Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nature ; 586(7831): 735-740, 2020 10.
Article in English | MEDLINE | ID: mdl-32879487

ABSTRACT

Innate immunity is associated with Alzheimer's disease1, but the influence of immune activation on the production of amyloid-ß is unknown2,3. Here we identify interferon-induced transmembrane protein 3 (IFITM3) as a γ-secretase modulatory protein, and establish a mechanism by which inflammation affects the generation of amyloid-ß. Inflammatory cytokines induce the expression of IFITM3 in neurons and astrocytes, which binds to γ-secretase and upregulates its activity, thereby increasing the production of amyloid-ß. The expression of IFITM3 is increased with ageing and in mouse models that express familial Alzheimer's disease genes. Furthermore, knockout of IFITM3 reduces γ-secretase activity and the formation of amyloid plaques in a transgenic mouse model (5xFAD) of early amyloid deposition. IFITM3 protein is upregulated in tissue samples from a subset of patients with late-onset Alzheimer's disease that exhibit higher γ-secretase activity. The amount of IFITM3 in the γ-secretase complex has a strong and positive correlation with γ-secretase activity in samples from patients with late-onset Alzheimer's disease. These findings reveal a mechanism in which γ-secretase is modulated by neuroinflammation via IFITM3 and the risk of Alzheimer's disease is thereby increased.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Immunity, Innate , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , Age of Onset , Aged, 80 and over , Aging/genetics , Aging/immunology , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Catalytic Domain , Disease Models, Animal , Female , HEK293 Cells , Humans , Inflammation , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/metabolism , RNA-Binding Proteins/genetics , Risk , Up-Regulation
2.
Mol Psychiatry ; 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383769

ABSTRACT

CD33 is a transmembrane receptor expressed on cells of myeloid lineage and regulates innate immunity. CD33 is a risk factor for Alzheimer's disease (AD) and targeting CD33 has been a promising strategy drug development. However, the mechanism of CD33's action is poorly understood. Here we investigate the mechanism of anti-CD33 antibody HuM195 (Lintuzumab) and its single-chain variable fragment (scFv) and examine their therapeutic potential. Treatment with HuM195 full-length antibody or its scFv increased phagocytosis of ß-amyloid 42 (Aß42) in human microglia and monocytes. This activation of phagocytosis was driven by internalization and degradation of CD33, thereby downregulating its inhibitory signal. HumM195 transiently induced CD33 phosphorylation and its signaling via receptor dimerization. However, this signaling decayed with degradation of CD33. scFv binding to CD33 leads to a degradation of CD33 without detection of the CD33 dimerization and signaling. Moreover, we found that treatments with either HuM195 or scFv promotes the secretion of IL33, a cytokine implicated in microglia reprogramming. Importantly, recombinant IL33 potentiates the uptake of Aß42 in monocytes. Collectively, our findings provide unanticipated mechanistic insight into the role of CD33 signaling in both monocytes and microglia and define a molecular basis for the development of CD33-based therapy of AD.

3.
Mol Psychiatry ; 27(10): 4264-4273, 2022 10.
Article in English | MEDLINE | ID: mdl-35764706

ABSTRACT

Hypoxic-ischemic injury has been linked with increased risk for developing Alzheimer's disease (AD). The underlying mechanism of this association is poorly understood. Here, we report distinct roles for hypoxia-inducible factor-1α (Hif-1α) in the regulation of BACE1 and γ-secretase activity, two proteases involved in the production of amyloid-beta (Aß). We have demonstrated that Hif-1α upregulates both BACE1 and γ-secretase activity for Aß production in brain hypoxia-induced either by cerebral hypoperfusion or breathing 10% O2. Hif-1α binds to γ-secretase, which elevates the amount of active γ-secretase complex without affecting the level of individual subunits in hypoxic-ischemic mouse brains. Additionally, the expression of full length Hif-1α increases BACE1 and γ-secretase activity in primary neuronal culture, whereas a transcriptionally incompetent Hif-1α variant only activates γ-secretase. These findings indicate that Hif-1α transcriptionally upregulates BACE1 and nontranscriptionally activates γ-secretase for Aß production in hypoxic-ischemic conditions. Consequently, Hif-1α-mediated Aß production may be an adaptive response to hypoxic-ischemic injury, subsequently leading to increased risk for AD. Preventing the interaction of Hif-1α with γ-secretase may therefore be a promising therapeutic strategy for AD treatment.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Hypoxia-Inducible Factor 1, alpha Subunit , Animals , Mice , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
4.
Mol Psychiatry ; 26(10): 5620-5635, 2021 10.
Article in English | MEDLINE | ID: mdl-32792660

