Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Fluids Barriers CNS ; 17(1): 62, 2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33054787

ABSTRACT

BACKGROUND: Preclinical models to determine blood to brain transport ability of therapeutics are often ambiguous. In this study a method is developed that relies on CNS target-engagement and is able to rank brain-penetrating capacities. This method led to the discovery of an anti-transferrin receptor nanobody that is able to deliver a biologically active peptide to the brain via receptor-mediated transcytosis. METHODS: Various nanobodies against the mouse transferrin receptor were fused to neurotensin and injected peripherally in mice. Neurotensin is a neuropeptide that causes hypothermia when present in the brain but is unable to reach the brain from the periphery. Continuous body temperature measurements were used as a readout for brain penetration of nanobody-neurotensin fusions after its peripheral administration. Full temperature curves were analyzed using two-way ANOVA with Dunnett multiple comparisons tests. RESULTS: One anti-transferrin receptor nanobody coupled to neurotensin elicited a drop in body temperature following intravenous injection. Epitope binning indicated that this nanobody bound a distinct transferrin receptor epitope compared to the non-crossing nanobodies. This brain-penetrating nanobody was used to characterize the in vivo hypothermia model. The hypothermic effect caused by neurotensin is dose-dependent and could be used to directly compare peripheral administration routes and various nanobodies in terms of brain exposure. CONCLUSION: This method led to the discovery of an anti-transferrin receptor nanobody that can reach the brain via receptor-mediated transcytosis after peripheral administration. This method could be used to assess novel proteins for brain-penetrating capabilities using a target-engaging readout.


Subject(s)
Body Temperature/drug effects , Brain/drug effects , Neurotensin/pharmacology , Receptors, Transferrin/immunology , Single-Domain Antibodies/pharmacology , Transcytosis/physiology , Animals , Camelids, New World , Female , Male , Mice , Neurotensin/administration & dosage , Receptors, Neurotensin/drug effects , Single-Domain Antibodies/administration & dosage
2.
Methods Enzymol ; 584: 59-97, 2017.
Article in English | MEDLINE | ID: mdl-28065273

ABSTRACT

The study of membrane protein function and structure requires their successful detection, expression, solubilization, and/or reconstitution, which poses a challenging task and relies on the availability of suitable tools. Several research groups have successfully applied Nanobodies in the purification, as well as the functional and structural characterization of membrane proteins. Nanobodies are small, single-chain antibody fragments originating from camelids presenting on average a longer CDR3 which enables them to bind in cavities and clefts (such as active and allosteric sites). Notably, Nanobodies generally bind conformational epitopes making them very interesting tools to stabilize, dissect, and characterize specific protein conformations. In the clinic, several Nanobodies are under evaluation either as potential drug candidates or as diagnostic tools. In recent years, we have successfully generated high-affinity, conformation-sensitive anti-γ-secretase Nanobodies. γ-Secretase is a multimeric membrane protease involved in processing of the amyloid precursor protein with high clinical relevance as mutations in its catalytic subunit (Presenilin) cause early-onset Alzheimer's disease. Advancing our knowledge on the mechanisms governing γ-secretase intramembrane proteolysis through various strategies may lead to novel therapeutic avenues for Alzheimer's disease. In this chapter, we present the strategies we have developed and applied for the screening and characterization of anti-γ-secretase Nanobodies. These protocols could be of help in the generation of Nanobodies targeting other membrane proteins.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/chemistry , Molecular Biology/methods , Presenilins/chemistry , Single-Domain Antibodies/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Amyloid Precursor Protein Secretases/immunology , Amyloid Precursor Protein Secretases/therapeutic use , Catalytic Domain/genetics , Epitopes/immunology , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mutation , Presenilins/genetics , Presenilins/immunology , Presenilins/therapeutic use , Protein Conformation , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Single-Chain Antibodies/therapeutic use , Single-Domain Antibodies/immunology , Single-Domain Antibodies/therapeutic use , Structure-Activity Relationship
3.
Oncogene ; 34(11): 1363-74, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-24681957

