Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 77
Filtrer
1.
Eur J Cell Biol ; 103(2): 151406, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38547677

RÉSUMÉ

Despite extensive research, targeted delivery of substances to the brain still poses a great challenge due to the selectivity of the blood-brain barrier (BBB). Most molecules require either carrier- or receptor-mediated transport systems to reach the central nervous system (CNS). These transport systems form attractive routes for the delivery of therapeutics into the CNS, yet the number of known brain endothelium-enriched receptors allowing the transport of large molecules into the brain is scarce. Therefore, to identify novel BBB targets, we combined transcriptomic analysis of human and murine brain endothelium and performed a complex screening of BBB-enriched genes according to established selection criteria. As a result, we propose the high-affinity cationic amino acid transporter 1 (SLC7A1) as a novel candidate for transport of large molecules across the BBB. Using RNA sequencing and in situ hybridization assays, we demonstrated elevated SLC7A1 gene expression in both human and mouse brain endothelium. Moreover, we confirmed SLC7A1 protein expression in brain vasculature of both young and aged mice. To assess the potential of SLC7A1 as a transporter for larger proteins, we performed internalization and transcytosis studies using a radiolabelled or fluorophore-labelled anti-SLC7A1 antibody. Our results showed that SLC7A1 internalised a SLC7A1-specific antibody in human colorectal carcinoma (HCT116) cells. Moreover, transcytosis studies in both immortalised human brain endothelial (hCMEC/D3) cells and primary mouse brain endothelial cells clearly demonstrated that SLC7A1 effectively transported the SLC7A1-specific antibody from luminal to abluminal side. Therefore, here in this study, we present for the first time the SLC7A1 as a novel candidate for transport of larger molecules across the BBB.


Sujet(s)
Barrière hémato-encéphalique , Transporteur-1 d'acides aminés cationiques , Barrière hémato-encéphalique/métabolisme , Animaux , Humains , Souris , Transporteur-1 d'acides aminés cationiques/métabolisme , Transporteur-1 d'acides aminés cationiques/génétique , Cellules endothéliales/métabolisme , Souris de lignée C57BL
2.
Cell Mol Life Sci ; 81(1): 139, 2024 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-38480559

RÉSUMÉ

Neurotoxic amyloid-ß (Aß) peptides cause neurodegeneration in Alzheimer's disease (AD) patients' brains. They are released upon proteolytic processing of the amyloid precursor protein (APP) extracellularly at the ß-secretase site and intramembranously at the γ-secretase site. Several AD mouse models were developed to conduct respective research in vivo. Most of these classical models overexpress human APP with mutations driving AD-associated pathogenic APP processing. However, the resulting pattern of Aß species in the mouse brains differs from those observed in AD patients' brains. Particularly mutations proximal to the ß-secretase cleavage site (e.g., the so-called Swedish APP (APPswe) fostering Aß1-x formation) lead to artificial Aß production, as N-terminally truncated Aß peptides are hardly present in these mouse brains. Meprin ß is an alternative ß-secretase upregulated in brains of AD patients and capable of generating N-terminally truncated Aß2-x peptides. Therefore, we aimed to generate a mouse model for the production of so far underestimated Aß2-x peptides by conditionally overexpressing meprin ß in astrocytes. We chose astrocytes as meprin ß was detected in this cell type in close proximity to Aß plaques in AD patients' brains. The meprin ß-overexpressing mice showed elevated amyloidogenic APP processing detected with a newly generated neo-epitope-specific antibody. Furthermore, we observed elevated Aß production from endogenous APP as well as AD-related behavior changes (hyperlocomotion and deficits in spatial memory). The novel mouse model as well as the established tools and methods will be helpful to further characterize APP cleavage and the impact of different Aß species in future studies.


Sujet(s)
Maladie d'Alzheimer , Amyloid precursor protein secretases , Humains , Souris , Animaux , Amyloid precursor protein secretases/génétique , Amyloid precursor protein secretases/métabolisme , Astrocytes/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Peptides bêta-amyloïdes/métabolisme , Maladie d'Alzheimer/métabolisme , Protéolyse , Encéphale/métabolisme
3.
Redox Biol ; 67: 102943, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37883843

