. Indeed, TGF-beta1 associates with lipoproteins isolated from human plasma, cultured liver cells, or astrocytes, and its bioactivity was highest in high-density lipoprotein preparations. Importantly, lipoproteins containing the apoE3 isoform had higher TGF-beta levels and bioactivity than those containing apoE4, a major genetic risk factor for atherosclerosis and Alzheimer's disease. Because TGF-beta1 can be protective in these diseases an association with apoE3 may be beneficial. Association of TGF-beta with different types of lipoproteins may facilitate its diffusion, regulate signaling, and offer additional specificity for this important growth factor.
- Published
- 2009
- Full Text
- View/download PDF
27. Collagen VI protects neurons against Abeta toxicity.
- Author
-
Cheng JS, Dubal DB, Kim DH, Legleiter J, Cheng IH, Yu GQ, Tesseur I, Wyss-Coray T, Bonaldo P, and Mucke L
- Subjects
- Alzheimer Disease physiopathology, Amyloid beta-Peptides genetics, Animals, Astrocytes cytology, Astrocytes metabolism, Collagen Type VI genetics, Dentate Gyrus metabolism, Dentate Gyrus pathology, Dentate Gyrus physiopathology, Humans, Mice, Mice, Transgenic, Neurons pathology, Oligonucleotide Array Sequence Analysis, Peptide Fragments genetics, Protein Binding physiology, Signal Transduction physiology, Transforming Growth Factor beta metabolism, Alzheimer Disease metabolism, Alzheimer Disease pathology, Amyloid beta-Peptides metabolism, Collagen Type VI metabolism, Neurons metabolism, Peptide Fragments metabolism
- Abstract
Amyloid-beta (Abeta) peptides, widely presumed to cause Alzheimer's disease, increased mouse neuronal expression of collagen VI through a mechanism involving transforming growth factor signaling. Reduction of collagen VI augmented Abeta neurotoxicity, whereas treatment of neurons with soluble collagen VI blocked the association of Abeta oligomers with neurons, enhanced Abeta aggregation and prevented neurotoxicity. These results identify collagen VI as an important component of the neuronal injury response and demonstrate its neuroprotective potential.
- Published
- 2009
- Full Text
- View/download PDF
28. Genes contributing to prion pathogenesis.
- Author
-
Tamgüney G, Giles K, Glidden DV, Lessard P, Wille H, Tremblay P, Groth DF, Yehiely F, Korth C, Moore RC, Tatzelt J, Rubinstein E, Boucheix C, Yang X, Stanley P, Lisanti MP, Dwek RA, Rudd PM, Moskovitz J, Epstein CJ, Cruz TD, Kuziel WA, Maeda N, Sap J, Ashe KH, Carlson GA, Tesseur I, Wyss-Coray T, Mucke L, Weisgraber KH, Mahley RW, Cohen FE, and Prusiner SB
- Subjects
- Amyloid beta-Protein Precursor genetics, Animals, Gene Dosage, Gene Silencing, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Prions genetics, Receptors, Interleukin-1 Type I genetics, Superoxide Dismutase genetics, Superoxide Dismutase-1, Survival Analysis, Prion Diseases genetics, Prions metabolism
- Abstract
Prion diseases are caused by conversion of a normally folded, non-pathogenic isoform of the prion protein (PrP(C)) to a misfolded, pathogenic isoform (PrP(Sc)). Prion inoculation experiments in mice expressing homologous PrP(C) molecules on different genetic backgrounds displayed different incubation times, indicating that the conversion reaction may be influenced by other gene products. To identify genes that contribute to prion pathogenesis, we analysed incubation times of prions in mice in which the gene product was inactivated, knocked out or overexpressed. We tested 20 candidate genes, because their products either colocalize with PrP, are associated with Alzheimer's disease, are elevated during prion disease, or function in PrP-mediated signalling, PrP glycosylation, or protein maintenance. Whereas some of the candidates tested may have a role in the normal function of PrP(C), our data show that many genes previously implicated in prion replication have no discernible effect on the pathogenesis of prion disease. While most genes tested did not significantly affect survival times, ablation of the amyloid beta (A4) precursor protein (App) or interleukin-1 receptor, type I (Il1r1), and transgenic overexpression of human superoxide dismutase 1 (SOD1) prolonged incubation times by 13, 16 and 19 %, respectively.
