Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 242
Filter
1.
J Labelled Comp Radiopharm ; 66(4-6): 145-154, 2023.
Article in English | MEDLINE | ID: mdl-36931890

ABSTRACT

The generation of amyloid beta peptides that aggregate in the brain is believed to play a major role in Alzheimer's disease. In theory, the inhibition of beta-site amyloid precursor protein-cleaving enzyme 1 (BACE1), which catalyzes the initial rate-limiting step in amyloid beta production, may slow or stop Alzheimer's disease. Herein, we report the preparation of two potent BACE1 inhibitors, BI 1147560 (1) and BI 1181181 (2), labeled with carbon-14 and with deuterium. The use of advanced key chiral intermediates like 3 and 5 shortened the carbon-14 syntheses of these two compounds to five and six steps, respectively, and helped in preparing them with very high chemical purity and enantiomeric excess without deviating from the process chemistry route. For the deuterium synthesis, oxetan-3-ylmethanamine [2 H6 ]-7 and 2-fluoro-2-methylpropan-1-amine [2 H6 ]-9 were prepared then used with the chiral intermediate 5 to furnish deuterium labeled 1 and 2, respectively.


Subject(s)
Alzheimer Disease , Humans , Amyloid beta-Peptides , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/physiology , Carbon Radioisotopes , Deuterium , Enzyme Inhibitors
2.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Article in English | MEDLINE | ID: mdl-34526403

ABSTRACT

The spleen contains phenotypically and functionally distinct conventional dendritic cell (cDC) subpopulations, termed cDC1 and cDC2, which each can be divided into several smaller and less well-characterized subsets. Despite advances in understanding the complexity of cDC ontogeny by transcriptional programming, the significance of posttranslational modifications in controlling tissue-specific cDC subset immunobiology remains elusive. Here, we identified the cell-surface-expressed A-disintegrin-and-metalloproteinase 10 (ADAM10) as an essential regulator of cDC1 and cDC2 homeostasis in the splenic marginal zone (MZ). Mice with a CD11c-specific deletion of ADAM10 (ADAM10ΔCD11c) exhibited a complete loss of splenic ESAMhi cDC2A because ADAM10 regulated the commitment, differentiation, and survival of these cells. The major pathways controlled by ADAM10 in ESAMhi cDC2A are Notch, signaling pathways involved in cell proliferation and survival (e.g., mTOR, PI3K/AKT, and EIF2 signaling), and EBI2-mediated localization within the MZ. In addition, we discovered that ADAM10 is a molecular switch regulating cDC2 subset heterogeneity in the spleen, as the disappearance of ESAMhi cDC2A in ADAM10ΔCD11c mice was compensated for by the emergence of a Clec12a+ cDC2B subset closely resembling cDC2 generally found in peripheral lymph nodes. Moreover, in ADAM10ΔCD11c mice, terminal differentiation of cDC1 was abrogated, resulting in severely reduced splenic Langerin+ cDC1 numbers. Next to the disturbed splenic cDC compartment, ADAM10 deficiency on CD11c+ cells led to an increase in marginal metallophilic macrophage (MMM) numbers. In conclusion, our data identify ADAM10 as a molecular hub on both cDC and MMM regulating their transcriptional programming, turnover, homeostasis, and ability to shape the anatomical niche of the MZ.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Dendritic Cells/metabolism , Membrane Proteins/metabolism , ADAM10 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , Animals , Antigen-Presenting Cells/metabolism , CD11c Antigen/metabolism , Cell Differentiation , Cell Proliferation , Female , Homeostasis , Lymphoid Tissue/metabolism , Macrophages/metabolism , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Signal Transduction , Spleen/cytology , Spleen/metabolism
3.
Int J Mol Sci ; 22(13)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201472

