Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
CNS Neurosci Ther ; 27(2): 174-185, 2021 02.
Article in English | MEDLINE | ID: mdl-32961023

ABSTRACT

INTRODUCTION: Presenilin enhancer2 (Pen-2) is an essential subunit of γ-secretase, which is a key protease responsible for the cleavage of amyloid precursor protein (APP) and Notch. Mutations on Pen-2 cause familial Alzheimer disease (AD). However, it remains unknown whether Pen-2 regulates neuronal survival and neuroinflammation in the adult brain. METHODS: Forebrain neuron-specific Pen-2 conditional knockout (Pen-2 cKO) mice were generated for this study. Pen-2 cKO mice expressing Notch1 intracellular domain (NICD) conditionally in cortical neurons were also generated. RESULTS: Loss of Pen-2 causes astrogliosis followed by age-dependent cortical atrophy and neuronal loss. Loss of Pen-2 results in microgliosis and enhanced inflammatory responses in the cortex. Expression of NICD in Pen-2 cKO cortices ameliorates neither neurodegeneration nor neuroinflammation. CONCLUSIONS: Pen-2 is required for neuronal survival in the adult cerebral cortex. The Notch signaling may not be involved in neurodegeneration caused by loss of Pen-2.


Subject(s)
Aging/metabolism , Amyloid Precursor Protein Secretases/deficiency , Cerebral Cortex/metabolism , Gliosis/metabolism , Neurons/metabolism , Receptors, Notch/deficiency , Aging/genetics , Aging/pathology , Amyloid Precursor Protein Secretases/genetics , Animals , Atrophy , Cerebral Cortex/pathology , Disease Progression , Female , Gene Deletion , Gliosis/genetics , Gliosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/pathology , Receptors, Notch/genetics
2.
J Neuroimmunol ; 349: 577423, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33074142

ABSTRACT

Following peripheral nerve injury, macrophages are recruited to the injury site from circulation to clear cellular debris. Injured ß-secretase 1 (BACE1) knockout mice have enhanced macrophage recruitment and debris clearance, which may be due to BACE1 activity in macrophages or the hypomyelination observed in BACE1 knockout mice. To assess if BACE1 expression by macrophages mediates enhanced macrophage recruitment we utilized mice with macrophage specific deletion of BACE1 and saw no increase in macrophage recruitment following injury. This study suggests that expression of BACE1 by macrophages may not be essential for increased recruitment observed previously in global BACE1 KO mice.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Aspartic Acid Endopeptidases/deficiency , Macrophages/metabolism , Nerve Regeneration/physiology , Peripheral Nerve Injuries/metabolism , Animals , Cells, Cultured , Female , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Nerve Injuries/pathology
3.
Biochem Biophys Res Commun ; 528(3): 440-446, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32507599

ABSTRACT

Previous studies have shown that the occurrence of atherosclerosis is closely related to changes of α2, 6-sialic acid transferase I (ST6Gal-I). Bace1 has been identified as a protease responsible for the cleavage and secretion of Golgi-resident ST6Gal-I. There have been only a few attempts to clarify the direct connection between Bace1 and atherosclerosis. The purpose of this study was to investigate the relationship between Bace1 gene and atherosclerosis. Expressions of Bace1 protein and mRNA in ApoE-/- mice fed on high-fat diet were evaluated and the development of atherosclerosis was assessed in Bace1-/- mice fed on high-fat diet. In vitro, the expression of Bace1 gene was detected in foam cell model and the formation of foam cells was examined after knocking down Bace1 by siRNA. We observed a significant increase in Bace1 expression in the aortic root in the model of atherosclerosis in ApoE-/-mice. The expression of Bace1 protein and mRNA levels had a remarkable increase in high-fat group. After knocking out the Bace1 gene, serum lipid levels were significantly lower and intimal thickness was obvious thinner than those in wild-type mice with high-fat diet. Expression of Bace1 protein and mRNA levels were significantly elevated in foam cell. The formation of foam cells was blocked when Bace1 was knocked down by siRNA interferes. Our results suggested that elevated Bace1 gene had a positive role in the progression of atherosclerosis. Affecting the glycosyltransferase may be one of its mechanisms.


