1
|
Zhang Y, Chen H, Li R, Sterling K, Song W. Amyloid β-based therapy for Alzheimer's disease: challenges, successes and future. Signal Transduct Target Ther 2023; 8:248. [PMID: 37386015 PMCID: PMC10310781 DOI: 10.1038/s41392-023-01484-7] [Citation(s) in RCA: 287] [Impact Index Per Article: 143.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 07/01/2023] Open
Abstract
Amyloid β protein (Aβ) is the main component of neuritic plaques in Alzheimer's disease (AD), and its accumulation has been considered as the molecular driver of Alzheimer's pathogenesis and progression. Aβ has been the prime target for the development of AD therapy. However, the repeated failures of Aβ-targeted clinical trials have cast considerable doubt on the amyloid cascade hypothesis and whether the development of Alzheimer's drug has followed the correct course. However, the recent successes of Aβ targeted trials have assuaged those doubts. In this review, we discussed the evolution of the amyloid cascade hypothesis over the last 30 years and summarized its application in Alzheimer's diagnosis and modification. In particular, we extensively discussed the pitfalls, promises and important unanswered questions regarding the current anti-Aβ therapy, as well as strategies for further study and development of more feasible Aβ-targeted approaches in the optimization of AD prevention and treatment.
Collapse
Affiliation(s)
- Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Huaqiu Chen
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China.
| |
Collapse
|
2
|
Imbimbo BP, Ippati S, Watling M, Imbimbo C. Role of monomeric amyloid-β in cognitive performance in Alzheimer's disease: Insights from clinical trials with secretase inhibitors and monoclonal antibodies. Pharmacol Res 2023; 187:106631. [PMID: 36586644 DOI: 10.1016/j.phrs.2022.106631] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
According to the β-amyloid (Aβ) hypothesis of Alzheimer's disease (AD), brain Aβ accumulation is the primary cascade event leading to cognitive deficit and dementia. Numerous anti-Aβ drugs either inhibiting production or aggregation of Aβ or stimulating its clearance have failed to show clinical benefit in large scale AD trials, with β- and γ-secretase inhibitors consistently worsening cognitive and clinical decline. In June 2021, the FDA approved aducanumab, an anti-Aβ monoclonal antibody for early AD based on its ability to reduce brain amyloid plaques, while two other amyloid-clearing antibodies (lecanemab and donanemab) have recently produced encouraging cognitive and clinical results. We reviewed AD trials using PubMed, meeting abstracts and ClinicalTrials.gov and evaluated the effects of such drugs on cerebrospinal fluid (CSF) Aβ levels, correlating them with cognitive effects. We found that β-secretase and γ-secretase inhibitors produce detrimental cognitive effects by significantly reducing CSF Aβ levels. We speculate that monoclonal antibodies targeting Aβ protofibrils, fibrils or plaques may improve cognitive performance in early AD by increasing soluble Aβ levels through Aβ aggregate disassembly and/or stabilization of existing Aβ monomers.These findings suggest that the real culprit in AD may be decreased levels of soluble monomeric Aβ due to sequestration into brain Aβ aggregates and plaques.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy.
| | - Stefania Ippati
- San Raffaele Scientific Institute, San Raffaele Hospital, 20132 Milan, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Ltd, Reading, UK
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
3
|
Alexander C, Li T, Hattori Y, Chiu D, Frost GR, Jonas L, Liu C, Anderson CJ, Wong E, Park L, Iadecola C, Li YM. Hypoxia Inducible Factor-1α binds and activates γ-secretase for Aβ production under hypoxia and cerebral hypoperfusion. Mol Psychiatry 2022; 27:4264-4273. [PMID: 35764706 PMCID: PMC9722522 DOI: 10.1038/s41380-022-01676-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023]
Abstract
Hypoxic-ischemic injury has been linked with increased risk for developing Alzheimer's disease (AD). The underlying mechanism of this association is poorly understood. Here, we report distinct roles for hypoxia-inducible factor-1α (Hif-1α) in the regulation of BACE1 and γ-secretase activity, two proteases involved in the production of amyloid-beta (Aβ). We have demonstrated that Hif-1α upregulates both BACE1 and γ-secretase activity for Aβ production in brain hypoxia-induced either by cerebral hypoperfusion or breathing 10% O2. Hif-1α binds to γ-secretase, which elevates the amount of active γ-secretase complex without affecting the level of individual subunits in hypoxic-ischemic mouse brains. Additionally, the expression of full length Hif-1α increases BACE1 and γ-secretase activity in primary neuronal culture, whereas a transcriptionally incompetent Hif-1α variant only activates γ-secretase. These findings indicate that Hif-1α transcriptionally upregulates BACE1 and nontranscriptionally activates γ-secretase for Aβ production in hypoxic-ischemic conditions. Consequently, Hif-1α-mediated Aβ production may be an adaptive response to hypoxic-ischemic injury, subsequently leading to increased risk for AD. Preventing the interaction of Hif-1α with γ-secretase may therefore be a promising therapeutic strategy for AD treatment.