- Published
- 2008
- Full Text
- View/download PDF
29. A role for TGF-beta signaling in neurodegeneration: evidence from genetically engineered models.
- Author
-
Tesseur I and Wyss-Coray T
- Subjects
- Animals, Disease Models, Animal, Humans, Mice, Animals, Genetically Modified, Neurodegenerative Diseases metabolism, Signal Transduction physiology, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism
- Abstract
Neurodegenerative diseases including Alzheimer's disease (AD) and Parkinson's disease (PD) afflict growing numbers of people but treatments are not available or ineffective. These diseases are characterized by the loss of specific neuronal populations, the accumulation of protein aggregates inside and sometimes outside neurons, and an activation of immune pathways in the brain. The causes of sporadic forms of AD or PD are not known but it has been postulated that reduced trophic support to neurons together with age dependent increases in cellular stress lead to chronic injury and ultimately the demise of neurons. TGF-betas are neuroprotective factors and organizers of injury responses and as such might have a role in neurodegenerative disease. We review here the evidence mostly from genetically manipulated mice that links the TGF-beta signaling pathway to neuronal phenotypes and neurodegeneration. Although many of these mutant models did not produce overt CNS phenotypes or adult brain were not studied due to embryonic lethality, there is growing support for a role of TGF-beta signaling in neuronal maintenance, function, and degeneration. Future studies will have to determine whether dysregulation of TGF-beta signaling in neurodegenerative diseases is significant and whether this signaling pathway may even be a target for treatment.
- Published
- 2006
- Full Text
- View/download PDF
30. Deficiency in neuronal TGF-beta signaling promotes neurodegeneration and Alzheimer's pathology.
- Author
-
Tesseur I, Zou K, Esposito L, Bard F, Berber E, Can JV, Lin AH, Crews L, Tremblay P, Mathews P, Mucke L, Masliah E, and Wyss-Coray T
- Subjects
- Aging physiology, Alzheimer Disease genetics, Amyloid beta-Peptides genetics, Amyloid beta-Peptides metabolism, Animals, Cells, Cultured, Dendrites metabolism, Dendrites pathology, Gliosis metabolism, Gliosis pathology, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Nerve Degeneration genetics, Neuroblastoma metabolism, Neuroblastoma pathology, Protein Serine-Threonine Kinases, Receptor, Transforming Growth Factor-beta Type II, Receptors, Transforming Growth Factor beta metabolism, Alzheimer Disease metabolism, Alzheimer Disease pathology, Nerve Degeneration metabolism, Nerve Degeneration pathology, Signal Transduction, Transforming Growth Factor beta metabolism
- Abstract
Alzheimer's disease (AD) is characterized by progressive neurodegeneration and cerebral accumulation of the beta-amyloid peptide (Abeta), but it is unknown what makes neurons susceptible to degeneration. We report that the TGF-beta type II receptor (TbetaRII) is mainly expressed by neurons, and that TbetaRII levels are reduced in human AD brain and correlate with pathological hallmarks of the disease. Reducing neuronal TGF-beta signaling in mice resulted in age-dependent neurodegeneration and promoted Abeta accumulation and dendritic loss in a mouse model of AD. In cultured cells, reduced TGF-beta signaling caused neuronal degeneration and resulted in increased levels of secreted Abeta and beta-secretase-cleaved soluble amyloid precursor protein. These results show that reduced neuronal TGF-beta signaling increases age-dependent neurodegeneration and AD-like disease in vivo. Increasing neuronal TGF-beta signaling may thus reduce neurodegeneration and be beneficial in AD.
- Published
- 2006
- Full Text
- View/download PDF
31. Highly sensitive and specific bioassay for measuring bioactive TGF-beta.
- Author
-
Tesseur I, Zou K, Berber E, Zhang H, and Wyss-Coray T
- Subjects
- Alkaline Phosphatase genetics, Animals, Brain Chemistry, CHO Cells, Cell Line, Coculture Techniques methods, Cricetinae, Culture Media chemistry, Female, Fibroblasts chemistry, Fibroblasts cytology, Genes, Reporter genetics, Mice, Mice, Mutant Strains, Sensitivity and Specificity, Signal Transduction physiology, Transforming Growth Factor beta blood, Transforming Growth Factor beta genetics, Biological Assay methods, Transforming Growth Factor beta analysis
- Abstract
Background: Transforming Growth Factor-beta (TGF-beta) regulates key biological processes during development and in adult tissues and has been implicated in many diseases. To study the biological functions of TGF-beta, sensitive, specific, and convenient bioassays are necessary. Here we describe a new cell-based bioassay that fulfills these requirements., Results: Embryonic fibroblasts from Tgfb1-/- mice were stably transfected with a reporter plasmid consisting of TGF-beta responsive Smad-binding elements coupled to a secreted alkaline phosphatase reporter gene (SBE-SEAP). Clone MFB-F11 showed more than 1000-fold induction after stimulation with 1 ng/ml TGF-beta1, and detected as little as 1 pg/ml TGF-beta1. MFB-F11 cells were highly induced by TGF-beta1, TGF-beta2 and TGF-beta3, but did not show induction with related family members activin, nodal, BMP-2 and BMP-6 or with trophic factors bFGF and BDNF. MFB-F11 cells can detect and quantify TGF-beta in biological samples without prior enrichment of TGF-betas, and can detect biologically activated TGF-beta in a cell co-culture system., Conclusion: MFB-F11 cells can be used to rapidly and specifically measure TGF-beta with high sensitivity.