ABSTRACT

Amyloid-ß peptide (Aß) accumulation in the brain is a hallmark of Alzheimer's Disease. An important mechanism of Aß clearance in the brain is uptake and degradation by microglia. Presenilin 1 (PS1) is the catalytic subunit of γ-secretase, an enzyme complex responsible for the maturation of multiple substrates, such as Aß. Although PS1 has been extensively studied in neurons, the role of PS1 in microglia is incompletely understood. Here we report that microglia containing phospho-deficient mutant PS1 display a slower kinetic response to micro injury in the brain in vivo and the inability to degrade Aß oligomers due to a phagolysosome dysfunction. An Alzheimer's mouse model containing phospho-deficient PS1 show severe Aß accumulation in microglia as well as the postsynaptic protein PSD95. Our results demonstrate a novel mechanism by which PS1 modulates microglial function and contributes to Alzheimer's -associated phenotypes.


Subject(s)
Alzheimer Disease , Microglia , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Mice , Microglia/metabolism , Phosphorylation , Presenilin-1/genetics , Presenilin-1/metabolism
5.
Proc Natl Acad Sci U S A ; 116(13): 6385-6390, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30850537

ABSTRACT

The mechanism by which γ-secretase activating protein (GSAP) regulates γ-secretase activity has not yet been elucidated. Here, we show that knockout of GSAP in cultured cells directly reduces γ-secretase activity for Aß production, but not for Notch1 cleavage, suggesting that GSAP may induce a conformational change contributing to the specificity of γ-secretase. Furthermore, using an active-site-directed photoprobe with double cross-linking moieties, we demonstrate that GSAP modifies the orientation and/or distance of the PS1 N-terminal fragment and the PS1 C-terminal fragment, a region containing the active site of γ-secretase. This work offers insight into how GSAP regulates γ-secretase specificity.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Presenilin-1/chemistry , Proteins/metabolism , CRISPR-Cas Systems , Catalytic Domain , Gene Knockdown Techniques , HEK293 Cells , Humans , Kinetics , Peptide Fragments/metabolism , Proteins/genetics , Receptor, Notch1
6.
J Biol Chem ; 290(19): 12135-46, 2015 May 08.
Article in English | MEDLINE | ID: mdl-25795784

ABSTRACT

Proenzyme maturation is a general mechanism to control the activation of enzymes. Catalytically active members of the A Disintegrin And Metalloprotease (ADAM) family of membrane-anchored metalloproteases are synthesized as proenzymes, in which the latency is maintained by their autoinhibitory pro-domains. A proteolytic processing then transforms the proenzyme into a catalytically active form. The removal of the pro-domain of ADAMs is currently thought to depend on processing at a canonical consensus site for the proprotein convertase Furin (RXXR) between the pro- and the catalytic domain. Here, we demonstrate that this previously described canonical site is a secondary cleavage site to a prerequisite cleavage in a newly characterized upstream PC site embedded within the pro-domain sequence. The novel upstream regulatory site is important for the maturation of several ADAM proenzymes. Mutations in the upstream regulatory site of ADAM17, ADAM10, and ADAM9 do not prevent pro-domain processing between the pro- and metalloprotease domain, but nevertheless, cause significantly reduced catalytic activity. Thus, our results have uncovered a novel functionally relevant PC processing site in the N-terminal part of the pro-domain that is important for the activation of these ADAMs. These results suggest that the novel PC site is part of a general mechanism underlying proenzyme maturation of ADAMs that is independent of processing at the previously identified canonical Furin cleavage site.