RÉSUMÉ

Accumulation of misfolded proteins or perturbation of calcium homeostasis leads to endoplasmic reticulum (ER) stress and is linked to the pathogenesis of neurodegenerative diseases. Hence, understanding the ability of neuronal cells to cope with chronic ER stress is of fundamental interest. Interestingly, several brain areas uphold functions that enable them to resist challenges associated with neurodegeneration. Here, we established novel clonal mouse hippocampal (HT22) cell lines that are resistant to prolonged (chronic) ER stress induced by thapsigargin (TgR) or tunicamycin (TmR) as in vitro models to study the adaption to ER stress. Morphologically, we observed a significant increase in vesicular und autophagosomal structures in both resistant lines and 'giant lysosomes', especially striking in TgR cells. While autophagic activity increased under ER stress, lysosomal function appeared slightly impaired; in both cell lines, we observed enhanced ER-phagy. However, proteomic analyses revealed that various protein clusters and signaling pathways were differentially regulated in TgR versus TmR cells in response to chronic ER stress. Additionally, bioenergetic analyses in both resistant cell lines showed a shift toward aerobic glycolysis ('Warburg effect') and a defective complex I of the oxidative phosphorylation (OXPHOS) machinery. Furthermore, ER stress-resistant cells differentially activated the unfolded protein response (UPR) comprising IRE1α and ATF6 pathways. These findings display the wide portfolio of adaptive responses of neuronal cells to chronic ER stress. ER stress-resistant neuronal cells could be the basis to uncover molecular modulators of adaptation, resistance, and neuroprotection as potential pharmacological targets for preventing neurodegeneration.


Sujet(s)
Endoribonucleases , Protein-Serine-Threonine Kinases , Souris , Animaux , Protein-Serine-Threonine Kinases/métabolisme , Endoribonucleases/génétique , Protéomique , Stress du réticulum endoplasmique , Réponse aux protéines mal repliées , Réticulum endoplasmique/métabolisme
4.
J Biol Chem ; 299(8): 104997, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37394008

RÉSUMÉ

Presenilin-1 (PSEN1) is the catalytic subunit of the intramembrane protease γ-secretase and undergoes endoproteolysis during its maturation. Heterozygous mutations in the PSEN1 gene cause early-onset familial Alzheimer's disease (eFAD) and increase the proportion of longer aggregation-prone amyloid-ß peptides (Aß42 and/or Aß43). Previous studies had suggested that PSEN1 mutants might act in a dominant-negative fashion by functional impediment of wild-type PSEN1, but the exact mechanism by which PSEN1 mutants promote pathogenic Aß production remains controversial. Using dual recombinase-mediated cassette exchange (dRMCE), here we generated a panel of isogenic embryonic and neural stem cell lines with heterozygous, endogenous expression of PSEN1 mutations. When catalytically inactive PSEN1 was expressed alongside the wild-type protein, we found the mutant accumulated as a full-length protein, indicating that endoproteolytic cleavage occurred strictly as an intramolecular event. Heterozygous expression of eFAD-causing PSEN1 mutants increased the Aß42/Aß40 ratio. In contrast, catalytically inactive PSEN1 mutants were still incorporated into the γ-secretase complex but failed to change the Aß42/Aß40 ratio. Finally, interaction and enzyme activity assays demonstrated the binding of mutant PSEN1 to other γ-secretase subunits, but no interaction between mutant and wild-type PSEN1 was observed. These results establish that pathogenic Aß production is an intrinsic property of PSEN1 mutants and strongly argue against a dominant-negative effect in which PSEN1 mutants would compromise the catalytic activity of wild-type PSEN1 through conformational effects.


Sujet(s)
Maladie d'Alzheimer , Amyloid precursor protein secretases , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Amyloid precursor protein secretases/métabolisme , Protéines mutantes/génétique , Mutation , Fragments peptidiques/métabolisme , Préséniline-1/métabolisme , Animaux , Souris
5.
J Cell Biochem ; 124(7): 1040-1049, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37288821

RÉSUMÉ

The acute ischemic stroke therapy of choice is the application of Alteplase, a drug containing the enzyme tissue-type plasminogen activator (tPa) which rapidly destabilizes blood clots. A central hallmark of stroke pathology is blood-brain barrier (BBB) breakdown associated with tight junction (TJ) protein degradation, which seems to be significantly more severe under therapeutic conditions. The exact mechanisms how tPa facilitates BBB breakdown are not entirely understood. There is evidence that an interaction with the lipoprotein receptor-related protein 1 (LRP1), allowing tPa transport across the BBB into the central nervous system, is necessary for this therapeutic side effect. Whether tPa-mediated disruption of BBB integrity is initiated directly on microvascular endothelial cells or other brain cell types is still elusive. In this study we could not observe any changes of barrier properties in microvascular endothelial cells after tPa incubation. However, we present evidence that tPa causes changes in microglial activation and BBB breakdown after LRP1-mediated transport across the BBB. Using a monoclonal antibody targeting the tPa binding sites of LRP1 decreased tPa transport across an endothelial barrier. Our results indicate that limiting tPa transport from the vascular system into the brain by coapplication of a LRP1-blocking monoclonal antibody might be a novel approach to minimize tPa-related BBB damage during acute stroke therapy.


