251
|
Baranger K, Bonnet AE, Girard SD, Paumier JM, García-González L, Elmanaa W, Bernard A, Charrat E, Stephan D, Bauer C, Moschke K, Lichtenthaler SF, Roman FS, Checler F, Khrestchatisky M, Rivera S. MT5-MMP Promotes Alzheimer's Pathogenesis in the Frontal Cortex of 5xFAD Mice and APP Trafficking in vitro. Front Mol Neurosci 2017; 9:163. [PMID: 28119565 PMCID: PMC5223243 DOI: 10.3389/fnmol.2016.00163] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/16/2016] [Indexed: 12/18/2022] Open
Abstract
We previously reported that deficiency of membrane-type five matrix metalloproteinase (MT5-MMP) prevents amyloid pathology in the cortex and hippocampus of 5xFAD mice, and ameliorates the functional outcome. We have now investigated whether the integrity of another important area affected in Alzheimer's disease (AD), the frontal cortex, was also preserved upon MT5-MMP deficiency in 4-month old mice at prodromal stages of the pathology. We used the olfactory H-maze (OHM) to show that learning impairment associated with dysfunctions of the frontal cortex in 5xFAD was prevented in bigenic 5xFAD/MT5-MMP-/- mice. The latter exhibited concomitant drastic reductions of amyloid beta peptide (Aβ) assemblies (soluble, oligomeric and fibrillary) and its immediate precursor, C99. Simultaneously, astrocyte reactivity and tumor necrosis factor alpha (TNF-α) levels were also lowered. Moreover, MT5-MMP deficiency induced a decrease in N-terminal soluble fragments of amyloid precursor protein (APP), including soluble APPα (sAPPα), sAPPβ and the MT5-MMP-linked fragment of 95 kDa, sAPP95. However, the lack of MT5-MMP did not affect the activity of β- and γ-secretases. In cultured HEKswe cells, transiently expressed MT5-MMP localized to early endosomes and increased the content of APP and Aβ40 in these organelles, as well as Aβ levels in cell supernatants. This is the first evidence that the pro-amyloidogenic features of MT5-MMP lie, at least in part, on the ability of the proteinase to promote trafficking into one of the amyloidogenic subcellular loci. Together, our data further support the pathogenic role of MT5-MMP in AD and that its inhibition improves the functional and pathological outcomes, in this case in the frontal cortex. These data also support the idea that MT5-MMP could become a novel therapeutic target in AD.
Collapse
Affiliation(s)
- Kévin Baranger
- Aix Marseille Université, CNRS, NICN UMR 7259 Marseille, France
| | | | | | | | | | - Wejdane Elmanaa
- Université Côte d'Azur, INSERM, CNRS, IPMC, Laboratory of excellence DistALZ, Sophia-Antipolis Valbonne, France
| | - Anne Bernard
- Aix Marseille Université, CNRS, NICN UMR 7259 Marseille, France
| | - Eliane Charrat
- Aix Marseille Université, CNRS, NICN UMR 7259 Marseille, France
| | | | - Charlotte Bauer
- Université Côte d'Azur, INSERM, CNRS, IPMC, Laboratory of excellence DistALZ, Sophia-Antipolis Valbonne, France
| | - Katrin Moschke
- German Center for Neurodegenerative Diseases (DZNE) Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE)Munich, Germany; Neuroproteomics, Klinikum rechts der Isar, and Institute for Advanced Study, Technische Universität München (TUM)Munich, Germany; Munich Cluster for Systems Neurology (SyNergy)Munich, Germany
| | | | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, Laboratory of excellence DistALZ, Sophia-Antipolis Valbonne, France
| | | | - Santiago Rivera
- Aix Marseille Université, CNRS, NICN UMR 7259 Marseille, France
| |
Collapse
|
252
|
Hammad H, Vanderkerken M, Pouliot P, Deswarte K, Toussaint W, Vergote K, Vandersarren L, Janssens S, Ramou I, Savvides SN, Haigh JJ, Hendriks R, Kopf M, Craessaerts K, de Strooper B, Kearney JF, Conrad DH, Lambrecht BN. Transitional B cells commit to marginal zone B cell fate by Taok3-mediated surface expression of ADAM10. Nat Immunol 2017; 18:313-320. [PMID: 28068307 DOI: 10.1038/ni.3657] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 12/06/2016] [Indexed: 12/17/2022]
Abstract
Notch2 and B cell antigen receptor (BCR) signaling determine whether transitional B cells become marginal zone B (MZB) or follicular B (FoB) cells in the spleen, but it is unknown how these pathways are related. We generated Taok3-/- mice, lacking the serine/threonine kinase Taok3, and found cell-intrinsic defects in the development of MZB but not FoB cells. Type 1 transitional (T1) B cells required Taok3 to rapidly respond to ligation by the Notch ligand Delta-like 1. BCR ligation by endogenous or exogenous ligands induced the surface expression of the metalloproteinase ADAM10 on T1 B cells in a Taok3-dependent manner. T1 B cells expressing surface ADAM10 were committed to becoming MZB cells in vivo, whereas T1 B cells lacking expression of ADAM10 were not. Thus, during positive selection in the spleen, BCR signaling causes immature T1 B cells to become receptive to Notch ligands via Taok3-mediated surface expression of ADAM10.
Collapse
Affiliation(s)
- Hamida Hammad
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Matthias Vanderkerken
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Philippe Pouliot
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Kim Deswarte
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Wendy Toussaint
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Karl Vergote
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Lana Vandersarren
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Sophie Janssens
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Ioanna Ramou
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,The Laboratory for Protein Biochemistry and Biomolecular Engineering (L-Probe), Ghent University, Ghent, Belgium
| | - Savvas N Savvides
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,The Laboratory for Protein Biochemistry and Biomolecular Engineering (L-Probe), Ghent University, Ghent, Belgium
| | - Jody J Haigh
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium
| | - Rudi Hendriks
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Manfred Kopf
- Institute for Molecular Health Sciences, ETH, Zürich, Switzerland
| | - Katleen Craessaerts
- VIB Center for Brain and Disease, VIB, Leuven, Belgium.,Center for Human Genetics, KULeuven, Leuven, Belgium
| | - Bart de Strooper
- VIB Center for Brain and Disease, VIB, Leuven, Belgium.,Center for Human Genetics, KULeuven, Leuven, Belgium
| | - John F Kearney
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Daniel H Conrad
- Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Bart N Lambrecht
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium.,Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
253
|
Aberrant proteolytic processing and therapeutic strategies in Alzheimer disease. Adv Biol Regul 2017; 64:33-38. [PMID: 28082052 DOI: 10.1016/j.jbior.2017.01.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 12/24/2016] [Accepted: 01/04/2017] [Indexed: 01/18/2023]
Abstract
Amyloid-β peptide (Aβ) and tau are major components of senile plaques and neurofibrillary tangles, respectively, deposited in the brains of Alzheimer disease (AD) patients. Aβ is derived from amyloid-β precursor protein that is sequentially cleaved by two aspartate proteases, β- and γ-secretases. Secreted Aβ is then catabolized by several proteases. Several lines of evidence suggest that accumulation of Aβ by increased production or decreased degradation induces the tau-mediated neuronal toxicity and symptomatic manifestations of AD. Thus, the dynamics of cerebral Aβ, called as "Aβ economy", would be the mechanistic basis of AD pathogenesis. Partial loss of γ-secretase activity leads to the increased generation of toxic Aβ isoforms, indicating that activation of γ-secretase would provide a beneficial effect for AD. After extensive discovery and development efforts, BACE1, which is a β-secretase enzyme, has emerged as a prime drug target for lowering brain Aβ levels. Recent studies revealed the decreased clearance of Aβ in sporadic AD patients, suggesting the importance of the catabolic mechanism in the pathogenesis of AD. I will discuss with these proteolytic mechanisms involved in the regulation of Aβ economy, and development of effective treatment and diagnostics for AD.
Collapse
|
254
|
Zeng J, Li T, Gong M, Jiang W, Yang T, Chen J, Liu Y, Chen L. Marginal Vitamin A Deficiency Exacerbates Memory Deficits Following Aβ1-42 Injection in Rats. Curr Alzheimer Res 2017; 14:562-570. [PMID: 28017127 PMCID: PMC5421133 DOI: 10.2174/1567205013666161223162110] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/09/2016] [Accepted: 12/16/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Although clinical vitamin A deficiency (VAD), which is a public health problem developing throughout the world, has been well controlled, marginal vitamin A deficiency (MVAD) is far more prevalent, especially among pregnant women and preschool children in China. Increasing evidence suggests that VAD is involved in the pathogenesis of Alzheimer's disease (AD). However, whether MVAD, beginning early in life, increases the risk of developing AD has yet to be determined. OBJECTIVE The goal of this study was to investigate the long-term effects of MVAD on the pathogenesis of AD in rats. METHOD An MVAD model was generated from maternal MVAD rats and maintained with an MVAD diet after weaning. The males were bilaterally injected with aggregated amyloid β (Aβ)1-42 into the CA3 area of the hippocampus, and the AD-associated cognitive and neuropathological phenotypes were examined. RESULTS We found that MVAD feeding significantly aggravated Aβ1-42-induced learning and memory deficits in the Morris water maze test. MVAD did not induce the mRNA expression of retinoic acid receptors (RARs), a disintegrin and metalloprotease 10 (ADAM10) or insulin-degrading enzyme (IDE) in Aβ1-42-injected rats. Moreover, RARα and RARγ mRNA were positively correlated with ADAM10 mRNA, whereas RARβ mRNA was positively correlated with IDE mRNA. CONCLUSION Our study suggests that MVAD beginning from the embryonic period perturbs the ADassociated genes, resulting in an enhanced risk of developing AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Li Chen
- Address correspondence to this author at the No.136 Zhongshan Er Road, Yuzhong District, Chongqing, 400014 P.R. China; Tel: 86-23-61966251; Fax: 86-23-61966253; E-mail:
| |
Collapse
|
255
|
Overexpression of MTERF4 promotes the amyloidogenic processing of APP by inhibiting ADAM10. Biochem Biophys Res Commun 2017; 482:928-934. [DOI: 10.1016/j.bbrc.2016.11.135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 11/24/2016] [Indexed: 12/21/2022]
|
256
|
He T, Santhanam AVR, Lu T, d'Uscio LV, Katusic ZS. Role of prostacyclin signaling in endothelial production of soluble amyloid precursor protein-α in cerebral microvessels. J Cereb Blood Flow Metab 2017; 37:106-122. [PMID: 26661245 PMCID: PMC5363732 DOI: 10.1177/0271678x15618977] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/20/2015] [Accepted: 10/30/2015] [Indexed: 12/20/2022]
Abstract
We tested hypothesis that activation of the prostacyclin (PGI2) receptor (IP receptor) signaling pathway in cerebral microvessels plays an important role in the metabolism of amyloid precursor protein (APP). In human brain microvascular endothelial cells activation of IP receptor with the stable analogue of PGI2, iloprost, stimulated expression of amyloid precursor protein and a disintegrin and metalloprotease 10 (ADAM10), resulting in an increased production of the neuroprotective and anticoagulant molecule, soluble APPα (sAPPα). Selective agonist of IP receptor, cicaprost, and adenylyl cyclase activator, forskolin, also enhanced expression of amyloid precursor protein and ADAM10. Notably, in cerebral microvessels of IP receptor knockout mice, protein levels of APP and ADAM10 were reduced. In addition, iloprost increased protein levels of peroxisome proliferator-activated receptor δ (PPARδ) in human brain microvascular endothelial cells. PPARδ-siRNA abolished iloprost-augmented protein expression of ADAM10. In contrast, GW501516 (a selective agonist of PPARδ) upregulated ADAM10 and increased production of sAPPα. Genetic deletion of endothelial PPARδ (ePPARδ-/-) in mice significantly reduced cerebral microvascular expression of ADAM10 and production of sAPPα. In vivo treatment with GW501516 increased sAPPα content in hippocampus of wild type mice but not in hippocampus of ePPARδ-/- mice. Our findings identified previously unrecognized role of IP-PPARδ signal transduction pathway in the production of sAPPα in cerebral microvasculature.