Collapse
Affiliation(s)
- Courtney Alexander
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Thomas Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Danica Chiu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Georgia R Frost
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Lauren Jonas
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Chenge Liu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Corey J Anderson
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Eitan Wong
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA.
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA.
| |
Collapse
|
4
|
Jeong H, Shin H, Hong S, Kim Y. Physiological Roles of Monomeric Amyloid-β and Implications for Alzheimer's Disease Therapeutics. Exp Neurobiol 2022; 31:65-88. [PMID: 35673997 PMCID: PMC9194638 DOI: 10.5607/en22004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) progressively inflicts impairment of synaptic functions with notable deposition of amyloid-β (Aβ) as senile plaques within the extracellular space of the brain. Accordingly, therapeutic directions for AD have focused on clearing Aβ plaques or preventing amyloidogenesis based on the amyloid cascade hypothesis. However, the emerging evidence suggests that Aβ serves biological roles, which include suppressing microbial infections, regulating synaptic plasticity, promoting recovery after brain injury, sealing leaks in the blood-brain barrier, and possibly inhibiting the proliferation of cancer cells. More importantly, these functions were found in in vitro and in vivo investigations in a hormetic manner, that is to be neuroprotective at low concentrations and pathological at high concentrations. We herein summarize the physiological roles of monomeric Aβ and current Aβ-directed therapies in clinical trials. Based on the evidence, we propose that novel therapeutics targeting Aβ should selectively target Aβ in neurotoxic forms such as oligomers while retaining monomeric Aβ in order to preserve the physiological functions of Aβ monomers.
Collapse
Affiliation(s)
- Hyomin Jeong
- Division of Integrated Science and Engineering, Underwood International College, Yonsei University, Incheon 21983, Korea
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Heewon Shin
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Seungpyo Hong
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Frontier Lab, Yonsei University, Seoul 03722, Korea
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - YoungSoo Kim
- Division of Integrated Science and Engineering, Underwood International College, Yonsei University, Incheon 21983, Korea
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Frontier Lab, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
5
|
Iqubal A, Rahman SO, Ahmed M, Bansal P, Haider MR, Iqubal MK, Najmi AK, Pottoo FH, Haque SE. Current Quest in Natural Bioactive Compounds for Alzheimer's Disease: Multi-Targeted-Designed-Ligand Based Approach with Preclinical and Clinical Based Evidence. Curr Drug Targets 2021; 22:685-720. [PMID: 33302832 DOI: 10.2174/1389450121999201209201004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/12/2020] [Accepted: 08/23/2020] [Indexed: 12/06/2022]
Abstract
Alzheimer's disease is a common and most chronic neurological disorder (NDs) associated with cognitive dysfunction. Pathologically, Alzheimer's disease (AD) is characterized by the presence of β-amyloid (Aβ) plaques, hyper-phosphorylated tau proteins, and neurofibrillary tangles, however, persistence oxidative-nitrative stress, endoplasmic reticulum stress, mitochondrial dysfunction, inflammatory cytokines, pro-apoptotic proteins along with altered neurotransmitters level are common etiological attributes in its pathogenesis. Rivastigmine, memantine, galantamine, and donepezil are FDA approved drugs for symptomatic management of AD, whereas tacrine has been withdrawn because of hepatotoxic profile. These approved drugs only exert symptomatic relief and exhibit poor patient compliance. In the current scenario, the number of published evidence shows the