Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Mol Cell Cardiol ; 89(Pt B): 335-48, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26454161

ABSTRACT

KCNQ1 (Kv7.1) proteins form a homotetrameric channel, which produces a voltage-dependent K(+) current. Co-assembly of KCNQ1 with the auxiliary ß-subunit KCNE1 strongly up-regulates this current. In cardiac myocytes, KCNQ1/E1 complexes are thought to give rise to the delayed rectifier current IKs, which contributes to cardiac action potential repolarization. We report here that the type I membrane protein BACE1 (ß-site APP-cleaving enzyme 1), which is best known for its detrimental role in Alzheimer's disease, but is also, as reported here, present in cardiac myocytes, serves as a novel interaction partner of KCNQ1. Using HEK293T cells as heterologous expression system to study the electrophysiological effects of BACE1 and KCNE1 on KCNQ1 in different combinations, our main findings were the following: (1) BACE1 slowed the inactivation of KCNQ1 current producing an increased initial response to depolarizing voltage steps. (2) Activation kinetics of KCNQ1/E1 currents were significantly slowed in the presence of co-expressed BACE1. (3) BACE1 impaired reconstituted cardiac IKs when cardiac action potentials were used as voltage commands, but interestingly augmented the IKs of ATP-deprived cells, suggesting that the effect of BACE1 depends on the metabolic state of the cell. (4) The electrophysiological effects of BACE1 on KCNQ1 reported here were independent of its enzymatic activity, as they were preserved when the proteolytically inactive variant BACE1 D289N was co-transfected in lieu of BACE1 or when BACE1-expressing cells were treated with the BACE1-inhibiting compound C3. (5) Co-immunoprecipitation and fluorescence recovery after photobleaching (FRAP) supported our hypothesis that BACE1 modifies the biophysical properties of IKs by physically interacting with KCNQ1 in a ß-subunit-like fashion. Strongly underscoring the functional significance of this interaction, we detected BACE1 in human iPSC-derived cardiomyocytes and murine cardiac tissue and observed decreased IKs in atrial cardiomyocytes of BACE1-deficient mice.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Aspartic Acid Endopeptidases/deficiency , Ion Channel Gating , KCNQ1 Potassium Channel/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , Action Potentials , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Female , HEK293 Cells , Humans , Immunoprecipitation , Kinetics , Male , Mice , Multiprotein Complexes/metabolism , Phenotype , Protein Binding , Proteolysis
2.
J Neurosci ; 35(8): 3298-311, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25716831

ABSTRACT

The ß-secretase BACE1 is widely known for its pivotal role in the amyloidogenic pathway leading to Alzheimer's disease, but how its action on transmembrane proteins other than the amyloid precursor protein affects the nervous system is only beginning to be understood. We report here that BACE1 regulates neuronal excitability through an unorthodox, nonenzymatic interaction with members of the KCNQ (Kv7) family that give rise to the M-current, a noninactivating potassium current with slow kinetics. In hippocampal neurons from BACE1(-/-) mice, loss of M-current enhanced neuronal excitability. We relate the diminished M-current to the previously reported epileptic phenotype of BACE1-deficient mice. In HEK293T cells, BACE1 amplified reconstituted M-currents, altered their voltage dependence, accelerated activation, and slowed deactivation. Biochemical evidence strongly suggested that BACE1 physically associates with channel proteins in a ß-subunit-like fashion. Our results establish BACE1 as a physiologically essential constituent of regular M-current function and elucidate a striking new feature of how BACE1 impacts on neuronal activity in the intact and diseased brain.