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Atherosclerosis/etiology , Foam Cells/metabolism , Amyloid Precursor Protein Secretases/deficiency , Animals , Aorta/metabolism , Aorta/pathology , Aspartic Acid Endopeptidases/deficiency , Atherosclerosis/genetics , Atherosclerosis/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Foam Cells/pathology , Gene Knockdown Techniques , In Vitro Techniques , Male , Mice , Mice, Knockout , Mice, Knockout, ApoE , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Up-Regulation
4.
Angiogenesis ; 23(3): 443-458, 2020 08.
Article in English | MEDLINE | ID: mdl-32385775

ABSTRACT

During the initiation of pregnancy, the vasculature of the implantation site expands rapidly, yet little is known about this process or its role in fertility. Here, we report that endothelial-specific deletion of a disintegrin and metalloprotease 10 (ADAM10), an essential regulator of Notch signaling, results in severe subfertility in mice. We found that implantation sites develop until 5.5 days post conception (dpc) but are resorbed by 6.5 dpc in A10ΔEC mice. Analysis of the mutant implantation sites showed impaired decidualization and abnormal vascular patterning compared to controls. Moreover, RNA-seq analysis revealed changes in endothelial cell marker expression consistent with defective ADAM10/Notch signaling in samples from A10ΔEC mice, suggesting that this signaling pathways is essential for the physiological function of endometrial endothelial cells during early pregnancy. Our findings raise the possibility that impaired endothelial cell function could be a cause for repeated pregnancy loss (RPL) and infertility in humans.


Subject(s)
ADAM10 Protein/deficiency , Amyloid Precursor Protein Secretases/deficiency , Decidua/metabolism , Fertility , Gene Deletion , Membrane Proteins/deficiency , Receptors, Notch/metabolism , Signal Transduction , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Female , Membrane Proteins/metabolism , Mice , Mice, Knockout , Pregnancy , Receptors, Notch/genetics
5.
Sci Rep ; 9(1): 19877, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882662

ABSTRACT

BACE1 is the first enzyme involved in APP processing, thus it is a strong therapeutic target candidate for Alzheimer's disease. The observation of deleterious phenotypes in BACE1 Knock-out (KO) mouse models (germline and conditional) raised some concerns on the safety and tolerability of BACE1 inhibition. Here, we have employed a tamoxifen inducible BACE1 conditional Knock-out (cKO) mouse model to achieve a controlled partial depletion of BACE1 in adult mice. Biochemical and behavioural characterization was performed at two time points: 4-5 months (young mice) and 12-13 months (aged mice). A ~50% to ~70% BACE1 protein reduction in hippocampus and cortex, respectively, induced a significant reduction of BACE1 substrates processing and decrease of Aßx-40 levels at both ages. Hippocampal axonal guidance and peripheral nerve myelination were not affected. Aged mice displayed a CA1 long-term potentiation (LTP) deficit that was not associated with memory impairment. Our findings indicate that numerous phenotypes observed in germline BACE1 KO reflect a fundamental role of BACE1 during development while other phenotypes, observed in adult cKO, may be absent when partially rather than completely deleting BACE1. However, we demonstrated that partial depletion of BACE1 still induces CA1 LTP impairment, supporting a role of BACE1 in synaptic plasticity in adulthood.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Aspartic Acid Endopeptidases/deficiency , Axon Guidance/genetics , CA1 Region, Hippocampal , Cerebral Cortex , Gene Deletion , Neuronal Plasticity/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/pathology , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Mice , Mice, Knockout
6.
ChemMedChem ; 14(22): 1894-1910, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31657130