Sujet(s)
Accident vasculaire cérébral ischémique , Accident vasculaire cérébral , Humains , Activateur tissulaire du plasminogène/effets indésirables , Activateur tissulaire du plasminogène/métabolisme , Cellules endothéliales/métabolisme , Accident vasculaire cérébral ischémique/induit chimiquement , Accident vasculaire cérébral ischémique/complications , Accident vasculaire cérébral ischémique/traitement médicamenteux , Protéine-1 apparentée au récepteur des LDL/métabolisme , Protéine-1 apparentée au récepteur des LDL/usage thérapeutique , Accident vasculaire cérébral/traitement médicamenteux , Accident vasculaire cérébral/anatomopathologie , Anticorps monoclonaux/usage thérapeutique , Lipoprotéines LDL
6.
Pharmaceutics ; 15(4)2023 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-37111752

RÉSUMÉ

Currently, many neurological disorders lack effective treatment options due to biological barriers that effectively separate the central nervous system (CNS) from the periphery. CNS homeostasis is maintained by a highly selective exchange of molecules, with tightly controlled ligand-specific transport systems at the blood-brain barrier (BBB) playing a key role. Exploiting or modifying these endogenous transport systems could provide a valuable tool for targeting insufficient drug delivery into the CNS or pathological changes in the microvasculature. However, little is known about how BBB transcytosis is continuously regulated to respond to temporal or chronic changes in the environment. The aim of this mini-review is to draw attention to the sensitivity of the BBB to circulating molecules derived from peripheral tissues, which may indicate a fundamental endocrine-operating regulatory system of receptor-mediated transcytosis at the BBB. We present our thoughts in the context of the recent observation that low-density lipoprotein receptor-related protein 1 (LRP1)-mediated clearance of brain amyloid-ß (Aß) across the BBB is negatively regulated by peripheral proprotein convertase subtilisin/kexin type 9 (PCSK9). We hope that our conclusions will inspire future investigations of the BBB as dynamic communication interface between the CNS and periphery, whose peripheral regulatory mechanisms could be easily exploited for therapeutic purposes.

7.
Cell Mol Life Sci ; 79(3): 168, 2022 Mar 02.
Article de Anglais | MEDLINE | ID: mdl-35235058

RÉSUMÉ

ß-Site amyloid precursor protein (APP) cleaving enzyme-1 (BACE1) is the major described ß-secretase to generate Aß peptides in Alzheimer's disease (AD). However, all therapeutic attempts to block BACE1 activity and to improve AD symptoms have so far failed. A potential candidate for alternative Aß peptides generation is the metalloproteinase meprin ß, which cleaves APP predominantly at alanine in p2 and in this study we can detect an increased meprin ß expression in AD brain. Here, we report the generation of the transgenic APP/lon mouse model of AD lacking the functional Mep1b gene (APP/lon × Mep1b-/-). We examined levels of canonical and truncated Aß species using urea-SDS-PAGE, ELISA and immunohistochemistry in brains of APP/lon mouse × Mep1b-/-. Additionally, we investigated the cognitive abilities of these mice during the Morris water maze task. Aß1-40 and 1-42 levels are reduced in APP/lon mice when meprin ß is absent. Immunohistochemical staining of mouse brain sections revealed that N-terminally truncated Aß2-x peptide deposition is decreased in APP/lon × Mep1b-/- mice. Importantly, loss of meprin ß improved cognitive abilities and rescued learning behavior impairments in APP/lon mice. These observations indicate an important role of meprin ß within the amyloidogenic pathway and Aß production in vivo.


Sujet(s)
Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Encéphale/métabolisme , Apprentissage , Troubles de la mémoire/anatomopathologie , Metalloendopeptidases/déficit , Sujet âgé , Amyloid precursor protein secretases/métabolisme , Animaux , Astrocytes/métabolisme , Encéphale/anatomopathologie , Croisements génétiques , Modèles animaux de maladie humaine , Femelle , Protéine gliofibrillaire acide/métabolisme , Humains , Mâle , Metalloendopeptidases/métabolisme , Souris knockout , Peptides/métabolisme , Maturation post-traductionnelle des protéines
8.
Cell Mol Life Sci ; 79(4): 212, 2022 Mar 27.
Article de Anglais | MEDLINE | ID: mdl-35344086