Subject(s)
Action Potentials , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Hippocampus/metabolism , KCNQ Potassium Channels/metabolism , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Cells, Cultured , Female , HEK293 Cells , Hippocampus/cytology , Hippocampus/physiology , Humans , KCNQ Potassium Channels/genetics , Male , Mice , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/physiology
3.
Biochem Biophys Res Commun ; 457(3): 334-40, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25576872

ABSTRACT

The lysosomal integral membrane protein type-2 (LIMP-2/SCARB2) has been identified as a receptor for enterovirus 71 uptake and mannose-6-phosphate-independent lysosomal trafficking of the acid hydrolase ß-glucocerebrosidase. Here we show that LIMP-2 undergoes proteolytic cleavage mediated by lysosomal cysteine proteases. Heterologous expression and in vitro studies suggest that cathepsin-F is mainly responsible for the lysosomal processing of wild-type LIMP-2. Furthermore, examination of purified lysosomes revealed that LIMP-2 undergoes proteolysis in vivo. Mutations in the gene encoding cathepsin-F (CTSF) have recently been associated with type-B-Kufs-disease, an adult form of neuronal ceroid-lipofuscinosis. In this study we show that disease-causing cathepsin-F mutants fail to cleave LIMP-2. Our findings provide evidence that LIMP-2 represents an in vivo substrate of cathepsin-F with relevance for understanding the pathophysiology of type-B-Kufs-disease.


Subject(s)
Cathepsin F/genetics , Cathepsin F/metabolism , Lysosomal Membrane Proteins/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Receptors, Scavenger/metabolism , Animals , CD36 Antigens/chemistry , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Line , HEK293 Cells , Humans , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Lysosomes/metabolism , Mice , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Proteolysis , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
4.
Cell Mol Life Sci ; 71(2): 331-48, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23760205

ABSTRACT

Programmed necrosis is important in many (patho)physiological settings. For specific therapeutic intervention, however, a better knowledge is required whether necrosis occurs through one single "core program" or through several independent pathways. Previously, the poly(ADP-ribose) polymerase (PARP) pathway has been suggested as a crucial element of tumor necrosis factor (TNF)-mediated necroptosis. Here, we show that TNF-induced necroptosis and the PARP pathway represent distinct and independent routes to programmed necrosis. First, DNA-alkylating agents such as 1-methyl-3-nitro-1-nitrosoguanidine (MNNG) or methyl methanesulfonate rapidly activate the PARP pathway, whereas this is a late and secondary event in TNF-induced necroptosis. Second, inhibition of the PARP pathway does not protect against TNF-induced necroptosis, e.g., the PARP-1 inhibitor 3-AB prevented MNNG- but not TNF-induced adenosine-5'-triposphate depletion, translocation of apoptosis-inducing factor, and necrosis. Likewise, olaparib, a more potent and selective PARP-1 inhibitor failed to block TNF-induced necroptosis, identical to knockdown/knockout of PARP-1, pharmacologic and genetic interference with c-Jun N-terminal kinases and calpain/cathepsin proteases as further components of the PARP pathway. Third, interruption of TNF-induced necroptosis by interference with ceramide generation, RIP1 or RIP3 function or by the radical scavenger butylated hydroxyanisole did not prevent programmed necrosis through the PARP pathway. In summary, our results suggest that the currently established role of the PARP pathway in TNF-induced necroptosis needs to be revised, with consequences for the design of future therapeutic strategies.


Subject(s)
Apoptosis/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Benzamides/pharmacology , Calpain/metabolism , Cathepsins/metabolism , Cell Line , Ceramides/metabolism , Free Radical Scavengers/pharmacology , Guanidines/pharmacology , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Jurkat Cells , MCF-7 Cells , Methyl Methanesulfonate/pharmacology , Mice , Necrosis , Nuclear Pore Complex Proteins/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , RNA Interference , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
5.
Obesity (Silver Spring) ; 21(12): E626-33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23596049