Subject(s)
ADAM Proteins/genetics , Amyloid Precursor Protein Secretases/genetics , Membrane Proteins/genetics , Mutation , Protein Processing, Post-Translational , ADAM10 Protein , ADAM17 Protein , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Circular Dichroism , Cloning, Molecular , Escherichia coli/metabolism , Fibroblasts/metabolism , Furin/chemistry , HEK293 Cells , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Protein Structure, Tertiary , Recombinant Proteins/genetics , Sequence Homology, Amino Acid
7.
Proc Natl Acad Sci U S A ; 114(27): 6885-6887, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28645897
8.
ACS Nano ; 16(5): 7269-7283, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35420796

ABSTRACT

Amyloid-beta (Aß) deposition occurs in the early stages of Alzheimer's disease (AD), but the early detection of Aß is a persistent challenge. Herein, we engineered a near-infrared optical nanosensor capable of detecting Aß intracellularly in live cells and intracranially in vivo. The sensor is composed of single-walled carbon nanotubes functionalized with Aß wherein Aß-Aß interactions drive the response. We found that the Aß nanosensors selectively responded to Aß via solvatochromic modulation of the near-infrared emission of the nanotube. The sensor tracked Aß accumulation in live cells and, upon intracranial administration in a genetic model of AD, signaled distinct responses in aged mice. This technology enables the interrogation of molecular mechanisms underlying Aß neurotoxicity in the development of AD in living systems.


Subject(s)
Alzheimer Disease , Nanotubes, Carbon , Animals , Mice , Amyloid beta-Peptides , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics
9.
Cancer Discov ; 12(5): 1314-1335, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35262173

ABSTRACT

Brain metastasis is a significant cause of morbidity and mortality in multiple cancer types and represents an unmet clinical need. The mechanisms that mediate metastatic cancer growth in the brain parenchyma are largely unknown. Melanoma, which has the highest rate of brain metastasis among common cancer types, is an ideal model to study how cancer cells adapt to the brain parenchyma. Our unbiased proteomics analysis of melanoma short-term cultures revealed that proteins implicated in neurodegenerative pathologies are differentially expressed in melanoma cells explanted from brain metastases compared with those derived from extracranial metastases. We showed that melanoma cells require amyloid beta (Aß) for growth and survival in the brain parenchyma. Melanoma-secreted Aß activates surrounding astrocytes to a prometastatic, anti-inflammatory phenotype and prevents phagocytosis of melanoma by microglia. Finally, we demonstrate that pharmacologic inhibition of Aß decreases brain metastatic burden. SIGNIFICANCE: Our results reveal a novel mechanistic connection between brain metastasis and Alzheimer's disease, two previously unrelated pathologies; establish Aß as a promising therapeutic target for brain metastasis; and demonstrate suppression of neuroinflammation as a critical feature of metastatic adaptation to the brain parenchyma. This article is highlighted in the In This Issue feature, p. 1171.


Subject(s)
Brain Neoplasms , Melanoma , Amyloid beta-Peptides/therapeutic use , Astrocytes/metabolism , Brain Neoplasms/genetics , Humans , Melanoma/drug therapy , Neoplasm Metastasis , Neuroinflammatory Diseases
10.
J Exp Med ; 218(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34156424

ABSTRACT

Biochemical, pathogenic, and human genetic data confirm that GSAP (γ-secretase activating protein), a selective γ-secretase modulatory protein, plays important roles in Alzheimer's disease (AD) and Down's syndrome. However, the molecular mechanism(s) underlying GSAP-dependent pathogenesis remains largely elusive. Here, through unbiased proteomics and single-nuclei RNAseq, we identified that GSAP regulates multiple biological pathways, including protein phosphorylation, trafficking, lipid metabolism, and mitochondrial function. We demonstrated that GSAP physically interacts with the Fe65-APP complex to regulate APP trafficking/partitioning. GSAP is enriched in the mitochondria-associated membrane (MAM) and regulates lipid homeostasis through the amyloidogenic processing of APP. GSAP deletion generates a lipid environment unfavorable for AD pathogenesis, leading to improved mitochondrial function and the rescue of cognitive deficits in an AD mouse model. Finally, we identified a novel GSAP single-nucleotide polymorphism that regulates its brain transcript level and is associated with an increased AD risk. Together, our findings indicate that GSAP impairs mitochondrial function through its MAM localization and that lowering GSAP expression reduces pathological effects associated with AD.