- Published
- 2006
- Full Text
- View/download PDF
32. Small molecule tgf-beta mimetics as potential neuroprotective factors.
- Author
-
Zhang H, Zou K, Tesseur I, and Wyss-Coray T
- Subjects
- Alzheimer Disease drug therapy, Animals, Biomimetic Materials therapeutic use, Cell Line, Dose-Response Relationship, Drug, Drug Evaluation, Preclinical methods, Mice, Mice, Knockout, Neuroprotective Agents therapeutic use, Rats, Transforming Growth Factor beta therapeutic use, Biomimetic Materials pharmacology, Neuroprotective Agents pharmacology, Transforming Growth Factor beta pharmacology
- Abstract
Neurodegenerative and dementing illnesses are becoming an increasing social and economical burden as the number of older people continues to grow in industrialized countries. Current knowledge of the processes leading to these diseases is still limited, and very few effective treatments are available. Because neurodegeneration is associated with an activation of injury and innate immune responses in the brain, drugs that could mimic the beneficial aspects of this response are potential therapeutic candidates. The cytokine transforming growth factor (TGF)-beta1 is an organizer of the brain's response to injury and is known to be neuroprotective. Previous studies from our lab also showed that TGF-beta1 can reduce accumulation of beta-amyloid peptide (Abeta), which appears to be central to Alzheimer's disease (AD) pathogenesis, and we therefore initiated a search for small molecule chemical compounds that could mimic this effect. We report here the identification of several such TGF-beta mimetics detected in an in vitro screen of a library with 5000 chemically diverse compounds. If active in vivo, these mimetics could be developed into candidates for the treatment of neurodegeneration.
- Published
- 2005
- Full Text
- View/download PDF
33. Neuron-specific apolipoprotein e4 proteolysis is associated with increased tau phosphorylation in brains of transgenic mice.
- Author
-
Brecht WJ, Harris FM, Chang S, Tesseur I, Yu GQ, Xu Q, Dee Fish J, Wyss-Coray T, Buttini M, Mucke L, Mahley RW, and Huang Y
- Subjects
- Aging metabolism, Animals, Apolipoprotein E3, Apolipoprotein E4, Apolipoproteins E genetics, Astrocytes cytology, Astrocytes metabolism, Brain cytology, Brain drug effects, Brain Chemistry genetics, Female, Gene Targeting, Glial Fibrillary Acidic Protein genetics, Humans, Kainic Acid pharmacology, Mice, Mice, Transgenic, Neurons cytology, Neurons drug effects, Neurotoxins pharmacology, Peptide Fragments metabolism, Phosphopyruvate Hydratase genetics, Phosphorylation, Promoter Regions, Genetic, Alzheimer Disease genetics, Apolipoproteins E metabolism, Brain metabolism, Neurons metabolism, tau Proteins metabolism
- Abstract
Apolipoprotein E (apoE) is found in amyloid plaques and neurofibrillary tangles (NFTs) in Alzheimer's disease (AD) brains, but its role in their pathogenesis is unclear. Previously, we found C-terminal-truncated fragments of apoE in AD brains and showed that such fragments can cause neurodegeneration and can induce NFT-like inclusions in cultured neuronal cells and in transgenic mice. Here, we analyzed apoE fragmentation in brain tissue homogenates from transgenic mice expressing apoE3 or apoE4 in neurons [neuron-specific enolase (NSE)-apoE] or astrocytes [glial fibrillary acidic protein (GFAP)-apoE] by Western blotting. The C-terminal-truncated fragments of apoE accumulated, in an age-dependent manner, in the brains of NSE-apoE4 and, to a significantly lesser extent, NSE-apoE3 mice; however, no fragments were detected in GFAP-apoE3 or GFAP-apoE4 mice. In NSE-apoE mice, the pattern of apoE fragmentation resembled that seen in AD brains, and the fragmentation was specific for certain brain regions, occurring in the neocortex and hippocampus, which are vulnerable to AD-related neurodegeneration, but not in the less vulnerable cerebellum. Excitotoxic challenge with kainic acid significantly increased apoE fragmentation in NSE-apoE4 but not NSE-apoE3 mice. Phosphorylated tau (p-tau) also accumulated in an age-dependent manner in NSE-apoE4 mice and, to a much lesser extent, in NSE-apoE3 mice but not in GFAP-apoE3 or GFAP-apoE4 mice. Intraneuronal p-tau inclusions in the hippocampus were prominent in 21-month-old NSE-apoE4 mice but barely detectable in NSE-apoE3 mice. Thus, the accumulation of potentially pathogenic C-terminal-truncated fragments of apoE depends on both the isoform and the cellular source of apoE. Neuron-specific proteolytic cleavage of apoE4 is associated with increased phosphorylation of tau and may play a key role in the development of AD-related neuronal deficits.