ABSTRACT

OBJECTIVE: Beta-site amyloid precursor protein cleaving enzyme (BACE1) is highly expressed in pancreatic ß-cells. The BACE1 gene is located in a region associated with a high diabetes risk in PIMA Indians. DESIGN AND METHODS: INS-1E cells were used to study the impact of siRNA-mediated BACE1 knockdown and glucose metabolism was characterized in Bace1(-/-) mice. BACE1 gene was sequenced in DNA samples from 48 subjects and 13 representative single nucleotide polymorphisms (SNPs) were then genotyped for association studies in 1,527 Caucasians. RESULTS: Reduction of Bace1 expression results in a significant decrease in insulin mRNA expression in INS-1E cells. Bace1(-/-) mice display significantly lower body weight, lower plasma insulin concentrations, but normal glucose tolerance and insulin sensitivity. In a case-control study including 538 healthy controls and 989 patients with type 2 diabetes (T2D), one SNP (rs535860) was significantly associated with T2D (P < 3.5 × 10(-5) , adjusted for age, sex, and BMI). CONCLUSIONS: Reduced Bace1 expression causes impaired insulin expression in pancreatic ß-cells of Bace1(-/-) mice, suggesting that BACE1 plays a role in the regulation of insulin biogenesis. The functionally relevant rs535860 SNP may decrease BACE1 expression by creating a new miR-661 binding site and could therefore contribute to T2D development.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Gene Expression Regulation , Insulin/blood , RNA, Messenger/metabolism , Adult , Aged , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Body Mass Index , Case-Control Studies , Diabetes Mellitus, Type 2/genetics , Female , Genes, Reporter , Genotype , Glucose Tolerance Test , Humans , Insulin/genetics , Insulin Resistance , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Polymorphism, Single Nucleotide , RNA, Messenger/genetics
6.
Gut ; 62(4): 520-30, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22442160

ABSTRACT

OBJECTIVE: Cathepsin K is a lysosomal cysteine protease that has pleiotropic roles in bone resorption, arthritis, atherosclerosis, blood pressure regulation, obesity and cancer. Recently, it was demonstrated that cathepsin K-deficient (Ctsk(-/-) ) mice are less susceptible to experimental autoimmune arthritis and encephalomyelitis, which implies a functional role for cathepsin K in chronic inflammatory responses. Here, the authors address the relevance of cathepsin K in the intestinal immune response during chronic intestinal inflammation. DESIGN: Chronic colitis was induced by administration of 2% dextran sodium sulphate (DSS) in distilled water. Mice were assessed for disease severity, histopathology and endoscopic appearance. Furthermore, DSS-exposed Ctsk(-/-) mice were treated by rectal administration of recombinant cathepsin K. Intestinal microflora was assessed by real-time PCR and 16srDNA molecular fingerprinting of ileal and colonic mucosal and faecal samples. RESULTS: Using Ctsk(-/-) mice, the authors demonstrate a protective role of cathepsin K against chronic DSS colitis. Dissecting the underlying mechanisms the authors found cathepsin K to be present in intestinal goblet cells and the mucin layer. Furthermore, a direct cathepsin K-mediated bactericidal activity against intestinal bacteria was demonstrated, which potentially explains the alteration of intestinal microbiota observed in Ctsk(-/-) mice. Rectal administration of recombinant cathepsin K in DSS-treated Ctsk(-/-) mice ameliorates the severity of intestinal inflammation. CONCLUSION: These data identify extracellular cathepsin K as an intestinal antibacterial factor with anti-inflammatory potential and suggest that topical administration of cathepsin K might provide a therapeutic option for patients with inflammatory bowel disease.


Subject(s)
Cathepsin K/pharmacology , Colitis/drug therapy , Colitis/microbiology , Animals , Blotting, Western , Cathepsin K/metabolism , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate , Disease Models, Animal , Endoscopy, Gastrointestinal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
7.
Exp Brain Res ; 217(3-4): 331-41, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22120156