ABSTRACT

PINK1 (phosphatase and tensin homolog deleted on chromosome 10 (PTEN)-induced kinase 1), a Parkinson's disease-associated gene, was identified originally because of its induction by the tumor-suppressor PTEN. PINK1 promotes cell survival and potentially metastatic functions and protects against cell stressors including chemotherapeutic agents. However, the mechanisms underlying PINK1 function in cancer cell biology are unclear. Here, using several model systems, we show that PINK1 deletion significantly reduced cancer-associated phenotypes including cell proliferation, colony formation and invasiveness, which were restored by human PINK1 overexpression. Results show that PINK1 deletion causes major defects in cell cycle progression in immortalized mouse embryonic fibroblasts (MEFs) from PINK1(-/-) mice, and in BE(2)-M17 cells stably transduced with short hairpin RNA against PINK1. Detailed cell cycle analyses of MEF cell lines from several PINK1(-/-) mice demonstrate an increased proportion of cells in G2/M and decreased number of cells in G1 following release from nocodazole block. This was concomitant with increased double and multi-nucleated cells, a reduced ability to undergo cytokinesis and to re-enter G1, and significant alterations in cell cycle markers, including failure to increase cyclin D1, all indicative of mitotic arrest. PINK1(-/-) cells also demonstrated ineffective cell cycle exit following serum deprivation. Cell cycle defects associated with PINK1 deficiency occur at points critical for cell division, growth and stress resistance in cancer cells were rescued by ectopic expression of human PINK1 and demonstrated PINK1 kinase dependence. The importance of PINK1 for cell cycle control is further supported by results showing that cell cycle deficits induced by PINK1 deletion were linked mechanistically to aberrant mitochondrial fission and its regulation by dynamin-related protein-1 (Drp1), known to be critical for progression of mitosis. Our data indicate that PINK1 has tumor-promoting properties and demonstrates a new function for PINK1 as a regulator of the cell cycle.


Subject(s)
Cell Division/genetics , Gene Expression Regulation, Neoplastic/genetics , M Phase Cell Cycle Checkpoints/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Cyclin D1/biosynthesis , Cytokinesis/genetics , Dynamins , GTP Phosphohydrolases/metabolism , HeLa Cells , Humans , MCF-7 Cells , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Neoplasm Invasiveness/genetics , Parkinson Disease/genetics , Protein Kinases/biosynthesis , RNA Interference , RNA, Small Interfering
4.
Diabetologia ; 56(7): 1547-56, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23595248

ABSTRACT

AIMS/HYPOTHESIS: We analysed the genomic organisation of miR-153, a microRNA embedded in genes that encode two of the major type 1 diabetes autoantigens, islet-associated protein (IA)-2 and IA-2ß. We also identified miR-153 target genes that correlated with IA-2ß localisation and function. METHODS: A bioinformatics approach was used to identify miR-153's genomic organisation. To analyse the co-regulation of miR-153 and IA-2ß, quantitative PCR analysis of miR-153 and Ia-2ß (also known as Ptprn2) was performed after a glucose stimulation assay in MIN6B cells and isolated murine pancreatic islets, and also in wild-type Ia-2 (also known as Ptprn), Ia-2ß single knockout and Ia-2/Ia-2ß double knockout mouse brain and pancreatic islets. Bioinformatics identification of miR-153 target genes and validation via luciferase reporter assays, western blotting and quantitative PCR were also carried out. RESULTS: Two copies of miR-153, miR-153-1 and miR-153-2, are localised in intron 19 of Ia-2 and Ia-2ß, respectively. In rodents, only miR-153-2 is conserved. We demonstrated that expression of miR-153-2 and Ia-2ß in rodents is partially co-regulated as demonstrated by a strong reduction of miR-153 expression levels in Ia-2ß knockout and Ia-2/Ia-2ß double knockout mice. miR-153 levels were unaffected in Ia-2 knockout mice. In addition, glucose stimulation, which increases Ia-2 and Ia-2ß expression, also significantly increased expression of miR-153. Several predicted targets of miR-153 were reduced after glucose stimulation in vitro, correlating with the increase in miR-153 levels. CONCLUSIONS/INTERPRETATION: This study suggests the involvement of miR-153, IA-2ß and miR-153 target genes in a regulatory network, which is potentially relevant to insulin and neurotransmitter release.