ABSTRACT

The ubiquitously expressed transmembrane protein a disintegrin and metalloproteinase 10 (ADAM10) functions as a "molecular scissor", by cleaving the extracellular regions from its membrane protein substrates in a process termed ectodomain shedding. ADAM10 is known to have over 100 substrates including Notch, amyloid precursor protein, cadherins, and growth factors, and is important in health and implicated in diseases such as cancer and Alzheimer's. The tetraspanins are a superfamily of membrane proteins that interact with specific partner proteins to regulate their intracellular trafficking, lateral mobility, and clustering at the cell surface. We and others have shown that ADAM10 interacts with a subgroup of six tetraspanins, termed the TspanC8 subgroup, which are closely related by protein sequence and comprise Tspan5, Tspan10, Tspan14, Tspan15, Tspan17, and Tspan33. Recent evidence suggests that different TspanC8/ADAM10 complexes have distinct substrates and that ADAM10 should not be regarded as a single scissor, but as six different TspanC8/ADAM10 scissor complexes. This review discusses the published evidence for this "six scissor" hypothesis and the therapeutic potential this offers.


Subject(s)
ADAM10 Protein/physiology , Tetraspanins/physiology , Amyloid Precursor Protein Secretases/physiology , Animals , Cadherins/metabolism , Humans , Membrane Proteins/metabolism , Membrane Proteins/physiology , Molecular Targeted Therapy/methods , Tetraspanins/chemistry
4.
Sci Rep ; 11(1): 11414, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34075077

ABSTRACT

ADAM10 and ADAM17 are proteases that affect multiple signalling pathways by releasing molecules from the cell surface. As their substrate specificities partially overlaps, we investigated their concurrent role in liver regeneration and fibrosis, using three liver-specific deficient mouse lines: ADAM10- and ADAM17-deficient lines, and a line deficient for both proteases. In the model of partial hepatectomy, double deficient mice exhibited decreased AKT phosphorylation, decreased release of EGFR activating factors and lower shedding of HGF receptor c-Met. Thus, simultaneous ablation of ADAM10 and ADAM17 resulted in inhibited EGFR signalling, while HGF/c-Met signalling pathway was enhanced. In contrast, antagonistic effects of ADAM10 and ADAM17 were observed in the model of chronic CCl4 intoxication. While ADAM10-deficient mice develop more severe fibrosis manifested by high ALT, AST, ALP and higher collagen deposition, combined deficiency of ADAM10 and ADAM17 surprisingly results in comparable degree of liver damage as in control littermates. Therefore, ADAM17 deficiency is not protective in fibrosis development per se, but can ameliorate the damaging effect of ADAM10 deficiency on liver fibrosis development. Furthermore, we show that while ablation of ADAM17 resulted in decreased shedding of TNF RI, ADAM10 deficiency leads to increased levels of soluble TNF RI in serum. In conclusion, hepatocyte-derived ADAM10 and ADAM17 are important regulators of growth receptor signalling and TNF RI release, and pathological roles of these proteases are dependent on the cellular context.


Subject(s)
ADAM10 Protein/physiology , ADAM17 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , Liver Diseases , Liver Regeneration , Liver , Membrane Proteins/physiology , Animals , Cells, Cultured , Fibrosis/metabolism , Liver/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Primary Cell Culture
5.
J Biol Chem ; 296: 100372, 2021.
Article in English | MEDLINE | ID: mdl-33548223

ABSTRACT

Neural cell adhesion molecules 1 (NCAM1) and 2 (NCAM2) belong to the cell adhesion molecules of the immunoglobulin superfamily and have been shown to regulate formation, maturation, and maintenance of synapses. NCAM1 and NCAM2 undergo proteolysis, but the identity of all the proteases involved and how proteolysis is used to regulate their functions are not known. We report here that NCAM1 and NCAM2 are BACE1 substrates in vivo. NCAM1 and NCAM2 overexpressed in HEK cells were both cleaved by metalloproteinases or BACE1, and NCAM2 was also processed by γ-secretase. We identified the BACE1 cleavage site of NCAM1 (at Glu 671) and NCAM2 (at Glu 663) using mass spectrometry and site-directed mutagenesis. Next, we assessed BACE1-mediated processing of NCAM1 and NCAM2 in the mouse brain during aging. NCAM1 and NCAM2 were cleaved in the olfactory bulb of BACE1+/+ but not BACE1-/- mice at postnatal day 10 (P10), 4 and 12 months of age. In the hippocampus, a BACE1-specific soluble fragment of NCAM1 (sNCAM1ß) was only detected at P10. However, we observed an accumulation of full-length NCAM1 in hippocampal synaptosomes in 4-month-old BACE1-/- mice. We also found that polysialylated NCAM1 (PSA-NCAM1) levels were increased in BACE1-/- mice at P10 and demonstrated that BACE1 cleaves both NCAM1 and PSA-NCAM1 in vitro. In contrast, we did not find evidence for BACE1-dependent NCAM2 processing in the hippocampus at any age analyzed. In summary, our data demonstrate that BACE1 differentially processes NCAM1 and NCAM2 depending on the region of brain, subcellular localization, and age in vivo.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , CD56 Antigen/metabolism , Neural Cell Adhesion Molecules/metabolism , Amyloid Precursor Protein Secretases/physiology , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/physiology , Brain/metabolism , CD56 Antigen/physiology , Cell Adhesion Molecules/metabolism , Female , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecules/physiology , Neurons/metabolism , Sialic Acids/metabolism , Spatio-Temporal Analysis , Synapses/metabolism
6.
J Gerontol A Biol Sci Med Sci ; 76(1): 23-31, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32154567