ABSTRACT

Alzheimer's disease causing mutations in the amyloid precursor protein (APP) or in the Presenilin 1 (PS1) or Presenilin 2 (PS2) genes increase the production of amyloid peptides (Aß) that precipitate in amyloid plaques. Since amyloid plaques are also a prominent feature of sporadic Alzheimer's disease (AD), abnormal proteolysis of APP and the generation of amyloid beta (Aß) are key events in the pathogenesis of AD. The proteases (secretases) that cleave APP are therefore important therapeutic targets, both for the rare familial forms but likely also for the sporadic forms of AD. The identification and understanding of the (neuro)biological functions of the α-, ß-, and presenilin/γ-secretase (complexes) is important for the development of drugs and the delineation of their associated side effects. The potential impact of this type of research exceeds the AD field since the function of these secretases are also linked to cellular pathways like ectodomain shedding of growth factors and regulated intramembrane proteolysis of receptors in developmental biology, tissue homeostasis, and tumorigenesis. The generation of mice deficient in presenilin 1, presenilin 2, the α-secretase ADAM10, and the ß-secretases BACE1 and BACE2 were instrumental for the elucidation of the physiological functions of these proteases. Using these mouse models understanding how these secretases regulate amyloid peptide formation and how they exert their diverse biological functions could be significantly increased. This review attempts to summarize selected aspects of the current view of the multiple roles such proteases play in health and disease.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , ADAM Proteins/physiology , ADAM10 Protein , Animals , Aspartic Acid Endopeptidases/physiology , Humans , Membrane Proteins/physiology , Mice , Models, Neurological , Neural Pathways/physiology , Presenilin-1/physiology , Presenilin-2/physiology
8.
Pflugers Arch ; 461(3): 355-71, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21246381

ABSTRACT

In cerebellar Purkinje cells, the ß4-subunit of voltage-dependent Na(+) channels has been proposed to serve as an open-channel blocker giving rise to a "resurgent" Na(+) current (I (NaR)) upon membrane repolarization. Notably, the ß4-subunit was recently identified as a novel substrate of the ß-secretase, BACE1, a key enzyme of the amyloidogenic pathway in Alzheimer's disease. Here, we asked whether BACE1-mediated cleavage of ß4-subunit has an impact on I (NaR) and, consequently, on the firing properties of Purkinje cells. In cerebellar tissue of BACE1-/- mice, mRNA levels of Na(+) channel α-subunits 1.1, 1.2, and 1.6 and of ß-subunits 1-4 remained unchanged, but processing of ß4 peptide was profoundly altered. Patch-clamp recordings from acutely isolated Purkinje cells of BACE1-/- and WT mice did not reveal any differences in steady-state properties and in current densities of transient, persistent, and resurgent Na(+) currents. However, I (NaR) was found to decay significantly faster in BACE1-deficient Purkinje cells than in WT cells. In modeling studies, the altered time course of I (NaR) decay could be replicated when we decreased the efficiency of open-channel block. In current-clamp recordings, BACE1-/- Purkinje cells displayed lower spontaneous firing rate than normal cells. Computer simulations supported the hypothesis that the accelerated decay kinetics of I (NaR) are responsible for the slower firing rate. Our study elucidates a novel function of BACE1 in the regulation of neuronal excitability that serves to tune the firing pattern of Purkinje cells and presumably other neurons endowed with I (NaR).


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Aspartic Acid Endopeptidases/physiology , Purkinje Cells/physiology , Sodium Channels/metabolism , Amyloid Precursor Protein Secretases/deficiency , Animals , Aspartic Acid Endopeptidases/deficiency , Cerebellum/metabolism , Mice , Patch-Clamp Techniques , Purkinje Cells/drug effects , Voltage-Gated Sodium Channel beta-4 Subunit
9.
J Neurosci ; 30(36): 12210-8, 2010 Sep 08.
Article in English | MEDLINE | ID: mdl-20826683