RÉSUMÉ

Despite the neurodegenerative disorder Alzheimer's disease (AD) is the most common form of dementia in late adult life, there is currently no therapy available to prevent the onset or slow down the progression of AD. The progressive cognitive decline in AD correlates with a successive accumulation of cerebral amyloid-ß (Aß) due to impaired clearance mechanisms. A significant percentage is removed by low-density lipoprotein receptor-related protein 1 (LRP1)-mediated transport across the blood-brain barrier (BBB) into the periphery. Circulating proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to members of the low-density lipoprotein receptor protein family at the cell surface and targets them for lysosomal degradation, which reduces the number of functional receptors. However, the adverse impact of PCSK9 on LRP1-mediated brain Aß clearance remains elusive. By using an established BBB model, we identified reduced LRP1-mediated brain-to-blood Aß clearance due to PCSK9 across different endothelial monolayer in vitro. Consequently, the repetitive application of FDA-approved monoclonal anti-PCSK9 antibodies into 5xFAD mice decreased the cerebral Aß burden across variants and aggregation state, which was not reproducible in brain endothelial-specific LRP1-/- 5xFAD mice. The peripheral PCSK9 inhibition reduced Aß pathology in prefrontal cortex and hippocampus-brain areas critically involved in memory processing-and prevented disease-related impairment in hippocampus-dependent memory formation. Our data suggest that peripheral inhibition of PCSK9 by already available therapeutic antibodies may be a novel and easily applicable potential AD treatment.


Sujet(s)
Barrière hémato-encéphalique , Proprotéine convertase 9 , Peptides bêta-amyloïdes/métabolisme , Animaux , Transport biologique , Barrière hémato-encéphalique/métabolisme , Humains , Souris , Proprotéine convertase 9/métabolisme
9.
Handb Exp Pharmacol ; 273: 247-266, 2022.
Article de Anglais | MEDLINE | ID: mdl-33580390

RÉSUMÉ

The accumulation of neurotoxic amyloid-beta (Aß) in the brain is one of the characteristic hallmarks of Alzheimer's disease (AD). Aß-peptide brain homeostasis is governed by its production and various clearance mechanisms. The blood-brain barrier provides a large surface area for influx and efflux mechanisms into and out of the brain. Different transporters and receptors have been implicated to play crucial roles in Aß clearance from brain. Besides Aß transport, the blood-brain barrier tightly regulates the brain's microenvironment; however, vascular alterations have been shown in patients with AD. Here, we summarize how the blood-brain barrier changes during aging and in disease and focus on recent findings of how the ABC transporter P-glycoprotein (ABCB1/P-gp) and the receptor low-density lipoprotein receptor-related protein 1 (LRP1) play a role in Aß clearance from brain.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Barrière hémato-encéphalique/métabolisme , Encéphale/métabolisme , Humains , Fragments peptidiques/métabolisme , Récepteurs aux lipoprotéines LDL/métabolisme
10.
Int J Mol Sci ; 22(9)2021 May 08.
Article de Anglais | MEDLINE | ID: mdl-34066779

RÉSUMÉ

The mechanisms underlying the transport of leptin into the brain are still largely unclear. While the leptin receptor has been implicated in the transport process, recent evidence has suggested an additional role of LRP2 (megalin). To evaluate the function of LRP2 for leptin transport across the blood-brain barrier (BBB), we developed a novel leptin-luciferase fusion protein (pLG), which stimulated leptin signaling and was transported in an in vitro BBB model based on porcine endothelial cells. The LRP inhibitor RAP did not affect leptin transport, arguing against a role of LRP2. In line with this, the selective deletion of LRP2 in brain endothelial cells and epithelial cells of the choroid plexus did not influence bodyweight, body composition, food intake, or energy expenditure of mice. These findings suggest that LRP2 at the BBB is not involved in the transport of leptin into the brain, nor in the development of obesity as has previously been described.


Sujet(s)
Barrière hémato-encéphalique/métabolisme , Leptine/métabolisme , Protéine-2 apparentée au récepteur des LDL/métabolisme , Obésité/métabolisme , Obésité/anatomopathologie , Animaux , Sites de fixation , Composition corporelle , Poids , Cellules CHO , Plexus choroïde/métabolisme , Cricetulus , Cellules endothéliales/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Femelle , Luciferases/métabolisme , Mâle , Modèles biologiques , Phosphorylation , Transport des protéines , Récepteurs à la leptine/métabolisme , Protéines de fusion recombinantes/métabolisme , Suidae
11.
Fluids Barriers CNS ; 18(1): 27, 2021 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-34147102

RÉSUMÉ

The entry of blood-borne molecules into the brain is restricted by the blood-brain barrier (BBB). Various physical, transport and immune properties tightly regulate molecule movement between the blood and the brain to maintain brain homeostasis. A recent study utilizing a pan-endothelial, constitutive Tie2-Cre showed that paracellular passage of blood proteins into the brain is governed by endocytic and cell signaling protein low-density lipoprotein receptor-related protein 1 (LRP1). Taking advantage of conditional Slco1c1-CreERT2 specific to CNS endothelial cells and choroid plexus epithelial cells we now supplement previous results and show that brain endothelial Lrp1 ablation results in protease-mediated tight junction degradation, P-glycoprotein (P-gp) reduction and a loss of BBB integrity.