Collapse
Affiliation(s)
- Tongrong He
- Department of Anesthesiology and Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Anantha Vijay R Santhanam
- Department of Anesthesiology and Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Tong Lu
- Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Livius V d'Uscio
- Department of Anesthesiology and Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Zvonimir S Katusic
- Department of Anesthesiology and Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
257
|
Oi R, Koizumi H, Maeda I, Noguchi A, Tatsunami S, Iwatani T, Kawamoto H, Tsugawa K, Takagi M. Clinicopathological Significance of TARBP2, APP, and ZNF395 in Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2016; 10:211-221. [PMID: 27980417 PMCID: PMC5147454 DOI: 10.4137/bcbcr.s40820] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 01/29/2023]
Abstract
The double-stranded RNA-binding protein TARBP2 has been suggested to act as an upstream regulator of breast cancer metastasis by destabilizing transcripts of the possible metastasis suppressors amyloid precursor protein (APP) and ZNF395. We examined this hypothesis by immunostaining of TARBP2, APP, and ZNF395 in 200 breast cancer specimens using tissue microarrays and analyzed the relationships between expression levels and clinicopathological parameters and prognosis. Increased TARBP2 overexpression was associated with shorter overall survival and disease-free survival, and increased but not reduced APP expression correlated with lower overall survival and disease-free survival. ZNF395 expression levels had no prognostic value, but reduced expression correlated with reduced lymph node metastasis. There was no significant relationship between TARBP2 overexpression and reduced APP and/or ZNF395 expression. Patients with tumors with higher TARBP2 or APP expression had unfavorable prognoses. Although reduced ZNF395 expression was significantly related to reduced lymph node metastasis, further studies are needed to clarify the role of TARBP2/APP/ZNF395 in breast cancer.
Collapse
Affiliation(s)
- Ryoko Oi
- Department of Pathology, St. Marianna University School of Medicine, Kawasaki, Japan.; Division of Breast and Endocrine Surgery, Department of Surgery, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hirotaka Koizumi
- Department of Pathology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Ichiro Maeda
- Department of Pathology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Akira Noguchi
- Department of Pathology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Shinobu Tatsunami
- Unit of Medical Informatics, Faculty of Education and Culture, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tsuguo Iwatani
- Division of Breast and Endocrine Surgery, Department of Surgery, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hisanori Kawamoto
- Division of Breast and Endocrine Surgery, Department of Surgery, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Koichiro Tsugawa
- Division of Breast and Endocrine Surgery, Department of Surgery, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Masayuki Takagi
- Department of Pathology, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
258
|
Koo JH, Kang EB, Oh YS, Yang DS, Cho JY. Treadmill exercise decreases amyloid-β burden possibly via activation of SIRT-1 signaling in a mouse model of Alzheimer's disease. Exp Neurol 2016; 288:142-152. [PMID: 27889467 DOI: 10.1016/j.expneurol.2016.11.014] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/27/2016] [Accepted: 11/22/2016] [Indexed: 02/07/2023]
Abstract
Accumulation of amyloid-β (Aβ) correlates significantly with progressive cognitive deficits, a main symptom of Alzheimer's disease (AD). Although treadmill exercise reduces Aβ levels, the molecular mechanisms underlying the effects are not fully understood. We hypothesize that treadmill exercise decreases Aβ production and alleviates cognitive deficits by activating the non-amyloidogenic pathway via SIRT-1 signaling. Treadmill exercise improved cognitive deficits and alleviated neurotoxicity. Most importantly, treadmill exercise increased SIRT-1 level, which subsequently resulted in increased ADAM-10 level by down-regulation of ROCK-1 and upregulation of RARβ, ultimately facilitating the non-amyloidogenic pathway. Treadmill exercise-induced activation in SIRT-1 level also elevated PGC-1α level and reduced BACE-1 and C-99 level, resulting in inhibition of the amyloidogenic pathway. Treadmill exercise may thus inhibit Aβ production via upregulation of SIRT-1, which biases amyloid precursor protein processing toward the non-amyloidogenic pathway. This study provides novel and valuable insight into the molecular mechanisms possibly by which treadmill exercise reduces Aβ production.
Collapse
Affiliation(s)
- Jung-Hoon Koo
- Department of Exercise Biochemistry, Korea National Sport University, Seoul 138-763, Republic of Korea
| | - Eun-Bum Kang
- Department of Exercise Biochemistry, Korea National Sport University, Seoul 138-763, Republic of Korea
| | - Yoo-Sung Oh
- Department of Exercise Prescription, University of Seoul, Seoul 163, Republic of Korea
| | - Dae-Seung Yang
- Department of Taekwondo Studies, University of Gachon, Gyeonggido, Republic of Korea.
| | - Joon-Yong Cho
- Department of Exercise Biochemistry, Korea National Sport University, Seoul 138-763, Republic of Korea.
| |
Collapse
|
259
|
Mbah NE, Overmeyer JH, Maltese WA. Disruption of endolysosomal trafficking pathways in glioma cells by methuosis-inducing indole-based chalcones. Cell Biol Toxicol 2016; 33:263-282. [PMID: 27822587 DOI: 10.1007/s10565-016-9369-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/26/2016] [Indexed: 12/21/2022]
Abstract
Methuosis is a form of non-apoptotic cell death involving massive vacuolization of macropinosome-derived endocytic compartments, followed by a decline in metabolic activity and loss of membrane integrity. To explore the induction of methuosis as a potential therapeutic strategy for killing cancer cells, we have developed small molecules (indole-based chalcones) that trigger this form of cell death in glioblastoma and other cancer cell lines. Here, we report that in addition to causing fusion and expansion of macropinosome compartments, the lead compound, 3-(5-methoxy-2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propen-1-one (MOMIPP), disrupts vesicular trafficking at the lysosomal nexus, manifested by impaired degradation of EGF and LDL receptors, defective processing of procathepsins, and accumulation of autophagosomes. In contrast, secretion of the ectodomain derived from a prototypical type-I membrane glycoprotein, β-amyloid precursor protein, is increased rather than diminished. A closely related MOMIPP analog, which causes substantial vacuolization without reducing cell viability, also impedes cathepsin processing and autophagic flux, but has more modest effects on receptor degradation. A third analog, which causes neither vacuolization nor loss of viability, has no effect on endolysosomal trafficking. The results suggest that differential cytotoxicity of structurally similar indole-based chalcones is related, at least in part, to the severity of their effects on endolysosomal trafficking pathways.
Collapse
Affiliation(s)
- Nneka E Mbah
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA
| | - Jean H Overmeyer
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA
| | - William A Maltese
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA.
| |
Collapse
|
260
|
Seipold L, Damme M, Prox J, Rabe B, Kasparek P, Sedlacek R, Altmeppen H, Willem M, Boland B, Glatzel M, Saftig P. Tetraspanin 3: A central endocytic membrane component regulating the expression of ADAM10, presenilin and the amyloid precursor protein. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:217-230. [PMID: 27818272 DOI: 10.1016/j.bbamcr.2016.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 12/31/2022]
Abstract
Despite existing knowledge about the role of the A Disintegrin and Metalloproteinase 10 (ADAM10) as the α-secretase involved in the non-amyloidogenic processing of the amyloid precursor protein (APP) and Notch signalling we have only limited information about its regulation. In this study, we have identified ADAM10 interactors using a split ubiquitin yeast two hybrid approach. Tetraspanin 3 (Tspan3), which is highly expressed in the murine brain and elevated in brains of Alzheimer´s disease (AD) patients, was identified and confirmed to bind ADAM10 by co-immunoprecipitation experiments in mammalian cells in complex with APP and the γ-secretase protease presenilin. Tspan3 expression increased the cell surface levels of its interacting partners and was mainly localized in early and late endosomes. In contrast to the previously described ADAM10-binding tetraspanins, Tspan3 did not affect the endoplasmic reticulum to plasma membrane transport of ADAM10. Heterologous Tspan3 expression significantly increased the appearance of carboxy-terminal cleavage products of ADAM10 and APP, whereas N-cadherin ectodomain shedding appeared unaffected. Inhibiting the endocytosis of Tspan3 by mutating a critical cytoplasmic tyrosine-based internalization motif led to increased surface expression of APP and ADAM10. After its downregulation in neuroblastoma cells and in brains of Tspan3-deficient mice, ADAM10 and APP levels appeared unaltered possibly due to a compensatory increase in the expression of Tspans 5 and 7, respectively. In conclusion, our data suggest that Tspan3 acts in concert with other tetraspanins as a stabilizing factor of active ADAM10, APP and the γ-secretase complex at the plasma membrane and within the endocytic pathway.
Collapse
Affiliation(s)
- Lisa Seipold
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Markus Damme
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Johannes Prox
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Björn Rabe
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Petr Kasparek
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Division BIOCEV, Institute of Molecular Genetics of the CAS, v. v. i., Vestec, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Division BIOCEV, Institute of Molecular Genetics of the CAS, v. v. i., Vestec, Czech Republic
| | - Hermann Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Willem
- Biomedical Center, Ludwig-Maximilians-University, 81337 Munich, Germany
| | - Barry Boland
- Department of Pharmacology and Therapeutics, School of Medicine, University College Cork, Cork, Ireland
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Olshausenstrasse 40, D-24098 Kiel, Germany.
| |
Collapse
|
261
|
Andersen OM, Rudolph IM, Willnow TE. Risk factor SORL1: from genetic association to functional validation in Alzheimer's disease. Acta Neuropathol 2016; 132:653-665. [PMID: 27638701 PMCID: PMC5073117 DOI: 10.1007/s00401-016-1615-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/12/2016] [Accepted: 09/05/2016] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) represents one of the most dramatic threats to healthy aging and devising effective treatments for this devastating condition remains a major challenge in biomedical research. Much has been learned about the molecular concepts that govern proteolytic processing of the amyloid precursor protein to amyloid-β peptides (Aβ), and how accelerated accumulation of neurotoxic Aβ peptides underlies neuronal cell death in rare familial but also common sporadic forms of this disease. Out of a plethora of proposed modulators of amyloidogenic processing, one protein emerged as a key factor in AD pathology, a neuronal sorting receptor termed SORLA. Independent approaches using human genetics, clinical pathology, or exploratory studies in animal models all converge on this receptor that is now considered a central player in AD-related processes by many. This review will provide a comprehensive overview of the evidence implicating SORLA-mediated protein sorting in neurodegenerative processes, and how receptor gene variants in the human population impair functional receptor expression in sporadic but possibly also in autosomal-dominant forms of AD.
Collapse
Affiliation(s)
- Olav M Andersen
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Ole Worms Alle 3, Aarhus C, 8000, Aarhus, Denmark.
| | - Ina-Maria Rudolph
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Strasse 10, 13125, Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Strasse 10, 13125, Berlin, Germany.
| |
Collapse
|
262
|
Schuck F, Wolf D, Fellgiebel A, Endres K. Increase of α-Secretase ADAM10 in Platelets Along Cognitively Healthy Aging. J Alzheimers Dis 2016; 50:817-26. [PMID: 26757187 DOI: 10.3233/jad-150737] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
ADAM10 is one of the key players in ectodomain-shedding of the amyloid-β protein precursor (AβPP). Previous research with postmortem tissue has shown reduced expression and activity of ADAM10 within the central nervous system (CNS) of Alzheimer's disease (AD) patients. Determination of cerebral ADAM10 in living humans is hampered by its transmembrane property; only the physiological AβPP cleavage product generated by ADAM10, sAβPPα, can be assessed in cerebrospinal fluid. Establishment of surrogate markers in easily accessible material therefore is crucial. It has been demonstrated that ADAM10 is expressed in platelets and that platelet amount is decreased in AD patients. Just recently it has been shown that platelet ADAM10 and cognitive performance of AD patients positively correlate. In contrast to AD patients, to our knowledge almost no information has been published regarding ADAM10 expression during normal aging. We investigated ADAM10 amount and activity in platelets of cognitively healthy individuals from three different age groups ranging from 22-85 years. Interestingly, we observed an age-dependent increase in ADAM10 levels and activity in platelets.