ABSTRACT

The ß-site amyloid precursor protein cleaving enzyme 1 (BACE1, also known as ß-secretase) is a promising target for the treatment of Alzheimer's disease. A pKa lowering approach over the initial leads was adopted to mitigate hERG inhibition and P-gp efflux, leading to the design of 6-CF3 dihydrothiazine 8 (N-(3-((4S,6S)-2-amino-4-methyl-6-(trifluoromethyl)-5,6-dihydro-4H-1,3-thiazin-4-yl)-4-fluorophenyl)-5-cyanopicolinamide). Optimization of 8 led to the discovery of 15 (N-(3-((4S,6S)-2-amino-4-methyl-6-(trifluoromethyl)-5,6-dihydro-4H-1,3-thiazin-4-yl)-4-fluorophenyl)-5-(fluoromethoxy)pyrazine-2-carboxamide) with an excellent balance of potency, hERG inhibition, P-gp efflux, and metabolic stability. Oral administration of 8 elicited robust Aß reduction in dog even at 0.16 mg/kg. Reflecting the reduced hERG inhibitory activity, no QTc prolongation was observed at high doses. The potential for reactive metabolite formation of 15 was realized in a nucleophile trapping assay using [14 C]-KCN in human liver microsomes. Utilizing covalent binding (CVB) in human hepatocytes and the maximum projected human dosage, the daily CVB burden of 15 was calculated to be at an acceptable value of below 1 mg/day. However, hepatotoxicity was observed when 15 was subjected to a two-week tolerance study in dog, which prevented further evaluation of this compound.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Oxazines/pharmacology , Thiazines/pharmacology , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/deficiency , Aspartic Acid Endopeptidases/metabolism , Dogs , Dose-Response Relationship, Drug , Drug Design , Hepatocytes/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Oxazines/chemistry , Rats , Structure-Activity Relationship , Thiazines/administration & dosage , Thiazines/chemistry
7.
Brain Struct Funct ; 224(3): 1279-1290, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30701309

ABSTRACT

BACE1 is a ß-secretase involved in the cleavage of amyloid precursor protein and the pathogenesis of Alzheimer's disease (AD). The entorhinal cortex and the dentate gyrus are important for learning and memory, which are affected in the early stages of AD. Since BACE1 is a potential target for AD therapy, it is crucial to understand its physiological role in these brain regions. Here, we examined the function of BACE1 in the dentate gyrus. We show that loss of BACE1 in the dentate gyrus leads to increased granule cell excitability, indicated by enhanced efficiency of synaptic potentials to generate granule cell spikes. The increase in granule cell excitability was accompanied by prolonged paired-pulse inhibition, altered network gamma oscillations, and impaired synaptic plasticity at entorhinal-dentate synapses of the perforant path. In summary, this is the first detailed electrophysiological study of BACE1 deletion at the network level in vivo. The results suggest that BACE1 is important for normal dentate gyrus network function. This has implications for the use of BACE1 inhibitors as therapeutics for AD therapy, since BACE1 inhibition could similarly disrupt synaptic plasticity and excitability in the entorhinal-dentate circuitry.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Aspartic Acid Endopeptidases/deficiency , Biological Clocks/genetics , Dentate Gyrus/cytology , Neuronal Plasticity/genetics , Perforant Pathway/cytology , Action Potentials/genetics , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Biophysics , Electric Stimulation , Excitatory Postsynaptic Potentials/genetics , Male , Mice , Mice, Knockout , Neurons , Reaction Time/genetics , Time Factors
8.
Sci Transl Med ; 10(459)2018 09 19.
Article in English | MEDLINE | ID: mdl-30232227

ABSTRACT

ß-Site APP (amyloid precursor protein) cleaving enzyme 1 (BACE1) is the ß-secretase enzyme that initiates production of the toxic amyloid-ß peptide that accumulates in the brains of patients with Alzheimer's disease (AD). Hence, BACE1 is a prime therapeutic target, and several BACE1 inhibitor drugs are currently being tested in clinical trials for AD. However, the safety of BACE1 inhibition is unclear. Germline BACE1 knockout mice have multiple neurological phenotypes, although these could arise from BACE1 deficiency during development. To address this question, we report that tamoxifen-inducible conditional BACE1 knockout mice in which the Bace1 gene was ablated in the adult largely lacked the phenotypes observed in germline BACE1 knockout mice. However, one BACE1-null phenotype was induced after Bace1 gene deletion in the adult mouse brain. This phenotype showed reduced length and disorganization of the hippocampal mossy fiber infrapyramidal bundle, the axonal pathway of dentate gyrus granule cells that is maintained by neurogenesis in the mouse brain. This defect in axonal organization correlated with reduced BACE1-mediated cleavage of the neural cell adhesion protein close homolog of L1 (CHL1), which has previously been associated with axon guidance. Although our results indicate that BACE1 inhibition in the adult mouse brain may avoid phenotypes associated with BACE1 deficiency during embryonic and postnatal development, they also suggest that BACE1 inhibitor drugs developed for treating AD may disrupt the organization of an axonal pathway in the hippocampus, an important structure for learning and memory.