Subject(s)
Alzheimer Disease/pathology , Homeostasis , Lipid Metabolism , Mitochondria/metabolism , Proteins/metabolism , Aging/pathology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Base Sequence , Disease Models, Animal , Hippocampus/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Models, Biological , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Open Field Test , Phosphorylation , Protein Binding , Protein Transport , Proteins/genetics , Transcription, Genetic
11.
Front Aging Neurosci ; 12: 614690, 2020.
Article in English | MEDLINE | ID: mdl-33343338

ABSTRACT

Described as the "proteasome of the membrane" or the "scissors in the membrane," γ-secretase has notoriously complicated biology, and even after decades of research, the full extent of its regulatory mechanism remains unclear. γ-Secretase is an intramembrane aspartyl protease complex composed of four obligatory subunits: Nicastrin (NCT), Presenilin (PS), Presenilin Enhancer-2 (Pen-2), and Anterior pharynx-defective-1 (Aph-1). γ-Secretase cleaves numerous type 1 transmembrane substrates, with no apparent homology, and plays major roles in broad biological pathways such as development, neurogenesis, and cancer. Notch and the amyloid precursor protein (APP) and are undoubtedly the best-studied γ-secretase substrates because of their role in cancer and Alzheimer's disease (AD) and therefore became the focus of increasing studies as an attractive therapeutic target. The regulation of γ-secretase is intricate and involves the function of multiple cellular entities. Recently, γ-secretase modulatory proteins (GSMPs), which are non-essential subunits and yet modulate γ-secretase activity and specificity, have emerged as an important component in guiding γ-secretase. GSMPs are responsive to cellular and environmental changes and therefore, provide another layer of regulation of γ-secretase. This type of enzymatic regulation allows for a rapid and fine-tuning of γ-secretase activity when appropriate signals appear enabling a temporal level of regulation. In this review article, we discuss the latest developments on GSMPs and implications on the development of effective therapeutics for γ-secretase-associated diseases such as AD and cancer.

12.
Sci Adv ; 6(1): eaay3566, 2020 01.
Article in English | MEDLINE | ID: mdl-31911948

ABSTRACT

In this study, we investigated the roles of Epac1 in pathological angiogenesis and its potential as a novel therapeutic target for the treatment of vasoproliferative diseases. Genetic deletion of Epac1 ameliorated pathological angiogenesis in mouse models of oxygen-induced retinopathy (OIR) and carotid artery ligation. Moreover, genetic deletion or pharmacological inhibition of Epac1 suppressed microvessel sprouting from ex vivo aortic ring explants. Mechanistic studies revealed that Epac1 acted as a previously unidentified inhibitor of the γ-secretase/Notch signaling pathway via interacting with γ-secretase and regulating its intracellular trafficking while enhancing vascular endothelial growth factor signaling to promote pathological angiogenesis. Pharmacological administration of an Epac-specific inhibitor suppressed OIR-induced neovascularization in wild-type mice, recapitulating the phenotype of genetic Epac1 knockout. Our results demonstrate that Epac1 signaling is critical for the progression of pathological angiogenesis but not for physiological angiogenesis and that the newly developed Epac-specific inhibitors are effective in combating proliferative retinopathy.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Neovascularization, Pathologic/genetics , Retinal Neovascularization/genetics , Animals , Cell Movement/genetics , Disease Models, Animal , Humans , Mice , Mice, Knockout , Neovascularization, Pathologic/pathology , Receptors, Notch/genetics , Retinal Neovascularization/pathology , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics
13.
Sci Rep ; 6: 35598, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27982031