neuroprotective potential of naturally occurring bioactive molecules via their antioxidant, anti-inflammatory, antiapoptotic and neurotransmitter modulatory properties. Despite their potent therapeutic implications, concerns have arisen in context to their efficacy and probable clinical outcome. Thus, to overcome these glitches, many heterocyclic and cyclic hydrocarbon compounds inspired by natural sources have been synthesized and showed improved therapeutic activity. Computational studies (molecular docking) have been used to predict the binding affinity of these natural bioactive as well as synthetic compounds derived from natural sources for the acetylcholine esterase, α/β secretase Nuclear Factor kappa- light-chain-enhancer of activated B cells (NF-kB), Nuclear factor erythroid 2-related factor 2(Nrf2) and other neurological targets. Thus, in this review, we have discussed the molecular etiology of AD, focused on the pharmacotherapeutics of natural products, chemical and pharmacological aspects and multi-targeted designed ligands (MTDLs) of synthetic and semisynthetic molecules derived from the natural sources along with some important on-going clinical trials.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, JamiaHamdard, New Delhi-110062, India
| | - Syed Obaidur Rahman
- Department of Pharmaceutical Medicine, School of Pharmaceutical Education and Research, JamiaHamdard, New Delhi-110062, India
| | - Musheer Ahmed
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, JamiaHamdard, New Delhi-110062, India
| | - Pratichi Bansal
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, JamiaHamdard, New Delhi-110062, India
| | - Md Rafi Haider
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, JamiaHamdard, New Delhi-110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, JamiaHamdard, New Delhi-110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, JamiaHamdard, New Delhi-110062, India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal, University, P.O.BOX 1982, Damman, 31441, Saudi Arabia
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, JamiaHamdard, New Delhi-110062, India
| |
Collapse
|
6
|
Maigler KC, Buhr TJ, Park CS, Miller SA, Kozlowski DA, Marr RA. Assessment of the Effects of Altered Amyloid-Beta Clearance on Behavior following Repeat Closed-Head Brain Injury in Amyloid-Beta Precursor Protein Humanized Mice. J Neurotrauma 2021; 38:665-676. [PMID: 33176547 DOI: 10.1089/neu.2020.6989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Traumatic brain injury (TBI) increases the risk for dementias including Alzheimer's disease (AD) and chronic traumatic encephalopathy. Further, both human and animal model data indicate that amyloid-beta (Aβ) peptide accumulation and its production machinery are upregulated by TBI. Considering the clear link between chronic Aβ elevation and AD as well as tau pathology, the role(s) of Aβ in TBI is of high importance. Endopeptidases, including the neprilysin (NEP)-like enzymes, are key mediators of Aβ clearance and may affect susceptibility to pathology post-TBI. Here, we use a "humanized" mouse model of Aβ production, which expresses normal human amyloid-beta precursor protein (APP) under its natural transcriptional regulation and exposed them to a more clinically relevant repeated closed-head TBI paradigm. These transgenic mice also were crossed with mice deficient for the Aβ degrading enzymes NEP or NEP2 to assess models of reduced cerebral Aβ clearance in our TBI model. Our results show that the presence of the human form of Aβ did not exacerbate motor (Rotarod) and spatial learning/memory deficits (Morris water maze) post-injuries, while potentially reduced anxiety (Open Field) was observed. NEP and NEP2 deficiency also did not exacerbate these deficits post-injuries and was associated with protection from motor (NEP and NEP2) and spatial learning/memory deficits (NEP only). These data suggest that normally regulated expression of wild-type human APP/Aβ does not contribute to deficits acutely after TBI and may be protective at this stage of injury.