Subject(s)
Aging/metabolism , Amyloid Precursor Protein Secretases/deficiency , Aspartic Acid Endopeptidases/deficiency , Axons/metabolism , Hippocampus/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Animals, Newborn , Apoptosis , Aspartic Acid Endopeptidases/metabolism , Cognition , Epilepsy/pathology , Epilepsy/physiopathology , Gene Deletion , Hippocampus/pathology , Hippocampus/physiopathology , Long-Term Potentiation , Memory Disorders/pathology , Memory Disorders/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/metabolism , Neurogenesis , Phenotype , Substrate Specificity
9.
J Biophotonics ; 11(12): e201800174, 2018 12.
Article in English | MEDLINE | ID: mdl-29968387

ABSTRACT

Mutations in NCSTN gene (encoding for nicastrin protein) are associated with hidradenitis suppurativa (HS), a chronic inflammatory disease involving hair follicles. HS is clinically handled with drugs but the most severe cases are treated with surgery. Photobiomodulation (PBM) therapy, already used in the treatment of skin diseases such as acne, herpes virus lesions, ultraviolet damage, vitiligo, hypertrophic scar, keloid, burn, psoriasis and diabetic chronic wounds, could be beneficial as an adjuvant supportive treatment to promote and foster the healing process after skin excision in HS. The effects of PBM therapy in promoting the wound closure are evaluated in a HaCaT cells NCSTN-/-, assessing cell metabolism, migration rate, proliferation and cell cycle progression. In our experimental model, PBM exerts a potent action on metabolism of mutated keratinocytes, incrementing adenosine triphosphate (ATP) production at 2 hours, while after 24 hours an increase of metabolism with a decrement of intracellular ATP levels were recorded. Moreover, PBM speeds up the wound closure, inducing cells' migration without affecting their proliferation.Based on our findings, we suggest the use of PBM in HS patients, who undergo major surgery with large skin excision.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Gene Knockout Techniques , Low-Level Light Therapy , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Wound Healing/radiation effects , Cell Cycle/radiation effects , Cell Line , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Humans , Keratinocytes/metabolism , Keratinocytes/radiation effects
10.
J Exp Med ; 215(3): 927-940, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29444819

ABSTRACT

BACE1 initiates the generation of the ß-amyloid peptide, which likely causes Alzheimer's disease (AD) when accumulated abnormally. BACE1 inhibitory drugs are currently being developed to treat AD patients. To mimic BACE1 inhibition in adults, we generated BACE1 conditional knockout (BACE1fl/fl) mice and bred BACE1fl/fl mice with ubiquitin-CreER mice to induce deletion of BACE1 after passing early developmental stages. Strikingly, sequential and increased deletion of BACE1 in an adult AD mouse model (5xFAD) was capable of completely reversing amyloid deposition. This reversal in amyloid deposition also resulted in significant improvement in gliosis and neuritic dystrophy. Moreover, synaptic functions, as determined by long-term potentiation and contextual fear conditioning experiments, were significantly improved, correlating with the reversal of amyloid plaques. Our results demonstrate that sustained and increasing BACE1 inhibition in adults can reverse amyloid deposition in an AD mouse model, and this observation will help to provide guidance for the proper use of BACE1 inhibitors in human patients.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Amyloid/metabolism , Aspartic Acid Endopeptidases/deficiency , Cognition , Gene Deletion , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Behavior, Animal , Disease Models, Animal , Integrases/metabolism , Learning , Long-Term Potentiation , Mice, Transgenic , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology
11.
Mol Neurobiol ; 55(1): 709-717, 2018 01.
Article in English | MEDLINE | ID: mdl-28004339