- Published
- 2004
- Full Text
- View/download PDF
34. Astroglial regulation of apolipoprotein E expression in neuronal cells. Implications for Alzheimer's disease.
- Author
-
Harris FM, Tesseur I, Brecht WJ, Xu Q, Mullendorff K, Chang S, Wyss-Coray T, Mahley RW, and Huang Y
- Subjects
- Animals, Blotting, Western, Butadienes pharmacology, Cell Line, Tumor, Cells, Cultured, Culture Media, Conditioned pharmacology, Enzyme Inhibitors pharmacology, Hippocampus metabolism, Humans, Immunohistochemistry, Mice, Mitogen-Activated Protein Kinases metabolism, Models, Biological, Models, Genetic, Nitriles pharmacology, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Up-Regulation, Alzheimer Disease metabolism, Apolipoproteins E biosynthesis, Astrocytes metabolism, Gene Expression Regulation, Neuroglia metabolism, Neurons metabolism
- Abstract
Although apolipoprotein (apo) E is synthesized in the brain primarily by astrocytes, neurons in the central nervous system express apoE, albeit at lower levels than astrocytes, in response to various physiological and pathological conditions, including excitotoxic stress. To investigate how apoE expression is regulated in neurons, we transfected Neuro-2a cells with a 17-kilobase human apoE genomic DNA construct encoding apoE3 or apoE4 along with upstream and downstream regulatory elements. The baseline expression of apoE was low. However, conditioned medium from an astrocytic cell line (C6) or from apoE-null mouse primary astrocytes increased the expression of both isoforms by 3-4-fold at the mRNA level and by 4-10-fold at the protein level. These findings suggest that astrocytes secrete a factor or factors that regulate apoE expression in neuronal cells. The increased expression of apoE was almost completely abolished by incubating neurons with U0126, an inhibitor of extracellular signal-regulated kinase (Erk), suggesting that the Erk pathway controls astroglial regulation of apoE expression in neuronal cells. Human neuronal precursor NT2/D1 cells expressed apoE constitutively; however, after treatment of these cells with retinoic acid to induce differentiation, apoE expression diminished. Cultured mouse primary cortical and hippocampal neurons also expressed low levels of apoE. Astrocyte-conditioned medium rapidly up-regulated apoE expression in fully differentiated NT2 neurons and in cultured mouse primary cortical and hippocampal neurons. Thus, neuronal expression of apoE is regulated by a diffusible factor or factors released from astrocytes, and this regulation depends on the activity of the Erk kinase pathway in neurons.