Sujet(s)
Barrière hémato-encéphalique/métabolisme , Encéphale/métabolisme , Perméabilité capillaire/physiologie , Cellules endothéliales/métabolisme , Protéine-1 apparentée au récepteur des LDL/déficit , Jonctions serrées/métabolisme , Animaux , Cellules cultivées , Protéine-1 apparentée au récepteur des LDL/génétique , Souris , Souris knockout , Souris transgéniques , Jonctions serrées/génétique
12.
J Cereb Blood Flow Metab ; 41(1): 31-44, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32065075

RÉSUMÉ

The metalloprotease meprin ß (Mep1b) is capable of cleaving cell-adhesion molecules in different tissues (e.g. skin, kidney and intestine) and is dysregulated in several diseases associated with barrier breakdown (Alzheimer´s disease, kidney disruption, inflammatory bowel disease). In this study, we demonstrate that Mep1b is a novel regulator of tight junction (TJ) composition and blood-brain barrier (BBB) integrity in brain endothelium. In Mep1b-transfected mouse brain endothelial cells (bEnd.3), we observed a reduction of the TJ protein claudin-5, decreased transendothelial electrical resistance (TEER) and an elevated permeability to paracellular diffusion marker [14C]-inulin. Analysis of global Mep1b knock-out (Mep1b-/-) mice showed increased TJ protein expression (claudin-5, occludin, ZO-1) in cerebral microvessels and increased TEER in cultivated primary mouse brain endothelial compared to wild-type (wt) mice. Furthermore, we investigated the IgG levels in cerebrospinal fluid (CSF) and the brain water content as additional permeability markers and detected lower IgG levels and reduced brain water content in Mep1b-/- mice compared to wt mice. Showing opposing features in overexpression and knock-out, we conclude that Mep1b plays a role in regulating brain endothelial TJ-proteins and therefore affecting BBB tightness in vitro and in vivo.


Sujet(s)
Barrière hémato-encéphalique/physiopathologie , Encéphale/métabolisme , Cellules endothéliales/métabolisme , Metalloendopeptidases/métabolisme , Protéines de la jonction serrée/métabolisme , Animaux , Humains , Souris
13.
Glia ; 68(12): 2517-2549, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-32579270

RÉSUMÉ

The Alzheimer disease-associated multifunctional low-density lipoprotein receptor-related protein-1 is expressed in the brain. Recent studies uncovered a role of this receptor for the appropriate functioning of neural stem cells, oligodendrocytes, and neurons. The constitutive knock-out (KO) of the receptor is embryonically lethal. To unravel the receptors' role in the developing brain we generated a mouse mutant by specifically targeting radial glia stem cells of the dorsal telencephalon. The low-density lipoprotein receptor-related protein-1 lineage-restricted KO female and male mice, in contrast to available models, developed a severe neurological phenotype with generalized seizures during early postnatal development. The mechanism leading to a buildup of hyperexcitability and emergence of seizures was traced to a failure in adequate astrocyte development and deteriorated postsynaptic density integrity. The detected impairments in the astrocytic lineage: precocious maturation, reactive gliosis, abolished tissue plasminogen activator uptake, and loss of functionality emphasize the importance of this glial cell type for synaptic signaling in the developing brain. Together, the obtained results highlight the relevance of astrocytic low-density lipoprotein receptor-related protein-1 for glutamatergic signaling in the context of neuron-glia interactions and stage this receptor as a contributing factor for epilepsy.


Sujet(s)
Cellules épendymogliales , Animaux , Astrocytes , Femelle , Lipoprotéines LDL , Mâle , Souris , Prosencéphale , Récepteurs aux lipoprotéines , Crises épileptiques , Activateur tissulaire du plasminogène
14.
Autophagy ; 16(5): 811-825, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-31282275