ABSTRACT

The accumulation of amyloid-ß (Aß) is a characteristic event in the pathogenesis of Alzheimer's disease (AD). Aquaporin 1 (AQP1) is a membrane water channel protein belonging to the AQP family. AQP1 levels are elevated in the cerebral cortex during the early stages of AD, but the role of AQP1 in AD pathogenesis is unclear. We first determined the expression and distribution of AQP1 in brain tissue samples of AD patients and two AD mouse models (3xTg-AD and 5xFAD). AQP1 accumulation was observed in vulnerable neurons in the cerebral cortex of AD patients, and in neurons affected by the Aß or tau pathology in the 3xTg-AD and 5xFAD mice. AQP1 levels increased in neurons as aging progressed in the AD mouse models. Stress stimuli increased AQP1 in primary cortical neurons. In response to cellular stress, AQP1 appeared to translocate to endocytic compartments of ß- and γ-secretase activities. Ectopic expression of AQP1 in human neuroblastoma cells overexpressing amyloid precussir protein (APP) with the Swedish mutations reduced ß-secretase (BACE1)-mediated cleavage of APP and reduced Aß production without altering the nonamyloidogenic pathway. Conversely, knockdown of AQP1 enhanced BACE1 activity and Aß production. Immunoprecipitation experiments showed that AQP1 decreased the association of BACE1 with APP. Analysis of a human database showed that the amount of Aß decreases as the expression of AQP1 increases. These results suggest that the upregulation of AQP1 is an adaptive response of neurons to stress that reduces Aß production by inhibiting the binding between BACE1 and APP.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/physiology , Amyloid/biosynthesis , Aquaporin 1/physiology , Alzheimer Disease/metabolism , Animals , Aquaporin 1/metabolism , Disease Models, Animal , Humans , Mice , Neurons/metabolism
8.
Life Sci ; 246: 117430, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32061671

ABSTRACT

Angiopoietin-1 (Ang-1), a regulatory angiogenesis protein and it has been found to be involved in the occurrence and progression of Alzheimer's disease. However, it was still to be addressed the distinctly role and the molecular mechanisms of Ang-1 affects Alzheimer's disease. Our data suggest that Ang-1 aggravated the accumulation of Aß42 and cognitive decline in APP/PS1 mice. The upregulation of APPß is essential for Aß42 production in N2a cells overexpressing the mutational human APP gene (N2a/APP695 cells), while downregulation of PEN2 could reduce APP expression. Silencing of FOXA2 lead to inhibition of APP expression, as well as decrease of Aß42 contents. In conclusion, Ang-1 has an accelerative effect on Alzheimer's disease by increasing the secretion of Aß42 via FOXA2/PEN2/APP pathway.