Collapse
Affiliation(s)
- Kathleen C Maigler
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Trevor J Buhr
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Christopher S Park
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Steven A Miller
- Department of Psychology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Dorothy A Kozlowski
- Department of Biological Sciences and Neuroscience Program, DePaul University, Chicago, Illinois, USA
| | - Robert A Marr
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| |
Collapse
|
7
|
Kent SA, Spires-Jones TL, Durrant CS. The physiological roles of tau and Aβ: implications for Alzheimer's disease pathology and therapeutics. Acta Neuropathol 2020; 140:417-447. [PMID: 32728795 PMCID: PMC7498448 DOI: 10.1007/s00401-020-02196-w] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 01/18/2023]
Abstract
Tau and amyloid beta (Aβ) are the prime suspects for driving pathology in Alzheimer's disease (AD) and, as such, have become the focus of therapeutic development. Recent research, however, shows that these proteins have been highly conserved throughout evolution and may have crucial, physiological roles. Such functions may be lost during AD progression or be unintentionally disrupted by tau- or Aβ-targeting therapies. Tau has been revealed to be more than a simple stabiliser of microtubules, reported to play a role in a range of biological processes including myelination, glucose metabolism, axonal transport, microtubule dynamics, iron homeostasis, neurogenesis, motor function, learning and memory, neuronal excitability, and DNA protection. Aβ is similarly multifunctional, and is proposed to regulate learning and memory, angiogenesis, neurogenesis, repair leaks in the blood-brain barrier, promote recovery from injury, and act as an antimicrobial peptide and tumour suppressor. This review will discuss potential physiological roles of tau and Aβ, highlighting how changes to these functions may contribute to pathology, as well as the implications for therapeutic development. We propose that a balanced consideration of both the physiological and pathological roles of tau and Aβ will be essential for the design of safe and effective therapeutics.
Collapse
Affiliation(s)
- Sarah A. Kent
- Translational Neuroscience PhD Programme, Centre for Discovery Brain Sciences and the UK Dementia Research Institute, The University of Edinburgh, 1 George Square, Edinburgh, EH8 9JZ Scotland, UK
| | - Tara L. Spires-Jones
- Centre for Discovery Brain Sciences and the UK Dementia Research Institute, The University of Edinburgh, 1 George Square, Edinburgh, EH8 9JZ Scotland, UK
| | - Claire S. Durrant
- Centre for Discovery Brain Sciences and the UK Dementia Research Institute, The University of Edinburgh, 1 George Square, Edinburgh, EH8 9JZ Scotland, UK
| |
Collapse
|
8
|
Brothers HM, Gosztyla ML, Robinson SR. The Physiological Roles of Amyloid-β Peptide Hint at New Ways to Treat Alzheimer's Disease. Front Aging Neurosci 2018; 10:118. [PMID: 29922148 PMCID: PMC5996906 DOI: 10.3389/fnagi.2018.00118] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022] Open
Abstract
Amyloid-ß (Aß) is best known as the misfolded peptide that is involved in the pathogenesis of Alzheimer's disease (AD), and it is currently the primary therapeutic target in attempts to arrest the course of this disease. This notoriety has overshadowed evidence that Aß serves several important physiological functions. Aß is present throughout the lifespan, it has been found in all vertebrates examined thus far, and its molecular sequence shows a high degree of conservation. These features are typical of a factor that contributes significantly to biological fitness, and this suggestion has been supported by evidence of functions that are beneficial for the brain. The putative roles of Aß include protecting the body from infections, repairing leaks in the blood-brain barrier, promoting recovery from injury, and regulating synaptic function. Evidence for these beneficial roles comes from in vitro and in vivo studies, which have shown that the cellular production of Aß rapidly increases in response to a physiological challenge and often diminishes upon recovery. These roles are further supported by the adverse outcomes of clinical trials that have attempted to deplete Aß in order to treat AD. We suggest that anti-Aß therapies will produce fewer adverse effects if the known triggers of Aß deposition (e.g., pathogens, hypertension, and diabetes) are addressed first.
Collapse
Affiliation(s)
- Holly M Brothers
- Department of Psychology, The Ohio State University Columbus, Columbus, OH, United States
| | - Maya L Gosztyla
- Department of Neuroscience, The Ohio State University Columbus, Columbus, OH, United States
| | - Stephen R Robinson
- Discipline of Psychology, School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Robinson SR, Brothers HM, Gosztyla ML. Consequences of redefining Alzheimer's disease in terms of amyloid burden without regard to cognitive decline. Neural Regen Res 2018; 13:2098-2099. [PMID: 30323134 PMCID: PMC6199922 DOI: 10.4103/1673-5374.241456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Stephen R Robinson
- Discipline of Psychology, School of Health and Biomedical Sciences, RMIT University, Bundoora; Institute for Breathing and Sleep, Austin Health, Heidelberg, Victoria, Australia
| | - Holly M Brothers
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Maya L Gosztyla
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
10
|
Peters-Libeu C, Campagna J, Mitsumori M, Poksay KS, Spilman P, Sabogal A, Bredesen DE, John V. sAβPPα is a Potent Endogenous Inhibitor of BACE1. J Alzheimers Dis 2016; 47:545-55. [PMID: 26401691 DOI: 10.3233/jad-150282] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Proteolytic cleavage of the amyloid-β protein precursor (AβPP) by the enzyme BACE1 (BACE) is the initial step in production of amyloid-β peptide (Aβ), and as such has been a major target of Alzheimer's disease (AD) drug discovery efforts. Overproduction of Aβ results in neuronal cell death and accumulation of amyloid plaques in AD and in traumatic brain injury, and is also associated with stroke due to cerebral amyloid angiopathy. Herein we report for the first time that sAβPPα, the product of the cleavage of AβPP by α-secretase, is a potent endogenous direct inhibitor of the BACE enzyme, and that its inhibition is likely by an allosteric mechanism. Furthermore, using small-angle X-ray scattering, we show that sAβPPβ, which is identical to sAβPPα except for a 16-amino acid truncation at the carboxy terminus, adopts a completely different structure than sAβPPα and does not inhibit BACE. Our data thus reveal a novel mechanistic role played by sAβPPα in regulating overproduction of Aβ and restoring neuronal homeostasis and neuroprotection. Identification of sAβPPα as a direct BACE inhibitor may lead to design of new therapeutics targeting pathologies associated with overproduction of Aβ.
Collapse
Affiliation(s)
| | - Jesus Campagna
- Drug Discovery Laboratory, Department of Neurology, UCLA, Los Angeles, CA, USA
| | | | | | - Patricia Spilman
- Drug Discovery Laboratory, Department of Neurology, UCLA, Los Angeles, CA, USA
| | - Alex Sabogal
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Dale E Bredesen
- Buck Institute for Research on Aging, Novato, CA, USA.,Drug Discovery Laboratory, Department of Neurology, UCLA, Los Angeles, CA, USA.,Easton Center for Alzheimer's Disease Research, UCLA, Los Angeles, CA, USA
| | - Varghese John
- Buck Institute for Research on Aging, Novato, CA, USA.,Drug Discovery Laboratory, Department of Neurology, UCLA, Los Angeles, CA, USA.,Easton Center for Alzheimer's Disease Research, UCLA, Los Angeles, CA, USA
| |
Collapse
|
11
|
Robinson S, Winer JL, Berkner J, Chan LAS, Denson JL, Maxwell JR, Yang Y, Sillerud LO, Tasker RC, Meehan WP, Mannix R, Jantzie LL. Imaging and serum biomarkers reflecting the functional efficacy of extended erythropoietin treatment in rats following infantile traumatic brain injury. J Neurosurg Pediatr 2016; 17:739-55. [PMID: 26894518 PMCID: PMC5369240 DOI: 10.3171/2015.10.peds15554] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Traumatic brain injury (TBI) is a leading cause of death and severe morbidity for otherwise healthy full-term infants around the world. Currently, the primary treatment for infant TBI is supportive, as no targeted therapies exist to actively promote recovery. The developing infant brain, in particular, has a unique response to injury and the potential for repair, both of which vary with maturation. Targeted interventions and objective measures of therapeutic efficacy are needed in this special population. The authors hypothesized that MRI and serum biomarkers can be used to quantify outcomes following infantile TBI in a preclinical rat model and that the potential efficacy of the neuro-reparative agent erythropoietin (EPO) in promoting recovery can be tested using these biomarkers as surrogates for functional outcomes. METHODS With institutional approval, a controlled cortical