ABSTRACT

BACE1 encodes for the beta-site amyloid precursor protein cleaving enzyme 1 or ß-secretase. Genetic deletion of Bace1 leads to behavioral alterations and affects midbrain dopaminergic signaling and memory processes. In order to further understand the role of BACE1 in brain function and behavior, we performed microarray transcriptome profiling and gene pathway analysis in the hippocampus of BACE1-deficient mice compared to wild type. We identified a total of 91 differentially expressed genes (DEGs), mostly enriched in pathways related to the immune and inflammation systems, particularly IL-9 and NF-κB activation pathways. Serum levels of IL-9 were elevated in BACE1-deficient mice. Our network analysis supports an intimate connection between immune response via NF-κB and BACE1 signaling through the NRG1/Akt1 pathway. Our findings warrant future mechanistic studies to determine if BACE1 signaling and the IL-9 pathway interact to alter behavior and brain function. This study opens new avenues in the investigation of hippocampus-related neuroimmunological and neuroinflammation-associated disorders.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Aspartic Acid Endopeptidases/deficiency , Immune System/metabolism , Signal Transduction , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Chemokines/blood , Gene Expression Profiling , Gene Regulatory Networks , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism
12.
CNS Neurosci Ther ; 23(11): 885-893, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28960838

ABSTRACT

AIMS: This study was to investigate whether cell proliferation and adult neurogenesis are affected at early neurodegenerative stage when neuron loss has not begun to display. METHODS AND RESULTS: Forebrain-specific nicastrin (NCT) conditional knockout (cKO) mice were generated by crossing NCTf/f with CaMKIIα-Cre Tg mice. BrdU was used as a lineage tracer to label proliferating neural progenitor cells (NPCs). Immunohistochemistry (IHC) on BrdU indicated that the total number of BrdU positive (+) cells was increased in NCT cKO mice. IHC on doublecortin (DCX) showed that the total number of DCX+ cells was also increased in NCT cKO mice. NCT cKO mice displayed significant astrogliosis as well. However, NCT cKO mice at 3 months did not show significant neuronal death or synaptic loss. CONCLUSIONS: NCT-dependent γ-secretase activity plays an important role in cell proliferation and immature neuron generation. Enhanced neurogenesis and astrogliosis may be early cellular events prior to the occurrence of neuronal death in neurodegenerative disease.


Subject(s)
Astrocytes/physiology , Cell Proliferation/physiology , Gliosis/physiopathology , Neurodegenerative Diseases/physiopathology , Neurogenesis/physiology , Neurons/physiology , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Animals , Astrocytes/pathology , Cell Death/physiology , Disease Models, Animal , Disease Progression , Doublecortin Protein , Gliosis/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice, Transgenic , Neurodegenerative Diseases/pathology , Neurons/pathology , Synapses/pathology , Synapses/physiology
13.
J Immunol ; 199(7): 2305-2315, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28814605

ABSTRACT

The proper regulation of ICOS and ICOS ligand (ICOSL) has been shown to be essential for maintaining proper immune homeostasis. Loss of either protein results in defective humoral immunity, and overexpression of ICOS results in aberrant Ab production resembling lupus. How ICOSL is regulated in response to ICOS interaction is still unclear. We demonstrate that a disintegrin and metalloproteinase (ADAM)10 is the primary physiological sheddase of ICOSL in mice and humans. Using an in vivo system in which ADAM10 is deleted only on B cells, elevated levels of ICOSL were seen. This increase is also seen when ADAM10 is deleted from human B cell lines. Identification of the primary sheddase has allowed the characterization of a novel mechanism of ICOS regulation. In wild-type mice, interaction of ICOS/ICOSL results in ADAM10-induced shedding of ICOSL on B cells and moderate ICOS internalization on T cells. When this shedding is blocked, excessive ICOS internalization occurs. This results in severe defects in T follicular helper development and TH2 polarization, as seen in a house dust mite exposure model. In addition, enhanced TH1 and TH17 immune responses are seen in experimental autoimmune encephalomyelitis. Blockade of ICOSL rescues T cell ICOS surface expression and rescues, at least in part, T follicular helper numbers and the abnormal Ab production previously reported in these mice. Overall, we propose a novel regulation of the ICOS/ICOSL axis, with ADAM10 playing a direct role in regulating ICOSL, as well as indirectly regulating ICOS, thus controlling ICOS/ICOSL-dependent responses.