RÉSUMÉ

CTSD (cathepsin D) is one of the major lysosomal proteases indispensable for the maintenance of cellular proteostasis by turning over substrates of endocytosis, phagocytosis and autophagy. Consequently, CTSD deficiency leads to a strong impairment of the lysosomal-autophagy machinery. In mice and humans CTSD dysfunction underlies the congenital variant (CLN10) of neuronal ceroid lipofuscinosis (NCL). NCLs are distinct lysosomal storage disorders (LSDs) sharing various hallmarks, namely accumulation of protein aggregates and ceroid lipofuscin leading to neurodegeneration and blindness. The most established and clinically approved approach to treat LSDs is enzyme replacement therapy (ERT) aiming to replace the defective hydrolase with an exogenously applied recombinant protein. Here we reveal that recombinant human pro-CTSD produced in a mammalian expression system can be efficiently taken up by a variety of cell models, is correctly targeted to lysosomes and processed to the active mature form of the protease. In proof-of-principle experiments we provide evidence that recombinant human CTSD (rhCTSD) can improve the biochemical phenotype of CTSD-deficient hippocampal slice cultures in vitro and retinal cells in vivo. Furthermore, we demonstrate that dosing of rhCTSD in the murine CLN10 model leads to a correction of lysosomal hypertrophy, storage accumulation and impaired autophagic flux in the viscera and central nervous system (CNS). We establish that direct delivery of the recombinant protease to the CNS is required for improvement of neuropathology and lifespan extension. Together these data support the continuation of the pre-clinical studies for the application of rhCTSD in the treatment of NCL.Abbreviations: AIF1/IBA1: allograft inflammatory factor 1; BBB: blood brain barrier; CNS: central nervous system; CTSB: cathepsin B; CTSD: cathepsin D; CTSL: cathepsin L; ERT: enzyme replacement therapy; GFAP: glial fibrillary acidic protein; INL: inner nuclear layer; LAMP1: lysosomal-associated membrane protein 1; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; LDL: low-density lipoprotein; LRP1: low density lipoprotein receptor-related protein 1; LSD: lysosomal storage disorder; MEFs: mouse embryonic fibroblasts; M6P: mannose 6-phosphate; mCTSD: mature CTSD; NCL: neuronal ceroid lipofuscinosis; ONL: outer nuclear layer; PB: phosphate buffer; proCTSD: pro-cathepsin D; LRPAP1: low density lipoprotein receptor-related protein associated protein 1; rhCTSD: human recombinant CTSD; SAPC: saposin C; SAPD: saposin D; ATP5G1: ATP synthase, H+ transporting, mitochondrial F0 complex, subunit C1 (subunit 9); SQSTM1/p62: sequestosome 1; TPP1: tripeptidyl peptidase I.


Sujet(s)
Autophagie/effets des médicaments et des substances chimiques , Cathepsine D/usage thérapeutique , Thérapie enzymatique substitutive , Céroïdes-lipofuscinoses neuronales/traitement médicamenteux , Céroïdes-lipofuscinoses neuronales/métabolisme , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Cathepsine D/métabolisme , Modèles animaux de maladie humaine , Thérapie enzymatique substitutive/méthodes , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Humains , Lysosomes/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Souris knockout , Tripeptidyl-peptidase-1
15.
Cell Mol Life Sci ; 77(2): 331-350, 2020 Jan.
Article de Anglais | MEDLINE | ID: mdl-31209506

RÉSUMÉ

Disintegrin and metalloproteinases (ADAMs) 10 and 17 can release the extracellular part of a variety of membrane-bound proteins via ectodomain shedding important for many biological functions. So far, substrate identification focused exclusively on membrane-anchored ADAM10 and ADAM17. However, besides known shedding of ADAM10, we identified ADAM8 as a protease capable of releasing the ADAM17 ectodomain. Therefore, we investigated whether the soluble ectodomains of ADAM10/17 (sADAM10/17) exhibit an altered substrate spectrum compared to their membrane-bound counterparts. A mass spectrometry-based N-terminomics approach identified 134 protein cleavage events in total and 45 common substrates for sADAM10/17 within the secretome of murine cardiomyocytes. Analysis of these cleavage sites confirmed previously identified amino acid preferences. Further in vitro studies verified fibronectin, cystatin C, sN-cadherin, PCPE-1 as well as sAPP as direct substrates of sADAM10 and/or sADAM17. Overall, we present the first degradome study for sADAM10/17, thereby introducing a new mode of proteolytic activity within the protease web.


Sujet(s)
Protéine ADAM10/métabolisme , Protéine ADAM17/métabolisme , Amyloid precursor protein secretases/métabolisme , Protéines membranaires/métabolisme , Metalloproteases/métabolisme , Acides aminés/métabolisme , Animaux , Lignée cellulaire , Cellules HEK293 , Humains , Souris , Myocytes cardiaques/métabolisme
16.
Mol Neurobiol ; 56(10): 7234-7245, 2019 Oct.
Article de Anglais | MEDLINE | ID: mdl-31004319

RÉSUMÉ

The low-density lipoprotein receptor-related protein-1 (LRP1) has a dual role in the metabolism of the amyloid precursor protein (APP). In cellular models, LRP1 enhances amyloid-ß (Aß) generation via APP internalization and thus its amyloidogenic processing. However, conditional knock-out studies in mice define LRP1 as an important mediator for the clearance of extracellular Aß from brain via cellular degradation or transcytosis across the blood-brain barrier (BBB). In order to analyze the net effect of LRP1 on production and clearance of Aß in vivo, we crossed mice with impaired LRP1 function with a mouse model of Alzheimer's disease (AD). Analysis of Aß metabolism showed that, despite reduced Aß clearance due to LRP1 inactivation in vivo, less Aß was found in cerebrospinal fluid (CSF) and brain interstitial fluid (ISF). Further analysis of APP metabolism revealed that impairment of LRP1 in vivo shifted APP processing from the Aß-generating amyloidogenic cleavage by beta-secretase to the non-amyloidogenic processing by alpha-secretase as shown by a decrease in extracellular Aß and an increase of soluble APP-α (sAPP-α). This shift in APP processing resulted in overall lower Aß levels and a reduction in plaque burden. Here, we present for the first time clear in vivo evidence that global impairment of LRP1's endocytosis function favors non-amyloidogenic processing of APP due to its reduced internalization and subsequently, reduced amyloidogenic processing. By inactivation of LRP1, the inhibitory effect on Aß generation overrules the simultaneous impaired Aß clearance, resulting in less extracellular Aß and reduced plaque deposition in a mouse model of AD.