- Published
- 2004
- Full Text
- View/download PDF
35. Loss of TGF-beta 1 leads to increased neuronal cell death and microgliosis in mouse brain.
- Author
-
Brionne TC, Tesseur I, Masliah E, and Wyss-Coray T
- Subjects
- Animals, Brain pathology, Cell Death physiology, Cell Survival physiology, Cells, Cultured, Gliosis pathology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Microglia pathology, Neurons pathology, Transforming Growth Factor beta biosynthesis, Transforming Growth Factor beta genetics, Transforming Growth Factor beta1, Brain metabolism, Gliosis metabolism, Microglia metabolism, Neurons metabolism, Transforming Growth Factor beta deficiency
- Abstract
TGF-beta1 is a key regulator of diverse biological processes in many tissues and cell types, but its exact function in the developing and adult mammalian CNS is still unknown. We report that lack of TGF-beta1 expression in neonatal Tgfb1(-/-) mice results in a widespread increase in degenerating neurons accompanied by reduced expression of synaptophysin and laminin and a prominent microgliosis. Lack of TGF-beta1 also strongly reduces survival of primary neurons cultured from Tgfb1(-/-) mice. TGF-beta1 deficiency in adult Tgfb1(-/+) mice results in increased neuronal susceptibility to excitotoxic injury, whereas astroglial overexpression of TGF-beta1 protects adult mice against neurodegeneration in acute, excitotoxic and chronic injury paradigms. This study reveals a nonredundant function for TGF-beta1 in maintaining neuronal integrity and survival of CNS neurons and in regulating microglial activation. Because individual TGF-beta1 expression levels in the brain vary considerably between humans, this finding could have important implications for susceptibility to neurodegeneration.
- Published
- 2003
- Full Text
- View/download PDF
36. Carboxyl-terminal-truncated apolipoprotein E4 causes Alzheimer's disease-like neurodegeneration and behavioral deficits in transgenic mice.
- Author
-
Harris FM, Brecht WJ, Xu Q, Tesseur I, Kekonius L, Wyss-Coray T, Fish JD, Masliah E, Hopkins PC, Scearce-Levie K, Weisgraber KH, Mucke L, Mahley RW, and Huang Y
- Subjects
- Aged, Animals, Apolipoprotein E4, Apolipoproteins E chemistry, Apolipoproteins E metabolism, Brain metabolism, Humans, Hydrolysis, Mice, Mice, Transgenic, Microscopy, Electron, Apolipoproteins E physiology, Behavior, Animal
- Abstract
Apolipoprotein (apo) E4 increases the risk and accelerates the onset of Alzheimer's disease (AD). However, the underlying mechanisms remain to be determined. We previously found that apoE undergoes proteolytic cleavage in AD brains and in cultured neuronal cells, resulting in the accumulation of carboxyl-terminal-truncated fragments of apoE that are neurotoxic. Here we show that this fragmentation is caused by proteolysis of apoE by a chymotrypsin-like serine protease that cleaves apoE4 more efficiently than apoE3. Transgenic mice expressing the carboxyl-terminal-cleaved product, apoE4(Delta272-299), at high levels in the brain died at 2-4 months of age. The cortex and hippocampus of these mice displayed AD-like neurodegenerative alterations, including abnormally phosphorylated tau (p-tau) and Gallyas silver-positive neurons that contained cytosolic straight filaments with diameters of 15-20 nm, resembling preneurofibrillary tangles. Transgenic mice expressing lower levels of the truncated apoE4 survived longer but showed impaired learning and memory at 6-7 months of age. Thus, carboxyl-terminal-truncated fragments of apoE4, which occur in AD brains, are sufficient to elicit AD-like neurodegeneration and behavioral deficits in vivo. Inhibiting their formation might inhibit apoE4-associated neuronal deficits.
- Published
- 2003
- Full Text
- View/download PDF
37. Modelling Alzheimer's disease in multiple transgenic mice.
- Author
-
Dewachter I, Moechars D, van Dorpe J, Tesseur I, Van den Haute C, Spittaels K, and Van Leuven F
- Subjects
- Alzheimer Disease etiology, Amyloid beta-Protein Precursor genetics, Animals, Apolipoprotein E4, Apolipoproteins E genetics, Disease Models, Animal, Humans, Membrane Proteins genetics, Mice, Mice, Transgenic, Mutation, Nerve Degeneration etiology, Nerve Degeneration genetics, Presenilin-1, tau Proteins genetics, Alzheimer Disease genetics
- Abstract
We have reported transgenic mice with neuronal overexpression of the clinical mutant beta-amyloid precursor protein (APP) known as London, which develop an AD-related phenotype [Moechers, Dewachter, Lorent, Reversé, Baekelandt, Nadiu, Tesseur, Spittaels, Van den Haute, Checler, et al. (1999) J. Biol. Chem. 274, 6483-6492]. Characterized early symptoms (3-9 months) include disturbed behaviour, neophobia, aggression, hypersensitivity to kainic acid, hyposensitivity to N-methyl-D-aspartate, defective cognition and memory, and decreased long-term potentiation. Late in life, at 12-15 months, amyloid plaques develop in the brain and correlate with increased levels of beta-amyloid (A beta)40/42 (the 40- and 42-amino-acid forms of A beta). The formation of amyloid plaques is dissociated in time from and not involved in the early phenotype. Hyperphosphorylated protein tau is present but no tangle pathology is observed. In double-transgenic mice, i.e. APP/London x Presenilin 1, the increased production of A beta 42 results in amyloid plaques developing by the age of 6 months. Transgenic mice with overexpression of either human apolipoprotein E4 (ApoE4) or human protein tau in central neurons develop severe axonopathy in the brain and spinal cord. Progressive degeneration of nerves and muscles is demonstrated by motor problems, wasting and premature death. Tau is hyperphosphorylated but there is no formation of filaments or neurofibrillary tangles. The tangle aspect of AD pathology is still missing from all current transgenic amyloid models. Its implementation will require insight into the cellular signalling pathways which regulate the microtubule-stabilizing function by phosphorylation of neuronal tau.