Subject(s)
B-Lymphocytes/immunology , Gene Expression Regulation , Inducible T-Cell Co-Stimulator Ligand/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , ADAM10 Protein/deficiency , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Homeostasis , Humans , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Ligand/immunology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Pyroglyphidae/immunology , Th1 Cells/immunology , Th17 Cells/immunology
14.
J Immunol ; 199(2): 666-676, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28600292

ABSTRACT

The recruitment of blood leukocytes across the endothelium to sites of tissue infection is central to inflammation, but also promotes chronic inflammatory diseases. A disintegrin and metalloproteinase 10 (ADAM10) is a ubiquitous transmembrane molecular scissor that is implicated in leukocyte transmigration by proteolytically cleaving its endothelial substrates. These include VE-cadherin, a homotypic adhesion molecule that regulates endothelial barrier function, and transmembrane chemokines CX3CL1 and CXCL16, which have receptors on leukocytes. However, a definitive role for endothelial ADAM10 in transmigration of freshly isolated primary leukocytes under flow has not been demonstrated, and the relative importance of distinct ADAM10 substrates is unknown. Emerging evidence suggests that ADAM10 can be regarded as six different molecular scissors with different substrate specificities, depending on which of six TspanC8 tetraspanins it is associated with, but TspanC8s remain unstudied in leukocyte transmigration. In the current study, ADAM10 knockdown on primary HUVECs was found to impair transmigration of freshly isolated human peripheral blood T lymphocytes, but not neutrophils or B lymphocytes, in an in vitro flow assay. This impairment was due to delayed transmigration rather than a complete block, and was overcome in the presence of neutrophils. Transmigration of purified lymphocytes was dependent on ADAM10 regulation of VE-cadherin, but not CX3CL1 and CXCL16. Tspan5 and Tspan17, the two most closely related TspanC8s by sequence, were the only TspanC8s that regulated VE-cadherin expression and were required for lymphocyte transmigration. Therefore endothelial Tspan5- and Tspan17-ADAM10 complexes may regulate inflammation by maintaining normal VE-cadherin expression and promoting T lymphocyte transmigration.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Antigens, CD/genetics , Cadherins/genetics , Membrane Proteins/metabolism , T-Lymphocytes/physiology , Tetraspanins/metabolism , Transendothelial and Transepithelial Migration , ADAM10 Protein/deficiency , ADAM10 Protein/genetics , ADAM10 Protein/immunology , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/immunology , Antigens, CD/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Cadherins/metabolism , Cells, Cultured , Chemokine CX3CL1/genetics , Chemokine CX3CL1/immunology , Chemokine CXCL16 , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Endothelial Cells/immunology , Endothelial Cells/physiology , Humans , Inflammation/immunology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/immunology , Neutrophils/immunology , Neutrophils/physiology , Receptors, Scavenger/genetics , Receptors, Scavenger/immunology , T-Lymphocytes/immunology , Tetraspanins/genetics , Tetraspanins/immunology
15.
Curr Alzheimer Res ; 14(4): 453-459, 2017.
Article in English | MEDLINE | ID: mdl-27784219

ABSTRACT

BACKGROUND: ß-secretase (BACE1) is a type 1 transmembrane protein implicated in Alzheimer's Disease (AD) pathogenesis. Cleavage of Amyloid Precursor Protein (APP), initiated by BACE1 and followed by γ-secretase, leads to the formation of toxic Aß peptides. Increased levels of BACE1 have been detected in the CSF of AD patients compared to age-matched healthy controls indicating that neurodegenerative conditions induce shedding of BACE1. OBJECTIVE: To mimic such conditions, we examined whether serum deprivation stimulates proteolysis-dependent secretion of BACE1. METHOD: Detection of BACE1 secretion in BACE1 overexpressing cells or ADAM10/ADAM17 knockout fibroblasts cultured under serum deprivation conditions, using western blot analysis. RESULTS: We found that serum deprivation of U251 neuroblastoma or HEK293T cells overexpressing BACE1 stimulated secretion of BACE1. Using ADAM10/ADAM17 knockout fibroblasts and inhibitors of both ADAM10 and ADAM17, we obtained data indicating that these proteases are involved in serum-starvation induced shedding of BACE1. This is unexpected since BACE1 is localized mainly in lipid rafts while ADAM10 is localized mainly in nonlipid raft domains. We hypothesized that serum deprivation results in alterations in the lipid composition of the membrane which can alter the localization of ADAM10 and BACE1. In support, we obtained results indicating that extraction of membrane cholesterol following incubation with methyl ß cyclodextrin potentiated the effect of serum deprivation. Secreted BACE1 was also found to be enzymatically active towards immunoprecipiated full length APP. CONCLUSION: Serum starvation induces ADAM10-mediated BACE1 secretion.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Serum/metabolism , Stress, Physiological/physiology , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/deficiency , ADAM10 Protein/genetics , ADAM17 Protein/antagonists & inhibitors , ADAM17 Protein/deficiency , ADAM17 Protein/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/genetics , Cell Line, Tumor , Cholesterol/metabolism , Culture Media, Serum-Free , Fibroblasts/drug effects , Fibroblasts/metabolism , HEK293 Cells , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/genetics , Stress, Physiological/drug effects , Surface-Active Agents/pharmacology , beta-Cyclodextrins/pharmacology
16.
J Alzheimers Dis ; 54(2): 535-47, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27472877