Subject(s)
Alzheimer Disease/etiology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Angiopoietin-1/physiology , Hepatocyte Nuclear Factor 3-beta/metabolism , Signal Transduction , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/physiology , Animals , Blotting, Western , Brain/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Hepatocyte Nuclear Factor 3-beta/physiology , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction
9.
Mol Biol Cell ; 31(1): 27-44, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31746668

ABSTRACT

Processing of amyloid precursor protein (APP) by the ß-secretase BACE1 is the initial step of the amyloidogenic pathway to generate amyloid-ß (Aß). Although newly synthesized BACE1 and APP are transported along the secretory pathway, it is not known whether BACE1 and APP share the same post-Golgi trafficking pathways or are partitioned into different transport routes. Here we demonstrate that BACE1 exits the Golgi in HeLa cells and primary neurons by a pathway distinct from the trafficking pathway for APP. By using the Retention Using Selective Hooks system, we show that BACE1 is transported from the trans-Golgi network to the plasma membrane in an AP-1- and Arf1/4-dependent manner. Subsequently, BACE1 is endocytosed to early and recycling endosomes. Perturbation of BACE1 post-Golgi trafficking results in an increase in BACE1 cleavage of APP and increased production of both Aß40 and Aß42. These findings reveal that Golgi exit of BACE1 and APP in primary neurons is tightly regulated, resulting in their segregation along different transport routes, which limits APP processing.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Protein Transport/physiology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/physiology , Cell Membrane/metabolism , Cell Movement , Endocytosis , Endosomes/metabolism , Golgi Apparatus/metabolism , HeLa Cells , Humans , Neurons/metabolism , trans-Golgi Network/metabolism , trans-Golgi Network/physiology
10.
EMBO J ; 38(23): e102345, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31701556

ABSTRACT

In Alzheimer's disease, BACE1 protease initiates the amyloidogenic processing of amyloid precursor protein (APP) that eventually results in synthesis of ß-amyloid (Aß) peptide. Aß deposition in turn causes accumulation of BACE1 in plaque-associated dystrophic neurites, thereby potentiating progressive Aß deposition once initiated. Since systemic pharmacological BACE inhibition causes adverse effects in humans, it is important to identify strategies that specifically normalize overt BACE1 activity around plaques. The microtubule-associated protein tau regulates axonal transport of proteins, and tau deletion rescues Aß-induced transport deficits in vitro. In the current study, long-term in vivo two-photon microscopy and immunohistochemistry were performed in tau-deficient APPPS1 mice. Tau deletion reduced plaque-associated axonal pathology and BACE1 accumulation without affecting physiological BACE1 expression distant from plaques. Thereby, tau deletion effectively decelerated formation of new plaques and reduced plaque compactness. The data revealed that tau reinforces Aß deposition, presumably by contributing to accumulation of BACE1 in plaque-associated dystrophies. Targeting tau-dependent mechanisms could become a suitable strategy to specifically reduce overt BACE1 activity around plaques, thereby avoiding adverse effects of systemic BACE inhibition.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid Precursor Protein Secretases/physiology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Gene Expression Regulation , Plaque, Amyloid/prevention & control , tau Proteins/antagonists & inhibitors , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Plaque, Amyloid/etiology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
11.
Sci Rep ; 9(1): 14086, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31575895

ABSTRACT

MMP20 cleaves cadherins and may facilitate cell movement, however MMP20 is not known to cleave tight junction or desmosome proteins. Ameloblasts had not previously been screened for membrane anchored proteases that could contribute to cell movement. Here we performed a PCR screen for proteolyticlly active A Disintegrin And Metalloproteinase (ADAM) family members. These proteinases are termed sheddases because they have a transmembrane domain and their catalytic domain on the cell surface can function to release anchored proteins. Significantly, ADAMs can be targeted to specific substrates on the cell membrane through their interaction with tetraspanins. Six ADAMs (ADAM8, 9, 10, 15, 17, 19) were expressed in mouse enamel organs. We show that Adam10 expression begins in the apical loop, continues through the secretory stage and abruptly ends at the transition stage when ameloblast migration ceases. ADAM10 cleaves cadherins and tight junction plus desmosome proteins and is well characterized for its role in cell movement. ADAM10 facilitated LS8 cell migration/invasion through a Matrigel coated membrane and we demonstrate that ADAM10, but not ADAM17 cleaves the RELT extracellular domain. This striking result is significant because RELT mutations cause amelogenesis imperfecta (AI) and this directly links ADAM10 to an important role in enamel development.