- Published
- 2001
- Full Text
- View/download PDF
38. Prominent axonopathy and disruption of axonal transport in transgenic mice expressing human apolipoprotein E4 in neurons of brain and spinal cord.
- Author
-
Tesseur I, Van Dorpe J, Bruynseels K, Bronfman F, Sciot R, Van Lommel A, and Van Leuven F
- Subjects
- Animals, Apolipoprotein E4, Apolipoproteins E genetics, Axons ultrastructure, Behavior, Animal, Brain pathology, Brain ultrastructure, Humans, Mice, Mice, Transgenic genetics, Neurons ultrastructure, Phosphorylation, Spinal Cord pathology, Spinal Cord ultrastructure, Thy-1 Antigens genetics, Thy-1 Antigens metabolism, tau Proteins metabolism, Apolipoproteins E metabolism, Axonal Transport, Axons pathology, Brain metabolism, Neurons metabolism, Spinal Cord metabolism
- Abstract
The epsilon 4 allele of the human apolipoprotein E gene (ApoE4) constitutes an important genetic risk factor for Alzheimer's disease. Recent experimental evidence suggests that human ApoE is expressed in neurons, in addition to being synthesized in glial cells. Moreover, brain regions in which neurons express ApoE seem to be most vulnerable to neurofibrillary pathology. The hypothesis that the expression pattern of human ApoE might be important for the pathogenesis of Alzheimer's disease was tested by generating transgenic mice that express human ApoE4 in neurons or in astrocytes of the central nervous system. Transgenic mice expressing human ApoE4 in neurons developed axonal degeneration and gliosis in brain and in spinal cord, resulting in reduced sensorimotor capacities. In these mice, axonal dilatations with accumulation of synaptophysin, neurofilaments, mitochondria, and vesicles were documented, suggesting impairment of axonal transport. In contrast, transgenic mice expressing human ApoE4 in astrocytes remained normal throughout life. These results suggest that expression of human ApoE in neurons of the central nervous system could contribute to impaired axonal transport and axonal degeneration. The possible contribution of hyperphosphorylation of protein Tau to the resulting phenotype is discussed.
- Published
- 2000
- Full Text
- View/download PDF
39. Prominent cerebral amyloid angiopathy in transgenic mice overexpressing the london mutant of human APP in neurons.
- Author
-
Van Dorpe J, Smeijers L, Dewachter I, Nuyens D, Spittaels K, Van Den Haute C, Mercken M, Moechars D, Laenen I, Kuiperi C, Bruynseels K, Tesseur I, Loos R, Vanderstichele H, Checler F, Sciot R, and Van Leuven F
- Subjects
- Aging physiology, Amyloid metabolism, Animals, Blood Vessels metabolism, Blood Vessels pathology, Blood Vessels ultrastructure, Cerebral Amyloid Angiopathy metabolism, Cerebrovascular Circulation, Humans, Hypercapnia physiopathology, Immunohistochemistry, Membrane Proteins genetics, Mice, Mice, Transgenic, Microscopy, Electron, Presenilin-1, Transgenes physiology, Amyloid beta-Protein Precursor genetics, Cerebral Amyloid Angiopathy genetics, Cerebral Amyloid Angiopathy pathology, Gene Expression, Mutation physiology
- Abstract
Deposition of amyloid beta-peptide (Abeta) in cerebral vessel walls (cerebral amyloid angiopathy, CAA) is very frequent in Alzheimer's disease and occurs also as a sporadic disorder. Here, we describe significant CAA in addition to amyloid plaques, in aging APP/Ld transgenic mice overexpressing the London mutant of human amyloid precursor protein (APP) exclusively in neurons. The number of amyloid-bearing vessels increased with age, from approximately 10 to >50 per coronal brain section in APP/Ld transgenic mice, aged 13 to 24 months. Vascular amyloid was preferentially deposited in arterioles and ranged from small focal to large circumferential depositions. Ultrastructural analysis allowed us to