impact (CCI) was delivered to postnatal Day (P)12 rats of both sexes (76 rats). On postinjury Day (PID)1, the 49 CCI rats designated for chronic studies were randomized to EPO (3000 U/kg/dose, CCI-EPO, 24 rats) or vehicle (CCI-veh, 25 rats) administered intraperitoneally on PID1-4, 6, and 8. Acute injury (PID3) was evaluated with an immunoassay of injured cortex and serum, and chronic injury (PID13-28) was evaluated with digitized gait analyses, MRI, and serum immunoassay. The CCI-veh and CCI-EPO rats were compared with shams (49 rats) primarily using 2-way ANOVA with Bonferroni post hoc correction. RESULTS Following CCI, there was 4.8% mortality and 55% of injured rats exhibited convulsions. Of the injured rats designated for chronic analyses, 8.1% developed leptomeningeal cyst-like lesions verified with MRI and were excluded from further study. On PID3, Western blot showed that EPO receptor expression was increased in the injured cortex (p = 0.008). These Western blots also showed elevated ipsilateral cortex calpain degradation products for αII-spectrin (αII-SDPs; p < 0.001), potassium chloride cotransporter 2 (KCC2-DPs; p = 0.037), and glial fibrillary acidic protein (GFAP-DPs; p = 0.002), as well as serum GFAP (serum GFAP-DPs; p = 0.001). In injured rats multiplex electrochemiluminescence analyses on PID3 revealed elevated serum tumor necrosis factor alpha (TNFα p = 0.01) and chemokine (CXC) ligand 1 (CXCL1). Chronically, that is, in PID13-16 CCI-veh rats, as compared with sham rats, gait deficits were demonstrated (p = 0.033) but then were reversed (p = 0.022) with EPO treatment. Diffusion tensor MRI of the ipsilateral and contralateral cortex and white matter in PID16-23 CCI-veh rats showed widespread injury and significant abnormalities of functional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD), and radial diffusivity (RD); MD, AD, and RD improved after EPO treatment. Chronically, P13-P28 CCI-veh rats also had elevated serum CXCL1 levels, which normalized in CCI-EPO rats. CONCLUSIONS Efficient translation of emerging neuro-reparative interventions dictates the use of age-appropriate preclinical models with human clinical trial-compatible biomarkers. In the present study, the authors showed that CCI produced chronic gait deficits in P12 rats that resolved with EPO treatment and that chronic imaging and serum biomarkers correlated with this improvement.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Newborn
- Biomarkers/blood
- Brain Injuries, Traumatic/blood
- Brain Injuries, Traumatic/complications
- Brain Injuries, Traumatic/diagnostic imaging
- Brain Injuries, Traumatic/drug therapy
- Calpain/metabolism
- Cerebral Cortex/drug effects
- Cerebral Cortex/metabolism
- Cytokines/blood
- Diffusion Magnetic Resonance Imaging
- Disease Models, Animal
- Epoetin Alfa/metabolism
- Erythropoietin/therapeutic use
- Female
- Gait Disorders, Neurologic/drug therapy
- Gait Disorders, Neurologic/etiology
- Gene Expression Regulation, Developmental/drug effects
- Glial Fibrillary Acidic Protein/metabolism
- Image Processing, Computer-Assisted
- Male
- Rats
- Receptors, Erythropoietin/metabolism
- Statistics, Nonparametric
- Symporters
- Time Factors
- K Cl- Cotransporters
Collapse
Affiliation(s)
- Shenandoah Robinson
- Brain Injury Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- F. M. Kirby Center for Neurobiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jesse L. Winer
- Brain Injury Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Justin Berkner
- Brain Injury Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Emergency Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lindsay A. S. Chan
- Brain Injury Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jesse L. Denson
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Jessie R. Maxwell
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Yirong Yang
- Department of Neurology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Laurel O. Sillerud
- Department of Neurology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Robert C. Tasker
- Brain Injury Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - William P. Meehan
- Brain Injury Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Sports Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rebekah Mannix
- Brain Injury Center, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Emergency Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lauren L. Jantzie
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, New Mexico
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico
| |
Collapse
|