Subject(s)
ADAM10 Protein/metabolism , Ameloblasts/metabolism , Amyloid Precursor Protein Secretases/metabolism , Dental Enamel/growth & development , Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , ADAM10 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , Animals , Blotting, Western , Cell Movement , Dental Enamel/metabolism , Fluorescent Antibody Technique , In Situ Hybridization , Membrane Proteins/physiology , Mice , Real-Time Polymerase Chain Reaction
12.
Nat Neurosci ; 22(7): 1075-1088, 2019 07.
Article in English | MEDLINE | ID: mdl-31209379

ABSTRACT

Microglia rapidly respond to changes in neural activity and inflammation to regulate synaptic connectivity. The extracellular signals, particularly neuron-derived molecules, that drive these microglial functions at synapses remain a key open question. Here we show that whisker lesioning, known to dampen cortical activity, induces microglia-mediated synapse elimination. This synapse elimination is dependent on signaling by CX3CR1, the receptor for microglial fractalkine (also known as CXCL1), but not complement receptor 3. Furthermore, mice deficient in CX3CL1 have profound defects in synapse elimination. Single-cell RNA sequencing revealed that Cx3cl1 is derived from cortical neurons, and ADAM10, a metalloprotease that cleaves CX3CL1 into a secreted form, is upregulated specifically in layer IV neurons and in microglia following whisker lesioning. Finally, inhibition of ADAM10 phenocopies Cx3cr1-/- and Cx3cl1-/- synapse elimination defects. Together, these results identify neuron-to-microglia signaling necessary for cortical synaptic remodeling and reveal that context-dependent immune mechanisms are utilized to remodel synapses in the mammalian brain.


Subject(s)
ADAM10 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , CX3C Chemokine Receptor 1/physiology , Chemokine CX3CL1/physiology , Membrane Proteins/physiology , Microglia/physiology , Sensorimotor Cortex/physiopathology , Touch/physiology , Vibrissae/injuries , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , CX3C Chemokine Receptor 1/deficiency , CX3C Chemokine Receptor 1/genetics , Cell Count , Female , Gene Expression Regulation , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfluidic Analytical Techniques , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sensorimotor Cortex/metabolism , Sensorimotor Cortex/pathology , Signal Transduction/physiology , Single-Cell Analysis , Transcriptome , Vibrissae/physiology
13.
J Neurosci ; 39(12): 2195-2207, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30692224

ABSTRACT

The transition of apical progenitors (APs) to basal progenitors (BPs) is an important neurogenic process during cortical expansion. Presenilin enhancer 2 (Pen-2, also named as Psenen) is a key subunit of γ-secretase and has been implicated in neurodevelopmental disease. However, it remains unknown how Pen-2 may regulate the maintenance of APs. To address this question, we generated a conditional KO (cKO) mouse in which Pen-2 is specifically inactivated in neural progenitor cells in the telencephalon. Both male and female embryos were used. We show that Pen-2 cKO cortices display remarkable depletion of Aps, but transient increase on BPs, compared with controls. We demonstrate that the proliferation rate of APs or BPs is not changed, but the switch of APs to BPs is dramatically accelerated in Pen-2 cKO cortices. Molecular analyses reveal decreased levels of Hes1 and Hes5 but increased levels of Ngn2 and NeuroD1 in Pen-2 KO cells. We report that expression of Notch1 intracellular domain in Pen-2 cKO cortices restores the population of APs and BPs. In summary, these findings highlight a central role of the Notch signaling in Pen-2-dependent maintenance of neural stem cells in the developing neocortex.SIGNIFICANCE STATEMENT Presenilin enhancer 2 (Pen-2) has been implicated in neurodevelopmental disease. However, mechanisms by which Pen-2 regulates cortical development are not understood. In this study, we generated neural progenitor cell-specific Pen-2 conditional KO mice. We observe depletion of apical progenitors and transiently increased the number of basal progenitors in the developing neocortex of Pen-2 mutant mice. Mechanistic analyses reveal decreased levels of Hes1 and Hes5, but increased levels of neurogenic transcription factors in Pen-2 mutant cortices, compared with controls. We demonstrate that reintroduction of Notch intracellular domain into mutant mice restores the population of apical progenitors to basal progenitors. The above findings strongly suggest that the Pen-2-Notch pathway plays an essential role in the maintenance of neural stem cells during cortical development.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Neocortex/embryology , Neural Stem Cells/physiology , Neurogenesis/physiology , Amyloid Precursor Protein Secretases/genetics , Animals , Cell Proliferation , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor, Notch1/physiology
14.
Angiogenesis ; 22(2): 237-250, 2019 05.
Article in English | MEDLINE | ID: mdl-30446855