ABSTRACT

Neuronal ELAV/Hu (nELAV) are RNA-binding proteins that mainly regulate gene expression by increasing the stability and/or translation rate of target mRNAs bearing ARE (adenine and uracil-rich elements) sequences. Among nELAV target transcripts there is ADAM10, an α-secretase involved in the non-amyloidogenic processing of the amyloid-ß protein precursor (AßPP) which leads to the production of the neuroprotective sAßPPα peptide. The aim of this study was to evaluate if nELAV depletion affects ADAM10 expression in human SH-SY5Y neuroblastoma cells. We also studied the effects of Bryostatin-1, a molecule able to activate nELAV protein cascade. The specific HuD/nELAV gene silencing decreased both nELAV and ADAM10 protein contents; similar results were obtained by Aß40 treatment in wild-type SH-SY5Y cells. In HuD-silenced cells, the exposure to Bryostatin-1 counteracted both nELAV and ADAM10 proteins downregulation, by restoring nELAV/ADAM10 basal levels. We also found that sAßPPα release, which seemed not to be compromised by Aß40 challenge or HuD-silencing, was favored by Bryostatin-1. Overall, these findings strongly suggest that a deficiency in nELAV content negatively affects ADAM10 expression and may play a role in neurodegenerative diseases, which may benefit by molecules activating ELAV cascade.


Subject(s)
ADAM10 Protein/deficiency , Amyloid Precursor Protein Secretases/deficiency , ELAV-Like Protein 1/metabolism , Membrane Proteins/deficiency , Neuroblastoma/metabolism , Protein Kinase C/metabolism , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/toxicity , Cell Line, Tumor , ELAV-Like Protein 1/genetics , Humans , Membrane Proteins/genetics , Neuroblastoma/genetics , Peptide Fragments/toxicity , Protein Kinase C/genetics
17.
Circ Res ; 119(4): 519-31, 2016 Aug 05.
Article in English | MEDLINE | ID: mdl-27354212

ABSTRACT

RATIONALE: Endothelial Notch signaling is critical for early vascular development and survival. Yet, previously described mice lacking endothelial a disintegrin and metalloproteinase 10 (ADAM10), a key regulator of Notch signaling, survived into adulthood with organ-specific vascular defects. These findings raised questions about whether these vascular defects were related to Notch signaling or other functions of ADAM10. OBJECTIVE: The aims of the study are to determine whether compensatory or redundant functions of ADAM17 in Notch signaling can explain the survival of Adam10ΔEC mice, explore the contribution of different Tie2-Cre transgenes to the differences in survival, and establish whether the Adam10ΔEC vascular phenotypes can be recapitulated by inactivation of Notch receptors in endothelial cells. METHODS AND RESULTS: Mice lacking ADAM10 and ADAM17 in endothelial cells (Adam10/Adam17ΔEC), which survived postnatally with organ-specific vascular defects, resembled Adam10ΔEC mice. In contrast, Adam10ΔEC mice generated with the Tie2Cre transgene previously used to inactivate endothelial Notch (Adam10ΔEC(Flv)) died by E10.5. Quantitative polymerase chain reaction analysis demonstrated that Cre-mediated recombination occurs earlier in Adam10ΔEC(Flv) mice than in the previously described Adam10ΔEC mice. Finally, mice lacking endothelial Notch1 (Notch1ΔEC) share some organ-specific vascular defects with Adam10ΔEC mice, whereas Notch4(-/-) mice lacking endothelial Notch1 (Notch1ΔEC/Notch4(-/-)) had defects in all vascular beds affected in Adam10ΔEC mice. CONCLUSIONS: Our results argue against a major role for ADAM17 in endothelial Notch signaling and clarify the difference in phenotypes of previously described mice lacking ADAM10 or Notch in endothelial cells. Most notably, these findings uncover new roles for Notch signaling in the development of organ-specific vascular beds.