ABSTRACT

Dysregulated activity of A Disintegrin And Metalloproteinase 17 (ADAM17)/TNFα Converting Enzyme (TACE) is associated with inflammatory disorders and cancer progression by releasing regulatory membrane-tethered proteins like TNFα, IL6R and EGFR ligands. Although specific inhibition of TACE is thought to be a viable strategy for inflammatory disorders and for malignancies treatment, the generation of effective inhibitors in vivo has been proven to be challenging. Here we report on the development of a protein inhibitor that leverages the endogenous modulator of TACE. We have generated a stable form of the auto-inhibitory TACE prodomain (TPD), which specifically inhibits in vitro and cell-surface TACE, but not the related ADAM10, and effectively modulated TNFα secretion in cells. TPD significantly attenuated TACE-mediated disease models of sepsis, rheumatoid arthritis (RA) and inflammatory bowel disease (IBD), and reduced TNFα in synovial fluids from RA patients. Our results demonstrate that intervening with endogenous ADAM sheddase modulatory mechanisms holds potential as a general strategy for the design of ADAM inhibitors.


Subject(s)
ADAM17 Protein/chemistry , Arthritis/drug therapy , Colitis/drug therapy , Enzyme Inhibitors/administration & dosage , Shock, Septic/drug therapy , ADAM10 Protein/metabolism , ADAM17 Protein/antagonists & inhibitors , Animals , Arthritis/chemically induced , Arthritis/metabolism , Cells, Cultured , Colitis/chemically induced , Colitis/metabolism , Collagen/adverse effects , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Lipopolysaccharides/adverse effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Protein Domains , Shock, Septic/chemically induced , Shock, Septic/metabolism , Trinitrobenzenesulfonic Acid/adverse effects
14.
JCI Insight ; 1(13)2016 Aug 18.
Article in English | MEDLINE | ID: mdl-27642633

ABSTRACT

Kidney fibrosis following kidney injury is an unresolved health problem and causes significant morbidity and mortality worldwide. In a study into its molecular mechanism, we identified essential causative features. Acute or chronic kidney injury causes sustained elevation of a disintegrin and metalloprotease 17 (ADAM17); of its cleavage-activated proligand substrates, in particular of pro-TNFα and the EGFR ligand amphiregulin (pro-AREG); and of the substrates' receptors. As a consequence, EGFR is persistently activated and triggers the synthesis and release of proinflammatory and profibrotic factors, resulting in macrophage/neutrophil ingress and fibrosis. ADAM17 hypomorphic mice, specific ADAM17 inhibitor-treated WT mice, or mice with inducible KO of ADAM17 in proximal tubule (Slc34a1-Cre) were significantly protected against these effects. In vitro, in proximal tubule cells, we show that AREG has unique profibrotic actions that are potentiated by TNFα-induced AREG cleavage. In vivo, in acute kidney injury (AKI) and chronic kidney disease (CKD, fibrosis) patients, soluble AREG is indeed highly upregulated in human urine, and both ADAM17 and AREG expression show strong positive correlation with fibrosis markers in related kidney biopsies. Our results indicate that targeting of the ADAM17 pathway represents a therapeutic target for human kidney fibrosis.


Subject(s)
ADAM17 Protein/metabolism , Acute Kidney Injury/pathology , Amphiregulin/pharmacology , Kidney Tubules, Proximal/metabolism , Kidney/pathology , Acute Kidney Injury/metabolism , Animals , Cells, Cultured , ErbB Receptors/metabolism , Fibrosis , Kidney/metabolism , Male , Mice , Mice, Knockout , Sodium-Phosphate Cotransporter Proteins, Type IIb
15.
Oncotarget ; 6(42): 44151-60, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26683521

ABSTRACT

Genetic deficiencies provide insights into gene function in humans. Here we describe a patient with a very rare genetic deficiency of ADAM17. We show that the patient's PBMCs had impaired cytokine secretion in response to LPS stimulation, correlating with the clinical picture of severe bacteremia from which the patient suffered. ADAM17 was shown to cleave CD16, a major NK killer receptor. Functional analysis of patient's NK cells demonstrated that his NK cells express normal levels of activating receptors and maintain high surface levels of CD16 following mAb stimulation. Activation of individual NK cell receptors showed that the patient's NK cells are more potent when activated directly by CD16, albeit no difference was observed in Antibody Depedent Cytotoxicity (ADCC) assays. Our data suggest that ADAM17 inhibitors currently considered for clinical use to boost CD16 activity should be cautiously applied, as they might have severe side effects resulting from impaired cytokine secretion.