Sujet(s)
Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Protéine-1 apparentée au récepteur des LDL/métabolisme , Motifs d'acides aminés , Animaux , Encéphale/métabolisme , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Protéine-1 apparentée au récepteur des LDL/composition chimique , Souris , Mutation/génétique , Plaque amyloïde/métabolisme
17.
FASEB J ; 33(6): 7490-7504, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30916990

RÉSUMÉ

Biologic activity of proteases is mainly characterized by the substrate specificity, tissue distribution, and cellular localization. The human metalloproteases meprin α and meprin ß share 41% sequence identity and exhibit a similar cleavage specificity with a preference for negatively charged amino acids. However, shedding of meprin α by furin on the secretory pathway makes it a secreted enzyme in comparison with the membrane-bound meprin ß. In this study, we identified human meprin α and meprin ß as forming covalently linked membrane-tethered heterodimers in the early endoplasmic reticulum, thereby preventing furin-mediated secretion of meprin α. Within this newly formed enzyme complex, meprin α was able to be activated on the cell surface and detected by cleavage of a novel specific fluorogenic peptide substrate. However, the known meprin ß substrates amyloid precursor protein and CD99 were not shed by membrane-tethered meprin α. On the other hand, being linked to meprin α, activation of or substrate cleavage by meprin ß on the cell surface was not altered. Interestingly, proteolytic activity of both proteases was increased in the heteromeric complex, indicating an increased proteolytic potential at the plasma membrane. Because meprins are susceptibility genes for inflammatory bowel disease (IBD), and to investigate the physiologic impact of the enzyme complex, we performed transcriptome analyses of intestinal mucosa from meprin-knockout mice. Comparison of the transcriptional gene analysis data with gene analyses of IBD patients revealed that different gene subsets were dysregulated if meprin α was expressed alone or in the enzyme complex, demonstrating the physiologic and pathophysiological relevance of the meprin heterodimer formation.-Peters, F., Scharfenberg, F., Colmorgen, C., Armbrust, F., Wichert, R., Arnold, P., Potempa, B., Potempa, J., Pietrzik, C. U., Häsler, R., Rosenstiel, P., Becker-Pauly, C. Tethering soluble meprin α in an enzyme complex to the cell surface affects IBD-associated genes.


Sujet(s)
Maladies inflammatoires intestinales/génétique , Metalloendopeptidases/métabolisme , Animaux , Membrane cellulaire/métabolisme , Cellules HeLa , Humains , Metalloendopeptidases/génétique , Souris , Souris knockout
18.
Acta Neuropathol ; 137(2): 239-257, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30426203

RÉSUMÉ

Brain accumulation and aggregation of amyloid-ß (Aß) peptides is a critical step in the pathogenesis of Alzheimer's disease (AD). Full-length Aß peptides (mainly Aß1-40 and Aß1-42) are produced through sequential proteolytic cleavage of the amyloid precursor protein (APP) by ß- and γ-secretases. However, studies of autopsy brain samples from AD patients have demonstrated that a large fraction of insoluble Aß peptides are truncated at the N-terminus, with Aß4-x peptides being particularly abundant. Aß4-x peptides are highly aggregation prone, but their origin and any proteases involved in their generation are unknown. We have identified a recognition site for the secreted metalloprotease ADAMTS4 (a disintegrin and metalloproteinase with thrombospondin motifs 4) in the Aß peptide sequence, which facilitates Aß4-x peptide generation. Inducible overexpression of ADAMTS4 in HEK293 cells resulted in the secretion of Aß4-40 but unchanged levels of Aß1-x peptides. In the 5xFAD mouse model of amyloidosis, Aß4-x peptides were present not only in amyloid plaque cores and vessel walls, but also in white matter structures co-localized with axonal APP. In the ADAMTS4-/- knockout background, Aß4-40 levels were reduced confirming a pivotal role of ADAMTS4 in vivo. Surprisingly, in the adult murine brain, ADAMTS4 was exclusively expressed in oligodendrocytes. Cultured oligodendrocytes secreted a variety of Aß species, but Aß4-40 peptides were absent in cultures derived from ADAMTS4-/- mice indicating that the enzyme was essential for Aß4-x production in this cell type. These findings establish an enzymatic mechanism for the generation of Aß4-x peptides. They further identify oligodendrocytes as a source of these highly amyloidogenic Aß peptides.