Subject(s)
ADAM10 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , Blood Circulation/physiology , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Receptor, Notch1/physiology , Receptors, Notch/physiology , Regional Blood Flow/physiology , Signal Transduction/physiology , ADAM10 Protein/deficiency , Amyloid Precursor Protein Secretases/deficiency , Animals , Endothelial Cells/physiology , Female , Membrane Proteins/deficiency , Mice , Mice, Knockout , Mice, Transgenic , Pregnancy , Proto-Oncogene Proteins/deficiency , Receptor, Notch1/deficiency , Receptor, Notch4 , Receptors, Notch/deficiency
18.
Oncotarget ; 7(14): 17431-41, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26942887

ABSTRACT

UNLABELLED: A Disintegrin And Metalloprotease (ADAM) 10 exerts essential roles during organ development and tissue integrity in different organs, mainly through activation of the Notch pathway. However, only little is known about its implication in liver tissue physiology. Here we show that in contrast to its role in other tissues, ADAM10 is dispensable for the Notch2-dependent biliary tree formation. However, we demonstrate that expression of bile acid transporters is dependent on ADAM10. Consequently, mice deficient for Adam10 in hepatocytes, cholangiocytes and liver progenitor cells develop spontaneous hepatocyte necrosis and concomitant liver fibrosis. We furthermore observed a strongly augmented ductular reaction in 15-week old ADAM10(Δhep/Δch) mice and demonstrate that c-Met dependent liver progenitor cell activation is enhanced. Additionally, liver progenitor cells are primed to hepatocyte differentiation in the absence of ADAM10. These findings show that ADAM10 is a novel central node controlling liver tissue homeostasis. HIGHLIGHTS: Loss of ADAM10 in murine liver results in hepatocyte necrosis and concomitant liver fibrosis. ADAM10 directly regulates expression of bile acid transporters but is dispensable for Notch2-dependent formation of the biliary system. Activation of liver progenitor cells is enhanced through increased c-Met signalling, in the absence of ADAM10. Differentiation of liver progenitor cells to hepatocytes is augmented in the absence of ADAM10.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Liver/metabolism , Membrane Proteins/metabolism , ADAM10 Protein/deficiency , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Animals , Carrier Proteins/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , Down-Regulation , Hepatocytes/metabolism , Hepatocytes/pathology , Homeostasis , Liver/cytology , Liver/pathology , Membrane Glycoproteins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Necrosis , Receptor, Notch2/metabolism , Signal Transduction
20.
Elife ; 52016 Jan 23.
Article in English | MEDLINE | ID: mdl-26802628

ABSTRACT

Metzincin metalloproteases have major roles in intercellular communication by modulating the function of membrane proteins. One of the proteases is the a-disintegrin-and-metalloprotease 10 (ADAM10) which acts as alpha-secretase of the Alzheimer's disease amyloid precursor protein. ADAM10 is also required for neuronal network functions in murine brain, but neuronal ADAM10 substrates are only partly known. With a proteomic analysis of Adam10-deficient neurons we identified 91, mostly novel ADAM10 substrate candidates, making ADAM10 a major protease for membrane proteins in the nervous system. Several novel substrates, including the neuronal cell adhesion protein NrCAM, are involved in brain development. Indeed, we detected mistargeted axons in the olfactory bulb of conditional ADAM10-/- mice, which correlate with reduced cleavage of NrCAM, NCAM and other ADAM10 substrates. In summary, the novel ADAM10 substrates provide a molecular basis for neuronal network dysfunctions in conditional ADAM10-/- mice and demonstrate a fundamental function of ADAM10 in the brain.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Axons/enzymology , Membrane Proteins/metabolism , Synapses/enzymology , ADAM10 Protein/deficiency , Amyloid Precursor Protein Secretases/deficiency , Animals , Axons/chemistry , Brain/embryology , Brain/enzymology , Gene Knockdown Techniques , Gene Knockout Techniques , Membrane Proteins/deficiency , Mice , Proteome/analysis , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...