ABSTRACT

Tumor necrosis factor alpha (TNF-alpha) is a potent cytokine in neurodegenerative disorders, but its precise role in particular brain disorders is ambiguous. In motor neuron (MN) disease of the mouse, exemplified by the model wobbler (WR), TNF-alpha causes upregulation of the metalloprotease-disintegrin ADAM8 (A8) in affected brain regions, spinal cord, and brainstem. The functional role of A8 during MN degeneration in the wobbler CNS was investigated by crossing WR with A8-deficient mice: a severely aggravated neuropathology was observed for A8-deficient WR compared with WR A8(+/-) mice, judged by drastically reduced survival [7 vs 81% survival at postnatal day 50 (P50)], accelerated force loss in the forelimbs, and terminal akinesis. In vitro protease assays using soluble A8 indicated specific cleavage of a TNF-alpha receptor 1 (p55 TNF-R1) but not a TNF-R2 peptide. Cleavage of TNF-R1 was confirmed in situ, because levels of soluble TNF-R1 were increased in spinal cords of standard WR compared with wild-type mice but not in A8-deficient WR mice. In isolated primary neurons and microglia, TNF-alpha-induced TNF-R1 shedding was dependent on the A8 gene dosage. Furthermore, exogenous TNF-alpha showed higher toxicity for cultured neurons from A8-deficient than for those from wild-type mice, demonstrating that TNF-R1 shedding by A8 is neuroprotective. Our results indicate an essential role for ADAM8 in modulating TNF-alpha signaling in CNS diseases: a feedback loop integrating TNF-alpha, ADAM8, and TNF-R1 shedding as a plausible mechanism for TNF-alpha mediated neuroprotection in situ and a rationale for therapeutic intervention.


Subject(s)
ADAM Proteins/metabolism , Antigens, CD/metabolism , Gene Expression Regulation/genetics , Membrane Proteins/metabolism , Neurodegenerative Diseases/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM Proteins/deficiency , ADAM Proteins/genetics , ADAM Proteins/pharmacology , Animals , Animals, Newborn , Antigens, CD/genetics , Antigens, CD/pharmacology , Cell Count/methods , Central Nervous System/metabolism , Central Nervous System/pathology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Humans , Kaplan-Meier Estimate , Leukocyte Common Antigens/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscular Atrophy/genetics , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/mortality , Neurodegenerative Diseases/pathology , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Type I/drug effects , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor-alpha/pharmacology
10.
Neurobiol Dis ; 33(2): 282-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19056495

ABSTRACT

The beta-site APP-cleaving enzyme 1 (BACE1) is widely known for its pivotal role in the amyloidogenic pathway leading to Alzheimer's disease. Here, we elaborate on the recent finding that auxiliary subunits of voltage-gated sodium channels (beta2 and beta4) are BACE substrates. BACE1 produced complex effects on sodium channel gating that could be only partially explained by beta2/beta4 cleavage. To characterize the unexpected non-proteolytic effect of BACE1, we examined HEK cells co-transfected with only Nav1.2 and either normal or catalytically inactive BACE1. Both BACE1 variants produced virtually identical effects on sodium channel gating, which would lead to enhanced cellular excitability. The non-proteolytic BACE1 effect on Nav1.2 current was confirmed in murine neuroblastoma cells, which express sodium channels endogenously, but lack beta2 and beta4. Our study reveals an important facet of BACE1 function that should help to decipher the role of BACE1 in normal and demented brain.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Nerve Tissue Proteins/metabolism , Sodium Channels/metabolism , Animals , Blotting, Western , Cell Line , Humans , Immunohistochemistry , Ion Channel Gating/physiology , Membrane Potentials , Mice , NAV1.2 Voltage-Gated Sodium Channel , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Voltage-Gated Sodium Channel beta-2 Subunit , Voltage-Gated Sodium Channel beta-4 Subunit
11.
Biol Reprod ; 78(6): 1038-48, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18287572

ABSTRACT

ADAM8 (a disintegrin and metalloprotease domain 8) is expressed in immune, neuronal, and bone progenitor cells and is thought to be involved in the tissue-remodeling process. Microarray analyses indicate that Adam8 is a potential target of the progesterone receptor (Pgr) in murine ovary. Further studies document that Adam8 mRNA and protein are expressed in granulosa cells and cumulus cells of periovulatory follicles whereas expression is significantly reduced in Pgr null mice that fail to ovulate. There is a reduced expression in granulosa cells from cultured, in vitro ovulated follicles exposed to inhibitors of progesterone or epidermal growth factor signaling while epiregulin induced its expression in the absence of hCG. In vitro studies with primary mouse granulosa cells document that Adam8 is induced in response to forskolin (Fo) and phorbol ester (PMA) or Fo and Amphiregulin treatment. To understand the transcriptional regulation of the Adam8, we amplified 1 kb of the mouse Adam8 promoter by PCR and subcloned it into a pGL3-luciferase reporter construct. The Adam8 promoter-luciferase constructs are induced by Fo and PMA treatment after transfection into rat granulosa cells, and cotransfection with a PGR-A expression vector further augment basal and Fo/PMA inducibility. Site-specific mutations within the -615/+50 promoter document that a GC-rich region, NF-1 (nuclear factor-1) site, and putative TATA box are critical for Adam8 promoter activation by Fo/PMA. Thus, ADAM8 is expressed in a stage-specific manner and is hormonally regulated in ovulating follicles by the coordinate actions of LH and PGR. To our knowledge, ADAM8 is the first member of the ADAM family shown to be hormonally regulated.