identify specific features contributing to weakening of the vessel wall and aneurysm formation, ie, disruption of the external elastic lamina, thinning of the internal elastic lamina, interruption of the smooth muscle layer, and loss of smooth muscle cells. Biochemically, the much lower Abeta42:Abeta40 ratio evident in vascular relative to plaque amyloid, demonstrated that in blood vessel walls Abeta40 was the more abundant amyloid peptide. The exclusive neuronal origin of transgenic APP, the high levels of Abeta in cerebrospinal fluid compared to plasma, and the specific neuroanatomical localization of vascular amyloid strongly suggest specific drainage pathways, rather than local production or blood uptake of Abeta as the primary mechanism underlying CAA. The demonstration in APP/Ld mice of rare vascular amyloid deposits that immunostained only for Abeta42, suggests that, similar to senile plaque formation, Abeta42 may be the first amyloid to be deposited in the vessel walls and that it entraps the more soluble Abeta40. Its ability to diffuse for larger distances along perivascular drainage pathways would also explain the abundance of Abeta40 in vascular amyloid. Consistent with this hypothesis, incorporation of mutant presenilin-1 in APP/Ld mice, which resulted in selectively higher levels of Abeta42, caused an increase in CAA and senile plaques. This mouse model will be useful in further elucidating the pathogenesis of CAA and Alzheimer's disease, and will allow testing of diagnostic and therapeutic strategies.
- Published
- 2000
- Full Text
- View/download PDF
40. Expression of human apolipoprotein E4 in neurons causes hyperphosphorylation of protein tau in the brains of transgenic mice.
- Author
-
Tesseur I, Van Dorpe J, Spittaels K, Van den Haute C, Moechars D, and Van Leuven F
- Subjects
- Aging, Animals, Apolipoprotein E4, Apolipoproteins E genetics, Brain pathology, DNA Primers chemistry, Humans, In Situ Hybridization, Inclusion Bodies metabolism, Inclusion Bodies pathology, Mice, Mice, Transgenic metabolism, Phosphorylation, RNA, Messenger metabolism, Survival Rate, Ubiquitins metabolism, Apolipoproteins E metabolism, Brain metabolism, Neurons metabolism, tau Proteins metabolism
- Abstract
Epidemiological studies have established that the epsilon 4 allele of the ApoE gene (ApoE4) constitutes an important risk factor for Alzheimer's disease and might influence the outcome of central nervous system injury. The mechanism by which ApoE4 contributes to the development of neurodegeneration remains unknown. To test one hypothesis or mode of action of ApoE, we generated transgenic mice that overexpressed human ApoE4 in different cell types in the brain, using four distinct gene promoter constructs. Many transgenic mice expressing ApoE4 in neurons developed motor problems accompanied by muscle wasting, loss of body weight, and premature death. Overexpression of human ApoE4 in neurons resulted in hyperphosphorylation of the microtubule-associated protein tau. In three independent transgenic lines from two different promoter constructs, increased phosphorylation of protein tau was correlated with ApoE4 expression levels. Hyperphosphorylation of protein tau increased with age. In the hippocampus, astrogliosis and ubiquitin-positive inclusions were demonstrated. These findings demonstrate that expression of ApoE in neurons results in hyperphosphorylation of protein tau and suggests a role for ApoE in neuronal cytoskeletal stability and metabolism.
- Published
- 2000
- Full Text
- View/download PDF
41. Analysis of expression of genes involved in apolipoprotein E-based lipoprotein metabolism in pregnant mice deficient in the receptor-associated protein, the low density lipoprotein receptor, or apolipoprotein E.