Subject(s)
Brain/metabolism , MicroRNAs/genetics , Pancreas/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 8/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Female , Male , Mice , Mice, Knockout , Receptor-Like Protein Tyrosine Phosphatases, Class 8/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Proc Natl Acad Sci U S A ; 105(28): 9775-80, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18626010

ABSTRACT

Regulated intramembrane proteolysis by gamma-secretase cleaves proteins in their transmembrane domain and is involved in important signaling pathways. At least four different gamma-secretase complexes have been identified, but little is known about their biological role and specificity. Previous work has demonstrated the involvement of the Aph1A-gamma-secretase complex in Notch signaling, but no specific function could be assigned to Aph1B/C-gamma-secretase. We demonstrate here that the Aph1B/C-gamma-secretase complex is expressed in brain areas relevant to schizophrenia pathogenesis and that Aph1B/C deficiency causes pharmacological and behavioral abnormalities that can be reversed by antipsychotic drugs. At the molecular level we find accumulation of Nrg1 fragments in the brain of Aph1BC(-/-) mice. Our observations gain clinical relevance by the demonstration that a Val-to-Leu mutation in the Nrg1 transmembrane domain, associated with increased risk for schizophrenia, affects gamma-secretase cleavage of Nrg1. This finding suggests that dysregulation of intramembrane proteolysis of Nrg1 could increase risk for schizophrenia and related disorders.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Antipsychotic Agents/pharmacology , Endopeptidases/deficiency , Gait Disorders, Neurologic/etiology , Neuregulin-1/metabolism , Protein Subunits/deficiency , Animals , Antipsychotic Agents/therapeutic use , Membrane Proteins , Mice , Mice, Knockout , Mutation, Missense/physiology , Schizophrenia/etiology
6.
Cell Death Differ ; 14(5): 1040-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17290285

ABSTRACT

The apoptosis-inducing Fas ligand (FasL) is a type II transmembrane protein that is involved in the downregulation of immune reactions by activation-induced cell death (AICD) as well as in T cell-mediated cytotoxicity. Proteolytic cleavage leads to the generation of membrane-bound N-terminal fragments and a soluble FasL (sFasL) ectodomain. sFasL can be detected in the serum of patients with dysregulated inflammatory diseases and is discussed to affect Fas-FasL-mediated apoptosis. Using pharmacological approaches in 293T cells, in vitro cleavage assays as well as loss and gain of function studies in murine embryonic fibroblasts (MEFs), we demonstrate that the disintegrin and metalloprotease ADAM10 is critically involved in the shedding of FasL. In primary human T cells, FasL shedding is significantly reduced after inhibition of ADAM10. The resulting elevated FasL surface expression is associated with increased killing capacity and an increase of T cells undergoing AICD. Overall, our findings suggest that ADAM10 represents an important molecular modulator of FasL-mediated cell death.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Fas Ligand Protein/metabolism , Membrane Proteins/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM10 Protein , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Cell Death/drug effects , Enzyme Inhibitors/pharmacology , Fas Ligand Protein/chemistry , Humans , Jurkat Cells , Membrane Proteins/antagonists & inhibitors , Mice , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Recombinant Proteins/metabolism , Solubility/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
7.
Hum Mutat ; 27(9): 888-96, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16917905

ABSTRACT

Since the first report showing that Alzheimer disease (AD) might be caused by mutations in the amyloid precursor protein gene (APP), 20 different missense mutations have been reported. The majority of early-onset AD mutations alter processing of APP increasing relative levels of Abeta42 peptide, either by increasing Abeta42 or decreasing Abeta40 peptide levels or both. In a diagnostic setting using direct sequence analysis, we identified in one patient with familial early-onset AD a novel mutation in APP (c.2172G>C), predicting a K724N substitution in the intracytosolic fragment. The mutation is located downstream of the epsilon-cleavage site of APP and is the furthermost C-terminal mutation reported to date. In vitro expression of APP K724N cDNA showed an increase in Abeta42 and a decrease in Abeta40 levels resulting in a near three-fold increase of the Abeta42/Abeta40 ratio. Further, in vivo amyloid positron emission tomography (PET) imaging revealed significantly increased cortical amyloid deposits, supporting that in human this novel APP mutation is likely causing disease.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Mutation, Missense , Aged , Alzheimer Disease/diagnosis , Alzheimer Disease/diagnostic imaging , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Animals , Belgium , Brain/diagnostic imaging , Cell Line , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mice , Middle Aged , Pedigree , Positron-Emission Tomography , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sequence Analysis, Protein
8.
Verh K Acad Geneeskd Belg ; 66(1): 29-58; discussion 58-9, 2004.
Article in German | MEDLINE | ID: mdl-15074081