Subject(s)
ADAM Proteins/genetics , ADAM Proteins/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ovary/enzymology , Animals , Base Sequence , Cells, Cultured , Chorionic Gonadotropin/pharmacology , DNA Primers/genetics , Epidermal Growth Factor/pharmacology , Female , Gene Expression Regulation, Enzymologic/drug effects , Granulosa Cells/drug effects , Granulosa Cells/enzymology , Luteinizing Hormone/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutagenesis, Site-Directed , Ovary/drug effects , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
12.
Science ; 314(5799): 664-6, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16990514

ABSTRACT

Although BACE1 (beta-site amyloid precursor protein-cleaving enzyme 1) is essential for the generation of amyloid-b peptide in Alzheimer's disease, its physiological function is unclear. We found that very high levels of BACE1 were expressed at time points when peripheral nerves become myelinated. Deficiency of BACE1 resulted in the accumulation of unprocessed neuregulin 1 (NRG1), an axonally expressed factor required for glial cell development and myelination. BACE1-/- mice displayed hypomyelination of peripheral nerves and aberrant axonal segregation of small-diameter afferent fibers, very similar to that seen in mice with mutations in type III NRG1 or Schwann cell-specific ErbB2 knockouts. Thus, BACE1 is required for myelination and correct bundling of axons by Schwann cells, probably through processing of type III NRG1.


Subject(s)
Axons/physiology , Endopeptidases/metabolism , Myelin Sheath/physiology , Nerve Tissue Proteins/metabolism , Neuregulin-1/metabolism , Schwann Cells/physiology , Sciatic Nerve/physiology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Endopeptidases/genetics , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Mice , Mice, Knockout , Motor Neurons/metabolism , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neuregulin-1/chemistry , Neuregulin-1/genetics , Neurons, Afferent/metabolism , Protein Isoforms , Protein Processing, Post-Translational , Sciatic Nerve/cytology , Spinal Cord/cytology , Spinal Cord/metabolism
13.
Dev Dyn ; 232(1): 221-31, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15580619

ABSTRACT

ADAM8 (a disintegrin and metalloprotease 8, also referred to as MS2/CD156a) is a membrane-anchored metalloprotease that was first identified in a macrophage cell line and has been implicated in neurodegenerative diseases. Here, we evaluated the expression of ADAM8 during mouse development and generated mice lacking ADAM8 (Adam8-/- mice). During early mouse development, ADAM8 is expressed by maternal cells in the decidua and by trophoblast derivatives of the embryo but not in the derivatives of the inner cell mass. At later stages, prominent expression of ADAM8 is seen in the embryo proper, in the gonadal ridge, thymus, developing cartilage and bone, brain and spinal cord, and in the mesenchyme in close proximity to the branch point between the jugular vein and developing lymphatic vessels. Examination of Adam8-/- mice, however, revealed no major defects in these or other structures during development or in adult tissues and no evident pathological phenotypes.


Subject(s)
Antigens, CD/biosynthesis , Antigens, CD/genetics , Gene Deletion , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , ADAM Proteins , Alleles , Animals , Blotting, Western , Bone Development , Bone and Bones , Female , Gene Expression Regulation, Developmental , Genetic Vectors , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , Lac Operon , Macrophages/metabolism , Male , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Genetic , Mutation , Time Factors , Tissue Distribution , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...