Collapse
|
263
|
Fendiline inhibits proliferation and invasion of pancreatic cancer cells by interfering with ADAM10 activation and β-catenin signaling. Oncotarget 2016; 6:35931-48. [PMID: 26440150 PMCID: PMC4742152 DOI: 10.18632/oncotarget.5933] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/14/2015] [Indexed: 12/20/2022] Open
Abstract
ADAM10 (A Disintegrin and Metalloprotease Domain 10) affects the pathophysiology of various cancers, and we had shown that inhibition of ADAM10 sensitizes pancreatic cancer cells to gemcitabine. ADAM10 is activated in response to calcium influx, and here we examined if calcium channel blockers (CCB) would impede ADAM10 activation and affect biology of pancreatic cancer cells. We find that the CCB, fendiline, significantly reduces proliferation, migration, invasion, and anchorage independent growth of pancreatic cancer cells. This was associated with ADAM10 inhibition and its localization at the actin-rich membrane protrusions. Further, fendiline-treated cells formed cadherin-catenin positive tight adherens junctions and elicited defective protein trafficking and recycling. Furthermore, the expression of β-catenin target genes, cyclinD1, c-Myc and CD44, were significantly decreased, suggesting that fendiline might prevent cell proliferation and migration by inhibiting ADAM10 function, cadherin proteolysis and stabilization of cadherin-catenin interaction at the plasma membrane. This will subsequently diminish β-catenin intracellular signaling and repress TCF/LEF target gene expression. Supporting this notion, RNAi-directed downregulation of ADAM10 in cancer cells decreased the expression of cyclinD1, c-Myc and CD44. Furthermore, analysis of human pancreatic tumor tissue microarrays and lysates showed elevated levels of ADAM10, suggesting that aberrant activation of ADAM10 plays a fundamental role in growth and metastasis of PDACs and inhibiting this pathway might be a viable strategy to combat PDACs.
Collapse
|
264
|
Müller SA, Scilabra SD, Lichtenthaler SF. Proteomic Substrate Identification for Membrane Proteases in the Brain. Front Mol Neurosci 2016; 9:96. [PMID: 27790089 PMCID: PMC5062031 DOI: 10.3389/fnmol.2016.00096] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/21/2016] [Indexed: 12/26/2022] Open
Abstract
Cell-cell communication in the brain is controlled by multiple mechanisms, including proteolysis. Membrane-bound proteases generate signaling molecules from membrane-bound precursor proteins and control the length and function of cell surface membrane proteins. These proteases belong to different families, including members of the “a disintegrin and metalloprotease” (ADAM), the beta-site amyloid precursor protein cleaving enzymes (BACE), membrane-type matrix metalloproteases (MT-MMP) and rhomboids. Some of these proteases, in particular ADAM10 and BACE1 have been shown to be essential not only for the correct development of the mammalian brain, but also for myelination and maintaining neuronal connections in the adult nervous system. Additionally, these proteases are considered as drug targets for brain diseases, including Alzheimer’s disease (AD), schizophrenia and cancer. Despite their biomedical relevance, the molecular functions of these proteases in the brain have not been explored in much detail, as little was known about their substrates. This has changed with the recent development of novel proteomic methods which allow to identify substrates of membrane-bound proteases from cultured cells, primary neurons and other primary brain cells and even in vivo from minute amounts of mouse cerebrospinal fluid (CSF). This review summarizes the recent advances and highlights the strengths of the individual proteomic methods. Finally, using the example of the Alzheimer-related proteases BACE1, ADAM10 and γ-secretase, as well as ADAM17 and signal peptide peptidase like 3 (SPPL3), we illustrate how substrate identification with novel methods is instrumental in elucidating broad physiological functions of these proteases in the brain and other organs.
Collapse
Affiliation(s)
- Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE)Munich, Germany; Neuroproteomics, Klinikum rechts der Isar, Technische Universität MünchenMunich, Germany
| | - Simone D Scilabra
- German Center for Neurodegenerative Diseases (DZNE)Munich, Germany; Neuroproteomics, Klinikum rechts der Isar, Technische Universität MünchenMunich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE)Munich, Germany; Neuroproteomics, Klinikum rechts der Isar, Technische Universität MünchenMunich, Germany; Institute for Advanced Study, Technische Universität MunichGarching, Germany; Munich Cluster for Systems Neurology (SyNergy)Munich, Germany
| |
Collapse
|
265
|
Höfling C, Morawski M, Zeitschel U, Zanier ER, Moschke K, Serdaroglu A, Canneva F, von Hörsten S, De Simoni M, Forloni G, Jäger C, Kremmer E, Roßner S, Lichtenthaler SF, Kuhn P. Differential transgene expression patterns in Alzheimer mouse models revealed by novel human amyloid precursor protein-specific antibodies. Aging Cell 2016; 15:953-63. [PMID: 27470171 PMCID: PMC5013031 DOI: 10.1111/acel.12508] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2016] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is histopathologically characterized by neurodegeneration, the formation of intracellular neurofibrillary tangles and extracellular Aβ deposits that derive from proteolytic processing of the amyloid precursor protein (APP). As rodents do not normally develop Aβ pathology, various transgenic animal models of AD were designed to overexpress human APP with mutations favouring its amyloidogenic processing. However, these mouse models display tremendous differences in the spatial and temporal appearance of Aβ deposits, synaptic dysfunction, neurodegeneration and the manifestation of learning deficits which may be caused by age-related and brain region-specific differences in APP transgene levels. Consequentially, a comparative temporal and regional analysis of the pathological effects of Aβ in mouse brains is difficult complicating the validation of therapeutic AD treatment strategies in different mouse models. To date, no antibodies are available that properly discriminate endogenous rodent and transgenic human APP in brains of APP-transgenic animals. Here, we developed and characterized rat monoclonal antibodies by immunohistochemistry and Western blot that detect human but not murine APP in brains of three APP-transgenic mouse and one APP-transgenic rat model. We observed remarkable differences in expression levels and brain region-specific expression of human APP among the investigated transgenic mouse lines. This may explain the differences between APP-transgenic models mentioned above. Furthermore, we provide compelling evidence that our new antibodies specifically detect endogenous human APP in immunocytochemistry, FACS and immunoprecipitation. Hence, we propose these antibodies as standard tool for monitoring expression of endogenous or transfected APP in human cells and APP expression in transgenic animals.
Collapse
Affiliation(s)
- Corinna Höfling
- Paul Flechsig Institute for Brain ResearchUniversity of LeipzigLeipzigGermany
| | - Markus Morawski
- Paul Flechsig Institute for Brain ResearchUniversity of LeipzigLeipzigGermany
| | - Ulrike Zeitschel
- Paul Flechsig Institute for Brain ResearchUniversity of LeipzigLeipzigGermany
| | - Elisa R. Zanier
- Department of NeuroscienceIRCCSIstituto di Ricerche Farmacologiche Mario NegriMilanoItaly
| | - Katrin Moschke
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| | - Alperen Serdaroglu
- Institute for Advanced StudyTechnische Universität MünchenGarchingGermany
- Institut für Pathologie und Pathologische AnatomieTechnische Universität MünchenMunichGermany
| | - Fabio Canneva
- Department of Experimental TherapyPräklinisches Experimentelles Tierzentrum (PETZ)Universitätsklinikum ErlangenErlangenGermany
| | - Stephan von Hörsten
- Department of Experimental TherapyPräklinisches Experimentelles Tierzentrum (PETZ)Universitätsklinikum ErlangenErlangenGermany
| | - Maria‐Grazia De Simoni
- Department of NeuroscienceIRCCSIstituto di Ricerche Farmacologiche Mario NegriMilanoItaly
| | - Gianluigi Forloni
- Department of NeuroscienceIRCCSIstituto di Ricerche Farmacologiche Mario NegriMilanoItaly
| | - Carsten Jäger
- Paul Flechsig Institute for Brain ResearchUniversity of LeipzigLeipzigGermany
| | - Elisabeth Kremmer
- Helmholtz Zentrum MünchenGerman Research Center for Environmental HealthInstitute of Molecular ImmunologyMunichGermany
| | - Steffen Roßner
- Paul Flechsig Institute for Brain ResearchUniversity of LeipzigLeipzigGermany
| | - Stefan F. Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Institute for Advanced StudyTechnische Universität MünchenGarchingGermany
- Neuroproteomics, Klinikum rechts der IsarTechnische Universität MünchenMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Peer‐Hendrik Kuhn
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Institute for Advanced StudyTechnische Universität MünchenGarchingGermany
- Institut für Pathologie und Pathologische AnatomieTechnische Universität MünchenMunichGermany
- Neuroproteomics, Klinikum rechts der IsarTechnische Universität MünchenMunichGermany
| |
Collapse
|
266
|
Mao Y, Guo Z, Zheng T, Jiang Y, Yan Y, Yin X, Chen Y, Zhang B. Intranasal insulin alleviates cognitive deficits and amyloid pathology in young adult APPswe/PS1dE9 mice. Aging Cell 2016; 15:893-902. [PMID: 27457264 PMCID: PMC5013027 DOI: 10.1111/acel.12498] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2016] [Indexed: 11/26/2022] Open
Abstract
Brain insulin signaling deficits contribute to multiple pathological features of Alzheimer's disease (AD). Although intranasal insulin has shown efficacy in patients with AD, the underlying mechanisms remain largely unillustrated. Here, we demonstrate that intranasal insulin improves cognitive deficits, ameliorates defective brain insulin signaling, and strongly reduces β‐amyloid (Aβ) production and plaque formation after 6 weeks of treatment in 4.5‐month‐old APPswe/PS1dE9 (APP/PS1) mice. Furthermore, c‐Jun N‐terminal kinase activation, which plays a pivotal role in insulin resistance and AD pathologies, is significantly inhibited. The alleviation of amyloid pathology by intranasal insulin results mainly from enhanced nonamyloidogenic processing and compromised amyloidogenic processing of amyloid precursor protein (APP), and from a reduction in apolipoprotein E protein which is involved in Aβ metabolism. In addition, intranasal insulin effectively promotes hippocampal neurogenesis in APP/PS1 mice. This study, exploring the mechanisms underlying the beneficial effects of intranasal insulin on Aβ pathologies in vivo for the first time, highlights important preclinical evidence that intranasal insulin is potentially an effective therapeutic method for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yan‐Fang Mao
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Zhangyu Guo
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Tingting Zheng
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Yasi Jiang
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Yaping Yan
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Xinzhen Yin
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Yanxing Chen
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Baorong Zhang
- Department of Neurology the Second Affiliated Hospital College of Medicine Zhejiang University Hangzhou Zhejiang China
| |
Collapse
|
267
|
Marchesi N, Amadio M, Colombrita C, Govoni S, Ratti A, Pascale A. PKC Activation Counteracts ADAM10 Deficit in HuD-Silenced Neuroblastoma Cells. J Alzheimers Dis 2016; 54:535-47. [DOI: 10.3233/jad-160299] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Nicoletta Marchesi
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Marialaura Amadio
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Claudia Colombrita
- Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, ‘Dino Ferrari’ Center, University of Milan, Milan, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Antonia Ratti
- Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, ‘Dino Ferrari’ Center, University of Milan, Milan, Italy
| | - Alessia Pascale
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| |
Collapse
|
268
|
Shi Y, Zuo D, Wang X, Han M, Wu Y. shRNA‑mediated silencing of TARBP2 inhibits NCI‑H1299 non‑small cell lung cancer cell invasion and migration via the JNK/STAT3/AKT pathway. Mol Med Rep 2016; 14:3725-30. [PMID: 27599909 DOI: 10.3892/mmr.2016.5723] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 03/23/2016] [Indexed: 11/06/2022] Open
Abstract
Metastasis is a major cause of lung cancer-associated mortality. The current study aimed to investigate the effects and mechanisms of TAR (human immunodeficiency virus 1) RNA binding protein 2 (TARBP2) in the invasion and migration of non‑small cell lung cancer in vitro. The highly metastatic cell clone H1299/M02 was obtained by TARBP2 overexpression. Expression of TARBP2 in H1299/M02 was also downregulated to different levels via small hairpin RNAs (shRNAs). Subsequent to TARBP2 silencing, the proliferation of H1299/M02 cells was predominantly unaffected, while invasion and migration were significantly inhibited. A positive correlation was observed between invasion and migration and the level of TARBP2 silencing in vitro. Western blotting and reverse transcription‑quantitative polymerase chain reaction indicated that the protein expression levels of amyloid β (A4) precursor protein (APP) and zinc finger protein 395 (ZNF395) were upregulated, while expression levels of pro‑metastatic proteins including interleukin (IL)‑1β, IL‑8, cyclooxygenase (COX)‑2, matrix metalloproteinase 2 (MMP2) and MMP9 were downregulated. Phosphorylation of c‑Jun N‑terminal kinase (JNK), signal transducer and activator of transcription 3 (STAT3) and protein kinase B (AKT) were also inhibited. Overexpression of TARBP2 was suggested to be involved in the metastasis of H1299/M02 cells. Silencing of TARBP2 was able to upregulate levels of APP and ZNF395, in addition to inhibiting metastasis‑promoting cytokines, the JNK/STAT3/AKT pathway and COX‑2 to attenuate the invasion and migration of cancer cells.
Collapse
Affiliation(s)
- Yue Shi
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Duo Zuo
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Xia Wang
- Department of Gastrointestinal Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer and Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Meng Han
- Department of Clinical Laboratory, The First Center Hospital, Tianjin 300192, P.R. China
| | - Yan Wu
- Department of Clinical Laboratory, TEAD Community Health Service Center, Tianjin 300457, P.R. China
| |
Collapse
|
269
|
Kanatsu K, Tomita T. Membrane trafficking and proteolytic activity of γ-secretase in Alzheimer’s disease. Biol Chem 2016; 397:827-35. [DOI: 10.1515/hsz-2016-0146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/04/2016] [Indexed: 12/26/2022]
Abstract
Abstract
γ-Secretase is an intramembrane-cleaving protease that generates various forms of amyloid-β peptides (Aβ) that accumulate in the brains of Alzheimer’s disease (AD) patients. The intracellular trafficking and subcellular localization of γ-secretase are linked to both qualitative and quantitative changes in Aβ production. However, the precise intracellular localization of γ-secretase as well as its detailed regulatory mechanisms have remained elusive. Recent genetic studies on AD provide ample evidence that alteration of the subcellular localization of γ-secretase contributes to the pathogenesis of AD. Here we review our current understanding of the intracellular membrane trafficking of γ-secretase, the association between its localization and proteolytic activity, and the possibility of γ-secretase as a therapeutic target against AD.
Collapse
|
270
|
Paschkowsky S, Hamzé M, Oestereich F, Munter LM. Alternative Processing of the Amyloid Precursor Protein Family by Rhomboid Protease RHBDL4. J Biol Chem 2016; 291:21903-21912. [PMID: 27563067 DOI: 10.1074/jbc.m116.753582] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Indexed: 12/13/2022] Open
Abstract
The amyloid precursor protein (APP) is an ubiquitously expressed cell surface protein and a key molecule in the etiology of Alzheimer disease. Amyloidogenic processing of APP through secretases leads to the generation of toxic amyloid β (Aβ) peptides, which are regarded as the molecular cause of the disease. We report here an alternative processing pathway of APP through the mammalian intramembrane rhomboid protease RHBDL4. RHBDL4 efficiently cleaves APP inside the cell, thus bypassing APP from amyloidogenic processing, leading to reduced Aβ levels. RHBDL4 cleaves APP multiple times in the ectodomain, resulting in several N- and C-terminal fragments that are not further degraded by classical APP secretases. Knockdown of endogenous RHBDL4 results in decreased levels of C-terminal fragments derived from endogenous APP. Similarly, we found the APP family members APLP1 and APLP2 to be substrates of RHBDL4. We conclude that RHBDL4-mediated APP processing provides insight into APP and rhomboid physiology and qualifies for further investigations to elaborate its impact on Alzheimer disease pathology.
Collapse
Affiliation(s)
| | - Mehdi Hamzé
- From the Department of Pharmacology and Therapeutics and
| | - Felix Oestereich
- From the Department of Pharmacology and Therapeutics and Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3G 0B1, Canada
| | | |
Collapse
|
271
|
Wilkins HM, Swerdlow RH. Amyloid precursor protein processing and bioenergetics. Brain Res Bull 2016; 133:71-79. [PMID: 27545490 DOI: 10.1016/j.brainresbull.2016.08.009] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 02/08/2023]
Abstract
The processing of amyloid precursor protein (APP) to amyloid beta (Aβ) is of great interest to the Alzheimer's disease (AD) field. Decades of research define how APP is altered to form Aβ, and how Aβ generates oligomers, protofibrils, and fibrils. Numerous signaling pathways and changes in cell physiology are known to influence APP processing. Existing data additionally indicate a relationship exists between mitochondria, bioenergetics, and APP processing. Here, we review data that address whether mitochondrial function and bioenergetics modify APP processing and Aβ production.
Collapse
Affiliation(s)
- Heather M Wilkins
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA; University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA; University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA; Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA.
| |
Collapse
|
272
|
Wang XF, Liu DX, Liang Y, Xing LL, Zhao WH, Qin XX, Shang DS, Li B, Fang WG, Cao L, Zhao WD, Chen YH. Cystatin C Shifts APP Processing from Amyloid-β Production towards Non-Amyloidgenic Pathway in Brain Endothelial Cells. PLoS One 2016; 11:e0161093. [PMID: 27532339 PMCID: PMC4988779 DOI: 10.1371/journal.pone.0161093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 07/31/2016] [Indexed: 12/15/2022] Open
Abstract
Amyloid-β (Aβ), the major component of neuritic plaques in Alzheimer's disease (AD), is derived from sequential proteolytic cleavage of amyloid protein precursor (APP) by secretases. In this study, we found that cystatin C (CysC), a natural cysteine protease inhibitor, is able to reduce Aβ40 secretion in human brain microvascular endothelial cells (HBMEC). The CysC-induced Aβ40 reduction was caused by degradation of β-secretase BACE1 through the ubiquitin/proteasome pathway. In contrast, we found that CysC promoted secretion of soluble APPα indicating the activated non-amyloidogenic processing of APP in HBMEC. Further results revealed that α-secretase ADAM10, which was transcriptionally upregulated in response to CysC, was required for the CysC-induced sAPPα secretion. Knockdown of SIRT1 abolished CysC-triggered ADAM10 upregulation and sAPPα production. Taken together, our results demonstrated that exogenously applied CysC can direct amyloidogenic APP processing to non-amyloidgenic pathway in brain endothelial cells, mediated by proteasomal degradation of BACE1 and SIRT1-mediated ADAM10 upregulation. Our study unveils previously unrecognized protective role of CysC in APP processing.
Collapse
Affiliation(s)
- Xia-Fei Wang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Dong-Xin Liu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Yue Liang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Li-Li Xing
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Wen-Hui Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Xiao-Xue Qin
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - De-Shu Shang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Bo Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Wen-Gang Fang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Liu Cao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang 110122, China
| |
Collapse
|
273
|
Shackleton B, Crawford F, Bachmeier C. Inhibition of ADAM10 promotes the clearance of Aβ across the BBB by reducing LRP1 ectodomain shedding. Fluids Barriers CNS 2016; 13:14. [PMID: 27503326 PMCID: PMC4977753 DOI: 10.1186/s12987-016-0038-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/01/2016] [Indexed: 12/22/2022] Open
Abstract
Background Transport across the blood–brain barrier (BBB) is an important mediator of beta-amyloid (Aβ) accumulation in the brain and a contributing factor in the pathogenesis of Alzheimer’s disease (AD). One of the receptors responsible for the transport of Aβ in the BBB is the low density lipoprotein receptor-related protein 1 (LRP1). LRP1 is susceptible to proteolytic shedding at the cell surface, which prevents endocytic transport of ligands. Previously, we reported a strong inverse correlation between LRP1 shedding in the brain and Aβ transit across the BBB. Several proteases contribute to the ectodomain shedding of LRP1 including the α-secretase, a desintegrin and metalloproteinase domain containing protein 10 (ADAM10). Methods The role of ADAM10 in the shedding of LRP1 and Aβ BBB clearance was assessed through pharmacological inhibition of ADAM10 in an in vitro model of the BBB and through the use of ADAM10 endothelial specific knock-out mice. In addition, an acute treatment paradigm with an ADAM10 inhibitor was also tested in an AD mouse model to assess the effect of ADAM10 inhibition on LRP1 shedding and Aβbrain accumulation. Results In the current studies, inhibition of ADAM10 reduced LRP1 shedding in brain endothelial cultures and increased Aβ42 transit across an in vitro model of the BBB. Similarly, transgenic ADAM10 endothelial knockout mice displayed lower LRP1 shedding in the brain and significantly enhanced Aβ clearance across the BBB compared to wild-type animals. Acute treatment with the ADAM10-selective inhibitor GI254023X in an AD mouse model substantially reduced brain LRP1 shedding and increased Aβ40 levels in the plasma, indicating enhanced Aβ transit from the brain to the periphery. Furthermore, both soluble and insoluble Aβ40 and Aβ42 brain levels were decreased following GI254023X treatment, but these effects lacked statistical significance. Conclusions These studies demonstrate a role for ADAM10 in the ectodomain shedding of LRP1 in the brain and the clearance of Aβ across the BBB, which may provide a novel strategy for attenuating Aβ accumulation in the AD brain.
Collapse
Affiliation(s)
- B Shackleton
- The Roskamp Institute, Sarasota, FL, 34243, USA. .,The Open University, Milton Keynes, Buckinghamshire, MK7 6AA, UK.
| | - F Crawford
- The Roskamp Institute, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, Buckinghamshire, MK7 6AA, UK
| | - C Bachmeier
- The Roskamp Institute, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, Buckinghamshire, MK7 6AA, UK
| |
Collapse
|
274
|
Dreymueller D, Ludwig A. Considerations on inhibition approaches for proinflammatory functions of ADAM proteases. Platelets 2016; 28:354-361. [PMID: 27460023 DOI: 10.1080/09537104.2016.1203396] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Proteases of the disintegrin and metalloproteinase (ADAM) family mediate the proteolytic shedding of various surface molecules including cytokine precursors, adhesion molecules, growth factors, and receptors. Within the vasculature ADAM10 and ADAM17 regulate endothelial permeability, transendothelial leukocyte migration, and the adhesion of leukocytes and platelets. In vivo studies show that both proteases are implicated in several inflammatory pathologies, for example, edema formation, leukocyte infiltration, and thrombosis. However, both proteases also contribute to developmental and regenerative processes. Thus, although ADAMs can be regarded as valuable drug targets in many aspects, the danger of severe side effects is clearly visible. To circumvent these side effects, traditional inhibition approaches have to be improved to target ADAMs at the right time in the right place. Moreover, the inhibitors need to be more selective for the target protease and if possible also for the substrate. Antibodies recognizing the active conformation of ADAMs or small molecules blocking exosites of ADAM proteases may represent inhibitors with the desired selectivities.
Collapse
Affiliation(s)
- Daniela Dreymueller
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| | - Andreas Ludwig
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| |
Collapse
|
275
|
Functions of the Alzheimer's Disease Protease BACE1 at the Synapse in the Central Nervous System. J Mol Neurosci 2016; 60:305-315. [PMID: 27456313 PMCID: PMC5059407 DOI: 10.1007/s12031-016-0800-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 07/07/2016] [Indexed: 02/06/2023]
Abstract
Inhibition of the protease β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) is a promising treatment strategy for Alzheimer's disease, and a number of BACE inhibitors are currently progressing through clinical trials. The strategy aims to decrease production of amyloid-β (Aβ) peptide from the amyloid precursor protein (APP), thus reducing or preventing Aβ toxicity. Over the last decade, it has become clear that BACE1 proteolytically cleaves a number of substrates in addition to APP. These substrates are not known to be involved in the pathogenesis of Alzheimer's disease but have other roles in the developing and/or mature central nervous system. Consequently, BACE inhibition and knockout in mice results in synaptic and other neuronal dysfunctions and the key substrates responsible for these deficits are still being elucidated. Of the BACE1 substrates that have been validated to date, a number may contribute to the synaptic deficits seen with BACE blockade, including neuregulin 1, close homologue of L1 and seizure-related gene 6. It is important to understand the impact that BACE blockade may have on these substrates and other proteins detected in substrate screens and, if necessary, develop substrate-selective BACE inhibitors.
Collapse
|
276
|
Alzheimer disease: modeling an Aβ-centered biological network. Mol Psychiatry 2016; 21:861-71. [PMID: 27021818 DOI: 10.1038/mp.2016.38] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 01/15/2023]
Abstract
In genetically complex diseases, the search for missing heritability is focusing on rare variants with large effect. Thanks to next generation sequencing technologies, genome-wide characterization of these variants is now feasible in every individual. However, a lesson from current studies is that collapsing rare variants at the gene level is often insufficient to obtain a statistically significant signal in case-control studies, and that network-based analyses are an attractive complement to classical approaches. In Alzheimer disease (AD), according to the prevalent amyloid cascade hypothesis, the pathology is driven by the amyloid beta (Aβ) peptide. In past years, based on experimental studies, several hundreds of proteins have been shown to interfere with Aβ production, clearance, aggregation or toxicity. Thanks to a manual curation of the literature, we identified 335 genes/proteins involved in this biological network and classified them according to their cellular function. The complete list of genes, or its subcomponents, will be of interest in ongoing AD genetic studies.
Collapse
|
277
|
Welt T, Kulic L, Hoey SE, McAfoose J, Späni C, Chadha AS, Fisher A, Nitsch RM. Acute Effects of Muscarinic M1 Receptor Modulation on AβPP Metabolism and Amyloid-β Levels in vivo: A Microdialysis Study. J Alzheimers Dis 2016; 46:971-82. [PMID: 25881909 DOI: 10.3233/jad-150152] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Indirect modulation of cholinergic activity by cholinesterase inhibition is currently a widely established symptomatic treatment for Alzheimer's disease (AD). Selective activation of certain muscarinic receptor subtypes has emerged as an alternative cholinergic-based amyloid-lowering strategy for AD, as selective muscarinic M1 receptor agonists can reduce amyloid-β (Aβ) production by shifting endoproteolytic amyloid-β protein precursor (AβPP) processing toward non-amyloidogenic pathways. In this study, we addressed the hypothesis that acute stimulation of muscarinic M1 receptors can inhibit Aβ production in awake and freely moving AβPP transgenic mice. By combining intracerebral microdialysis with retrodialysis, we determined hippocampal Aβ concentrations during simultaneous pharmacological modulation of brain M1 receptor function. Infusion with a M1 receptor agonist AF102B resulted in a rapid reduction of interstitial fluid (ISF) Aβ levels while treatment with the M1 antagonist dicyclomine increased ISF Aβ levels reaching significance within 120 minutes of treatment. The reduction in Aβ levels was associated with PKCα and ERK activation resulting in increased levels of the α-secretase ADAM17 and a shift in AβPP processing toward the non-amyloidogenic processing pathway. In contrast, treatment with the M1 receptor antagonist dicyclomine caused a decrease in levels of phosphorylated ERK that was independent of PKCα, and led to an elevation of β-secretase levels associated with increased amyloidogenic AβPP processing. The results of this study demonstrate rapid effects of in vivo M1 receptor modulation on the ISF pool of Aβ and suggest that intracerebral microdialysis with retrodialysis is a useful technical approach for monitoring acute treatment effects of muscarinic receptor modulators on AβPP/Aβ metabolism.
Collapse
Affiliation(s)
- Tobias Welt
- Division of Psychiatry Research, University of Zürich Campus Schlieren, Switzerland
| | - Luka Kulic
- Division of Psychiatry Research, University of Zürich Campus Schlieren, Switzerland.,Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Sarah E Hoey
- Division of Psychiatry Research, University of Zürich Campus Schlieren, Switzerland
| | - Jordan McAfoose
- Division of Psychiatry Research, University of Zürich Campus Schlieren, Switzerland
| | - Claudia Späni
- Division of Psychiatry Research, University of Zürich Campus Schlieren, Switzerland
| | | | - Abraham Fisher
- Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Roger M Nitsch
- Division of Psychiatry Research, University of Zürich Campus Schlieren, Switzerland
| |
Collapse
|
278
|
Fisher JR, Wallace CE, Tripoli DL, Sheline YI, Cirrito JR. Redundant Gs-coupled serotonin receptors regulate amyloid-β metabolism in vivo. Mol Neurodegener 2016; 11:45. [PMID: 27315796 PMCID: PMC4912779 DOI: 10.1186/s13024-016-0112-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 06/14/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The aggregation of amyloid-β (Aβ) into insoluble plaques is a hallmark pathology of Alzheimer's disease (AD). Previous work has shown increasing serotonin levels with selective serotonin re-uptake inhibitor (SSRI) compounds reduces Aβ in the brain interstitial fluid (ISF) in a mouse model of AD and in the cerebrospinal fluid of humans. We investigated which serotonin receptor (5-HTR) subtypes and downstream effectors were responsible for this reduction. RESULTS Agonists of 5-HT4R, 5-HT6R, and 5-HT7R significantly reduced ISF Aβ, but agonists of other receptor subtypes did not. Additionally, inhibition of Protein Kinase A (PKA) blocked the effects of citalopram, an SSRI, on ISF Aβ levels. Serotonin signaling does not appear to change gene expression to reduce Aβ levels in acute timeframes, but likely acts within the cytoplasm to increase α-secretase enzymatic activity. Broad pharmacological inhibition of putative α-secretases increased ISF Aβ and blocked the effects of citalopram. CONCLUSIONS In total, these studies map the major signaling components linking serotonin receptors to suppression of brain ISF Aβ. These results suggest the reduction in ISF Aβ is mediated by a select group of 5-HTRs and open future avenues for targeted therapy of AD.
Collapse
Affiliation(s)
- Jonathan R Fisher
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Clare E Wallace
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Danielle L Tripoli
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Yvette I Sheline
- Departments of Psychiatry, Radiology, and Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - John R Cirrito
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA. .,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA. .,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA. .,Present Address: Washington University, Neurology, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO, 63110, USA.
| |
Collapse
|
279
|
Letronne F, Laumet G, Ayral AM, Chapuis J, Demiautte F, Laga M, Vandenberghe ME, Malmanche N, Leroux F, Eysert F, Sottejeau Y, Chami L, Flaig A, Bauer C, Dourlen P, Lesaffre M, Delay C, Huot L, Dumont J, Werkmeister E, Lafont F, Mendes T, Hansmannel F, Dermaut B, Deprez B, Hérard AS, Dhenain M, Souedet N, Pasquier F, Tulasne D, Berr C, Hauw JJ, Lemoine Y, Amouyel P, Mann D, Déprez R, Checler F, Hot D, Delzescaux T, Gevaert K, Lambert JC. ADAM30 Downregulates APP-Linked Defects Through Cathepsin D Activation in Alzheimer's Disease. EBioMedicine 2016; 9:278-292. [PMID: 27333034 PMCID: PMC4972530 DOI: 10.1016/j.ebiom.2016.06.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 01/12/2023] Open
Abstract
Although several ADAMs (A disintegrin-like and metalloproteases) have been shown to contribute to the amyloid precursor protein (APP) metabolism, the full spectrum of metalloproteases involved in this metabolism remains to be established. Transcriptomic analyses centred on metalloprotease genes unraveled a 50% decrease in ADAM30 expression that inversely correlates with amyloid load in Alzheimer's disease brains. Accordingly, in vitro down- or up-regulation of ADAM30 expression triggered an increase/decrease in Aβ peptides levels whereas expression of a biologically inactive ADAM30 (ADAM30(mut)) did not affect Aβ secretion. Proteomics/cell-based experiments showed that ADAM30-dependent regulation of APP metabolism required both cathepsin D (CTSD) activation and APP sorting to lysosomes. Accordingly, in Alzheimer-like transgenic mice, neuronal ADAM30 over-expression lowered Aβ42 secretion in neuron primary cultures, soluble Aβ42 and amyloid plaque load levels in the brain and concomitantly enhanced CTSD activity and finally rescued long term potentiation alterations. Our data thus indicate that lowering ADAM30 expression may favor Aβ production, thereby contributing to Alzheimer's disease development.
Collapse
Affiliation(s)
- Florent Letronne
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Geoffroy Laumet
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Anne-Marie Ayral
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Julien Chapuis
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Florie Demiautte
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Mathias Laga
- Department of Medical Protein Research, VIB, Ghent, Belgium; Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Michel E Vandenberghe
- CEA, DSV, I2BM, MIRCen, Fontenay aux Roses, France; CNRS, UMR 9199, Fontenay aux Roses, France
| | - Nicolas Malmanche
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Florence Leroux
- Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France; INSERM U1177, Drugs and Molecules for Living Systems, F5900 Lille, France
| | - Fanny Eysert
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Yoann Sottejeau
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Linda Chami
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275 CNRS, Laboratoire d'Excellence Distalz, Nice, France; Université de Nice-Sophia-Antipolis, Valbonne, France
| | - Amandine Flaig
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Charlotte Bauer
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275 CNRS, Laboratoire d'Excellence Distalz, Nice, France; Université de Nice-Sophia-Antipolis, Valbonne, France
| | - Pierre Dourlen
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Marie Lesaffre
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Charlotte Delay
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Ludovic Huot
- Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France; Center for Infection and Immunity of Lille, CNRS UMR 8204, INSERM 1019, Lille, France
| | - Julie Dumont
- Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France; INSERM U1177, Drugs and Molecules for Living Systems, F5900 Lille, France
| | | | | | - Tiago Mendes
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Franck Hansmannel
- INSERM, U954, Vandoeuvre-lès-Nancy, France; Department of Hepato-Gastroenterology, University Hospital of Nancy, Université Henri Poincaré 1, Vandoeuvre-lès-Nancy, France
| | - Bart Dermaut
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France
| | - Benoit Deprez
- Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France; INSERM U1177, Drugs and Molecules for Living Systems, F5900 Lille, France
| | - Anne-Sophie Hérard
- CEA, DSV, I2BM, MIRCen, Fontenay aux Roses, France; CNRS, UMR 9199, Fontenay aux Roses, France
| | - Marc Dhenain
- CEA, DSV, I2BM, MIRCen, Fontenay aux Roses, France; CNRS, UMR 9199, Fontenay aux Roses, France
| | - Nicolas Souedet
- CEA, DSV, I2BM, MIRCen, Fontenay aux Roses, France; CNRS, UMR 9199, Fontenay aux Roses, France
| | - Florence Pasquier
- Univ. Lille, Inserm, U1171, - Degenerative & Vascular Cognitive Disorders, Laboratoire d'Excellence Distalz, F-59000 Lille, France; CHR&U, Lille, France
| | - David Tulasne
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Claudine Berr
- INSERM, U1061, Université de Montpellier I, Hôpital La Colombière, Montpellier, France
| | - Jean-Jacques Hauw
- APHP-Raymond Escourolle Neuropathology Laboratory, la salpétrière Hospital, Paris, France
| | - Yves Lemoine
- Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France; Center for Infection and Immunity of Lille, CNRS UMR 8204, INSERM 1019, Lille, France
| | - Philippe Amouyel
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France; CHR&U, Lille, France
| | - David Mann
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Salford Royal Hospital, Salford, UK
| | - Rebecca Déprez
- Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France; INSERM U1177, Drugs and Molecules for Living Systems, F5900 Lille, France
| | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275 CNRS, Laboratoire d'Excellence Distalz, Nice, France; Université de Nice-Sophia-Antipolis, Valbonne, France
| | - David Hot
- Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France; Center for Infection and Immunity of Lille, CNRS UMR 8204, INSERM 1019, Lille, France
| | - Thierry Delzescaux
- CEA, DSV, I2BM, MIRCen, Fontenay aux Roses, France; CNRS, UMR 9199, Fontenay aux Roses, France
| | - Kris Gevaert
- Department of Medical Protein Research, VIB, Ghent, Belgium; Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Jean-Charles Lambert
- INSERM, U1167, Laboratoire d'Excellence Distalz, F59000 Lille, France; Institut Pasteur de Lille, F59000 Lille, France; Univ. Lille, F59000 Lille, France.