ABSTRACT

The coronary vasculature is crucial for normal heart function, yet much remains to be learned about its development, especially the maturation of coronary arterial endothelium. Here, we show that endothelial inactivation of ADAM10, a key regulator of Notch signaling, leads to defects in coronary arterial differentiation, as evidenced by dysregulated genes related to Notch signaling and arterial identity. Moreover, transcriptome analysis indicated reduced EGFR signaling in A10ΔEC coronary endothelium. Further analysis revealed that A10ΔEC mice have enlarged dysfunctional hearts with abnormal myocardial compaction, and increased expression of venous and immature endothelium markers. These findings provide the first evidence for a potential role for endothelial ADAM10 in cardioprotective homeostatic EGFR signaling and implicate ADAM10/Notch signaling in coronary arterial cell specification, which is vital for normal heart development and function. The ADAM10/Notch signaling pathway thus emerges as a potential therapeutic target for improving the regenerative capacity and maturation of the coronary vasculature.


Subject(s)
ADAM10 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , Cell Differentiation/genetics , Coronary Vessels/physiology , Endothelial Cells/physiology , Endothelium, Vascular/physiology , Membrane Proteins/physiology , Animals , Coronary Vessels/cytology , Coronary Vessels/growth & development , Endothelium, Vascular/growth & development , Female , Heart/growth & development , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics
15.
eNeuro ; 5(4)2018.
Article in English | MEDLINE | ID: mdl-30079376

ABSTRACT

ß-Site amyloid precursor protein cleaving enzyme 1 (BACE1) is required for the production of ß-amyloid (Aß), one of the major pathogenic molecules of Alzheimer's disease (AD), and is therefore being actively pursued as a drug target for AD. Adult hippocampal neurogenesis (AHN) is a lifelong process that is known to be important for learning and memory and may have the potential to regenerate damaged neural tissue. In this study, we examined whether BACE1 regulates AHN, which holds important implications for its suitability as a drug target in AD. Cohorts of 2-month-old wild-type (BACE1+/+), heterozygous, and homozygous BACE1 knockout mice (BACE1+/- and BACE1-/-, respectively) were injected with 5-bromo-2'-deoxyuridine (BrdU) and sacrificed 1 day later to examine the impact of loss of BACE1 on neural precursor cell (NPC) proliferation in the adult brain. Parallel cohorts of mice were sacrificed 4 weeks after BrdU injection to determine the effects of BACE1 on survival and differentiation of newborn NPCs. We found that NPC proliferation was increased in BACE1-/- mice compared to BACE1+/+ mice, while no difference was observed in NPC survival across genotypes. Differentiation of NPCs to neuronal lineage was impaired in BACE1-/- mice. However, no differences were observed in astrogenesis, the proportion of immature neurons, or the production of oligodendrocytes across genotypes. Importantly, corresponding with a decrease in neuronal differentiation in the absence of a complementary increase in an alternate cell fate, BACE1-/- mice were found to have a pool of undifferentiated NPCs in the hippocampus compared to BACE1+/+ and BACE1+/- mice.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Aspartic Acid Endopeptidases/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Hippocampus/physiology , Neurogenesis/physiology , Neurons/physiology , Age Factors , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Front Immunol ; 9: 687, 2018.
Article in English | MEDLINE | ID: mdl-29696016