- Author
-
Umans L, Overbergh L, Serneels L, Tesseur I, and Van Leuven F
- Subjects
- Animals, Blotting, Northern, Blotting, Western, Cloning, Molecular, DNA Probes, Female, Gene Targeting, Heymann Nephritis Antigenic Complex, Mice, Mice, Inbred C57BL, Mice, Knockout, Pregnancy, RNA, Messenger biosynthesis, Apolipoproteins E deficiency, Apolipoproteins E genetics, Gene Expression Regulation genetics, Lipoproteins metabolism, Membrane Glycoproteins deficiency, Membrane Glycoproteins genetics, Receptors, LDL deficiency, Receptors, LDL genetics
- Abstract
Mice deficient in receptor-associated protein (RAP) were phenotypically normal, but in contrast to results previously reported in RAP(-/-) mice, nearly 50% of the offspring died at or shortly after birth. To attempt to determine the reason for this, we analyzed the regulation of expression of genes involved in apolipoprotein E (apoE)-based mechanisms in RAP-deficient mice and compared this to results in mice deficient in low density lipoprotein receptor (LDLR) or apoE. The major finding concerned a large increase in hepatic lipoprotein receptor-related protein (LRP) mRNA and LDLR mRNA levels in pregnant RAP knockout mice. This is in contrast to the down-regulation of LRP mRNA and LDLR mRNA, which is normally seen in wild-type mice. Also in LDLR knockout mice, a significant up-regulation in expression of LRP mRNA was demonstrated. In apoE knockout mice, hepatic LRP mRNA did not change significantly, while hepatic LDLR mRNA expression was increased. In placenta and uterus, the deficiency of RAP did not markedly affect the expression of LRP and LDLR. Lipoprotein lipase mRNA and apoE mRNA increased during pregnancy in all mice, independent of their genetic status. The current study does not directly explain the increased mortality of RAP(-/-) pups. The data demonstrate, however, important relative changes in expression of the genes analyzed, an indication that LRP and LDLR play an important role in lipid metabolism during pregnancy.
- Published
- 1999
- Full Text
- View/download PDF
42. Disruption of the Kluyveromyces lactis GGS1 gene causes inability to grow on glucose and fructose and is suppressed by mutations that reduce sugar uptake.
- Author
-
Luyten K, de Koning W, Tesseur I, Ruiz MC, Ramos J, Cobbaert P, Thevelein JM, and Hohmann S
- Subjects
- Amino Acid Sequence, Base Sequence, Cloning, Molecular, Glucosyltransferases chemistry, Glycolysis, Kluyveromyces chemistry, Kluyveromyces growth & development, Kluyveromyces metabolism, Molecular Sequence Data, Mutation, Restriction Mapping, Sequence Alignment, Sequence Homology, Amino Acid, Sequence Homology, Nucleic Acid, Fructose metabolism, Genes, Fungal, Genes, Suppressor, Glucose metabolism, Glucosyltransferases genetics, Kluyveromyces genetics
- Abstract
In the yeast Saccharomyces cerevisiae the GGS1 gene is essential for growth on glucose or other readily fermentable sugars. GGS1 is the same gene as TPS1 which was identified as encoding a subunit of the trehalose-6-phosphate synthase/phosphatase complex and it is allelic to the fdp1, byp1, glc6 and cif1 mutations. Its precise function in the regulation of sugar catabolism is unknown. We have cloned the GGS1 homologue from the distantly related yeast Kluyveromyces lactis. The KlGGS1 gene is 74% and 79% identical at the nucleotide and amino acid sequence level, respectively, to the S. cerevisiae counterpart. We also compared the sequence with the partly homologous products of the S. cerevisiae genes TPS2 and TSL1 which code for the larger subunits of the trehalose synthase complex and with a TSL1 homologue, TPS3, of unknown function. Multiple alignment of these sequences revealed several particularly well conserved elements. Disruption of GGS1 in K. lactis caused the same pleiotropic phenotype as in S. cerevisiae, i.e. inability to grow on glucose or fructose and strongly reduced trehalose content. We have also studied short-term glucose-induced regulatory effects related to cAMP and cAMP-dependent protein kinase, i.e. the cAMP signal, trehalase activation, trehalose mobilization and inactivation of fructose-1,6-bisphosphatase. These effects occur very rapidly in S. cerevisiae and are absent in the Scggs1 mutant. In K. lactis all these effects were much slower and largely unaffected by the Klggs1 mutation. On the other hand, glucose strongly induced pyruvate decarboxylase and activated the potassium transport system in K. lactis and both effects were absent in the Klggs1 mutant. Addition of glucose to galactose-grown cells of the Klggs1 mutant caused, as in S. cerevisiae, intracellular accumulation of free glucose and of sugar phosphates and a rapid drop of the ATP and inorganic phosphate levels. Glucose transport kinetics were the same for the wild type and the Klggs1 mutant in both derepressed cells and in cells incubated with glucose. We have isolated phenotypic revertants of the Klggs1 mutant for growth on fructose. The suppressors that we characterized had, to different extents, diminished glucose uptake in derepressed cells but cells incubated in glucose showed very different characteristics. The suppressor mutations prevented deregulation of glycolysis in the Klggs1 mutant but not the accumulation of free glucose. The mutants with higher residual uptake activity showed partially restored induction of pyruvate decarboxylase and activation of potassium transport.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1993
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.