ABSTRACT

Alzheimer's disease is the most frequent degenerative disorder of the central nervous system. A focus on the familial, monogenetic disease subtypes has put researchers on the trail of the primary pathogenetic agent, the Abeta-peptide. The production of this peptide is being controlled by a number of proteolytic enzymes commonly known as the "secretases". An intensive study of the molecular and cell biological functions of these secretases is undertaken with the aim of generating drugs to cure Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Amyloid beta-Protein Precursor/physiology , Endopeptidases/physiology , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Endopeptidases/genetics , Humans , Protease Inhibitors/therapeutic use
10.
Neuron ; 32(4): 579-89, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11719200

ABSTRACT

The carboxyl terminus of presenilin 1 and 2 (PS1 and PS2) binds to the neuron-specific cell adhesion molecule telencephalin (TLN) in the brain. PS1 deficiency results in the abnormal accumulation of TLN in a yet unidentified intracellular compartment. The first transmembrane domain and carboxyl terminus of PS1 form a binding pocket with the transmembrane domain of TLN. Remarkably, APP binds to the same regions via part of its transmembrane domain encompassing the critical residues mutated in familial Alzheimer's disease. Our data surprisingly indicate a spatial dissociation between the binding site and the proposed catalytic site near the critical aspartates in PSs. They provide important experimental evidence to support a ring structure model for PS.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/chemistry , Nerve Tissue Proteins/metabolism , Amyloid beta-Protein Precursor/chemistry , Animals , Binding Sites , Cell Differentiation , Gene Expression , Hippocampus/cytology , Membrane Glycoproteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/chemistry , Neurons/cytology , Neurons/metabolism , Presenilin-1 , Presenilin-2 , Protein Structure, Tertiary , Two-Hybrid System Techniques
11.
J Neurochem ; 78(5): 1168-78, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11553691

ABSTRACT

The gamma-secretase cleavage is the last step in the generation of the beta-amyloid peptide (Abeta) from the amyloid precursor protein (APP). The Abeta precipitates in the amyloid plaques in the brain of Alzheimer's disease patients. The fate of the intracellular APP carboxy-terminal stub generated together with Abeta has been, in contrast, only poorly documented. The analogies between the processing of APP and other transmembrane proteins like SREBP and Notch suggests that this intracellular fragment could have important signalling functions. We demonstrate here that APP-C59 is rapidly degraded (half-life approximately 5 min) when overexpressed in baby hamster kidney cells or primary cultures of neurones by a mechanism that is not inhibited by endosomal/lysosomal or proteasome inhibitors. Furthermore, APP-C59 binds to the DNA binding protein Fe65, although this does not increase the half-life of APP-C59. Finally, we demonstrate that a fraction of APP-C59 becomes redistributed to the nuclear detergent-insoluble pellet, in which the transcription factor SP1 is also present. Overall our results reinforce the analogy between Notch and APP processing, and suggest that the APP intracellular domain, like the Notch intracellular domain, could have a role in signalling events from the plasma membrane to the nucleus.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Nucleus/enzymology , Endopeptidases/metabolism , Neurons/enzymology , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases , Cell Fractionation , Cells, Cultured , Cricetinae , Cytoplasm/metabolism , Genetic Vectors , Kidney/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Neurons/cytology , Presenilin-1 , Receptors, Notch , Semliki forest virus/genetics , Sp1 Transcription Factor/metabolism , Subcellular Fractions , Transfection
12.
J Cell Biol ; 154(4): 731-40, 2001 Aug 20.
Article in English | MEDLINE | ID: mdl-11502763