ABSTRACT

The follicular (FO) versus marginal zone (MZ) B cell fate decision in the spleen depends upon BCR, BAFF, and Notch2 signaling. Whether or how Gi signaling affects this fate decision is unknown. Here, we show that direct contact with Notch ligand expressing stromal cells (OP9-Delta-like 1) cannot promote normal MZ B cell development when progenitor B cells lack Gαi proteins, or if Gi signaling is disabled. Consistent with faulty ADAM10-dependent Notch2 processing, Gαi-deficient transitional B cells had low ADAM10 membrane expression levels and reduced Notch2 target gene expression. Immunoblotting Gαi-deficient B cell lysates revealed a reduction in mature, processed ADAM10. Suggesting that Gαi signaling promotes ADAM10 membrane expression, stimulating normal transitional B cells with CXCL12 raised it, while inhibiting Gαi nucleotide exchange blocked its upregulation. Surprisingly, inhibiting Gαi nucleotide exchange in transitional B cells also impaired the upregulation of ADAM10 that occurs following antigen receptor crosslinking. These results indicate that Gαi signaling supports ADAM10 maturation and activity in transitional B cells, and ultimately Notch2 signaling to promote MZ B cell development.


Subject(s)
ADAM10 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , B-Lymphocytes/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Membrane Proteins/physiology , Animals , Cells, Cultured , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Pertussis Toxin/pharmacology , Receptor, Notch2/physiology , Signal Transduction , Spleen/cytology
17.
Mol Psychiatry ; 23(7): 1542-1554, 2018 07.
Article in English | MEDLINE | ID: mdl-29703946

ABSTRACT

Potassium channel Kv2.1 regulates potassium current in cortical neurons and potassium efflux is necessary for cell apoptosis. As a major component of delayed rectifier current potassium channels, Kv2.1 forms clusters in the membrane of hippocampal neurons. BACE2 is an aspartyl protease to cleave APP to prevent the generation of Aß, a central component of neuritic plaques in Alzheimer's brain. We now identified Kv2.1 as a novel substrate of BACE2. We found that BACE2 cleaved Kv2.1 at Thr376, Ala717, and Ser769 sites and disrupted Kv2.1 clustering on cell membrane, resulting in decreased Ik of Kv2.1 and a hyperpolarizing shift in primary neurons. Furthermore, we discovered that the BACE2-cleaved Kv2.1 forms, Kv2.1-1-375, Kv2.1-1-716, and Kv2.1-1-768, depressed the delayed rectifier Ik surge and reduced neuronal apoptosis. Our study suggests that BACE2 plays a neuroprotective role by cleavage of Kv2.1 to prevent the outward potassium currents, a potential new target for Alzheimer's treatment.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/physiology , Aspartic Acid Endopeptidases/metabolism , Aspartic Acid Endopeptidases/physiology , Shab Potassium Channels/physiology , Amino Acid Sequence , Animals , Apoptosis/physiology , Cell Membrane/metabolism , HEK293 Cells , Hippocampus/metabolism , Humans , Neurons/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Primary Cell Culture , Rats , Shab Potassium Channels/metabolism , Substrate Specificity
18.
Neurobiol Aging ; 62: 130-145, 2018 02.
Article in English | MEDLINE | ID: mdl-29149631

ABSTRACT

Regulation of the amyloid precursor protein (APP) processing by α- and ß-secretases is of special interest to Alzheimer's disease (AD), as these proteases prevent or mediate amyloid beta formation, respectively. Neuroinflammation is also implicated in AD. Our data demonstrate that the endogenous mediator of inflammation prostaglandin J2 (PGJ2) promotes full-length APP (FL-APP) processing by α- and ß-secretases. The decrease in FL-APP was independent of proteasomal, lysosomal, calpain, caspase, and γ-secretase activities. Moreover, PGJ2-treatment promoted cleavage of secreted APP, specifically sAPPα and sAPPß, generated by α and ß-secretase, respectively. Notably, PGJ2-treatment induced caspase-dependent cleavage of sAPPß. Mechanistically, PGJ2-treatment selectively diminished mature (O- and N-glycosylated) but not immature (N-glycosylated only) FL-APP. PGJ2-treatment also increased the overall levels of protein O-GlcNAcylation, which occurs within the nucleocytoplasmic compartment. It is known that APP undergoes O-GlcNAcylation and that the latter protects proteins from proteasomal degradation. Our results suggest that by increasing protein O-GlcNAcylation levels, PGJ2 renders mature APP less prone to proteasomal degradation, thus shunting APP toward processing by α- and ß-secretases.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/metabolism , Prostaglandin D2/analogs & derivatives , Animals , Caspases/physiology , Cells, Cultured , Cytoplasm/metabolism , Female , Glycosylation , Humans , Inflammation/etiology , Inflammation/metabolism , Male , Prostaglandin D2/physiology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Rats, Sprague-Dawley , Tumor Cells, Cultured
19.
Nat Med ; 23(12): 1405-1415, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29058717