Sujet(s)
Protéine ADAMTS4/métabolisme , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Oligodendroglie/métabolisme , Maladie d'Alzheimer/anatomopathologie , Amyloid precursor protein secretases/métabolisme , Animaux , Encéphale/métabolisme , Encéphale/anatomopathologie , Modèles animaux de maladie humaine , Cellules HEK293 , Humains , Souris , Oligodendroglie/anatomopathologie , Fragments peptidiques/métabolisme , Plaque amyloïde/anatomopathologie
19.
Brain Behav Immun ; 73: 21-33, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30041013

RÉSUMÉ

The accumulation of neurotoxic amyloid-beta (Aß) in the brain is a characteristic hallmark of Alzheimer's disease (AD). The blood-brain barrier (BBB) provides a large surface area and has been shown to be an important mediator for removal of brain Aß. Both, the ABC transporter P-glycoprotein (ABCB1/P-gp) and the receptor low-density lipoprotein receptor-related protein 1 (LRP1) have been implicated to play crucial roles in Aß efflux from brain. Here, with immunoprecipitation experiments, co-immunostainings and dual inhibition of ABCB1/P-gp and LRP1, we show that both proteins are functionally linked, mediating a concerted transcytosis of Aß through endothelial cells. Late-onset AD risk factor Phosphatidylinositol binding clathrin assembly protein (PICALM) is associated with both ABCB1/P-gp and LRP1 representing a functional link and guiding both proteins through the brain endothelium. Together, our results give more mechanistic insight on Aß transport across the BBB and show that the functional interplay of different clearance proteins is needed for the rapid removal of Aß from the brain.


Sujet(s)
Glycoprotéine P/métabolisme , Protéines d'assemblage monomériques de la clathrine/physiologie , Récepteurs aux lipoprotéines LDL/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Glycoprotéine P/physiologie , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/physiologie , Animaux , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/physiologie , Encéphale/métabolisme , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Cellules endothéliales/physiologie , Protéine-1 apparentée au récepteur des LDL , Mâle , Souris , Souris knockout , Protéines d'assemblage monomériques de la clathrine/métabolisme , Fragments peptidiques/métabolisme , Culture de cellules primaires , Récepteurs aux lipoprotéines LDL/physiologie , Suidae , Transcytose/physiologie , Protéines suppresseurs de tumeurs/physiologie
20.
Bioorg Med Chem Lett ; 28(8): 1417-1422, 2018 05 01.
Article de Anglais | MEDLINE | ID: mdl-29463448

RÉSUMÉ

Rhomboids are intramembrane serine proteases with diverse physiological functions in organisms ranging from archaea to humans. Crystal structure analysis has provided a detailed understanding of the catalytic mechanism, and rhomboids have been implicated in various disease contexts. Unfortunately, the design of specific rhomboid inhibitors has lagged behind, and previously described small molecule inhibitors displayed insufficient potency and/or selectivity. Using a computer-aided approach, we focused on the discovery of novel scaffolds with reduced liabilities and the possibility for broad structural variations. Docking studies with the E. coli rhomboid GlpG indicated that 2-styryl substituted benzoxazinones might comprise novel rhomboid inhibitors. Protease in vitro assays confirmed activity of 2-styryl substituted benzoxazinones against GlpG but not against the soluble serine protease α-chymotrypsin. Furthermore, mass spectrometry analysis demonstrated covalent modification of the catalytic residue Ser201, corroborating the predicted mechanism of inhibition and the formation of an acyl enzyme intermediate. In conclusion, 2-styryl substituted benzoxazinones are a novel rhomboid inhibitor scaffold with ample opportunity for optimization.


Sujet(s)
Benzoxazines/composition chimique , Inhibiteurs de la sérine protéinase/composition chimique , Styrènes/composition chimique , Animaux , Benzoxazines/synthèse chimique , Domaine catalytique , Bovins , Chymotrypsine/composition chimique , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/composition chimique , Protéines de liaison à l'ADN/génétique , Drosophila/composition chimique , Protéines de Drosophila/métabolisme , Découverte de médicament , Endopeptidases/composition chimique , Endopeptidases/génétique , Dosages enzymatiques , Escherichia coli/enzymologie , Protéines Escherichia coli/antagonistes et inhibiteurs , Protéines Escherichia coli/composition chimique , Protéines Escherichia coli/génétique , Humains , Protéines membranaires/antagonistes et inhibiteurs , Protéines membranaires/composition chimique , Protéines membranaires/génétique , Simulation de docking moléculaire , Mutation , Sérine/composition chimique , Inhibiteurs de la sérine protéinase/synthèse chimique , Styrènes/synthèse chimique , Facteur de croissance transformant alpha/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...