ABSTRACT

We investigated the relationship between PS1 and gamma-secretase processing of amyloid precursor protein (APP) in primary cultures of neurons. Increasing the amount of APP at the cell surface or towards endosomes did not significantly affect PS1-dependent gamma-secretase cleavage, although little PS1 is present in those subcellular compartments. In contrast, almost no gamma-secretase processing was observed when holo-APP or APP-C99, a direct substrate for gamma-secretase, were specifically retained in the endoplasmic reticulum (ER) by a double lysine retention motif. Nevertheless, APP-C99-dilysine (KK) colocalized with PS1 in the ER. In contrast, APP-C99 did not colocalize with PS1, but was efficiently processed by PS1-dependent gamma-secretase. APP-C99 resides in a compartment that is negative for ER, intermediate compartment, and Golgi marker proteins. We conclude that gamma-secretase cleavage of APP-C99 occurs in a specialized subcellular compartment where little or no PS1 is detected. This suggests that at least one other factor than PS1, located downstream of the ER, is required for the gamma-cleavage of APP-C99. In agreement, we found that intracellular gamma-secretase processing of APP-C99-KK both at the gamma40 and the gamma42 site could be restored partially after brefeldin A treatment. Our data confirm the "spatial paradox" and raise several questions regarding the PS1 is gamma-secretase hypothesis.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Compartmentation , Endopeptidases/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Protein Processing, Post-Translational , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/isolation & purification , Animals , Aspartic Acid Endopeptidases , Cells, Cultured , Endopeptidases/isolation & purification , Endoplasmic Reticulum , Golgi Apparatus , Membrane Proteins/isolation & purification , Mice , Mutation , Neurons/cytology , Neurons/ultrastructure , Presenilin-1 , Protein Transport/genetics
13.
J Biol Chem ; 276(46): 42645-57, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11526104

ABSTRACT

Urea-based beta-amyloid (Abeta) SDS-polyacrylamide gel electrophoresis and immunoblots were used to analyze the generation of Abeta peptides in conditioned medium from primary mouse neurons and a neuroglioma cell line, as well as in human cerebrospinal fluid. A comparable and highly conserved pattern of Abeta peptides, namely, 1-40/42 and carboxyl-terminal-truncated 1-37, 1-38, and 1-39, was found. Besides Abeta1-42, we also observed a consistent elevation of amino-terminal-truncated Abeta2-42 in a detergent-soluble pool in brains of subjects with Alzheimer's disease. Abeta2-42 was also specifically elevated in cerebrospinal fluid samples of Alzheimer's disease patients. To decipher the contribution of potential different gamma-secretases (presenilins (PSs)) in generating the amino-terminal- and carboxyl-terminal-truncated Abeta peptides, we overexpressed beta-amyloid precursor protein (APP)-trafficking mutants in PS1+/+ and PS1-/- neurons. As compared with APP-WT (primary neurons from control or PS1-deficient mice infected with Semliki Forest virus), PS1-/- neurons and PS1+/+ neurons overexpressing APP-Deltact (a slow-internalizing mutant) show a decrease of all secreted Abeta peptide species, as expected, because this mutant is processed mainly by alpha-secretase. This drop is even more pronounced for the APP-KK construct (APP mutant carrying an endoplasmic reticulum retention motif). Surprisingly, Abeta2-42 is significantly less affected in PS1-/- neurons and in neurons transfected with the endocytosis-deficient APP-Deltact construct. Our data confirm that PS1 is closely involved in the production of Abeta1-40/42 and the carboxyl-terminal-truncated Abeta1-37, Abeta1-38, and Abeta1-39, but the amino-terminal-truncated and carboxyl-terminal-elongated Abeta2-42 seems to be less affected by PS1 deficiency. Moreover, our results indicate that the latter Abeta peptide species could be generated by a beta(Asp/Ala)-secretase activity.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/chemistry , Peptide Fragments/biosynthesis , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/chemistry , Aged , Aged, 80 and over , Alzheimer Disease/cerebrospinal fluid , Amino Acid Sequence , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Cell Line , Cells, Cultured , DNA, Complementary/metabolism , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Endopeptidases , Endoplasmic Reticulum/metabolism , Humans , Immunoblotting , Mice , Mice, Knockout , Middle Aged , Molecular Sequence Data , Mutation , Neurons/metabolism , Peptides/chemistry , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Semliki forest virus/genetics , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
14.
Hum Mol Genet ; 10(16): 1665-71, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11487570

ABSTRACT

Release of amyloid beta (Abeta) from the amyloid precursor protein (APP) requires cleavages by beta- and gamma-secretases and plays a crucial role in Alzheimer's disease (AD) pathogenesis. Missense mutations in the APP gene causing familial AD are clustered around the beta-, alpha- and particular gamma-secretase cleavage sites. We systematically compare in primary neurons the effect on APP processing of a series of clinical APP mutations (two of which not characterized before) located in close proximity to the gamma-secretase cleavage site. We confirm and extend previous observations showing that all these mutations (T714I, V715M, V715A, I716V, V717I and V717L) affect gamma-secretase cleavage causing an increased relative ratio of Abeta42 to Abeta40. Taking advantage of these extended series of APP mutations we were able to demonstrate an inverse correlation between these ratios and the age at onset of the disease in the different families. In addition, a subset of mutations caused the accumulation of APP C-terminal fragments indicating that these mutations also influence the stability of APP C-terminal fragments. However, it is unlikely that these fragments contribute significantly to the disease process.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Endopeptidases/metabolism , Mutation , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases , Binding Sites , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Humans , Mice , Neurons/metabolism , Peptide Fragments/metabolism , Precipitin Tests , Transduction, Genetic
15.
J Biol Chem ; 276(41): 37743-6, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11477090