Subject(s)
ADAM Proteins/deficiency , Cytokines/metabolism , Immunologic Deficiency Syndromes/enzymology , Killer Cells, Natural/enzymology , Leukocytes, Mononuclear/enzymology , Lymphocyte Activation , ADAM Proteins/genetics , ADAM Proteins/immunology , ADAM17 Protein , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Child, Preschool , Cytokines/immunology , Fatal Outcome , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Genetic Predisposition to Disease , Humans , Immunity, Innate , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/pharmacology , Male , Phenotype , Receptors, IgG/immunology , Receptors, IgG/metabolism
16.
Nat Med ; 21(11): 1307-17, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26457757

ABSTRACT

Retention of long-term repopulating hematopoietic stem cells (LT-HSCs) in the bone marrow is essential for hematopoiesis and for protection from myelotoxic injury. We report that signaling cascades that are traditionally viewed as coagulation related also control retention of endothelial protein C receptor-positive (EPCR(+)) LT-HSCs in the bone marrow and their recruitment to the blood via two pathways mediated by protease activated receptor 1 (PAR1). Thrombin-PAR1 signaling induces nitric oxide (NO) production, leading to EPCR shedding mediated by tumor necrosis factor-α-converting enzyme (TACE), enhanced CXCL12-CXCR4-induced motility and rapid stem and progenitor cell mobilization. Conversely, bone marrow blood vessels provide a microenvironment enriched with activated protein C (aPC) that retains EPCR(+) LT-HSCs by limiting NO generation, reducing Cdc42 activity and enhancing integrin VLA4 affinity and adhesion. Inhibition of NO production by aPC-EPCR-PAR1 signaling reduces progenitor cell egress from the bone marrow, increases retention of bone marrow NO(low) EPCR(+) LT-HSCs and protects mice from chemotherapy-induced hematological failure and death. Our study reveals new roles for PAR1 and EPCR in controlling NO production to balance maintenance and recruitment of bone marrow EPCR(+) LT-HSCs, with potential clinical relevance for stem cell transplantation.


Subject(s)
Hematopoietic Stem Cells/metabolism , Nitric Oxide/metabolism , Protein C/metabolism , Receptor, PAR-1/metabolism , Receptors, Cell Surface/metabolism , Thrombin/metabolism , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Bone Marrow/metabolism , Cell Adhesion , Cell Movement , Chemokine CXCL12/metabolism , Endothelial Protein C Receptor , Hematopoietic Stem Cells/cytology , Integrin alpha4beta1/metabolism , Mice , Mice, Inbred C57BL , Receptors, CXCR4/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/metabolism
17.
Nat Commun ; 5: 4058, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24898499

ABSTRACT

During embryonic development, axons can gain and lose sensitivity to guidance cues, and this flexibility is essential for the correct wiring of the nervous system. Yet, the underlying molecular mechanisms are largely unknown. Here we show that receptor cleavage by ADAM (A Disintegrin And Metalloprotease) metalloproteases promotes murine sensory axons loss of responsiveness to the chemorepellant Sema3A. Genetic ablation of ADAM10 and ADAM17 disrupts the developmental downregulation of Neuropilin-1 (Nrp1), the receptor for Sema3A, in sensory axons. Moreover, this is correlated with gain of repulsive response to Sema3A. Overexpression of Nrp1 in neurons reverses axonal desensitization to Sema3A, but this is hampered in a mutant Nrp1 with high susceptibility to cleavage. Lastly, we detect guidance errors of proprioceptive axons in ADAM knockouts that are consistent with enhanced response to Sema3A. Our results provide the first evidence for involvement of ADAMs in regulating developmental switch in responsiveness to axonal guidance cues.


Subject(s)
ADAM Proteins/genetics , Axons/metabolism , Gene Expression Regulation, Developmental , Neuropilin-1/genetics , Semaphorin-3A/metabolism , Sensory Receptor Cells/metabolism , ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases/genetics , Animals , Membrane Proteins/genetics , Mice , Neuropilin-1/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL