1
|
Saito J, Dave JM, Lau FD, Greif DM. Presenilin-1 in smooth muscle cells facilitates hypermuscularization in elastin aortopathy. iScience 2024; 27:108636. [PMID: 38226162 PMCID: PMC10788461 DOI: 10.1016/j.isci.2023.108636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/16/2023] [Accepted: 12/01/2023] [Indexed: 01/17/2024] Open
Abstract
Smooth muscle cell (SMC) accumulation is central to the pathogenesis of elastin-defective arterial diseases, including supravalvular aortic stenosis (SVAS). We previously demonstrated that elastin insufficiency activates Notch signaling in aortic SMCs. Activation of Notch is catalyzed by the enzyme gamma-secretase, but the role of catalytic subunits presenilin (PSEN)-1 or PSEN-2 in elastin aortopathy is not defined. Genetic approaches reveal that endothelial cell-specific Psen1 deletion does not improve elastin aortopathy whereas the deletion of either Psen1 in SMCs or Psen2 globally attenuates Notch pathway and SMC proliferation, mitigating aortic disease. With SMC-specific Psen1 deletion in elastin nulls, these rescue effects are more robust and in fact, survival is increased. SMC deletion of Psen1 also attenuates hypermuscularization in newborns heterozygous for the elastin null gene, which genetically mimics SVAS. Similarly, the pharmacological inhibition of PSEN-1 mitigates SMC accumulation in elastin aortopathy. These findings put forth SMC PSEN-1 as a potential therapeutic target in SVAS.
Collapse
Affiliation(s)
- Junichi Saito
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- Stem Cell Center, Yale University, New Haven, CT 06511, USA
| | - Jui M. Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- Stem Cell Center, Yale University, New Haven, CT 06511, USA
| | - Freddy Duarte Lau
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- Stem Cell Center, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
2
|
Eccles MK, Main N, Carlessi R, Armstrong AM, Sabale M, Roberts-Mok B, Tirnitz-Parker JEE, Agostino M, Groth D, Fraser PE, Verdile G. Quantitative comparison of presenilin protein expression reveals greater activity of PS2-γ-secretase. FASEB J 2024; 38:e23396. [PMID: 38156414 DOI: 10.1096/fj.202300954rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 12/30/2023]
Abstract
γ-secretase processing of amyloid precursor protein (APP) has long been of interest in the pathological progression of Alzheimer's disease (AD) due to its role in the generation of amyloid-β. The catalytic component of the enzyme is the presenilins of which there are two homologues, Presenilin-1 (PS1) and Presenilin-2 (PS2). The field has focussed on the PS1 form of this enzyme, as it is typically considered the more active at APP processing. However, much of this work has been completed without appropriate consideration of the specific levels of protein expression of PS1 and PS2. We propose that expression is an important factor in PS1- and PS2-γ-secretase activity, and that when this is considered, PS1 does not have greater activity than PS2. We developed and validated tools for quantitative assessment of PS1 and PS2 protein expression levels to enable the direct comparison of PS in exogenous and endogenous expression systems, in HEK-293 PS1 and/or PS2 knockout cells. We show that exogenous expression of Myc-PS1-NTF is 5.5-times higher than Myc-PS2-NTF. Quantitating endogenous PS protein levels, using a novel PS1/2 fusion standard we developed, showed similar results. When the marked difference in PS1 and PS2 protein levels is considered, we show that compared to PS1-γ-secretase, PS2-γ-secretase has equal or more activity on APP and Notch1. This study has implications for understanding the PS1- and PS2-specific contributions to substrate processing, and their potential influence in AD pathogenesis.
Collapse
Affiliation(s)
- Melissa K Eccles
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
| | - Nathan Main
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
| | - Rodrigo Carlessi
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
| | - Ayeisha Milligan Armstrong
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
| | - Miheer Sabale
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Brigid Roberts-Mok
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
| | - Janina E E Tirnitz-Parker
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
| | - Mark Agostino
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
| | - David Groth
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Giuseppe Verdile
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Bentley, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
3
|
Wang S, Dong K, Zhang J, Chen C, Shuai H, Yu X. Raw Inonotus obliquus polysaccharide counteracts Alzheimer's disease in a transgenic mouse model by activating the ubiquitin-proteosome system. Nutr Res Pract 2023; 17:1128-1142. [PMID: 38053824 PMCID: PMC10694425 DOI: 10.4162/nrp.2023.17.6.1128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/27/2023] [Accepted: 09/14/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND/OBJECTIVES Inonotus obliquus has been used as antidiabetic herb around the world, especially in the Russian and Scandinavian countries. Diabetes is widely believed to be a key factor in Alzheimer's disease (AD), which is widely considered to be type III diabetes. To investigate whether I. obliquus can also ameliorate AD, it would be interesting to identify new clues for AD treatment. We tested the anti-AD effects of raw Inonotus obliquus polysaccharide (IOP) in a mouse model of AD (3×Tg-AD transgenic mice). MATERIALS/METHODS SPF-grade 3×Tg-AD mice were randomly divided into three groups (Control, Metformin, and raw IOP groups, n = 5 per group). β-Amyloid deposition in the brain was analyzed using immunohistochemistry for AD characterization. Gene and protein expression of pertinent factors of the ubiquitin-proteasome system (UPS) was determined using real-time quantitative polymerase chain reaction and Western blotting. RESULTS Raw IOP significantly reduced the accumulation of amyloid aggregates and facilitated UPS activity, resulting in a significant reduction in AD-related symptoms in an AD mouse model. The presence of raw IOP significantly enhanced the expression of ubiquitin, E1, and Parkin (E3) at both the mRNA and protein levels in the mouse hippocampus. The mRNA level of ubiquitin carboxyl-terminal hydrolase isozyme L1, a key factor involved in UPS activation, also increased by approximately 50%. CONCLUSIONS Raw IOP could contribute to AD amelioration via the UPS pathway, which could be considered as a new potential strategy for AD treatment, although we could not exclude other mechanisms involved in counteracting AD processing.
Collapse
Affiliation(s)
- Shumin Wang
- School of Basic Medicine, Dali University, Dali 671000, China
| | - Kaiye Dong
- Department of Ophthalmology, The First Affiliated Hospital of Dali University, Dali 671000, China
| | - Ji Zhang
- College of Clinical Medicine, Dali University, Dali 671000, China
| | - Chaochao Chen
- College of Clinical Medicine, Dali University, Dali 671000, China
| | - Hongyan Shuai
- School of Basic Medicine, Dali University, Dali 671000, China
| | - Xin Yu
- School of Basic Medicine, Dali University, Dali 671000, China
| |
Collapse
|
4
|
Narlawar R, Serneels L, Gaffric C, Gijsen HJM, De Strooper B, Bischoff F. Discovery of brain permeable 2-Azabicyclo[2.2.2]octane sulfonamides as a novel class of presenilin-1 selective γ-secretase inhibitors. Eur J Med Chem 2023; 260:115725. [PMID: 37657269 DOI: 10.1016/j.ejmech.2023.115725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/04/2023] [Accepted: 08/13/2023] [Indexed: 09/03/2023]
Abstract
This paper describes the rational design, synthesis, structure-activity relationship (SAR), and biological profile of presenilin-1 (PSEN-1) complex selective γ-secretase inhibitors, assessed for selectivity using a unique set of four γ-secretase subtype complexes. A set of known PSEN-1 selective γ-Secretase inhibitors (GSIs) was analyzed to understand the pharmacophoric features required for selective inhibition. Conformational modeling suggests that a characteristic 'U' shape orientation between aromatic sulfone/sulfonamide and aryl ring is crucial for PSEN-1 selectivity and potency. Using these insights, a series of brain-penetrant 2-azabicyclo[2,2,2]octane sulfonamides was devised and synthesized as a new class of PSEN-1 selective inhibitors. Compounds 13c and 13k displayed high potency towards PSEN1-APH1B complex but moderate selectivity towards PSEN2 complexes. However, compound (+)-13b displayed low nanomolar potency towards the PSEN1-APH1B complex, little (∼4-fold) selectivity towards PSEN1-APH1A, and high selectivity (>350-fold) versus PSEN2 complexes. Excellent brain penetration, no significant CYP inhibition, or cardiotoxicity, good solubility, and permeability make (+)-13b an excellent candidate for further lead optimization.
Collapse
Affiliation(s)
- Rajeshwar Narlawar
- Discovery Chemistry, Therapeutics Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium; KU Leuven, Department of Neurosciences, Leuven Institute for Neuroscience and Disease, (LIND), Leuven, Belgium.
| | | | - Celia Gaffric
- Discovery Chemistry, Therapeutics Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Harrie J M Gijsen
- Discovery Chemistry, Therapeutics Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Bart De Strooper
- KU Leuven, Department of Neurosciences, Leuven Institute for Neuroscience and Disease, (LIND), Leuven, Belgium; VIB Center for Brain and Disease Research, Leuven, Belgium; Dementia Research Institute, University College London, London, UK
| | - François Bischoff
- Discovery Chemistry, Therapeutics Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium.
| |
Collapse
|
5
|
Nordvall G, Lundkvist J, Sandin J. Gamma-secretase modulators: a promising route for the treatment of Alzheimer's disease. Front Mol Neurosci 2023; 16:1279740. [PMID: 37908487 PMCID: PMC10613654 DOI: 10.3389/fnmol.2023.1279740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/15/2023] [Indexed: 11/02/2023] Open
Abstract
Recent clinical data with three therapeutic anti-Aβ antibodies have demonstrated that removal of Aβ-amyloid plaques in early Alzheimer's disease (AD) can attenuate disease progression. This ground-breaking progress in AD medicine has validated both the amyloid cascade hypothesis and Aβ-amyloid as therapeutic targets. These results also strongly support therapeutic approaches that aim to reduce the production of amyloidogenic Aβ to prevent the formation of Aβ-pathology. One such strategy, so-called gamma-secretase modulators (GSM), has been thoroughly explored in preclinical settings but has yet to be fully tested in clinical trials. Recent scientific progress has shed new light on the role of Aβ in Alzheimer's disease and suggests that GSMs exhibit specific pharmacological features that hold great promise for the prevention and treatment of Alzheimer's disease. In this short review, we discuss the data that support why it is important to continue to progress in this class of compounds.
Collapse
Affiliation(s)
- Gunnar Nordvall
- AlzeCure Pharma AB, Huddinge, Sweden
- Department of Neurobiology, Care Sciences, and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Johan Lundkvist
- AlzeCure Pharma AB, Huddinge, Sweden
- Department of Neurobiology, Care Sciences, and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Sinfonia Biotherapeutics AB, Huddinge, Sweden
| | - Johan Sandin
- AlzeCure Pharma AB, Huddinge, Sweden
- Department of Neurobiology, Care Sciences, and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
Serneels L, Narlawar R, Benito LP, Municoy M, Guallar V, T'Syen D, Dewilde M, Bischoff F, Fraiponts E, Tresadern G, Roevens PWM, Gijsen HJM, De Strooper B. Selective inhibitors of the PSEN1-gamma-secretase complex. J Biol Chem 2023:104794. [PMID: 37164155 DOI: 10.1016/j.jbc.2023.104794] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/12/2023] Open
Abstract
Clinical development of γ-secretases, a family of intramembrane cleaving proteases, as therapeutic targets for a variety of disorders including cancer and Alzheimer's disease, was aborted because of serious mechanism based side effects in phase III trials of unselective inhibitors. Selective inhibition of specific γ-secretase complexes, containing either PSEN1 or PSEN2 as catalytic subunit and APH1A or APH1B as supporting subunits, do provide a feasible therapeutic window in preclinical models of these disorders. We explore here the pharmacophoric features required for PSEN1 versus PSEN2 selective inhibition. We synthesized a series of brain penetrant 2-azabicyclo[2,2,2]octane sulfonamides and identified a compound with low nanomolar potency and high selectivity (>250-fold) towards the PSEN1-APH1B sub-complex versus PSEN2 sub-complexes. We used modelling and site directed mutagenesis to identify critical amino acids along the entry part of this inhibitor into the catalytic site of PSEN1. Specific targeting one of the different γ-secretase complexes might provide safer drugs in the future.
Collapse
Affiliation(s)
- Lutgarde Serneels
- VIB Center for Brain and Disease Research and KU Leuven, Department of Neurosciences, Leuven, Belgium
| | - Rajeshwar Narlawar
- VIB Center for Brain and Disease Research and KU Leuven, Department of Neurosciences, Leuven, Belgium; Discovery Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Laura Perez Benito
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Marti Municoy
- Nostrum Biodiscovery, Jordi Girona 29, Nexus II D128, 08034, Barcelona, Spain
| | - Victor Guallar
- Barcelona Supercomputing Center, Jordi Girona 29, E-08034 Barcelona, Spain; ICREA, Passeig Lluís Companys 23, E-08010 Barcelona, Spain
| | - Dries T'Syen
- VIB Center for Brain and Disease Research and KU Leuven, Department of Neurosciences, Leuven, Belgium
| | - Maarten Dewilde
- VIB Center for Brain and Disease Research and KU Leuven, Department of Neurosciences, Leuven, Belgium
| | - François Bischoff
- Discovery Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Erwin Fraiponts
- Charles River Laboratories, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Gary Tresadern
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Peter W M Roevens
- Campus Strategy & Partnerships, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Harrie J M Gijsen
- Discovery Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Bart De Strooper
- VIB Center for Brain and Disease Research and KU Leuven, Department of Neurosciences, Leuven, Belgium; Dementia Research Institute, University College London, London, UK.
| |
Collapse
|
7
|
Petit D, Fernández SG, Zoltowska KM, Enzlein T, Ryan NS, O'Connor A, Szaruga M, Hill E, Vandenberghe R, Fox NC, Chávez-Gutiérrez L. Aβ profiles generated by Alzheimer's disease causing PSEN1 variants determine the pathogenicity of the mutation and predict age at disease onset. Mol Psychiatry 2022; 27:2821-2832. [PMID: 35365805 PMCID: PMC9156411 DOI: 10.1038/s41380-022-01518-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023]
Abstract
Familial Alzheimer's disease (FAD), caused by mutations in Presenilin (PSEN1/2) and Amyloid Precursor Protein (APP) genes, is associated with an early age at onset (AAO) of symptoms. AAO is relatively consistent within families and between carriers of the same mutations, but differs markedly between individuals carrying different mutations. Gaining a mechanistic understanding of why certain mutations manifest several decades earlier than others is extremely important in elucidating the foundations of pathogenesis and AAO. Pathogenic mutations affect the protease (PSEN/γ-secretase) and the substrate (APP) that generate amyloid β (Aβ) peptides. Altered Aβ metabolism has long been associated with AD pathogenesis, with absolute or relative increases in Aβ42 levels most commonly implicated in the disease development. However, analyses addressing the relationships between these Aβ42 increments and AAO are inconsistent. Here, we investigated this central aspect of AD pathophysiology via comprehensive analysis of 25 FAD-linked Aβ profiles. Hypothesis- and data-driven approaches demonstrate linear correlations between mutation-driven alterations in Aβ profiles and AAO. In addition, our studies show that the Aβ (37 + 38 + 40) / (42 + 43) ratio offers predictive value in the assessment of 'unclear' PSEN1 variants. Of note, the analysis of PSEN1 variants presenting additionally with spastic paraparesis, indicates that a different mechanism underlies the aetiology of this distinct clinical phenotype. This study thus delivers valuable assays for fundamental, clinical and genetic research as well as supports therapeutic interventions aimed at shifting Aβ profiles towards shorter Aβ peptides.
Collapse
Affiliation(s)
- Dieter Petit
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Sara Gutiérrez Fernández
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Katarzyna Marta Zoltowska
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Thomas Enzlein
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack Str. 10, 68163, Mannheim, Germany
| | - Natalie S Ryan
- UK Dementia Research Institute at UCL, Queen Square, WC1N 3BG, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Antoinette O'Connor
- UK Dementia Research Institute at UCL, Queen Square, WC1N 3BG, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Maria Szaruga
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Elizabeth Hill
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Herestraat 49 box 1027, 3000, Leuven, Belgium
- Neurology Department, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Nick C Fox
- UK Dementia Research Institute at UCL, Queen Square, WC1N 3BG, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Lucía Chávez-Gutiérrez
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium.
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium.
| |
Collapse
|
8
|
Neuroprotective Effects of Probiotic-Supplemented Diet on Cognitive Behavior of 3xTg-AD Mice. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:4602428. [PMID: 35035837 PMCID: PMC8754595 DOI: 10.1155/2022/4602428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/08/2021] [Accepted: 12/18/2021] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is recognized as one of the most common types of senile dementia. AD patients first suffer memory loss for recent events (short-term memory impairment). As the disease progresses, they are deprived of self-awareness. This study aims to explore the effects of a probiotic-supplemented diet on the cognitive behaviors and pathological features of mouse models of Alzheimer's disease (AD). Mice in the control group and the 3xTg-AD group were fed a regular diet and a probiotic-supplemented diet, respectively, for 20 weeks. Behavioral experiments like Morris's water maze and Y maze were conducted. Then, feces of mice were collected for 16S sRNA gene sequencing for microorganisms. In the end, soluble and insoluble Aβ40 and Aβ42 in the hippocampus and cortex of mice in each group were quantitatively analyzed with a double-antibody Sandwich ELISA. The expression levels of tau protein and gliocyte in the hippocampus and cortex were detected using the Western Blot method. The result of the Morris water maze experiment indicated that, in the place navigation test, the mice in the 3xTg-AD group experienced a significant decline in the learning ability and a longer escape latency and in the space exploration test, the swimming time of mice in the 3xTg-AD group in the target quadrant decreased and after being treated with the probiotic diet, mice in the 3xTg-AD group had improved learning and memory ability. The result of Y maze showed that the probiotic diet can improve the spontaneous alternation accuracy of mice in the 3xTg-AD group. The result of 16s rRNA gene sequencing showed that, compared with mice in the WT group, those in the 3xTg-AD group experienced a change in the intestinal flora. The Western Blot result displayed a decreased expression level of tau (pS202) (P < 0.05) and decreased expression levels of Iba-1 and GFAP (P < 0.05). The result of the ELISA experiment showed decreased levels of soluble and insoluble Aβ40 and Aβ42 in 3xTg-AD mice (P < 0.05). In conclusion, a probiotic diet can prevent and treat AD by improving the intestinal flora of 3xTg-AD.
Collapse
|
9
|
Ioppolo A, Eccles M, Groth D, Verdile G, Agostino M. Evaluation of Virtual Screening Strategies for the Identification of γ-Secretase Inhibitors and Modulators. Molecules 2021; 27:176. [PMID: 35011410 PMCID: PMC8746326 DOI: 10.3390/molecules27010176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 11/17/2022] Open
Abstract
γ-Secretase is an intramembrane aspartyl protease that is important in regulating normal cell physiology via cleavage of over 100 transmembrane proteins, including Amyloid Precursor Protein (APP) and Notch family receptors. However, aberrant proteolysis of substrates has implications in the progression of disease pathologies, including Alzheimer's disease (AD), cancers, and skin disorders. While several γ-secretase inhibitors have been identified, there has been toxicity observed in clinical trials associated with non-selective enzyme inhibition. To address this, γ-secretase modulators have been identified and pursued as more selective agents. Recent structural evidence has provided an insight into how γ-secretase inhibitors and modulators are recognized by γ-secretase, providing a platform for rational drug design targeting this protease. In this study, docking- and pharmacophore-based screening approaches were evaluated for their ability to identify, from libraries of known inhibitors and modulators with decoys with similar physicochemical properties, γ-secretase inhibitors and modulators. Using these libraries, we defined strategies for identifying both γ-secretase inhibitors and modulators incorporating an initial pharmacophore-based screen followed by a docking-based screen, with each strategy employing distinct γ-secretase structures. Furthermore, known γ-secretase inhibitors and modulators were able to be identified from an external set of bioactive molecules following application of the derived screening strategies. The approaches described herein will inform the discovery of novel small molecules targeting γ-secretase.
Collapse
Affiliation(s)
- Alicia Ioppolo
- Curtin Health and Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.I.); (M.E.); (D.G.); (G.V.)
| | - Melissa Eccles
- Curtin Health and Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.I.); (M.E.); (D.G.); (G.V.)
| | - David Groth
- Curtin Health and Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.I.); (M.E.); (D.G.); (G.V.)
| | - Giuseppe Verdile
- Curtin Health and Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.I.); (M.E.); (D.G.); (G.V.)
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Mark Agostino
- Curtin Health and Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.I.); (M.E.); (D.G.); (G.V.)
- Curtin Institute for Computation, Curtin University, Bentley, WA 6102, Australia
| |
Collapse
|
10
|
Govaerts I, Prieto C, Vandersmissen C, Gielen O, Jacobs K, Provost S, Nittner D, Maertens J, Boeckx N, De Keersmaecker K, Segers H, Cools J. PSEN1-selective gamma-secretase inhibition in combination with kinase or XPO-1 inhibitors effectively targets T cell acute lymphoblastic leukemia. J Hematol Oncol 2021; 14:97. [PMID: 34167562 PMCID: PMC8223323 DOI: 10.1186/s13045-021-01114-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/15/2021] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND T cell acute lymphoblastic leukemia (T-ALL) is a high-risk subtype that comprises 10-15% of childhood and 20-25% of adult ALL cases. Over 70% of T-ALL patients harbor activating mutations in the NOTCH1 signaling pathway and are predicted to be sensitive to gamma-secretase inhibitors. We have recently demonstrated that selective inhibition of PSEN1-containing gamma-secretase complexes can overcome the dose-limiting toxicity associated with broad gamma-secretase inhibitors. In this study, we developed combination treatment strategies with the PSEN1-selective gamma-secretase inhibitor MRK-560 and other targeted agents (kinase inhibitors ruxolitinib and imatinib; XPO-1 inhibitor KPT-8602/eltanexor) for the treatment of T-ALL. METHODS We treated T-ALL cell lines in vitro and T-ALL patient-derived xenograft (PDX) models in vivo with MRK-560 alone or in combination with other targeted inhibitors (ruxolitinib, imatinib or KPT-8602/eltanexor). We determined effects on proliferation of the cell lines and leukemia development and survival in the PDX models. RESULTS All NOTCH1-signaling-dependent T-ALL cell lines were sensitive to MRK-560 and its combination with ruxolitinib or imatinib in JAK1- or ABL1-dependent cell lines synergistically inhibited leukemia proliferation. We also observed strong synergy between MRK-560 and KPT-8602 (eltanexor) in all NOTCH1-dependent T-ALL cell lines. Such synergy was also observed in vivo in a variety of T-ALL PDX models with NOTCH1 or FBXW7 mutations. Combination treatment significantly reduced leukemic infiltration in vivo and resulted in a survival benefit when compared to single treatment groups. We did not observe weight loss or goblet cell hyperplasia in single drug or combination treated mice when compared to control. CONCLUSIONS These data demonstrate that the antileukemic effect of PSEN1-selective gamma-secretase inhibition can be synergistically enhanced by the addition of other targeted inhibitors. The combination of MRK-560 with KPT-8602 is a highly effective treatment combination, which circumvents the need for the identification of additional mutations and provides a clear survival benefit in vivo. These promising preclinical data warrant further development of combination treatment strategies for T-ALL based on PSEN1-selective gamma-secretase inhibition.
Collapse
Affiliation(s)
- Inge Govaerts
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Cristina Prieto
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Charlien Vandersmissen
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Olga Gielen
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Kris Jacobs
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | - Sarah Provost
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
| | | | - Johan Maertens
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
- Department of Hematology, UZ Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Nancy Boeckx
- Department of Laboratory Medicine, UZ Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Kim De Keersmaecker
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Heidi Segers
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Pediatric Oncology, UZ Leuven, Leuven, Belgium
| | - Jan Cools
- Center for Human Genetics, KU Leuven, Leuven, Belgium.
- Center for Cancer Biology, VIB, Leuven, Belgium.
- Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven, Belgium.
| |
Collapse
|
11
|
Allen F, Maillard I. Therapeutic Targeting of Notch Signaling: From Cancer to Inflammatory Disorders. Front Cell Dev Biol 2021; 9:649205. [PMID: 34124039 PMCID: PMC8194077 DOI: 10.3389/fcell.2021.649205] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Over the past two decades, the Notch signaling pathway has been investigated as a therapeutic target for the treatment of cancers, and more recently in the context of immune and inflammatory disorders. Notch is an evolutionary conserved pathway found in all metazoans that is critical for proper embryonic development and for the postnatal maintenance of selected tissues. Through cell-to-cell contacts, Notch orchestrates cell fate decisions and differentiation in non-hematopoietic and hematopoietic cell types, regulates immune cell development, and is integral to shaping the amplitude as well as the quality of different types of immune responses. Depriving some cancer types of Notch signals has been shown in preclinical studies to stunt tumor growth, consistent with an oncogenic function of Notch signaling. In addition, therapeutically antagonizing Notch signals showed preclinical potential to prevent or reverse inflammatory disorders, including autoimmune diseases, allergic inflammation and immune complications of life-saving procedures such allogeneic bone marrow and solid organ transplantation (graft-versus-host disease and graft rejection). In this review, we discuss some of these unique approaches, along with the successes and challenges encountered so far to target Notch signaling in preclinical and early clinical studies. Our goal is to emphasize lessons learned to provide guidance about emerging strategies of Notch-based therapeutics that could be deployed safely and efficiently in patients with immune and inflammatory disorders.
Collapse
Affiliation(s)
- Frederick Allen
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Ivan Maillard
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
12
|
Majumder S, Crabtree JS, Golde TE, Minter LM, Osborne BA, Miele L. Targeting Notch in oncology: the path forward. Nat Rev Drug Discov 2021; 20:125-144. [PMID: 33293690 DOI: 10.1038/s41573-020-00091-3] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
Notch signalling is involved in many aspects of cancer biology, including angiogenesis, tumour immunity and the maintenance of cancer stem-like cells. In addition, Notch can function as an oncogene and a tumour suppressor in different cancers and in different cell populations within the same tumour. Despite promising preclinical results and early-phase clinical trials, the goal of developing safe, effective, tumour-selective Notch-targeting agents for clinical use remains elusive. However, our continually improving understanding of Notch signalling in specific cancers, individual cancer cases and different cell populations, as well as crosstalk between pathways, is aiding the discovery and development of novel investigational Notch-targeted therapeutics.
Collapse
Affiliation(s)
- Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Judy S Crabtree
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Todd E Golde
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Lisa M Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Barbara A Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
13
|
Neuroprotective Effects of 2-Substituted 1, 3-Selenazole Amide Derivatives on Amyloid-Beta-Induced Toxicity in a Transgenic Caenorhabditis Elegans Model of Alzheimer's Disease. Neurotox Res 2021; 39:841-850. [PMID: 33400180 DOI: 10.1007/s12640-020-00321-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease is an age-related neurodegenerative disease, associated with the presence of extracellular amyloid-β (Aβ) plaques and neurofibrillary tangles. Although the pathogenesis of AD remains unclear, the characteristic feature of AD was reported to be the buildup of Aβ plaques. In this study, we extensively investigated the neuroprotective effects of 2-substituted 1,3-selenazole amide derivatives (CHF11) on Aβ1-42 transgenic Caenorhabditis elegans CL4176. Results showed that worms fed with CHF11 exhibited remarkably reduced paralysis, decreased levels of toxic Aβ oligomers and Aβ plaque deposition, as well as less ROS production in comparison with the untreated worms. The effective concentrations of CHF11 were arranged in the descending order of 100 µM > 10 µM > 1 µM. Real-time PCR analysis showed that there was no significant difference in Aβ expression between CHF11-administered group and the blank control group, suggesting that CHF11-induced reduction in toxic protein deposition may be regulated at the post-transcriptional level. In the meantime, the gene expressions of hsf-1 and its downstream target hsp-12.6 were significantly increased, indicating that CHF11 against Aβ toxicity may involve in HSF-1 signaling pathway in worms. In conclusion, CHF11 exhibits a significant protective effect against β-amyloid-induced toxicity in CL4176 by reducing β-amyloid aggregation and ROS production, which may involve in HSF-1 and downstream target HSP-12.6 pathway.
Collapse
|
14
|
Notch Pathway: A Journey from Notching Phenotypes to Cancer Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1287:201-222. [PMID: 33034034 DOI: 10.1007/978-3-030-55031-8_13] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Notch is a key evolutionary conserved pathway, which has fascinated and engaged the work of investigators in an uncountable number of biological fields, from development of metazoans to immunotherapy for cancer. The study of Notch has greatly contributed to the understanding of cancer biology and a substantial effort has been spent in designing Notch-targeting therapies. Due to its broad involvement in cancer, targeting Notch would allow to virtually modulate any aspect of the disease. However, this means that Notch-based therapies must be highly specific to avoid off-target effects. This review will present the newest mechanistic and therapeutic advances in the Notch field and discuss the promises and challenges of this constantly evolving field.
Collapse
|
15
|
Uddin MS, Kabir MT, Rahman MS, Behl T, Jeandet P, Ashraf GM, Najda A, Bin-Jumah MN, El-Seedi HR, Abdel-Daim MM. Revisiting the Amyloid Cascade Hypothesis: From Anti-Aβ Therapeutics to Auspicious New Ways for Alzheimer's Disease. Int J Mol Sci 2020; 21:5858. [PMID: 32824102 PMCID: PMC7461598 DOI: 10.3390/ijms21165858] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder related to age, characterized by the cerebral deposition of fibrils, which are made from the amyloid-β (Aβ), a peptide of 40-42 amino acids. The conversion of Aβ into neurotoxic oligomeric, fibrillar, and protofibrillar assemblies is supposed to be the main pathological event in AD. After Aβ accumulation, the clinical symptoms fall out predominantly due to the deficient brain clearance of the peptide. For several years, researchers have attempted to decline the Aβ monomer, oligomer, and aggregate levels, as well as plaques, employing agents that facilitate the reduction of Aβ and antagonize Aβ aggregation, or raise Aβ clearance from brain. Unluckily, broad clinical trials with mild to moderate AD participants have shown that these approaches were unsuccessful. Several clinical trials are running involving patients whose disease is at an early stage, but the preliminary outcomes are not clinically impressive. Many studies have been conducted against oligomers of Aβ which are the utmost neurotoxic molecular species. Trials with monoclonal antibodies directed against Aβ oligomers have exhibited exciting findings. Nevertheless, Aβ oligomers maintain equivalent states in both monomeric and aggregation forms; so, previously administered drugs that precisely decrease Aβ monomer or Aβ plaques ought to have displayed valuable clinical benefits. In this article, Aβ-based therapeutic strategies are discussed and several promising new ways to fight against AD are appraised.
Collapse
Affiliation(s)
- Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
| | - Md. Tanvir Kabir
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh;
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims CEDEX 2, France;
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland;
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Hesham R. El-Seedi
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China;
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 23 Uppsala, Sweden
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
16
|
Lessard CB, Rodriguez E, Ladd TB, Minter LM, Osborne BA, Miele L, Golde TE, Ran Y. Individual and combined presenilin 1 and 2 knockouts reveal that both have highly overlapping functions in HEK293T cells. J Biol Chem 2019; 294:11276-11285. [PMID: 31167792 DOI: 10.1074/jbc.ra119.008041] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/30/2019] [Indexed: 01/13/2023] Open
Abstract
Presenilins 1 and 2 (PS1 and 2) are the catalytic subunits of γ-secretase, a multiprotein protease that cleaves amyloid protein precursor and other type I transmembrane proteins. Previous studies with mouse models or cells have indicated differences in PS1 and PS2 functions. We have recently reported that clinical γ-secretase inhibitors (GSIs), initially developed to manage Alzheimer's disease and now being considered for other therapeutic interventions, are both pharmacologically and functionally distinct. Here, using CRISPR/Cas9-based gene editing, we established human HEK 293T cell lines in which endogenous PS1, PS2, or both have been knocked out. Using these knockout lines to examine differences in PS1- and PS2-mediated cleavage events, we confirmed that PS2 generates more intracellular β-amyloid than does PS1. Moreover, we observed subtle differences in PS1- and PS2-mediated cleavages of select substrates. In exploring the question of whether differences in activity among clinical GSIs could be attributed to differential inhibition of PS1 or PS2, we noted that select GSIs inhibit PS1 and PS2 activities on specific substrates with slightly different potencies. We also found that endoproteolysis of select PS1 FAD-linked variants in human cells is more efficient than what has been previously reported for mouse cell lines. Overall, these results obtained with HEK293T cells suggest that selective PS1 or PS2 inhibition by a given GSI does not explain the previously observed differences in functional and pharmacological properties among various GSIs.
Collapse
Affiliation(s)
- Christian B Lessard
- Department of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Edgardo Rodriguez
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, Florida 32610
| | - Thomas B Ladd
- Department of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Lisa M Minter
- Department of Veterinary and Animal Sciences, Center for Bioactive Delivery, Institute for Applied Life Sciences, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Barbara A Osborne
- Department of Veterinary and Animal Sciences, Center for Bioactive Delivery, Institute for Applied Life Sciences, and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, Massachusetts 01003
| | - Lucio Miele
- Department of Genetics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112
| | - Todd E Golde
- Department of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Yong Ran
- Department of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
17
|
Habets RA, de Bock CE, Serneels L, Lodewijckx I, Verbeke D, Nittner D, Narlawar R, Demeyer S, Dooley J, Liston A, Taghon T, Cools J, de Strooper B. Safe targeting of T cell acute lymphoblastic leukemia by pathology-specific NOTCH inhibition. Sci Transl Med 2019; 11:11/494/eaau6246. [DOI: 10.1126/scitranslmed.aau6246] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 01/18/2019] [Accepted: 04/29/2019] [Indexed: 12/15/2022]
Abstract
Given the high frequency of activating NOTCH1 mutations in T cell acute lymphoblastic leukemia (T-ALL), inhibition of the γ-secretase complex remains an attractive target to prevent ligand-independent release of the cytoplasmic tail and oncogenic NOTCH1 signaling. However, four different γ-secretase complexes exist, and available inhibitors block all complexes equally. As a result, these cause severe “on-target” gastrointestinal tract, skin, and thymus toxicity, limiting their therapeutic application. Here, we demonstrate that genetic deletion or pharmacologic inhibition of the presenilin-1 (PSEN1) subclass of γ-secretase complexes is highly effective in decreasing leukemia while avoiding dose-limiting toxicities. Clinically, T-ALL samples were found to selectively express only PSEN1-containing γ-secretase complexes. The conditional knockout of Psen1 in developing T cells attenuated the development of a mutant NOTCH1-driven leukemia in mice in vivo but did not abrogate normal T cell development. Treatment of T-ALL cell lines with the selective PSEN1 inhibitor MRK-560 effectively decreased mutant NOTCH1 processing and led to cell cycle arrest. These observations were extended to T-ALL patient-derived xenografts in vivo, demonstrating that MRK-560 treatment decreases leukemia burden and increased overall survival without any associated gut toxicity. Therefore, PSEN1-selective compounds provide a potential therapeutic strategy for safe and effective targeting of T-ALL and possibly also for other diseases in which NOTCH signaling plays a role.
Collapse
|
18
|
Voytyuk I, De Strooper B, Chávez-Gutiérrez L. Modulation of γ- and β-Secretases as Early Prevention Against Alzheimer's Disease. Biol Psychiatry 2018; 83:320-327. [PMID: 28918941 DOI: 10.1016/j.biopsych.2017.08.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 01/18/2023]
Abstract
The genetic evidence implicating amyloid-β in the initial stage of Alzheimer's disease is unequivocal. However, the long biochemical and cellular prodromal phases of the disease suggest that dementia is the result of a series of molecular and cellular cascades whose nature and connections remain unknown. Therefore, it is unlikely that treatments directed at amyloid-β will have major clinical effects in the later stages of the disease. We discuss the two major candidate therapeutic targets to lower amyloid-β in a preventive mode, i.e., γ- and β-secretase; the rationale behind these two targets; and the current state of the field.
Collapse
Affiliation(s)
- Iryna Voytyuk
- KU Leuven Department for Neurosciences, Leuven, Belgium; VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Bart De Strooper
- KU Leuven Department for Neurosciences, Leuven, Belgium; VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium; UK Dementia Research Institute, University College, London, United Kingdom.
| | - Lucía Chávez-Gutiérrez
- KU Leuven Department for Neurosciences, Leuven, Belgium; VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| |
Collapse
|
19
|
Siebel C, Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol Rev 2017; 97:1235-1294. [PMID: 28794168 DOI: 10.1152/physrev.00005.2017] [Citation(s) in RCA: 674] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Notch signaling is an evolutionarily highly conserved signaling mechanism, but in contrast to signaling pathways such as Wnt, Sonic Hedgehog, and BMP/TGF-β, Notch signaling occurs via cell-cell communication, where transmembrane ligands on one cell activate transmembrane receptors on a juxtaposed cell. Originally discovered through mutations in Drosophila more than 100 yr ago, and with the first Notch gene cloned more than 30 yr ago, we are still gaining new insights into the broad effects of Notch signaling in organisms across the metazoan spectrum and its requirement for normal development of most organs in the body. In this review, we provide an overview of the Notch signaling mechanism at the molecular level and discuss how the pathway, which is architecturally quite simple, is able to engage in the control of cell fates in a broad variety of cell types. We discuss the current understanding of how Notch signaling can become derailed, either by direct mutations or by aberrant regulation, and the expanding spectrum of diseases and cancers that is a consequence of Notch dysregulation. Finally, we explore the emerging field of Notch in the control of tissue homeostasis, with examples from skin, liver, lung, intestine, and the vasculature.
Collapse
Affiliation(s)
- Chris Siebel
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Urban Lendahl
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
20
|
Eketjäll S, Janson J, Kaspersson K, Bogstedt A, Jeppsson F, Fälting J, Haeberlein SB, Kugler AR, Alexander RC, Cebers G. AZD3293: A Novel, Orally Active BACE1 Inhibitor with High Potency and Permeability and Markedly Slow Off-Rate Kinetics. J Alzheimers Dis 2016; 50:1109-23. [PMID: 26890753 PMCID: PMC4927864 DOI: 10.3233/jad-150834] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A growing body of pathological, biomarker, genetic, and mechanistic data suggests that amyloid accumulation, as a result of changes in production, processing, and/or clearance of brain amyloid-β peptide (Aβ) concentrations, plays a key role in the pathogenesis of Alzheimer’s disease (AD). Beta-secretase 1 (BACE1) mediates the first step in the processing of amyloid-β protein precursor (AβPP) to Aβ peptides, with the soluble N terminal fragment of AβPP (sAβPPβ) as a direct product, and BACE1 inhibition is an attractive target for therapeutic intervention to reduce the production of Aβ. Here, we report the in vitro and in vivo pharmacological profile of AZD3293, a potent, highly permeable, orally active, blood-brain barrier (BBB) penetrating, BACE1 inhibitor with unique slow off-rate kinetics. The in vitro potency of AZD3293 was demonstrated in several cellular models, including primary cortical neurons. In vivo in mice, guinea pigs, and dogs, AZD3293 displayed significant dose- and time-dependent reductions in plasma, cerebrospinal fluid, and brain concentrations of Aβ40, Aβ42, and sAβPPβ. The in vitro potency of AZD3293 in mouse and guinea pig primary cortical neuronal cells was correlated to the in vivo potency expressed as free AZD3293 concentrations in mouse and guinea pig brains. In mice and dogs, the slow off-rate from BACE1 may have translated into a prolongation of the observed effect beyond the turnover rate of Aβ. The preclinical data strongly support the clinical development of AZD3293, and patients with AD are currently being recruited into a combined Phase 2/3 study to test the disease-modifying properties of AZD3293.
Collapse
Affiliation(s)
- Susanna Eketjäll
- AstraZeneca Translational Sciences Centre, Science for Life Laboratory, Personal Healthcare and Biomarkers, AstraZeneca, Solna, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Juliette Janson
- AstraZeneca Translational Sciences Centre, Science for Life Laboratory, Personal Healthcare and Biomarkers, AstraZeneca, Solna, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
| | | | - Anna Bogstedt
- AstraZeneca Translational Sciences Centre, Science for Life Laboratory, Personal Healthcare and Biomarkers, AstraZeneca, Solna, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Fredrik Jeppsson
- CNS and Pain iMed, AstraZeneca, Södertälje, Sweden.,Operations Global Quality, AstraZeneca, Södertälje, Sweden
| | | | | | - Alan R Kugler
- Neuroscience iMed, IMED Biotech Unit, AstraZeneca, Cambridge, MA, USA
| | | | - Gvido Cebers
- Neuroscience iMed, IMED Biotech Unit, AstraZeneca, Cambridge, MA, USA
| |
Collapse
|
21
|
Wu WL, Burnett DA, Clader J, Greenlee WJ, Jiang Q, Hyde LA, Del Vecchio RA, Cohen-Williams ME, Song L, Lee J, Terracina G, Zhang Q, Nomeir A, Parker EM, Zhang L. Design and synthesis of water soluble β-aminosulfone analogues of SCH 900229 as γ-secretase inhibitors. Bioorg Med Chem Lett 2016; 26:5836-5841. [PMID: 27836402 DOI: 10.1016/j.bmcl.2016.04.095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 11/16/2022]
Abstract
In this paper we describe our strategy to improve the aqueous solubility of SCH 900229, a potent PS1-selective γ-secretase inhibitor for the treatment of Alzheimer's disease. Incorporation of ionizable amino groups into the side chain terminal generates water soluble β-aminosulfone analogues of SCH 900229 that maintain robust in vitro potency and in vivo efficacy.
Collapse
Affiliation(s)
- Wen-Lian Wu
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Duane A Burnett
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - John Clader
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - William J Greenlee
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Qin Jiang
- Albany Molecular Research, Inc., 26 Corporate Cir, Albany, NY 12212, USA
| | - Lynn A Hyde
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | | | | | - Lixin Song
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Julie Lee
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Giuseppe Terracina
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Qi Zhang
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Amin Nomeir
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Eric M Parker
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Lili Zhang
- Merck Research Laboratories, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| |
Collapse
|
22
|
Kumar R, Juillerat-Jeanneret L, Golshayan D. Notch Antagonists: Potential Modulators of Cancer and Inflammatory Diseases. J Med Chem 2016; 59:7719-37. [DOI: 10.1021/acs.jmedchem.5b01516] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Rajesh Kumar
- Transplantation
Center and Transplantation Immunopathology Laboratory, Department
of Medicine and ‡University Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH-1011 Lausanne, Switzerland
| | - Lucienne Juillerat-Jeanneret
- Transplantation
Center and Transplantation Immunopathology Laboratory, Department
of Medicine and ‡University Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH-1011 Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation
Center and Transplantation Immunopathology Laboratory, Department
of Medicine and ‡University Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH-1011 Lausanne, Switzerland
| |
Collapse
|
23
|
Sala Frigerio C, De Strooper B. Alzheimer's Disease Mechanisms and Emerging Roads to Novel Therapeutics. Annu Rev Neurosci 2016; 39:57-79. [PMID: 27050320 DOI: 10.1146/annurev-neuro-070815-014015] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ten years of remarkable progress in understanding the fundamental biochemistry of Alzheimer's disease have been followed by ten years of remarkable and increasing clinical insight into the natural progression of the disorder. The concept of a long, intermediary, prodromal phase between the first appearance of amyloid plaques and tangles and the manifestation of dementia is now well established. The major challenge for the next decade is to chart the many cellular processes that underlie this phase and link the biochemical alterations to the clinical manifestation of Alzheimer's disease. We discuss here how genetics, new cell culture systems, and improved animal models will fuel this work. We anticipate that the resulting novel insights will provide a basis for further drug development for this terrible disease.
Collapse
Affiliation(s)
- Carlo Sala Frigerio
- VIB Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, Leuven 3000, Belgium; , .,Center for Human Genetics, KU Leuven, Leuven 3000, Belgium.,Leuven Research Institute for Neuroscience & Disease (LIND), KU Leuven, Leuven 3000, Belgium
| | - Bart De Strooper
- VIB Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, Leuven 3000, Belgium; , .,Center for Human Genetics, KU Leuven, Leuven 3000, Belgium.,Leuven Research Institute for Neuroscience & Disease (LIND), KU Leuven, Leuven 3000, Belgium.,Institute of Neurology, University College London, WC1N 3BG London, United Kingdom
| |
Collapse
|
24
|
Barage SH, Sonawane KD. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer's disease. Neuropeptides 2015; 52:1-18. [PMID: 26149638 DOI: 10.1016/j.npep.2015.06.008] [Citation(s) in RCA: 388] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 06/24/2015] [Accepted: 06/24/2015] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease is an irreversible, progressive neurodegenerative disorder. Various therapeutic approaches are being used to improve the cholinergic neurotransmission, but their role in AD pathogenesis is still unknown. Although, an increase in tau protein concentration in CSF has been described in AD, but several issues remains unclear. Extensive and accurate analysis of CSF could be helpful to define presence of tau proteins in physiological conditions, or released during the progression of neurodegenerative disease. The amyloid cascade hypothesis postulates that the neurodegeneration in AD caused by abnormal accumulation of amyloid beta (Aβ) plaques in various areas of the brain. The amyloid hypothesis has continued to gain support over the last two decades, particularly from genetic studies. Therefore, current research progress in several areas of therapies shall provide an effective treatment to cure this devastating disease. This review critically evaluates general biochemical and physiological functions of Aβ directed therapeutics and their relevance.
Collapse
Affiliation(s)
- Sagar H Barage
- Department of Biotechnology, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India
| | - Kailas D Sonawane
- Structural Bioinformatics Unit, Department of Biochemistry, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India; Department of Microbiology, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India.
| |
Collapse
|
25
|
Loss of Presenilin 2 Function Is Associated with Defective LPS-Mediated Innate Immune Responsiveness. Mol Neurobiol 2015; 53:3428-3438. [PMID: 26081153 DOI: 10.1007/s12035-015-9285-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 06/03/2015] [Indexed: 10/23/2022]
Abstract
The importance of presenilin-dependent γ-secretase protease activities in the development, neurogenesis, and immune system is highlighted by the diversity of its substrates and characterization of Psen1- and Psen2-deficient transgenic animals. Functional differences between presenilin 1 (PS1) and presenilin 2 (PS2) are incompletely understood. In this study, we have identified a Psen2-specific function, not shared by Psen1 in Toll-like receptor signaling. We show that immortalized fibroblasts and bone marrow-derived macrophages from Psen2- but not Psen1-deficient mice display reduced responsiveness to lipopolysaccharide (LPS) with decreased nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) activity and diminished pro-inflammatory cytokine production. In whole animal in vivo responses, Psen2-deficient animals have abnormal systemic production of LPS-stimulated pro-inflammatory cytokines. Mechanistically, we demonstrate that Psen2 deficiency is paralleled by reduced transcription of tlr4 mRNA and loss of LPS-induced tlr4 mRNA transcription regulation. These observations illustrate a novel PS2-dependent means of modulating LPS-mediated immune responses and identify a functional distinction between PS1 and PS2 in innate immunity.
Collapse
|
26
|
Wu G, Wu Z, Na S, Hershey JC. Quantitative assessment of Aβ peptide in brain, cerebrospinal fluid and plasma following oral administration of γ-secretase inhibitor MRK-560 in rats. Int J Neurosci 2015; 125:616-24. [DOI: 10.3109/00207454.2014.952730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
27
|
De Strooper B, Chávez Gutiérrez L. Learning by Failing: Ideas and Concepts to Tackle γ-Secretases in Alzheimer's Disease and Beyond. Annu Rev Pharmacol Toxicol 2015; 55:419-37. [DOI: 10.1146/annurev-pharmtox-010814-124309] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Bart De Strooper
- VIB Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, BE-3000 Leuven, Belgium
- Center for Human Genetics, Laboratory for the Research of Neurodegenerative Diseases, KU Leuven, BE-3000 Leuven, Belgium; ,
| | - Lucía Chávez Gutiérrez
- VIB Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, BE-3000 Leuven, Belgium
- Center for Human Genetics, Laboratory for the Research of Neurodegenerative Diseases, KU Leuven, BE-3000 Leuven, Belgium; ,
| |
Collapse
|
28
|
|
29
|
Georgievska B, Gustavsson S, Lundkvist J, Neelissen J, Eketjäll S, Ramberg V, Bueters T, Agerman K, Juréus A, Svensson S, Berg S, Fälting J, Lendahl U. Revisiting the peripheral sink hypothesis: inhibiting BACE1 activity in the periphery does not alter β-amyloid levels in the CNS. J Neurochem 2014; 132:477-86. [PMID: 25156639 DOI: 10.1111/jnc.12937] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/03/2014] [Indexed: 11/28/2022]
Abstract
Aggregation of amyloid beta (Aβ) peptides and the subsequent neural plaque formation is a central aspect of Alzheimer's disease. Various strategies to reduce Aβ load in the brain are therefore intensely pursued. It has been hypothesized that reducing Aβ peptides in the periphery, that is in organs outside the brain, would be a way to diminish Aβ levels and plaque load in the brain. In this report, we put this peripheral sink hypothesis to test by investigating how selective inhibition of Aβ production in the periphery using a β-secretase (BACE)1 inhibitor or reduced BACE1 gene dosage affects Aβ load in the brain. Selective inhibition of peripheral BACE1 activity in wild-type mice or mice over-expressing amyloid precursor protein (APPswe transgenic mice; Tg2576) reduced Aβ levels in the periphery but not in the brain, not even after chronic treatment over several months. In contrast, a BACE1 inhibitor with improved brain disposition reduced Aβ levels in both brain and periphery already after acute dosing. Mice heterozygous for BACE1, displayed a 62% reduction in plasma Aβ40, whereas brain Aβ40 was only lowered by 11%. These data suggest that reduction of Aβ in the periphery is not sufficient to reduce brain Aβ levels and that BACE1 is not the rate-limiting enzyme for Aβ processing in the brain. This provides evidence against the peripheral sink hypothesis and suggests that a decrease in Aβ via BACE1 inhibition would need to be carried out in the brain. Aggregation of amyloid beta (Aβ) peptides in the brain is a central aspect of Alzheimer's disease. In this study, we demonstrate that inhibition of Aβ formation by BACE1 inhibitors needs to be carried out in the brain and that reduction of Aβ in the periphery is not sufficient to reduce brain Aβ levels. This information is useful for developing future Aβ-targeting therapies for Alzheimer's disease.
Collapse
|
30
|
Potential therapeutic strategies for Alzheimer's disease targeting or beyond β-amyloid: insights from clinical trials. BIOMED RESEARCH INTERNATIONAL 2014; 2014:837157. [PMID: 25136630 PMCID: PMC4124758 DOI: 10.1155/2014/837157] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 06/23/2014] [Accepted: 06/25/2014] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with two hallmarks: β-amyloid plagues and neurofibrillary tangles. It is one of the most alarming illnesses to elderly people. No effective drugs and therapies have been developed, while mechanism-based explorations of therapeutic approaches have been intensively investigated. Outcomes of clinical trials suggested several pitfalls in the choice of biomarkers, development of drug candidates, and interaction of drug-targeted molecules; however, they also aroused concerns on the potential deficiency in our understanding of pathogenesis of AD, and ultimately stimulated the advent of novel drug targets tests. The anticipated increase of AD patients in next few decades makes development of better therapy an urgent issue. Here we attempt to summarize and compare putative therapeutic strategies that have completed clinical trials or are currently being tested from various perspectives to provide insights for treatments of Alzheimer's disease.
Collapse
|
31
|
Acx H, Chávez-Gutiérrez L, Serneels L, Lismont S, Benurwar M, Elad N, De Strooper B. Signature amyloid β profiles are produced by different γ-secretase complexes. J Biol Chem 2013; 289:4346-55. [PMID: 24338474 DOI: 10.1074/jbc.m113.530907] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
γ-Secretase complexes are involved in the generation of amyloid-β (Aβ) in the brain. Therefore, γ-secretase has been proposed as a potential therapeutic target in Alzheimer disease (AD). Targeting γ-secretase activity in AD requires the pharmacological dissociation of the processing of physiological relevant substrates and the generation of "toxic" Aβ. Previous reports suggest the differential targeting of γ-secretase complexes, based on their subunit composition, as a valid strategy. However, little is known about the biochemical properties of the different complexes, and key questions regarding their Aβ product profiles should be first addressed. Here, we expressed, purified, and analyzed, under the same conditions, the endopeptidase and carboxypeptidase-like activities of the four γ-secretase complexes present in humans. We find that the nature of the catalytic subunit in the complex affects both activities. Interestingly, PSEN2 complexes discriminate between the Aβ40 and Aβ38 production lines, indicating that Aβ generation in one or the other pathway can be dissociated. In contrast, the APH1 subunit mainly affects the carboxypeptidase-like activity, with APH1B complexes favoring the generation of longer Aβ peptides. In addition, we determined that expression of a single human γ-secretase complex in cell lines retains the intrinsic attributes of the protease while present in the membrane, providing validation for the in vitro studies. In conclusion, our data show that each γ-secretase complex produces a characteristic Aβ signature. The qualitative and quantitative differences between different γ-secretase complexes could be used to advance drug development in AD and other disorders.
Collapse
Affiliation(s)
- Hermien Acx
- From the Center for the Biology of Disease, Flemish Institute for Biology (VIB), 3000 Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
32
|
Golde TE, Koo EH, Felsenstein KM, Osborne BA, Miele L. γ-Secretase inhibitors and modulators. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1828:2898-907. [PMID: 23791707 PMCID: PMC3857966 DOI: 10.1016/j.bbamem.2013.06.005] [Citation(s) in RCA: 216] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/04/2013] [Indexed: 12/11/2022]
Abstract
γ-Secretase is a fascinating, multi-subunit, intramembrane cleaving protease that is now being considered as a therapeutic target for a number of diseases. Potent, orally bioavailable γ-secretase inhibitors (GSIs) have been developed and tested in humans with Alzheimer's disease (AD) and cancer. Preclinical studies also suggest the therapeutic potential for GSIs in other disease conditions. However, due to inherent mechanism based-toxicity of non-selective inhibition of γ-secretase, clinical development of GSIs will require empirical testing with careful evaluation of benefit versus risk. In addition to GSIs, compounds referred to as γ-secretase modulators (GSMs) remain in development as AD therapeutics. GSMs do not inhibit γ-secretase, but modulate γ-secretase processivity and thereby shift the profile of the secreted amyloid β peptides (Aβ) peptides produced. Although GSMs are thought to have an inherently safe mechanism of action, their effects on substrates other than the amyloid β protein precursor (APP) have not been extensively investigated. Herein, we will review the current state of development of GSIs and GSMs and explore pertinent biological and pharmacological questions pertaining to the use of these agents for select indications. This article is part of a Special Issue entitled: Intramembrane Proteases.
Collapse
Affiliation(s)
- Todd E Golde
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | | | | | | | | |
Collapse
|
33
|
Suárez-Calvet M, Belbin O, Pera M, Badiola N, Magrané J, Guardia-Laguarta C, Muñoz L, Colom-Cadena M, Clarimón J, Lleó A. Autosomal-dominant Alzheimer's disease mutations at the same codon of amyloid precursor protein differentially alter Aβ production. J Neurochem 2013; 128:330-9. [PMID: 24117942 DOI: 10.1111/jnc.12466] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Revised: 09/20/2013] [Accepted: 10/24/2013] [Indexed: 02/02/2023]
Abstract
Autosomal-dominant Alzheimer's disease (ADAD) is a genetic disorder caused by mutations in Amyloid Precursor Protein (APP) or Presenilin (PSEN) genes. Studying the mechanisms underlying these mutations can provide insight into the pathways that lead to AD pathology. The majority of biochemical studies on APP mutations to-date have focused on comparing mechanisms between mutations at different codons. It has been assumed that amino acid position is a major determinant of protein dysfunction and clinical phenotype. However, the differential effect of mutations at the same codon has not been sufficiently addressed. In the present study we compared the effects of the aggressive ADAD-associated APP I716F mutation with I716V and I716T on APP processing in human neuroglioma and CHO-K1 cells. All APP I716 mutations increased the ratio of Aβ42/40 and changed the product line preference of γ-secretase towards Aβ38 production. In addition, the APP I716F mutation impaired the ε-cleavage and the fourth cleavage of γ-secretase and led to abnormal APP β-CTF accumulation at the plasma membrane. Taken together, these data indicate that APP mutations at the same codon can induce diverse abnormalities in APP processing, some resembling PSEN1 mutations. These differential effects could explain the clinical differences observed among ADAD patients bearing different APP mutations at the same position. The amyloid precursor protein (APP) I716F mutation is associated with autosomal dominant Alzheimer's disease with the youngest age-at-onset for the APP locus. Here, we describe that this mutation, when compared to two other familial Alzheimer's disease mutations at the same codon (I716V and I716T), interfered distinctly with γ-secretase cleavage. While all three mutations direct γ-secretase cleavage towards the 48→38 production line, the APP I716F mutation also impaired the ε-cleavage and the fourth cleavage of γ-secretase, resembling a PSEN1 mutation. These features may contribute to the aggressiveness of this mutation.
Collapse
Affiliation(s)
- Marc Suárez-Calvet
- Department of Neurology, Memory Disorders Unit, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Alzheimer Laboratory, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chen XF, Zhang YW, Xu H, Bu G. Transcriptional regulation and its misregulation in Alzheimer's disease. Mol Brain 2013; 6:44. [PMID: 24144318 PMCID: PMC3854070 DOI: 10.1186/1756-6606-6-44] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 10/15/2013] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder characterized by loss of memory and cognitive function. A key neuropathological event in AD is the accumulation of amyloid-β (Aβ) peptide. The production and clearance of Aβ in the brain are regulated by a large group of genes. The expression levels of these genes must be fine-tuned in the brain to keep Aβ at a balanced amount under physiological condition. Misregulation of AD genes has been found to either increase AD risk or accelerate the disease progression. In recent years, important progress has been made in uncovering the regulatory elements and transcriptional factors that guide the expression of these genes. In this review, we describe the mechanisms of transcriptional regulation for the known AD genes and the misregualtion that leads to AD susceptibility.
Collapse
Affiliation(s)
- Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, 361102 Xiamen, Fujian, People's Republic of China.
| | | | | | | |
Collapse
|
35
|
Abstract
The cytokine TNF-α is a major drug target for rheumatoid arthritis, an inflammatory joint disorder. An alternative approach is to target the protease TNF-α convertase (TACE), which releases TNF-α from cells. However, because TACE cleaves other proteins involved in development and cancer, a tissue-specific inhibition of TACE in immune cells appears mandatory. In this issue of the JCI, Issuree et al. report that iRHOM2 is a TACE activator in immune cells. Loss of iRHOM2 largely protects mice from inflammatory arthritis, making iRHOM2 a potential drug target for this condition.
Collapse
|