| |
Collapse
|
280
|
Yan R, Fan Q, Zhou J, Vassar R. Inhibiting BACE1 to reverse synaptic dysfunctions in Alzheimer's disease. Neurosci Biobehav Rev 2016; 65:326-40. [PMID: 27044452 PMCID: PMC4856578 DOI: 10.1016/j.neubiorev.2016.03.025] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 12/21/2022]
Abstract
Over the past two decades, many studies have identified significant contributions of toxic β-amyloid peptides (Aβ) to the etiology of Alzheimer's disease (AD), which is the most common age-dependent neurodegenerative disease. AD is also recognized as a disease of synaptic failure. Aβ, generated by sequential proteolytic cleavages of amyloid precursor protein (APP) by BACE1 and γ-secretase, is one of major culprits that cause this failure. In this review, we summarize current findings on how BACE1-cleaved APP products impact learning and memory through proteins localized on glutamatergic, GABAergic, and dopaminergic synapses. Considering the broad effects of Aβ on all three types of synapses, BACE1 inhibition emerges as a practical approach for ameliorating Aβ-mediated synaptic dysfunctions. Since BACE1 inhibitory drugs are currently in clinical trials, this review also discusses potential complications arising from BACE1 inhibition. We emphasize that the benefits of BACE1 inhibitory drugs will outweigh the concerns.
Collapse
Affiliation(s)
- Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - John Zhou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Robert Vassar
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
281
|
Glatzel M, Linsenmeier L, Dohler F, Krasemann S, Puig B, Altmeppen HC. Shedding light on prion disease. Prion 2016; 9:244-56. [PMID: 26186508 DOI: 10.1080/19336896.2015.1065371] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Proteolytic processing regulates key processes in health and disease. The cellular prion protein (PrP(C)) is subject to at least 3 cleavage events, α-cleavage, β-cleavage and shedding. In contrast to α- and β-cleavage where there is an ongoing controversy on the identity of relevant proteases, the metalloprotease ADAM10 represents the only relevant PrP sheddase. Here we focus on the roles that ADAM10-mediated shedding of PrP(C) and its pathogenic isoform (PrP(Sc)) might play in regulating their physiological and pathogenic functions, respectively. As revealed by our recent study using conditional ADAM10 knockout mice (Altmeppen et al., 2015), shedding of PrP seems to be involved in key processes of prion diseases. These aspects and several open questions arising from them are discussed. Increased knowledge on this topic can shed new light on prion diseases and other neurodegenerative conditions as well.
Collapse
Affiliation(s)
- Markus Glatzel
- a Institute of Neuropathology; University Medical Center Hamburg-Eppendorf ; Hamburg , Germany
| | | | | | | | | | | |
Collapse
|
282
|
Kretner B, Trambauer J, Fukumori A, Mielke J, Kuhn PH, Kremmer E, Giese A, Lichtenthaler SF, Haass C, Arzberger T, Steiner H. Generation and deposition of Aβ43 by the virtually inactive presenilin-1 L435F mutant contradicts the presenilin loss-of-function hypothesis of Alzheimer's disease. EMBO Mol Med 2016; 8:458-65. [PMID: 26988102 PMCID: PMC5119496 DOI: 10.15252/emmm.201505952] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
As stated by the prevailing amyloid cascade hypothesis, Alzheimer's disease (AD) is caused by the aggregation and cerebral deposition of long amyloid‐β peptide (Aβ) species, which are released from a C‐terminal amyloid precursor protein fragment by γ‐secretase. Mutations in its catalytic subunit presenilin‐1 (PS1) increase the Aβ42 to Aβ40 ratio and are the major cause of familial AD (FAD). An opposing hypothesis states that loss of essential presenilin functions underlies the disease. A major argument for this hypothesis is the observation that the nearly inactive PS1 L435F mutant, paradoxically, causes FAD. We now show that the very little Aβ generated by PS1 L435F consists primarily of Aβ43, a highly amyloidogenic species which was overlooked in previous studies of this mutant. We further demonstrate that the generation of Aβ43 is not due to a trans‐dominant effect of this mutant on WT presenilin. Furthermore, we found Aβ43‐containing plaques in brains of patients with this mutation. The aberrant generation of Aβ43 by this particular mutant provides a direct objection against the presenilin hypothesis.
Collapse
Affiliation(s)
- Benedikt Kretner
- Biomedical Center, Metabolic Biochemistry, Ludwig-Maximilians-University Munich, Munich, Germany DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Johannes Trambauer
- Biomedical Center, Metabolic Biochemistry, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Akio Fukumori
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Janina Mielke
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peer-Hendrik Kuhn
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany Neuroproteomics, Klinikum rechts der Isar and Institute for Advanced Study, Technische Universität München, Munich, Germany
| | - Elisabeth Kremmer
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Armin Giese
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany Neuroproteomics, Klinikum rechts der Isar and Institute for Advanced Study, Technische Universität München, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Haass
- Biomedical Center, Metabolic Biochemistry, Ludwig-Maximilians-University Munich, Munich, Germany DZNE - German Center for Neurodegenerative Diseases, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Arzberger
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Harald Steiner
- Biomedical Center, Metabolic Biochemistry, Ludwig-Maximilians-University Munich, Munich, Germany DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| |
Collapse
|
283
|
Qian M, Shen X, Wang H. The Distinct Role of ADAM17 in APP Proteolysis and Microglial Activation Related to Alzheimer's Disease. Cell Mol Neurobiol 2016; 36:471-82. [PMID: 26119306 DOI: 10.1007/s10571-015-0232-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/23/2015] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with the symptom of cognitive impairment. The deposition of amyloid β (Aβ) peptide is believed to be the primary cause to neuronal dystrophy and eventually dementia. Aβ is the proteolytic product from its precursor amyloid precursor protein (APP) by β- and γ- secretase. An optional cleavage by α-secretase happens inside the Aβ domain. ADAM17 is supposed to be the regulated α-secretase of APP. Enhanced activity of ADAM17 leads to the increasing secretion of neuroprotective soluble APP α fragment and reduction of Aβ generation, which may be benefit to the disease. ADAM17 is then considered the potential therapeutic target for AD. Microglia activation and neuroinflammation is another important event in AD pathogenesis. Interestingly, ADAM17 also participates in the cleavage of many other membrane-bound proteins, especially some inflammatory factors related to microglia activation. The facilitating role of ADAM17 in inflammation and further neuronal damage has also been illustrated. In results, the activation of ADAM17 as the solution to AD may be a tricky task. The comprehensive consideration and evaluation has to be carried out carefully before the final treatment. In the present review, the distinct role of ADAM17 in AD-related APP shedding and neuroinflammatory microglial activation will be carefully discussed.
Collapse
Affiliation(s)
- Meng Qian
- Key Lab of Inflammation and Immunoregulation, School of Medicine, Hangzhou Normal University, Xuelin Street 16, Hangzhou, 310036, China
| | - Xiaoqiang Shen
- Key Lab of Inflammation and Immunoregulation, School of Medicine, Hangzhou Normal University, Xuelin Street 16, Hangzhou, 310036, China
| | - Huanhuan Wang
- Key Lab of Inflammation and Immunoregulation, School of Medicine, Hangzhou Normal University, Xuelin Street 16, Hangzhou, 310036, China.
| |
Collapse
|
284
|
Mohamed T, Shakeri A, Rao PP. Amyloid cascade in Alzheimer's disease: Recent advances in medicinal chemistry. Eur J Med Chem 2016; 113:258-72. [DOI: 10.1016/j.ejmech.2016.02.049] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 12/21/2022]
|
285
|
Mullooly M, McGowan PM, Crown J, Duffy MJ. The ADAMs family of proteases as targets for the treatment of cancer. Cancer Biol Ther 2016; 17:870-80. [PMID: 27115328 DOI: 10.1080/15384047.2016.1177684] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The ADAMs (a disintegrin and metalloproteases) are transmembrane multidomain proteins implicated in multiple biological processes including proteolysis, cell adhesion, cell fusion, cell proliferation and cell migration. Of these varied activities, the best studied is their role in proteolysis. However, of the 22 ADAMs believed to be functional in humans, only approximately a half possess matrix metalloproteinase (MMP)-like protease activity. In contrast to MMPs which are mostly implicated in the degradation of extracellular matrix proteins, the main ADAM substrates are the ectodomains of type I and type II transmembrane proteins. These include growth factor/cytokine precursors, growth factor/cytokine receptors and adhesion proteins. Recently, several different ADAMs, especially ADAM17, have been shown to play a role in the development and progression of multiple cancer types. Consistent with this role in cancer, targeting ADAM17 with either low molecular weight inhibitors or monoclonal antibodies was shown to have anti-cancer activity in multiple preclinical systems. Although early phase clinical trials have shown no serious side effects with a dual ADAM10/17 low molecular weight inhibitor, the consequences of long-term treatment with these agents is unknown. Furthermore, efficacy in clinical trials remains to be shown.
Collapse
Affiliation(s)
- Maeve Mullooly
- a National Institutes of Health , Bethesda , MD , USA.,b UCD School of Medicine and Medical Science , Conway Institute of Biomolecular and Biomedical Research, University College Dublin , Ireland
| | - Patricia M McGowan
- b UCD School of Medicine and Medical Science , Conway Institute of Biomolecular and Biomedical Research, University College Dublin , Ireland.,c Education and Research Center , St. Vincent's University Hospital , Dublin , Ireland
| | - John Crown
- d Department of Medical Oncology , St. Vincent's University Hospital , Dublin , Ireland
| | - Michael J Duffy
- b UCD School of Medicine and Medical Science , Conway Institute of Biomolecular and Biomedical Research, University College Dublin , Ireland.,e UCD Clinical Research Center , St. Vincent's University Hospital , Dublin , Ireland
| |
Collapse
|
286
|
Vincent B. Regulation of the α-secretase ADAM10 at transcriptional, translational and post-translational levels. Brain Res Bull 2016; 126:154-169. [PMID: 27060611 DOI: 10.1016/j.brainresbull.2016.03.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/25/2016] [Accepted: 03/30/2016] [Indexed: 12/19/2022]
Abstract
A tremendous gain of interest in the biology of ADAM10 emerged during the past 15 years when it has first been shown that this protease was able to target the α-site of the β-amyloid precursor protein (βAPP) and later confirmed as the main physiological α-secretase activity. However, beside its well-established implication in the so-called non-amyloidogenic processing of βAPP and its probable protective role against Alzheimer's disease (AD), this metalloprotease also cleaves many other substrates, thereby being implicated in various physiological as well as pathological processes such as cancer and inflammation. Thus, in view of possible effective therapeutic interventions, a full comprehension of how ADAM10 is up and down regulated is required. This review discusses our current knowledge concerning the implication of this enzyme in AD as well as its more recently established roles in other brain disorders and provides a detailed up-date on its various transcriptional, translational and post-translational modulations.
Collapse
Affiliation(s)
- Bruno Vincent
- Mahidol University, Institute of Molecular Biosciences, Nakhon Pathom 73170, Thailand; Centre National de la Recherche Scientifique, 2 rue Michel Ange, 75016 Paris, France.
| |
Collapse
|
287
|
Neural stem/progenitor cells in Alzheimer's disease. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2016; 89:23-35. [PMID: 27505014 PMCID: PMC4797833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and a worldwide health challenge. Different therapeutic approaches are being developed to reverse or slow the loss of affected neurons. Another plausible therapeutic way that may complement the studies is to increase the survival of existing neurons by mobilizing the existing neural stem/progenitor cells (NSPCs) - i.e. "induce their plasticity" - to regenerate lost neurons despite the existing pathology and unfavorable environment. However, there is controversy about how NSPCs are affected by the unfavorable toxic environment during AD. In this review, we will discuss the use of stem cells in neurodegenerative diseases and in particular how NSPCs affect the AD pathology and how neurodegeneration affects NSPCs. In the end of this review, we will discuss how zebrafish as a useful model organism with extensive regenerative ability in the brain might help to address the molecular programs needed for NSPCs to respond to neurodegeneration by enhanced neurogenesis.
Collapse
|
288
|
Yoon SY, Kim DH. Alzheimer's disease genes and autophagy. Brain Res 2016; 1649:201-209. [PMID: 27016058 DOI: 10.1016/j.brainres.2016.03.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 03/09/2016] [Accepted: 03/13/2016] [Indexed: 11/15/2022]
Abstract
Autophagy is a process to degrade and recycle cellular constituents via the lysosome for regulating cellular homeostasis. Its dysfunction is now considered to be involved in many diseases, including neurodegenerative diseases. Many features reflecting autophagy impairment, such as autophagosome accumulation and lysosomal dysfunction, have been also revealed to be involved in Alzheimer's disease (AD). Recent genetic studies such as genome-wide association studies in AD have identified a number of novel genes associated with AD. Some of the identified genes have demonstrated dysfunction in autophagic processes in AD, while others remain under investigation. Since autophagy is strongly regarded to be one of the major pathogenic mechanisms of AD, it is necessary to review how the AD-associated genes are related to autophagy. We anticipate our current review to be a starting point for future studies regarding AD-associated genes and autophagy. This article is part of a Special Issue entitled SI:Autophagy.
Collapse
Affiliation(s)
- Seung-Yong Yoon
- Alzheimer's Disease Experts Lab (ADEL), Asan Institute of Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Brain Science, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Republic of Korea; Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Dong-Hou Kim
- Alzheimer's Disease Experts Lab (ADEL), Asan Institute of Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Brain Science, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Republic of Korea; Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
289
|
Plummer S, Van den Heuvel C, Thornton E, Corrigan F, Cappai R. The Neuroprotective Properties of the Amyloid Precursor Protein Following Traumatic Brain Injury. Aging Dis 2016; 7:163-79. [PMID: 27114849 PMCID: PMC4809608 DOI: 10.14336/ad.2015.0907] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 09/07/2015] [Indexed: 01/16/2023] Open
Abstract
Despite the significant health and economic burden that traumatic brain injury (TBI) places on society, the development of successful therapeutic agents have to date not translated into efficacious therapies in human clinical trials. Injury to the brain is ongoing after TBI, through a complex cascade of primary and secondary injury events, providing a valuable window of opportunity to help limit and prevent some of the severe consequences with a timely treatment. Of note, it has been suggested that novel treatments for TBI should be multifactorial in nature, mimicking the body's own endogenous repair response. Whilst research has historically focused on the role of the amyloid precursor protein (APP) in the pathogenesis of Alzheimer's disease, recent advances in trauma research have demonstrated that APP offers considerable neuroprotective properties following TBI, suggesting that APP is an ideal therapeutic candidate. Its acute upregulation following TBI has been shown to serve a beneficial role following trauma and has lead to significant advances in understanding the neuroprotective and neurotrophic functions of APP and its metabolites. Research has focused predominantly on the APP derivative sAPPα, which has consistently demonstrated neuroprotective and neurotrophic functions both in vitro and in vivo following various traumatic insults. Its neuroprotective activity has been narrowed down to a 15 amino acid sequence, and this region is linked to both heparan binding and growth-factor-like properties. It has been proposed that APP binds to heparan sulfate proteoglycans to exert its neuroprotective action. APP presents us with a novel therapeutic compound that could overcome many of the challenges that have stalled development of efficacious TBI treatments previously.
Collapse
Affiliation(s)
- Stephanie Plummer
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Corinna Van den Heuvel
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Emma Thornton
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Frances Corrigan
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Roberto Cappai
- Department of Pathology, the University of Melbourne, Victoria, Australia
| |
Collapse
|
290
|
Schönherr C, Bien J, Isbert S, Wichert R, Prox J, Altmeppen H, Kumar S, Walter J, Lichtenthaler SF, Weggen S, Glatzel M, Becker-Pauly C, Pietrzik CU. Generation of aggregation prone N-terminally truncated amyloid β peptides by meprin β depends on the sequence specificity at the cleavage site. Mol Neurodegener 2016; 11:19. [PMID: 26895626 PMCID: PMC4759862 DOI: 10.1186/s13024-016-0084-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 02/08/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The metalloprotease meprin β cleaves the Alzheimer's Disease (AD) relevant amyloid precursor protein (APP) as a β-secretase reminiscent of BACE-1, however, predominantly generating N-terminally truncated Aβ2-x variants. RESULTS Herein, we observed increased endogenous sAPPα levels in the brains of meprin β knock-out (ko) mice compared to wild-type controls. We further analyzed the cellular interaction of APP and meprin β and found that cleavage of APP by meprin β occurs prior to endocytosis. The N-terminally truncated Aβ2-40 variant shows increased aggregation propensity compared to Aβ1-40 and acts even as a seed for Aβ1-40 aggregation. Additionally, we observed that different APP mutants affect the catalytic properties of meprin β and that, interestingly, meprin β is unable to generate N-terminally truncated Aβ peptides from Swedish mutant APP (APPswe). CONCLUSION Concluding, we propose that meprin β may be involved in the generation of N-terminally truncated Aβ2-x peptides of APP, but acts independently from BACE-1.
Collapse
Affiliation(s)
- Caroline Schönherr
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Duesbergweg 6, 55128, Mainz, Germany
| | - Jessica Bien
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Duesbergweg 6, 55128, Mainz, Germany
| | - Simone Isbert
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Duesbergweg 6, 55128, Mainz, Germany
| | - Rielana Wichert
- Institute of Biochemistry, Unit for Degradomics of the Protease Web, Christian-Albrechts-University, Otto-Hahn-Platz 9, 24118, Kiel, Germany
| | - Johannes Prox
- Institute of Biochemistry, Unit for Degradomics of the Protease Web, Christian-Albrechts-University, Otto-Hahn-Platz 9, 24118, Kiel, Germany
| | - Hermann Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Sathish Kumar
- Department of Neurology, Molecular Cell Biology, University of Bonn, 53127, Bonn, Germany
| | - Jochen Walter
- Department of Neurology, Molecular Cell Biology, University of Bonn, 53127, Bonn, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, 81675, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sascha Weggen
- Department of Neuropathology, Heinrich Heine University, 40225, Duesseldorf, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Christoph Becker-Pauly
- Institute of Biochemistry, Unit for Degradomics of the Protease Web, Christian-Albrechts-University, Otto-Hahn-Platz 9, 24118, Kiel, Germany.
| | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University of Mainz, Duesbergweg 6, 55128, Mainz, Germany.
| |
Collapse
|
291
|
Cervera-Carles L, Clarimón J. Genetic and Epigenetic Architecture of Alzheimer’s Dementia. CURRENT GENETIC MEDICINE REPORTS 2016. [DOI: 10.1007/s40142-016-0086-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
292
|
Kuhn PH, Colombo AV, Schusser B, Dreymueller D, Wetzel S, Schepers U, Herber J, Ludwig A, Kremmer E, Montag D, Müller U, Schweizer M, Saftig P, Bräse S, Lichtenthaler SF. Systematic substrate identification indicates a central role for the metalloprotease ADAM10 in axon targeting and synapse function. eLife 2016; 5. [PMID: 26802628 PMCID: PMC4786429 DOI: 10.7554/elife.12748] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/22/2016] [Indexed: 12/11/2022] Open
Abstract
Metzincin metalloproteases have major roles in intercellular communication by modulating the function of membrane proteins. One of the proteases is the a-disintegrin-and-metalloprotease 10 (ADAM10) which acts as alpha-secretase of the Alzheimer's disease amyloid precursor protein. ADAM10 is also required for neuronal network functions in murine brain, but neuronal ADAM10 substrates are only partly known. With a proteomic analysis of Adam10-deficient neurons we identified 91, mostly novel ADAM10 substrate candidates, making ADAM10 a major protease for membrane proteins in the nervous system. Several novel substrates, including the neuronal cell adhesion protein NrCAM, are involved in brain development. Indeed, we detected mistargeted axons in the olfactory bulb of conditional ADAM10-/- mice, which correlate with reduced cleavage of NrCAM, NCAM and other ADAM10 substrates. In summary, the novel ADAM10 substrates provide a molecular basis for neuronal network dysfunctions in conditional ADAM10-/- mice and demonstrate a fundamental function of ADAM10 in the brain.
Collapse
Affiliation(s)
- Peer-Hendrik Kuhn
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Institut für Pathologie und Pathologische Anatomie, Technische Universität München, Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany
| | - Alessio Vittorio Colombo
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany
| | - Benjamin Schusser
- Department of Animal Science, Institute for Animal Physiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniela Dreymueller
- Institute of Pharmacology and Toxicology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Sebastian Wetzel
- Institute of Biochemistry, Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Ute Schepers
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Julia Herber
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Elisabeth Kremmer
- German Research Center for Environmental Health, Institute of Molecular Tumor immunology, Helmholtz Zentrum München, Munich, Germany
| | - Dirk Montag
- Neurogenetics, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Ulrike Müller
- Department of Functional Genomics, Institute for Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Michaela Schweizer
- Service-Gruppe für Elektronenmikroskopie, Zentrum für Molekulare Neurobiologie, Hamburg, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Stefan Bräse
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Stefan F Lichtenthaler
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
293
|
Leriche G, Chen AC, Kim S, Selkoe DJ, Yang J. Fluorescent Analogue of Batimastat Enables Imaging of α-Secretase in Living Cells. ACS Chem Neurosci 2016; 7:40-5. [PMID: 26559179 DOI: 10.1021/acschemneuro.5b00283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The ADAM family of metalloproteases cleave a diverse range of transmembrane substrates, resulting in the release of their soluble ectodomains. This process of protein shedding, termed α-secretase processing, is involved in many facets of both normal and disease related cellular function. While the processing of substrates has been well documented, the regulation and trafficking of the ADAMs are less well understood. Tools that allow for the study of ADAMs under their native environment will allow for a better understanding of their regulation and activity. Here we describe the design and evaluation of a novel fluorescent analogue of a well-characterized ADAM inhibitor, Batimastat. This probe exhibited similar activity for inhibiting α-secretase processing in cells as did Batimastat. Importantly, this probe specifically labeled ADAMs fluorescently in both fixed and living cells, enabling the possibility to study the trafficking of α-secretase proteins in a dynamic environment.
Collapse
Affiliation(s)
- Geoffray Leriche
- Department
of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093-0358, United States
| | - Allen C. Chen
- Ann
Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sumin Kim
- Ann
Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Dennis J. Selkoe
- Ann
Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jerry Yang
- Department
of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093-0358, United States
| |
Collapse
|
294
|
Baranger K, Marchalant Y, Bonnet AE, Crouzin N, Carrete A, Paumier JM, Py NA, Bernard A, Bauer C, Charrat E, Moschke K, Seiki M, Vignes M, Lichtenthaler SF, Checler F, Khrestchatisky M, Rivera S. MT5-MMP is a new pro-amyloidogenic proteinase that promotes amyloid pathology and cognitive decline in a transgenic mouse model of Alzheimer's disease. Cell Mol Life Sci 2016; 73:217-36. [PMID: 26202697 PMCID: PMC4700096 DOI: 10.1007/s00018-015-1992-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/08/2015] [Accepted: 07/10/2015] [Indexed: 01/22/2023]
Abstract
Membrane-type 5-matrix metalloproteinase (MT5-MMP) is a proteinase mainly expressed in the nervous system with emerging roles in brain pathophysiology. The implication of MT5-MMP in Alzheimer's disease (AD), notably its interplay with the amyloidogenic process, remains elusive. Accordingly, we crossed the genetically engineered 5xFAD mouse model of AD with MT5-MMP-deficient mice and examined the impact of MT5-MMP deficiency in bigenic 5xFAD/MT5-MMP(-/-) mice. At early stages (4 months) of the pathology, the levels of amyloid beta peptide (Aβ) and its amyloid precursor protein (APP) C-terminal fragment C99 were largely reduced in the cortex and hippocampus of 5xFAD/MT5-MMP(-/-), compared to 5xFAD mice. Reduced amyloidosis in bigenic mice was concomitant with decreased glial reactivity and interleukin-1β (IL-1β) levels, and the preservation of long-term potentiation (LTP) and spatial learning, without changes in the activity of α-, β- and γ-secretases. The positive impact of MT5-MMP deficiency was still noticeable at 16 months of age, as illustrated by reduced amyloid burden and gliosis, and a better preservation of the cortical neuronal network and synaptophysin levels in bigenic mice. MT5-MMP expressed in HEKswe cells colocalized and co-immunoprecipitated with APP and significantly increased the levels of Aβ and C99. MT5-MMP also promoted the release of a soluble APP fragment of 95 kDa (sAPP95) in HEKswe cells. sAPP95 levels were significantly reduced in brain homogenates of 5xFAD/MT5-MMP(-/-) mice, supporting altogether the idea that MT5-MMP influences APP processing. MT5-MMP emerges as a new pro-amyloidogenic regulator of APP metabolism, whose deficiency alleviates amyloid pathology, neuroinflammation and cognitive decline.
Collapse
Affiliation(s)
- Kévin Baranger
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Yannick Marchalant
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
- Psychology Department, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Amandine E Bonnet
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Nadine Crouzin
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Alex Carrete
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | | | - Nathalie A Py
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Anne Bernard
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Charlotte Bauer
- Labex DistAlz, IPMC UMR 7275 CNRS-UNS, 06560, Valbonne, France
| | - Eliane Charrat
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Katrin Moschke
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, Munich, Germany
- Klinikum rechts der Isar, and Institute for Advanced Study, Technische Universität München (TUM), 81675, Munich, Germany
| | - Mothoharu Seiki
- Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Michel Vignes
- UMR5247 IBMM CNRS University of Montpellier 1 and University of Montpellier 2, 34095, Montepellier, France
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, Munich, Germany
- Klinikum rechts der Isar, and Institute for Advanced Study, Technische Universität München (TUM), 81675, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
| | | | | | - Santiago Rivera
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France.
| |
Collapse
|
295
|
Sjödin S, Andersson KKA, Mercken M, Zetterberg H, Borghys H, Blennow K, Portelius E. APLP1 as a cerebrospinal fluid biomarker for γ-secretase modulator treatment. ALZHEIMERS RESEARCH & THERAPY 2015; 7:77. [PMID: 26689589 PMCID: PMC4687145 DOI: 10.1186/s13195-015-0160-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/28/2015] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Alzheimer's disease brains are characterized by extracellular plaques containing the aggregated amyloid β42 (Aβ42) peptide and intraneuronal tangles containing hyperphosphorylated tau. Aβ42 is produced by sequential processing of the amyloid precursor protein (APP) by β-secretase followed by γ-secretase. Substantial efforts have been put into developing pharmaceuticals preventing the production or increasing the clearance of Aβ42. However, treatments inhibiting γ-secretase have proven disappointing due to off-target effects. To circumvent these effects, γ-secretase modulators (GSMs) have been developed, which rather than inhibiting γ-secretase shift its preference into producing less aggregation-prone shorter Aβ peptides. Belonging to the same family of proteins as APP, amyloid-like protein 1 (APLP1) is also a substrate for γ-secretase. Herein we investigated whether the GSM E2012 affects APLP1 processing in the central nervous system by measuring APLP1 peptide levels in cerebrospinal fluid (CSF) before and after E2012 treatment in dogs. METHODS An in-house monoclonal APLP1 antibody, AP1, was produced and utilized for immunopurification of APLP1 from human and dog CSF in a hybrid immuno-affinity mass spectrometric method. Seven dogs received a single dose of 20 or 80 mg/kg of E2012 in a randomized cross-over design and CSF was collected prior to and 4, 8 and 24 hours after dosing. RESULTS We have identified 14 CSF APLP1 peptides in humans and 12 CSF APLP1 peptides in dogs. Of these, seven were reproducibly detectable in dogs who received E2012. We found a dose-dependent relative increase of the CSF peptides APLP1β17, 1β18 and 1β28 accompanied with a decrease of 1β25 and 1β27 in response to E2012 treatment. All peptides reverted to baseline over the time of sample collection. CONCLUSION We show an in vivo effect of the GSM E2012 on the processing of APLP1 which is measurable in CSF. These data suggest that APLP1 peptides may be used as biomarkers to monitor drug effects of GSMs on γ-secretase processing in clinical trials. However, this requires further investigation in larger cohorts, including studies in man.
Collapse
Affiliation(s)
- Simon Sjödin
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden.
| | - Kerstin K A Andersson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden.
| | - Marc Mercken
- Janssen Research and Development, Beerse, Belgium.
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden. .,UCL Institute of Neurology, University College London, London, UK.
| | | | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden.
| | - Erik Portelius
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden.
| |
Collapse
|
296
|
Chen AC, Kim S, Shepardson N, Patel S, Hong S, Selkoe DJ. Physical and functional interaction between the α- and γ-secretases: A new model of regulated intramembrane proteolysis. J Cell Biol 2015; 211:1157-76. [PMID: 26694839 PMCID: PMC4687875 DOI: 10.1083/jcb.201502001] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 11/11/2015] [Indexed: 12/20/2022] Open
Abstract
Many single-transmembrane proteins are sequentially cleaved by ectodomain-shedding α-secretases and the γ-secretase complex, a process called regulated intramembrane proteolysis (RIP). These cleavages are thought to be spatially and temporally separate. In contrast, we provide evidence for a hitherto unrecognized multiprotease complex containing both α- and γ-secretase. ADAM10 (A10), the principal neuronal α-secretase, interacted and cofractionated with γ-secretase endogenously in cells and mouse brain. A10 immunoprecipitation yielded γ-secretase proteolytic activity and vice versa. In agreement, superresolution microscopy showed that portions of A10 and γ-secretase colocalize. Moreover, multiple γ-secretase inhibitors significantly increased α-secretase processing (r = -0.86) and decreased β-secretase processing of β-amyloid precursor protein. Select members of the tetraspanin web were important both in the association between A10 and γ-secretase and the γ → α feedback mechanism. Portions of endogenous BACE1 coimmunoprecipitated with γ-secretase but not A10, suggesting that β- and α-secretases can form distinct complexes with γ-secretase. Thus, cells possess large multiprotease complexes capable of sequentially and efficiently processing transmembrane substrates through a spatially coordinated RIP mechanism.
Collapse
Affiliation(s)
- Allen C Chen
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Sumin Kim
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Nina Shepardson
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Sarvagna Patel
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Soyon Hong
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
297
|
Fedele E, Rivera D, Marengo B, Pronzato MA, Ricciarelli R. Amyloid β: Walking on the dark side of the moon. Mech Ageing Dev 2015; 152:1-4. [DOI: 10.1016/j.mad.2015.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/31/2015] [Accepted: 09/03/2015] [Indexed: 12/20/2022]
|
298
|
Montoliu-Gaya L, Villegas S. Protein structures in Alzheimer's disease: The basis for rationale therapeutic design. Arch Biochem Biophys 2015; 588:1-14. [DOI: 10.1016/j.abb.2015.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/24/2015] [Accepted: 10/09/2015] [Indexed: 01/06/2023]
|
299
|
Xie H, Xiao Z, Huang J. C6 Glioma-Secreted NGF and FGF2 Regulate Neuronal APP Processing Through Up-Regulation of ADAM10 and Down-Regulation of BACE1, Respectively. J Mol Neurosci 2015; 59:334-42. [PMID: 26614345 DOI: 10.1007/s12031-015-0690-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/19/2015] [Indexed: 01/05/2023]
Abstract
Excessive accumulation of amyloid-β (Aβ) caused by cleavage of amyloid precursor protein (APP) is thought to be the primary cause of Alzheimer's disease (AD). Two key enzymes ADAM10 and BACE1 are involved in the initial cleavage of APP, resulting in the onset of two pathways, the amyloidogenic pathway and the non-amyloidogenic pathway, respectively. Altering APP metabolism towards the non-amyloidogenic pathway is thought to reduce Aβ production. It has been reported that, in vivo, exogenous neurotrophic factors make APP apt to entering the non-amyloidogenic pathway. Since astrocytes secrete a battery of neurotrophic factors, we investigated the role of astrocyte-derived factors in the dynamics of Aβ generation in neural cells. Results show that C6 glioma cell-conditioned medium (GCM), obtained from cultured astrocyte-derived C6 glioma cells, inhibit Aβ1-42 production and shift APP processing towards the non-amyloidogenic pathway in APPswe-HEK293 cells. Such effect is attributed to two key APP cleavage enzymes, ADAM10 and BACE1. Two neurotrophic factors in the GCM, nerve growth factor and fibroblast growth factor 2, are responsible for the up-regulation of ADAM10 and down-regulation of BACE1, respectively. Our findings enhance our understanding of the relationship between astrocytes and Aβ generation, indicating that stimulation of astrocytic neurotrophic factors could slow AD progression.
Collapse
Affiliation(s)
- Huiping Xie
- College of Life Sciences, Wuhan University, Room 5105, Wuhan, Hubei, 430072, People's Republic of China
| | - Zhimin Xiao
- College of Life Sciences, Wuhan University, Room 5105, Wuhan, Hubei, 430072, People's Republic of China.,Sanofi, Cambridge, MA, 02142, USA
| | - Jian Huang
- College of Life Sciences, Wuhan University, Room 5105, Wuhan, Hubei, 430072, People's Republic of China.
| |
Collapse
|
300
|
Delta-secretase cleaves amyloid precursor protein and regulates the pathogenesis in Alzheimer's disease. Nat Commun 2015; 6:8762. [PMID: 26549211 PMCID: PMC4659940 DOI: 10.1038/ncomms9762] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 09/25/2015] [Indexed: 01/15/2023] Open
Abstract
The age-dependent deposition of amyloid-β peptides, derived from amyloid precursor protein (APP), is a neuropathological hallmark of Alzheimer's disease (AD). Despite age being the greatest risk factor for AD, the molecular mechanisms linking ageing to APP processing are unknown. Here we show that asparagine endopeptidase (AEP), a pH-controlled cysteine proteinase, is activated during ageing and mediates APP proteolytic processing. AEP cleaves APP at N373 and N585 residues, selectively influencing the amyloidogenic fragmentation of APP. AEP is activated in normal mice in an age-dependent manner, and is strongly activated in 5XFAD transgenic mouse model and human AD brains. Deletion of AEP from 5XFAD or APP/PS1 mice decreases senile plaque formation, ameliorates synapse loss, elevates long-term potentiation and protects memory. Blockade of APP cleavage by AEP in mice alleviates pathological and behavioural deficits. Thus, AEP acts as a δ-secretase, contributing to the age-dependent pathogenic mechanisms in AD.
Collapse
|