ABSTRACT

Maladaptive wound healing responses to chronic tissue injury result in organ fibrosis. Fibrosis, which entails excessive extracellular matrix (ECM) deposition and tissue remodeling by activated myofibroblasts, leads to loss of proper tissue architecture and organ function; however, the molecular mediators of myofibroblast activation have yet to be fully identified. Here we identify soluble ephrin-B2 (sEphrin-B2) as a new profibrotic mediator in lung and skin fibrosis. We provide molecular, functional and translational evidence that the ectodomain of membrane-bound ephrin-B2 is shed from fibroblasts into the alveolar airspace after lung injury. Shedding of sEphrin-B2 promotes fibroblast chemotaxis and activation via EphB3 and/or EphB4 receptor signaling. We found that mice lacking ephrin-B2 in fibroblasts are protected from skin and lung fibrosis and that a disintegrin and metalloproteinase 10 (ADAM10) is the major ephrin-B2 sheddase in fibroblasts. ADAM10 expression is increased by transforming growth factor (TGF)-ß1, and ADAM10-mediated sEphrin-B2 generation is required for TGF-ß1-induced myofibroblast activation. Pharmacological inhibition of ADAM10 reduces sEphrin-B2 levels in bronchoalveolar lavage and prevents lung fibrosis in mice. Consistent with the mouse data, ADAM10-sEphrin-B2 signaling is upregulated in fibroblasts from human subjects with idiopathic pulmonary fibrosis. These results uncover a new molecular mechanism of tissue fibrogenesis and identify sEphrin-B2, its receptors EphB3 and EphB4 and ADAM10 as potential therapeutic targets in the treatment of fibrotic diseases.


Subject(s)
ADAM10 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , Ephrin-B2/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Lung/pathology , Membrane Proteins/physiology , Myofibroblasts/physiology , Skin Diseases/genetics , Skin/pathology , Animals , Cells, Cultured , Exocytosis/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/pathology , Protein Transport/genetics , Skin/metabolism , Skin Diseases/metabolism , Skin Diseases/pathology
20.
Neurobiol Aging ; 60: 129-140, 2017 12.
Article in English | MEDLINE | ID: mdl-28946017

ABSTRACT

The cleavage of amyloid precursor protein (APP) by ß-site APP cleaving enzyme 1 (BACE1) is the rate-limiting step in beta amyloid generation during Alzheimer's disease (AD) pathogenesis. In AD brains, BACE1 is abnormally accumulated in endocytic compartments, where the acidic pH is optimal for its activity. However, mechanisms regulating the endosome-to-trans-Golgi network (TGN) retrieval of BACE1 remain unclear. Here, we show that partitioning defective 3 (Par3) facilitates BACE1 retrograde trafficking from endosomes to the TGN. Par3 functions through aPKC-mediated phosphorylation of BACE1 on Ser498, which in turn promotes the interaction between BACE1 and phosphofurin acidic cluster sorting protein 1 and facilitates the retrograde trafficking of BACE1 to the TGN. In human AD brains, there is a significant decrease in Ser498 phosphorylation of BACE1 suggesting that defective phosphorylation-dependent retrograde transport of BACE1 is important in AD pathogenesis. Together, our studies provide mechanistic insight into a novel role for Par3 and aPKC in regulating the retrograde endosome-to-TGN trafficking of BACE1 and shed light on the mechanisms of AD pathogenesis.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Carrier Proteins/physiology , Endosomes/metabolism , Golgi Apparatus/metabolism , Protein Kinase C/physiology , Vesicular Transport Proteins/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/physiology , Brain/metabolism , Cells, Cultured , Humans , Nerve Tissue Proteins , Phosphorylation , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...