ABSTRACT

We showed previously that PrPc undergoes constitutive and phorbol ester-regulated cleavage inside the 106-126 toxic domain of the protein, leading to the production of a fragment referred to as N1. Here we show by a pharmacological approach that o-phenanthroline, a general zinc-metalloprotease inhibitors, as well as BB3103 and TAPI, the inhibitors of metalloenzymes ADAM10 (A disintegrin and metalloprotease); and TACE, tumor necrosis factor alpha-converting enzyme; ADAM17), respectively, drastically reduce N1 formation. We set up stable human embryonic kidney 293 transfectants overexpressing human ADAM10 and TACE, and we demonstrate that ADAM10 contributes to constitutive N1 production whereas TACE mainly participates in regulated N1 formation. Furthermore, constitutive N1 secretion is drastically reduced in fibroblasts deficient for ADAM10 whereas phorbol 12,13-dibutyrate-regulated N1 production is fully abolished in TACE-deficient cells. Altogether, our data demonstrate for the first time that disintegrins could participate in the catabolism of glycosyl phosphoinositide-anchored proteins such as PrPc. Second, our study identifies ADAM10 and ADAM17 as the protease candidates responsible for normal cleavage of PrPc. Therefore, these disintegrins could be seen as putative cellular targets of a therapeutic strategy aimed at increasing normal PrPc breakdown and thereby depleting cells of the putative 106-126 "toxic" domain of PrPc.


Subject(s)
Endopeptidases/metabolism , Metalloendopeptidases/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , PrPC Proteins/metabolism , ADAM Proteins , ADAM17 Protein , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Cell Line , Humans , Hydrolysis
16.
J Biol Chem ; 276(35): 33273-81, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11432849

ABSTRACT

Mutations in presenilin (PS) genes cause early-onset familial Alzheimer's disease by increasing production of the amyloidogenic form of amyloid beta peptides ending at residue 42 (Abeta42). PS is an evolutionarily conserved multipass transmembrane protein, and all known PS proteins contain a proline-alanine-leucine-proline (PALP) motif starting at proline (P) 414 (amino acid numbering based on human PS2) at the C terminus. Furthermore, missense mutations that replace the first proline of PALP with leucine (P414L) lead to a loss-of-function of PS in Drosophila melanogaster and Caenorhabditis elegans. To elucidate the roles of the PALP motif in PS structure and function, we analyzed neuro2a as well as PS1/2 null fibroblast cell lines transfected with human PS harboring mutations at the PALP motif. P414L mutation in PS2 (and its equivalent in PS1) abrogated stabilization, high molecular weight complex formation, and entry to Golgi/trans-Golgi network of PS proteins, resulting in failure of Abeta42 overproduction on familial Alzheimer's disease mutant basis as well as of site-3 cleavage of Notch. These data suggest that the first proline of the PALP motif plays a crucial role in the stabilization and formation of the high molecular weight complex of PS, the latter being the active form with intramembrane proteolytic activities.


Subject(s)
Endopeptidases/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mutation, Missense , Proline , Alzheimer Disease/genetics , Amino Acid Sequence , Amino Acid Substitution , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Caenorhabditis/genetics , Caenorhabditis/metabolism , Cattle , Cell Line , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Kinetics , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Presenilin-1 , Presenilin-2 , Rats , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
17.
Amyloid ; 8(2): 124-42, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11409035

ABSTRACT

The extracellular deposition of short amyloid peptides in the brain of patients is thought to be a central event in the pathogenesis of Alzheimer's Disease. The generation of the amyloid peptide occurs via a regulated cascade of cleavage events in its precursor protein, A beta PP. At least three enzymes are responsible for A beta PP proteolysis and have been tentatively named alpha-, beta- and gamma-secretases. The recent identification of several of these secretases is a major leap in the understanding how these secretases regulate amyloid peptide formation. Members of the ADAM family of metalloproteases are involved in the non-amyloidogenic alpha-secretase pathway. The amyloidogenic counterpart pathway is initiated by the recently cloned novel aspartate protease named BACE. The available data are conclusive and crown BACE as the long-sought beta-secretase. This enzyme is a prime candidate drug target for the development of therapy aiming to lower the amyloid burden in the disease. Finally, the gamma-secretases are intimately linked to the function of the presenilins. These multi-transmembrane domain proteins remain intriguing study objects. The hypothesis that the presenilins constitute a complete novel type of protease family, and are cleaving A beta PP within the transmembrane region, remains an issue of debate. Several questions remain unanswered and direct proof that they exert catalytic activity is still lacking. The subcellular localization of presenilins in neurons, their integration in functional multiprotein complexes and the recent identification of additional modulators of gamma-secretase, like nicastrin, indicate already that several players are involved. Nevertheless, the rapidly increasing knowledge in this area is already paving the road towards selective inhibitors of this secretase as well. It is hoped that such drugs, possibly in concert with the experimental vaccination therapies that are currently tested, will lead to a cure of this inexorable disease.


Subject(s)
Alzheimer Disease/drug therapy , Endopeptidases/drug effects , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/drug effects , Aspartic Acid Endopeptidases/metabolism , Humans
19.
J Biol Chem ; 276(6): 4211-7, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11071887

ABSTRACT

The amyloid peptide is the main constituent of the amyloid plaques in brain of Alzheimer's disease patients. This peptide is generated from the amyloid precursor protein by two consecutive cleavages. Cleavage at the N terminus is performed by the recently discovered beta-secretase (Bace). This aspartyl protease contains a propeptide that has to be removed to obtain mature Bace. Furin and other members of the furin family of prohormone convertases are involved in this process. Surprisingly, beta-secretase activity, neither at the classical Asp(1) position nor at the Glu(11) position of amyloid precursor protein, seems to be controlled by this maturation step. Furthermore, we show that Glu(11) cleavage is a function of the expression level of Bace, that it depends on the membrane anchorage of Bace, and that Asp(1) cleavage can be followed by Glu(11) cleavage. Our data suggest that pro-Bace could be active as a beta-secretase in the early biosynthetic compartments of the cell and could be involved in the generation of the intracellular pool of the amyloid peptide. We conclude that modulation of the conversion of pro-Bace to mature Bace is not a relevant drug target to treat Alzheimer's disease.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Subtilisins/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/biosynthesis , Furin , Hippocampus/enzymology , Hippocampus/metabolism , Humans , Protein Processing, Post-Translational
20.
J Mol Neurosci ; 17(2): 171-81, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11816790

ABSTRACT

Signaling via notch receptors and their ligands is an evolutionary ancient and highly conserved mechanism governing cell-fate decisions throughout the animal kingdom. Upon ligand binding, notch receptors are subject to a two-step proteolysis essential for signal transduction. First, the ectodomain is removed by an enzyme cleaving near the outer-membrane surface ("site2"). Consecutively, the notch intracellular domain is liberated by a second protease cutting within the transmembrane sequence ("site3"). The intracellular domain is then transferred to the nucleus to act as a transcriptional coactivator. The proteases involved in notch receptor activation are shared with other proteins undergoing regulated intramembrane proteolysis, with intriguing parallels to APP. Specifically, site3 cleavage of Notch, as well as gamma-secretase processing of APP depend both critically on presenilins 1 and 2. Moreover, ADAM 10 and ADAM 17, the proteases proposed to perform site2 cleavage, are also the most probable candidate alpha-secretases to cleave APP. While the biological significance of APP processing remains to be further elucidated, interference with notch signaling has been shown to have severe consequences both in small animal models as well as in humans. Thus, a growing number of long known genetic syndromes like Alagille syndrome or Fallot's tetralogy can be caused by mutations of genes relevant for the notch signaling pathway. Likewise, the anticipated interference of gamma-secretase inhibitors with site3 cleavage may turn out to be a major obstacle for this therapeutic approach to Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Endopeptidases/metabolism , Membrane Proteins/metabolism , Receptors, Cell Surface , Signal Transduction/genetics , Transcription Factors , ADAM Proteins , ADAM17 Protein , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Endopeptidases/genetics , Humans , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Presenilin-1 , Presenilin-2 , Receptor, Notch1
SELECTION OF CITATIONS
SEARCH DETAIL
...